Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 7.329
1.
Pak J Pharm Sci ; 37(1): 53-63, 2024 Jan.
Article En | MEDLINE | ID: mdl-38741400

The study focused on the neuroprotective role of Sorghum bicolor and vitamin C in the amelioration of oxidative stress and anxiety-like behavoiur induced by tramadol in male albino rats. The study design involved 7 groups and a control group with 5 male albino rats in each group. Tramadol (40 mg/kg) treatment was administered for 21 days. Tramadol 40mg/kg was administered in all groups. Pretreatment with varying doses of Sorghum bicolor and Vitamin C was done in three of the groups. Behavioral assessment of anxiety and locomotors actions of the groups were compared using Elevated Plus Maze (EPM) and Open Field Test (OFT). In conclusion, Sorghum bicolor and Vitamin C tramadol ameliorated oxidative stress and anxiety-like behaviour induced by tramadol. Pretreatment with Sorghum bicolor or vitamin C (100mg) can also reduced anxiogenic responses in male albino rats that are induced by chronic tramadol use.


Anxiety , Ascorbic Acid , Behavior, Animal , Oxidative Stress , Sorghum , Tramadol , Animals , Tramadol/pharmacology , Oxidative Stress/drug effects , Male , Ascorbic Acid/pharmacology , Anxiety/prevention & control , Anxiety/chemically induced , Anxiety/drug therapy , Rats , Behavior, Animal/drug effects , Antioxidants/pharmacology , Brain/drug effects , Brain/metabolism , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , Rats, Wistar , Analgesics, Opioid/pharmacology , Anti-Anxiety Agents/pharmacology , Maze Learning/drug effects
2.
Pak J Pharm Sci ; 37(1): 129-137, 2024 Jan.
Article En | MEDLINE | ID: mdl-38741409

Stress is described as a noxious stimulus that affects the health of an individual and alters body homeostasis resulting in changes the individual behavioural and metabolic condition. Synthesis of drug from plants has main interest due the significant medicinal values. The recent investigation was designed to examine the pharmacological impacts of Ficus carica leaves extract on stress. In this experiment, the rodents were randomly distributed as (n=6) control rats were kept at standard condition, second group of rats were exposed with different stressors and Third group of rodents was exposed to stress and treated with extract of ficus carica leaves at the dose of 100 mg/kg. Acute behavioural alteration was observed after 7 days and prolonged impact was monitored after the 28 days. The current finding showed that administration of Ficus carica leaves extract produced anxiolytic behaviours and decreased depression like symptoms in CUMS treated rats. It also increased stimulatory, ambulatory, locomotor activity and enhanced spatial working memory and recognition memory in CUMS exposed rats. So, it can be concluded from recent study that leaves of Ficus carica can be utilized as secure drug for curing physiological stress with less side effect profile.


Behavior, Animal , Disease Models, Animal , Ficus , Plant Extracts , Plant Leaves , Stress, Psychological , Animals , Ficus/chemistry , Plant Extracts/pharmacology , Behavior, Animal/drug effects , Stress, Psychological/drug therapy , Male , Rats , Rats, Wistar , Anti-Anxiety Agents/pharmacology , Depression/drug therapy
3.
Neurosci Biobehav Rev ; 161: 105648, 2024 Jun.
Article En | MEDLINE | ID: mdl-38565340

This narrative review describes the research on the effects of the association between environmental context and medications, suggesting the benefit of specific design interventions in adjunction to pharmacotherapy. The literature on Evidence-Based Design (EBD) studies and Neuro-Architecture show how contact with light, nature, and specific physical features of urban and interior architecture may enhance the effects of analgesic, anxiolytics, and antidepressant drugs. This interaction mirrors those already known between psychedelics, drugs of abuse, and setting. Considering that the physical feature of space is a component of the complex placebo configuration, the aim is to highlight those elements of built or natural space that may help to improve drug response in terms of efficacy, tolerability, safety, and compliance. Ecocebo, the integration of design approaches such as EBD and Neuro-Architecture may thus contribute to a more efficient, cost-sensitive, and sustainable pharmacotherapy.


Antidepressive Agents , Humans , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Analgesics/pharmacology , Environment
4.
Behav Pharmacol ; 35(4): 156-160, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38651975

Exposure to chronic caffeine during adolescence has been shown to produce decreased anxiety-like behaviors in rats as well as decreased immobility in the forced swim test (FST) suggesting an antidepressant-like effect. The effects of chronic caffeine on anxiety, however, have been found to be test-dependent and sexually dimorphic. In addition, decreased immobility in the FST has been argued to reflect a shift toward active coping behavior as opposed to an antidepressant-like effect. In order to further characterize the effects of adolescent caffeine exposure, the present experiment assessed the effects of caffeine on marble burying behavior in a two-zone marble burying task. There was no difference in the amount of time rats spent in the two zones failing to support a shift in coping strategy. Caffeine-exposed rats spent less time engaged in marble burying activity and buried slightly fewer marbles, suggesting an anxiolytic effect of caffeine. In addition, caffeine treated rats spent less time engaged in nondirected burying and slightly more time actively engaging with the marbles; however, these effects appeared to be sexually dimorphic as they were driven by larger changes in the females. Overall, these results support an anxiolytic effect of adolescent caffeine, with female behavior appearing to be more affected by caffeine than males.


Anxiety , Behavior, Animal , Caffeine , Animals , Caffeine/pharmacology , Caffeine/administration & dosage , Male , Anxiety/drug therapy , Female , Rats , Behavior, Animal/drug effects , Central Nervous System Stimulants/pharmacology , Anti-Anxiety Agents/pharmacology , Rats, Sprague-Dawley , Motor Activity/drug effects
5.
Pharmacol Biochem Behav ; 239: 173770, 2024 Jun.
Article En | MEDLINE | ID: mdl-38636813

The population of most countries in the world is increasing and understanding risk factors that can influence the health of the older population is critical. Older adults consume alcohol often in a risky, binge manner. Previous work has demonstrated that aged rats are more sensitive to many of the effects of acute ethanol. In the current project aged, adult, and adolescent female and male rats were tested on the elevated plus maze and open field following either a 1.0 g/kg alcohol injection or a saline injection. We report sex- and age-dependent effects whereas aged female rats, but not aged male rats, showed an increased anxiolytic effect of alcohol in the elevated plus maze while aged male rats, but not aged female rats, showed increased stimulatory movement in the open field. In addition, significant age effects were found for both female and male rats. It is proposed that the sex- and age-dependent effects reported in the current studies may be due to differential levels of alcohol-induced allopregnanolone for the anxiolytic effects and differential levels of alcohol-induced dopamine for the stimulatory effects. The current work provides insights into factors influencing alcohol consumption in older adults.


Aging , Anti-Anxiety Agents , Ethanol , Motor Activity , Animals , Male , Female , Rats , Ethanol/administration & dosage , Ethanol/pharmacology , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/administration & dosage , Aging/psychology , Motor Activity/drug effects , Behavior, Animal/drug effects , Anxiety/psychology , Anxiety/drug therapy , Age Factors , Sex Characteristics , Maze Learning/drug effects , Sex Factors
6.
Sci Rep ; 14(1): 9799, 2024 04 29.
Article En | MEDLINE | ID: mdl-38684743

This study investigated the potential anxiolytic properties of flavan-3-ols and aromatic resins through a combined computational and experimental approach. Network pharmacology techniques were utilized to identify potential anxiolytic targets and compounds by analyzing protein-protein interactions and KEGG pathway data. Molecular docking and simulation studies were conducted to evaluate the binding interactions and stability of the identified targets. Behavioral tests, including the elevated plus maze test, open field test, light-dark test, actophotometer, and holeboard test, were used to assess anxiolytic activity. The compound-target network analysis revealed complex interactions involving 306 nodes and 526 edges, with significant interactions observed and an average node degree of 1.94. KEGG pathway analysis highlighted pathways such as neuroactive ligand-receptor interactions, dopaminergic synapses, and serotonergic synapses as being involved in anxiety modulation. Docking studies on EGCG (Epigallocatechin gallate) showed binding energies of -9.5 kcal/mol for MAOA, -9.2 kcal/mol for SLC6A4, and -7.4 kcal/mol for COMT. Molecular dynamic simulations indicated minimal fluctuations, suggesting the formation of stable complexes between small molecules and proteins. Behavioral tests demonstrated a significant reduction in anxiety-like behavior, as evidenced by an increased number of entries into and time spent in the open arm of the elevated plus maze test, light-dark test, open field center activity, hole board head dips, and actophotometer beam interruptions (p < 0.05 or p < 0.01). This research provides a comprehensive understanding of the multi-component, multi-target, and multi-pathway intervention mechanisms of flavan-3-ols and aromatic resins in anxiety treatment. Integrated network and behavioral analyses collectively support the anxiolytic potential of these compounds and offer valuable insights for future research in this area.


Anti-Anxiety Agents , Anxiety , Catechin , Catechin/analogs & derivatives , Flavonoids , Molecular Docking Simulation , Animals , Flavonoids/chemistry , Flavonoids/pharmacology , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/chemistry , Anxiety/drug therapy , Catechin/pharmacology , Catechin/chemistry , Molecular Dynamics Simulation , Male , Network Pharmacology , Monoamine Oxidase/metabolism , Monoamine Oxidase/chemistry , Behavior, Animal/drug effects , Catechol O-Methyltransferase/metabolism , Catechol O-Methyltransferase/chemistry , Mice , Protein Binding
7.
Int J Neuropsychopharmacol ; 27(4)2024 Apr 01.
Article En | MEDLINE | ID: mdl-38577951

BACKGROUND: Anxiety disorders are highly prevalent and socio-economically costly. Novel pharmacological treatments for these disorders are needed because many patients do not respond to current agents or experience unwanted side effects. However, a barrier to treatment development is the variable and large placebo response rate seen in trials of novel anxiolytics. Despite this, the mechanisms that drive placebo responses in anxiety disorders have been little investigated, possibly due to low availability of convenient experimental paradigms. We aimed to develop and test a novel protocol for inducing placebo anxiolysis in the 7.5% CO2 inhalational model of generalized anxiety in healthy volunteers. METHODS: Following a baseline 20-minute CO2 challenge, 32 healthy volunteers were administered a placebo intranasal spray labelled as either the anxiolytic "lorazepam" or "saline." Following this, participants surreptitiously underwent a 20-minute inhalation of normal air. Post-conditioning, a second dose of the placebo was administered, after which participants completed another CO2 challenge. RESULTS: Participants administered sham "lorazepam" reported significant positive expectations of reduced anxiety (P = .001), but there was no group-level placebo effect on anxiety following CO2 challenge post-conditioning (Ps > .350). Surprisingly, we found many participants exhibited unexpected worsening of anxiety, despite positive expectations. CONCLUSIONS: Contrary to our hypothesis, our novel paradigm did not induce a placebo response, on average. It is possible that effects of 7.5% CO2 inhalation on prefrontal cortex function or behavior in line with a Bayesian predictive coding framework attenuated the effect of expectations on subsequent placebo response. Future studies are needed to explore these possibilities.


Anti-Anxiety Agents , Anxiety , Carbon Dioxide , Placebo Effect , Humans , Carbon Dioxide/administration & dosage , Carbon Dioxide/pharmacology , Male , Female , Adult , Young Adult , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/administration & dosage , Administration, Inhalation , Anxiety/drug therapy , Anxiety/chemically induced , Lorazepam/pharmacology , Lorazepam/administration & dosage , Double-Blind Method
8.
Anesthesiology ; 140(6): 1134-1152, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38498811

BACKGROUND: Dexmedetomidine has repeatedly shown to improve anxiety, but the precise neural mechanisms underlying this effect remain incompletely understood. This study aims to explore the role of corticotropin-releasing hormone-producing hypothalamic paraventricular nucleus (CRHPVN) neurons in mediating the anxiolytic effects of dexmedetomidine. METHODS: A social defeat stress mouse model was used to evaluate the anxiolytic effects induced by dexmedetomidine through the elevated plus maze, open-field test, and measurement of serum stress hormone levels. In vivo Ca2+ signal fiber photometry and ex vivo patch-clamp recordings were used to determine the excitability of CRHPVN neurons and investigate the specific mechanism involved. CRHPVN neuron modulation was achieved through chemogenetic activation or inhibition. RESULTS: Compared with saline, dexmedetomidine (40 µg/kg) alleviated anxiety-like behaviors. Additionally, dexmedetomidine reduced CRHPVN neuronal excitability. Chemogenetic activation of CRHPVN neurons decreased the time spent in the open arms of the elevated plus maze and in the central area of the open-field test. Conversely, chemogenetic inhibition of CRHPVN neurons had the opposite effect. Moreover, the suppressive impact of dexmedetomidine on CRHPVN neurons was attenuated by the α2-receptor antagonist yohimbine. CONCLUSIONS: The results indicate that the anxiety-like effects of dexmedetomidine are mediated via α2-adrenergic receptor-triggered inhibition of CRHPVN neuronal excitability in the hypothalamus.


Anxiety , Corticotropin-Releasing Hormone , Dexmedetomidine , Neurons , Paraventricular Hypothalamic Nucleus , Stress, Psychological , Animals , Dexmedetomidine/pharmacology , Mice , Corticotropin-Releasing Hormone/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Neurons/drug effects , Male , Anxiety/drug therapy , Stress, Psychological/psychology , Mice, Inbred C57BL , Anti-Anxiety Agents/pharmacology , Disease Models, Animal
9.
J Agric Food Chem ; 72(12): 6189-6202, 2024 Mar 27.
Article En | MEDLINE | ID: mdl-38501577

The hexapeptide YPVEPF with strong sleep-enhancing effects could be detected in rat brain after a single oral administration as we previously proved. In this study, the mechanism and molecular effects of YPVEPF in the targeted stress-induced anxiety mice were first investigated, and its key active structure was further explored. The results showed that YPVEPF could significantly prolong sleep duration and improve the anxiety indexes, including prolonging the time spent in the open arms and in the center. Meanwhile, YPVEPF showed strong sleep-enhancing effects by significantly increasing the level of the GABA/Glu ratio, 5-HT, and dopamine in brain and serum and regulating the anabolism of multiple targets, but the effects could be blocked by bicuculline and WAY100135. Moreover, the molecular simulation results showed that YPVEPF could stably bind to the vital GABAA and 5-HT1A receptors due to the vital structure of Tyr-Pro-Xaa-Xaa-Pro-, and the electrostatic and van der Waals energy played dominant roles in stabilizing the conformation. Therefore, YPVEPF displayed sleep-enhancing and anxiolytic effects by regulating the GABA-Glu metabolic pathway and serotoninergic system depending on distinctive self-folding structures with Tyr and two Pro repeats.


Anti-Anxiety Agents , Sleep Initiation and Maintenance Disorders , gamma-Aminobutyric Acid/analogs & derivatives , Rats , Mice , Animals , Caseins/metabolism , Receptors, GABA-A/metabolism , Serotonin , Anti-Anxiety Agents/pharmacology , Anxiety
10.
Exp Brain Res ; 242(4): 913-920, 2024 Apr.
Article En | MEDLINE | ID: mdl-38451318

Self-care behavior covers individual's health, life and well-being to maintain the necessary activities. The aim of this study is to examine the self-care and possible anxiolytic effects of high-intensity interval exercise (HIIT). Eight-week-old Wistar Albino male rats were divided into Control (n = 8), and Exercise (n = 8). Rat exercised for 38 min a day, 5 days a week, for 8 weeks The animals were then subjected to open field test and splash test, and the behaviors were video recorded. Student t test and Shapiro-Wilk test were used as statistical tests. In the exercise group, spray-induced grooming behavior increased significantly in terms of duration and frequency (p < 0.05), but no significant difference was observed in the latency of grooming (p > 0.05). In the open-field test, the total distance traveled, which is a locomotor activity parameter, did not change between the groups. Anxiolytic-like behaviors such as total rearing behavior, unsupported rearing, central time, and central region entries increased remarkably in the exercise group vs. control (p < 0.0001). Freezing as an anxiogenic behavior decreased in the exercise group positively (p < 0.0001). Intermittent high-intensity exercise improved and increased self-care behaviors. Further, the present study shows that HIIT has beneficial effects on different aspects of behaviors such as exploratory behaviors, increasing anxiolytic behaviors, and reducing anxiogenic behavior. The present study is a preclinical study that will pave the way for new studies.


Anti-Anxiety Agents , High-Intensity Interval Training , Rats , Animals , Humans , Anti-Anxiety Agents/pharmacology , Rats, Wistar , Self Care , Anxiety/therapy
11.
Neurosci Lett ; 826: 137723, 2024 Mar 15.
Article En | MEDLINE | ID: mdl-38467272

Cannabidiol (CBD), a non-psychoactive compound derived from the cannabis plant, has been confirmed to induce anxiolytic-like and antipsychotic-like effects. However, the exact mechanisms remain unclear. This study substantiated CBD's interaction with the 5-HT1A receptor (5-HT1AR) in vitro (CHO cells expressing human 5-HT1AR) and in vivo (rat lower lip retraction test, LLR test). We then assessed the impact of CBD in mice using the stress-induced hyperthermia (SIH) model and the phencyclidine (PCP)-induced negative symptoms of schizophrenia model, respectively. Concurrently, we investigated whether WAY-100635, a typical 5-HT1AR antagonist, could attenuate these effects. Furthermore, the neurotransmitter changes through high-performance liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) were studied. Results revealed that CBD exhibits selective 5-HT1AR agonists-mediated effects in the rat lower lip retraction test, aligning with the robust agonistic (EC50 = 1.75 µM) profile observed in CHO cells. CBD at 3 mg/kg significantly reduced SIH (ΔT), a response that WAY-100635 abolished. Chronic administration of CBD at 100 mg/kg mitigated the increase in PCP-induced immobility time in the forced swim test (FST) and tail suspension test (TST). Moreover, it induced significant alterations in gamma-aminobutyric acid (GABA) and norepinephrine (NE) levels within the hippocampus (HPC). Thus, we concluded that the 5-HT1AR mediates CBD's anxiolytic-like effects. Additionally, CBD's effects on the negative symptoms of schizophrenia may be linked to changes in GABA and NE levels in the hippocampus. These findings offer novel insights for advancing the exploration of CBD's anxiolytic-like and antipsychotic-like effects.


