Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 809
Filter
1.
Chemosphere ; 358: 142124, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677614

ABSTRACT

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.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase 5 , Metformin , Neocortex , Metformin/pharmacology , Animals , Female , Pregnancy , Neocortex/drug effects , Cyclin-Dependent Kinase 5/metabolism , Rats , Cell Proliferation/drug effects , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , MAP Kinase Signaling System/drug effects , Neurons/drug effects , Rats, Sprague-Dawley , Cell Differentiation/drug effects , Neurogenesis/drug effects , Cell Movement/drug effects , Apoptosis/drug effects , Signal Transduction/drug effects
2.
eNeuro ; 11(5)2024 May.
Article in English | MEDLINE | ID: mdl-38664009

ABSTRACT

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.


Subject(s)
4-Aminopyridine , Neocortex , Transcriptome , Animals , Neocortex/metabolism , Neocortex/drug effects , Female , Male , Mice , 4-Aminopyridine/pharmacology , Seizures/genetics , Seizures/metabolism , Seizures/physiopathology , Sequence Analysis, RNA/methods , Epilepsy/genetics , Epilepsy/metabolism , Epilepsy/physiopathology , Mice, Inbred C57BL
3.
Neurochem Int ; 152: 105251, 2022 01.
Article in English | MEDLINE | ID: mdl-34861326

ABSTRACT

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.


Subject(s)
Annexin A1/metabolism , Dementia/metabolism , Neocortex/metabolism , Parkinson Disease/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Anti-Inflammatory Agents/pharmacology , Biomarkers/blood , Dementia/drug therapy , Female , Humans , Male , Middle Aged , Neocortex/drug effects , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Parkinson Disease/drug therapy , Parkinson Disease/pathology
4.
Exp Neurol ; 347: 113916, 2022 01.
Article in English | MEDLINE | ID: mdl-34752784

ABSTRACT

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.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , Hippocampus/metabolism , Neocortex/metabolism , Pilocarpine/toxicity , Receptors, Glutamate/biosynthesis , Temporal Lobe/metabolism , Animals , Dose-Response Relationship, Drug , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/pathology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Gene Expression , Hippocampus/drug effects , Hippocampus/pathology , Male , Neocortex/drug effects , Neocortex/pathology , Rats , Rats, Sprague-Dawley , Receptors, Glutamate/genetics , Temporal Lobe/drug effects , Temporal Lobe/pathology
5.
Development ; 148(17)2021 09 01.
Article in English | MEDLINE | ID: mdl-34414407

ABSTRACT

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.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Dendrites/metabolism , Extracellular Matrix Proteins/metabolism , Interneurons/cytology , Neocortex/growth & development , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Animals , Calbindin 2/metabolism , Calcium/metabolism , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/pharmacology , Dendrites/drug effects , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/pharmacology , Hypertrophy , Interneurons/drug effects , Interneurons/metabolism , Mice , Mice, Knockout , Neocortex/cytology , Neocortex/drug effects , Neocortex/pathology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/pharmacology , Neuropeptide Y/metabolism , Parvalbumins/metabolism , Receptors, GABA-B/metabolism , Receptors, Glutamate/metabolism , Reelin Protein , Serine Endopeptidases/deficiency , Serine Endopeptidases/pharmacology
6.
Sci Data ; 8(1): 180, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34267214

ABSTRACT

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.


Subject(s)
Brain Waves , Memory Consolidation , Neocortex/physiology , Neurons/physiology , Sleep , Analgesics/pharmacology , Anesthesia , Animals , Female , Ketamine/pharmacology , Neocortex/cytology , Neocortex/drug effects , Rats , Rats, Wistar , Xylazine/pharmacology
7.
Neuropharmacology ; 196: 108676, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34216585

ABSTRACT

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.


Subject(s)
Alzheimer Disease/diagnostic imaging , Amyloid beta-Peptides/metabolism , Brain/diagnostic imaging , Monoamine Oxidase/metabolism , Plaque, Amyloid/diagnostic imaging , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Aniline Compounds , Animals , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Hippocampus/diagnostic imaging , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Mice, Transgenic , Monoamine Oxidase Inhibitors/pharmacology , Neocortex/diagnostic imaging , Neocortex/drug effects , Neocortex/metabolism , Plaque, Amyloid/metabolism , Positron-Emission Tomography , Presenilin-1/genetics , Quinolines , Radiopharmaceuticals , Selegiline/pharmacology , Thalamus/diagnostic imaging , Thalamus/drug effects , Thalamus/metabolism
8.
Neurobiol Learn Mem ; 183: 107484, 2021 09.
Article in English | MEDLINE | ID: mdl-34175450

ABSTRACT

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.


Subject(s)
Acetylcholine/metabolism , Basal Nucleus of Meynert/physiology , Cholinergic Neurons/physiology , Mental Recall/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Antibodies, Monoclonal/pharmacology , Basal Nucleus of Meynert/drug effects , Basal Nucleus of Meynert/metabolism , Cholinergic Agents/pharmacology , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Cholinesterase Inhibitors/pharmacology , Donepezil/pharmacology , Mental Recall/drug effects , Neocortex/drug effects , Neocortex/metabolism , Neocortex/physiology , Rats , Saporins/pharmacology
9.
Brain Res ; 1759: 147370, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33600830

ABSTRACT

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.


Subject(s)
Inflammation Mediators/immunology , Interneurons/immunology , Neuroimmunomodulation/physiology , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/immunology , Animals , Female , Glutamate Decarboxylase/deficiency , Glutamate Decarboxylase/genetics , Hippocampus/drug effects , Hippocampus/immunology , Hippocampus/metabolism , Inflammation Mediators/metabolism , Interferon Inducers/toxicity , Interneurons/drug effects , Interneurons/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neocortex/drug effects , Neocortex/immunology , Neocortex/metabolism , Neuroimmunomodulation/drug effects , Poly I-C/toxicity , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism
11.
Ann Neurol ; 89(2): 226-241, 2021 02.
Article in English | MEDLINE | ID: mdl-33068018

ABSTRACT

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.


Subject(s)
Brain Waves/physiology , Neocortex/physiopathology , Pyramidal Cells/physiology , Spasms, Infantile/physiopathology , Thalamus/physiopathology , Animals , Disease Models, Animal , Electrocorticography , Female , Humans , Infant , Male , Neocortex/drug effects , Pyramidal Cells/drug effects , Rats , Rats, Wistar , Seizures/chemically induced , Seizures/physiopathology , Sodium Channel Blockers/toxicity , Spasm/chemically induced , Spasm/physiopathology , Spasms, Infantile/chemically induced , Tetrodotoxin/toxicity , Thalamus/drug effects
12.
J Comp Neurol ; 529(7): 1628-1641, 2021 05 01.
Article in English | MEDLINE | ID: mdl-32975324

ABSTRACT

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.


Subject(s)
Neocortex/physiology , Proto-Oncogene Proteins c-fos/metabolism , Animals , Callithrix , GABA Antagonists/pharmacology , Male , Neocortex/drug effects , Pentylenetetrazole/pharmacology , Rats , Rats, Wistar
13.
Neurotox Res ; 38(4): 957-966, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33025361

ABSTRACT

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.


Subject(s)
Alzheimer Disease/metabolism , Drug Delivery Systems/methods , Phosphatidylethanolamines/administration & dosage , Receptors, Estrogen/metabolism , Alzheimer Disease/chemically induced , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/toxicity , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Mice , Neocortex/drug effects , Neocortex/metabolism , Neurons/drug effects , Neurons/metabolism , Peptide Fragments/toxicity
14.
Neuron ; 108(6): 1113-1129.e6, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33080227

ABSTRACT

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.


Subject(s)
Cell Proliferation/physiology , Lateral Ventricles/cytology , Neocortex/cytology , Neural Stem Cells/cytology , Receptor, Serotonin, 5-HT2A/metabolism , Animals , CRISPR-Cas Systems , Cell Proliferation/drug effects , Ferrets , Gene Expression Regulation, Developmental , Lateral Ventricles/drug effects , Lateral Ventricles/metabolism , Mice , Neocortex/drug effects , Neocortex/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurogenesis/physiology , Receptor, Serotonin, 5-HT2A/genetics , Serotonin/pharmacology
15.
Epilepsia ; 61(12): 2811-2824, 2020 12.
Article in English | MEDLINE | ID: mdl-33063874

ABSTRACT

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.


Subject(s)
Neocortex/injuries , Status Epilepticus/etiology , Animals , Cobalt/toxicity , Disease Models, Animal , Dose-Response Relationship, Drug , Electroencephalography , Female , Homocysteine/toxicity , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Neocortex/drug effects , Neuroimaging , Status Epilepticus/diagnostic imaging , Status Epilepticus/physiopathology , Video Recording
16.
Epilepsia ; 61(9): e129-e134, 2020 09.
Article in English | MEDLINE | ID: mdl-32929741

ABSTRACT

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.


Subject(s)
Brain/drug effects , Dihydrotestosterone/analogs & derivatives , Enzyme Inhibitors/pharmacology , Neurosteroids/metabolism , Status Epilepticus/metabolism , Status Epilepticus/physiopathology , 5-alpha-Dihydroprogesterone/metabolism , Animals , Brain/metabolism , Brain/physiopathology , Chromatography, Liquid , Dihydrotestosterone/pharmacology , Electrocorticography , Excitatory Amino Acid Agonists/toxicity , Hippocampus/drug effects , Hippocampus/metabolism , Kainic Acid/toxicity , Male , Neocortex/drug effects , Neocortex/metabolism , Pregnanolone/metabolism , Pregnenolone/metabolism , Progesterone/metabolism , Rats , Receptors, GABA-A , Status Epilepticus/chemically induced , Tandem Mass Spectrometry , Time Factors
17.
Neurotoxicology ; 80: 41-51, 2020 09.
Article in English | MEDLINE | ID: mdl-32561249

ABSTRACT

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.


Subject(s)
Fumonisins/toxicity , Hippocampus/drug effects , Neocortex/drug effects , Nerve Net/drug effects , Neurons/drug effects , Action Potentials , Animals , Cells, Cultured , Hippocampus/metabolism , In Vitro Techniques , Male , Mice , Neocortex/metabolism , Neurons/metabolism , Rats, Wistar , Time Factors
18.
CNS Neurol Disord Drug Targets ; 19(4): 264-275, 2020.
Article in English | MEDLINE | ID: mdl-32496992

ABSTRACT

BACKGROUND: Mammalian central neurons regulate their intracellular pH (pHi) strongly and even slight pHi-fluctuations can influence inter-/intracellular signaling, synaptic plasticity and excitability. OBJECTIVE: For the first time, we investigated topiramate´s (TPM) influence on pHi-behavior of human central neurons representing a promising target for anticonvulsants and antimigraine drugs. METHODS: In slice-preparations of tissue resected from the middle temporal gyrus of five adults with intractable temporal lobe epilepsy, BCECF-AM-loaded neocortical pyramidal-cells were investigated by fluorometry. The pHi-regulation was estimated by using the recovery-slope from intracellular acidification after an Ammonium-Prepulse (APP). RESULTS: Among 17 pyramidal neurons exposed to 50 µM TPM, seven (41.24%) responded with an altered resting-pHi (7.02±0.12), i.e., acidification of 0.01-0.03 pH-units. The more alkaline the neurons, the greater the TPM-related acidifications (r=0.7, p=0.001, n=17). The recovery from APPacidification was significantly slowed under TPM (p<0.001, n=5). Further experiments using nominal bicarbonate-free (n=2) and chloride-free (n=2) conditions pointed to a modulation of the HCO3 -- driven pHi-regulation by TPM, favoring a stimulation of the passive Cl-/HCO3 --antiporter (CBT) - an acid-loader predominantly in more alkaline neurons. CONCLUSION: TPM modulated the bicarbonate-driven pHi-regulation, just as previously described in adult guinea-pig hippocampal neurons. We discussed the significance of the resulting subtle acidifications for beneficial antiepileptic, antimigraine and neuroprotective effects as well as for unwanted cognitive deficits.


Subject(s)
Acid-Base Equilibrium/drug effects , Anticonvulsants/pharmacology , Bicarbonates/metabolism , Chloride-Bicarbonate Antiporters/drug effects , Hydrogen-Ion Concentration , Neocortex/drug effects , Pyramidal Cells/drug effects , Topiramate/pharmacology , Adult , Chloride-Bicarbonate Antiporters/metabolism , Epilepsy, Temporal Lobe/surgery , Female , Fluorometry , Hippocampus/pathology , Humans , Male , Malformations of Cortical Development , Neocortex/chemistry , Neocortex/cytology , Neocortex/metabolism , Neurons/chemistry , Neurons/drug effects , Neurons/metabolism , Pyramidal Cells/chemistry , Pyramidal Cells/metabolism , Sclerosis , Temporal Lobe/chemistry , Temporal Lobe/cytology , Temporal Lobe/drug effects , Temporal Lobe/metabolism , Young Adult
19.
Epilepsia ; 61(6): e60-e65, 2020 06.
Article in English | MEDLINE | ID: mdl-32385935

ABSTRACT

Because oral pharmacological treatment of neocortical focal epilepsy is limited due to common systemic side effects and relatively low drug concentrations reached at the epileptic foci locally, application of antiepileptic agents directly onto the neocortical focus may enhance treatment tolerability and efficacy. We describe the effects of cortically applied sodium valproate (VPA) in two patients with pharmacoresistant neocortical focal epilepsy who were selected for epilepsy surgery after a circumscribed epileptic focus had been determined by invasive presurgical evaluation using subdural electrodes. Local VPA modified epileptic activity as electrocorticographically recorded from the chronic focus in both patients. In addition, VPA induced local increase of the inhibitory neurotransmitter γ-aminobutyric acid (GABA) in cortical tissue samples, whereas the excitatory glutamate was possibly decreased. In this clinical pilot study, we could show antiepileptic effects of cortically applied VPA in humans by electrocorticographic and neurochemical parameters.


Subject(s)
Anticonvulsants/administration & dosage , Drug Resistant Epilepsy/physiopathology , Electrocorticography/methods , Epilepsies, Partial/physiopathology , Neocortex/physiopathology , Valproic Acid/administration & dosage , Drug Resistant Epilepsy/drug therapy , Electrocorticography/drug effects , Epilepsies, Partial/drug therapy , Female , Humans , Intraoperative Neurophysiological Monitoring/methods , Male , Middle Aged , Neocortex/chemistry , Neocortex/drug effects , Valproic Acid/therapeutic use
20.
Bull Exp Biol Med ; 168(4): 521-524, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32152848

ABSTRACT

The effect of glyproline-containing peptide MGHPPGP (Met-Glu-His-Phe-Pro-Gly-Pro) was studied in experiments on male Wistar rats with modeled traumatic brain injury. The peptide was administered intraperitoneally in a dose of 1 mg/kg in 3 h and on days 2, 3, 4, 5 after injury. We evaluated morphometric parameters of the epithelial cells of the tongue, small intestine, and liver cells (AgNOR staining), neuronal layers II and V of the neocortex of the parietal lobe and hippocampal CA1 field (staining with gallocyanin) were evaluated in the post-traumatic period. Traumatic brain injury (TBI) was induced in rats by using the impact model (WDM; weight drop method). MGHPPGP peptide corrected the activity indicators of the nuclear organizer regions in epitheliocytes of the tongue.


Subject(s)
Brain Injuries, Traumatic/drug therapy , CA1 Region, Hippocampal/drug effects , Epithelial Cells/drug effects , Neocortex/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oligopeptides/pharmacology , Animals , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Epithelial Cells/ultrastructure , Hepatocytes/drug effects , Hepatocytes/ultrastructure , Injections, Intraperitoneal , Intestine, Small/drug effects , Liver/drug effects , Male , Neurons/ultrastructure , Parietal Lobe/drug effects , Rats , Rats, Wistar , Tongue/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...