Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 633
Filter
1.
J Biol Chem ; 300(1): 105528, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38043794

ABSTRACT

Parasitic flatworms cause various clinical and veterinary infections that impart a huge burden worldwide. The most clinically impactful infection is schistosomiasis, a neglected tropical disease caused by parasitic blood flukes. Schistosomiasis is treated with praziquantel (PZQ), an old drug introduced over 40 years ago. New drugs are urgently needed, as while PZQ is broadly effective it suffers from several limitations including poor efficacy against juvenile worms, which may prevent it from being completely curative. An old compound that retains efficacy against juvenile worms is the benzodiazepine meclonazepam (MCLZ). However, host side effects caused by benzodiazepines preclude development of MCLZ as a drug and MCLZ lacks an identified parasite target to catalyze rational drug design for engineering out human host activity. Here, we identify a transient receptor potential ion channel of the melastatin subfamily, named TRPMMCLZ, as a parasite target of MCLZ. MCLZ potently activates Schistosoma mansoni TRPMMCLZ through engagement of a binding pocket within the voltage-sensor-like domain of the ion channel to cause worm paralysis, tissue depolarization, and surface damage. TRPMMCLZ reproduces all known features of MCLZ action on schistosomes, including a lower activity versus Schistosoma japonicum, which is explained by a polymorphism within this voltage-sensor-like domain-binding pocket. TRPMMCLZ is distinct from the TRP channel targeted by PZQ (TRPMPZQ), with both anthelmintic chemotypes targeting unique parasite TRPM paralogs. This advances TRPMMCLZ as a novel druggable target that could circumvent any target-based resistance emerging in response to current mass drug administration campaigns centered on PZQ.


Subject(s)
Anthelmintics , Clonazepam , Schistosomiasis mansoni , TRPM Cation Channels , Animals , Humans , Anthelmintics/pharmacology , Benzodiazepines/pharmacology , Benzodiazepinones/pharmacology , Clonazepam/analogs & derivatives , Clonazepam/pharmacology , Praziquantel/pharmacology , Schistosoma mansoni/drug effects , Schistosoma mansoni/metabolism , Schistosomiasis mansoni/drug therapy , TRPM Cation Channels/agonists
2.
Neurol Sci ; 44(1): 115-128, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36112279

ABSTRACT

BACKGROUND: The objectives of this review and meta-analysis of polysomnographic data are those to focus on the clinical use of clonazepam for the management of sleep disorders by re-analyzing clinical trials and randomized clinical trials which have been published in peer-reviewed journals. METHODS: A review of the literature including clinical trials and randomized controlled trials was performed in PubMed®, in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement protocol. A random effects model meta-analysis was then carried out for the four more frequently reported polysomnographic measures: total sleep time, sleep latency, sleep efficiency, and periodic leg movement during sleep (PLMS) index. RESULTS: A total of 33 articles were retrieved and screened in full text, of which 18 met the criteria for review; among the latter, nine met the criteria for meta-analysis. The studies included in the review involved patients with insomnia, REM sleep behavior disorder, sleep bruxism, and restless leg syndrome or PLMS which reported, most often, an increase in total sleep time with clonazepam. A clear sleep-promoting effect of clonazepam was found also by meta-analysis. DISCUSSION AND CONCLUSIONS: Our results indicate that the pharmacological treatment of sleep disorders with clonazepam must always be personalized according to the type of patient, the risk of addiction and the concomitant presence of respiratory disorders are key factors to take into account. However, in light of the clinical evidence of the few studies in the literature on the different types of disorders, more studies on the use of clonazepam (also in association with first choice treatments) are definitely needed.


Subject(s)
Clonazepam , Restless Legs Syndrome , Humans , Clonazepam/therapeutic use , Clonazepam/pharmacology , Polysomnography/methods , Restless Legs Syndrome/complications , Leg , Sleep
3.
Mol Biol Rep ; 49(9): 9041-9049, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35690686

ABSTRACT

BACKGROUND: Newly identified multifunctional peptidergic modulators of stress responses: neuromedin U (NMU) and neuropeptide S (NPS) are involved in the wide spectrum of brain functions. However, there are no reports dealing with potential molecular relationships between the action of diverse anxiolytic or antidepressant drugs and NMU and NPS signaling in the brain. The present work was therefore focused on local expression of the aforementioned stress-related neuropeptides in the rat brain after long-term treatment with escitalopram and clonazepam. METHODS: Studies were carried out on adult, male Sprague-Dawley rats that were divided into 3 groups: animals injected with saline (control) and experimental individuals treated with escitalopram (at single dose 5 mg/kg daily), and clonazepam (at single dose 0.5 mg/kg). All individuals were sacrificed under anaesthesia and the whole brain excised. Total mRNA was isolated from homogenized samples of amygdala, hippocampus, hypothalamus, thalamus, cerebellum and brainstem. Real time-PCR method was used for estimation of related NPS, NPS receptor (NPSR), NMU, NMU and receptor 2 (NMUR2) mRNA expression. The whole brains were also sliced for general immunohistochemical assessment of the neuropeptides expression. RESULTS: Chronic administration of clonazepam resulted in an increase of NMU mRNA expression and formation of NMU-expressing fibers in the amygdala, while escitalopram produced a significant decrease in NPSR mRNA level in hypothalamus. Long-term escitalopram administration affects the local expression of examined neuropeptides mRNA in a varied manner depending on the brain structure. CONCLUSIONS: Pharmacological effects of escitalopram may be connected with local at least partially NPSR-related alterations in the NPS/NMU/NMUR2 gene expression at the level selected rat brain regions. A novel alternative mode of SSRI action can be therefore cautiously proposed.


Subject(s)
Anxiety , Brain , Clonazepam , Escitalopram , GABA Modulators , Neuropeptides , Receptors, Neuropeptide , Receptors, Neurotransmitter , Animals , Anxiety/drug therapy , Brain/drug effects , Brain/metabolism , Clonazepam/pharmacology , Clonazepam/therapeutic use , Escitalopram/pharmacology , Escitalopram/therapeutic use , GABA Modulators/pharmacology , GABA Modulators/therapeutic use , Male , Neuropeptides/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide/metabolism , Receptors, Neurotransmitter/metabolism
4.
Biosensors (Basel) ; 12(2)2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35200335

ABSTRACT

Rapid eye movement (REM) sleep behavior disorder (RBD) is associated with Parkinson's disease (PD). In this study, a smartwatch-based sensor is utilized as a convenient tool to detect the abnormal RBD phenomenon in PD patients. Instead, a questionnaire with sleep quality assessment and sleep physiological indices, such as sleep stage, activity level, and heart rate, were measured in the smartwatch sensors. Therefore, this device can record comprehensive sleep physiological data, offering several advantages such as ubiquity, long-term monitoring, and wearable convenience. In addition, it can provide the clinical doctor with sufficient information on the patient's sleeping patterns with individualized treatment. In this study, a three-stage sleep staging method (i.e., comprising sleep/awake detection, sleep-stage detection, and REM-stage detection) based on an accelerometer and heart-rate data is implemented using machine learning (ML) techniques. The ML-based algorithms used here for sleep/awake detection, sleep-stage detection, and REM-stage detection were a Cole-Kripke algorithm, a stepwise clustering algorithm, and a k-means clustering algorithm with predefined criteria, respectively. The sleep staging method was validated in a clinical trial. The results showed a statistically significant difference in the percentage of abnormal REM between the control group (1.6 ± 1.3; n = 18) and the PD group (3.8 ± 5.0; n = 20) (p = 0.04). The percentage of deep sleep stage in our results presented a significant difference between the control group (38.1 ± 24.3; n = 18) and PD group (22.0 ± 15.0, n = 20) (p = 0.011) as well. Further, our results suggested that the smartwatch-based sensor was able to detect the difference of an abnormal REM percentage in the control group (1.6 ± 1.3; n = 18), PD patient with clonazepam (2.0 ± 1.7; n = 10), and without clonazepam (5.7 ± 7.1; n = 10) (p = 0.007). Our results confirmed the effectiveness of our sensor in investigating the sleep stage in PD patients. The sensor also successfully determined the effect of clonazepam on reducing abnormal REM in PD patients. In conclusion, our smartwatch sensor is a convenient and effective tool for sleep quantification analysis in PD patients.


Subject(s)
Clonazepam/pharmacology , Parkinson Disease , REM Sleep Behavior Disorder , Algorithms , Humans , Parkinson Disease/diagnosis , REM Sleep Behavior Disorder/complications , REM Sleep Behavior Disorder/diagnosis , Sleep
5.
Biochem Pharmacol ; 192: 114711, 2021 10.
Article in English | MEDLINE | ID: mdl-34324871

ABSTRACT

Ample evidence indicates that maternal immune activation (MIA) during gestation is linked to an increased risk for neurodevelopmental and psychiatric disorders, such as autism spectrum disorder (ASD), anxiety and depression, in offspring. However, the underlying mechanism for such a link remains largely elusive. Here, we performed RNA sequencing (RNA-seq) to examine the transcriptional profiles changes in mice in response to MIA and identified that the expression of Scn1a gene, encoding the pore-forming α-subunit of the brain voltage-gated sodium channel type-1 (NaV1.1) primarily in fast-spiking inhibitory interneurons, was significantly decreased in the medial prefrontal cortex (mPFC) of juvenile offspring after MIA. Moreover, diminished excitatory drive onto interneurons causes reduction of spontaneous gamma-aminobutyric acid (GABA)ergic neurotransmission in the mPFC of MIA offspring, leading to hyperactivity in this brain region. Remarkably, treatment with low-dose benzodiazepines clonazepam, an agonist of GABAA receptors, completely prevented the behavioral abnormalities, including stereotypies, social deficits, anxiety- and depression-like behavior, via increasing inhibitory neurotransmission as well as decreasing neural activity in the mPFC of MIA offspring. Our results demonstrate that decreased expression of NaV1.1 in the mPFC leads to abnormalities in maternal inflammation-related behaviors and provides a potential therapeutic strategy for the abnormal behavioral phenotypes observed in the offspring exposed to MIA.


Subject(s)
Clonazepam/therapeutic use , GABA Modulators/therapeutic use , GABAergic Neurons/immunology , Mental Disorders/immunology , Prenatal Exposure Delayed Effects/immunology , Synaptic Transmission/immunology , Animals , Clonazepam/pharmacology , Female , GABA Modulators/pharmacology , GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Agonists/therapeutic use , GABAergic Neurons/chemistry , GABAergic Neurons/drug effects , Male , Mental Disorders/chemically induced , Mental Disorders/prevention & control , Mice , Mice, Inbred C57BL , NAV1.1 Voltage-Gated Sodium Channel/biosynthesis , NAV1.1 Voltage-Gated Sodium Channel/immunology , Poly I-C/toxicity , Prefrontal Cortex/drug effects , Prefrontal Cortex/immunology , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/prevention & control , Receptors, GABA-A/immunology , Synaptic Transmission/drug effects
6.
Can J Microbiol ; 67(8): 599-612, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33481681

ABSTRACT

Bacterial resistance has become one of the most serious public health problems, globally, and drug repurposing is being investigated to speed up the identification of effective drugs. The aim of this study was to investigate the repurposing of escitalopram oxalate and clonazepam drugs individually, and in combination with the antibiotics ciprofloxacin and sulfamethoxazole-trimethoprim, to treat multidrug-resistant (MDR) microorganisms and to evaluate the potential chemical nuclease activity. The minimum inhibitory concentration, minimum bactericidal concentration, fractional inhibitory concentration index, and tolerance level were determined for each microorganism tested. In vitro antibacterial activity was evaluated against 47 multidrug-resistant clinical isolates and 11 standard bacterial strains from the American Type Culture Collection. Escitalopram oxalate was mainly active against Gram-positive bacteria, and clonazepam was active against both Gram-positive and Gram-negative bacteria. When associated with the two antibiotics mentioned, they had a significant synergistic effect. Clonazepam cleaved plasmid DNA, and the mechanisms involved were oxidative and hydrolytic. These results indicate the potential for repurposing these non-antibiotic drugs to treat bacterial infections. However, further studies on the mechanism of action of these drugs should be performed to ensure their safe use.


Subject(s)
Ciprofloxacin , Gram-Negative Bacteria , Anti-Bacterial Agents/pharmacology , Ciprofloxacin/pharmacology , Citalopram/pharmacology , Clonazepam/pharmacology , DNA , Drug Repositioning , Drug Resistance, Multiple, Bacterial , Gram-Positive Bacteria , Humans , Microbial Sensitivity Tests , Plasmids/genetics , Sulfamethoxazole/pharmacology , Trimethoprim/pharmacology
7.
Int J Mol Sci ; 23(1)2021 Dec 21.
Article in English | MEDLINE | ID: mdl-35008465

ABSTRACT

Benzodiazepines (BZDs) produce versatile pharmacological actions through positive modulation of GABAA receptors (GABAARs). A previous study has demonstrated that high concentrations of diazepam potentiate GABA currents on the α1ß2γ2 and α1ß2 GABAARs in a flumazenil-insensitive manner. In this study, the high-concentration effects of BZDs and their sensitivity to flumazenil were determined on synaptic (α1ß2γ2, α2ß2γ2, α5ß2γ2) and extra-synaptic (α4ß2δ) GABAARs using the voltage-clamp electrophysiology technique. The in vivo evaluation of flumazenil-insensitive BZD effects was conducted in mice via the loss of righting reflex (LORR) test. Diazepam induced biphasic potentiation on the α1ß2γ2, α2ß2γ2 and α5ß2γ2 GABAARs, but did not affect the α4ß2δ receptor. In contrast to the nanomolar component of potentiation, the second potentiation elicited by micromolar diazepam was insensitive to flumazenil. Midazolam, clonazepam, and lorazepam at 200 µM exhibited similar flumazenil-insensitive effects on the α1ß2γ2, α2ß2γ2 and α5ß2γ2 receptors, whereas the potentiation induced by 200 µM zolpidem or triazolam was abolished by flumazenil. Both the GABAAR antagonist pentylenetetrazol and Fa173, a proposed transmembrane site antagonist, abolished the potentiation induced by 200 µM diazepam. Consistent with the in vitro results, flumazenil antagonized the zolpidem-induced LORR, but not that induced by diazepam or midazolam. Pentylenetetrazol and Fa173 antagonized the diazepam-induced LORR. These findings support the existence of non-classical BZD binding sites on certain GABAAR subtypes and indicate that the flumazenil-insensitive effects depend on the chemical structures of BZD ligands.


Subject(s)
Benzodiazepines/pharmacology , Flumazenil/pharmacology , Receptors, GABA-A/metabolism , Animals , Animals, Outbred Strains , Clonazepam/pharmacology , Diazepam/pharmacology , Female , GABA Antagonists/pharmacology , Male , Mice , Midazolam/pharmacology , Xenopus laevis/metabolism , gamma-Aminobutyric Acid/metabolism
8.
Biol Pharm Bull ; 43(12): 1993-1996, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-33028749

ABSTRACT

The effect of KB-R7943, an inhibitor of the plasmalemmal Na+-Ca2+ exchanger, on mitochondrial Ca2+ transporters was examined with membrane-permeabilized cardiomyocyte-derived H9c2 cells expressing the fluorescent Ca2+ indicator, yellow cameleon 3.1, in the mitochondria. KB-R7943, as well as ruthenium red, inhibited the rise in mitochondrial Ca2+ on increasing the extramitochondrial Ca2+ concentration from 0 nM to 300 nM. CGP-37157, but not KB-R7943, inhibited the decline in mitochondrial Ca2+on return to Ca2+ free extramitochondrial solution. These results indicated that KB-R7943 has inhibitory effects on the mitochondrial Ca2+ uniporter, but not on the mitochondrial Na+-Ca2+ exchanger.


Subject(s)
Calcium Channels/drug effects , Calcium/metabolism , Sodium-Calcium Exchanger/drug effects , Thiourea/analogs & derivatives , Animals , Cell Membrane/metabolism , Cells, Cultured , Clonazepam/analogs & derivatives , Clonazepam/pharmacology , Mitochondria/metabolism , Myocytes, Cardiac/drug effects , Rats , Sodium/metabolism , Sodium-Calcium Exchanger/metabolism , Thiazepines/pharmacology , Thiourea/pharmacology
9.
Pharmacol Biochem Behav ; 197: 173014, 2020 10.
Article in English | MEDLINE | ID: mdl-32795565

ABSTRACT

Misuse of prescription medications has risen to popularity. Reasons for this practice include the self-medication of sleep and psychiatric disorders and attempts to counteract the dysphoric side effects of stimulant drugs. Clonazepam, a commonly prescribed benzodiazepine, has been increasingly used as a countermeasure to cocaine side-effects, including sleep reduction and anxiety. As both substances may impair sleep and aggravate psychiatric conditions, this study aimed to evaluate the long-term effects of the interaction of clonazepam and cocaine on anxiety-like behavior, and the short-term effects of this drug combination on sleep using male Wistar rats. Animals received saline, cocaine (15 mg/kg), clonazepam (1.25 mg/kg) or both drugs for 16 days. Sleep recording was performed on the first day of treatment to evaluate acute treatment effects. One day after the end of the treatment period, the open field and elevated plus-maze tests were used to assess anxiety-like behavior. Blood samples were collected for analysis of corticosterone levels. Rats receiving both drugs presented an increase in impulsivity when moving between arms in the elevated plus-maze and a reduction in exploratory behavior in the open field test. These findings suggest the presence of a withdrawal behavioral syndrome, which can manifest as a paradoxical increase in exploratory activity after a period without receiving the drug and may indicate the development of dependence. Combined treatment reduced paradoxical sleep time and increased its onset latency. There was no significant difference regarding corticosterone levels across any group. Our results contribute to the understanding of the risks of combining cocaine and clonazepam. Association of these drugs may impair sleep architecture and aggravate the dependence symptoms already seen when these substances are used separately. These findings may be useful in helping to counteract the impairments resulting from the combined use of these 2 substances and to raise awareness of these associated risks.


Subject(s)
Anxiety/chemically induced , Behavior, Animal/drug effects , Clonazepam/adverse effects , Clonazepam/pharmacology , Cocaine/adverse effects , Cocaine/pharmacology , Sleep, REM/drug effects , Substance Withdrawal Syndrome/etiology , Animals , Anxiety/blood , Clonazepam/administration & dosage , Cocaine/administration & dosage , Corticosterone/blood , Exploratory Behavior/drug effects , Male , Open Field Test/drug effects , Rats , Rats, Wistar , Substance Withdrawal Syndrome/blood
10.
Int J Mol Sci ; 21(9)2020 Apr 30.
Article in English | MEDLINE | ID: mdl-32366006

ABSTRACT

Benzodiazepines (BZDs) are widely used in patients of all ages. Unlike adults, neonatal animals treated with BZDs exhibit a variety of behavioral deficits later in life; however, the mechanisms underlying these deficits are poorly understood. This study aims to examine whether administration of clonazepam (CZP; 1 mg/kg/day) in 7-11-day-old rats affects Gama aminobutyric acid (GABA)ergic receptors in both the short and long terms. Using RT-PCR and quantitative autoradiography, we examined the expression of the selected GABAA receptor subunits (α1, α2, α4, γ2, and δ) and the GABAB B2 subunit, and GABAA, benzodiazepine, and GABAB receptor binding 48 h, 1 week, and 2 months after treatment discontinuation. Within one week after CZP cessation, the expression of the α2 subunit was upregulated, whereas that of the δ subunit was downregulated in both the hippocampus and cortex. In the hippocampus, the α4 subunit was downregulated after the 2-month interval. Changes in receptor binding were highly dependent on the receptor type, the interval after treatment cessation, and the brain structure. GABAA receptor binding was increased in almost all of the brain structures after the 48-h interval. BZD-binding was decreased in many brain structures involved in the neuronal networks associated with emotional behavior, anxiety, and cognitive functions after the 2-month interval. Binding of the GABAB receptors changed depending on the interval and brain structure. Overall, the described changes may affect both synaptic development and functioning and may potentially cause behavioral impairment.


Subject(s)
Clonazepam/pharmacology , Receptors, GABA-A/metabolism , Receptors, GABA-B/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Benzodiazepines/pharmacology , Brain/drug effects , Brain/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Protein Binding , Rats , Rats, Inbred WF , Real-Time Polymerase Chain Reaction
11.
Br J Pharmacol ; 177(21): 4975-4989, 2020 11.
Article in English | MEDLINE | ID: mdl-32415690

ABSTRACT

BACKGROUND AND PURPOSE: Resurgence in the use of chloroquine as a potential treatment for COVID-19 has seen recent cases of fatal toxicity due to unintentional overdoses. Protocols for the management of poisoning recommend diazepam, although there are uncertainties in its pharmacology and efficacy in this context. The aim was to assess the effects of diazepam in experimental models of chloroquine cardiotoxicity. EXPERIMENTAL APPROACH: In vitro experiments involved cardiac tissues isolated from rats and incubated with chloroquine alone or in combination with diazepam. In vivo models of toxicity involved chloroquine administered intravenously to pentobarbitone-anaesthetised rats and rabbits. Randomised, controlled treatment studies in rats assessed diazepam, clonazepam and Ro5-4864 administered: (i) prior, (ii) during and (iii) after chloroquine and the effects of diazepam: (iv) at high dose, (v) in urethane-anaesthetised rats and (vi) co-administered with adrenaline. KEY RESULTS: Chloroquine decreased the developed tension of left atria, prolonged the effective refractory period of atria, ventricular tissue and right papillary muscles, and caused dose-dependent impairment of haemodynamic and electrocardiographic parameters. Cardiac arrhythmias indicated impairment of atrioventricular conduction. Studies (i), (ii) and (v) showed no differences between treatments and control. Diazepam increased heart rate in study (iv) and as with clonazepam also prolonged the QTc interval in study (iii). Combined administration of diazepam and adrenaline in study (vi) improved cardiac contractility but caused hypokalaemia. CONCLUSION AND IMPLICATIONS: Neither diazepam nor other ligands for benzodiazepine binding sites protect against or attenuate chloroquine cardiotoxicity. However, diazepam may augment the effects of positive inotropes in reducing chloroquine cardiotoxicity. LINKED ARTICLES: This article is part of a themed issue on The Pharmacology of COVID-19. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.21/issuetoc.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Cardiotoxicity/etiology , Chloroquine/poisoning , Diazepam/pharmacology , Animals , Arrhythmias, Cardiac/prevention & control , Benzodiazepinones/pharmacology , COVID-19 , Cardiotoxicity/prevention & control , Clonazepam/pharmacology , Coronavirus Infections/drug therapy , Diazepam/administration & dosage , Dose-Response Relationship, Drug , Drug Overdose , Electrocardiography , Female , Hypokalemia/chemically induced , Male , Pandemics , Pneumonia, Viral/drug therapy , Rabbits , Random Allocation , Rats , Rats, Wistar
12.
Pharmacol Rep ; 72(2): 322-330, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32048251

ABSTRACT

BACKGROUND: The goal of our study was to examine the long-term effect of vigabatrin (VGB), a γ-aminobutyric acid aminotransferase (GABA-AT) inhibitor on clonazepam (CLO), ethosuximide (ETX) and valproate (VPA) anticonvulsive activity against pentylenetetrazole (PTZ)-induced seizures in mice. METHODS: VGB was administered for 3 and 7 days. Convulsions were evoked by PTZ at its CD97 (99 mg/kg). The influence of CLO, ETX and VPA alone or in combination with VGB on motor performance and long-term memory was analyzed. γ-aminobutyric acid (GABA) concentration in mice brain and plasma as well as glutamate decarboxylase (GAD) activity was measured. RESULTS: After 3 days of treatment, VGB in doses up to 500 mg/kg increased PTZ-induced seizure threshold, whereas after 7 days VGB (at the dose of 125 mg/kg) inhibited clonic seizures in experimental mice. 7 days of VGB administration did not change the protective effect of CLO, ETX and VPA against PTZ-induced seizures. 7 days of VGB treatment at a subthreshold dose of 75 mg/kg decreased TD50 of ETX and CLO in the chimney test, but did not affect TD50 value for VPA. 7 days of VGB administration in combination with AEDs did not affect long-term memory in mice. VGB after 3 days or 7 days of administration increased brain GABA concentration. GAD activity was decreased after 3 and 7 days of VGB administration. CONCLUSIONS: The presented results confirm anticonvulsive activity of VGB through GABA metabolism alteration and suggest care when combining VGB with ETX or CLO in the therapy.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , Anticonvulsants/pharmacology , Brain/drug effects , Seizures/drug therapy , Vigabatrin/pharmacology , gamma-Aminobutyric Acid/metabolism , Animals , Brain/metabolism , Clonazepam/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Therapy, Combination , Ethosuximide/pharmacology , Mice , Pentylenetetrazole/pharmacology , Seizures/metabolism , Time Factors , Valproic Acid/pharmacology , Vigabatrin/administration & dosage
13.
Cereb Cortex ; 30(5): 3240-3258, 2020 05 14.
Article in English | MEDLINE | ID: mdl-31828304

ABSTRACT

Mammalian target of rapamycin (mTOR) signaling plays essential roles in brain development. Hyperactive mTOR is an essential pathological mechanism in autism spectrum disorder (ASD). Here, we show that tripartite motif protein 32 (TRIM32), as a maintainer of mTOR activity through promoting the proteasomal degradation of G protein signaling protein 10 (RGS10), regulates the proliferation of medial/lateral ganglionic eminence (M/LGE) progenitors. Deficiency of TRIM32 results in an impaired generation of GABAergic interneurons and autism-like behaviors in mice, concomitant with an elevated autophagy, which can be rescued by treatment embryonically with 3BDO, an mTOR activator. Transplantation of M/LGE progenitors or treatment postnatally with clonazepam, an agonist of the GABAA receptor, rescues the hyperexcitability and the autistic behaviors of TRIM32-/- mice, indicating a causal contribution of GABAergic disinhibition. Thus, the present study suggests a novel mechanism for ASD etiology in that TRIM32 deficiency-caused hypoactive mTOR, which is linked to an elevated autophagy, leads to autism-like behaviors via impairing generation of GABAergic interneurons. TRIM32-/- mouse is a novel autism model mouse.


Subject(s)
Autistic Disorder/genetics , Cell Proliferation/genetics , GABAergic Neurons/metabolism , Interneurons/metabolism , Neural Stem Cells/metabolism , Neurogenesis/genetics , TOR Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , Autistic Disorder/metabolism , Autophagy/drug effects , Autophagy/genetics , Behavior, Animal/drug effects , Behavior, Animal/physiology , Clonazepam/pharmacology , GABA-A Receptor Agonists/pharmacology , GABAergic Neurons/drug effects , Interneurons/drug effects , Mice , Mice, Knockout , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Proteasome Endopeptidase Complex/metabolism , RGS Proteins/metabolism
14.
Life Sci ; 239: 117033, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31697950

ABSTRACT

AIMS: Benzodiazepines (BZDs) produce various pharmacological actions by binding to and allosterically regulating GABAA receptors. Several in vitro studies have demonstrated diazepam, the prototypic BZD, produces a high-dose action that cannot be countered with the classical BZD-binding site antagonist flumazenil. Here, we investigate the existence and behavioral relevance of non-classical BZD binding sites in zebrafish larvae. MAIN METHODS: Zebrafish larvae were treated with a series of BZDs alone or combined with flumazenil, bicuculline (a non-selective GABAA receptor antagonist), or RO 15-4513 (a general BZD antagonist and a proposed antagonist interacting with α+/ß- interfaces in α4/6/ß3δ receptors), and their locomotor activities and behavioral phenotypes were recorded. KEY FINDINGS: Diazepam-induced hypolocomotion (sedation-like state) at low doses (10 and 20 mg L-1) was effectively antagonized by flumazenil or bicuculline, while diazepam-induced immobility (anesthesia-like state) at higher dose (30 mg L-1) was prevented by bicuculline (3 mg L-1) but not flumazenil, even at doses up to 150 mg L-1. Ro 15-4513 also failed to efficiently antagonize diazepam-induced immobility. Immobility induced by high dose of another 1,4-BZD, clonazepam, was also resistant to flumazenil. SIGNIFICANCE: These results provide direct in vivo evidence for non-classical BZD-binding sites, which may be located at the second transmembrane domain of GABAA receptors and contribute to BZD-induced anesthesia.


Subject(s)
Benzodiazepines/metabolism , Benzodiazepines/pharmacology , Flumazenil/pharmacology , GABA Modulators/pharmacology , Motor Activity/drug effects , Receptors, GABA-A/drug effects , Animals , Azides/pharmacology , Benzodiazepines/toxicity , Bicuculline/pharmacology , Binding Sites/drug effects , Clonazepam/pharmacology , Dose-Response Relationship, Drug , Female , Flumazenil/toxicity , GABA Antagonists/pharmacology , GABA Modulators/toxicity , Larva , Male , Zebrafish
15.
Cereb Cortex ; 29(8): 3666-3682, 2019 07 22.
Article in English | MEDLINE | ID: mdl-31237323

ABSTRACT

FOXG1 syndrome is a severe encephalopathy that exhibit intellectual disability, emotional disorder, and limited social communication. To elucidate the contribution of somatostatin-expressing interneurons (SST-INs) to the cellular basis underlying FOXG1 syndrome, here, by crossing SST-cre with a Foxg1fl/fl line, we selectively ablated Foxg1. Loss of Foxg1 resulted in an obvious reduction in the number of SST-INs, accompanied by an altered ratio of subtypes. Foxg1-deficient SST-INs exhibited decreased membrane excitability and a changed ratio of electrophysiological firing patterns, which subsequently led to an excitatory/inhibitory imbalance. Moreover, cognitive defects, limited social interactions, and depression-like behaviors were detected in Foxg1 cKO mice. Treatment with low-dose of clonazepam effectively alleviated the defects. These results identify a link of SST-IN development to the aberrant emotion, cognition, and social capacities in patients. Our findings identify a novel role of Foxg1 in SST-IN development and put new insights into the cellular basis of FOXG1 syndrome.


Subject(s)
Behavior, Animal/physiology , Cognition/physiology , Depression/genetics , Emotions/physiology , Forkhead Transcription Factors/genetics , Interneurons/metabolism , Nerve Tissue Proteins/genetics , Social Behavior , Animals , Behavior, Animal/drug effects , Brain Diseases/genetics , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Clonazepam/pharmacology , Cognition/drug effects , Emotions/drug effects , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , GABA Modulators/pharmacology , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Intellectual Disability/genetics , Interneurons/drug effects , Mice , Mice, Knockout , Patch-Clamp Techniques , Social Communication Disorder/genetics , Somatostatin/metabolism , Syndrome
17.
J Neurol Sci ; 401: 81-86, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-31035190

ABSTRACT

BACKGROUND: Clonazepam is considered to be a first-line treatment for rapid eye movement sleep-related behavior disorder (RBD) in Parkinson's disease (PD). The purpose of this study was to determine the short-term efficacy and safety of clonazepam for the treatment of probable RBD (pRBD) in patients with PD. METHODS: We conducted a four-week, randomized, double-blind, placebo-controlled trial of clonazepam (0.5 mg/day at bedtime) compared to a placebo for RBD symptoms in patients with PD. Patients aged 30 years or older who had a caregiver that could observe RBD symptoms were recruited between April 2015 and February 2016. The primary outcome was the Clinical Global Impressions-Improvement (CGII) score at week four, and we compared scores between the clonazepam and placebo groups. RESULTS: A total of 40 patients were enrolled, with 20 assigned to receive clonazepam and 20 to receive the placebo. The CGI-I score at four weeks indicated an improvement in RBD symptoms in both the clonazepam (median score [minimum, maximum] = 2 [1,5]) and placebo (3 [1,6]) groups, with no significant difference between the groups (p = .253). The secondary outcomes were not significantly different between the clonazepam and placebo groups. CONCLUSIONS: Both clonazepam and placebo tended toward improvement on pRBD symptoms in patients with PD. No firm conclusion on efficacy of clonazepam was drawn due to limitations in the study design. This study emphasized the importance of conducting future large-scale, randomized trials with better assessment tools and polysomnography to provide evidence for the benefit of clonazepam.


Subject(s)
Clonazepam/therapeutic use , Parkinson Disease/drug therapy , Parkinson Disease/epidemiology , REM Sleep Behavior Disorder/drug therapy , REM Sleep Behavior Disorder/epidemiology , Aged , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Clonazepam/pharmacology , Double-Blind Method , Female , Follow-Up Studies , Humans , Male , Middle Aged , Parkinson Disease/physiopathology , Polysomnography/drug effects , Polysomnography/methods , REM Sleep Behavior Disorder/physiopathology , Republic of Korea/epidemiology
18.
J Mol Neurosci ; 65(1): 110-117, 2018 May.
Article in English | MEDLINE | ID: mdl-29737465

ABSTRACT

Anxiety disorders are highly prevalent and considered a major public health concern worldwide. Current anxiolytics are of limited efficacy and associated with various side effects. Our novel herbal treatment (NHT), composed of four constituents, was shown to reduce anxiety-like behavior while precluding a common side effect caused by current anxiolytics, i.e., sexual dysfunction. Nevertheless, NHT's mechanism of action is yet to be determined. There is evidence that some medicinal herbs interact with the GABAergic system. Therefore, we aimed to examine whether NHT's anxiolytic-like effect is exerted by alterations in GABAA receptor density in the hippocampus, prefrontal cortex, and hypothalamus. The effects of 3-weeks treatment with NHT on anxiety-like behavior and locomotion were assessed using the elevated plus maze (EPM) and the open field test (OFT), respectively. Regional GABAA receptor levels were analyzed using [3H] RO15-1788 high-affinity binding assays. In stressed mice, NHT reduced anxiety-like behavior similarly to the benzodiazepine, clonazepam, while locomotion remained intact. Lack of changes or minor changes in regional GABAA receptor density in the brain were induced by NHT or clonazepam. In naive mice, performance in the EPM, locomotion and GABAA receptor densities were not altered by treatment with NHT or clonazepam. These findings support NHT as an efficacious and safe anxiolytic, although the GABAergic involvement remains to be further elucidated.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Brain/metabolism , Plant Extracts/pharmacology , Receptors, GABA-A/metabolism , Animals , Anti-Anxiety Agents/therapeutic use , Anxiety/metabolism , Brain/drug effects , Clonazepam/pharmacology , Clonazepam/therapeutic use , Male , Maze Learning , Mice , Mice, Inbred C57BL , Plant Extracts/therapeutic use , Receptors, GABA-A/genetics
19.
Cell Calcium ; 72: 1-17, 2018 06.
Article in English | MEDLINE | ID: mdl-29748128

ABSTRACT

Interstitial cells of Cajal (ICC-MY) are pacemakers that generate and propagate electrical slow waves in gastrointestinal (GI) muscles. Slow waves appear to be generated by the release of Ca2+ from intracellular stores and activation of Ca2+-activated Cl- channels (Ano1). Conduction of slow waves to smooth muscle cells coordinates rhythmic contractions. Mitochondrial Ca2+ handling is currently thought to be critical for ICC pacemaking. Protonophores, inhibitors of the electron transport chain (FCCP, CCCP or antimycin) or mitochondrial Na+/Ca2+ exchange blockers inhibited slow waves in several GI muscles. Here we utilized Ca2+ imaging of ICC in small intestinal muscles in situ to determine the effects of mitochondrial drugs on Ca2+ transients in ICC. Muscles were obtained from mice expressing a genetically encoded Ca2+ indicator (GCaMP3) in ICC. FCCP, CCCP, antimycin, a uniporter blocker, Ru360, and a mitochondrial Na+/Ca2+ exchange inhibitor, CGP-37157 inhibited Ca2+ transients in ICC-MY. Effects were not due to depletion of ATP, as oligomycin did not affect Ca2+ transients. Patch-clamp experiments were performed to test the effects of the mitochondrial drugs on key pacemaker conductances, Ano1 and T-type Ca2+ (CaV3.2), in HEK293 cells. Antimycin blocked Ano1 and reduced CaV3.2 currents. CCCP blocked CaV3.2 current but did not affect Ano1 current. Ano1 and Cav3.2 currents were inhibited by CGP-37157. Inhibitory effects of mitochondrial drugs on slow waves and Ca2+ signalling in ICC can be explained by direct antagonism of key pacemaker conductances in ICC that generate and propagate slow waves. A direct obligatory role for mitochondria in pacemaker activity is therefore questionable.


Subject(s)
Biological Clocks , Calcium Signaling , Electric Conductivity , Interstitial Cells of Cajal/metabolism , Intestine, Small/cytology , Mitochondria/metabolism , Adenosine Triphosphate/metabolism , Animals , Anoctamin-1/metabolism , Antimycin A/analogs & derivatives , Antimycin A/pharmacology , Calcium Channels, T-Type/metabolism , Calcium Signaling/drug effects , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Clonazepam/analogs & derivatives , Clonazepam/pharmacology , HEK293 Cells , Humans , Interstitial Cells of Cajal/cytology , Ion Channel Gating/drug effects , Mice, Inbred C57BL , Mitochondria/drug effects , Muscles/cytology , Ruthenium Compounds/pharmacology , Thiazepines/pharmacology
20.
Biol Psychiatry ; 83(6): 518-529, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29150182

ABSTRACT

BACKGROUND: Platelet-activating factor acetylhydrolase 1B1 (LIS1), a critical mediator of neuronal migration in developing brain, is expressed throughout life. However, relatively little is known about LIS1 function in the mature brain. We previously demonstrated that LIS1 involvement in the formation and turnover of synaptic protrusions and synapses of young brain after neuronal migration is complete. Here we examine the requirement for LIS1 to maintain hippocampal circuit function in adulthood. METHODS: Effects of conditional Lis1 inactivation in excitatory pyramidal neurons, starting in juvenile mouse brain, were probed using high-resolution approaches combining mouse genetics, designer receptor exclusively activated by designer drug technology to specifically manipulate CA1 pyramidal neuron excitatory activity, electrophysiology, hippocampus-selective behavioral testing, and magnetic resonance imaging tractography to examine the connectivity of LIS1-deficient neurons. RESULTS: We found progressive excitatory and inhibitory postsynaptic dysfunction as soon as 10 days after conditional inactivation of Lis1 targeting CA1 pyramidal neurons. Surprisingly, by postnatal day 60 it also caused CA1 histological disorganization, with a selective decline in parvalbumin-expressing interneurons and further reduction in inhibitory neurotransmission. Accompanying these changes were behavioral and cognitive deficits that could be rescued by either designer receptor exclusively activated by designer drug-directed specific increases in CA1 excitatory transmission or pharmacological enhancement of gamma-aminobutyric acid transmission. Lagging behind electrophysiological changes was a progressive, selective decline in neural connectivity, affecting hippocampal efferent pathways documented by magnetic resonance imaging tractography. CONCLUSIONS: LIS1 supports synaptic function and plasticity of mature CA1 neurons. Postjuvenile loss of LIS1 disrupts the structure and cellular composition of the hippocampus, its connectivity with other brain regions, and cognition dependent on hippocampal circuits.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Cognition/physiology , Hippocampus/cytology , Microtubule-Associated Proteins/metabolism , Neurons/physiology , Synapses/physiology , 1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Animals , Animals, Newborn , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Movement/genetics , Clonazepam/pharmacology , Cognition/drug effects , Conditioning, Psychological/physiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Fear/physiology , GABA Modulators/pharmacology , Hippocampus/diagnostic imaging , Locomotion/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Neurons/drug effects , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Recognition, Psychology/physiology , Synapses/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...