Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 373
Filter
1.
Int J Mol Sci ; 22(24)2021 Dec 19.
Article in English | MEDLINE | ID: mdl-34948401

ABSTRACT

Theta oscillations generated in hippocampal (HPC) and cortical neuronal networks are involved in various aspects of brain function, including sensorimotor integration, movement planning, memory formation and attention. Disruptions of theta rhythms are present in individuals with brain disorders, including epilepsy and Alzheimer's disease. Theta rhythm generation involves a specific interplay between cellular (ion channel) and network (synaptic) mechanisms. HCN channels are theta modulators, and several medications are known to enhance their activity. We investigated how different doses of lamotrigine (LTG), an HCN channel modulator, and antiepileptic and neuroprotective agent, would affect HPC theta rhythms in acute HPC slices (in vitro) and anaesthetized rats (in vivo). Whole-cell patch clamp recordings revealed that LTG decreased GABAA-fast transmission in CA3 cells, in vitro. In addition, LTG directly depressed CA3 and CA1 pyramidal neuron excitability. These effects were partially blocked by ZD 7288, a selective HCN blocker, and are consistent with decreased excitability associated with antiepileptic actions. Lamotrigine depressed HPC theta oscillations in vitro, also consistent with its neuronal depressant effects. In contrast, it exerted an opposite, enhancing effect, on theta recorded in vivo. The contradictory in vivo and in vitro results indicate that LTG increases ascending theta activating medial septum/entorhinal synaptic inputs that over-power the depressant effects seen in HPC neurons. These results provide new insights into LTG actions and indicate an opportunity to develop more precise therapeutics for the treatment of dementias, memory disorders and epilepsy.


Subject(s)
Action Potentials/drug effects , Anticonvulsants/pharmacology , Hippocampus/drug effects , Lamotrigine/pharmacology , Theta Rhythm/drug effects , Animals , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/physiology , Hippocampus/cytology , Hippocampus/physiology , Male , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Wistar , Synapses/drug effects , Synapses/physiology , gamma-Aminobutyric Acid/metabolism
2.
PLoS Comput Biol ; 17(10): e1009435, 2021 10.
Article in English | MEDLINE | ID: mdl-34597293

ABSTRACT

In the hippocampus, episodic memories are thought to be encoded by the formation of ensembles of synaptically coupled CA3 pyramidal cells driven by sparse but powerful mossy fiber inputs from dentate gyrus granule cells. The neuromodulators acetylcholine and noradrenaline are separately proposed as saliency signals that dictate memory encoding but it is not known if they represent distinct signals with separate mechanisms. Here, we show experimentally that acetylcholine, and to a lesser extent noradrenaline, suppress feed-forward inhibition and enhance Excitatory-Inhibitory ratio in the mossy fiber pathway but CA3 recurrent network properties are only altered by acetylcholine. We explore the implications of these findings on CA3 ensemble formation using a hierarchy of models. In reconstructions of CA3 pyramidal cells, mossy fiber pathway disinhibition facilitates postsynaptic dendritic depolarization known to be required for synaptic plasticity at CA3-CA3 recurrent synapses. We further show in a spiking neural network model of CA3 how acetylcholine-specific network alterations can drive rapid overlapping ensemble formation. Thus, through these distinct sets of mechanisms, acetylcholine and noradrenaline facilitate the formation of neuronal ensembles in CA3 that encode salient episodic memories in the hippocampus but acetylcholine selectively enhances the density of memory storage.


Subject(s)
Acetylcholine/pharmacology , CA3 Region, Hippocampal , Memory , Norepinephrine/pharmacology , Animals , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/physiology , Computational Biology , Memory/drug effects , Memory/physiology , Mice , Mice, Inbred C57BL , Models, Neurological , Neuronal Plasticity/drug effects , Neurons/drug effects , Pyramidal Cells/drug effects
3.
Cell Rep ; 36(7): 109513, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34407417

ABSTRACT

Ketamine produces rapid antidepressant action in patients with major depression or treatment-resistant depression. Studies have identified brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin receptor kinase B (TrkB), as necessary for the antidepressant effects and underlying ketamine-induced synaptic potentiation in the hippocampus. Here, we delete BDNF or TrkB in presynaptic CA3 or postsynaptic CA1 regions of the Schaffer collateral pathway to investigate the rapid antidepressant action of ketamine. The deletion of Bdnf in CA3 or CA1 blocks the ketamine-induced synaptic potentiation. In contrast, ablation of TrkB only in postsynaptic CA1 eliminates the ketamine-induced synaptic potentiation. We confirm BDNF-TrkB signaling in CA1 is required for ketamine's rapid behavioral action. Moreover, ketamine application elicits dynamin1-dependent TrkB activation and downstream signaling to trigger rapid synaptic effects. Taken together, these data demonstrate a requirement for BDNF-TrkB signaling in CA1 neurons in ketamine-induced synaptic potentiation and identify a specific synaptic locus in eliciting ketamine's rapid antidepressant effects.


Subject(s)
Antidepressive Agents/pharmacology , Ketamine/pharmacology , Receptor, trkB/metabolism , Signal Transduction , Synapses/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/metabolism , Dynamins/metabolism , Endocytosis/drug effects , HEK293 Cells , Humans , Mice , Neurons/metabolism , Signal Transduction/drug effects , Synapses/drug effects
4.
Neurosci Lett ; 763: 136181, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34416345

ABSTRACT

Motopsin is a serine protease that plays a crucial role in synaptic functions. Loss of motopsin function causes severe intellectual disability in humans. In this study, we evaluated the role of motopsin in the neuropathological development of cognitive impairments following chemotherapy, also known as chemobrain. Motopsin knockout (KO) and wild-type (WT) mice were intravenously injected with doxorubicin (Dox) or saline four times every 8 days and were evaluated for open field, novel object recognition, and passive avoidance tests. Parvalbumin-positive neurons in the hippocampus were immunohistochemically analyzed. Dox administration significantly decreased the total distance in the open field test in both WT and motopsin KO mice without affecting the duration spent in the center square. A significant interaction between the genotype and drug treatment was detected in the recognition index (the rate to investigate a novel object) in the novel object recognition test, although Dox treatment did not affect the total investigation time. Additionally, Dox treatment significantly decreased the recognition index in WT mice, whereas it tended to increase the recognition index in motopsin KO mice. Dox treatment did not affect the latency to enter a dark compartment in either WT or motopsin KO mice in the passive avoidance test. Interestingly, Dox treatment increased the parvalbumin-positive neurons in the stratum oriens of the hippocampus CA1 region of only WT mice, not motopsin KO mice. Our data suggest that motopsin deficiency imparted partial insensitivity to Dox-induced hippocampal impairments. Alternatively, motopsin may contribute to the neuropathology of chemobrain.


Subject(s)
CA1 Region, Hippocampal/pathology , CA3 Region, Hippocampal/pathology , Chemotherapy-Related Cognitive Impairment/pathology , Doxorubicin/adverse effects , Serine Endopeptidases/deficiency , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/drug effects , Chemotherapy-Related Cognitive Impairment/etiology , Disease Models, Animal , Humans , Locomotion/drug effects , Male , Mice , Mice, Knockout , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/pathology , Neurons/drug effects , Neurons/pathology , Parvalbumins/metabolism , Serine Endopeptidases/genetics
5.
Bull Exp Biol Med ; 171(3): 327-332, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34297297

ABSTRACT

We studied the prolonged action of kainic acid on glutamatergic neurons in the dorsal hippocampus and the endocannabinoid-dependent protection against neurodegeneration. The pyramidal neurons of the CA3 field of the hippocampus, as well as granular and mossy cells of the dentate gyrus were examined. Light and electron microscopy revealed substantial damage to the components of the protein-synthesizing (rough endoplasmic reticulum, Golgi apparatus, and polyribosomes) and catabolic (lysosomes, autophagosomes, multivesicular structures, and lipofuscin formations) systems in all cells. Pyramidal and mossy neurons die mainly by the necrotic pathway. The death of granular cells occurred through both apoptosis and necrosis. The most vulnerable cells are mossy neurons located in the hilus. Activation of the endocannabinoid system induced by intracerebral injection of URB597, an inhibitor of degradation of endocannabinoid anandamide, protected the normal structure of the hippocampus and prevented neuronal damage and death induced by KA.


Subject(s)
Arachidonic Acids/metabolism , Endocannabinoids/metabolism , Excitatory Amino Acid Agonists/pharmacology , Kainic Acid/pharmacology , Nerve Degeneration/pathology , Polyunsaturated Alkamides/metabolism , Pyramidal Cells/drug effects , Status Epilepticus/pathology , Animals , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Benzamides/pharmacology , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/pathology , Carbamates/pharmacology , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/ultrastructure , Male , Microscopy, Electron , Necrosis/metabolism , Necrosis/pathology , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Rats , Rats, Wistar , Status Epilepticus/chemically induced , Status Epilepticus/metabolism
6.
Int J Mol Sci ; 22(9)2021 May 01.
Article in English | MEDLINE | ID: mdl-34062911

ABSTRACT

Hypoxic-ischemic encephalopathy (HIE) remains to be a major cause of long-term neurodevelopmental deficits in term neonates. Hypothermia offers partial neuroprotection warranting research for additional therapies. Kynurenic acid (KYNA), an endogenous product of tryptophan metabolism, was previously shown to be beneficial in rat HIE models. We sought to determine if the KYNA analog SZR72 would afford neuroprotection in piglets. After severe asphyxia (pHa = 6.83 ± 0.02, ΔBE = -17.6 ± 1.2 mmol/L, mean ± SEM), anesthetized piglets were assigned to vehicle-treated (VEH), SZR72-treated (SZR72), or hypothermia-treated (HT) groups (n = 6, 6, 6; Tcore = 38.5, 38.5, 33.5 °C, respectively). Compared to VEH, serum KYNA levels were elevated, recovery of EEG was faster, and EEG power spectral density values were higher at 24 h in the SZR72 group. However, instantaneous entropy indicating EEG signal complexity, depression of the visual evoked potential (VEP), and the significant neuronal damage observed in the neocortex, the putamen, and the CA1 hippocampal field were similar in these groups. In the caudate nucleus and the CA3 hippocampal field, neuronal damage was even more severe in the SZR72 group. The HT group showed the best preservation of EEG complexity, VEP, and neuronal integrity in all examined brain regions. In summary, SZR72 appears to enhance neuronal activity after asphyxia but does not ameliorate early neuronal damage in this HIE model.


Subject(s)
Asphyxia Neonatorum/drug therapy , Brain Ischemia/drug therapy , Kynurenic Acid/analogs & derivatives , Neurons/metabolism , Animals , Asphyxia Neonatorum/metabolism , Asphyxia Neonatorum/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , CA1 Region, Hippocampal/diagnostic imaging , CA1 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/diagnostic imaging , CA3 Region, Hippocampal/drug effects , Disease Models, Animal , Electroencephalography , Evoked Potentials, Visual/drug effects , Humans , Kynurenic Acid/pharmacology , Neurons/drug effects , Neurons/pathology , Rats , Translational Research, Biomedical
7.
Int J Mol Sci ; 22(9)2021 May 04.
Article in English | MEDLINE | ID: mdl-34064311

ABSTRACT

Dephosphorylation of target proteins at serine/threonine residues is one of the most crucial mechanisms regulating their activity and, consequently, the cellular functions. The role of phosphatases in synaptic plasticity, especially in long-term depression or depotentiation, has been reported. We studied serine/threonine phosphatase activity during the protein synthesis blocker (PSB)-induced impairment of long-term potentiation (LTP). Established protein phosphatase 2B (PP2B, calcineurin) inhibitor cyclosporin A prevented the LTP early phase (E-LTP) decline produced by pretreatment of hippocampal slices with cycloheximide or anisomycin. For the first time, we directly measured serine/threonine phosphatase activity during E-LTP, and its significant increase in PSB-treated slices was demonstrated. Nitric oxide (NO) donor SNAP also heightened phosphatase activity in the same manner as PSB, and simultaneous application of anisomycin + SNAP had no synergistic effect. Direct measurement of the NO production in hippocampal slices by the NO-specific fluorescent probe DAF-FM revealed that PSBs strongly stimulate the NO concentration in all studied brain areas: CA1, CA3, and dentate gyrus (DG). Cyclosporin A fully abolished the PSB-induced NO production in the hippocampus, suggesting a close relationship between nNOS and PP2B activity. Surprisingly, cyclosporin A alone impaired short-term plasticity in CA1 by decreasing paired-pulse facilitation, which suggests bi-directionality of the influences of PP2B in the hippocampus. In conclusion, we proposed a minimal model of signaling events that occur during LTP induction in normal conditions and the PSB-treated slices.


Subject(s)
CA1 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/metabolism , Calcineurin/genetics , Long-Term Potentiation/genetics , Synaptic Potentials/genetics , Animals , Anisomycin/pharmacology , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/drug effects , Calcineurin/metabolism , Calcineurin Inhibitors/pharmacology , Cycloheximide/pharmacology , Cyclosporine/pharmacology , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Gene Expression Regulation , Long-Term Potentiation/drug effects , Male , Microtomy , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Nitric Oxide/chemistry , Nitric Oxide/pharmacology , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Wistar , S-Nitroso-N-Acetylpenicillamine/chemistry , S-Nitroso-N-Acetylpenicillamine/pharmacology , Synaptic Potentials/drug effects , Tissue Culture Techniques
8.
Molecules ; 26(6)2021 Mar 13.
Article in English | MEDLINE | ID: mdl-33805696

ABSTRACT

Experimental evidence indicates that the activation of ionotropic glutamate receptors plays an important role in neurological disorders' models such as epilepsy, cerebral ischemia and trauma. The glutamate receptor agonist kainic acid (KA) induces seizures and excitotoxic cell death in the CA3 region of the hippocampus. Thymoquinone (TQ) is the most important component of the essential oil obtained from black cumin (Nigella sativa L.) seeds. It has many pharmacological actions including antioxidant, anti-inflammatory, and anti-apoptotic effects. TQ was used in an in vitro experimental model of primary cultures where excitotoxicity was induced. Briefly, rat organotypic hippocampal slices were exposed to 5 µM KA for 24 h. Cell death in the CA3 subregions of slices was quantified by measuring propidium iodide fluorescence. The cross-talk between TQ, ER stress and apoptotic pathways was investigated by Western blot. In untreated slices TQ (10 µM) induced a significant increase on the PSD95 levels and it decreased the excitotoxic injury induced by KA. Additionally, TQ was able to ameliorate the KA-induced increase in unfolded proteins GRP78 and GRP94 expression. Finally, TQ was able to partially rescue the reduction of the KA-induced apoptotic pathway activation. Our results suggest that TQ modulates the processes leading to post-kainate neuronal death in the CA3 hippocampal area.


Subject(s)
Benzoquinones/pharmacology , CA3 Region, Hippocampal/drug effects , Neuroprotective Agents/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , CA3 Region, Hippocampal/pathology , CA3 Region, Hippocampal/physiopathology , Disease Models, Animal , Disks Large Homolog 4 Protein/metabolism , Endoplasmic Reticulum Stress/drug effects , Epilepsy/chemically induced , Epilepsy/drug therapy , Epilepsy/physiopathology , Excitatory Amino Acid Agonists/toxicity , Female , In Vitro Techniques , Kainic Acid/toxicity , Male , Neuronal Plasticity/drug effects , Rats , Rats, Wistar
9.
PLoS One ; 16(3): e0240074, 2021.
Article in English | MEDLINE | ID: mdl-33711021

ABSTRACT

Cholinergic modulation of hippocampal network function is implicated in multiple behavioral and cognitive states. Activation of nicotinic and muscarinic acetylcholine receptors affects neuronal excitability, synaptic transmission and rhythmic oscillations in the hippocampus. In this work, we studied the ability of the cholinergic system to sustain hippocampal epileptiform activity independently from glutamate and GABA transmission. Simultaneous CA3 and CA1 field potential recordings were obtained during the perfusion of hippocampal slices with the aCSF containing AMPA, NMDA and GABA receptor antagonists. Under these conditions, spontaneous epileptiform discharges synchronous between CA3 and CA1 were recorded. Epileptiform discharges were blocked by addition of the calcium-channel blocker Cd2+ and disappeared in CA1 after a surgical cut between CA3 and CA1. Cholinergic antagonist mecamylamine abolished CA3-CA1 synchronous epileptiform discharges, while antagonists of α7 and α4ß2 nAChRs, MLA and DhßE, had no effect. Our results suggest that activation of nicotinic acetylcholine receptors can sustain CA3-CA1 synchronous epileptiform activity independently from AMPA, NMDA and GABA transmission. In addition, mecamylamine, but not α7 and α4ß2 nAChRs antagonists, reduced bicuculline-induced seizure-like activity. The ability of mecamylamine to decrease hippocampal network synchronization might be associated with its therapeutic effects in a wide variety of CNS disorders including addiction, depression and anxiety.


Subject(s)
CA1 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/drug effects , Mecamylamine/pharmacology , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/metabolism , Animals , Bicuculline/pharmacology , CA1 Region, Hippocampal/physiology , CA3 Region, Hippocampal/physiology , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , In Vitro Techniques , Mecamylamine/therapeutic use , Nicotinic Antagonists/therapeutic use , Patch-Clamp Techniques , Rats , Rats, Wistar , Receptors, Nicotinic/chemistry , Seizures/prevention & control , Seizures/veterinary , Synaptic Transmission/drug effects
10.
Biomed Pharmacother ; 135: 111230, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33434853

ABSTRACT

Diabetes mellitus is mainly associated with degeneration of the central nervous system, which eventually leads to cognitive deficit. Although some studies suggest that exercise can improve the cognitive decline associated with diabetes, the potential effects of endurance training (ET) accompanied by Matricaria chamomilla (M.ch) flowers extract on cognitive impairment in type 2 diabetes has been poorly understood. Forty male Wistar rats were randomized into 5 equal groups of 8: healthy-sedentary (H-sed), diabetes-sedentary (D-sed), diabetes-endurance training (D-ET), diabetes-Matricaria chamomilla. (D-M.ch), and diabetes-endurance training-Matricaria chamomilla. (D-ET-M.ch). Nicotinamide (110 mg/kg, i.p.) and Streptozotocin (65 mg/kg, i.p.) were utilized to initiate type 2 diabetes. Then, ET (5 days/week) and M.ch (200 mg/kg body weight/daily) were administered for 12 weeks. After 12 weeks of the experiment, cognitive functions were assessed using the Morris Water Maze (MWM) test and a passive avoidance paradigm using a shuttle box device. Subsequently, using crystal violet staining, neuron necrosis was examined in the CA3 area of the hippocampus. Diabetic rats showed cognitive impairment following an increase in the number of necrotic cells in region CA3 of the hippocampal tissue. Also, diabetes increased serum levels of lipid peroxidation and decreased total antioxidant capacity in serum and hippocampal tissue. ET + M.ch treatment prevented the necrosis of neurons in the hippocampal tissue. Following positive changes in hippocampal tissue and serum antioxidant enzyme levels, an improvement was observed in the cognitive impairment of the diabetic rats receiving ET + M.ch. Therefore the results showed that treatment with ET + M.ch could ameliorate memory and inactive avoidance in diabetic rats. Hence, the use of ET + M.ch interventions is proposed as a new therapeutic perspective on the death of hippocampal neurons and cognitive deficit caused by diabetes.


Subject(s)
Behavior, Animal/drug effects , CA3 Region, Hippocampal/drug effects , Cognition/drug effects , Cognitive Dysfunction/prevention & control , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 2/therapy , Endurance Training , Matricaria , Physical Conditioning, Animal , Plant Extracts/pharmacology , Animals , CA3 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/pathology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Cognitive Dysfunction/psychology , Combined Modality Therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/psychology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/psychology , Flowers , Lipid Peroxidation , Male , Matricaria/chemistry , Morris Water Maze Test/drug effects , Necrosis , Oxidative Stress/drug effects , Plant Extracts/isolation & purification , Rats, Wistar
11.
Neuroreport ; 32(4): 306-311, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33470771

ABSTRACT

Fast ripples (FRs) are found in the hippocampus of epileptic brains, and this fast electrical activity has been described as a biomarker of the epileptogenic process itself. Results from our laboratory, such as the observation of decreased seizure rates and FR incidence at a specific citalopram dose, have suggested that serotonin (5-HT) may play a key role in the FR generation process. Therefore, to gather more details about the state of the serotoninergic system in the hippocampus under an epileptogenic process, we studied the immunoreactivity of three 5-HT receptors (5-HT1A, 5-HT2 and 5-HT7) as well as the extracellular levels of 5-HT in the hippocampal tissue of epileptic rats with FR. Wistar rats (210-300 g) were injected with a single dose of pilocarpine hydrochloride (2.4 mg/2 µl) in the right lateral ventricle and video-monitored 24 h/d to detect spontaneous and recurrent seizures; microelectrodes were implanted in the dentate gyrus (DG) and CA3 and CA1 regions of these rats ipsilateral to the pilocarpine injection site 1 day after the first spontaneous seizure was observed, and only rats who suffered FR events were used in this work. Thirty-three days after the first spontaneous seizure, an immunostaining procedure and high performance liquid chromatography were performed to measure the 5-HT levels. A general depletion of the 5-HT and 5-HIIA levels in hippocampal tissue from epileptic animals compared with those in controls was observed; in addition, a general decrease in immunoreactivity for the three receptors was found, especially in the DG, which may support the establishment of an excitatory/inhibitory imbalance in the trisynaptic circuit that underlies the FR generation process.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , Hippocampus/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Serotonin, 5-HT2/metabolism , Receptors, Serotonin/metabolism , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/metabolism , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Disease Models, Animal , Epilepsy, Temporal Lobe/chemically induced , Hippocampus/drug effects , Hydroxyindoleacetic Acid/metabolism , Immunohistochemistry , Muscarinic Agonists/toxicity , Pilocarpine/toxicity , Rats , Receptor, Serotonin, 5-HT1A/drug effects , Receptors, Serotonin/drug effects , Receptors, Serotonin, 5-HT2/drug effects , Serotonin/metabolism
12.
Behav Brain Res ; 397: 112933, 2021 01 15.
Article in English | MEDLINE | ID: mdl-32991927

ABSTRACT

Increasing evidence shows the close relationship between hippocampal glutamatergic and serotonergic systems through the modulation of behavioral responses. This study aimed to investigate the possible involvement of 5-HT4 receptors in the CA3 hippocampal region in anxiolytic-like effects induced by D-AP5 (a competitive antagonist of the glutamate NMDA [N-Methyl-D-aspartate] receptor). Male Wistar rats were placed in the elevated plus maze (EPM) apparatus that is used to assess anxiety-related behaviors, and the percentages of open arm time (%OAT) and open arm entries (%OAE) which are associated with anxiety-related behaviors were measured. The close arm entries (CAE) which is correlated with locomotor activity was also evaluated. The results showed that, intra-CA3 injection of D-AP5 (0.4 µg/rat), RS67333 (1.2 µg/rat; a 5-HT4 receptor agonist), and RS23597-190 (1.2 µg/rat; a 5-HT4 receptor antagonist) increased %OAT and %OAE, indicating the anxiolytic-like effect of these drugs. Also, only RS23597-190 (1.2 µg/rat) decreased CAE. Intra-CA3 injection of sub-threshold dose of RS67333 (0.012 µg/rat) or RS23597-190 (0.012 µg/rat), 5 min before the injection of D-AP5 (0.2 µg/rat) increased %OAT, indicating potentiating the anxiolytic-like effect of D-AP5. The isobolographic analyses also showed the additive or synergistic anxiolytic-like effect of intra-CA3 co-administration of D-AP5 with RS67333 or RS23597-190, respectively. In conclusion, CA3 5-HT4 receptors are involved in D-AP5-induced anxiolytic-like behaviors in rats.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , CA3 Region, Hippocampal/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Serotonin 5-HT4 Receptor Agonists/pharmacology , Serotonin 5-HT4 Receptor Antagonists/pharmacology , Aniline Compounds/pharmacology , Animals , Anti-Anxiety Agents/administration & dosage , Behavior, Animal/drug effects , Disease Models, Animal , Drug Interactions , Drug Therapy, Combination , Excitatory Amino Acid Antagonists/administration & dosage , Locomotion/drug effects , Male , Piperidines/pharmacology , Rats , Rats, Wistar , Receptors, Serotonin, 5-HT4 , Serotonin 5-HT4 Receptor Agonists/administration & dosage , Serotonin 5-HT4 Receptor Antagonists/administration & dosage
13.
J Chem Neuroanat ; 113: 101837, 2021 04.
Article in English | MEDLINE | ID: mdl-32534024

ABSTRACT

Amyloid ß-peptides (Aß) are considered as a major hallmark of Alzheimer's disease (AD) that can induce synaptic loss and apoptosis in brain regions, particularly in the cortex and the hippocampus. Evidence suggests that crocin, as the major component of saffron, can exhibit neuromodulatory effects in AD. However, specific data related to their efficacy to attenuate the synaptic loss and neuronal death in animal models of AD are limited. Hence, we investigated the efficacy of crocin in the CA3 and dentate gyrus (DG) regions of the hippocampus and also in frontal cortex neurons employing a rat model of AD. Male Wistar rats were randomly divided into control, sham, AD model, crocin, and AD model + crocin groups, with 8 rats per group. AD model was established by injecting Aß1-42 into the frontal cortex rats, and thereafter the rats were administrated by crocin (30 mg/kg) for a duration of 12-day. The number of live cells, neuronal arborization and apoptosis were measured using a Cresyl violet, Golgi-Cox and TUNEL staining, respectively. Results showed that, the number of live cells in the hippocampus pyramidal neurons in the CA3 and granular cells in the DG regions of the AD rats significantly decreased, which was significantly rescued by crocin. Compared with the control group, the axonal, spine and dendrites arborization in the frontal cortex and CA3 region of the AD model group significantly decreased. The crocin could significantly reverse this arborization loss in the AD rats (P < 0.05). The apoptotic cell number in the CA3 and DG regions in the AD model group was significantly higher than that of the control group (P < 0.05), while crocin significantly decreased the apoptotic cell number in the AD group (P < 0.05). Conclusion. Crocin can improve the synaptic loss and neuronal death of the AD rats possibly by reducing the neuronal apoptosis.


Subject(s)
Alzheimer Disease/pathology , CA3 Region, Hippocampal/drug effects , Carotenoids/pharmacology , Dentate Gyrus/drug effects , Frontal Lobe/drug effects , Pyramidal Cells/drug effects , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Apoptosis/drug effects , CA3 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/pathology , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Disease Models, Animal , Frontal Lobe/metabolism , Frontal Lobe/pathology , Male , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Rats , Rats, Wistar
14.
Neural Plast ; 2020: 8869526, 2020.
Article in English | MEDLINE | ID: mdl-33381164

ABSTRACT

Recent evidence indicates that soluble amyloid-ß (Aß) species induce imbalances in excitatory and inhibitory transmission, resulting in neural network functional impairment and cognitive deficits during early stages of Alzheimer's disease (AD). To evaluate the in vivo effects of two soluble Aß species (Aß 25-35 and Aß 1-40) on commissural CA3-to-CA1 (cCA3-to-CA1) synaptic transmission and plasticity, and CA1 oscillatory activity, we used acute intrahippocampal microinjections in adult anaesthetized male Wistar rats. Soluble Aß microinjection increased cCA3-to-CA1 synaptic variability without significant changes in synaptic efficiency. High-frequency CA3 stimulation was rendered inefficient by soluble Aß intrahippocampal injection to induce long-term potentiation and to enhance synaptic variability in CA1, contrasting with what was observed in vehicle-injected subjects. Although soluble Aß microinjection significantly increased the relative power of γ-band and ripple oscillations and significantly shifted the average vector of θ-to-γ phase-amplitude coupling (PAC) in CA1, it prevented θ-to-γ PAC shift induced by high-frequency CA3 stimulation, opposite to what was observed in vehicle-injected animals. These results provide further evidence that soluble Aß species induce synaptic dysfunction causing abnormal synaptic variability, impaired long-term plasticity, and deviant oscillatory activity, leading to network activity derailment in the hippocampus.


Subject(s)
Amyloid beta-Peptides/pharmacology , Brain Waves/drug effects , CA1 Region, Hippocampal/diagnostic imaging , CA3 Region, Hippocampal/drug effects , Neuronal Plasticity/drug effects , Peptide Fragments/pharmacology , Synapses/drug effects , Animals , Electric Stimulation , Male , Neural Pathways/drug effects , Neurons/drug effects , Rats , Rats, Wistar , Synaptic Transmission/drug effects
15.
PLoS One ; 15(11): e0242309, 2020.
Article in English | MEDLINE | ID: mdl-33180836

ABSTRACT

Neuronal activity within the physiologic range stimulates lactate production that, via metabolic pathways or operating through an array of G-protein-coupled receptors, regulates intrinsic excitability and synaptic transmission. The recent discovery that lactate exerts a tight control of ion channels, neurotransmitter release, and synaptic plasticity-related intracellular signaling cascades opens up the possibility that lactate regulates synaptic potentiation at central synapses. Here, we demonstrate that extracellular lactate (1-2 mM) induces glutamatergic potentiation on the recurrent collateral synapses of hippocampal CA3 pyramidal cells. This potentiation is independent of lactate transport and further metabolism, but requires activation of NMDA receptors, postsynaptic calcium accumulation, and activation of a G-protein-coupled receptor sensitive to cholera toxin. Furthermore, perfusion of 3,5- dihydroxybenzoic acid, a lactate receptor agonist, mimics this form of synaptic potentiation. The transduction mechanism underlying this novel form of synaptic plasticity requires G-protein ßγ subunits, inositol-1,4,5-trisphosphate 3-kinase, PKC, and CaMKII. Activation of these signaling cascades is compartmentalized in a synapse-specific manner since lactate does not induce potentiation at the mossy fiber synapses of CA3 pyramidal cells. Consistent with this synapse-specific potentiation, lactate increases the output discharge of CA3 neurons when recurrent collaterals are repeatedly activated during lactate perfusion. This study provides new insights into the cellular mechanisms by which lactate, acting via a membrane receptor, contributes to the memory formation process.


Subject(s)
CA3 Region, Hippocampal/physiology , Excitatory Postsynaptic Potentials/drug effects , Lactic Acid/pharmacology , Synapses/metabolism , Animals , CA3 Region, Hippocampal/drug effects , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cholera Toxin/pharmacology , Male , Neuronal Plasticity , Oxamic Acid/pharmacology , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction
16.
Int J Mol Sci ; 21(20)2020 Oct 14.
Article in English | MEDLINE | ID: mdl-33066466

ABSTRACT

Major depressive disorder is typically treated with selective serotonin reuptake inhibitors (SSRIs), however, SSRIs take approximately six weeks to produce therapeutic effects, if any. Not surprisingly, there has been great interest in findings that low doses of ketamine, a non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist, produce rapid and long-lasting antidepressant effects. Preclinical studies show that the antidepressant-like effects of ketamine are dependent upon availability of serotonin, and that ketamine increases extracellular serotonin, yet the mechanism by which this occurs is unknown. Here we examined the role of the high-affinity, low-capacity serotonin transporter (SERT), and the plasma membrane monoamine transporter (PMAT), a low-affinity, high-capacity transporter for serotonin, as mechanisms contributing to ketamine's ability to increase extracellular serotonin and produce antidepressant-like effects. Using high-speed chronoamperometry to measure real-time clearance of serotonin from CA3 region of hippocampus in vivo, we found ketamine robustly inhibited serotonin clearance in wild-type mice, an effect that was lost in mice constitutively lacking SERT or PMAT. As expected, in wild-type mice, ketamine produced antidepressant-like effects in the forced swim test. Mapping onto our neurochemical findings, the antidepressant-like effects of ketamine were lost in mice lacking SERT or PMAT. Future research is needed to understand how constitutive loss of either SERT or PMAT, and compensation that occurs in other systems, is sufficient to void ketamine of its ability to inhibit serotonin clearance and produce antidepressant-like effects. Taken together with existing literature, a critical role for serotonin, and its inhibition of uptake via SERT and PMAT, cannot be ruled out as important contributing factors to ketamine's antidepressant mechanism of action. Combined with what is already known about ketamine's action at NMDA receptors, these studies help lead the way to the development of drugs that lack ketamine's abuse potential but have superior efficacy in treating depression.


Subject(s)
Antidepressive Agents/pharmacology , Equilibrative Nucleoside Transport Proteins/metabolism , Ketamine/pharmacology , Serotonin Plasma Membrane Transport Proteins/metabolism , Animals , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/metabolism , Equilibrative Nucleoside Transport Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics
17.
Int J Mol Sci ; 21(20)2020 Oct 20.
Article in English | MEDLINE | ID: mdl-33092287

ABSTRACT

The neurosteroid allopregnanolone (AL) has many beneficial functions in the brain. This study tested the hypothesis that AL administered for three days into the third brain ventricle would affect the enzymatic activity of the DNA base excision repair (BER) pathway in the hippocampal CA1 and CA3 fields and the central amygdala in luteal-phase sheep under both natural and stressful conditions. Acute stressful stimuli, including isolation and partial movement restriction, were used on the last day of infusion. The results showed that stressful stimuli increased N-methylpurine DNA glycosylase (MPG), thymine DNA glycosylase (TDG), 8-oxoguanine glycosylase (OGG1), and AP-endonuclease 1 (APE1) mRNA expression, as well as repair activities for 1,N6-ethenoadenine (εA), 3,N4-ethenocytosine (εC), and 8-oxoguanine (8-oxoG) compared to controls. The stimulated events were lower in stressed and AL-treated sheep compared to sheep that were only stressed (except MPG mRNA expression in the CA1 and amygdala, as well as TDG mRNA expression in the CA1). AL alone reduced mRNA expression of all DNA repair enzymes (except TDG in the amygdala) relative to controls and other groups. DNA repair activities varied depending on the tissue-AL alone stimulated the excision of εA in the amygdala, εC in the CA3 and amygdala, and 8-oxoG in all tissues studied compared to controls. However, the excision efficiency of lesioned bases in the AL group was lower than in the stressed and stressed and AL-treated groups, with the exception of εA in the amygdala. In conclusion, the presented modulating effect of AL on the synthesis of BER pathway enzymes and their repair capacity, both under natural and stressful conditions, indicates another functional role of this neurosteroid in brain structures.


Subject(s)
Amygdala/drug effects , CA1 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/drug effects , DNA Repair/genetics , Gene Expression Regulation, Enzymologic/drug effects , Pregnanolone/pharmacology , Amygdala/enzymology , Amygdala/metabolism , Animals , CA1 Region, Hippocampal/enzymology , CA1 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/enzymology , CA3 Region, Hippocampal/metabolism , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Female , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sheep , Thymine DNA Glycosylase/genetics , Thymine DNA Glycosylase/metabolism
18.
Toxicology ; 442: 152542, 2020 09.
Article in English | MEDLINE | ID: mdl-32735850

ABSTRACT

Heavy metal neurotoxicity is one of the major challenges in today's era due to the large scale and widespread mechanisation of the production. However, the causative factors responsible for neurotoxicity are neither known nor do we have the availability of therapeutic approaches to deal with it. One of the major causative agents of neurotoxicity is a non-essential transition heavy metal, Cadmium (Cd), that reaches the central nervous system (CNS) through the nasal mucosa and olfactory pathway causing adverse structural and functional effects. In this study, we explored the neuroprotective efficacy of plant derived Curcumin which is reported to have pleiotropic biological activity including anti-oxidant, anti-inflammatory, anti-apoptotic, anti-carcinogenic and anti-angiogenic effects. Four different concentrations of curcumin (20, 40, 80 and 160 mg/kg of the body weight) were used to assess the behavioural, biochemical, hippocampal proteins (BDNF, CREB, DCX and Synapsin II) and histological changes in Swiss Albino mice that were pre-treated with Cd (2.5 mg/kg). The findings showed that Cd exposure led to the behavioural impairment through oxidative stress, reduction of hippocampal neurogenesis associated proteins, and degeneration of CA3 and cortical neurons. However, treatment of different curcumin concentrations had effectively restored the behavioural changes in Cd-exposed mice through regulation of oxidative stress and up-regulation of hippocampal proteins in a dose-dependent manner. Significantly, a dose of 160 mg/kg body weight was found to be glaringly effective. From this study, we infer that curcumin reverses the adverse effects of neurotoxicity induced by Cd and promotes neurogenesis.


Subject(s)
Brain-Derived Neurotrophic Factor/drug effects , Cadmium Poisoning/prevention & control , Curcumin/pharmacology , Cyclic AMP Response Element-Binding Protein/drug effects , Hippocampus/drug effects , Neurogenesis/drug effects , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/prevention & control , Signal Transduction/drug effects , Animals , Anxiety/chemically induced , Anxiety/prevention & control , Anxiety/psychology , Behavior, Animal/drug effects , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/drug effects , Cadmium Poisoning/psychology , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Doublecortin Protein , Hippocampus/cytology , Hippocampus/metabolism , Maze Learning/drug effects , Mice , Neurotoxicity Syndromes/psychology , Oxidative Stress/drug effects , Psychomotor Performance/drug effects
19.
Curr Biol ; 30(18): 3556-3569.e5, 2020 09 21.
Article in English | MEDLINE | ID: mdl-32707066

ABSTRACT

Prenatal alcohol exposure (PAE) leads to profound deficits in spatial memory and synaptic and cellular alterations to the hippocampus that last into adulthood. Neurons in the hippocampus called place cells discharge as an animal enters specific places in an environment, establish distinct ensemble codes for familiar and novel places, and are modulated by local theta rhythms. Spatial memory is thought to critically depend on the integrity of hippocampal place cell firing. Therefore, we tested the hypothesis that hippocampal place cell firing is impaired after PAE by performing in vivo recordings from the hippocampi (CA1 and CA3) of moderate PAE and control adult rats. Our results show that hippocampal CA3 neurons from PAE rats have reduced spatial tuning. Second, CA1 and CA3 neurons from PAE rats are less likely to orthogonalize their firing between directions of travel on a linear track and between changes in contextual stimuli in an open arena compared to control neurons. Lastly, reductions in the number of hippocampal place cells exhibiting significant theta rhythmicity and phase precession were observed, which may suggest changes to hippocampal microcircuit function. Together, the reduced spatial tuning and sensitivity to contextual changes provide a neural systems-level mechanism to explain spatial memory impairment after moderate PAE.


Subject(s)
Action Potentials , Alcohol Drinking/adverse effects , CA1 Region, Hippocampal/pathology , CA3 Region, Hippocampal/pathology , Maternal Exposure/adverse effects , Prenatal Exposure Delayed Effects/pathology , Theta Rhythm/drug effects , Animals , CA1 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/drug effects , Female , Male , Neurons/drug effects , Neurons/pathology , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Rats , Rats, Long-Evans , Spatial Memory
20.
Can J Physiol Pharmacol ; 98(10): 725-732, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32516556

ABSTRACT

Levetiracetam (LEV) has been demonstrated to improve cognitive function. Hippocampal theta rhythm (4-12 Hz) is associated with a variety of cognitively related behaviors, such as exploration in both humans and animal models. We investigated the effects of LEV on the theta rhythm in the rat hippocampal CA3 in hippocampal slices in vitro. We found that LEV increased the theta power in a dose-dependent manner. The increase in theta power can be blocked by GABAA receptor (GABAAR) or NMDA receptor (NMDAR) antagonists but not by AMPA receptor antagonist, indicating the involvement of GABAAR and NMDAR in the induction of theta activity. Interestingly, LEV enhancement of theta power can be also blocked by taurine or GABA-A agonist THIP, indicating that LEV induction of theta may be related to the indirect boosting of GABA action via reduction of extrasynaptic GABAAR activation. Furthermore, the increased theta power can be partially reduced by the mACh receptor (mAChR) antagonist atropine but not by nACh receptor antagonists, suggesting that mAChR activation provides excitatory input into local network responsible for LEV-induced theta. Our study demonstrated that LEV induced a novel theta oscillation in vitro, which may have implications in the treatment of the neuronal disorders with impaired theta oscillation and cognitive function.


Subject(s)
CA3 Region, Hippocampal/drug effects , Levetiracetam/pharmacology , Theta Rhythm/drug effects , Animals , CA3 Region, Hippocampal/metabolism , In Vitro Techniques , Male , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Receptors, Muscarinic/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...