Anti-Anxiety Agents , Antipsychotic Agents , Cannabidiol , Cricetinae , Mice , Rats , Humans , Animals , Antipsychotic Agents/pharmacology , Anti-Anxiety Agents/pharmacology , Cannabidiol/pharmacology , Serotonin , Cricetulus , Chromatography, Liquid , Tandem Mass Spectrometry , gamma-Aminobutyric Acid
12.
Physiol Behav ; 277: 114506, 2024 Apr 01.
Article En | MEDLINE | ID: mdl-38432442

The cannabinoid system plays a key role in stress-related emotional symptoms such as anxiety. Citicoline is a supplemental substance with neuroprotective properties that alleviates anxiety-related behaviors. There is a relation between the actions of cannabinoids and cholinergic systems. So, we decided to evaluate the effects of intracerebroventricular (i.c.v.) infusion of cannabinoid CB1 receptor agents on citicoline-produced response to anxiety-like behaviors in the non-acute restraint stress (NARS) and acute restraint stress (ARS) mice. For i.c.v. microinjection of drugs, a guide cannula was inserted in the left lateral ventricle. ARS was induced by movement restraint for 4 h. Anxiety-related behaviors were assessed using an elevated plus maze (EPM). The results showed that induction of ARS for 4 h decreased the percentage of time spent in the open arms (%OAT) and the percentage of entries to the open arms (%OAE) without affecting locomotor activity, showing anxiogenic-like behaviors. i.c.v. infusion of ACPA (1 µg/mouse) induced an anxiolytic-like effect due to the enhancement of %OAT in the NARS and ARS mice. Nonetheless, i.c.v. microinjection of AM251 (1 µg/mouse) decreased %OAT in the NARS and ARS mice which suggested an anxiogenic-like response. Intraperitoneal (i.p.) administration of citicoline (80 mg/kg) induced an anxiolytic-like effect by the augmentation of %OAT in the ARS mice. Furthermore, when ACPA and citicoline were co-administrated, ACPA potentiated the anxiolytic-like effect induced by citicoline in the NARS and ARS mice. On the other hand, when AM251 and the citicoline were co-injected, AM251 reversed the anxiolytic-like response induced by the citicoline in the NARS and ARS mice. The results of this research exhibited an additive effect between citicoline and ACPA on the induction of anxiolytic-like response in the NARS and ARS mice. Our results indicated an interaction between citicoline and cannabinoid CB1 receptor drugs on the control of anxiety-like behaviors in the NARS and ARS mice.


Anti-Anxiety Agents , Cannabinoids , Mice , Animals , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Cytidine Diphosphate Choline , Receptor, Cannabinoid, CB1 , Anxiety/etiology , Anxiety/chemically induced , Cannabinoids/pharmacology
13.
Vet Anaesth Analg ; 51(3): 244-252, 2024.
Article En | MEDLINE | ID: mdl-38555213

OBJECTIVE: To evaluate the effect of oral tasipimidine on dog handling, ease of catheter placement and propofol and isoflurane requirements for anaesthesia. STUDY DESIGN: Placebo-controlled, randomized, blinded, experimental trial. ANIMALS: A group of seven adult Beagle dogs weighing (mean ± standard deviation) 13.1 ± 2.7 kg with a mean age of 18.6 ± 1 months. METHODS: The dogs underwent four treatments before induction of anaesthesia with propofol. PP: placebo orally (PO) 60 minutes before induction of anaesthesia followed by placebo (NaCl 0.9%) intravenously (IV). TP: tasipimidine 30 µg kg-1 (PO) 60 minutes before induction of anaesthesia followed by placebo (NaCl 0.9%) IV. TMP: tasipimidine 30 µg kg-1 PO 60 minutes before induction of anaesthesia followed by methadone 0.2 mg kg-1 IV. TMPD: tasipimidine 30 µg kg-1 PO 60 minutes before induction of anaesthesia followed by methadone 0.2 mg kg-1 and dexmedetomidine 1 µg kg-1 IV followed by a dexmedetomidine constant rate infusion of 1 µg kg-1 hour-1. Sedation, response to catheter placement, intubation quality, time to loss of consciousness, time to intubation, required dose of propofol and minimum alveolar isoflurane concentration preventing motor movement (MACNM) were determined. A mixed-model analysis or the Friedman and Mann-Whitney test were used; p-value < 0.05. RESULTS: Response to catheter placement did not differ between treatments. Tasipimidine alone reduced the propofol dose by 30%. Addition of methadone or methadone and dexmedetomidine reduced the propofol dose by 48% and 50%, respectively. Isoflurane MACNM was reduced by 19% in tasipimidine-medicated dogs, whereas in combination with methadone or methadone and dexmedetomidine, isoflurane MACNM was reduced by 35%. CONCLUSIONS AND CLINICAL RELEVANCE: An anxiolytic dose of tasipimidine induced mild signs of sedation in dogs and reduced propofol and isoflurane requirements to induce and maintain anaesthesia, which needs to be considered in an anaesthetic plan.


Anti-Anxiety Agents , Imidazoles , Propofol , Animals , Dogs , Male , Anti-Anxiety Agents/administration & dosage , Anti-Anxiety Agents/pharmacology , Propofol/administration & dosage , Propofol/pharmacology , Female , Isoflurane/administration & dosage , Anesthetics, Intravenous/administration & dosage , Anesthetics, Intravenous/pharmacology , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/pharmacology , Dexmedetomidine/administration & dosage , Dexmedetomidine/pharmacology , Quinolizines/administration & dosage , Quinolizines/pharmacology , Anesthetics, Inhalation/administration & dosage , Anesthetics, Inhalation/pharmacology
14.
Molecules ; 29(6)2024 Mar 21.
Article En | MEDLINE | ID: mdl-38543047

Close to 19% of the world population suffers from anxiety. Current medications for this chronic mental disorder have improved treatment over the last half century or more, but the newer anxiolytics have proved disappointing, and enormous challenges remain. Nitric oxide (NO), an intra- and inter-cellular messenger in the brain, is involved in the pathogenesis of anxiety. In particular, excessive NO production might contribute to its pathology. This implies that it might be useful to reduce nitrergic activity; therefore, molecules aiming to downregulate NO production such as NO synthase inhibitors (NOSIs) might be candidates. Here, it was intended to critically review advances in research on these emerging molecules for the treatment of anxiety disorders. Current assessment indicates that, although NOSIs are implicated in anxiety, their potential anti-anxiety action remains to be established.


Anti-Anxiety Agents , Nitric Oxide , Humans , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Anxiety Disorders/drug therapy , Anxiety/drug therapy , Anxiety/etiology , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use
15.
J Neurosci Res ; 102(3): e25306, 2024 Mar.
Article En | MEDLINE | ID: mdl-38468573

Finasteride is used in female-pattern hair loss, hirsutism, and polycystic ovarian syndrome. It inhibits 5α-reductase, which is an important enzyme in the biosynthesis of neurosteroids. The effects of finasteride treatment on mental health in female patients as well as the effects of repeated/chronic finasteride administration in female rodents are still unknown. Accordingly, in our study, we administered finasteride (10, 30, or 100 mg/Kg, s.c.) for 6 days in female rats and evaluated behavior, plasma steroid levels, and synaptic plasticity. Depression-like behavior was evaluated using forced swim test (FST) and splash test. Anxiety-like behavior was evaluated using novelty-suppressed feeding task (NSFT), elevated plus maze (EPM), open field test (OFT), and light-dark test (LDT). Plasma steroid levels were assessed using ELISA and synaptic plasticity by field potential recordings. We observed that finasteride decreased total immobility duration in FST, indicating antidepressant-like effect and decreased the latency to first bite in NSFT, showing anxiolytic-like effect. We also found a significant increase in plasma estradiol and a significant decrease in plasma corticosterone level. Furthermore, field potential recordings showed that finasteride increased hippocampal long-term potentiation. These results indicate that repeated finasteride administration in female rats may have antidepressant- and anxiolytic-like effect, which might be mediated by enhanced estradiol levels or decreased corticosterone levels. Further studies are required to validate the molecular mechanisms underlying the effects of finasteride in female rats. Understanding the mechanisms will help us in developing novel neurosteroid-based therapeutics in the treatment of neuropsychiatric disorders in women.


Anti-Anxiety Agents , Finasteride , Humans , Rats , Female , Animals , Finasteride/adverse effects , Anti-Anxiety Agents/pharmacology , Corticosterone , Depression/drug therapy , Steroids , Estradiol , Antidepressive Agents/pharmacology , Neuronal Plasticity
16.
Neurochem Int ; 175: 105720, 2024 May.
Article En | MEDLINE | ID: mdl-38458538

The anteroventral bed nucleus of stria terminalis (avBNST) is a key brain region which involves negative emotional states, such as anxiety. The most neurons in the avBNST are GABAergic, and it sends GABAergic projections to the ventral tegmental area (VTA) and the dorsal raphe nucleus (DRN), respectively. The VTA and DRN contain dopaminergic and serotonergic cell groups in the midbrain which regulate anxiety-like behaviors. However, it is unclear the role of GABAergic projections from the avBNST to the VTA and the DRN in the regulation of anxiety-like behaviors, particularly in Parkinson's disease (PD)-related anxiety. In the present study, unilateral 6-hydroxydopamine (6-OHDA) lesions of the substantia nigra pars compacta in rats induced anxiety-like behaviors, and decreased level of dopamine (DA) in the basolateral amygdala (BLA). Chemogenetic activation of avBNSTGABA-VTA or avBNSTGABA-DRN pathway induced anxiety-like behaviors and decreased DA or 5-HT release in the BLA in sham and 6-OHDA rats, while inhibition of avBNSTGABA-VTA or avBNSTGABA-DRN pathway produced anxiolytic-like effects and increased level of DA or 5-HT in the BLA. These findings suggest that avBNST inhibitory projections directly regulate dopaminergic neurons in the VTA and serotonergic neurons in the DRN, and the avBNSTGABA-VTA and avBNSTGABA-DRN pathways respectively exert impacts on PD-related anxiety-like behaviors.


Anti-Anxiety Agents , Parkinson Disease , Septal Nuclei , Rats , Animals , Dorsal Raphe Nucleus/metabolism , Ventral Tegmental Area/metabolism , Serotonin/metabolism , Septal Nuclei/metabolism , Oxidopamine/toxicity , Anxiety , Parkinson Disease/metabolism , Dopamine/metabolism , Anti-Anxiety Agents/pharmacology , Dopaminergic Neurons/metabolism , gamma-Aminobutyric Acid/metabolism
17.
Eur J Pharmacol ; 969: 176454, 2024 Apr 15.
Article En | MEDLINE | ID: mdl-38417607

Estrogen hormone replacement therapy (EHRT), improving women's life quality at menopause, reduces anxiety and depression symptoms associated with ovarian hormonal decline. However, its potential adverse effects, like thromboembolism and cancer risk, limit its use. Prolame is a synthetic 17ß-amino estrogen with antithrombotic actions that exerts anxiolytic- and antidepressant-like effects on young adult ovariectomized female rats. It is unknown if prolame's effects may be observed in age and endocrine conditions emulating menopause. This study aimed to identify the antidepressant- and anxiolytic-like effects of prolame and E2 (used as a reference estrogen treatment) in middle-aged female rats coursing with irregular cycles, in two different conditions: ovariectomized or gonadally intact. Results were compared with those from young adult ovariectomized rats. Prolame (60 or 120 µg/kg), 17ß-estradiol (E2, 40 or 80 µg/kg), or vehicle were chronically administered, and their effects were evaluated in the elevated plus-maze, defensive burying behavior test, open field test, and forced swimming test. Uterotrophic actions were estimated by uterine weight related to body weight. Prolame and E2 produced robust anxiolytic- and antidepressant-like effects in young adult ovariectomized rats, but these effects were absent in gonadally intact middle-aged rats. Interestingly, only prolame induced anxiolytic- and antidepressant-like effects in middle-aged ovariectomized rats. Uterotrophic effects of prolame were weaker than E2 effects, notably in middle-aged females. Altogether, present data support the notion that prolame has the potential to be considered an EHRT with relevant psychoactive actions and with apparently lower adverse-side effects, especially in middle-aged populations.


Anti-Anxiety Agents , Estrenes , Humans , Rats , Female , Animals , Middle Aged , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Rats, Wistar , Estradiol/pharmacology , Estradiol/therapeutic use , Estrogens/pharmacology , Estrogens/therapeutic use , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Ovariectomy/adverse effects
18.
Eur J Pharmacol ; 969: 176394, 2024 Apr 15.
Article En | MEDLINE | ID: mdl-38331342

TSPO, translocator protein (18 kDa) ligands have demonstrated consistent antidepression and anxiolytic effects in several preclinical studies. This study aimed to examine whether YL-IPA08[N-ethyl-N-(2-pyridinylmethyl)-2-(3,4-ichlorophenyl) -7-methylimidazo [1,2-a] pyridine-3-acetamide hydrochloride], a potent and selective TSPO ligand synthesized by our institute, could alleviate anxiety-related behaviors induced by electric shock (ES) and investigate its underlying mechanism. As expected, we showed that chronic treatment with YL-IPA08 significantly reversed anxiety-related behaviors induced by electrical stimulation (0.5 mA, 12 times, duration 1s, interval 10s) exposure. Using the analysis of RNA-sequencing (RNA-seq) technology, it was found that the differential genes associated with the anxiolytic effect of YL-IPA08 were mainly related to synaptic plasticity. Furthermore, YL-IPA08 restored the decreased levels of brain-derived neurotrophic factor (BDNF), synapse-related protein (e.g. synapsin-1 and post-synaptic density95, PSD95), and the number of doublecortin (DCX) + neurons in the hippocampus of post-ES mice. In addition, YL-IPA08 also enhanced the dendritic complexity and dendritic spine density of hippocampal dentate gyrus (DG) granule neurons. Meanwhile, the induction of long-term potentiation (LTP) was significantly enhanced by YL-IPA08. In summary, the findings from the current study showed that YL-IPA08 exerted a clear anxiolytic effect, which might be partially mediated by promoting hippocampal neuroplasticity.


Anti-Anxiety Agents , Imidazoles , Mice , Animals , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Ligands , Hippocampus , Pyridines/pharmacology , Neuronal Plasticity
19.
Behav Brain Res ; 463: 114899, 2024 Apr 12.
Article En | MEDLINE | ID: mdl-38342379

Juvenile stress (JS) is a known risk factor for the development of alcohol use disorder (AUD) and post-traumatic stress disorder (PTSD), both of which are frequently co-morbid. Data suggest there may be common, genetically-influenced biological responses to stress that contribute to the development of both AUD and PTSD. The present study investigated the impact of JS on contextual fear learning and extinction, as well as corticosterone (CORT) responses before and after JS, before and after contextual fear conditioning (CFC), and after fear extinction in male and female high-alcohol-preferring (HAP2) and low-alcohol-preferring (LAP2) mouse lines. We also measured unconditioned anxiety-related behavior in the light-dark-transition test before CFC. HAP2 and LAP2 mice did not differ in fear acquisition, but HAP2 mice showed faster fear extinction compared to LAP2 mice. No effects of JS were seen in HAP2 mice, whereas in LAP2 mice, JS reduced fear acquisition in males and facilitated fear extinction in females. Females showed greater fear-related behavior relative to males, regardless of subgroup. HAP2 males demonstrated more anxiolytic-like responses than LAP2 males and LAP2 females demonstrated more anxiolytic-like responses than LAP2 males in the light-dark transition test. HAP2 and LAP2 mice did not differ in CORT during the juvenile stage; however, adult LAP2 mice showed greater CORT levels than HAP2 mice at baseline and after CFC and extinction testing. These findings build upon prior work in these unique mouse lines that differ in genetic propensity toward alcohol preference and provide new information regarding contextual fear learning and extinction mechanisms theorized to contribute to co-morbid AUD and PTSD.


Alcoholism , Anti-Anxiety Agents , Mice , Female , Male , Animals , Fear , Anti-Anxiety Agents/pharmacology , Extinction, Psychological , Ethanol/pharmacology , Alcoholism/genetics , Anxiety
20.
Neurosci Lett ; 824: 137688, 2024 Feb 28.
Article En | MEDLINE | ID: mdl-38360146

This research was done to examine the combination of citalopram, an antidepressant drug, and omega-3 in a mice model of depression. Mice received citalopram (1 and 2 mg/kg) or omega-3 (10 and 20 mg/kg) daily over 30 days. Then, they were exposed to acute and chronic restraint stress to assess the possible increasing effect of omega-3 on the antidepressant and anxiolytic effects of citalopram. Elevated plus-maze (EPM) and forced swimming test (FST) were used to assess anxiety and depression symptoms in non-restraint stress (NRS), acute restraint stress (ARS), and chronic restraint stress (CRS) mice. The results indicated that induction of acute and chronic restraint stress reduced %OAT (Open arm time) and %OAE (Open arm entrance) in the EPM test but enhanced immobility time in the FST, showing anxiogenic- and depressive-like effects. These stresses reduced the stability of pyramidal neurons in the prefrontal cortex (PFC) and hippocampus. Aone and combination administration with citalopram and omega-3 induced anxiolytic- and antidepressant-like effects in NRS, ARS, and CRS mice. This combination usage increased the stability of pyramidal neurons in the PFC and hippocampus. These results suggested an interaction between citalopram and omega-3 upon the induction of anxiolytic- and antidepressant-like effects as well as augmentation of the ratio of pyramidal live to dark neurons in the PFC and hippocampus of the ARS and CRS mice.


Anti-Anxiety Agents , Fatty Acids, Omega-3 , Mice , Animals , Citalopram/pharmacology , Anti-Anxiety Agents/pharmacology , Depression/drug therapy , Fatty Acids, Omega-3/pharmacology , Behavior, Animal , Anxiety/drug therapy , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Brain , Pyramidal Cells
...