Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.530
Filtrer
1.
Neurobiol Dis ; 199: 106611, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39032797

RÉSUMÉ

Ultrastructural studies of contusive spinal cord injury (SCI) in mammals have shown that the most prominent acute changes in white matter are periaxonal swelling and separation of myelin away from their axon, axonal swelling, and axonal spheroid formation. However, the underlying cellular and molecular mechanisms that cause periaxonal swelling and the functional consequences are poorly understood. We hypothesized that periaxonal swelling and loss of connectivity between the axo-myelinic interface impedes neurological recovery by disrupting conduction velocity, and glial to axonal trophic support resulting in axonal swelling and spheroid formation. Utilizing in vivo longitudinal imaging of Thy1YFP+ axons and myelin labeled with Nile red, we reveal that periaxonal swelling significantly increases acutely following a contusive SCI (T13, 30 kdyn, IH Impactor) versus baseline recordings (laminectomy only) and often precedes axonal spheroid formation. In addition, using longitudinal imaging to determine the fate of myelinated fibers acutely after SCI, we show that ∼73% of myelinated fibers present with periaxonal swelling at 1 h post SCI and âˆ¼ 51% of those fibers transition to axonal spheroids by 4 h post SCI. Next, we assessed whether cation-chloride cotransporters present within the internode contributed to periaxonal swelling and whether their modulation would increase white matter sparing and improve neurological recovery following a moderate contusive SCI (T9, 50 kdyn). Mechanistically, activation of the cation-chloride cotransporter KCC2 did not improve neurological recovery and acute axonal survival, but did improve chronic tissue sparing. In distinction, the NKKC1 antagonist bumetanide improved neurological recovery, tissue sparing, and axonal survival, in part through preventing periaxonal swelling and disruption of the axo-myelinic interface. Collectively, these data reveal a novel neuroprotective target to prevent periaxonal swelling and improve neurological recovery after SCI.


Sujet(s)
Axones , Récupération fonctionnelle , Membre-2 de la famille-12 des transporteurs de solutés , Traumatismes de la moelle épinière , Substance blanche , Animaux , Traumatismes de la moelle épinière/traitement médicamenteux , Traumatismes de la moelle épinière/anatomopathologie , Substance blanche/effets des médicaments et des substances chimiques , Substance blanche/anatomopathologie , Récupération fonctionnelle/effets des médicaments et des substances chimiques , Récupération fonctionnelle/physiologie , Membre-2 de la famille-12 des transporteurs de solutés/métabolisme , Axones/effets des médicaments et des substances chimiques , Axones/anatomopathologie , Femelle , Gaine de myéline/anatomopathologie , Gaine de myéline/effets des médicaments et des substances chimiques , Gaine de myéline/métabolisme , Souris , Inhibiteurs du symport chlorure potassium sodium/pharmacologie , Bumétanide/pharmacologie
2.
Biopharm Drug Dispos ; 45(3): 138-148, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38823029

RÉSUMÉ

Bumetanide is used widely as a tool and off-label treatment to inhibit the Na-K-2Cl cotransporter NKCC1 in the brain and thereby to normalize intra-neuronal chloride levels in several brain disorders. However, following systemic administration, bumetanide only poorly penetrates into the brain parenchyma and does not reach levels sufficient to inhibit NKCC1. The low brain penetration is a consequence of both the high ionization rate and plasma protein binding, which restrict brain entry by passive diffusion, and of brain efflux transport. In previous studies, bumetanide was determined in the whole brain or a few brain regions, such as the hippocampus. However, the blood-brain barrier and its efflux transporters are heterogeneous across brain regions, so it cannot be excluded that bumetanide reaches sufficiently high brain levels for NKCC1 inhibition in some discrete brain areas. Here, bumetanide was determined in 14 brain regions following i.v. administration of 10 mg/kg in rats. Because bumetanide is much more rapidly eliminated by rats than humans, its metabolism was reduced by pretreatment with piperonyl butoxide. Significant, up to 5-fold differences in regional bumetanide levels were determined with the highest levels in the midbrain and olfactory bulb and the lowest levels in the striatum and amygdala. Brain:plasma ratios ranged between 0.004 (amygdala) and 0.022 (olfactory bulb). Regional brain levels were significantly correlated with local cerebral blood flow. However, regional bumetanide levels were far below the IC50 (2.4 µM) determined previously for rat NKCC1. Thus, these data further substantiate that the reported effects of bumetanide in rodent models of brain disorders are not related to NKCC1 inhibition in the brain.


Sujet(s)
Encéphale , Bumétanide , Animaux , Bumétanide/pharmacologie , Bumétanide/pharmacocinétique , Bumétanide/administration et posologie , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Mâle , Rats , Inhibiteurs du symport chlorure potassium sodium/pharmacocinétique , Inhibiteurs du symport chlorure potassium sodium/pharmacologie , Inhibiteurs du symport chlorure potassium sodium/administration et posologie , Rat Sprague-Dawley , Distribution tissulaire , Membre-2 de la famille-12 des transporteurs de solutés/métabolisme , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques
3.
Aging (Albany NY) ; 16(11): 9959-9971, 2024 06 07.
Article de Anglais | MEDLINE | ID: mdl-38850525

RÉSUMÉ

This meta-analysis aimed to describe the efficacy of bumetanide in improving infarct volume, brain edema, and behavioral outcomes in animal models of cerebral ischemia. Embase, PubMed and Web of Science databases were searched from their inception to February 2024 (INPLASY:202430023). Data on the animal species, stroke model, drug dose, time of treatment, method of administration, study quality, and outcomes were extracted and pooled in a meta-analysis. The combined standardized mean difference (SMD) or mean difference (MD) estimates and 95% confidence intervals (CIs) were calculated using random- or fixed-effects models. Thirteen eligible studies involving >200 animals fulfilled the inclusion criteria and were included in this meta-analysis. Meta-analyses demonstrated that bumetanide treatment significantly reduced cerebral infarct volume (SMD: -0.42; 95% CI: -0.75, -0.09; p < 0.01; n = 186 animals) and consistently relieved brain edema (SMD: -1.39; 95% CI: -2.06, -0.72; p < 0.01; n = 64 animals). Subgroup analyses demonstrated that bumetanide treatment reduced infarct volume in transient but not permanent cerebral ischemia models. When administered after the stroke, it was more effective than treatment initiation before the stroke. Eight studies assessed the effect of bumetanide on behavioral function and the results showed that bumetanide treatment significantly improved neurobehavioral deficits (SMD: -2.35; 95% CI: -2.72, -1.97; p < 0.01; n = 250 animals). We conclude that bumetanide appears to be effective in reducing infarct volume and brain edema and improving behavioral recovery in animal models of cerebral ischemia. This mechanism needs to be confirmed through further investigation.


Sujet(s)
Bumétanide , Modèles animaux de maladie humaine , Accident vasculaire cérébral ischémique , Bumétanide/usage thérapeutique , Bumétanide/pharmacologie , Animaux , Accident vasculaire cérébral ischémique/traitement médicamenteux , Accident vasculaire cérébral ischémique/anatomopathologie , Oedème cérébral/traitement médicamenteux , Inhibiteurs du symport chlorure potassium sodium/usage thérapeutique , Inhibiteurs du symport chlorure potassium sodium/pharmacologie , Neuroprotecteurs/usage thérapeutique , Neuroprotecteurs/pharmacologie
4.
Pflugers Arch ; 476(5): 847-859, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38421407

RÉSUMÉ

Increases in the current threshold occur in optic nerve axons with the application of infra-red laser light, whose mechanism is only partly understood. In isolated rat optic nerve, laser light was applied near the site of electrical stimulation, via a flexible fibre optic. Paired applications of light produced increases in threshold that were reduced on the second application, the response recovering with increasing delays, with a time constant of 24 s. 3-min duration single applications of laser light gave rise to a rapid increase in threshold followed by a fade, whose time-constant was between 40 and 50 s. After-effects were sometimes apparent following the light application, where the resting threshold was reduced. The increase in threshold was partially blocked by 38.6 mM Li+ in combination with 5  µ M bumetanide, a manoeuvre increasing refractoriness and consistent with axonal depolarization. Assessing the effect of laser light on the nerve input resistance ruled out a previously suggested fall in myelin resistance as contributing to threshold changes. These data appear consistent with an axonal membrane potential that partly relies on temperature-dependent electroneutral Na+ influx, and where fade in the response to the laser may be caused by a gradually diminishing Na+ pump-induced hyperpolarization, in response to falling intracellular [Na+].


Sujet(s)
Axones , Lasers , Nerf optique , Sodium , Animaux , Rats , Nerf optique/métabolisme , Sodium/métabolisme , Axones/métabolisme , Axones/physiologie , Axones/effets des radiations , Potentiels de membrane/physiologie , Mâle , Bumétanide/pharmacologie , Rat Sprague-Dawley
5.
Neuromuscul Disord ; 35: 33-38, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38232533

RÉSUMÉ

The aim of this study was to establish whether bumetanide can abort an acute attack of weakness in patients with HypoPP. This was a randomised, double-blind, cross-over, placebo-controlled phase II clinical trial. Focal attack of weakness was induced by isometric exercise of ADM followed by rest (McManis protocol). Participants had two study visits and received either placebo or 2 mg bumetanide at attack onset (defined as 40 % decrement in the abductor digiti minimi CMAP amplitude from peak). CMAP measurements assessed attack severity and duration. Nine participants completed both visits. CMAP percentage of peak amplitudes in the bumetanide (40.6 %) versus placebo (34.9 %) group at 1hr following treatment did not differ significantly (estimated effect difference 5.9 % (95 % CI: (-5.7 %; 17.5 %), p = 0.27, primary outcome). CMAP amplitudes assessed by the area under the curve for early (0-2hr post-treatment) and late (2-4 h post-treatment) efficacy were not statistically different between bumetanide and placebo (early effect estimate 0.043, p = 0.3; late effect estimate 0.085, p = 0.1). Two participants recovered from the attack following bumetanide intake; none recovered following placebo. Bumetanide was well tolerated but not efficacious to rescue a focal attack in an immobilised hand in the majority of patients, although data supports further studies of this agent.


Sujet(s)
Paralysie périodique hypokaliémique , Humains , Bumétanide/pharmacologie , Bumétanide/usage thérapeutique , Muscles squelettiques , Main , Membre supérieur , Méthode en double aveugle
6.
Anesth Analg ; 138(1): 198-209, 2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-36753442

RÉSUMÉ

BACKGROUND: General anesthetics (eg, propofol and volatile anesthetics) enhance the slow-delta oscillations of the cortical electroencephalogram (EEG), which partly results from the enhancement of (γ-aminobutyric acid [GABA]) γ-aminobutyric acid-ergic (GABAergic) transmission. There is a GABAergic excitatory-inhibitory shift during postnatal development. Whether general anesthetics can enhance slow-delta oscillations in the immature brain has not yet been unequivocally determined. METHODS: Perforated patch-clamp recording was used to confirm the reversal potential of GABAergic currents throughout GABAergic development in acute brain slices of neonatal rats. The power density of the electrocorticogram and the minimum alveolar concentrations (MAC) of isoflurane and/or sevoflurane were measured in P4-P21 rats. Then, the effects of bumetanide, an inhibitor of the Na + -K + -2Cl - cotransporter (NKCC1) and K + -Cl - cotransporter (KCC2) knockdown on the potency of volatile anesthetics and the power density of the EEG were determined in vivo. RESULTS: Reversal potential of GABAergic currents were gradually hyperpolarized from P4 to P21 in cortical pyramidal neurons. Bumetanide enhanced the hypnotic effects of volatile anesthetics at P5 (for MAC LORR , isoflurane: 0.63% ± 0.07% vs 0.81% ± 0.05%, 95% confidence interval [CI], -0.257 to -0.103, P < .001; sevoflurane: 1.46% ± 0.12% vs 1.66% ± 0.09%, 95% CI, -0.319 to -0.081, P < .001); while knockdown of KCC2 weakened their hypnotic effects at P21 in rats (for MAC LORR , isoflurane: 0.58% ± 0.05% to 0.77% ± 0.20%, 95% CI, 0.013-0.357, P = .003; sevoflurane: 1.17% ± 0.04% to 1.33% ± 0.04%, 95% CI, 0.078-0.244, P < .001). For cortical EEG, slow-delta oscillations were the predominant components of the EEG spectrum in neonatal rats. Isoflurane and/or sevoflurane suppressed the power density of slow-delta oscillations rather than enhancement of it until GABAergic maturity. Enhancement of slow-delta oscillations under volatile anesthetics was simulated by preinjection of bumetanide at P5 (isoflurane: slow-delta changed ratio from -0.31 ± 0.22 to 1.57 ± 1.15, 95% CI, 0.67-3.08, P = .007; sevoflurane: slow-delta changed ratio from -0.46 ± 0.25 to 0.95 ± 0.97, 95% CI, 0.38-2.45, P = .014); and suppressed by KCC2-siRNA at P21 (isoflurane: slow-delta changed ratio from 16.13 ± 5.69 to 3.98 ± 2.35, 95% CI, -18.50 to -5.80, P = .002; sevoflurane: slow-delta changed ratio from 0.13 ± 2.82 to 3.23 ± 2.49, 95% CI, 3.02-10.79, P = .003). CONCLUSIONS: Enhancement of cortical EEG slow-delta oscillations by volatile anesthetics may require mature GABAergic inhibitory transmission during neonatal development.


Sujet(s)
Anesthésie , Anesthésiques généraux , Anesthésiques par inhalation , Isoflurane , Éthers méthyliques , Symporteurs , Rats , Animaux , Isoflurane/pharmacologie , Sévoflurane/pharmacologie , Animaux nouveau-nés , Bumétanide/pharmacologie , Acide gamma-amino-butyrique/pharmacologie , Électroencéphalographie , Hypnotiques et sédatifs , Anesthésiques par inhalation/pharmacologie
7.
Epilepsia ; 64(12): 3389-3403, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37779224

RÉSUMÉ

OBJECTIVE: A pathological excitatory action of the major inhibitory neurotransmitter γ-aminobutyric acid (GABA) has been observed in epilepsy. Blocking the Cl- importer NKCC1 with bumetanide is expected to reduce the neuronal intracellular Cl- concentration ([Cl- ]i ) and thereby attenuate the excitatory GABA response. Accordingly, several clinical trials of bumetanide for epilepsy were conducted. Although NKCC1 is expressed in both neurons and glial cells, an involvement of glial NKCC1 in seizures has not yet been reported. Astrocytes maintain high [Cl- ]i with NKCC1, and this gradient promotes Cl- efflux via the astrocytic GABAA receptor (GABAA R). This Cl- efflux buffers the synaptic cleft Cl- concentration to maintain the postsynaptic Cl- gradient during intense firing of GABAergic neurons, thereby sustaining its inhibitory action during seizure. In this study, we investigated the function of astrocytic NKCC1 in modulating the postsynaptic action of GABA in acute seizure models. METHODS: We used the astrocyte-specific conditional NKCC1 knockout (AstroNKCC1KO) mice. The seizurelike events (SLEs) in CA1 pyramidal neurons were triggered by tetanic stimulation of stratum radiatum in acute hippocampus slices. The SLE underlying GABAA R-mediated depolarization was evaluated by applying the GABAA R antagonist bicuculline. The pilocarpine-induced seizure in vivo was monitored in adult mice by the Racine scale. The SLE duration and tetanus stimulation intensity threshold and seizure behavior in AstroNKCC1KO mice and wild-type (WT) mice were compared. RESULTS: The AstroNKCC1KO mice were prone to seizures with lower threshold and longer duration of SLEs and larger GABAA R-mediated depolarization underlying the SLEs, accompanied by higher Racine-scored seizures. Bumetanide reduced these indicators of seizure in AstroNKCC1KO mice (which still express neuronal NKCC1), but not in the WT, both in vitro and in vivo. SIGNIFICANCE: Astrocytic NKCC1 inhibits GABA-mediated excitatory action during seizures, whereas neuronal NKCC1 has the converse effect, suggesting opposing actions of bumetanide on these cells.


Sujet(s)
Bumétanide , Épilepsie , Membre-2 de la famille-12 des transporteurs de solutés , Animaux , Souris , Astrocytes , Bumétanide/pharmacologie , Bumétanide/usage thérapeutique , Épilepsie/traitement médicamenteux , Acide gamma-amino-butyrique/métabolisme , Neurones , Récepteurs GABA-A/physiologie , Crises épileptiques , Inhibiteurs du symport chlorure potassium sodium/pharmacologie , Inhibiteurs du symport chlorure potassium sodium/usage thérapeutique , Membre-2 de la famille-12 des transporteurs de solutés/génétique , Synapses , Chlorures/métabolisme
8.
Cell Mol Life Sci ; 80(8): 209, 2023 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-37458846

RÉSUMÉ

The corticosteroid hormone, aldosterone, markedly enhances K+ secretion throughout the colon, a mechanism critical to its role in maintaining overall K+ balance. Previous studies demonstrated that basolateral NKCC1 was up-regulated by aldosterone in the distal colon specifically to support K+ secretion-which is distinct from the more well-established role of NKCC1 in supporting luminal Cl- secretion. However, considerable segmental variability exists between proximal and distal colonic ion transport processes, especially concerning their regulation by aldosterone. Furthermore, delineating such region-specific effects has important implications for the management of various gastrointestinal pathologies. Experiments were therefore designed to determine whether aldosterone similarly up-regulates NKCC1 in the proximal colon to support K+ secretion. Using dietary Na+ depletion as a model of secondary hyperaldosteronism in rats, we found that proximal colon NKCC1 expression was indeed enhanced in Na+-depleted (i.e., hyperaldosteronemic) rats. Surprisingly, electrogenic K+ secretion was not detectable by short-circuit current (ISC) measurements in response to either basolateral bumetanide (NKCC1 inhibitor) or luminal Ba2+ (non-selective K+ channel blocker), despite enhanced K+ secretion in Na+-depleted rats, as measured by 86Rb+ fluxes. Expression of BK and IK channels was also found to be unaltered by dietary Na+ depletion. However, bumetanide-sensitive basal and agonist-stimulated Cl- secretion (ISC) were significantly enhanced by Na+ depletion, as was CFTR Cl- channel expression. These data suggest that NKCC1-dependent secretory pathways are differentially regulated by aldosterone in proximal and distal colon. Development of therapeutic strategies in treating pathologies related to aberrant colonic K+/Cl- transport-such as pseudo-obstruction or ulcerative colitis-may benefit from these findings.


Sujet(s)
Aldostérone , Bumétanide , Animaux , Rats , Aldostérone/pharmacologie , Aldostérone/métabolisme , Bumétanide/pharmacologie , Bumétanide/métabolisme , Chlorures/métabolisme , Côlon , Potassium/métabolisme , Sodium/métabolisme
9.
Sci Adv ; 9(28): eadi5716, 2023 07 14.
Article de Anglais | MEDLINE | ID: mdl-37436983

RÉSUMÉ

Moseng et al. recently reported four cryo-electron microscopy structures of the human Na-K-2Cl cotransporter-1 (hNKCC1), both in the absence and presence of bound loop diuretic (furosemide or bumetanide). This research article included high-resolution structural information for a previously undefined structure of apo-hNKCC1 containing both the transmembrane and cytosolic carboxyl-terminal domains. The manuscript also demonstrated various conformational states of this cotransporter induced by diuretic drugs. On the basis of the structural information, the authors proposed a scissor-like inhibition mechanism that involves a coupled movement between the cytosolic and transmembrane domains of hNKCC1. This work has provided important insights into the mechanism of inhibition and substantiated the concept of a long-distance coupling involving movements of both the transmembrane and carboxyl-terminal cytoplasmic domains for inhibition.


Sujet(s)
Bumétanide , Furosémide , Humains , Cryomicroscopie électronique , Bumétanide/pharmacologie , Cytosol , Conformation moléculaire
10.
Sci Adv ; 9(28): eadh0821, 2023 07 14.
Article de Anglais | MEDLINE | ID: mdl-37436990

RÉSUMÉ

Flygaard, Habeck and Nissen question claims on bumetanide and furosemide binding to sodium-potassium-chloride cotransporter NKCC1.


Sujet(s)
Bumétanide , Furosémide , Bumétanide/pharmacologie , Conformation moléculaire
11.
Neurotox Res ; 41(6): 526-545, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37378827

RÉSUMÉ

Neonatal hypoxia-ischemia (HI) is one of the main causes of tissue damage, cell death, and imbalance between neuronal excitation and inhibition and synaptic loss in newborns. GABA, the major inhibitory neurotransmitter of the central nervous system (CNS) in adults, is excitatory at the onset of neurodevelopment and its action depends on the chloride (Cl-) cotransporters NKCC1 (imports Cl-) and KCC2 (exports Cl-) expression. Under basal conditions, the NKCC1/KCC2 ratio decreases over neurodevelopment. Thus, changes in this ratio caused by HI may be related to neurological disorders. The present study evaluated the effects of bumetanide (NKCC cotransporters inhibitor) on HI impairments in two neurodevelopmental periods. Male Wistar rat pups, 3 (PND3) and 11 (PND11) days old, were submitted to the Rice-Vannucci model. Animals were divided into 3 groups: SHAM, HI-SAL, and HI-BUM, considering each age. Bumetanide was administered intraperitoneally at 1, 24, 48, and 72 h after HI. NKCC1, KCC2, PSD-95, and synaptophysin proteins were analyzed after the last injection by western blot. Negative geotaxis, righting reflex, open field, object recognition test, and Morris water maze task were performed to assess neurological reflexes, locomotion, and memory function. Tissue atrophy and cell death were evaluated by histology. Bumetanide prevented neurodevelopmental delay, hyperactivity, and declarative and spatial memory deficits. Furthermore, bumetanide reversed HI-induced brain tissue damage, reduced neuronal death and controlled GABAergic tone, maintained the NKCC1/KCC2 ratio, and synaptogenesis close to normality. Thereby, bumetanide appears to play an important therapeutic role in the CNS, protecting the animals against HI damage and improving functional performance.


Sujet(s)
Bumétanide , Hypoxie-ischémie du cerveau , Rats , Animaux , Mâle , Bumétanide/pharmacologie , Bumétanide/usage thérapeutique , Rat Wistar , Membre-2 de la famille-12 des transporteurs de solutés/métabolisme , Ischémie/traitement médicamenteux , Hypoxie/traitement médicamenteux , Hypoxie-ischémie du cerveau/complications , Hypoxie-ischémie du cerveau/traitement médicamenteux , Encéphale/métabolisme , Cognition , Animaux nouveau-nés
12.
Brain ; 146(10): 4247-4261, 2023 10 03.
Article de Anglais | MEDLINE | ID: mdl-37082944

RÉSUMÉ

Although the Na-K-Cl cotransporter (NKCC1) inhibitor bumetanide has prominent positive effects on the pathophysiology of many neurological disorders, the mechanism of action is obscure. Attention paid to elucidating the role of Nkcc1 has mainly been focused on neurons, but recent single cell mRNA sequencing analysis has demonstrated that the major cellular populations expressing NKCC1 in the cortex are non-neuronal. We used a combination of conditional transgenic animals, in vivo electrophysiology, two-photon imaging, cognitive behavioural tests and flow cytometry to investigate the role of Nkcc1 inhibition by bumetanide in a mouse model of controlled cortical impact (CCI). Here, we found that bumetanide rescues parvalbumin-positive interneurons by increasing interneuron-microglia contacts shortly after injury. The longitudinal phenotypic changes in microglia were significantly modified by bumetanide, including an increase in the expression of microglial-derived BDNF. These effects were accompanied by the prevention of CCI-induced decrease in hippocampal neurogenesis. Treatment with bumetanide during the first week post-CCI resulted in significant recovery of working and episodic memory as well as changes in theta band oscillations 1 month later. These results disclose a novel mechanism for the neuroprotective action of bumetanide mediated by an acceleration of microglial activation dynamics that leads to an increase in parvalbumin interneuron survival following CCI, possibly resulting from increased microglial BDNF expression and contact with interneurons. Salvage of interneurons may normalize ambient GABA, resulting in the preservation of adult neurogenesis processes as well as contributing to bumetanide-mediated improvement of cognitive performance.


Sujet(s)
Bumétanide , Inhibiteurs du symport chlorure potassium sodium , Souris , Animaux , Bumétanide/pharmacologie , Inhibiteurs du symport chlorure potassium sodium/pharmacologie , Microglie/métabolisme , Facteur neurotrophique dérivé du cerveau/métabolisme , Parvalbumines/métabolisme , Parvalbumines/pharmacologie , Membre-2 de la famille-12 des transporteurs de solutés , Interneurones/métabolisme , Neurogenèse
13.
Sci Rep ; 13(1): 5685, 2023 04 17.
Article de Anglais | MEDLINE | ID: mdl-37069177

RÉSUMÉ

Angelman syndrome is a neurodevelopmental disorder caused by loss of function of the maternally expressed UBE3A gene. Treatments for the main manifestations, including cognitive dysfunction or epilepsy, are still under development. Recently, the Cl- importer Na+-K+-Cl- cotransporter 1 (NKCC1) and the Cl- exporter K+-Cl- cotransporter 2 (KCC2) have garnered attention as therapeutic targets for many neurological disorders. Dysregulation of neuronal intracellular Cl- concentration ([Cl-]i) is generally regarded as one of the mechanisms underlying neuronal dysfunction caused by imbalanced expression of these cation-chloride cotransporters (CCCs). Here, we analyzed the regulation of [Cl-]i and the effects of bumetanide, an NKCC1 inhibitor, in Angelman syndrome models (Ube3am-/p+ mice). We observed increased NKCC1 expression and decreased KCC2 expression in the hippocampi of Ube3am-/p+ mice. The average [Cl-]i of CA1 pyramidal neurons was not significantly different but demonstrated greater variance in Ube3am-/p+ mice. Tonic GABAA receptor-mediated Cl- conductance was reduced, which may have contributed to maintaining the normal average [Cl-]i. Bumetanide administration restores cognitive dysfunction in Ube3am-/p+ mice. Seizure susceptibility was also reduced regardless of the genotype. These results suggest that an imbalanced expression of CCCs is involved in the pathophysiological mechanism of Ube3am-/p+ mice, although the average [Cl-]i is not altered. The blockage of NKCC1 may be a potential therapeutic strategy for patients with Angelman syndrome.


Sujet(s)
Syndrome d'Angelman , Épilepsie , Symporteurs , Souris , Animaux , Symporteurs des ions sodium-potassium-chlorure/génétique , Symporteurs des ions sodium-potassium-chlorure/métabolisme , Bumétanide/pharmacologie , Syndrome d'Angelman/traitement médicamenteux , Syndrome d'Angelman/génétique , Chlorures/métabolisme , Symporteurs/génétique , Symporteurs/métabolisme , Épilepsie/traitement médicamenteux , Épilepsie/génétique , Récepteurs GABA-A
14.
Epilepsy Behav ; 142: 109189, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-37037061

RÉSUMÉ

Birth asphyxia and the resulting hypoxic-ischemic encephalopathy (HIE) are highly associated with perinatal and neonatal death, neonatal seizures, and an adverse later-life outcome. Currently used drugs, including phenobarbital and midazolam, have limited efficacy to suppress neonatal seizures. There is a medical need to develop new therapies that not only suppress neonatal seizures but also prevent later-life consequences. We have previously shown that the loop diuretic bumetanide does not potentiate the effects of phenobarbital in a rat model of birth asphyxia. Here we compared the effects of bumetanide (0.3 or 10 mg/kg i.p.), midazolam (1 mg/kg i.p.), and a combination of bumetanide and midazolam on neonatal seizures and later-life outcomes in this model. While bumetanide at either dose was ineffective when administered alone, the higher dose of bumetanide markedly potentiated midazolam's effect on neonatal seizures. Median bumetanide brain levels (0.47-0.53 µM) obtained with the higher dose were in the range known to inhibit the Na-K-Cl-cotransporter NKCC1 but it remains to be determined whether brain NKCC1 inhibition was underlying the potentiation of midazolam. When behavioral and cognitive alterations were examined over three months after asphyxia, treatment with the bumetanide/midazolam combination, but not with bumetanide or midazolam alone, prevented impairment of learning and memory. Furthermore, the combination prevented the loss of neurons in the dentate hilus and aberrant mossy fiber sprouting in the CA3a area of the hippocampus. The molecular mechanisms that explain that bumetanide potentiates midazolam but not phenobarbital in the rat model of birth asphyxia remain to be determined.


Sujet(s)
Asphyxie néonatale , Épilepsie , Humains , Nouveau-né , Rats , Animaux , Bumétanide/usage thérapeutique , Bumétanide/pharmacologie , Midazolam/usage thérapeutique , Anticonvulsivants/usage thérapeutique , Anticonvulsivants/pharmacologie , Asphyxie/complications , Asphyxie/traitement médicamenteux , Naissance à terme , Membre-2 de la famille-12 des transporteurs de solutés , Phénobarbital/usage thérapeutique , Phénobarbital/pharmacologie , Épilepsie/traitement médicamenteux , Asphyxie néonatale/complications , Asphyxie néonatale/traitement médicamenteux , Crises épileptiques/traitement médicamenteux , Crises épileptiques/étiologie
15.
J Physiol ; 601(8): 1425-1447, 2023 04.
Article de Anglais | MEDLINE | ID: mdl-36847245

RÉSUMÉ

Current anti-spastic medication significantly compromises motor recovery after spinal cord injury (SCI), indicating a critical need for alternative interventions. Because a shift in chloride homeostasis decreases spinal inhibition and contributes to hyperreflexia after SCI, we investigated the effect of bumetanide, an FDA-approved sodium-potassium-chloride intruder (NKCC1) antagonist, on presynaptic and postsynaptic inhibition. We compared its effect with step-training as it is known to improve spinal inhibition by restoring chloride homeostasis. In SCI rats, a prolonged bumetanide treatment increased postynaptic inhibition but not presynaptic inhibition of the plantar H-reflex evoked by posterior biceps and semitendinosus (PBSt) group I afferents. By using in vivo intracellular recordings of motoneurons, we further show that a prolonged bumetanide increased postsynaptic inhibition by hyperpolarizing the reversal potential for inhibitory postsynaptic potentials (IPSPs) after SCI. However, in step-trained SCI rats an acute delivery of bumetanide decreased presynaptic inhibition of the H-reflex, but not postsynaptic inhibition. These results suggest that bumetanide might be a viable option to improve postsynaptic inhibition after SCI, but it also decreases the recovery of presynaptic inhibition with step-training. We discuss whether the effects of bumetanide are mediated by NKCC1 or by off-target effects. KEY POINTS: After spinal cord injury (SCI), chloride homeostasis is dysregulated over time in parallel with the decrease in presynaptic inhibition of Ia afferents and postsynaptic inhibition of motoneurons, and the development of spasticity. While step-training counteracts these effects, it cannot always be implemented in the clinic because of comorbidities. An alternative intervention is to use pharmacological strategies to decrease spasticity without hindering the recovery of motor function with step-training. Here we found that, after SCI, a prolonged bumetanide (an FDA-approved antagonist of the sodium-potassium-chloride intruder, NKCC1) treatment increases postsynaptic inhibition of the H-reflex, and it hyperpolarizes the reversal potential for inhibitory postsynaptic potentials in motoneurons. However, in step-trained SCI, an acute delivery of bumetanide decreases presynaptic inhibition of the H-reflex, but not postsynaptic inhibition. Our results suggest that bumetanide has the potential to decrease spastic symptoms related to a decrease in postsynaptic but not presynaptic inhibition after SCI.


Sujet(s)
Bumétanide , Traumatismes de la moelle épinière , Rats , Animaux , Bumétanide/pharmacologie , Moelle spinale/physiologie , Chlorures , Traumatismes de la moelle épinière/traitement médicamenteux , Motoneurones/physiologie , Spasticité musculaire
16.
Mol Pain ; 19: 17448069231159855, 2023.
Article de Anglais | MEDLINE | ID: mdl-36760008

RÉSUMÉ

Previous studies have confirmed the relationship between chloride homeostasis and pain. However, the role of sodium potassium chloride co-transporter isoform 1 (NKCC1) in dorsal horn and dorsal root ganglion neurons (DRGs) in spinal cord injury (SCI)-induced neuropathic pain (NP) remains inconclusive. Therefore, we aimed to explore whether suppression of NKCC1 in the spinal cord and DRGs alleviate the NP of adult rats with thoracic spinal cord contusion. Thirty adult female Sprague-Dawley rats (8 week-old, weighing 250-280 g) were randomly divided into three groups with ten animals in each group (sham, SCI, and bumetanide groups). The paw withdrawal mechanical threshold and paw withdrawal thermal latency were recorded before injury (baseline) and on post-injury days 14, 21, 28, and 35. At the end of experiment, western blotting (WB) analysis, quantitative real-time Polymerase Chain Reaction (PCR) and immunofluorescence were performed to quantify NKCC1 expression. Our results revealed that NKCC1 protein expression in the spinal cord and DRGs was significantly up-regulated in rats with SCI. Intraperitoneal treatment of bumetanide (an NKCC1 inhibitor) reversed the expression of NKCC1 in the dorsal horn and DRGs and ameliorated mechanical ectopic pain and thermal hypersensitivities in the SCI rats. Our study demonstrated the occurrence of NKCC1 overexpression in the spinal cord and DRGs in a rodent model of NP and indicated that changes in the peripheral nervous system also play a major role in promoting pain sensitization after SCI.


Sujet(s)
Névralgie , Traumatismes de la moelle épinière , Rats , Femelle , Animaux , Rat Sprague-Dawley , Bumétanide/métabolisme , Bumétanide/pharmacologie , Ganglions sensitifs des nerfs spinaux/métabolisme , Corne dorsale de la moelle spinale/métabolisme , Traumatismes de la moelle épinière/métabolisme , Névralgie/métabolisme , Moelle spinale/métabolisme , Hyperalgésie/métabolisme
17.
ACS Chem Neurosci ; 14(6): 1146-1155, 2023 03 15.
Article de Anglais | MEDLINE | ID: mdl-36802490

RÉSUMÉ

Sevoflurane (Sevo) is one of the most commonly used general anesthetics for infants and young children. We investigated whether Sevo impairs neurological functions, myelination, and cognition via the γ-aminobutyric acid A receptor (GABAAR) and Na+-K+-2Cl- cotransporter (NKCC1) in neonatal mice. On postnatal days 5-7, mice were exposed to 3% Sevo for 2 h. On postnatal day 14, mouse brains were dissected, and oligodendrocyte precursor cell line level lentivirus knockdown of GABRB3, immunofluorescence, and transwell migration assays were performed. Finally, behavioral tests were conducted. Multiple Sevo exposure groups exhibited increased neuronal apoptosis levels and decreased neurofilament protein levels in the mouse cortex compared with the control group. Sevo exposure inhibited the proliferation, differentiation, and migration of the oligodendrocyte precursor cells, thereby affecting their maturation process. Electron microscopy revealed that Sevo exposure reduced myelin sheath thickness. The behavioral tests showed that multiple Sevo exposures induced cognitive impairment. GABAAR and NKCC1 inhibition provided protection against Sevo-induced neurotoxicity and cognitive dysfunction. Thus, bicuculline and bumetanide can protect against Sevo-induced neuronal injury, myelination impairment, and cognitive dysfunction in neonatal mice. Furthermore, GABAAR and NKCC1 may be mediators of Sevo-induced myelination impairment and cognitive dysfunction.


Sujet(s)
Anesthésiques par inhalation , Bumétanide , Animaux , Souris , Sévoflurane/pharmacologie , Bumétanide/pharmacologie , Bicuculline/pharmacologie , Animaux nouveau-nés , Cognition , Acide gamma-amino-butyrique , Anesthésiques par inhalation/toxicité
18.
Epilepsy Behav ; 139: 109057, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36586153

RÉSUMÉ

Loop diuretics such as furosemide and bumetanide, which act by inhibiting the Na-K-2Cl cotransporter NKCC2 at the thick ascending limb of the loop of Henle, have been shown to exert anti-seizure effects. However, the exact mechanism of this effect is not known. For bumetanide, it has been suggested that inhibition of the NKCC isoform NKCC1 in the membrane of brain neurons may be involved; however, NKCC1 is expressed by virtually all cell types in the brain, which makes any specific targeting of neuronal NKCC1 by bumetanide impossible. In addition, bumetanide only poorly penetrates the brain. We have previously shown that loop diuretics azosemide and torasemide also potently inhibit NKCC1. In contrast to bumetanide and furosemide, azosemide and torasemide lack a carboxylic group, which should allow them to better penetrate through biomembranes by passive diffusion. Because of the urgent medical need to develop new treatments for neonatal seizures and their adverse outcome, we evaluated the effects of azosemide and torasemide, administered alone or in combination with phenobarbital or midazolam, in a rat model of birth asphyxia and neonatal seizures. Neither diuretic suppressed the seizures when administered alone but torasemide potentiated the anti-seizure effect of midazolam. Brain levels of torasemide were below those needed to inhibit NKCC1. In addition to suppressing seizures, the combination of torasemide and midazolam, but not midazolam alone, prevented the cognitive impairment of the post-asphyxial rats at 3 months after asphyxia. Furthermore, aberrant mossy fiber sprouting in the hippocampus was more effectively prevented by the combination. We assume that either an effect on NKCC1 at the blood-brain barrier and/or cells in the periphery or the NKCC2-mediated diuretic effect of torasemide are involved in the present findings. Our data suggest that torasemide may be a useful option for improving the treatment of neonatal seizures and their adverse outcome.


Sujet(s)
Épilepsie , Inhibiteurs du symport chlorure potassium sodium , Rats , Animaux , Inhibiteurs du symport chlorure potassium sodium/usage thérapeutique , Inhibiteurs du symport chlorure potassium sodium/pharmacologie , Bumétanide/usage thérapeutique , Bumétanide/pharmacologie , Torasémide , Furosémide/usage thérapeutique , Furosémide/pharmacologie , Asphyxie , Membre-2 de la famille-12 des transporteurs de solutés/métabolisme , Diurétiques/usage thérapeutique , Diurétiques/pharmacologie
19.
Neuroscience ; 510: 95-108, 2023 02 01.
Article de Anglais | MEDLINE | ID: mdl-36493910

RÉSUMÉ

Aquaporin-4 (AQP4) regulates retinal water homeostasis and participates in retinal oedema pathophysiology. ß-dystroglycan (ß-DG) is responsible for AQP4 polarization and can be cleaved by matrix metalloproteinase-9 (MMP9). Retinal oedema induced by ischemia-reperfusion (I/R) injury is an early complication. Bumetanide (BU) has potential efficacy against cytotoxic oedema. Our study investigated the effects of ß-DG cleavage on AQP4 and the roles of BU in a rat retinal I/R injury model. The model was induced by applying 110 mm Hg intraocular pressure to the anterior eye chamber. BU and U0126 (a selective ERK inhibitor) were intraperitoneally administered 15 and 30 min, respectively, before I/R induction. Rhodamine isothiocyanate extravasation detection, quantitative real-time PCR, transmission electron microscopy, hematoxylin-eosin staining, immunofluorescence staining, western blotting, and TUNEL staining were performed. AQP4 lost its polarization in the retinal perivascular domain as a result of ß-DG cleavage. BU rescued AQP4 depolarization, suppressed AQP4 protein expression, attenuated retinal cytotoxic oedema, and downregulated ß-DG and AQP4 mRNA expression. BU suppressed glial responses and mitochondria-mediated apoptotic protein expression, including that of Caspase-3 and Cyto C, raised the Bcl-2/Bax ratio, and lowered the number of apoptotic cells in the retina. Both BU and U0126 downregulated p-ERK and MMP9 expression. Thus, BU treatment suppressed ß-DG cleavage, recovered AQP4 polarization partially via inhibiting ERK/MMP9 signaling pathway, and possess potential neuroprotective efficacy in the rat retinal ischemia-reperfusion injury model.


Sujet(s)
Oedème papillaire , Lésion d'ischémie-reperfusion , Animaux , Rats , Aquaporine-4/métabolisme , Bumétanide/pharmacologie , Dystroglycanes/génétique , Dystroglycanes/métabolisme , Oedème , Matrix metalloproteinase 9/métabolisme , Neuroprotection , Lésion d'ischémie-reperfusion/métabolisme , Rétine/métabolisme
20.
Appl Neuropsychol Child ; 12(1): 88-95, 2023.
Article de Anglais | MEDLINE | ID: mdl-34860628

RÉSUMÉ

The behavioral phenotypes emerge from cognitive architecture comprising attention, executive functions, and primary communication skills that all have shown remarkable deficits in Down's Syndrome (DS). These states arise from the proper functional interactions of the contributing neurotransmission and neuromodulation systems and other coding platforms. Gamma-aminobutyric acid (GABA) is an integral part of the neural interaction and regulation networks that its reverse action leads to broad detrimental consequences. This inhibitory substance needs an appropriate balance of co-transporters that largely shape the ionic milieu. Bumetanide, a specific NKCC1 inhibitor used for an eighteen-month interval, showed promising effects in restoring some behavior deficits in a fourteen-year-old boy diagnosed with genetically confirmed mosaic Down's Syndrome.


Sujet(s)
Syndrome de Down , Humains , Syndrome de Down/complications , Bumétanide/pharmacologie , Trisomie , Mosaïcisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE