Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Adv Ther ; 41(4): 1351-1371, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38443647

ABSTRACT

Stiripentol (Diacomit®) (STP) is an orally active antiseizure medication (ASM) indicated as adjunctive therapy, for the treatment of seizures associated with Dravet syndrome (DS), a severe form of childhood epilepsy, in conjunction with clobazam and, in some regions valproic acid. Since the discovery of STP, several mechanisms of action (MoA) have been described that may explain its specific effect on seizures associated with DS. STP is mainly considered as a potentiator of gamma-aminobutyric acid (GABA) neurotransmission: (i) via uptake blockade, (ii) inhibition of degradation, but also (iii) as a positive allosteric modulator of GABAA receptors, especially those containing α3 and δ subunits. Blockade of voltage-gated sodium and T-type calcium channels, which is classically associated with anticonvulsant and neuroprotective properties, has also been demonstrated for STP. Finally, several studies indicate that STP could regulate glucose energy metabolism and inhibit lactate dehydrogenase. STP is also an inhibitor of several cytochrome P450 enzymes involved in the metabolism of other ASMs, contributing to boost their anticonvulsant efficacy as add-on therapy. These different MoAs involved in treatment of DS and recent data suggest a potential for STP to treat other neurological or non-neurological diseases.


Subject(s)
Dioxolanes , Epilepsies, Myoclonic , Humans , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Dioxolanes/pharmacology , Dioxolanes/therapeutic use , Seizures/drug therapy , Epilepsies, Myoclonic/drug therapy , gamma-Aminobutyric Acid
2.
Epilepsia ; 63(5): 1200-1210, 2022 05.
Article in English | MEDLINE | ID: mdl-35184274

ABSTRACT

OBJECTIVE: Stiripentol (STP; Diacomit®) is an antiepileptic drug indicated for Dravet syndrome that has been identified as a γ-aminobutyric acid (GABAergic) positive allosteric modulator. Dravet syndrome is characterized by multiple seizure types: generalized tonic-clonic, focal, myoclonic, and absence seizures. In addition to its antiepileptic effects on tonic-clonic seizures, STP has also been reported to reduce the frequency of atypical absence seizures in patients. Our study focused on STP potential effects on absence seizures, to better characterize its full spectrum of mechanisms of action. METHODS: STP effects on absence seizures were quantified by electroencephalographic recording in two animal models: rats treated with a low dose of pentylenetetrazol (20 mg/kg ip) and rats from the WAG/Rij strain. In addition, we characterized STP effects on T-type calcium channel activity. Peak currents were recorded with manual patch clamp on cells transfected with cDNA encoding for the human isoform for Cav 3.1, Cav 3.2, and Cav 3.3. RESULTS: STP administered before pentylenetetrazol almost completely abolished the generation of spike-and-wave discharges (SWDs) at the dose of 300 mg/kg. At this dose, STP also statistically significantly decreased SWD cumulated duration and number in WAG/Rij rats. Its antiepileptic effect was maintained in WAG/Rij rats, whose seizures were aggravated by the GABA agonist THIP (gaboxadol hydrochloride). Furthermore, electrophysiological recordings showed that STP inhibits T-type calcium channel peak activity, with a higher specificity for the Cav 3.3 subtype. SIGNIFICANCE: In addition to its previously characterized anticonvulsive properties, these data highlight a new mechanism of action of STP on abnormal thalamocortical activity. This strong antiabsence effect on seizures is correlated with an inhibition of T-type calcium channels. This new mechanism of action could be implicated in the specificity of STP therapeutic effects in Dravet syndrome.


Subject(s)
Calcium Channels, T-Type , Epilepsies, Myoclonic , Epilepsy, Absence , Animals , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Dioxolanes , Disease Models, Animal , Electroencephalography , Epilepsies, Myoclonic/drug therapy , Epilepsy, Absence/drug therapy , Epilepsy, Absence/genetics , Humans , Pentylenetetrazole/toxicity , Rats , Rats, Wistar , Seizures/chemically induced , Seizures/drug therapy
3.
CNS Neurol Disord Drug Targets ; 19(3): 227-240, 2020.
Article in English | MEDLINE | ID: mdl-32479250

ABSTRACT

AIM: The aim of this study is to examine the effect of etifoxine on ß-amyloid-induced toxicity models. BACKGROUND: Etifoxine is an anxiolytic compound with a dual mechanism of action; it is a positive allosteric modulator of GABAergic receptors as well as a ligand for the 18 kDa mitochondrial Translocator Protein (TSPO). TSPO has recently raised interest in Alzheimer's Disease (AD), and experimental studies have shown that some TSPO ligands could induce neuroprotective effects in animal models. OBJECTIVE: In this study, we examined the potential protective effect of etifoxine in an in vitro and an in vivo model of amyloid beta (Aß)-induced toxicity in its oligomeric form, which is a crucial factor in AD pathologic mechanisms. METHODS: Neuronal cultures were intoxicated with Aß1-42, and the effects of etifoxine on oxidative stress, Tau-hyperphosphorylation and synaptic loss were quantified. In a mice model, behavioral deficits induced by intracerebroventricular administration of Aß25-35 were measured in a spatial memory test, the spontaneous alternation and in a contextual memory test, the passive avoidance test. RESULTS: In neuronal cultures intoxicated with Aß1-42, etifoxine dose-dependently decreased oxidative stress (methionine sulfoxide positive neurons), tau-hyperphosphorylation and synaptic loss (ratio PSD95/synaptophysin). In a mice model, memory impairments were fully alleviated by etifoxine administered at anxiolytic doses (12.5-50mg/kg). In addition, markers of oxidative stress and apoptosis were decreased in the hippocampus of these animals. CONCLUSION: Our results have shown that in these two models, etifoxine could fully prevent neurotoxicity and pathological changes induced by Aß. These results confirm that TSPO ligands could offer an interesting therapeutic approach to Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/metabolism , Anti-Anxiety Agents/therapeutic use , Oxazines/therapeutic use , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/toxicity , Animals , Apoptosis/drug effects , Disease Models, Animal , Hippocampus/drug effects , Male , Mice , Neurons/metabolism , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects
4.
Pharmacol Res ; 145: 104250, 2019 07.
Article in English | MEDLINE | ID: mdl-31059790

ABSTRACT

Etifoxine (EFX) is a non-benzodiazepine psychoactive drug which exhibits anxiolytic effects through a dual mechanism, by directly binding to GABAA receptors (GABAARs) and to the mitochondrial 18-kDa translocator protein, resulting in the potentiation of the GABAergic function. The ß subunit subtype plays a key role in the EFX-GABAAR interaction, however this does not explain the anxiolytic effects of this drug. Here, we combined behavioral and electrophysiological experiments to challenge the role of the GABAAR α subunit in the EFX mode of action. After single administrations of anxiolytic doses (25-50 mg/kg, intraperitoneal), EFX did not induce any neurological nor locomotor impairments, unlike the benzodiazepine bromazepam (0.5-1 mg/kg, intraperitoneal). We established the EFX pharmacological profile on heteropentameric GABAARs constructed with α1 to α6 subunit expressed in Xenopus oocyte. Unlike what is known for benzodiazepines, neither the γ nor δ subunits influenced EFX-mediated potentiation of GABA-evoked currents. EFX acted first as a partial agonist on α2ß3γ2S, α3ß3γ2S, α6ß3γ2S and α6ß3δ GABAARs, but not on α1ß3γ2S, α4ß3γ2S, α4ß3δ nor α5ß3γ2S GABAARs. Moreover, EFX exhibited much higher positive allosteric modulation towards α2ß3γ2S, α3ß3γ2S and α6ß3γ2S than for α1ß3γ2S, α4ß3γ2S and α5ß3γ2S GABAARs. At 20 µM, corresponding to brain concentration at anxiolytic doses, EFX increased GABA potency to the highest extent for α3ß3γ2S GABAARs. We built a docking model of EFX on α3ß3γ2S GABAARs, which is consistent with a binding site located between α and ß subunits in the extracellular domain. In conclusion, EFX preferentially potentiates α2ß3γ2S and α3ß3γ2S GABAARs, which might support its advantageous anxiolytic/sedative balance.


Subject(s)
Anti-Anxiety Agents/pharmacology , Oxazines/pharmacology , Protein Subunits/metabolism , Receptors, GABA-A/metabolism , Animals , Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Anxiety/metabolism , Anxiety/physiopathology , Female , Locomotion/drug effects , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Molecular , Oocytes/physiology , Oxazines/therapeutic use , Protein Subunits/genetics , Psychomotor Performance/drug effects , Receptors, GABA-A/genetics , Xenopus laevis
5.
Eur J Pharmacol ; 843: 316-322, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30552900

ABSTRACT

Inflammatory processes are critical promoting factors of chronic pain states, mostly by inducing peripheral and central sensitization of the nociceptive system. These processes are associated with a massive increase in glutamatergic transmission, sometimes facilitated by spinal disinhibition. In this study, we used etifoxine, a non-benzodiazepine anxiolytic known to amplify inhibition mediated by gamma-aminobutyric acid type A (GABAA) receptors in pain processing regions, either directly (through allosteric modulation) or indirectly (through the synthesis of endogenous neurosteroids). We used different models of local inflammation to evaluate the possible direct action of etifoxine on analgesia and edema. Pain symptom and edema measurements were performed after intraplantar carrageenan injection or after topical ear inflammation. We found that etifoxine treatment was associated with reduced plantar surface temperature 24 h after intraplantar carrageenan injection. In this model, etifoxine also alleviated thermal hot and mechanical hyperalgesia. A similar finding was observed while analyzing pain symptoms in the late phase of the formalin test. In a model of ear inflammation, etifoxine appeared to have a moderate anti-edemic effect after topical application. This slight action of etifoxine on the limitation of inflammatory processes could be mediated in part by cyclo-oxygenase 1 activity inhibition. Etifoxine appears as a promising therapeutic tool contributing to the limitation of inflammatory pain symptoms. Since etifoxine is already prescribed as an anxiolytic in several countries, it could be a good candidate for the prevention of inflammatory-driven edema and hyperalgesia, although the precise mechanism of action relative to its anti-inflammatory potential remains to be elucidated.


Subject(s)
Analgesics/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Edema/drug therapy , Hyperalgesia/drug therapy , Oxazines/therapeutic use , Pain/drug therapy , Animals , Carrageenan , Disease Models, Animal , Edema/chemically induced , Formaldehyde , Hyperalgesia/chemically induced , Male , Mice , Pain/chemically induced , Rats, Sprague-Dawley , Tetradecanoylphorbol Acetate
6.
J Neuroinflammation ; 13(1): 203, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27565146

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) results in important neurological impairments which occur through a cascade of deleterious physiological events over time. There are currently no effective treatments to prevent these consequences. TBI is followed not only by an inflammatory response but also by a profound reorganization of the GABAergic system and a dysregulation of translocator protein 18 kDa (TSPO). Etifoxine is an anxiolytic compound that belongs to the benzoxazine family. It potentiates GABAergic neurotransmission, either through a positive allosteric effect or indirectly, involving the activation of TSPO that leads to an increase in neurosteroids synthesis. In several models of peripheral nerve injury, etifoxine has been demonstrated to display potent regenerative and anti-inflammatory properties and to promote functional recovery. Prior study also showed etifoxine efficacy in reducing brain edema in rats. In light of these positive results, we used a rat model of TBI to explore etifoxine treatment effects in a central nervous system injury, from functional outcomes to the underlying mechanisms. METHODS: Male Sprague-Dawley rats received contusion (n = 18) or sham (n = 19) injuries centered laterally to bregma over the left sensorimotor cortex. They were treated with etifoxine (50 mg/kg, i.p.) or its vehicle 30 min following injury and every day during 7 days. Rats underwent behavioral testing to assess sensorimotor function. In another experiment, injured rats (n = 10) or sham rats (n = 10) received etifoxine (EFX) (50 mg/kg, i.p.) or its vehicle 30 min post-surgery. Brains were then dissected for analysis of neuroinflammation markers, glial activation, and neuronal degeneration. RESULTS: Brain-injured rats exhibited significant sensorimotor function deficits compared to sham-injured rats in the bilateral tactile adhesive removal test, the beam walking test, and the limb-use asymmetry test. After 2 days of etifoxine treatment, behavioral impairments were significantly reduced. Etifoxine treatment reduced pro-inflammatory cytokines levels without affecting anti-inflammatory cytokines levels in injured rats, reduced macrophages and glial activation, and reduced neuronal degeneration. CONCLUSIONS: Our results showed that post-injury treatment with etifoxine improved functional recovery and reduced neuroinflammation in a rat model of TBI. These findings suggest that etifoxine may have a therapeutic potential in the treatment of TBI.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Encephalitis/drug therapy , Gait Ataxia/drug therapy , Nerve Degeneration/drug therapy , Neuroglia/drug effects , Oxazines/therapeutic use , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Brain Injuries, Traumatic/complications , Cytokines/metabolism , Disease Models, Animal , Encephalitis/etiology , Functional Laterality/drug effects , Gait Ataxia/etiology , Glial Fibrillary Acidic Protein/metabolism , Locomotion/drug effects , Macrophages/drug effects , Male , Nerve Degeneration/etiology , Psychomotor Performance/drug effects , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects
7.
Clin Exp Pharmacol Physiol ; 43(1): 3-12, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26475417

ABSTRACT

Nefopam is a non-opioid, non-steroidal, centrally acting analgesic drug used to prevent postoperative pain, primarily in the context of multimodal analgesia. This paper reviews preclinical and clinical studies in which nefopam has been combined with opioids, non-steroidal anti-inflammatory compounds, and paracetamol. This report focuses on the literature during the last decade and discusses the translational efforts between animal and clinical studies in the context of multimodal or balanced analgesia. In preclinical rodent models of acute and inflammatory pain, nefopam combinations including opioids revealed a synergistic interaction or enhanced morphine analgesia in six out of seven studies. Nefopam combinations including non-steroidal anti-inflammatory drugs (NSAIDs) (aspirin, ketoprofen or nimesulide) or paracetamol likewise showed enhanced analgesic effects for the associated compound in all instances. Clinical studies have been performed in various types of surgeries involving different pain intensities. Nefopam combinations including opioids resulted in a reduction in morphine consumption in 8 out of 10 studies of severe or moderate pain. Nefopam combinations including NSAIDs (ketoprofen or tenoxicam) or paracetamol also demonstrated a synergic interaction or an enhancement of the analgesic effect of the associated compound. In conclusion, this review of nefopam combinations including various analgesic drugs (opioids, NSAIDs and paracetamol) reveals that enhanced analgesia was demonstrated in most preclinical and clinical studies, suggesting a role for nefopam in multimodal analgesia based on its distinct characteristics as an analgesic. Further clinical studies are needed to evaluate the analgesic effects of nefopam combinations including NSAIDs or paracetamol.


Subject(s)
Analgesia/methods , Analgesics, Non-Narcotic/pharmacology , Nefopam/pharmacology , Analgesics, Opioid/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Drug Interactions , Humans
8.
J Neurosci Res ; 94(2): 179-89, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26511438

ABSTRACT

A growing body of data has shown that recurrent epileptic seizures may be caused by an excessive release of the excitatory neurotransmitter glutamate in the brain. Glutamatergic overstimulation results in massive neuronal influxes of calcium and sodium through N-methyl-D-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, and kainic acid glutamate subtype receptors and also through voltage-gated calcium and sodium channels. These persistent and abnormal sodium and calcium entry points have deleterious consequences (neurotoxicity) for neuronal function. The therapeutic value of an antiepileptic drug would include not only control of seizure activity but also protection of neuronal tissue. The present study examines the in vitro neuroprotective effects of stiripentol, an antiepileptic compound with γ-aminobutyric acidergic properties, on neuronal-astroglial cultures from rat cerebral cortex exposed to oxygen-glucose deprivation (OGD) or to glutamate (40 µM for 20 min), two in vitro models of brain injury. In addition, the affinity of stiripentol for the different glutamate receptor subtypes and the interaction with the cell influx of Na(+) and of Ca(2+) enhanced by veratridine and NMDA, respectively, are assessed. Stiripentol (10-100 µM) included in the culture medium during OGD or with glutamate significantly increased the number of surviving neurons relative to controls. Stiripentol displayed no binding affinity for different subtypes of glutamate receptors (IC50 >100 µM) but significantly blocked the entry of Na(+) and Ca(2+) activated by veratridine and NMDA, respectively. These results suggest that Na(+) and Ca(2+) channels could contribute to the neuroprotective properties of sitiripentol.


Subject(s)
Calcium/metabolism , Dioxolanes/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Sodium/metabolism , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian , Fibrinolytic Agents/pharmacokinetics , Glucose/deficiency , Glutamic Acid/pharmacology , Hippocampus/cytology , Hirudins/pharmacokinetics , Mice, Inbred C57BL , Neurofilament Proteins/metabolism , Neuroglia/drug effects , Protein Binding/drug effects , Rats , Receptors, Glutamate/metabolism , Recombinant Proteins/pharmacokinetics , Tritium/pharmacokinetics , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacokinetics
9.
Pharmacology ; 94(5-6): 239-44, 2014.
Article in English | MEDLINE | ID: mdl-25427779

ABSTRACT

BACKGROUND/AIMS: Hypercholesterolemia is a major risk factor for coronary artery disease and probiotics have been suggested as tools to manage elevated cholesterol levels. METHODS: The present study investigated the ability of the biotherapeutic agent Saccharomyces boulardii (Sb-Biocodex) to reduce the hypercholesterolemia induced by a 0.1% cholesterol-enriched diet in the hamster. RESULTS: In a first experiment, chronic oral treatment with S. boulardii at 12 × 10(10) CFU/kg (3 g/kg) twice a day was started from the beginning of the cholesterol diet and continued for 14 days ('preventive protocol'). In the second experiment, S. boulardii was given 14 days after the beginning of the cholesterol diet when hypercholesterolemia had developed and continued for an additional 14 days ('curative protocol'). In the preventive protocol, administration of the yeast significantly reduced hypercholesterolemia (14%) induced by the cholesterol-enriched diet compared to the group receiving only the cholesterol diet. In the curative protocol, S. boulardii significantly reduced hypercholesterolemia (12%) induced by the cholesterol-enriched diet, too. Moreover, the yeast significantly decreased the serum triglyceride increase by 39%. CONCLUSION: S. boulardii possesses anti-hypercholesterolemic properties in the hamster worthy of further evaluation in clinical studies.


Subject(s)
Hypercholesterolemia/therapy , Probiotics/therapeutic use , Saccharomyces , Animals , Cholesterol/blood , Cholesterol/metabolism , Cricetinae , Hypercholesterolemia/blood , Liver/metabolism , Male , Probiotics/pharmacology , Triglycerides/blood
10.
Eur J Pharmacol ; 667(1-3): 100-4, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21664351

ABSTRACT

Citrulline malate (CM; CAS 54940-97-5, Stimol®) is known to limit the deleterious effect of asthenic state on muscle function, but its effect under healthy condition remains poorly documented. The aim of this longitudinal double-blind study was to investigate the effect of oral ingestion of CM on muscle mechanical performance and bioenergetics in normal rat. Gastrocnemius muscle function was investigated strictly non-invasively using nuclear magnetic resonance techniques. A standardized rest-stimulation- (5.7 min of repeated isometric contractions electrically induced by transcutaneous stimulation at a frequency of 3.3 Hz) recovery-protocol was performed twice, i.e., before (t(0)-24 h) and after (t(0)+48 h) CM (3 g/kg/day) or vehicle treatment. CM supplementation did not affect PCr/ATP ratio, [PCr], [Pi], [ATP] and intracellular pH at rest. During the stimulation period, it lead to a 23% enhancement of specific force production that was associated to significant decrease in both PCr (28%) and oxidative (32%) costs of contraction, but had no effect on the time-courses of phosphorylated compounds and intracellular pH. Furthermore, both the rate of PCr resynthesis during the post-stimulation period (VPCr(rec)) and the oxidative ATP synthesis capacity (Q(max)) remained unaffected by CM treatment. These data demonstrate that CM supplementation under healthy condition has an ergogenic effect associated to an improvement of muscular contraction efficiency.


Subject(s)
Citrulline/analogs & derivatives , Malates/administration & dosage , Malates/pharmacology , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Administration, Oral , Animals , Biomechanical Phenomena/drug effects , Citrulline/administration & dosage , Citrulline/pharmacology , Electric Stimulation , Energy Metabolism/drug effects , Male , Muscle, Skeletal/metabolism , Oxidation-Reduction/drug effects , Rats , Rats, Wistar
11.
Clin Exp Pharmacol Physiol ; 38(3): 170-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21226753

ABSTRACT

1. The aim of the present study was to explore the concept of multimodal anaesthesia using a combination of two non-opioid analgesics, namely nefopam, a centrally acting non-opioid that inhibits monoamine reuptake, and paracetamol, an inhibitor of central cyclo-oxygenases. The antinociceptive characteristics of the combination were evaluated using four different animal models of pain. 2. In the mouse writhing test, antinociceptive properties were observed with ED50 values of 1.5 ± 0.2 and 120.9 ± 14.8 mg/kg for nefopam and paracetamol, respectively. In the mouse formalin test, both compounds significantly inhibited the licking time of the injected hind paw, with ED50 values in the early phase of 4.5 ± 1.1 and 330.7 ± 80.3 mg/kg for nefopam and paracetamol, respectively, compared with 4.3 ± 0.2 and 206.1 ± 45.1 mg/kg for nefopam and paracetamol, respectively, in the inflammatory phase. Isobolographic analysis revealed that this drug combination was synergistic in the writhing test and additive in the formalin test. 3. In a rat incision model of postoperative thermal hyperalgesia, coadministration of nefopam at a non-analgesic dose (3 mg/kg) with paracetamol at a low analgesic dose (300 mg/kg) showed the appearance of a strong antihyperalgesic effect, maintained for at least 3 h. In rat carrageenan-induced tactile allodynia, the combination of low analgesic doses of nefopam (10 or 30 mg/kg) with a non-analgesic dose of paracetamol (30 mg/kg), significantly blocked allodynia with a longer duration of efficacy. 4. In conclusion, coadministration of nefopam with paracetamol is worthy of clinical evaluation.


Subject(s)
Acetaminophen/pharmacology , Analgesics/pharmacology , Nefopam/pharmacology , Pain/drug therapy , Analgesics, Non-Narcotic/pharmacology , Analgesics, Opioid/pharmacology , Animals , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination/methods , Hyperalgesia/drug therapy , Male , Mice , Pain Measurement/methods , Rats , Rats, Sprague-Dawley
12.
Eur Neuropsychopharmacol ; 21(6): 457-70, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20943351

ABSTRACT

Dysfunction of GABAergic transmission related to abnormal expression of GABA(A) receptor subunits in specific brain regions underlies some pathological anxiety states. Besides involvement of the benzodiazepine recognition site of GABA(A) receptor in the expression of anxiety-like behaviour, the roles of the ß(2)/ß(3) subunits are not well characterized. To address this issue, the experimental design of this study utilized the GABAergic compound etifoxine (with a preferential effectiveness after binding to a specific site at ß(2)/ß(3) subunits) tested in two inbred mouse strains: BALB/cByJ and C57BL/6J mice using three behavioural paradigms (light/dark box, elevated plus maze and restraint stress-induced small intestinal transit inhibition) and the t-butylbicyclophosphorothionate-induced convulsions model. Etifoxine plasma and brain levels and ß(2)/ß(3) mRNAs and protein expression levels in various brain regions were compared between the two strains. The two mouse strains differed markedly in basal anxiety level. Etifoxine exhibited more pronounced anxiolytic and anticonvulsant effects in the BALB/cByJ mice compared to the C57BL/6J mice. The etifoxine brain/plasma ratios of the two strains were not different. Beta2 subunit mRNA and protein expression levels were around 25 and 10% higher respectively in the anterodorsal nucleus of the thalamus and the CA3 field of hippocampus of BALB/cByJ mice compared to C57BL/6J mice. Beta3 subunit mRNA and protein expression levels did not differ between the two strains. Based on these results, it is suggested that overexpression of GABA(A) receptor ß(2) subunit in BALB/cByJ mice relative to C57BL/6j mice contributes to the dysfunction in GABA(A) transmission in regions of brain known to regulate responses to stress. The dysregulated GABA(A) function in BALB/cByJ mice may be corrected by the administration of etifoxine.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anxiety Disorders/drug therapy , Anxiety/drug therapy , Brain/physiopathology , Hippocampus/physiopathology , Oxazines/pharmacology , Receptors, GABA-A/metabolism , Seizures/drug therapy , Animals , Anti-Anxiety Agents/blood , Anti-Anxiety Agents/toxicity , Behavior, Animal/drug effects , Brain/physiology , Hippocampus/physiology , Intestine, Small/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Motor Activity/drug effects , Oxazines/blood , Oxazines/toxicity , Receptors, GABA-A/genetics , Seizures/physiopathology
13.
Alcohol ; 43(3): 197-206, 2009 May.
Article in English | MEDLINE | ID: mdl-19393860

ABSTRACT

Change in the function of gamma-aminobutyric acid(A) (GABA(A)) receptors attributable to alterations in receptor subunit composition is one of main molecular mechanisms with those affecting the glutamatergic system which accompany prolonged alcohol (ethanol) intake. These changes explain in part the central nervous system hyperexcitability consequently to ethanol administration cessation. Hyperexcitability associated with ethanol withdrawal is expressed by physical signs, such as tremors, convulsions, and heightened anxiety in animal models as well as in humans. The present work investigated the effects of anxiolytic compound etifoxine on ethanol-withdrawal paradigms in a mouse model. The benzodiazepine diazepam was chosen as reference compound. Ethanol was given to NMRI mice by a liquid diet at 3% for 8 days, then at 4% for 7 days. Under these conditions, ethanol blood level ranged between 0.5 and 2 g/L for a daily ethanol intake varying from 24 to 30 g/kg. These parameters permitted the emergence of ethanol-withdrawal symptoms once ethanol administration was terminated. Etifoxine (12.5-25 mg/kg) and diazepam (1-4 mg/kg) injected intraperitoneally 3h 30 min after ethanol removal, decreased the severity in handling-induced tremors and convulsions in the period of 4-6h after withdrawal from chronic ethanol treatment. In addition when administered at 30 and 15 min, respectively, before the light and dark box test, etifoxine (50mg/kg) and diazepam (1mg/kg) inhibited enhanced aversive response 8h after ethanol withdrawal. Etifoxine at 25 and 50 mg/kg doses was without effects on spontaneous locomotor activity and did not exhibit ataxic effects on the rota rod in animals not treated with ethanol. These findings demonstrate that the GABAergic compound etifoxine selectively reduces the physical signs and anxiety-like behavior associated with ethanol withdrawal in a mouse model and may hold promise in the treatment of ethanol-withdrawal syndrome in humans.


Subject(s)
Ethanol/adverse effects , Oxazines/therapeutic use , Seizures/prevention & control , Animals , Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Diazepam , Ethanol/blood , GABA Agonists/therapeutic use , Male , Mice , Models, Animal , Motor Activity/drug effects , Receptors, GABA-A/drug effects , Substance Withdrawal Syndrome/drug therapy
14.
Pharmacology ; 83(2): 116-21, 2009.
Article in English | MEDLINE | ID: mdl-19096234

ABSTRACT

In order to further elucidate the mechanism(s) of action of analgesic and antihyperalgesic nefopam, its interactions with the transient receptor potential vanilloid subtype 1 (TRPV1) were investigated. In sensory neurons of rat embryos, dorsal root ganglion (DRG) in culture, nefopam (3-30 mumol/l) and capsazepine (TRPV1 antagonist, 10 mumol/l) prevented intracellular calcium elevation and calcitonin gene-related peptide release induced by vanilloid agonist capsaicin. Unlike nefopam, capsazepine failed to inhibit these same responses induced by KCl excess. In vivo, nefopam (0.5 and 2 mg/kg, i.v.) and capsazepine (40 mg/kg, i.p.) reduced the licking response due to intraplantar injection of capsaicin in mice. These findings suggest that nefopam exerts its analgesic and antihyperalgesic effects through multiple mechanisms including blockade of TRPV1 in addition to voltage-dependent calcium channels in the DRG.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Nefopam/pharmacology , TRPV Cation Channels/pharmacology , Animals , Calcitonin Gene-Related Peptide/metabolism , Calcium/metabolism , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Cells, Cultured , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Hyperalgesia/drug therapy , Intracellular Fluid/metabolism , Mice , Potassium Chloride/pharmacology , Rats , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Sensory System Agents/pharmacology , TRPV Cation Channels/antagonists & inhibitors
15.
Eur J Pharmacol ; 602(1): 143-7, 2009 Jan 05.
Article in English | MEDLINE | ID: mdl-19036344

ABSTRACT

Although citrulline malate (CM; CAS 54940-97-5, Stimol) is used against fatigue states, its anti-asthenic effect remains poorly documented. The objective of this double-blind study was to evaluate the effect of oral ingestion of CM on a rat model of asthenia, using in situ (31)Phosphorus magnetic resonance spectroscopy ((31)P-MRS). Muscle weakness was induced by intraperitoneal injections of Klebsiella pneumoniae endotoxin (lipopolysaccharides at 3 mg/kg) at t(0) and t(0)+24 h. For each animal, muscle function was investigated strictly non-invasively before (t(0)-24 h) and during (t(0)+48 h) endotoxemia, through a standardized rest-stimulation-recovery protocol. The transcutaneous electrical stimulation protocol consisted of 5.7 min of repeated isometric contractions at a frequency of 3.3 Hz, and force production was measured with an ergometer. CM supplementation in endotoxemic animals prevented the basal phosphocreatine/ATP ratio reduction and normalized the intracellular pH (pH(i)) time-course during muscular activity as a sign of an effect at the muscle energetics level. In addition, CM treatment avoided the endotoxemia-induced decline in developed force. These results demonstrate the efficiency of CM for limiting skeletal muscle dysfunction in rats treated with bacterial endotoxin.


Subject(s)
Citrulline/analogs & derivatives , Endotoxemia/drug therapy , Endotoxemia/physiopathology , Malates/pharmacology , Muscle, Skeletal/drug effects , Administration, Oral , Animals , Citrulline/administration & dosage , Citrulline/pharmacology , Citrulline/therapeutic use , Double-Blind Method , Endotoxemia/chemically induced , Endotoxins/pharmacology , Energy Metabolism/drug effects , Energy Metabolism/physiology , Hydrogen-Ion Concentration , Klebsiella pneumoniae/chemistry , Magnetic Resonance Spectroscopy , Malates/administration & dosage , Malates/therapeutic use , Male , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Skeletal/physiology , Muscle, Skeletal/physiopathology , Physical Exertion/drug effects , Physical Exertion/physiology , Rats , Rats, Wistar , Time Factors
16.
Proc Natl Acad Sci U S A ; 105(51): 20505-10, 2008 Dec 23.
Article in English | MEDLINE | ID: mdl-19075249

ABSTRACT

Peripheral nerves show spontaneous regenerative responses, but recovery after injury or peripheral neuropathies (toxic, diabetic, or chronic inflammatory demyelinating polyneuropathy syndromes) is slow and often incomplete, and at present no efficient treatment is available. Using well-defined peripheral nerve lesion paradigms, we assessed the therapeutic usefulness of etifoxine, recently identified as a ligand of the translocator protein (18 kDa) (TSPO), to promote axonal regeneration, modulate inflammatory responses, and improve functional recovery. We found by histologic analysis that etifoxine therapy promoted the regeneration of axons in and downstream of the lesion after freeze injury and increased axonal growth into a silicone guide tube by a factor of 2 after nerve transection. Etifoxine also stimulated neurite outgrowth in PC12 cells, and the effect was even stronger than for specific TSPO ligands. Etifoxine treatment caused a marked reduction in the number of macrophages after cryolesion within the nerve stumps, which was rapid in the proximal and delayed in the distal nerve stumps. Functional tests revealed accelerated and improved recovery of locomotion, motor coordination, and sensory functions in response to etifoxine. This work demonstrates that etifoxine, a clinically approved drug already used for the treatment of anxiety disorders, is remarkably efficient in promoting acceleration of peripheral nerve regeneration and functional recovery. Its possible mechanism of action is discussed, with reference to the neurosteroid concept. This molecule, which easily enters nerve tissues and regulates multiple functions in a concerted manner, offers promise for the treatment of peripheral nerve injuries and axonal neuropathies.


Subject(s)
Nerve Regeneration/drug effects , Oxazines/pharmacology , Peripheral Nerves/physiology , Animals , Axons , Carrier Proteins/antagonists & inhibitors , GABA-A Receptor Antagonists , Locomotion , Macrophages , Male , Motor Activity , Oxazines/therapeutic use , PC12 Cells , Peripheral Nerve Injuries , Rats , Rats, Sprague-Dawley , Receptors, GABA-A , Recovery of Function/drug effects , Sensation
17.
Psychopharmacology (Berl) ; 197(4): 523-34, 2008 May.
Article in English | MEDLINE | ID: mdl-18236028

ABSTRACT

RATIONALE: A disordered regulation of neuroactive steroids release in response to acute stress could induce GABAergic dysfunctions underlying anxiety disorders. OBJECTIVES: First, we conducted studies indicating that a short immobilization stress in anxious Balb/cByJ mice produced an anticonvulsive effect. Second, the effects of different positive allosteric modulators (etifoxine, progesterone, clonazepam, and allopregnanolone) of GABA A receptors were compared in a mouse model mimicking the disruption of the acute stress-induced neuroactive steroids release with finasteride (types I and II 5alpha-reductase inhibitor). RESULTS: The acute stress-induced anticonvulsive effect, expressed by the threshold dose of t-butylbicyclophosphorothionate-producing clonic seizures, was time-dependent. The extent of the enhancement of acute stress-induced anticonvulsive effect was lowered in the presence of finasteride. The same effect was observed with PK11195, which behaves as an antagonist of the peripheral benzodiazepine receptor in the dose range used in this study. Picrotoxin reduced the acute stress anticonvulsive effect, proving that this effect operates through the GABA A receptor. Contrary to progesterone (up to 30 mg/kg), etifoxine (50 mg/kg), allopregnanolone (10 mg/kg), and clonazepam (10 microg/kg) inhibited the finasteride effect in stressed animals. The effect of etifoxine was blocked in the presence of finasteride and picrotoxin combined in stressed animals. CONCLUSIONS: These findings support the hypothesis suggesting an involvement of neuroactive steroids in the anticonvulsive effect of restraint stress. The dual and complementary mechanisms of action of etifoxine (directly on the GABA A receptor and indirectly via the neuroactive steroids) may represent a therapeutic benefit in the treatment of various anxiety disorders with abnormal production of neuroactive steroids.


Subject(s)
Anxiety/physiopathology , Receptors, GABA-A/physiology , Seizures/physiopathology , Stress, Psychological/physiopathology , Allosteric Regulation/drug effects , Animals , Bridged Bicyclo Compounds, Heterocyclic , Clonazepam/pharmacology , Convulsants , Drug Interactions , Finasteride/pharmacology , Isoquinolines/pharmacology , Male , Mice , Mice, Inbred BALB C , Oxazines/pharmacology , Picrotoxin/pharmacology , Pregnanolone/pharmacology , Progesterone/pharmacology , Receptors, GABA-A/drug effects , Restraint, Physical , Seizures/chemically induced
18.
Muscle Nerve ; 37(4): 496-504, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18260074

ABSTRACT

Although depletion in high-energy phosphorylated compounds and mitochondrial impairment have been reported in septic skeletal muscle at rest, their impact on energy metabolism has not been documented during exercise. In this study we aimed to investigate strictly gastrocnemius muscle function non-invasively, using magnetic resonance techniques in endotoxemic rats. Endotoxemia was induced by injecting animals intraperitoneally at t(0) and t(0) + 24 h with Klebsiella pneumoniae lipopolysaccharides (at 3 mg kg(-1)). Investigations were performed at t(0) + 48 h during a transcutaneous electrical stimulation protocol consisting of 5.7 min of repeated isometric contractions at a frequency of 3.3 HZ. Endotoxin treatment produced a depletion in basal phosphocreatine content and a pronounced reduction in oxidative adenosine triphosphate (ATP) synthesis capacity, whereas the resting ATP concentration remained unchanged. During the stimulation period, endotoxemia caused a decrease in force-generating capacity that was fully accounted for by the loss of muscle mass. It further induced an acceleration of glycolytic ATP production and an increased accumulation of adenosine diphosphate (ADP, an important mitochondrial regulator) that allowed a near-normal rate of oxidative ATP synthesis. Finally, endotoxemia did not affect the total rate of ATP production or the ATP cost of contraction throughout the whole stimulation period. These data demonstrate that, in an acute septic phase, metabolic alterations in resting muscle do not impact energy supply in exercising muscle, likely as a result of adaptive mechanisms.


Subject(s)
Endotoxemia/metabolism , Energy Metabolism/physiology , Muscle, Skeletal/metabolism , Physical Exertion/physiology , Adenosine Triphosphate/metabolism , Animals , Endotoxemia/chemically induced , Hydrogen-Ion Concentration , Klebsiella pneumoniae/chemistry , Lipopolysaccharides/pharmacology , Male , Muscle Contraction/physiology , Phosphorylation , Rats , Rats, Wistar , Sepsis/chemically induced , Sepsis/metabolism
19.
Muscle Nerve ; 36(4): 505-14, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17626288

ABSTRACT

In resting skeletal muscle, endotoxemia causes disturbances in energy metabolism that could potentially disturb intracellular pH (pH(i)) during muscular activity. We tested this hypothesis using in situ (31)P-magnetic resonance spectroscopy in contracting rat gastrocnemius muscle. Endotoxemia was induced by injecting rats intraperitoneally at t(0) and t(0) + 24 h with Klebsiella pneumoniae endotoxin (lipopolysaccharides at 3 mg/kg) or saline vehicle. Muscle function was investigated strictly noninvasively at t(0) + 48 h through a transcutaneous electrical stimulation protocol consisting of 5.7 minutes of repeated isometric contraction at 3.3 HZ, and force production was measured with an ergometer. At rest, endotoxin treatment did not affect pH(i) and adenosine triphosphate concentration, but significantly reduced phosphocreatine and glycogen contents. Endotoxemia produced both a reduction of isometric force production and a marked linear recovery (0.08 +/- 0.01 pH unit/min) of pH(i) during the second part of the stimulation period. This recovery was not due to any phenomenon of fiber inactivation linked to development of muscle fatigue, and was not associated with any change in intracellular proton buffering, net proton efflux from the cell, or proton turnovers through creatine kinase reaction and oxidative phosphorylation. This paradoxical pH(i) recovery in exercising rat skeletal muscle under endotoxemia is likely due to slowing of glycolytic flux following the reduction in intramuscular glycogen content. These findings may be useful in the follow-up of septic patients and in the assessment of therapies.


Subject(s)
Endotoxemia/metabolism , Endotoxemia/rehabilitation , Intracellular Membranes/metabolism , Muscle, Skeletal/pathology , Physical Conditioning, Animal/methods , Adenosine Triphosphate/metabolism , Analysis of Variance , Animals , Endotoxemia/physiopathology , Energy Metabolism , Glycogen/metabolism , Hydrogen-Ion Concentration , In Vitro Techniques , Magnetic Resonance Spectroscopy , Male , Muscle Contraction/physiology , Muscle, Skeletal/physiopathology , Rats , Rats, Wistar
20.
Brain Res ; 1138: 30-8, 2007 Mar 23.
Article in English | MEDLINE | ID: mdl-17274964

ABSTRACT

Recent data suggested the existence of a bidirectional relation between depression and neurodegenerative diseases resulting from cerebral ischemia injury. Glutamate, a major excitatory neurotransmitter, has long been recognised to play a key role in the pathophysiology of anoxia or ischemia, due to its excessive accumulation in the extracellular space and the subsequent activation of its receptors. A characteristic response to glutamate is the increase in cytosolic Na(+) and Ca(2+) levels which is due mainly to influx from the extracellular space, with a consequent cell swelling and oxidative metabolism dysfunction. The present study examined the in vitro effects of the antidepressant and type-A monoamine oxidase inhibitor, moclobemide, in neuronal-astroglial cultures from rat cerebral cortex exposed to anoxia (for 5 and 7 h) or to glutamate (2 mM for 6 h), two in vitro models of brain ischemia. In addition, the affinity of moclobemide for the different glutamate receptor subtypes and an interaction with the cell influx of Na(+) and of Ca(2+) enhanced by veratridine and K(+) excess, respectively, were evaluated. Moclobemide (10-100 microM) included in the culture medium during anoxia or with glutamate significantly increased in a concentration-dependent manner the amount of surviving neurons compared to controls. Moclobemide displayed no binding affinity for the different glutamate receptor subtypes (IC(50)>100 microM) and did not block up to 300 microM the entry of Na(+) and of Ca(2+) activated by veratridine and K(+), respectively. These results suggest that the neuroprotective properties of moclobemide imply neither the glutamate neurotransmission nor the Na(+) and Ca(2+) channels.


Subject(s)
Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Glutamic Acid/pharmacology , Hypoxia/pathology , Moclobemide/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Neuroprotective Agents/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Calcium/metabolism , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Moclobemide/metabolism , Monoamine Oxidase Inhibitors/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/metabolism , Potassium/pharmacology , Rats , Rats, Wistar , Receptors, Glutamate/metabolism , Sodium/pharmacokinetics , Veratridine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL