Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
Cells ; 12(9)2023 04 25.
Article in English | MEDLINE | ID: mdl-37174647

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) remains a significant risk factor for post-traumatic epilepsy (PTE). The pathophysiological mechanisms underlying the injury-induced epileptogenesis are under investigation. The dentate gyrus-a structure that is highly susceptible to injury-has been implicated in the evolution of seizure development. METHODS: Utilizing the murine unilateral focal control cortical impact (CCI) injury, we evaluated seizure onset using 24/7 EEG video analysis at 2-4 months post-injury. Cellular changes in the dentate gyrus and hilus of the hippocampus were quantified by unbiased stereology and Imaris image analysis to evaluate Prox1-positive cell migration, astrocyte branching, and morphology, as well as neuronal loss at four months post-injury. Isolation of region-specific astrocytes and RNA-Seq were performed to determine differential gene expression in animals that developed post-traumatic epilepsy (PTE+) vs. those animals that did not (PTE-), which may be associated with epileptogenesis. RESULTS: CCI injury resulted in 37% PTE incidence, which increased with injury severity and hippocampal damage. Histological assessments uncovered a significant loss of hilar interneurons that coincided with aberrant migration of Prox1-positive granule cells and reduced astroglial branching in PTE+ compared to PTE- mice. We uniquely identified Cst3 as a PTE+-specific gene signature in astrocytes across all brain regions, which showed increased astroglial expression in the PTE+ hilus. CONCLUSIONS: These findings suggest that epileptogenesis may emerge following TBI due to distinct aberrant cellular remodeling events and key molecular changes in the dentate gyrus of the hippocampus.


Subject(s)
Brain Injuries, Traumatic , Epilepsy, Post-Traumatic , Mice , Animals , Epilepsy, Post-Traumatic/etiology , Epilepsy, Post-Traumatic/pathology , Gliosis/complications , Brain Injuries, Traumatic/complications , Seizures , Interneurons/metabolism
2.
Int J Mol Sci ; 23(22)2022 Nov 16.
Article in English | MEDLINE | ID: mdl-36430654

ABSTRACT

Sucrose consumption impairs behavioral and cognitive functions that correlate with decreased neurogenesis in animal models. When consumed during early adolescence, this disaccharide promotes anxious and depressive behaviors, along with a reduction in the generation of new neurons in the dentate gyrus of the hippocampus. Data concerning sucrose consumption during late adolescence are lacking, and the effect of sucrose intake on the ventral dentate gyrus of the hippocampus (which modulates anxiety and depression) remains elusive. Here, we tested whether sucrose intake during late adolescence causes anxiety or impaired neurogenesis in the ventral dentate gyrus. Rats did not display anxiety-like behaviors neither at the light−dark box test nor at the open field exploration. However, there was a significant increase in proliferative cells in the subgranular zone of the ventral dentate gyrus in rats exposed to sucrose (p < 0.05). This increased proliferation corresponded to neural stem cells (Radial Type 1 cells) in the group exposed to sucrose until adulthood but was not present in rats exposed to sucrose only during late adolescence. Remarkably, the phosphorylation of ERK1/2 kinases was increased in the hippocampi of rats exposed to sucrose only during late adolescence, suggesting that the increased proliferation in this group could be mediated by the MAPK pathway. On the other hand, although no differences were found in the number of immature granular neurons, we observed more immature granular neurons with impaired dendritic orientation in both groups exposed to sucrose. Finally, GAD65/67 and BCL2 levels did not change between groups, suggesting an unaltered hippocampal GABAergic system and similar apoptosis, respectively. This information provides the first piece of evidence of how sucrose intake, starting in late adolescence, impacts ventral dentate gyrus neurogenesis and contributes to a better understanding of the effects of this carbohydrate on the brain at postnatal stages.


Subject(s)
Dentate Gyrus , Neural Stem Cells , Rats , Animals , Dentate Gyrus/metabolism , Sucrose/metabolism , Neurogenesis/physiology , Neural Stem Cells/metabolism , Anxiety
3.
Bull Exp Biol Med ; 168(6): 802-806, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32350713

ABSTRACT

The deposition of beta-amyloid (Aß) in the brain is detected in Alzheimer's disease and during ageing. Until now, ultrastructural studies of changes caused by Aß in the dentate gyrus are very scarce. The effects of Aß 1-42 injection into the CA1 field of rat hippocampus were studied by electron microscopy. In 2 weeks after injection of aggregated Aß in low concentrations, destructive changes were seen in the structure of dentate gyrus cells, which consisted in a decrease in the number of dentate gyrus neurons and axo-dendritic synapses. These changes were accompanied by enlargement of the endoplasmic reticulum cisterns and widening of the active zones of synapses. Thus, injection of aggregated Aß 1-42 into the hippocampus led to irreversible (a decrease in the number of neurons and axo-dendritic synapses, agglutination of synthetic vesicles) and adaptive changes (an increase in the sizes of endoplasmic reticulum cisterns and active zones of synapses) in dentate gyrus neurons aimed at the maintenance of functional activity of the nervous system.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/administration & dosage , CA1 Region, Hippocampal/ultrastructure , Dentate Gyrus/ultrastructure , Neurons/ultrastructure , Peptide Fragments/administration & dosage , Synapses/ultrastructure , Alzheimer Disease/chemically induced , Amyloid beta-Peptides/chemistry , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/pathology , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/pathology , Cytoplasmic Granules/ultrastructure , Dentate Gyrus/drug effects , Dentate Gyrus/pathology , Disease Models, Animal , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum/ultrastructure , Injections, Intraventricular , Lipofuscin/chemistry , Male , Microscopy, Electron , Neurons/drug effects , Neurons/pathology , Peptide Fragments/chemistry , Protein Aggregates , Rats , Rats, Wistar , Synapses/drug effects , Synapses/pathology
4.
Neurosci Lett ; 694: 192-197, 2019 02 16.
Article in English | MEDLINE | ID: mdl-30528876

ABSTRACT

The inability of central nervous system (CNS) to regenerate following traumatic brain injury (TBI) can be attributed to apoptotic cell death, inhibitory extrinsic environment, and the limited ability of neurons to regenerate. Thus, fostering the intrinsic regenerative potential and minimizing neuronal cell death could be a promising therapeutic approach. Pyrroloquinoline quinone (PQQ) was previously reported for its neuroprotective and regenerative potential on peripheral nerves. Here, we investigated the ability of PQQ to induce neurite re-growth in a wound healing model on cultured cerebellar granular neurons (CGNs), an integral part of cerebellar circuitry, as one of the most affected areas following TBI. The neuroprotective effect was also examined utilizing K+/FCS deprivation-induced apoptosis model in CGNs. Resveratrol (RVT), an effective promoter of neuroprotection and regeneration both centrally and peripherally was also investigated separately and in combination with PQQ to establish a possible synergistic effect. RVT (5 µM) and PQQ (0.5 µM) showed a tendency to promote neurite re-growth in the wound healing assay, however the effect observed was statistically insignificant. Higher concentrations of PQQ (1 and 2 µM) were found to be less effective. Resveratrol did not affect neurite length in CGNs culture; however, it did significantly increase the number of viable CGNs. For the neuroprotective effect; PQQ and RVT showed a significant increase in the survival of CGNs following K+/FCS deprivation of culture. Thus, both compounds showed a tendency to support neurite outgrowth in addition to a significant neuroprotective effect, but no synergistic effect was detected.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Cerebellum/drug effects , Neuronal Outgrowth/drug effects , Neuroprotective Agents/administration & dosage , PQQ Cofactor/administration & dosage , Resveratrol/administration & dosage , Animals , Brain Injuries, Traumatic/physiopathology , Cell Survival/drug effects , Cells, Cultured , Cerebellum/physiopathology , Mice , Wound Healing/drug effects
5.
J Exp Pharmacol ; 8: 43-52, 2016.
Article in English | MEDLINE | ID: mdl-27785113

ABSTRACT

BACKGROUND: Recent evidence suggests that many individuals who chew khat recreationally also drink ethanol to offset the stimulating effect of khat. The objective of this study was to describe the separate and interactive effects of chronic ethanol and khat exposure on key projection neurons in the cortex and hippocampus of young adult male rats. METHODS: Young adult male Sprague Dawley rats were divided into six treatment groups: 2 g/kg khat, 4 g/kg khat, 4 g/kg ethanol, combined khat and ethanol (4 g/kg each), a normal saline control, and an untreated group. Treatments were administered orally for 28 continuous days; brains were then harvested, sectioned, and routine hematoxylin-eosin staining was done. Following photomicrography, ImageJ® software captured data regarding neuron number and size. RESULTS: No differences occurred in counts of both granular and pyramidal projection neurons in the motor cortex and all four subfields of the hippocampal formation. Khat dose-dependently increased pyramidal neuron size in the motor cortex and the CA3 region, but had different effects on granular neuron size in the dentate gyrus and the motor cortex. Mean pyramidal neuron size for the ethanol-only treatment was larger than that for the 2 g/kg khat group, and the saline control group, in CA3 and in the motor cortex. Concomitant khat and ethanol increased granular neuron size in the motor cortex, compared to the 2 g/kg khat group, the 4 g/kg khat group, and the 4 g/kg ethanol group. In the CA3 region, the 4 g/kg ethanol group showed a larger mean pyramidal neuron size than the combined khat and ethanol group. CONCLUSION: These results suggest that concomitant khat and ethanol exposure changes granular and pyramidal projection neuron sizes differentially in the motor cortex and hippocampus, compared to the effects of chronic exposure to these two drugs separately.

6.
Biochem J ; 473(6): 779-87, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26772870

ABSTRACT

An γ-aminobutyric acid type B (GABAB)-receptor mediates slow and prolonged synaptic inhibition in the central nervous system, which represents an interesting target for the treatment of various diseases and disorders of the central nervous system. To date, only one activator of the GABAB-receptor, baclofen, is on the market for the treatment of spasticity. Inhibitors of the GABAB-receptor, such as antagonists, show anti-absence seizure activity and pro-cognitive properties. In a search for allosteric compounds of the GABAB-receptor, although several positive allosteric modulators have been developed, it is only recently that the first negative allosteric modulator (NAM), CLH304a (also named Compound 14), has been reported. In the present study, we provide further information on the mechanism of action of CLH304a, and also show the possibility of designing more NAMs, such as CLH391 and CLH393, based on the structure of CLH304a. First we show that CLH304a inhibits native GABAB-receptor activity in cultured cerebellar granular neurons. We then show that CLH304a has inverse agonist properties and non-competitively inhibits the effect of agonists, indicating that it binds at a different site to GABA. The GABAB-receptor is a mandatory heterodimer made of GB1 subunits, in which agonists bind, and GB2 subunits, which activate G-proteins. By using various combinations made up of wild-type and/or mutated GB1 and GB2 subunits, we show that CLH304a acts on the heptahelical domain of GB2 subunits. These data revealed the possibility of designing innovative NAMs acting in the heptahelical domain of the GB2 subunits, offering novel possibilities for therapeutic intervention based on GABAB-receptor inhibition.


Subject(s)
GABA Agonists/pharmacology , Keto Acids/pharmacology , Phenols/pharmacology , Receptors, GABA-B/metabolism , Signal Transduction , Allosteric Regulation , Animals , Binding Sites , GABA Agonists/chemistry , Gene Expression Regulation , HEK293 Cells , Humans , Keto Acids/chemistry , Mice , Models, Molecular , Molecular Structure , Phenols/chemistry , Protein Conformation , Protein Subunits , Receptors, GABA-B/genetics , Structure-Activity Relationship
7.
Iran J Pharm Res ; 14(4): 1123-35, 2015.
Article in English | MEDLINE | ID: mdl-26664379

ABSTRACT

Lithium (Li), a glycogen synthase kinase-3ß (GSK-3ß) inhibitor, has used to attenuate the cannabinoid-induced dependence/withdrawal signs, but molecular mechanisms related to this are unclear. Recent studies indicate the involvement of upstream extracellular signal kinase1/2 (ERK1/2) and downstream GSK-3ß pathways in the development of cannabinoid-induced dependence. This is mediated through cannabinoid receptor 1 (CB1) enriched in cerebellar granular neurons (CGNs). Accordingly, the present study aimed to investigate the mechanism of modulatory/neuroprotective effects of Li on a cannabinoid agonist (WIN 55,212-2 (WIN))-induced dependence, through quantitative analysis of some involved proteins such as ERK1/2, GSK-3ß and related signaling pathways including their phosphorylated forms; and cAMP level as the other molecular mechanisms leading to dependence, in CGNs model. The CGNs were prepared from 7-day-old Wistar rat pup in a 12-well plate, pretreated with Li (1mM) and an ERK1/2 inhibitor SL327 (SL, 10 µM). The WIN (1 µM) was added 30 minutes prior to treatment and AM251 (AM, 1 µM), as a cannabinoid antagonist was co-treated with WIN. The cAMP level, as an indicator of cannabinoid-induced dependence, was measured by ELISA following forskolin (FSK) stimulation. Western blot analyses determined the phosphorylated forms of ERK1/2 (p-ERK1/2), GSK-3ß (p-GSK-3ß) as well as their total expressions in various treatment times and doses in CGNs. WIN alone could down regulate the cAMP/p-ERK1/2 cascade compared to AM treatment. However, P-GSK-3ß was up-regulated with Li and WIN or with SL and Li pretreatment to AM-induced cellular response, which was the highest 60 minutes after CGNs exposure. Results further suggested the potential role of Li pretreatment to diminish the development of cannabinoid-induced dependence/neuronal injury through possible mechanisms of modulating the cAMP/p-ERK1/2 cascade independent of p-GSK-3ß signaling pathway in-vitro.

8.
Neurobiol Dis ; 68: 215-27, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24769160

ABSTRACT

G-protein coupled receptor 3 (GPR3), GPR6, and GPR12 belong to a family of constitutively active Gs-coupled receptors that activate 3'-5'-cyclic adenosine monophosphate (cAMP) and are highly expressed in the brain. Among these receptors, the endogenous expression of GPR3 in cerebellar granule neurons (CGNs) is increased following development. GPR3 is important for neurite outgrowth and neural maturation; however, the physiological functions of GPR3 remain to be fully elucidated. Here, we investigated the survival and antiapoptotic functions of GPR3 under normal and apoptosis-inducing culture conditions. Under normal culture conditions, CGNs from GPR3-knockout mice demonstrated lower survival than did CGNs from wild-type or GPR3-heterozygous mice. Cerebellar sections from GPR3-/- mice at P7, P14, and P21 revealed more caspase-3-positive neurons in the internal granular layer than in cerebellar sections from wild-type mice. Conversely, in a potassium-deprivation model of apoptosis, increased expression of these three receptors promoted neuronal survival. The antiapoptotic effect of GPR3 was also observed under hypoxic (1% O2/5% CO2) and reactive oxygen species (ROS)-induced apoptotic conditions. We further investigated the signaling pathways involved in the GPR3-mediated antiapoptotic effect. The addition of the PKA inhibitor KT5720, the MAP kinase inhibitor U0126, and the PI3 kinase inhibitor LY294002 abrogated the GPR3-mediated antiapoptotic effect in a potassium-deprivation model of apoptosis, whereas the PKC inhibitor Gö6976 did not affect the antiapoptotic function of GPR3. Furthermore, downregulation of endogenous GPR3 expression in CGNs resulted in a marked reduction in the basal levels of ERK and Akt phosphorylation under normal culture conditions. Finally, we used a transient middle cerebral artery occlusion (tMCAO) model in wild-type and GPR3-knockout mice to determine whether GPR3 expression modulates neuronal survival after brain ischemia. After tMCAO, GPR3-knockout mice exhibited a significantly larger infarct area than did wild-type mice. Collectively, these in vitro and in vivo results suggest that the developmental expression of constitutively active Gs-coupled GPR3 activates the ERK and Akt signaling pathways at the basal level, thereby protecting neurons from apoptosis that is induced by various stimuli.


Subject(s)
Apoptosis/genetics , Cerebellum/cytology , Gene Expression Regulation, Developmental/genetics , Neurons/metabolism , Receptors, G-Protein-Coupled/deficiency , Age Factors , Animals , Cardiotonic Agents/pharmacology , Cell Survival/genetics , Colforsin/pharmacology , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Developmental/drug effects , Ischemia/metabolism , Ischemia/pathology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
9.
Hippocampus ; 24(3): 257-69, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24132937

ABSTRACT

In the context of Alzheimer's disease (AD), hippocampal alterations have been well described in advanced stages of the pathology, when amyloid deposition, inflammation and glial activation occur, but less attention has been directed to studying early brain and behavioral changes. Using an animal model of AD, the transgenic PDAPP-J20 mouse at 5 months of age, when no amyloid plaques are present and low cerebral levels of amyloid peptides are detectable, we found structural, morphological, and cellular alterations in the hippocampus. Young transgenic mice showed a reduced hippocampal volume with less number of pyramidal and granular neurons, which additionally exhibited cell atrophy. The neurogenic capability in this zone, measured as DCX+ cells, was strongly diminished and associated to alterations in cell maturity. A decrease in presynaptic synaptophysin optical density was detected in mossy fibers reaching CA3 subfield but not in Golgi stained- CA1 dendritic spine density. Employing confocal microscopy and accurate stereological tools we also found a reduction in the number of GFAP+ cells, along with decreased astrocyte complexity, suggesting a potential detriment of neural support. According with untimely neuroglial alterations, young PDAPP mice failed in the novel location recognition test, that depends on hippocampal function. Moreover, multivariate statistical analysis of the behavioral outcome in the open-field test evidenced an elevated anxiety score in Tg mice compared with age-matched control mice. In line with this, the transgenic group showed a higher number of c-Fos+ nuclei in central and basolateral amygdala, a result that supports the early involvement of the emotionality factor in AD pathology. Applying an integrative approach, this work focuses on early structural, morphological and functional changes and provides new and compelling evidence of behavioral alterations that precede manifest AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid/chemistry , Anxiety/etiology , Astrocytes/pathology , Hippocampus/pathology , Memory Disorders/etiology , Neurons/pathology , Alzheimer Disease/psychology , Amygdala/pathology , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/genetics , Animals , Anxiety/pathology , Atrophy , Disease Models, Animal , Disease Progression , Doublecortin Protein , Exploratory Behavior , Hippocampus/chemistry , Humans , Memory Disorders/pathology , Mice , Mice, Transgenic , Nerve Tissue Proteins/analysis , Peptide Fragments/analysis , Peptide Fragments/genetics , Plaque, Amyloid/pathology , Proto-Oncogene Proteins c-fos/analysis , Recombinant Fusion Proteins/analysis , Spatial Behavior , Synaptophysin/analysis
SELECTION OF CITATIONS
SEARCH DETAIL