Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 622(7981): 195-201, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37730991

RESUMEN

Type A γ-aminobutyric acid receptors (GABAARs) are the principal inhibitory receptors in the brain and the target of a wide range of clinical agents, including anaesthetics, sedatives, hypnotics and antidepressants1-3. However, our understanding of GABAAR pharmacology has been hindered by the vast number of pentameric assemblies that can be derived from 19 different subunits4 and the lack of structural knowledge of clinically relevant receptors. Here, we isolate native murine GABAAR assemblies containing the widely expressed α1 subunit and elucidate their structures in complex with drugs used to treat insomnia (zolpidem (ZOL) and flurazepam) and postpartum depression (the neurosteroid allopregnanolone (APG)). Using cryo-electron microscopy (cryo-EM) analysis and single-molecule photobleaching experiments, we uncover three major structural populations in the brain: the canonical α1ß2γ2 receptor containing two α1 subunits, and two assemblies containing one α1 and either an α2 or α3 subunit, in which the single α1-containing receptors feature a more compact arrangement between the transmembrane and extracellular domains. Interestingly, APG is bound at the transmembrane α/ß subunit interface, even when not added to the sample, revealing an important role for endogenous neurosteroids in modulating native GABAARs. Together with structurally engaged lipids, neurosteroids produce global conformational changes throughout the receptor that modify the ion channel pore and the binding sites for GABA and insomnia medications. Our data reveal the major α1-containing GABAAR assemblies, bound with endogenous neurosteroid, thus defining a structural landscape from which subtype-specific drugs can be developed.


Asunto(s)
Microscopía por Crioelectrón , Neuroesteroides , Receptores de GABA-A , Ácido gamma-Aminobutírico , Animales , Ratones , Sitios de Unión/efectos de los fármacos , Depresión Posparto/tratamiento farmacológico , Flurazepam/farmacología , Ácido gamma-Aminobutírico/metabolismo , Hipnóticos y Sedantes/farmacología , Activación del Canal Iónico/efectos de los fármacos , Neuroesteroides/metabolismo , Neuroesteroides/farmacología , Fotoblanqueo , Pregnanolona/farmacología , Conformación Proteica/efectos de los fármacos , Subunidades de Proteína/química , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/metabolismo , Receptores de GABA-A/química , Receptores de GABA-A/efectos de los fármacos , Receptores de GABA-A/metabolismo , Receptores de GABA-A/ultraestructura , Trastornos del Inicio y del Mantenimiento del Sueño/tratamiento farmacológico , Zolpidem/farmacología
2.
Epilepsia ; 65(3): e41-e46, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38243753

RESUMEN

Trilostane is a 3ß-hydroxysteroid dehydrogenase/Δ5-4 isomerase inhibitor able to produce a manyfold increase in brain levels of various neurosteroids, including allopregnanolone. We previously found that treatment with trilostane can slow down epileptogenesis in the kainic acid (KA) model of temporal lobe epilepsy. It is unknown whether trilostane may have a similar effect on the progression of epilepsy severity, as observed in KA-treated rats. Consequently, we investigated the effects of trilostane (50 mg/kg/day, 1 week) in epileptic rats, given 64 days after KA administration. Seizures were monitored by video-electrocorticographic recordings before and during the treatment with trilostane or vehicle (sesame oil), and neurosteroid levels were measured in serum and cerebral tissue using liquid chromatography-electrospray tandem mass spectrometry after treatment. Pregnenolone sulfate, pregnenolone, progesterone, 5α-dihydroprogesterone, and allopregnanolone peripheral levels were massively increased by trilostane. With the only exception of hippocampal pregnenolone sulfate, the other neurosteroids augmented in both the neocortex and hippocampus. Only pregnanolone levels were not upregulated by trilostane. As expected, a significant increase in the seizure occurrence was observed in rats receiving the vehicle, but not in the trilostane group. This suggests that the increased availability of neurosteroids produced a disease-modifying effect in the brain of epileptic rats.


Asunto(s)
Epilepsia , Neuroesteroides , Ratas , Animales , Neuroesteroides/farmacología , Pregnanolona/farmacología , Epilepsia/inducido químicamente , Epilepsia/tratamiento farmacológico , Encéfalo , Convulsiones/inducido químicamente , Convulsiones/tratamiento farmacológico
3.
Cell Mol Life Sci ; 80(2): 42, 2023 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-36645496

RESUMEN

N-methyl-D-aspartate receptors (NMDARs) play vital roles in normal brain functions (i.e., learning, memory, and neuronal development) and various neuropathological conditions, such as epilepsy, autism, Parkinson's disease, Alzheimer's disease, and traumatic brain injury. Endogenous neuroactive steroids such as 24(S)-hydroxycholesterol (24(S)-HC) have been shown to influence NMDAR activity, and positive allosteric modulators (PAMs) derived from 24(S)-hydroxycholesterol scaffold can also enhance NMDAR function. This study describes the structural determinants and mechanism of action for 24(S)-hydroxycholesterol and two novel synthetic analogs (SGE-550 and SGE-301) on NMDAR function. We also show that these agents can mitigate the altered function caused by a set of loss-of-function missense variants in NMDAR GluN subunit-encoding GRIN genes associated with neurological and neuropsychiatric disorders. We anticipate that the evaluation of novel neuroactive steroid NMDAR PAMs may catalyze the development of new treatment strategies for GRIN-related neuropsychiatric conditions.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades del Sistema Nervioso , Neuroesteroides , Humanos , Receptores de N-Metil-D-Aspartato/metabolismo , Neuroesteroides/farmacología , Neuroesteroides/uso terapéutico , Hidroxicolesteroles/farmacología , Hidroxicolesteroles/uso terapéutico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/genética , Enfermedad de Alzheimer/tratamiento farmacológico , Esteroides/farmacología , Regulación Alostérica/fisiología
4.
Eur Arch Psychiatry Clin Neurosci ; 273(7): 1477-1487, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36574032

RESUMEN

There is need for novel fast acting treatment options in affective disorders. 3α-reduced neurosteroids such as allopregnanolone are powerful positive allosteric modulators of GABAA receptors and target also extrasynaptic receptors. Their synthesis is mediated by the translocator protein 18 kDa (TSPO). TSPO ligands not only promote endogenous neurosteroidogenesis, but also exert a broad spectrum of functions involving modulation of mitochondrial activity and acting as anti-inflammatory and neuroregenerative agents. Besides affective symptoms, in depression cognitive impairment can be frequently observed, which may be ameliorated through targeting of extrasynaptic GABAA receptors either via TSPO ligands or exogenously administered 3α-reduced neurosteroids. Interestingly, recent findings indicate an enhanced activation of the complement system, e.g., enhanced expression of C1q, both in depression and dementia. It is of note that benzodiazepines have been shown to reduce long-term potentiation and to cause cognitive decline. Intriguingly, TSPO may be crucial in mediating the effects of benzodiazepines on synaptic pruning. Here, we discuss how benzodiazepines and TSPO may interfere with synaptic pruning. Moreover, we highlight recent developments of TSPO ligands and 3α-reduced neurosteroids as therapeutic agents. Etifoxine is the only clinically available TSPO ligand so far and has been studied in anxiety disorders. Regarding 3α-reduced neurosteroids, brexanolone, an intravenous formulation of allopregnanolone, has been approved for the treatment of postpartum depression and zuranolone, an orally available 3α-reduced neurosteroid, is currently being studied in major depressive disorder and postpartum depression. As such, 3α-reduced neurosteroids and TSPO ligands may constitute promising treatment approaches for affective disorders.


Asunto(s)
Ansiolíticos , Depresión Posparto , Trastorno Depresivo Mayor , Neuroesteroides , Humanos , Femenino , Neuroesteroides/farmacología , Ansiolíticos/uso terapéutico , Pregnanolona/farmacología , Ligandos , Depresión , Depresión Posparto/tratamiento farmacológico , Trastorno Depresivo Mayor/tratamiento farmacológico , Neurotransmisores/farmacología , Neurotransmisores/metabolismo , Receptores de GABA-A , Benzodiazepinas , Proteínas Portadoras , Plasticidad Neuronal , Cognición , Ácido gamma-Aminobutírico , Receptores de GABA/metabolismo
5.
Int J Mol Sci ; 24(10)2023 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-37240402

RESUMEN

Neurosteroids and benzodiazepines are modulators of the GABAA receptors, thereby causing anxiolysis. Furthermore, benzodiazepines such as midazolam are known to cause adverse side-effects on cognition upon administration. We previously found that midazolam at nanomolar concentrations (10 nM) blocked long-term potentiation (LTP). Here, we aim to study the effect of neurosteroids and their synthesis using XBD173, which is a synthetic compound that promotes neurosteroidogenesis by binding to the translocator protein 18 kDa (TSPO), since they might provide anxiolytic activity with a favourable side-effect profile. By means of electrophysiological measurements and the use of mice with targeted genetic mutations, we revealed that XBD173, a selective ligand of the translocator protein 18 kDa (TSPO), induced neurosteroidogenesis. In addition, the exogenous application of potentially synthesised neurosteroids (THDOC and allopregnanolone) did not depress hippocampal CA1-LTP, the cellular correlate of learning and memory. This phenomenon was observed at the same concentrations that neurosteroids conferred neuroprotection in a model of ischaemia-induced hippocampal excitotoxicity. In conclusion, our results indicate that TSPO ligands are promising candidates for post-ischaemic recovery exerting neuroprotection, in contrast to midazolam, without detrimental effects on synaptic plasticity.


Asunto(s)
Midazolam , Neuroesteroides , Ratones , Animales , Midazolam/farmacología , Neuroesteroides/farmacología , Neuroprotección , Hipoglucemiantes/farmacología , Receptores de GABA-A/metabolismo , Benzodiazepinas/farmacología , Proteínas Portadoras , Ligandos , Potenciación a Largo Plazo , Ácido gamma-Aminobutírico/farmacología
6.
Mol Pharmacol ; 101(2): 68-77, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34853153

RESUMEN

The GABAA receptor is inhibited by the endogenous sulfated steroids pregnenolone sulfate (PS) and dehydroepiandrosterone sulfate (DHEAS). It has been proposed in previous work that these steroids act by enhancing desensitization of the receptor. Here, we have investigated the modulatory effects of the steroids on the human α1ß3γ2L GABAA receptor. Using electrophysiology and quantitative model-based data analysis, we show that exposure to the steroid promotes occupancy of a nonconducting state that retains high affinity to the transmitter but whose properties differ from those of the classic, transmitter-induced desensitized state. From the analysis of the inhibitory actions of two combined steroids, we infer that PS and DHEAS act through shared or overlapping binding sites. SIGNIFICANCE STATEMENT: Previous work has proposed that sulfated neurosteroids inhibit the GABAA receptor by enhancing the rate of entry into the desensitized state. This study shows that the inhibitory steroids pregnenolone sulfate and dehydroepiandrosterone sulfate act through a common interaction site by stabilizing a distinct nonconducting state.


Asunto(s)
Sulfato de Deshidroepiandrosterona/farmacología , Antagonistas del GABA/farmacología , Pregnenolona/farmacología , Receptores de GABA-A/metabolismo , Animales , Sulfato de Deshidroepiandrosterona/química , Relación Dosis-Respuesta a Droga , Femenino , Antagonistas del GABA/química , Humanos , Neuroesteroides/química , Neuroesteroides/farmacología , Pregnenolona/química , Estabilidad Proteica , Receptores de GABA-A/química , Xenopus laevis
7.
J Neurochem ; 160(1): 113-127, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34482541

RESUMEN

Stroke is the primary reason for death and disability worldwide, with few treatment strategies to date. Neurosteroids, which are natural molecules in the brain, have aroused great interest in the field of stroke. Neurosteroids are a kind of steroid that acts on the nervous system, and are synthesized in the mitochondria of neurons or glial cells using cholesterol or other steroidal precursors. Neurosteroids mainly include estrogen, progesterone (PROG), allopregnanolone, dehydroepiandrosterone (DHEA), and vitamin D (VD). Most of the preclinical studies have confirmed that neurosteroids can decrease the risk of stroke, and improve stroke outcomes. In the meantime, neurosteroids have been shown to have a positive therapeutic significance in some post-stroke complications, such as epilepsy, depression, anxiety, cardiac complications, movement disorders, and post-stroke pain. In this review, we report the historical background, modulatory mechanisms of neurosteroids in stroke and post-stroke complications, and emphasize on the application prospect of neurosteroids in stroke therapy.


Asunto(s)
Fármacos Neuroprotectores/farmacología , Neuroesteroides/farmacología , Accidente Cerebrovascular , Animales , Humanos
8.
Int J Mol Sci ; 23(21)2022 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-36361865

RESUMEN

Muscarinic acetylcholine receptors are membrane receptors involved in many physiological processes. Malfunction of muscarinic signaling is a cause of various internal diseases, as well as psychiatric and neurologic conditions. Cholesterol, neurosteroids, neuroactive steroids, and steroid hormones are molecules of steroid origin that, besides having well-known genomic effects, also modulate membrane proteins including muscarinic acetylcholine receptors. Here, we review current knowledge on the allosteric modulation of muscarinic receptors by these steroids. We give a perspective on the research on the non-genomic effects of steroidal compounds on muscarinic receptors and drug development, with an aim to ultimately exploit such knowledge.


Asunto(s)
Neuroesteroides , Neuroesteroides/farmacología , Hormonas/metabolismo , Receptores Muscarínicos , Esteroides/farmacología , Colesterol
9.
Int J Mol Sci ; 24(1)2022 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-36613951

RESUMEN

Muscarinic acetylcholine receptors expressed in the central nervous system mediate various functions, including cognition, memory, or reward. Therefore, muscarinic receptors represent potential pharmacological targets for various diseases and conditions, such as Alzheimer's disease, schizophrenia, addiction, epilepsy, or depression. Muscarinic receptors are allosterically modulated by neurosteroids and steroid hormones at physiologically relevant concentrations. In this review, we focus on the modulation of muscarinic receptors by neurosteroids and steroid hormones in the context of diseases and disorders of the central nervous system. Further, we propose the potential use of neuroactive steroids in the development of pharmacotherapeutics for these diseases and conditions.


Asunto(s)
Neuroesteroides , Neuroesteroides/farmacología , Sistema Nervioso Central , Esteroides/farmacología , Esteroides/fisiología , Hormonas , Receptores Muscarínicos , Colinérgicos
10.
Br J Anaesth ; 126(1): 245-255, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32859366

RESUMEN

BACKGROUND: The mechanisms underlying the role of T-type calcium channels (T-channels) in thalamocortical excitability and oscillations in vivo during neurosteroid-induced hypnosis are largely unknown. METHODS: We used patch-clamp electrophysiological recordings from acute brain slices ex vivo, recordings of local field potentials (LFPs) from the central medial thalamic nucleus in vivo, and wild-type (WT) and Cav3.1 knock-out mice to investigate the molecular mechanisms of hypnosis induced by the neurosteroid analogue (3ß,5ß,17ß)-3-hydroxyandrostane-17-carbonitrile (3ß-OH). RESULTS: Patch-clamp recordings showed that 3ß-OH inhibited isolated T-currents but had no effect on phasic or tonic γ-aminobutyric acid A currents. Also in acute brain slices, 3ß-OH inhibited the spike firing mode more profoundly in WT than in Cav3.1 knockout mice. Furthermore, 3ß-OH significantly hyperpolarised neurones, reduced the amplitudes of low threshold spikes, and diminished rebound burst firing only in WT mice. We found that 80 mg kg-1 i.p. injections of 3ß-OH induced hypnosis in >60% of WT mice but failed to induce hypnosis in the majority of mutant mice. A subhypnotic dose of 3ß-OH (20 mg kg-1 i.p.) accelerated induction of hypnosis by isoflurane only in WT mice, but had similar effects on the maintenance of isoflurane-induced hypnosis in both WT and Cav3.1 knockout mice. In vivo recordings of LFPs showed that a hypnotic dose of 3ß-OH increased δ, θ, α, and ß oscillations in WT mice in comparison with Cav3.1 knock-out mice. CONCLUSIONS: The Cav3.1 T-channel isoform is critical for diminished thalamocortical excitability and oscillations that underlie neurosteroid-induced hypnosis.


Asunto(s)
Androstanoles/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Canales de Calcio Tipo T/metabolismo , Hipnóticos y Sedantes/farmacología , Nitrilos/farmacología , Androstanoles/metabolismo , Animales , Fenómenos Electrofisiológicos , Hipnóticos y Sedantes/metabolismo , Masculino , Ratones , Ratones Noqueados , Modelos Animales , Neuroesteroides/metabolismo , Neuroesteroides/farmacología , Nitrilos/metabolismo
11.
Br J Anaesth ; 127(3): 435-446, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33972091

RESUMEN

BACKGROUND: We recently showed that a neurosteroid analogue, (3ß,5ß,17ß)-3-hydroxyandrostane-17-carbonitrile (3ß-OH), induced hypnosis in rats. The aim of the present study was to evaluate the hypnotic and anaesthetic potential of 3ß-OH further using electroencephalography. METHODS: We used behavioural assessment and cortical electroencephalogram (EEG) spectral power analysis to examine hypnotic and anaesthetic effects of 3ß-OH (30 and 60 mg kg-1) administered intraperitoneally or intravenously to young adult male and female rats. RESULTS: We found dose-dependent sex differences in 3ß-OH-induced hypnosis and EEG changes. Both male and female rats responded similarly to i.p. 3ß-OH 30 mg kg-1. However, at the higher dose (60 mg kg-1, i.p.), female rats had two-fold longer duration of spontaneous immobility than male rats (203.4 [61.6] min vs 101.3 [32.1] min), and their EEG was suppressed in the low-frequency range (2-6 Hz), in contrast to male rats. Although a sex-dependent hypnotic effect was not confirmed after 30 mg kg-1 i.v., female rats appeared more sensitive to 3ß-OH with relatively small changes within delta (1-4 Hz) and alpha (8-13 Hz) bands. Finally, 3ß-OH had a rapid onset of action and potent hypnotic/anaesthetic effect after 60 mg kg-1 i.v. in rats of both sexes; however, all female rats and only half of the male rats reached burst suppression, an EEG pattern usually associated with profound inhibition of thalamocortical networks. CONCLUSIONS: Based on its behavioural effects and EEG signature, 3ß-OH is a potent hypnotic in rats, with female rats being more sensitive than male rats.


Asunto(s)
Androstanoles/farmacología , Ondas Encefálicas/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Electrocorticografía , Pérdida de Tono Postural/efectos de los fármacos , Neuroesteroides/farmacología , Nitrilos/farmacología , Androstanoles/administración & dosificación , Animales , Corteza Cerebral/fisiopatología , Relación Dosis-Respuesta a Droga , Femenino , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Masculino , Neuroesteroides/administración & dosificación , Nitrilos/administración & dosificación , Ratas Sprague-Dawley , Factores Sexuales , Factores de Tiempo
12.
J Zoo Wildl Med ; 51(4): 841-847, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33480564

RESUMEN

Veterinary care of ferrets often requires chemical restraint. This study hypothesized that IM alfaxalone and butorphanol would result in clinically useful sedation without clinically relevant cardiorespiratory effects. Twelve healthy 15-mo-old ferrets of equal sexes weighing 0.75 to 1.66 kg were enrolled. Using a prospective, blinded design, ferrets randomly received either IM alfaxalone 2.5 mg/kg and butorphanol 0.2 mg/kg (low dose [LD]) or IM alfaxalone 5 mg/kg and butorphanol 0.2 mg/kg (high dose [HD]) (n = 6/group). Sedation times and induction and recovery scores were recorded by a blinded observer. Anesthetic monitor placement was attempted in all recumbent ferrets, and physiologic parameters and reflexes were recorded every 5 min until return of spontaneous movement. Data were assessed for normality using a Shapiro-Wilk normality test and analyzed by two-sample t test or Mann-Whitney U test; one ferret in HD was excluded. Ferrets in LD and HD exhibited moderate and marked sedation, with one of six and four of five ferrets tolerating monitor placement, respectively. Mean ± SD time to first effects, recumbency, and recovery in LD and HD was 2.30 ± 1.13 and 2.054 ± 1.12 (P = 0.7240), 2.87 ± 1.25 and 2.72 ± 1.41 (P = 0.8529), and 65.43 ± 32.43 and 52.30 ± 13.19 (P = 0.4212), respectively. Median (range) duration of recumbency in LD and HD was 31.12 (25.58-115.72) and 35.47 (28.27-44.42) min (P = 0.3290), respectively. Among monitored ferrets, transient mild hypotension and hypoxemia were observed. Intramuscular alfaxalone 5 mg/kg with butorphanol 0.2 mg/kg provided clinically useful sedation in ferrets with mild transient cardiorespiratory derangements.


Asunto(s)
Analgésicos Opioides/farmacología , Butorfanol/farmacología , Hurones , Neuroesteroides/farmacología , Pregnanodionas/farmacología , Analgésicos Opioides/administración & dosificación , Animales , Butorfanol/administración & dosificación , Sedación Consciente/veterinaria , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Femenino , Masculino , Neuroesteroides/administración & dosificación , Pregnanodionas/administración & dosificación
13.
Mol Pharmacol ; 98(4): 280-291, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32675382

RESUMEN

The ρ1 GABAA receptor is prominently expressed in the retina and is present at lower levels in several brain regions and other tissues. Although the ρ1 receptor is insensitive to many anesthetic drugs that modulate the heteromeric GABAA receptor, it maintains a rich and multifaceted steroid pharmacology. The receptor is negatively modulated by 5ß-reduced steroids, sulfated or carboxylated steroids, and ß-estradiol, whereas many 5α-reduced steroids potentiate the receptor. In this study, we analyzed modulation of the human ρ1 GABAA receptor by several neurosteroids, individually and in combination, in the framework of the coagonist concerted transition model. Experiments involving coapplication of two or more steroids revealed that the receptor contains at least three classes of distinct, nonoverlapping sites for steroids, one each for the inhibitory steroids pregnanolone (3α5ßP), 3α5ßP sulfate, and ß-estradiol. The site for 3α5ßP can accommodate the potentiating steroid 5αTHDOC. The findings are discussed with respect to receptor modulation by combinations of endogenous neurosteroids. SIGNIFICANCE STATEMENT: The study describes modulation of the ρ1 GABAA receptor by neurosteroids. The coagonist concerted transition model was used to determine overlap of binding sites for several inhibitory and potentiating steroids.


Asunto(s)
Desoxicorticosterona/análogos & derivados , Neuroesteroides/farmacología , Pregnanolona/farmacología , Receptores de GABA-A/química , Receptores de GABA-A/metabolismo , Xenopus laevis/genética , Animales , Animales Modificados Genéticamente , Sitios de Unión , Desoxicorticosterona/química , Desoxicorticosterona/farmacología , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Modelos Moleculares , Estructura Molecular , Neuroesteroides/química , Pregnanolona/química , Receptores de GABA-A/genética
14.
J Biol Chem ; 294(32): 12220-12230, 2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31239352

RESUMEN

Neuroactive steroids (NASs) are synthesized within the brain and exert profound effects on behavior. These effects are primarily believed to arise from the activities of NASs as positive allosteric modulators (PAMs) of the GABA-type A receptor (GABAAR). NASs also activate a family of G protein-coupled receptors known as membrane progesterone receptors (mPRs). Here, using surface-biotinylation assays and electrophysiology techniques, we examined mPRs' role in mediating the effects of NAS on the efficacy of GABAergic inhibition. Selective mPR activation enhanced phosphorylation of Ser-408 and Ser-409 (Ser-408/9) within the GABAAR ß3 subunit, which depended on the activity of cAMP-dependent protein kinase A (PKA) and protein kinase C (PKC). mPR activation did not directly modify GABAAR activity and had no acute effects on phasic or tonic inhibition. Instead, mPR activation induced a sustained elevation in tonic current, which was blocked by PKA and PKC inhibition. Substitution of Ser-408/9 to alanine residues also prevented the effects of mPR activation on tonic current. Furthermore, this substitution abolished the effects of sustained NAS exposure on tonic inhibition. Interestingly, the allosteric effects of NAS on GABAergic inhibition were independent of Ser-408/9 in the ß3 subunit. Additionally, although allosteric effects of NAS on GABAergic inhibition were sensitive to a recently developed "NAS antagonist," the sustained effects of NAS on tonic inhibition were not. We conclude that metabotropic effects of NAS on GABAergic inhibition are mediated by mPR-dependent modulation of GABAAR phosphorylation. We propose that this mechanism may contribute to the varying behavioral effects of NAS.


Asunto(s)
Neuroesteroides/metabolismo , Receptores de GABA-A/metabolismo , Regulación Alostérica/efectos de los fármacos , Animales , Membrana Celular/metabolismo , Potenciales Evocados/efectos de los fármacos , Antagonistas de Receptores de GABA-A/farmacología , Células HEK293 , Hipocampo/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutagénesis Sitio-Dirigida , Neuroesteroides/farmacología , Fosforilación/efectos de los fármacos , Proteína Quinasa C/metabolismo , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Receptores de GABA-A/química , Receptores de GABA-A/genética , Receptores de Progesterona/agonistas , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
15.
Front Neuroendocrinol ; 55: 100795, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31562849

RESUMEN

Complex mechanisms involved in neuropathic pain that represents a major health concern make its management complicated. Because neurosteroids are bioactive steroids endogenously synthesized in the nervous system, including in pain pathways, they appear relevant to develop effective treatments against neuropathic pain. Neurosteroids act in paracrine or autocrine manner through genomic mechanisms and/or via membrane receptors of neurotransmitters that pivotally modulate pain sensation. Basic studies which uncovered a direct link between neuropathic pain symptoms and endogenous neurosteroid production/regulation, paved the way for the investigations of neurosteroid therapeutic potential against pathological pain. Concordantly, antinociceptive properties of synthetic neurosteroids were evidenced in humans and animals. Neurosteroids promote peripheral analgesia mediated by T-type calcium and gamma-aminobutyric acid type A channels, counteract chemotherapy-induced neuropathic pain and ameliorate neuropathic symptoms of injured spinal cord animals by stimulating anti-inflammatory, remyelinating and neuroprotective processes. Together, these data open interesting perspectives for neurosteroid-based strategies to manage/alleviate efficiently neuropathic pain.


Asunto(s)
Dolor Crónico/metabolismo , Neuralgia/metabolismo , Neuroprotección/fisiología , Neuroesteroides/metabolismo , Nocicepción/fisiología , Manejo del Dolor , Dolor Crónico/tratamiento farmacológico , Humanos , Neuralgia/tratamiento farmacológico , Neuroesteroides/farmacología , Nocicepción/efectos de los fármacos
16.
Int J Mol Sci ; 21(24)2020 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-33321734

RESUMEN

Neurosteroids are a family of compounds that are synthesized in principal excitatory neurons and glial cells, and derive from the transformation of cholesterol into pregnenolone. The most studied neurosteroids-allopregnanolone and allotetrahydrodeoxycorticosterone (THDOC)-are known to modulate GABAA receptor-mediated transmission, thus playing a role in controlling neuronal network excitability. Given the role of GABAA signaling in epileptic disorders, neurosteroids have profound effects on seizure generation and play a role in the development of chronic epileptic conditions (i.e., epileptogenesis). We review here studies showing the effects induced by neurosteroids on epileptiform synchronization in in vitro brain slices, on epileptic activity in in vivo models, i.e., in animals that were made epileptic with chemoconvulsant treatment, and in epileptic patients. These studies reveal that neurosteroids can modulate ictogenesis and the occurrence of pathological network activity such as interictal spikes and high-frequency oscillations (80-500 Hz). Moreover, they can delay the onset of spontaneous seizures in animal models of mesial temporal lobe epilepsy. Overall, this evidence suggests that neurosteroids represent a new target for the treatment of focal epileptic disorders.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Epilepsia del Lóbulo Temporal/fisiopatología , Neuroesteroides/uso terapéutico , Animales , Anticonvulsivantes/farmacología , Ondas Encefálicas/efectos de los fármacos , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Epilepsia del Lóbulo Temporal/metabolismo , Humanos , Neuroesteroides/farmacología , Transmisión Sináptica/efectos de los fármacos
17.
Epilepsia ; 60(4): 730-743, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30895610

RESUMEN

OBJECTIVE: Sex differences are evident in the antiseizure activity of neurosteroids; however, the potential mechanisms remain unclear. In this study, we sought to determine whether differences in target extrasynaptic δ-subunit γ-aminobutyric acid type A (GABA-A) receptor expression and function underlie the sex differences in seizure susceptibility and the antiseizure activity of neurosteroids. METHODS: Sex differences in seizure susceptibility and protective activity of three distinct neurosteroids-allopregnanolone (AP), androstanediol (AD), and ganaxolone-were evaluated in the pilocarpine model of status epilepticus (SE) and kindling seizure test in mice. Immunocytochemistry was used for δGABA-A receptor expression analysis, and patch-clamp recordings in brain slices evaluated its functional currents. RESULTS: Sex differences were apparent in kindling epileptogenic seizures, with males exhibiting a faster progression to a fully kindled state. Neurosteroids AP, AD, or ganaxolone produced dose-dependent protection against SE and acute partial seizures. However, female mice exhibited strikingly enhanced sensitivity to the antiseizure activity of neurosteroids compared to males. Sex differences in neurosteroid protection were unrelated to pharmacokinetic factors, as plasma levels of neurosteroids associated with seizure protection were similar between sexes. Mice lacking extrasynaptic δGABA-A receptors did not exhibit sex differences in neurosteroid protection. Consistent with a greater abundance of extrasynaptic δGABA-A receptors, AP produced a significantly greater potentiation of tonic currents in dentate gyrus granule cells in females than males; however, such enhanced AP sensitivity was diminished in δGABA-A receptor knockout female mice. SIGNIFICANCE: Neurosteroids exhibit greater antiseizure potency in females than males, likely due to a greater abundance of extrasynaptic δGABA-A receptors that mediate neurosteroid-sensitive tonic currents and seizure protection. These findings indicate the potential to develop personalized gender-specific neurosteroid treatments for SE and epilepsy in men and women, including catamenial epilepsy.


Asunto(s)
Neuroesteroides/farmacología , Receptores de GABA-A/metabolismo , Convulsiones/metabolismo , Caracteres Sexuales , Estado Epiléptico/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Pregnanolona/análogos & derivados , Pregnanolona/farmacología
18.
Pediatr Res ; 85(1): 86-96, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30237570

RESUMEN

BACKGROUND: Children born preterm, especially boys, are at increased risk of developing attention deficit hyperactivity disorder (ADHD) and learning difficulties. We propose that neurosteroid-replacement therapy with ganaxolone (GNX) following preterm birth may mitigate preterm-associated neurodevelopmental impairment. METHODS: Time-mated sows were delivered preterm (d62) or at term (d69). Male preterm pups were randomized to ganaxolone (Prem-GNX; 2.5 mg/kg subcutaneously twice daily until term equivalence), or preterm control (Prem-CON). Surviving male juvenile pups underwent behavioural testing at d25-corrected postnatal age (CPNA). Brain tissue was collected at CPNA28 and mature myelinating oligodendrocytes of the hippocampus and subcortical white matter were quantified by immunostaining of myelin basic protein (MBP). RESULTS: Ganaxolone treatment returned the hyperactive behavioural phenotype of preterm-born juvenile males to a term-born phenotype. Deficits in MBP immunostaining of the preterm hippocampus and subcortical white matter were also ameliorated in animals receiving ganaxolone. However, during the treatment period weight gain was poor, and pups were sedated, ultimately increasing the neonatal mortality rate. CONCLUSION: Ganaxolone improved neurobehavioural outcomes in males suggesting that neonatal treatment may be an option for reducing preterm-associated neurodevelopmental impairment. However, dosing studies are required to reduce the burden of unwanted side effects.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/prevención & control , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Moduladores del GABA/farmacología , Locomoción/efectos de los fármacos , Neuroesteroides/farmacología , Pregnanolona/análogos & derivados , Nacimiento Prematuro , Animales , Animales Recién Nacidos , Trastorno por Déficit de Atención con Hiperactividad/metabolismo , Trastorno por Déficit de Atención con Hiperactividad/fisiopatología , Trastorno por Déficit de Atención con Hiperactividad/psicología , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Moduladores del GABA/toxicidad , Cobayas , Masculino , Proteína Básica de Mielina/metabolismo , Neuroesteroides/toxicidad , Pregnanolona/farmacología , Pregnanolona/toxicidad , Prueba de Estudio Conceptual , Receptores de GABA-A/efectos de los fármacos , Receptores de GABA-A/metabolismo , Conducta Social
19.
Gen Comp Endocrinol ; 284: 113051, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30339808

RESUMEN

The brain has traditionally been considered to be a target site of peripheral steroid hormones. On the other hand, extensive studies over the past thirty years have demonstrated that the brain is a site of biosynthesis of several steroids. Such steroids synthesized de novo from cholesterol in the brain are called neurosteroids. To investigate the biosynthesis and biological actions of neurosteroids in the brain, data on the regio- and temporal-specific synthesis of neurosteroids are needed. In the mid 1990s, the Purkinje cell, an important cerebellar neuron, was discovered as a major cell producing neurosteroids in the brain of vertebrates. It was the first demonstration of de novo neuronal biosynthesis of neurosteroids in the brain. Subsequently, neuronal biosynthesis of neurosteroids and biological actions of neurosteroids have become clear by the follow-up studies using the Purkinje cell as an excellent cellular model. Progesterone and estradiol, which are known as sex steroid hormones, are actively synthesized de novo from cholesterol in the Purkinje cell during development, when cerebellar neuronal circuit formation occurs. Importantly, progesterone and estradiol synthesized in the Purkinje cell promote dendritic growth, spinogenesis and synaptogenesis via their cognate nuclear receptors in the Purkinje cell. Neurotrophic factors may mediate these neurosteroid actions. Futhermore, allopregnanolone (3α,5α-tetrahydroprogesterone), a progesterone metabolite, is also synthesized in the cerebellum and acts on the survival of Purkinje cells. On the other hand, at the beginning of 2010s, the pineal gland, an endocrine organ located close to the cerebellum, was discovered as an important site of the biosynthesis of neurosteroids. Allopregnanolone, a major pineal neurosteroid, acts on the Purkinje cell for the survival of Purkinje cells by suppressing the expression of caspase-3, a crucial mediator of apoptosis. I as a recipient of Kobayashi Award from the Japan Society for Comparative Endocrinology in 2016 summarize the discovery of cerebellar and pineal neurosteroids and their biological actions on the growth and survival of Purkinje cells during development.


Asunto(s)
Distinciones y Premios , Cerebelo/metabolismo , Neuroesteroides/farmacología , Glándula Pineal/metabolismo , Células de Purkinje/citología , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células de Purkinje/efectos de los fármacos , Células de Purkinje/metabolismo
20.
Neurosci Biobehav Rev ; 152: 105327, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37499891

RESUMEN

Affective disorders such as depression and anxiety are among the most prevalent psychiatric illnesses and causes of disability worldwide. The recent FDA-approval of a novel antidepressant treatment, ZULRESSO® (Brexanolone), a synthetic neurosteroid has fueled interest into the role of neurosteroids in the pathophysiology of depression as well as the mechanisms mediating the antidepressant effects of these compounds. The majority of studies examining the impact of neurosteroids on affective states have relied on the administration of exogenous neurosteroids; however, neurosteroids can also be synthesized endogenously from cholesterol or steroid hormone precursors. Despite the well-established influence of exogenous neurosteroids on affective states, we still lack an understanding of the role of endogenous neurosteroids in modulating affective tone. This review aims to summarize the current literature supporting the influence of neurosteroids on affective states in clinical and preclinical studies, as well as recent evidence suggesting that endogenous neurosteroids may set a baseline affective tone.


Asunto(s)
Neuroesteroides , Humanos , Neuroesteroides/farmacología , Neuroesteroides/uso terapéutico , Antidepresivos/uso terapéutico , Ansiedad , Trastornos del Humor/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA