Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Br J Anaesth ; 130(2): 154-164, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36428160

RESUMEN

BACKGROUND: The novel synthetic neuroactive steroid (3ß,5ß,17ß)-3-hydroxyandrostane-17-carbonitrile (3ß-OH) blocks T-type calcium channels but does not directly modulate neuronal γ-aminobutyric acid type A (GABAA) currents like other anaesthetic neurosteroids. As 3ß-OH has sex-specific hypnotic effects in adult rats, we studied the mechanism contributing to sex differences in its effects. METHODS: We used a combination of behavioural loss of righting reflex, neuroendocrine, pharmacokinetic, in vitro patch-clamp electrophysiology, and in vivo electrophysiological approaches in wild-type mice and in genetic knockouts of the CaV3.1 T-type calcium channel isoform to study the mechanisms by which 3ß-OH and its metabolite produces sex-specific hypnotic effects. RESULTS: Adult male mice were less sensitive to the hypnotic effects of 3ß-OH compared with female mice, and these differences appeared during development. Adult males had higher 3ß-OH brain concentrations despite being less sensitive to its hypnotic effects. Females metabolised 3ß-OH into the active GABAA receptor positive allosteric modulator (3α,5ß,17ß)-3-hydroxyandrostane-17-carbonitrile (3α-OH) to a greater extent than males. The 3α-OH metabolite has T-channel blocking properties with sex-specific hypnotic and pharmacokinetic effects. Sex-dependent suppression of the cortical electroencephalogram is more pronounced with 3α-OH compared with 3ß-OH. CONCLUSIONS: The sex-specific differences in the hypnotic effect of 3ß-OH in mice are attributable to differences in its peripheral metabolism into the more potent hypnotic metabolite 3α-OH.


Asunto(s)
Canales de Calcio Tipo T , Neuroesteroides , Ratas , Ratones , Femenino , Masculino , Animales , Hipnóticos y Sedantes/farmacología , Esteroides/farmacología , Receptores de GABA-A
2.
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
3.
Neurobiol Dis ; 127: 554-562, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30951850

RESUMEN

Apoptosis is triggered in the developing mammalian brain by sedative, anesthetic or antiepileptic drugs during late gestation and early life. Whether human children are vulnerable to this toxicity mechanism remains unknown, as there are no imaging techniques to capture it. Apoptosis is characterized by distinct structural features, which affect the way damaged tissue scatters ultrasound compared to healthy tissue. We evaluated whether apoptosis, triggered by the anesthetic sevoflurane in the brains of neonatal rhesus macaques, can be detected using quantitative ultrasound (QUS). Neonatal (n = 15) rhesus macaques underwent 5 h of sevoflurane anesthesia. QUS images were obtained through the sagittal suture at 0.5 and 6 h. Brains were collected at 8 h and examined immunohistochemically to analyze apoptotic neuronal and oligodendroglial death. Significant apoptosis was detected in white and gray matter throughout the brain, including the thalamus. We measured a change in the effective scatterer size (ESS), a QUS biomarker derived from ultrasound echo signals obtained with clinical scanners, after sevoflurane-anesthesia in the thalamus. Although initial inclusion of all measurements did not reveal a significant correlation, when outliers were excluded, the change in the ESS between the pre- and post-anesthesia measurements correlated strongly and proportionally with the severity of apoptotic death. We report for the first time in vivo changes in QUS parameters, which may reflect severity of apoptosis in the brains of infant nonhuman primates. These findings suggest that QUS may enable in vivo studies of apoptosis in the brains of human infants following exposure to anesthetics, antiepileptics and other brain injury mechanisms.


Asunto(s)
Apoptosis/fisiología , Encéfalo/diagnóstico por imagen , Sevoflurano/farmacología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Femenino , Macaca mulatta , Masculino , Neuronas/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Ultrasonografía
4.
Horm Behav ; 90: 84-89, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28257758

RESUMEN

Women and non-human females have surprisingly high levels of circulating testosterone, yet the effects of androgens on non-reproductive behaviors, including cognition, of females are not well characterized. The current project used an aromatase inhibitor, letrozole, to block conversion of androgens to estrogens. Adult female rats were ovariectomized and administered either vehicle only, testosterone propionate only (400µg/kg, TP only), letrozole only (1mg/kg, Letro only), or the combination of letrozole and testosterone (TP+Letro) over 4weeks. A gonadally intact group was used for comparisons. During the last 3weeks, the animals were tested for working memory in both a spatial task (radial arm maze) and a non-spatial task (object recognition). At sacrifice, uterine weights and serum testosterone and estradiol were determined. Behavioral results were the intact animals showed better working memories on the object recognition task, but that there were no differences among the ovariectomized groups. In the radial arm maze task, groups with best to worst performance were TP only>Intact=TP+Letro>vehicle=Letro only. Highest to lowest serum titers, for testosterone, were TP+Letro>TP only>Intact=Letro only>vehicle and, for estradiol, Intact>TP only>Vehicle>Letro only=TP+Letro. Our interpretation is that testosterone enhanced spatial performance when bioavailability of both TP and E2 are high, and high testosterone can rescue spatial memory when E2 bioavailability is low.


Asunto(s)
Andrógenos/metabolismo , Inhibidores de la Aromatasa/farmacología , Cognición/efectos de los fármacos , Estrógenos/metabolismo , Animales , Estradiol/sangre , Femenino , Letrozol , Aprendizaje por Laberinto/efectos de los fármacos , Nitrilos/farmacología , Ratas , Ratas Long-Evans , Testosterona/sangre , Triazoles/farmacología , Útero/anatomía & histología , Útero/efectos de los fármacos
5.
Exp Biol Med (Maywood) ; 248(7): 578-587, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37309730

RESUMEN

Preclinical studies have established that neonatal exposure to contemporary sedative/hypnotic drugs causes neurotoxicity in the developing rodent and primate brains. Our group recently reported that novel neuroactive steroid (3ß,5ß,17ß)-3-hydroxyandrostane-17-carbonitrile (3ß-OH) induced effective hypnosis in both neonatal and adult rodents but did not cause significant neurotoxicity in vulnerable brain regions such as subiculum, an output region of hippocampal formation particularly sensitive to commonly used sedatives/hypnotics. Despite significant emphasis on patho-morphological changes, little is known about long-term effects on subicular neurophysiology after neonatal exposure to neuroactive steroids. Hence, we explored the lasting effects of neonatal exposure to 3ß-OH on sleep macrostructure as well as subicular neuronal oscillations in vivo and synaptic plasticity ex vivo in adolescent rats. At postnatal day 7, we exposed rat pups to either 10 mg/kg of 3ß-OH over a period of 12 h or to volume-matched cyclodextrin vehicle. At weaning age, a cohort of rats was implanted with a cortical electroencephalogram (EEG) and subicular depth electrodes. At postnatal day 30-33, we performed in vivo assessment of sleep macrostructure (divided into wake, non-rapid eye movement, and rapid eye movement sleep) and power spectra in cortex and subiculum. In a second cohort of 3ß-OH exposed animals, we conducted ex vivo studies of long-term potentiation (LTP) in adolescent rats. Overall, we found that neonatal exposure to 3ß-OH decreased subicular delta and sigma oscillations during non-rapid eye movement sleep without altering sleep macrostructure. Furthermore, we observed no significant changes in subicular synaptic plasticity. Interestingly, our previous study found that neonatal exposure to ketamine increased subicular gamma oscillations during non-rapid eye movement sleep and profoundly suppressed subicular LTP in adolescent rats. Together these results suggest that exposure to different sedative/hypnotic agents during a critical period of brain development may induce distinct functional changes in subiculum circuitry that may persist into adolescent age.


Asunto(s)
Neuroesteroides , Ratas , Animales , Neuroesteroides/farmacología , Ratas Sprague-Dawley , Hipocampo , Plasticidad Neuronal , Hipnóticos y Sedantes/farmacología
6.
J Neuroendocrinol ; 34(2): e13086, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35014105

RESUMEN

Since the 1990s, there has been waning interest in researching general anaesthetics (anaesthetics). Although currently used anaesthetics are mostly safe and effective, they are not without fault. In paediatric populations and neonatal animal models, they are associated with learning impairments and neurotoxicity. In an effort to research safer anaesthetics, we have gone back to re-examine neuroactive steroids as anaesthetics. Neuroactive steroids are steroids that have direct, local effects in the central nervous system. Since the discovery of their anaesthetic effects, neuroactive steroids have been consistently used in human or veterinary clinics as preferred anaesthetic agents. Although briefly abandoned for clinical use due to unwanted vehicle side effects, there has since been renewed interest in their therapeutic value. Neuroactive steroids are safe sedative/hypnotic and anaesthetic agents across various animal species. Importantly, unlike traditional anaesthetics, they do not cause extensive neurotoxicity in the developing rodent brain. Similar to traditional anaesthetics, neuroactive steroids are modulators of synaptic and extrasynaptic γ-aminobutyric acid type A (GABAA ) receptors and their interactions at the GABAA receptor are stereo- and enantioselective. Recent work has also shown that these agents act on other ion channels, such as high- and low-voltage-activated calcium channels. Through these mechanisms of action, neuroactive steroids modulate neuronal excitability, which results in characteristic burst suppression of the electroencephalogram, and a surgical plane of anaesthesia. However, in addition to their interactions with voltage and ligand gated ions channels, neuroactive steroids interact with membrane bound metabotropic receptors and xenobiotic receptors to facilitate signaling of prosurvival, antiapoptotic pathways. These pathways play a role in their neuroprotective effects in neuronal injury and may also prevent extensive apoptosis in the developing brain during anaesthesia. The current review explores the history of neuroactive steroids as anaesthetics in humans and animal models, their diverse mechanisms of action, and their neuroprotective properties.


Asunto(s)
Anestésicos , Neuroesteroides , Anestésicos/farmacología , Animales , Humanos , Hipnóticos y Sedantes/farmacología , Receptores de GABA-A , Esteroides
7.
Front Pharmacol ; 13: 850658, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35677453

RESUMEN

We recently reported that a neurosteroid analogue with T-channel-blocking properties (3ß,5ß,17ß)-3-hydroxyandrostane-17-carbonitrile (3ß-OH), induced hypnosis in rat pups without triggering neuronal apoptosis. Furthermore, we found that the inhibition of the CaV3.1 isoform of T-channels contributes to the hypnotic properties of 3ß-OH in adult mice. However, the specific mechanisms underlying the role of other subtypes of voltage-gated calcium channels in thalamocortical excitability and oscillations in vivo during 3ß-OH-induced hypnosis are largely unknown. Here, we used patch-clamp recordings from acute brain slices, in vivo electroencephalogram (EEG) recordings, and mouse genetics with wild-type (WT) and CaV2.3 knock-out (KO) mice to further investigate the molecular mechanisms of neurosteroid-induced hypnosis. Our voltage-clamp recordings showed that 3ß-OH inhibited recombinant CaV2.3 currents. In subsequent current-clamp recordings in thalamic slices ex vivo, we found that selective CaV2.3 channel blocker (SNX-482) inhibited stimulated tonic firing and increased the threshold for rebound burst firing in WT animals. Additionally, in thalamic slices we found that 3ß-OH inhibited spike-firing more profoundly in WT than in mutant mice. Furthermore, 3ß-OH reduced bursting frequencies in WT but not mutant animals. In ensuing in vivo experiments, we found that intra-peritoneal injections of 3ß-OH were less effective in inducing LORR in the mutant mice than in the WT mice, with expected sex differences. Furthermore, the reduction in total α, ß, and low γ EEG power was more profound in WT than in CaV2.3 KO females over time, while at 60 min after injections of 3ß-OH, the increase in relative ß power was higher in mutant females. In addition, 3ß-OH depressed EEG power more strongly in the male WT than in the mutant mice and significantly increased the relative δ power oscillations in WT male mice in comparison to the mutant male animals. Our results demonstrate for the first time the importance of the CaV2.3 subtype of voltage-gated calcium channels in thalamocortical excitability and the oscillations that underlie neurosteroid-induced hypnosis.

8.
Front Behav Neurosci ; 15: 703859, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34790103

RESUMEN

General anesthetics are neurotoxic to the developing rodent and primate brains leading to neurocognitive and socio-affective impairment later in life. In addition, sleep patterns are important predictors of cognitive outcomes. Yet, little is known about how anesthetics affect sleep-wake behaviors and their corresponding oscillations. Here we examine how neonatal general anesthesia affects sleep and wake behavior and associated neuronal oscillations. We exposed male and female rat pups to either 6 h of continuous isoflurane or sham anesthesia (compressed air) at the peak of their brain development (postnatal day 7). One cohort of animals was used to examine neurotoxic insult 2 h post-anesthesia exposure. At weaning age, a second cohort of rats was implanted with cortical electroencephalogram electrodes and allowed to recover. During adolescence, we measured sleep architecture (divided into wake, non-rapid eye movement, and rapid eye movement sleep) and electroencephalogram power spectra over a 24 h period. We found that exposure to neonatal isoflurane caused extensive neurotoxicity but did not disrupt sleep architecture in adolescent rats. However, these animals had a small but significant reduction in beta oscillations, specifically in the 12-20 Hz beta 1 range, associated with wake behavior. Furthermore, beta oscillations play a critical role in cortical development, cognitive processing, and homeostatic sleep drive. We speculate that dysregulation of beta oscillations may be implicated in cognitive and socio-affective outcomes associated with neonatal anesthesia.

9.
Neurosci Lett ; 753: 135854, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33785378

RESUMEN

Physical inactivity is positively associated with anxiety and depression. Considering physical inactivity, anxiety, and depression each have a genetic basis for inheritance, our lab used artificial selectively bred low-voluntary running (LVR) and wild type (WT) female Wistar rats to test if physical inactivity genes selected over multiple generations would lead to an anxiety or depressive-like phenotype. We performed next generation RNA sequencing and immunoblotting on the dentate gyrus to reveal key biological functions from heritable physical inactivity. LVR rats did not display depressive-like behavior. However, LVR rats did display anxiogenic behavior with gene networks associated with reduced neuronal development, proliferation, and function compared to WT counterparts. Additionally, immunoblotting revealed LVR deficits in neuronal development and function. To our knowledge, this is the first study to show that by selectively breeding for physical inactivity genes, anxiety-like genes were co-selected. The study also reveals molecular insights to the genetic influences that physical inactivity has on anxiety-like behavior.


Asunto(s)
Ansiedad/genética , Conducta Sedentaria , Selección Artificial/genética , Animales , Ansiedad/patología , Ansiedad/fisiopatología , Giro Dentado , Depresión/genética , Depresión/patología , Depresión/fisiopatología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , RNA-Seq , Ratas , Ratas Wistar , Carrera/fisiología
10.
Br J Pharmacol ; 177(8): 1735-1753, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31732978

RESUMEN

BACKGROUND AND PURPOSE: Neuroactive steroid (3ß,5ß,17ß)-3-hydroxyandrostane-17-carbonitrile (3ß-OH) is a novel hypnotic and voltage-dependent blocker of T-type calcium channels. Here, we examine its potential analgesic effects and adjuvant anaesthetic properties using a post-surgical pain model in rodents. EXPERIMENTAL APPROACH: Analgesic properties of 3ß-OH were investigated in thermal and mechanical nociceptive tests in sham or surgically incised rats and mice, with drug injected either systemically (intraperitoneal) or locally via intrathecal or intraplantar routes. Hypnotic properties of 3ß-OH and its use as an adjuvant anaesthetic in combination with isoflurane were investigated using behavioural experiments and in vivo EEG recordings in adolescent rats. KEY RESULTS: A combination of 1% isoflurane with 3ß-OH (60 mg·kg-1 , i.p.) induced suppression of cortical EEG and stronger thermal and mechanical anti-hyperalgesia during 3 days post-surgery, when compared to isoflurane alone and isoflurane with morphine. 3ß-OH exerted prominent enantioselective thermal and mechanical antinociception in healthy rats and reduced T-channel-dependent excitability of primary sensory neurons. Intrathecal injection of 3ß-OH alleviated mechanical hyperalgesia, while repeated intraplantar application alleviated both thermal and mechanical hyperalgesia in the rats after incision. Using mouse genetics, we found that CaV 3.2 T-calcium channels are important for anti-hyperalgesic effect of 3ß-OH and are contributing to its hypnotic effect. CONCLUSION AND IMPLICATIONS: Our study identifies 3ß-OH as a novel analgesic for surgical procedures. 3ß-OH can be used to reduce T-channel-dependent excitability of peripheral sensory neurons as an adjuvant for induction and maintenance of general anaesthesia while improving analgesia and lowering the amount of volatile anaesthetic needed for surgery.


Asunto(s)
Analgesia , Canales de Calcio Tipo T , Neuroesteroides , Animales , Hiperalgesia/tratamiento farmacológico , Hipnóticos y Sedantes , Ratones , Dolor Postoperatorio/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley , Roedores
11.
Front Syst Neurosci ; 14: 26, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32528257

RESUMEN

Exposure to sedative/hypnotic and anesthetic drugs, such as ketamine, during the critical period of synaptogenesis, causes profound neurotoxicity in the developing rodent and primate brains and is associated with poor cognitive outcomes later in life. The subiculum is especially vulnerable to acute neurotoxicity after neonatal exposure to sedative/hypnotic and anesthetic drugs. The subiculum acts as a relay center between the hippocampal complex and various cortical and subcortical brain regions and is also an independent generator of gamma oscillations. Gamma oscillations are vital in neuronal synchronization and play a role in learning and memory during wake and sleep. However, there has been little research examining long-term changes in subicular neurophysiology after neonatal exposure to ketamine. Here we explore the lasting effects of neonatal ketamine exposure on sleep macrostructure as well as subicular neuronal oscillations and synaptic plasticity in rats. During the peak of rodent synaptogenesis at postnatal day 7, rat pups were exposed to either 40 mg/kg of ketamine over 12 h or to volume matched saline vehicle. At weaning age, a subset of rats were implanted with a cortical and subicular electroencephalogram electrode, and at postnatal day 31, we performed in vivo experiments that included sleep macrostructure (divided into the wake, non-rapid eye movement, and rapid eye movement sleep) and electroencephalogram power spectra in cortex and subiculum. In a second subset of ketamine exposed animals, we conducted ex vivo studies of long-term potentiation (LTP) experiments in adolescent rats. Overall, we found that neonatal exposure to ketamine increased subicular gamma oscillations during non-rapid eye movement sleep but it did not alter sleep macrostructure. Also, we observed a significant decrease in subicular LTP. Gamma oscillations during non-rapid eye movement sleep are implicated in memory formation and consolidation, while LTP serves as a surrogate for learning and memory. Together these results suggest that lasting functional changes in subiculum circuitry may underlie neurocognitive impairments associated with neonatal exposure to anesthetic agents.

12.
Sci Rep ; 8(1): 5302, 2018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29593226

RESUMEN

Caffeine is the most frequently used medication in premature infants. It is the respiratory stimulant of choice for apnea associated with prematurity and has been called the silver bullet in neonatology because of many proven benefits and few known risks. Research has revealed that sedative/anesthetic drugs trigger apoptotic death of neurons and oligodendrocytes in developing mammalian brains. Here we evaluated the influence of caffeine on the neurotoxicity of anesthesia in developing nonhuman primate brains. Fetal macaques (n = 7-8/group), at a neurodevelopmental age comparable to premature human infants, were exposed in utero for 5 hours to no drug (control), isoflurane, or isoflurane + caffeine and examined for evidence of apoptosis. Isoflurane exposure increased apoptosis 3.3 fold for neurons and 3.4 fold for oligodendrocytes compared to control brains. Isoflurane + caffeine caused neuronal apoptosis to increase 8.0 fold compared to control levels but did not augment oligoapoptosis. Neuronal death was particularly pronounced in the basal ganglia and cerebellum. Higher blood levels of caffeine within the range considered therapeutic and safe for human infants correlated with increased neuroapoptosis. Caffeine markedly augments neurotoxicity of isoflurane in the fetal macaque brain and challenges the assumption that caffeine is safe for premature infants.


Asunto(s)
Cafeína/efectos adversos , Desarrollo Fetal/efectos de los fármacos , Isoflurano/efectos adversos , Anestesia/efectos adversos , Anestésicos por Inhalación/efectos adversos , Anestésicos por Inhalación/farmacología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Cafeína/farmacología , Muerte Celular/efectos de los fármacos , Femenino , Isoflurano/farmacología , Macaca mulatta/embriología , Masculino , Neuronas/fisiología , Oligodendroglía/efectos de los fármacos , Embarazo
13.
J Matern Fetal Neonatal Med ; 30(22): 2734-2741, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27924651

RESUMEN

OBJECTIVES: Caffeine (CAF) and sedative/anesthetic drugs (SADs) are often coadministered to premature infants in the neonatal intensive care unit (NICU). While SAD neurotoxicity in the developing brain is well established, it is not fully clear whether CAF interacts with SADs and whether this interaction is detrimental. Using a mouse model of prematurity, we hypothesized that CAF would increase apoptotic neurotoxicity when coadministered with SADs. METHODS: Postnatal day 3 mice were treated with vehicle or 80 mg/kg CAF prior to challenge with 6 mg/kg midazolam, 40 mg/kg ketamine, or 40 µg/kg fentanyl. Six hours later, pups were sacrificed for activated caspase 3 (AC3) immunohistochemistry, and number of AC3 positive cells per mm3 throughout neocortex, hippocampus, caudate, thalamus, and colliculi was analyzed. RESULTS: CAF caused a statistically significant increase in AC3 positive cells when coadministered with midazolam (p = 0.002), ketamine (p = 0.014), or fentanyl (p < 0.001). Our composite dataset suggests that the addition of CAF to these SADs has a supra-additive effect, causing more neurotoxicity than expected. CONCLUSIONS: CAF may augment the neurotoxic action of SADs indicated for neonatal sedation/anesthesia in the NICU by triggering widespread apoptosis in the developing brains of premature infants.


Asunto(s)
Anestésicos/efectos adversos , Apoptosis/efectos de los fármacos , Cafeína/efectos adversos , Hipnóticos y Sedantes/efectos adversos , Neuronas/efectos de los fármacos , Nacimiento Prematuro/patología , Anestésicos/administración & dosificación , Animales , Animales Recién Nacidos , Cafeína/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Humanos , Hipnóticos y Sedantes/administración & dosificación , Ratones , Ratones Endogámicos ICR , Neuronas/fisiología , Síndromes de Neurotoxicidad/patología , Embarazo , Nacimiento Prematuro/tratamiento farmacológico , Nacimiento Prematuro/psicología , Distribución Aleatoria
14.
Neurotoxicol Teratol ; 60: 63-68, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27876652

RESUMEN

Previously we reported that a 5-hour exposure of 6-day-old (P6) rhesus macaques to isoflurane triggers robust neuron and oligodendrocyte apoptosis. In an attempt to further describe the window of vulnerability to anesthetic neurotoxicity, we exposed P20 and P40 rhesus macaques to 5h of isoflurane anesthesia or no exposure (control animals). Brains were collected 3h later and examined immunohistochemically to analyze neuronal and glial apoptosis. Brains exposed to isoflurane displayed neuron and oligodendrocyte apoptosis distributed throughout cortex and white matter, respectively. When combining the two age groups (P20+P40), the animals exposed to isoflurane had 3.6 times as many apoptotic cells as the control animals. In the isoflurane group, approximately 66% of the apoptotic cells were oligodendrocytes and 34% were neurons. In comparison, in our previous studies on P6 rhesus macaques, approximately 52% of the dying cells were glia and 48% were neurons. In conclusion, the present data suggest that the window of vulnerability for neurons is beginning to close in the P20 and P40 rhesus macaques, but continuing for oligodendrocytes.


Asunto(s)
Envejecimiento/fisiología , Apoptosis/fisiología , Encéfalo/fisiología , Isoflurano/efectos adversos , Neuronas/fisiología , Oligodendroglía/fisiología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Femenino , Macaca mulatta , Masculino
15.
Curr Neuropharmacol ; 14(2): 165-76, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26903446

RESUMEN

Opioids are traditionally associated with pain, analgesia and drug abuse. It is now clear, however, that the opioids are central players in mood. The implications for mood disorders, particularly clinical depression, suggest a paradigm shift from the monoamine neurotransmitters to the opioids either alone or in interaction with monoamine neurons. We have a special interest in dynorphin, the last of the major endogenous opioids to be isolated and identified. Dynorphin is derived from the Greek word for power, dynamis, which hints at the expectation that the neuropeptide held for its discoverers. Yet, dynorphin and its opioid receptor subtype, kappa, has always taken a backseat to the endogenous b-endorphin and the exogenous morphine that both bind the mu opioid receptor subtype. That may be changing as the dynorphin/ kappa system has been shown to have different, often opposite, neurophysiological and behavioral influences. This includes major depressive disorder (MDD). Here, we have undertaken a review of dynorphin/ kappa neurobiology as related to behaviors, especially MDD. Highlights include the unique features of dynorphin and kappa receptors and the special relation of a plant-based agonist of the kappa receptor salvinorin A. In addition to acting as a kappa opioid agonist, we conclude that salvinorin A has a complex pharmacologic profile, with potential additional mechanisms of action. Its unique neurophysiological effects make Salvinorina A an ideal candidate for MDD treatment research.


Asunto(s)
Encéfalo/fisiopatología , Trastorno Depresivo Mayor/fisiopatología , Diterpenos de Tipo Clerodano/fisiología , Dinorfinas/fisiología , Receptores Opioides kappa/fisiología , Anhedonia/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Trastorno Depresivo Mayor/tratamiento farmacológico , Diterpenos de Tipo Clerodano/uso terapéutico , Humanos , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inhibidores
16.
Cell Rep ; 16(12): 3208-3218, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27612415

RESUMEN

Zika virus (ZIKV) is an emerging flavivirus that causes congenital abnormalities and Guillain-Barré syndrome. ZIKV infection also results in severe eye disease characterized by optic neuritis, chorioretinal atrophy, and blindness in newborns and conjunctivitis and uveitis in adults. We evaluated ZIKV infection of the eye by using recently developed mouse models of pathogenesis. ZIKV-inoculated mice developed conjunctivitis, panuveitis, and infection of the cornea, iris, optic nerve, and ganglion and bipolar cells in the retina. This phenotype was independent of the entry receptors Axl or Mertk, given that Axl(-/-), Mertk(-/-), and Axl(-/-)Mertk(-/-) double knockout mice sustained levels of infection similar to those of control animals. We also detected abundant viral RNA in tears, suggesting that virus might be secreted from lacrimal glands or shed from the cornea. This model provides a foundation for studying ZIKV-induced ocular disease, defining mechanisms of viral persistence, and developing therapeutic approaches for viral infections of the eye.


Asunto(s)
Panuveítis/virología , Lágrimas/virología , Esparcimiento de Virus/fisiología , Infección por el Virus Zika/virología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Tirosina Quinasas Receptoras/deficiencia , Tirosina Quinasa c-Mer/deficiencia , Tirosina Quinasa del Receptor Axl
17.
World J Psychiatry ; 5(1): 35-46, 2015 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-25815253

RESUMEN

A common remark among laypeople, and notably also among mental health workers, is that individuals with mental illnesses use drugs as self-medication to allay clinical symptoms and the side effects of drug treatments. Roots of the self-medication concept in psychiatry date back at least to the 1980s. Observations that rates of smokers in schizophrenic patients are multiple times the rates for regular smoking in the general population, as well as those with other disorders, proved particularly tempting for a self-medication explanation. Additional evidence came from experiments with animal models exposed to nicotine and the identification of neurobiological mechanisms suggesting self-medication with smoking is a plausible idea. More recently, results from studies comparing smoking and non-smoking schizophrenic patients have led to the questioning of the self-medication hypothesis. Closer examination of the literature points to the possibility that smoking is less beneficial on schizophrenic symptomology than generally assumed while clearly increasing the risk of cancer and other smoking-related diseases responsible for early mortality. It is a good time to examine the evidence for the self-medication concept as it relates to smoking. Our approach is to focus on data addressing direct or implied predictions of the hypothesis in schizophrenic smokers.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA