Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 812
Filtrar
1.
Commun Biol ; 7(1): 885, 2024 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-39033173

RESUMEN

Rhythmic brain activity is critical to many brain functions and is sensitive to neuromodulation, but so far very few studies have investigated this activity on the cellular level in vitro in human brain tissue samples. This study reveals and characterizes a novel rhythmic network activity in the human neocortex. Using intracellular patch-clamp recordings of human cortical neurons, we identify large rhythmic depolarizations (LRDs) driven by glutamate release but not by GABA. These LRDs are intricate events made up of multiple depolarizing phases, occurring at ~0.3 Hz, have large amplitudes and long decay times. Unlike human tissue, rat neocortex layers 2/3 exhibit no such activity under identical conditions. LRDs are mainly observed in a subset of L2/3 interneurons that receive substantial excitatory inputs and are likely large basket cells based on their morphology. LRDs are highly sensitive to norepinephrine (NE) and acetylcholine (ACh), two neuromodulators that affect network dynamics. NE increases LRD frequency through ß-adrenergic receptor activity while ACh decreases it via M4 muscarinic receptor activation. Multi-electrode array recordings show that NE enhances and synchronizes oscillatory network activity, whereas ACh causes desynchronization. Thus, NE and ACh distinctly modulate LRDs, exerting specific control over human neocortical activity.


Asunto(s)
Acetilcolina , Neocórtex , Norepinefrina , Humanos , Acetilcolina/farmacología , Norepinefrina/farmacología , Neocórtex/fisiología , Neocórtex/metabolismo , Neocórtex/citología , Neocórtex/efectos de los fármacos , Masculino , Femenino , Animales , Persona de Mediana Edad , Ratas , Anciano , Periodicidad , Neuronas/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Interneuronas/fisiología , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Adulto
2.
Med Gas Res ; 14(3): 115-120, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39073339

RESUMEN

Sevoflurane has been shown to increase the incidence of emergence delirium in children; however, the mechanism remains unclear. Sevoflurane increases cytoplasmic calcium concentration which in turn may play a role in emergence delirium. This study aimed to investigate the level of intracellular calcium in rats experiencing hyperexcitatory behavior after exposure to sevoflurane, as well as the role of magnesium in preventing this phenomenon. After ethical approval, 2-5-week-old Sprague-Dawley rats (n = 34) were insufflated with sevoflurane in a modified anesthesia chamber. One group received magnesium sulphate intraperitoneally. After termination of sevoflurane exposure, the occurrence of hyperexcitation was observed. Brain tissue samples from the rats were studied for intracellular calcium levels under a two-channel laser scanning confocal microscope and were quantitatively calculated using ratiometric calculation. The presence of inflammation or oxidative stress reaction was assessed using nuclear factor κB and malondialdehyde. The incidence of hyperexcitatory behavior post sevoflurane exposure was 9 in 16 rats in the observation group and none in the magnesium group. Tests for inflammation and oxidative stress were within normal limits in both groups. The rats showing hyperexcitation had a higher level of cytosol calcium concentration compared to the other groups. To conclude, the calcium concentration of neocortical neurons in Sprague-Dawley rats with hyperexcitatory behavior is increased after exposure to sevoflurane. Administration of magnesium sulphate can prevent the occurrence of hyperexcitation in experimental animals.


Asunto(s)
Calcio , Neocórtex , Neuronas , Ratas Sprague-Dawley , Sevoflurano , Animales , Sevoflurano/farmacología , Sevoflurano/efectos adversos , Calcio/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Neocórtex/efectos de los fármacos , Neocórtex/metabolismo , Masculino , Anestésicos por Inhalación/farmacología , Anestésicos por Inhalación/efectos adversos , Éteres Metílicos/farmacología , Éteres Metílicos/efectos adversos , Conducta Animal/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos
3.
Artículo en Inglés | MEDLINE | ID: mdl-38797491

RESUMEN

BACKGROUND AND PURPOSE: Chronic neuropathic pain (NP) is commonly associated with cognitive and emotional impairments. Cannabidiol (CBD) presents a broad spectrum of action with a potential analgesic effect. This work investigates the CBD effect on comorbidity between chronic NP, depression, and memory impairment. EXPERIMENTAL APPROACH: The connection between the neocortex and the hippocampus was investigated with biotinylated dextran amine (BDA) deposits in the prelimbic cortex (PrL). Wistar rats were submitted to chronic constriction injury (CCI) of the sciatic nerve and CA1 treatment with CBD (15, 30, 60 nmol). KEY RESULTS: BDA-labeled perikarya and terminal buttons were found in CA1 and dentate gyrus. CCI-induced mechanical and cold allodynia increased c-Fos protein expression in the PrL and CA1. The number of astrocytes in PrL and CA1 increased, and the number of neuroblasts decreased in CA1. Animals submitted to CCI procedure showed increasing depressive-like behaviors, such as memory impairment. CBD (60 nmol) treatment decreased mechanical and cold allodynia, attenuated depressive-associated behaviors, and improved memory performance. Cobalt chloride (CoCl2: 1 nM), WAY-100635 (0.37 nmol), and AM251 (100 nmol) intra-PrL reversed the effect of CA1 treatment with CBD (60 nmol) on nociceptive, cognitive, and depressive behaviors. CONCLUSION: CBD represents a promising therapeutic perspective in the pharmacological treatment of chronic NP and associated comorbidities such as depression and memory impairments. The CBD effects possibly recruit the CA1-PrL pathway, inducing neuroplasticity. CBD acute treatment into the CA1 produces functional and molecular morphological improvements.


Asunto(s)
Cannabidiol , Disfunción Cognitiva , Hipocampo , Neocórtex , Neuralgia , Ratas Wistar , Animales , Cannabidiol/farmacología , Cannabidiol/uso terapéutico , Masculino , Neuralgia/tratamiento farmacológico , Ratas , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Neocórtex/efectos de los fármacos , Hiperalgesia/tratamiento farmacológico , Vías Nerviosas/efectos de los fármacos , Síntomas Afectivos/tratamiento farmacológico , Síntomas Afectivos/etiología
4.
Chemosphere ; 358: 142124, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38677614

RESUMEN

Metformin, the most commonly prescribed drug for the treatment of diabetes, is increasingly used during pregnancy to address various disorders such as diabetes, obesity, preeclampsia, and metabolic diseases. However, its impact on neocortex development remains unclear. Here, we investigated the direct effects of metformin on neocortex development, focusing on ERK and p35/CDK5 regulation. Using a pregnant rat model, we found that metformin treatment during pregnancy induces small for gestational age (SGA) and reduces relative cortical thickness in embryos and neonates. Additionally, we discovered that metformin inhibits neural progenitor cell proliferation in the sub-ventricular zone (SVZ)/ventricular zone (VZ) of the developing neocortex, a process possibly mediated by ERK inactivation. Furthermore, metformin induces neuronal apoptosis in the SVZ/VZ area of the developing neocortex. Moreover, metformin retards neuronal migration, cortical lamination, and differentiation, potentially through p35/CDK5 inhibition in the developing neocortex. Remarkably, compensating for p35 through in utero electroporation partially rescues metformin-impaired neuronal migration and development. In summary, our study reveals that metformin disrupts neocortex development by inhibiting neuronal progenitor proliferation, neuronal migration, cortical layering, and cortical neuron maturation, likely via ERK and p35/CDK5 inhibition. Consequently, our findings advocate for caution in metformin usage during pregnancy, given its potential adverse effects on fetal brain development.


Asunto(s)
Proliferación Celular , Quinasa 5 Dependiente de la Ciclina , Metformina , Neocórtex , Metformina/farmacología , Animales , Femenino , Embarazo , Neocórtex/efectos de los fármacos , Quinasa 5 Dependiente de la Ciclina/metabolismo , Ratas , Proliferación Celular/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neuronas/efectos de los fármacos , Ratas Sprague-Dawley , Diferenciación Celular/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
5.
eNeuro ; 11(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38664009

RESUMEN

Seizures are generally associated with epilepsy but may also be a symptom of many other neurological conditions. A hallmark of a seizure is the intensity of the local neuronal activation, which can drive large-scale gene transcription changes. Such changes in the transcriptional profile likely alter neuronal function, thereby contributing to the pathological process. Therefore, there is a strong clinical imperative to characterize how gene expression is changed by seizure activity. To this end, we developed a simplified ex vivo technique for studying seizure-induced transcriptional changes. We compared the RNA sequencing profile in mouse neocortical tissue with up to 3 h of epileptiform activity induced by 4-aminopyridine (4AP) relative to control brain slices not exposed to the drug. We identified over 100 genes with significantly altered expression after 4AP treatment, including multiple genes involved in MAPK, TNF, and neuroinflammatory signaling pathways, all of which have been linked to epilepsy previously. Notably, the patterns in male and female brain slices were almost identical. Various immediate early genes were among those showing the largest upregulation. The set of down-regulated genes included ones that might be expected either to increase or to decrease neuronal excitability. In summary, we found the seizure-induced transcriptional profile complex, but the changes aligned well with an analysis of published epilepsy-associated genes. We discuss how simple models may provide new angles for investigating seizure-induced transcriptional changes.


Asunto(s)
4-Aminopiridina , Neocórtex , Transcriptoma , Animales , Neocórtex/metabolismo , Neocórtex/efectos de los fármacos , Femenino , Masculino , Ratones , 4-Aminopiridina/farmacología , Convulsiones/genética , Convulsiones/metabolismo , Convulsiones/fisiopatología , Análisis de Secuencia de ARN/métodos , Epilepsia/genética , Epilepsia/metabolismo , Epilepsia/fisiopatología , Ratones Endogámicos C57BL
6.
Neurochem Int ; 152: 105251, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34861326

RESUMEN

Inflammation is usually a tightly regulated process whose termination by mediators including Annexin A1 (AnxA1) results in the resolution of inflammatory responses. In neurodegenerative dementias, chronic neuroinflammation, along with accumulation of aggregated ß-amyloid (Aß) peptides and apoptosis, has long been recognized to be a pathological hallmark; but it is unclear whether a failure of inflammation resolution contributes to this pathophysiological process. In this study, we measured AnxA1 immunoreactivities in postmortem neocortex (Brodmann areas BA9 and BA40) of well characterized Alzheimer's disease (AD), Parkinson disease dementia (PDD) and dementia with Lewy bodies (DLB) patients as well as aged controls. Inactive cleaved AnxA1 was found to be elevated in AD and DLB in BA40. Levels of cleaved AnxA1 also positively correlated with amyloidogenic brain Aß, anti-inflammatory markers such as IL10 and IL13, as well as with the pro-apoptotic marker cleaved caspase-3 in BA40. Our findings suggest that elevated cleaved AnxA1 in neurodegenerative dementias may reflect a failure of inflammation resolution in certain regions of the diseased brain, and also support a mechanistic link between AnxA1 and amyloid pathology, neuroinflammation and apoptosis.


Asunto(s)
Anexina A1/metabolismo , Demencia/metabolismo , Neocórtex/metabolismo , Enfermedad de Parkinson/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Antiinflamatorios/farmacología , Biomarcadores/sangre , Demencia/tratamiento farmacológico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neocórtex/efectos de los fármacos , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/patología , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/patología
7.
Exp Neurol ; 347: 113916, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34752784

RESUMEN

Temporal lobe epilepsy (TLE) is the most common form of intractable epilepsy where hyperactive glutamate receptors may contribute to the complex epileptogenic network hubs distributed among different regions. This study was designed to investigate the region-specific molecular alterations of the glutamate receptors and associated excitatory synaptic transmission in pilocarpine rat model of TLE. We recorded spontaneous excitatory postsynaptic currents (EPSCs) from pyramidal neurons in resected rat brain slices of the hippocampus, anterior temporal lobe (ATL) and neocortex. We also performed mRNA and protein expression of the glutamate receptor subunits (NR1, NR2A, NR2B, and GLUR1-4) by qPCR and immunohistochemistry. We observed significant increase in the frequency and amplitude of spontaneous EPSCs in the hippocampal and ATL samples of TLE rats than in control rats. Additionally, the magnitude of the frequency and amplitude was increased in ATL samples compared to that of the hippocampal samples of TLE rats. The mRNA level of NR1 was upregulated in both the hippocampal as well as ATL samples and that of NR2A, NR2B were upregulated only in the hippocampal samples of TLE rats than in control rats. The mRNA level of GLUR4 was upregulated in both the hippocampal as well as ATL samples of TLE rats than in control rats. Immunohistochemical analysis demonstrated that the number of NR1, NR2A, NR2B, and GLUR4 immuno-positive cells were significantly higher in the hippocampal samples whereas number of NR1 and GLUR4 immuno-positive cells were significantly higher in the ATL samples of the TLE rats than in control rats. This study demonstrated the region-specific alterations of glutamate receptor subunits in pilocarpine model of TLE, suggesting possible cellular mechanisms contributing to generation of independent epileptogenic networks in different temporal lobe structures.


Asunto(s)
Epilepsia del Lóbulo Temporal/metabolismo , Hipocampo/metabolismo , Neocórtex/metabolismo , Pilocarpina/toxicidad , Receptores de Glutamato/biosíntesis , Lóbulo Temporal/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Epilepsia del Lóbulo Temporal/inducido químicamente , Epilepsia del Lóbulo Temporal/patología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Expresión Génica , Hipocampo/efectos de los fármacos , Hipocampo/patología , Masculino , Neocórtex/efectos de los fármacos , Neocórtex/patología , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato/genética , Lóbulo Temporal/efectos de los fármacos , Lóbulo Temporal/patología
8.
Development ; 148(17)2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34414407

RESUMEN

Reelin is a large secreted glycoprotein that regulates neuronal migration, lamination and establishment of dendritic architecture in the embryonic brain. Reelin expression switches postnatally from Cajal-Retzius cells to interneurons. However, reelin function in interneuron development is still poorly understood. Here, we have investigated the role of reelin in interneuron development in the postnatal neocortex. To preclude early cortical migration defects caused by reelin deficiency, we employed a conditional reelin knockout (RelncKO) mouse to induce postnatal reelin deficiency. Induced reelin deficiency caused dendritic hypertrophy in distal dendritic segments of neuropeptide Y-positive (NPY+) and calretinin-positive (Calr+) interneurons, and in proximal dendritic segments of parvalbumin-positive (Parv+) interneurons. Chronic recombinant Reelin treatment rescued dendritic hypertrophy in Relncko interneurons. Moreover, we provide evidence that RelncKO interneuron hypertrophy is due to presynaptic GABABR dysfunction. Thus, GABABRs in RelncKO interneurons were unable to block N-type (Cav2.2) Ca2+ channels that control neurotransmitter release. Consequently, the excessive Ca2+ influx through AMPA receptors, but not NMDA receptors, caused interneuron dendritic hypertrophy. These findings suggest that reelin acts as a 'stop-growth-signal' for postnatal interneuron maturation.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Dendritas/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Interneuronas/citología , Neocórtex/crecimiento & desarrollo , Proteínas del Tejido Nervioso/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Calbindina 2/metabolismo , Calcio/metabolismo , Moléculas de Adhesión Celular Neuronal/deficiencia , Moléculas de Adhesión Celular Neuronal/farmacología , Dendritas/efectos de los fármacos , Proteínas de la Matriz Extracelular/deficiencia , Proteínas de la Matriz Extracelular/farmacología , Hipertrofia , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Ratones , Ratones Noqueados , Neocórtex/citología , Neocórtex/efectos de los fármacos , Neocórtex/patología , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/farmacología , Neuropéptido Y/metabolismo , Parvalbúminas/metabolismo , Receptores de GABA-B/metabolismo , Receptores de Glutamato/metabolismo , Proteína Reelina , Serina Endopeptidasas/deficiencia , Serina Endopeptidasas/farmacología
9.
Sci Data ; 8(1): 180, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34267214

RESUMEN

Publicly available neural recordings obtained with high spatial resolution are scarce. Here, we present an electrophysiological dataset recorded from the neocortex of twenty rats anesthetized with ketamine/xylazine. The wideband, spontaneous recordings were acquired with a single-shank silicon-based probe having 128 densely-packed recording sites arranged in a 32 × 4 array. The dataset contains the activity of a total of 7126 sorted single units extracted from all layers of the cortex. Here, we share raw neural recordings, as well as spike times, extracellular spike waveforms and several properties of units packaged in a standardized electrophysiological data format. For technical validation of our dataset, we provide the distributions of derived single unit properties along with various spike sorting quality metrics. This large collection of in vivo data enables the investigation of the high-resolution electrical footprint of cortical neurons which in turn may aid their electrophysiology-based classification. Furthermore, the dataset might be used to study the laminar-specific neuronal activity during slow oscillation, a brain rhythm strongly involved in neural mechanisms underlying memory consolidation and sleep.


Asunto(s)
Ondas Encefálicas , Consolidación de la Memoria , Neocórtex/fisiología , Neuronas/fisiología , Sueño , Analgésicos/farmacología , Anestesia , Animales , Femenino , Ketamina/farmacología , Neocórtex/citología , Neocórtex/efectos de los fármacos , Ratas , Ratas Wistar , Xilazina/farmacología
10.
Neuropharmacology ; 196: 108676, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34216585

RESUMEN

The mouse model of beta-amyloid (Aß) deposition, APP/PS1-21, exhibits high brain uptake of the tau-tracer (S)-[18F]THK5117, although no neurofibrillary tangles are present in this mouse model. For this reason we investigated (S)-[18F]THK5117 off-target binding to Aß plaques and MAO-B enzyme in APP/PS1-21 transgenic (TG) mouse model of Aß deposition. APP/PS1-21 TG and wild-type (WT) control mice in four different age groups (2-26 months) were imaged antemortem by positron emission tomography with (S)-[18F]THK5117, and then brain autoradiography. Additional animals were used for immunohistochemical staining and MAO-B enzyme blocking study with deprenyl pre-treatment. Regional standardized uptake value ratios for the cerebellum revealed a significant temporal increase in (S)-[18F]THK5117 uptake in aged TG, but not WT, brain. Immunohistochemical staining revealed a similar increase in Aß plaques but not endogenous hyper-phosphorylated tau or MAO-B enzyme, and ex vivo autography showed that uptake of (S)-[18F]THK5117 co-localized with the amyloid pathology. Deprenyl hydrochloride pre-treatment reduced the binding of (S)-[18F]THK5117 in the neocortex, hippocampus, and thalamus. This study's findings suggest that increased (S)-[18F]THK5117 binding in aging APP/PS1-21 TG mice is mainly due to increasing Aß deposition, and to a lesser extent binding to MAO-B enzyme, but not hyper-phosphorylated tau.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico por imagen , Péptidos beta-Amiloides/metabolismo , Encéfalo/diagnóstico por imagen , Monoaminooxidasa/metabolismo , Placa Amiloide/diagnóstico por imagen , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Compuestos de Anilina , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Hipocampo/diagnóstico por imagen , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ratones , Ratones Transgénicos , Inhibidores de la Monoaminooxidasa/farmacología , Neocórtex/diagnóstico por imagen , Neocórtex/efectos de los fármacos , Neocórtex/metabolismo , Placa Amiloide/metabolismo , Tomografía de Emisión de Positrones , Presenilina-1/genética , Quinolinas , Radiofármacos , Selegilina/farmacología , Tálamo/diagnóstico por imagen , Tálamo/efectos de los fármacos , Tálamo/metabolismo
11.
Neurobiol Learn Mem ; 183: 107484, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34175450

RESUMEN

Retrieval deficit of long-term memory is a cardinal symptom of dementia and has been proposed to associate with abnormalities in the central cholinergic system. Difficulty in the retrieval of memory is experienced by healthy individuals and not limited to patients with neurological disorders that result in forgetfulness. The difficulty of retrieving memories is associated with various factors, such as how often the event was experienced or remembered, but it is unclear how the cholinergic system plays a role in the retrieval of memory formed by a daily routine (accumulated experience). To investigate this point, we trained rats moderately (for a week) or extensively (for a month) to detect a visual cue in a two-alternative forced-choice task. First, we confirmed the well-established memory in the extensively trained group was more resistant to the retrieval problem than recently acquired memory in the moderately trained group. Next, we tested the effect of a cholinesterase inhibitor, donepezil, on the retrieval of memory after a long no-task period in extensively trained rats. Pre-administration of donepezil improved performance and reduced the latency of task initiation compared to the saline-treated group. Finally, we lesioned cholinergic neurons of the nucleus basalis magnocellularis (NBM), which project to the entire neocortex, by injecting the cholinergic toxin 192 IgG-saporin. NBM-lesioned rats showed severely impaired task initiation and performance. These abilities recovered as the trials progressed, though they never reached the level observed in rats with intact NBM. These results suggest that acetylcholine released from the NBM contributes to the retrieval of well-established memory developed by a daily routine.


Asunto(s)
Acetilcolina/metabolismo , Núcleo Basal de Meynert/fisiología , Neuronas Colinérgicas/fisiología , Recuerdo Mental/fisiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Anticuerpos Monoclonales/farmacología , Núcleo Basal de Meynert/efectos de los fármacos , Núcleo Basal de Meynert/metabolismo , Colinérgicos/farmacología , Neuronas Colinérgicas/efectos de los fármacos , Neuronas Colinérgicas/metabolismo , Inhibidores de la Colinesterasa/farmacología , Donepezilo/farmacología , Recuerdo Mental/efectos de los fármacos , Neocórtex/efectos de los fármacos , Neocórtex/metabolismo , Neocórtex/fisiología , Ratas , Saporinas/farmacología
12.
Brain Res ; 1759: 147370, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33600830

RESUMEN

Genes and environment interact during intrauterine life, and potentially alter the developmental trajectory of the brain. This can result in life-long consequences on brain function. We have previously developed two transgenic mouse lines that suppress Gad1 expression in parvalbumin (PVALB) and neuropeptide Y (NPY) expressing interneuron populations using a bacterial artificial chromosome (BAC)-driven miRNA-based silencing technology. We were interested to assess if maternal immune activation (MIA), genetic interneuronal inhibition, and the combination of these two factors disrupt and result in long-term changes in neuroinflammatory gene expression, sterol biosynthesis, and acylcarnitine levels in the brain of maternally exposed offspring. Pregnant female WT mice were given a single intraperitoneal injection of saline or polyinosinic-polycytidilic acid [poly(I:C)] at E12.5. Brains of offspring were analyzed at postnatal day 90. We identified complex and persistent neuroinflammatory gene expression changes in the hippocampi of MIA-exposed offspring, as well in the hippocampi of Npy/Gad1 and Pvalb/Gad1 mice. In addition, both MIA and genetic inhibition altered the post-lanosterol sterol biosynthesis in the neocortex and disrupted the typical acylcarnitine profile. In conclusion, our findings suggest that both MIA and inhibition of interneuronal function have long-term consequences on critical homeostatic mechanisms of the brain, including immune function, sterol levels, and energy metabolism.


Asunto(s)
Mediadores de Inflamación/inmunología , Interneuronas/inmunología , Neuroinmunomodulación/fisiología , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/inmunología , Animales , Femenino , Glutamato Descarboxilasa/deficiencia , Glutamato Descarboxilasa/genética , Hipocampo/efectos de los fármacos , Hipocampo/inmunología , Hipocampo/metabolismo , Mediadores de Inflamación/metabolismo , Inductores de Interferón/toxicidad , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neocórtex/efectos de los fármacos , Neocórtex/inmunología , Neocórtex/metabolismo , Neuroinmunomodulación/efectos de los fármacos , Poli I-C/toxicidad , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/metabolismo
14.
J Comp Neurol ; 529(7): 1628-1641, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32975324

RESUMEN

Previous evidence indicated a potential mechanism that might support the fact that primates exhibit greater neural integration capacity as a result of the activation of different structures of the central nervous system, as compared to rodents. The current study aimed to provide further evidence to confirm previous findings by analyzing the patterns of c-Fos expression in more neocortical structures of rats and marmosets using a more robust quantitative technique and evaluating a larger number of brain areas. Nineteen Wistar rats and 21 marmosets (Callithrix jacchus) were distributed among control groups (animals without injections) and animals injected with pentylenetetrazol (PTZ) and euthanized at different time points after stimulus. Immunohistochemical detection of c-Fos was quantified using unbiased and efficient stereological cell counting in eight neocortical regions. Marmosets had a c-Fos expression that was notably more widely expressed (5× more cells) and longer lasting (up to 3 hr) than rats. c-Fos expression in rats presented similar patterns of expression according to the function of the brain cortical structures (associative, sensorial, and motor functions), which was not observed for marmosets (in which no clear pattern could be drawn, and a more diverse profile emerged). Our results provide evidence that the marmoset brain has a greater neuronal activation after intense stimulation by means of PTZ and a more complex pattern of brain activation. We speculate that these functional differences may contribute for the understanding of the different neuronal processing capacities of the neocortex in these mammals' orders.


Asunto(s)
Neocórtex/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Animales , Callithrix , Antagonistas del GABA/farmacología , Masculino , Neocórtex/efectos de los fármacos , Pentilenotetrazol/farmacología , Ratas , Ratas Wistar
15.
Ann Neurol ; 89(2): 226-241, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33068018

RESUMEN

OBJECTIVE: Epileptic spasms are a hallmark of severe seizure disorders. The neurophysiological mechanisms and the neuronal circuit(s) that generate these seizures are unresolved and are the focus of studies reported here. METHODS: In the tetrodotoxin model, we used 16-channel microarrays and microwires to record electrophysiological activity in neocortex and thalamus during spasms. Chemogenetic activation was used to examine the role of neocortical pyramidal cells in generating spasms. Comparisons were made to recordings from infantile spasm patients. RESULTS: Current source density and simultaneous multiunit activity analyses indicate that the ictal events of spasms are initiated in infragranular cortical layers. A dramatic pause of neuronal activity was recorded immediately prior to the onset of spasms. This preictal pause is shown to share many features with the down states of slow wave sleep. In addition, the ensuing interictal up states of slow wave rhythms are more intense in epileptic than control animals and occasionally appear sufficient to initiate spasms. Chemogenetic activation of neocortical pyramidal cells supported these observations, as it increased slow oscillations and spasm numbers and clustering. Recordings also revealed a ramp-up in the number of neocortical slow oscillations preceding spasms, which was also observed in infantile spasm patients. INTERPRETATION: Our findings provide evidence that epileptic spasms can arise from the neocortex and reveal a previously unappreciated interplay between brain state physiology and spasm generation. The identification of neocortical up states as a mechanism capable of initiating epileptic spasms will likely provide new targets for interventional therapies. ANN NEUROL 2021;89:226-241.


Asunto(s)
Ondas Encefálicas/fisiología , Neocórtex/fisiopatología , Células Piramidales/fisiología , Espasmos Infantiles/fisiopatología , Tálamo/fisiopatología , Animales , Modelos Animales de Enfermedad , Electrocorticografía , Femenino , Humanos , Lactante , Masculino , Neocórtex/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Ratas , Ratas Wistar , Convulsiones/inducido químicamente , Convulsiones/fisiopatología , Bloqueadores de los Canales de Sodio/toxicidad , Espasmo/inducido químicamente , Espasmo/fisiopatología , Espasmos Infantiles/inducido químicamente , Tetrodotoxina/toxicidad , Tálamo/efectos de los fármacos
16.
Neurotox Res ; 38(4): 957-966, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33025361

RESUMEN

Alzheimer's disease (AD) is a multifactorial and severe neurodegenerative disorder characterized by progressive memory decline, the presence of Aß plaques and tau tangles, brain atrophy, and neuronal loss. Available therapies provide moderate symptomatic relief but do not alter disease progression. This study demonstrated that PaPE-1, which has been designed to selectively activate non-nuclear estrogen receptors (ERs), has anti-AD capacity, as evidenced in a cellular model of the disease. In this model, the treatment of mouse neocortical neurons with Aß (5 and 10 µM) induced apoptosis (loss of mitochondrial membrane potential, activation of caspase-3, induction of apoptosis-related genes and proteins) accompanied by increases in levels of reactive oxygen species (ROS) and lactate dehydrogenase (LDH) as well as reduced cell viability. Following 24 h of exposure, PaPE-1 inhibited Aß-evoked effects, as shown by reduced parameters of neurotoxicity, oxidative stress, and apoptosis. Because PaPE-1 downregulated Aß-induced Fas/FAS expression but upregulated that of Aß-induced FasL, the role of PaPE-1 in controlling the external apoptotic pathway is controversial. However, PaPE-1 normalized Aß-induced loss of mitochondrial membrane potential and restored the BAX/BCL2 ratio, suggesting that the anti-AD capacity of PaPE-1 particularly relies on inhibition of the mitochondrial apoptotic pathway. These data provide new evidence for an anti-AD strategy that utilizes the selective targeting of non-nuclear ERs with PaPE-1.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Fosfatidiletanolaminas/administración & dosificación , Receptores de Estrógenos/metabolismo , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/toxicidad , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ratones , Neocórtex/efectos de los fármacos , Neocórtex/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fragmentos de Péptidos/toxicidad
17.
Epilepsia ; 61(12): 2811-2824, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33063874

RESUMEN

OBJECTIVE: To characterize neocortical onset status epilepticus (SE) in the C57BL/6J mouse. METHODS: We induced SE by administering homocysteine 16-18 hours after cobalt (Co) implantation. SE was monitored by video and electroencephalography (EEG). We evaluated brain structure with magnetic resonance imaging (MRI). Neurodegeneration was evaluated 72 hours after SE using Fluoro-Jade C staining. RESULTS: Cobalt triggered seizures in a dose-dependent manner (median effective dose, ED50  = 0.78 mg) and the latency to peak seizure frequency shortened with increased dose. Animals developed SE after homocysteine administration. SE began with early intermittent focal seizures, consisting of frontal onset rhythmic spike-wave discharges manifested as focal dystonia with clonus. These focal seizures then evolved into generalized continuous convulsive activity. Behavioral manifestations of SE included tonic stiffening, bilateral limb clonus, and bilateral tonic-clonic movements, which were accompanied by generalized rhythmic spike-wave discharges on EEG. After prolonged seizures, animals became comatose with intermittent bilateral myoclonic seizures or jerks. During this period, EEG showed seizures interspersed with generalized periodic discharges on a suppressed background. MRI obtained when animals were in a coma revealed edema, midline shift in frontal lobe around the Co implantation site, and ventricular effacement. Fluoro-Jade C staining revealed neurodegeneration in the cortex, amygdala, and thalamus. SIGNIFICANCE: We have developed a mouse model of severe, refractory cortical-onset SE, consisting of convulsions merging into a coma, EEG patterns of cortical seizures, and injury, with evidence of widespread neocortical edema and damage. This model replicates many features of acute seizures and SE resulting from traumatic brain injury, subarachnoid, and lobar hemorrhage.


Asunto(s)
Neocórtex/lesiones , Estado Epiléptico/etiología , Animales , Cobalto/toxicidad , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Electroencefalografía , Femenino , Homocisteína/toxicidad , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Neocórtex/efectos de los fármacos , Neuroimagen , Estado Epiléptico/diagnóstico por imagen , Estado Epiléptico/fisiopatología , Grabación en Video
18.
Neuron ; 108(6): 1113-1129.e6, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33080227

RESUMEN

Evolutionary expansion of the mammalian neocortex (Ncx) has been linked to increased abundance and proliferative capacity of basal progenitors (BPs) in the subventricular zone during development. BP proliferation is governed by both intrinsic and extrinsic signals, several of which have been identified. However, a role of neurotransmitters, a canonical class of extrinsic signaling molecules, in BP proliferation remains to be established. Here, we show that serotonin (5-HT), via its receptor HTR2A, promotes BP proliferation in an evolutionarily relevant manner. HTR2A is not expressed in embryonic mouse Ncx; accordingly, 5-HT does not increase mouse BP proliferation. However, ectopic HTR2A expression can increase mouse BP proliferation. Conversely, CRISPR/Cas9-mediated knockout of endogenous HTR2A in embryonic ferret Ncx reduces BP proliferation. Pharmacological activation of endogenous HTR2A in fetal human Ncx ex vivo increases BP proliferation via HER2/ERK signaling. Hence, 5-HT emerges as an important extrinsic pro-proliferative signal for BPs, which may have contributed to evolutionary Ncx expansion.


Asunto(s)
Proliferación Celular/fisiología , Ventrículos Laterales/citología , Neocórtex/citología , Células-Madre Neurales/citología , Receptor de Serotonina 5-HT2A/metabolismo , Animales , Sistemas CRISPR-Cas , Proliferación Celular/efectos de los fármacos , Hurones , Regulación del Desarrollo de la Expresión Génica , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/metabolismo , Ratones , Neocórtex/efectos de los fármacos , Neocórtex/metabolismo , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neurogénesis/efectos de los fármacos , Neurogénesis/fisiología , Receptor de Serotonina 5-HT2A/genética , Serotonina/farmacología
19.
Epilepsia ; 61(9): e129-e134, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32929741

RESUMEN

Neurosteroids can modulate γ-aminobutyric acid type A receptor-mediated inhibitory currents. Recently, we discovered that the neurosteroids progesterone, 5α-dihydroprogesterone, allopregnanolone, and pregnanolone are reduced in the cerebrospinal fluid of patients with status epilepticus (SE). However, it is undetermined whether neurosteroids influence SE. For this reason, first we evaluated whether the inhibitor of adrenocortical steroid production trilostane (50 mg/kg) could modify the levels of neurosteroids in the hippocampus and neocortex, and we found a remarkable increase in pregnenolone, progesterone, 5α-dihydroprogesterone, and allopregnanolone levels using liquid chromatography tandem mass spectrometry. Second, we characterized the dynamics of SE in the presence of the varied neurosteroidal milieu by a single intraperitoneal kainic acid (KA; 15 mg/kg) injection in trilostane-treated rats and their controls. Convulsions started in advance in the trilostane group, already appearing 90 minutes after the KA injection. In contrast to controls, convulsions prevalently developed as generalized seizures with loss of posture in the trilostane group. However, this effect was transient, and convulsions waned 2 hours before the control group. Moreover, electrocorticographic traces of convulsions were shorter in trilostane-treated rats, especially at the 180-minute (P < .001) and 210-minute (P < .01) time points. These findings indicate that endogenous neurosteroids remarkably modulate SE dynamics.


Asunto(s)
Encéfalo/efectos de los fármacos , Dihidrotestosterona/análogos & derivados , Inhibidores Enzimáticos/farmacología , Neuroesteroides/metabolismo , Estado Epiléptico/metabolismo , Estado Epiléptico/fisiopatología , 5-alfa-Dihidroprogesterona/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Cromatografía Liquida , Dihidrotestosterona/farmacología , Electrocorticografía , Agonistas de Aminoácidos Excitadores/toxicidad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ácido Kaínico/toxicidad , Masculino , Neocórtex/efectos de los fármacos , Neocórtex/metabolismo , Pregnanolona/metabolismo , Pregnenolona/metabolismo , Progesterona/metabolismo , Ratas , Receptores de GABA-A , Estado Epiléptico/inducido químicamente , Espectrometría de Masas en Tándem , Factores de Tiempo
20.
Neurotoxicology ; 80: 41-51, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32561249

RESUMEN

Fumonisin B1 (FB1) is a mycotoxin produced by microscopic fungi (mostly Fusarium species), which may infect our major crops. The toxin inhibits the development of these plants and may also have harmful effects on animals and humans consuming the infected crops. FB1 inhibits sphingolipid biosynthesis which leads to altered membrane characteristics and consequently, altered cellular functions. There are some indications that the toxin has inhibitory effects on neuronal activity in case of repeated consumption, presumably due to sphingolipid depletion. However, according to new literature data, FB1 may have acute excitatory neural effects, too, via different mechanisms of action. Therefore, in the present study, we addressed the neuronal network effects of FB1 following acute treatment, using different electrophysiological techniques in vitro and in vivo. Acute treatments with FB1 (10-100 µM) were carried out on brain slices, tissue cultures and live animals. After direct treatment of samples, electrically evoked or spontaneous field potentials were examined in the hippocampus and the neocortex of rat brain slices and in hippocampal cell cultures. In the hippocampus, a short-term increase in the excitability of neuronal networks and individual cells was observed in response to FB1 treatment. In some cases, the initially enhanced excitation was reversed presumably due to overactivation of neuronal networks. Normal spontaneous activity was found to be stimulated in hippocampal cell cultures. Seizure susceptibility was not affected in the neocortex of brain slices. For the verification of the results caused by direct treatment, effects of systemic administration of FB1 (7.5 mg/kg, i.p.) were also examined. Evoked field potentials recorded in vivo from the somatosensory cortex and cell activation measured by the c-fos technique in hippocampus and somatosensory cortex were analyzed. However, the hippocampal and cortical stimulatory effect detected in vitro could not be demonstrated by these in vivo assays. Altogether, the toxin enhanced the basic excitability of neurons and neuronal networks after direct treatment but there were no effects on the given brain areas after systemic treatment in vivo. Based on the observed in vitro FB1 effects and the lack of data on the penetration of FB1 across the blood-brain barrier, we assume that in vivo consequences of FB1 administration can be more prominent in case of perturbed blood-brain barrier functions.


Asunto(s)
Fumonisinas/toxicidad , Hipocampo/efectos de los fármacos , Neocórtex/efectos de los fármacos , Red Nerviosa/efectos de los fármacos , Neuronas/efectos de los fármacos , Potenciales de Acción , Animales , Células Cultivadas , Hipocampo/metabolismo , Técnicas In Vitro , Masculino , Ratones , Neocórtex/metabolismo , Neuronas/metabolismo , Ratas Wistar , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA