Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Methods ; 17(2): 242, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31969728

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Br J Anaesth ; 131(2): 208-211, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37173202

RESUMEN

Over the past two decades there has been an increase in reports of attention deficit-hyperactivity disorder and perhaps autism spectrum disorder that appear to coincide with a substantial number of general anaesthesia interventions during early stages of human brain development. Is there a link between anaesthesia exposure and neurocognitive effects considering the growing body of evidence in numerous animal species, including humans, that suggests long-lasting socio-affective behavioural impairments after early exposure to general anaesthesia? Could routinely used general anaesthetics contribute as environmental toxins? Here we present the case that this notion is worthy of further consideration.


Asunto(s)
Anestésicos Generales , Trastorno por Déficit de Atención con Hiperactividad , Trastorno del Espectro Autista , Animales , Humanos , Anestesia General/efectos adversos , Anestésicos Generales/efectos adversos
3.
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
4.
Neurobiol Dis ; 175: 105923, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36371060

RESUMEN

Preclinical models demonstrate that nearly all anesthetics cause widespread neuroapoptosis in the developing brains of infant rodents and non-human primates. Anesthesia-induced developmental apoptosis is succeeded by prolonged neuropathology in the surviving neurons and lasting cognitive impairments, suggesting that anesthetics interfere with the normal developmental trajectory of the brain. However, little is known about effects of anesthetics on stereotyped axonal pruning, an important developmental algorithm that sculpts neural circuits for proper function. Here, we proposed that neonatal ketamine exposure may interfere with stereotyped axonal pruning of the infrapyramidal bundle (IPB) of the hippocampal mossy fiber system and that impaired pruning may be associated with alterations in the synaptic transmission of CA3 neurons. To test this hypothesis, we injected postnatal day 7 (PND7) mouse pups with ketamine or vehicle over 6 h and then studied them at different developmental stages corresponding to IPB pruning (PND20-40). Immunohistochemistry with synaptoporin (a marker of mossy fibers) revealed that in juvenile mice treated with ketamine at PND7, but not in vehicle-treated controls, positive IPB fibers extended farther into the stratum pyramidale of CA3 region. Furthermore, immunofluorescent double labeling for synaptoporin and PSD-95 strongly suggested that the unpruned IPB caused by neonatal ketamine exposure makes functional synapses. Importantly, patch-clamp electrophysiology for miniature excitatory postsynaptic currents (mEPSCs) in acute brain slices ex vivo revealed increased frequency and amplitudes of mEPSCs in hippocampal CA3 neurons in ketamine-treated groups when compared to vehicle controls. We conclude that neonatal ketamine exposure interferes with normal neural circuit development and that this interference leads to lasting increase in excitatory synaptic transmission in hippocampus.


Asunto(s)
Anestésicos , Ketamina , Ratones , Animales , Ketamina/toxicidad , Transmisión Sináptica/fisiología , Hipocampo , Sinapsis/fisiología , Fibras Musgosas del Hipocampo , Anestésicos/farmacología
5.
Br J Anaesth ; 128(3): 411-413, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35115156

RESUMEN

Despite substantial advocacy for the scientific community to focus on sex-specific differences in biology, the role of sex hormones remains inadequately studied in the field of anaesthesia-induced developmental neurotoxicity. A recent study by Yang and colleagues published in this journal addresses the importance of studying sex hormones during critical stages of brain development. The authors demonstrate that exogenous testosterone administered to immature mice pups around the time of sevoflurane exposure increased brain levels of testosterone, attenuated tau phosphorylation, inhibited glycogen synthase kinase-3ß activation and its interaction/binding with tau, reversed sevoflurane-induced decreases in neuronal activation, and attenuated cognitive impairments. Their well-designed experiments suggest an important role that testosterone plays in balancing several important pathways crucial for neuronal protection and normal function of neuronal circuits in the male mammalian brain.


Asunto(s)
Testosterona , Proteínas tau , Animales , Encéfalo/metabolismo , Femenino , Masculino , Ratones , Fosforilación , Sevoflurano/farmacología , Testosterona/metabolismo , Testosterona/farmacología
6.
Br J Anaesth ; 128(2): 229-231, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34857358

RESUMEN

Growing animal and clinical data continue to point to general anaesthetics as being potentially detrimental to the very young brain. While we are trying to understand the mechanisms responsible for this worrisome phenomenon, we must consider the value of protective strategies that would enable use of currently available general anaesthetics while avoiding histopathological changes and long-lasting impairment in behavioural and cognitive development. Wali and colleagues1 report that the gestational hormone progesterone is a promising 'safening' agent that ameliorates systemic inflammation caused by sevoflurane, a commonly used inhaled anaesthetic, while preventing development of cognitive impairment and an anxious phenotype.


Asunto(s)
Anestésicos Generales , Neuroprotección , Animales , Encéfalo , Hormonas Esteroides Gonadales , Sevoflurano
7.
Br J Anaesth ; 129(4): 555-566, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35701270

RESUMEN

BACKGROUND: General anaesthesia in the neonatal period has detrimental effects on the developing mammalian brain. The impact of underlying inflammation on anaesthesia-induced developmental neurotoxicity remains largely unknown. METHODS: Postnatal day 7 (PND7) rats were randomly assigned to receive sevoflurane (3 vol% for 3 h) or carrier gas 12 h after bacterial lipopolysaccharide (LPS; 1 µg g-1) or vehicle injection. Pharmacological inhibition of caspase-1 by Vx-765 (two doses of 50 µg g-1 body weight) was used to investigate mechanistic pathways of neuronal injury. Histomorphological injury and molecular changes were quantified 2 h after the end of anaesthesia. Long-term functional deficits were tested at 5-8 weeks of age using a battery of behavioural tests in the memory and anxiety domains. RESULTS: Sevoflurane or LPS treatment increased activated caspase-3 and caspase-9 expression in the hippocampal subiculum and CA1, which was greater when sevoflurane was administered in the setting of LPS-induced inflammation. Neuronal injury induced by LPS+sevoflurane treatment resulted in sex-specific behavioural outcomes when rats were tested at 5-8 weeks of age, including learning and memory deficits in males and heightened anxiety-related behaviour in females. Hippocampal caspase-1 and NLRP1 (NLR family pyrin domain containing 1), but not NLRP3, were upregulated by LPS or LPS+sevoflurane treatment, along with related proinflammatory cytokines, interleukin (IL)-1ß, and IL-18. Pretreatment with Vx-765, a selective caspase-1 inhibitor, led to reduced IL-1ß in LPS and LPS+sevoflurane groups. Caspase-1 inhibition by Vx-765 significantly decreased activated caspase-3 and caspase-9 immunoreactivity in the subiculum. CONCLUSIONS: Systemic inflammation promotes developmental neurotoxicity by worsening anaesthesia-induced neuronal damage with sex-specific behavioural outcomes. This highlights the importance of studying anaesthesia-induced neurotoxicity in more clinically relevant settings.


Asunto(s)
Lipopolisacáridos , Síndromes de Neurotoxicidad , Animales , Animales Recién Nacidos , Caspasa 1 , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Citocinas/metabolismo , Inflamación/inducido químicamente , Interleucina-18/metabolismo , Lipopolisacáridos/toxicidad , Masculino , Mamíferos/metabolismo , Síndromes de Neurotoxicidad/etiología , Ratas , Sevoflurano/toxicidad
8.
Int J Mol Sci ; 23(3)2022 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-35163060

RESUMEN

Since its invention, general anesthesia has been an indispensable component of modern surgery. While traditionally considered safe and beneficial in many pathological settings, hundreds of preclinical studies in various animal species have raised concerns about the detrimental and long-lasting consequences that general anesthetics may cause to the developing brain. Clinical evidence of anesthetic neurotoxicity in humans continues to mount as we continue to contemplate how to move forward. Notwithstanding the alarming evidence, millions of children are being anesthetized each year, setting the stage for substantial healthcare burdens in the future. Hence, furthering our knowledge of the molecular underpinnings of anesthesia-induced developmental neurotoxicity is crucially important and should enable us to develop protective strategies so that currently available general anesthetics could be safely used during critical stages of brain development. In this mini-review, we provide a summary of select strategies with primary focus on the mechanisms of neuroprotection and potential for clinical applicability. First, we summarize a diverse group of chemicals with the emphasis on intracellular targets and signal-transduction pathways. We then discuss epigenetic and transgenerational effects of general anesthetics and potential remedies, and also anesthesia-sparing or anesthesia-delaying approaches. Finally, we present evidence of a novel class of anesthetics with a distinct mechanism of action and a promising safety profile.


Asunto(s)
Anestésicos/toxicidad , Desarrollo Infantil/efectos de los fármacos , Síndromes de Neurotoxicidad/prevención & control , Animales , Niño , Epigénesis Genética , Humanos , Mitocondrias/metabolismo , Síndromes de Neurotoxicidad/metabolismo
9.
Int J Mol Sci ; 23(3)2022 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-35163810

RESUMEN

Over the past three decades, we have been grappling with rapidly accumulating evidence that general anesthetics (GAs) may not be as innocuous for the young brain as we previously believed. The growing realization comes from hundreds of animal studies in numerous species, from nematodes to higher mammals. These studies argue that early exposure to commonly used GAs causes widespread apoptotic neurodegeneration in brain regions critical to cognition and socio-emotional development, kills a substantial number of neurons in the young brain, and, importantly, results in lasting disturbances in neuronal synaptic communication within the remaining neuronal networks. Notably, these outcomes are often associated with long-term impairments in multiple cognitive-affective domains. Not only do preclinical studies clearly demonstrate GA-induced neurotoxicity when the exposures occur in early life, but there is a growing body of clinical literature reporting similar cognitive-affective abnormalities in young children who require GAs. The need to consider alternative GAs led us to focus on synthetic neuroactive steroid analogues that have emerged as effective hypnotics, and analgesics that are apparently devoid of neurotoxic effects and long-term cognitive impairments. This would suggest that certain steroid analogues with different cellular targets and mechanisms of action may be safe alternatives to currently used GAs. Herein we summarize our current knowledge of neuroactive steroids as promising novel GAs.


Asunto(s)
Anestésicos Generales/efectos adversos , Red Nerviosa/efectos de los fármacos , Trastornos Neurocognitivos/inducido químicamente , Animales , Niño , Modelos Animales de Enfermedad , Humanos , Trastornos Neurocognitivos/psicología
10.
J Med Syst ; 46(4): 19, 2022 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-35244783

RESUMEN

Over half of hospital revenue results from perioperative patient care, thus emphasizing the importance of efficient resource utilization within a hospital's suite of operating rooms (ORs). Predicting surgical case duration, including Anesthesia-controlled time (ACT) and Surgical-controlled time (SCT) has been significantly detailed throughout the literature as a means to help manage and predict OR scheduling. However, this information has previously been divided by surgical specialty, and only limited benchmarking data regarding ACT and SCT exists. We hypothesized that advancing the granularity of the ACT and SCT from surgical specialty to specific Current Procedural Terminology (CPT®) codes will produce data that is more accurate, less variable, and therefore more useful for OR schedule modeling and management. This single center study was conducted using times from surgeries performed at the University of Colorado Hospital (UCH) between September 2018 - September 2019. Individual cases were categorized by surgical specialty based on the specialty of the primary attending surgeon and CPT codes were compiled from billing data. Times were calculated as defined by the American Association of Clinical Directors. I2 values were calculated to assess heterogeneity of mean ACT and SCT times while Levene's test was utilized to assess heterogeneity of ACT and SCT variances. Statistical analyses for both ACT and SCT were calculated using JMP Statistical Discovery Software from SAS (Cary, NC) and R v3.6.3 (Vienna, Austria). All surgical cases (n = 87,537) performed at UCH from September 2018 to September 2019 were evaluated and 30,091 cases were included in the final analysis. All surgical subspecialties, with the exception of Podiatry, showed significant variability in ACT and SCT values between CPT codes within each surgical specialty. Furthermore, the variances of ACT and SCT values were also highly variable between CPT codes within each surgical specialty. Finally, benchmarking values of mean ACT and SCT with corresponding standard deviations are provided. Because each mean ACT and SCT value varies significantly between different CPT codes within a surgical specialty, using this granularity of data will likely enable improved accuracy in surgical schedule modeling compared to using mean ACT and SCT values for each surgical specialty as a whole. Furthermore, because there was significant variability of ACT and SCT variances between CPT codes, incorporating variance into surgical schedule modeling may also improve accuracy. Future investigations should include real-time simulations, logistical modeling, and labor utilization analyses as well as validation of benchmarking times in private practice settings.


Asunto(s)
Anestesia , Current Procedural Terminology , Anestesia/métodos , Benchmarking , Humanos , Quirófanos , Tempo Operativo , Estados Unidos
11.
Curr Opin Anaesthesiol ; 35(4): 425-435, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35787582

RESUMEN

PURPOSE OF REVIEW: Steadily mounting evidence of anesthesia-induced developmental neurotoxicity has been a challenge in pediatric anesthesiology. Considering that presently used anesthetics have, in different animal models, been shown to cause lasting behavioral impairments when administered at the peak of brain development, the nagging question, 'Is it time for the development of a new anesthetic' must be pondered. RECENT FINDINGS: The emerging 'soft analogs' of intravenous anesthetics aim to overcome the shortcomings of currently available clinical drugs. Remimazolam, a novel ester-analog of midazolam, is a well tolerated intravenous drug with beneficial pharmacological properties. Two novel etomidate analogs currently in development are causing less adrenocortical suppression while maintaining equally favorable hemodynamic stability and rapid metabolism. Quaternary lidocaine derivatives are explored as more potent and longer lasting alternatives to currently available local anesthetics. Xenon, a noble gas with anesthetic properties, is being considered as an anesthetic-sparing adjuvant in pediatric population. Finally, alphaxalone is being reevaluated in a new drug formulation because of its favorable pharmacological properties. SUMMARY: Although a number of exciting anesthetic drugs are under development, there is currently no clear evidence to suggest their lack of neurotoxic properties in young brain. Well designed preclinical studies are needed to evaluate their neurotoxic potential.


Asunto(s)
Anestesia , Anestésicos , Síndromes de Neurotoxicidad , Anestesia/efectos adversos , Anestésicos/efectos adversos , Anestésicos Intravenosos , Animales , Niño , Humanos , Lidocaína/efectos adversos , Midazolam , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/prevención & control
12.
Curr Opin Anaesthesiol ; 35(4): 436-441, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35787588

RESUMEN

PURPOSE OF REVIEW: A family of neuronal voltage-gated calcium channels (VGCCs) have received only recently a significant consideration regarding the mechanisms of anesthesia because VGCC inhibition may be important in anesthetic action by decreasing neuronal excitability and presynaptic excitatory transmission. The T-type VGCCs channels (T-channels), although rarely involved in synaptic neurotransmitter release, play an important role in controlling neuronal excitability and in generating spontaneous oscillatory bursting of groups of neurons in the thalamus thought to be involved in regulating the state of arousal and sleep. Furthermore, these channels are important regulators of neuronal excitability in pain pathway. This review will provide an overview of historic perspective and the recent literature on the role of VGCCs and T-channel inhibition in particular in the mechanisms of action of anesthetics and analgesics. RECENT FINDINGS: Recent research in the field of novel mechanisms of hypnotic action of anesthetics revealed significant contribution of the Ca V 3.1 isoform of T-channels expressed in the thalamus. Furthermore, perioperative analgesia can be achieved by targeting Ca V 3.2 isoform of these channels that is abundantly expressed in pain pathways. SUMMARY: The review summarizes current knowledge regarding the contribution of T-channels in hypnosis and analgesia. Further preclinical and clinical studies are needed to validate their potential for developing novel anesthetics and new perioperative pain therapies.


Asunto(s)
Analgesia , Anestesia , Canales de Calcio/metabolismo , Canales de Calcio/uso terapéutico , Humanos , Dolor/tratamiento farmacológico , Dolor/etiología , Dolor/metabolismo , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/uso terapéutico
13.
Biol Reprod ; 105(3): 720-734, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34258621

RESUMEN

Each year, millions of infants and children are anesthetized for medical and surgical procedures. Yet, a substantial body of preclinical evidence suggests that anesthetics are neurotoxins that cause rapid and widespread apoptotic cell death in the brains of infant rodents and nonhuman primates. These animals have persistent impairments in cognition and behavior many weeks or months after anesthesia exposure, leading us to hypothesize that anesthetics do more than simply kill brain cells. Indeed, anesthetics cause chronic neuropathology in neurons that survive the insult, which then interferes with major aspects of brain development, synaptic plasticity, and neuronal function. Understanding the phenomenon of anesthesia-induced developmental neurotoxicity is of critical public health importance because clinical studies now report that anesthesia in human infancy is associated with cognitive and behavioral deficits. In our search for mechanistic explanations for why a young and pliable brain cannot fully recover from a relatively brief period of anesthesia, we have accumulated evidence that neonatal anesthesia can dysregulate epigenetic tags that influence gene transcription such as histone acetylation and DNA methylation. In this review, we briefly summarize the phenomenon of anesthesia-induced developmental neurotoxicity. We then discuss chronic neuropathology caused by neonatal anesthesia, including disturbances in cognition, socio-affective behavior, neuronal morphology, and synaptic plasticity. Finally, we present evidence of anesthesia-induced genetic and epigenetic dysregulation within the developing brain that may be transmitted intergenerationally to anesthesia-naïve offspring.


Asunto(s)
Anestesia/efectos adversos , Animales Recién Nacidos/genética , Epigenoma/efectos de los fármacos , Primates/genética , Animales , Humanos , Recién Nacido , Ratones , Ratas
14.
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
15.
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
16.
Int J Mol Sci ; 22(6)2021 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-33802812

RESUMEN

Targeting tumor vasculature through specific endothelial cell markers represents a promising approach for cancer treatment. Here our aim was to construct an antibiotic resistance gene-free plasmid encoding shRNAs to simultaneously target two endothelial cell markers, CD105 and CD146, and to test its functionality and therapeutic potential in vitro when delivered by gene electrotransfer (GET) and combined with irradiation (IR). Functionality of the plasmid was evaluated by determining the silencing of the targeted genes using qRT-PCR. Antiproliferative and antiangiogenic effects were determined by the cytotoxicity assay tube formation assay and wound healing assay in murine endothelial cells 2H-11. The functionality of the plasmid construct was also evaluated in malignant melanoma tumor cell line B16F10. Additionally, potential activation of immune response was measured by induction of DNA sensor STING and proinflammatory cytokines by qRT-PCR in endothelial cells 2H-11. We demonstrated that the plasmid construction was successful and can efficiently silence the expression of the two targeted genes. As a consequence of silencing, reduced migration rate and angiogenic potential was confirmed in 2H-11 endothelial cells. Furthermore, induction of DNA sensor STING and proinflammatory cytokines were determined, which could add to the therapeutic effectiveness when used in vivo. To conclude, we successfully constructed a novel plasmid DNA with two shRNAs, which holds a great promise for further in vivo testing.


Asunto(s)
Antígeno CD146/genética , Electroporación , Endoglina/genética , Silenciador del Gen , Plásmidos/genética , Radiación Ionizante , Transfección , Animales , Muerte Celular , Línea Celular , Citocinas/metabolismo , Células Endoteliales/efectos de la radiación , Proteínas de la Membrana , Ratones , Neovascularización Fisiológica/efectos de la radiación
17.
Br J Anaesth ; 124(3): e81-e91, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31980157

RESUMEN

Exposure to anaesthetic drugs during the fetal or neonatal period induces widespread neuronal apoptosis in the brains of rodents and non-human primates. Hundreds of published preclinical studies and nearly 20 clinical studies have documented cognitive and behavioural deficits many months or years later, raising the spectre that early life anaesthesia exposure is a long-term, perhaps permanent, insult that might affect the quality of life of millions of humans. Although the phenomenon of anaesthesia-induced developmental neurotoxicity is well characterised, there are important and lingering questions pertaining to sex differences and neurodevelopmental sequelae that might occur differentially in females and males. We review the relevant literature on sex differences in the field of anaesthesia-induced developmental neurotoxicity, and present an emerging pattern of potential sex-dependent neurodevelopmental abnormalities in rodent models of human infant anaesthesia exposure.


Asunto(s)
Anestésicos/efectos adversos , Trastornos del Neurodesarrollo/inducido químicamente , Animales , Ansiedad/inducido químicamente , Apoptosis/efectos de los fármacos , Trastorno por Déficit de Atención con Hiperactividad/inducido químicamente , Trastornos del Conocimiento/inducido químicamente , Metilación de ADN , Femenino , Humanos , Masculino , Síndromes de Neurotoxicidad/etiología , Caracteres Sexuales
18.
Br J Anaesth ; 124(5): 603-613, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32151384

RESUMEN

BACKGROUND: The most currently used general anaesthetics are potent potentiators of γ-aminobutyric acid A (GABAA) receptors and are invariably neurotoxic during the early stages of brain development in preclinical animal models. As causality between GABAA potentiation and anaesthetic-induced developmental neurotoxicity has not been established, the question remains whether GABAergic activity is crucial for promoting/enhancing neurotoxicity. Using the neurosteroid analogue, (3α,5α)-3-hydroxy-13,24-cyclo-18,21-dinorchol-22-en-24-ol (CDNC24), which potentiates recombinant GABAA receptors, we examined whether this potentiation is the driving force in inducing neurotoxicity during development. METHODS: The neurotoxic potential of CDNC24 was examined vis-à-vis propofol (2,6-diisopropylphenol) and alphaxalone (5α-pregnan-3α-ol-11,20-dione) at the peak of rat synaptogenesis. In addition to the morphological neurotoxicity studies of the subiculum and medial prefrontal cortex (mPFC), we assessed the extra-, pre-, and postsynaptic effects of these agents on GABAergic neurotransmission in acute subicular slices from rat pups. RESULTS: CDNC24, like alphaxalone and propofol, caused dose-dependent hypnosis in vivo, with a higher therapeutic index. CDNC24 and alphaxalone, unlike propofol, did not cause developmental neuroapoptosis in the subiculum and mPFC. Propofol potentiated post- and extrasynaptic GABAA currents as evidenced by increased spontaneous inhibitory postsynaptic current (sIPSC) decay time and prominent tonic currents, respectively. CDNC24 and alphaxalone had a similar postsynaptic effect, but also displayed a strong presynaptic effect as evidenced by decreased frequency of sIPSCs and induced moderate tonic currents. CONCLUSIONS: The lack of neurotoxicity of CDNC24 and alphaxalone may be at least partly related to suppression of presynaptic GABA release in the developing brain.


Asunto(s)
Encéfalo/efectos de los fármacos , Hipnóticos y Sedantes/toxicidad , Pregnanodionas/toxicidad , Esteroides/toxicidad , Animales , Apoptosis/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Relación Dosis-Respuesta a Droga , Agonistas de Receptores de GABA-A/administración & dosificación , Agonistas de Receptores de GABA-A/farmacología , Agonistas de Receptores de GABA-A/toxicidad , Hipocampo/efectos de los fármacos , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Hipocampo/patología , Hipnóticos y Sedantes/administración & dosificación , Hipnóticos y Sedantes/farmacología , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/patología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/crecimiento & desarrollo , Corteza Prefrontal/patología , Pregnanodionas/administración & dosificación , Pregnanodionas/farmacología , Propofol/administración & dosificación , Propofol/farmacología , Propofol/toxicidad , Ratas Sprague-Dawley , Receptores de GABA-A/metabolismo , Esteroides/administración & dosificación , Esteroides/farmacología , Sinapsis/efectos de los fármacos , Sinapsis/fisiología
19.
Br J Anaesth ; 124(5): 585-593, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32145876

RESUMEN

In March 2019, SmartTots, a public-private partnership between the US Food and Drug Administration and the International Anesthesia Research Society, hosted a meeting attended by research experts, anaesthesia journal editors, and government agency representatives to discuss the continued need for rigorous preclinical research and the importance of establishing reporting standards for the field of anaesthetic perinatal neurotoxicity. This group affirmed the importance of preclinical research in the field, and welcomed novel and mechanistic approaches to answer some of the field's largest questions. The attendees concluded that summarising the benefits and disadvantages of specific model systems, and providing guidance for reporting results, would be helpful for designing new experiments and interpreting results across laboratories. This expert opinion report is a summary of these discussions, and includes a focused review of current animal models and reporting standards for the field of perinatal anaesthetic neurotoxicity. This will serve as a practical guide and road map for novel and rigorous experimental work.


Asunto(s)
Anestésicos/efectos adversos , Investigación Biomédica/normas , Evaluación Preclínica de Medicamentos/normas , Síndromes de Neurotoxicidad/etiología , Informe de Investigación/normas , Animales , Investigación Biomédica/métodos , Niño , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Humanos , Asociación entre el Sector Público-Privado
20.
Neurobiol Learn Mem ; 165: 106834, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-29550366

RESUMEN

Fifteen years ago Olney and colleagues began using animal models to evaluate the effects of anesthetic and sedative agents (ASAs) on neurodevelopment. The results from ongoing studies indicate that, under certain conditions, exposure to these drugs during development induces an acute elevated apoptotic neurodegenerative response in the brain and long-term functional impairments. These animal models have played a significant role in bringing attention to the possible adverse effects of exposing the developing brain to ASAs when few concerns had been raised previously in the medical community. The apoptotic degenerative response resulting from neonatal exposure to ASAs has been replicated in many studies in both rodents and non-human primates, suggesting that a similar effect may occur in humans. In both rodents and non-human primates, significantly increased levels of apoptotic degeneration are often associated with functional impairments later in life. However, behavioral deficits following developmental ASA exposure have not been consistently reported even when significantly elevated levels of apoptotic degeneration have been documented in animal models. In the present work, we review this literature and propose a rodent model for assessing potential functional deficits following neonatal ASA exposure with special reference to experimental design and procedural issues. Our intent is to improve test sensitivity and replicability for detecting subtle behavioral effects, and thus enhance the translational significance of ASA models.


Asunto(s)
Anestesia/efectos adversos , Trastornos del Neurodesarrollo/inducido químicamente , Anestésicos/efectos adversos , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA