Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Exp Biol Med (Maywood) ; 248(7): 641-655, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37309741

RESUMEN

General anesthetics are potent neurotoxins when given during early development, causing apoptotic deletion of substantial number of neurons and persistent neurocognitive and behavioral deficits in animals and humans. The period of intense synaptogenesis coincides with the peak of susceptibility to deleterious effects of anesthetics, a phenomenon particularly pronounced in vulnerable brain regions such as subiculum. With steadily accumulating evidence confirming that clinical doses and durations of anesthetics may permanently alter the physiological trajectory of brain development, we set out to investigate the long-term consequences on dendritic morphology of subicular pyramidal neurons and expression on genes regulating the complex neural processes such as neuronal connectivity, learning, and memory. Using a well-established model of anesthetic neurotoxicity in rats and mice neonatally exposed to sevoflurane, a volatile general anesthetic commonly used in pediatric anesthesia, we report that a single 6 h of continuous anesthesia administered at postnatal day (PND) 7 resulted in lasting dysregulation in subicular mRNA levels of cAMP responsive element modulator (Crem), cAMP responsive element-binding protein 1 (Creb1), and Protein phosphatase 3 catalytic subunit alpha, a subunit of calcineurin (Ppp3ca) (calcineurin) when examined during juvenile period at PND28. Given the critical role of these genes in synaptic development and neuronal plasticity, we deployed a set of histological measurements to investigate the implications of anesthesia-induced dysregulation of gene expression on morphology and complexity of surviving subicular pyramidal neurons. Our results indicate that neonatal exposure to sevoflurane induced lasting rearrangement of subicular dendrites, resulting in higher orders of complexity and increased branching with no significant effects on the soma of pyramidal neurons. Correspondingly, changes in dendritic complexity were paralleled by the increased spine density on apical dendrites, further highlighting the scope of anesthesia-induced dysregulation of synaptic development. We conclude that neonatal sevoflurane induced persistent genetic and morphological dysregulation in juvenile rodents, which could indicate heightened susceptibility toward cognitive and behavioral disorders we are beginning to recognize as sequelae of early-in-life anesthesia.


Asunto(s)
Anestésicos por Inhalación , Éteres Metílicos , Humanos , Niño , Animales , Ratas , Ratones , Sevoflurano/toxicidad , Sevoflurano/metabolismo , Calcineurina/metabolismo , Calcineurina/farmacología , Animales Recién Nacidos , Anestésicos por Inhalación/toxicidad , Éteres Metílicos/toxicidad , Hipocampo/metabolismo
2.
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
3.
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
4.
J Matern Fetal Neonatal Med ; 30(18): 2156-2162, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27677376

RESUMEN

OBJECTIVES: Glucocorticoids (GCs) are used to improve respiratory mechanics in preterm infants despite clinical evidence linking neonatal GC therapy to cerebellar pathology. In developing mouse cerebellum, the GC dexamethasone (DEX) causes rapid GC-induced neural progenitor cell apoptosis (GINA). Focusing on pharmacological neuroprotection strategies, we investigated whether dexmedetomidine (DMT) protects against GINA. METHODS: Neonatal mice were pretreated with DMT prior to DEX challenge. Additionally, we tested clonidine and yohimbine in vivo to determine mechanism of DMT neuroprotection. For in vitro studies, cerebellar neural progenitor cells were pretreated with DMT before DEX challenge. RESULTS: In vivo, DMT attenuated GINA at 1 µg/kg and above, p < 0.0001. Clonidine significantly attenuated GINA, p < 0.0001, while yohimbine reversed DMT neuroprotection, p < 0.0001, suggesting DMT neuroprotection is likely mediated via adrenergic signaling. In vitro, DMT neuroprotection was achieved at 10 µM and above, p < 0.001, indicating DMT rescue is cell autonomous. CONCLUSIONS: DMT affords dose-dependent neuroprotection from GINA at clinically relevant doses, an effect that is cell autonomous and likely mediated by α2 adrenergic receptor agonism. DMT co-administration with GCs may be an effective strategy to protect the neonatal brain from GINA while retaining the beneficial effects of GCs on respiratory mechanics.


Asunto(s)
Apoptosis/efectos de los fármacos , Cerebelo/efectos de los fármacos , Dexmedetomidina/farmacología , Glucocorticoides/efectos adversos , Fármacos Neuroprotectores/farmacología , Animales , Animales Recién Nacidos , Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ratones , Ratones Endogámicos ICR , Distribución Aleatoria , Respiración/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos
5.
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
6.
Neurobiol Dis ; 83: 35-43, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26319366

RESUMEN

The external granule layer (EGL) is a proliferative region that produces over 90% of the neurons in the cerebellum but can also malignantly transform into a cerebellar tumor called the medulloblastoma (the most common malignant brain tumor in children). Current dogma considers Hedgehog stimulation a potent proliferative signal for EGL neural progenitor cells (NPCs) and medulloblastomas. However, the Hedgehog pathway also acts as a survival signal in the neural tube where it regulates dorsoventral patterning by controlling NPC apoptosis. Here we show that Hedgehog stimulation is also a potent survival signal in the EGL and medulloblastomas that produces a massive apoptotic response within hours of signal loss in mice. This toxicity can be produced by numerous Hedgehog antagonists (vismodegib, cyclopamine, and jervine) and is Bax/Bak dependent but p53 independent. Finally, since glucocorticoids can also induce EGL and medulloblastoma apoptosis, we show that Hedgehog's effects on apoptosis can occur independent of glucocorticoid stimulation. This effect may play a major role in cerebellar development by directing where EGL proliferation occurs thereby morphologically sculpting growth. It may also be a previously unknown major therapeutic effect of Hedgehog antagonists during medulloblastoma therapy. Results are discussed in terms of their implications for both cerebellar development and medulloblastoma treatment.


Asunto(s)
Apoptosis , Cerebelo/crecimiento & desarrollo , Cerebelo/metabolismo , Proteínas Hedgehog/fisiología , Meduloblastoma/metabolismo , Células-Madre Neurales/metabolismo , Animales , Caspasa 3/metabolismo , Fluocinolona Acetonida/administración & dosificación , Fluocinolona Acetonida/análogos & derivados , Fluocinolona Acetonida/metabolismo , Genes p53 , Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA