Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 373
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
J Neurochem ; 155(1): 62-80, 2020 10.
Article in English | MEDLINE | ID: mdl-32491248

ABSTRACT

Alzheimer's disease (AD) is the leading type of dementia worldwide. With an increasing burden of an aging population coupled with the lack of any foreseeable cure, AD warrants the current intense research effort on the toxic effects of an increased concentration of beta-amyloid (Aß) in the brain. Glutamate is the main excitatory brain neurotransmitter and it plays an essential role in the function and health of neurons and neuronal excitability. While previous studies have shown alterations in expression of glutamatergic signaling components in AD, the underlying mechanisms of these changes are not well understood. This is the first comprehensive anatomical study to characterize the subregion- and cell layer-specific long-term effect of Aß1-42 on the expression of specific glutamate receptors and transporters in the mouse hippocampus, using immunohistochemistry with confocal microscopy. Outcomes are examined 30 days after Aß1-42 stereotactic injection in aged male C57BL/6 mice. We report significant decreases in density of the glutamate receptor subunit GluA1 and the vesicular glutamate transporter (VGluT) 1 in the conus ammonis 1 region of the hippocampus in the Aß1-42 injected mice compared with artificial cerebrospinal fluid injected and naïve controls, notably in the stratum oriens and stratum radiatum. GluA1 subunit density also decreased within the dentate gyrus dorsal stratum moleculare in Aß1-42 injected mice compared with artificial cerebrospinal fluid injected controls. These changes are consistent with findings previously reported in the human AD hippocampus. By contrast, glutamate receptor subunits GluA2, GluN1, GluN2A, and VGluT2 showed no changes in expression. These findings indicate that Aß1-42 induces brain region and layer specific expression changes of the glutamatergic receptors and transporters, suggesting complex and spatial vulnerability of this pathway during development of AD neuropathology. Read the Editorial Highlight for this article on page 7. Cover Image for this issue: https://doi.org/10.1111/jnc.14763.


Subject(s)
Amyloid beta-Peptides/toxicity , Hippocampus/metabolism , Peptide Fragments/toxicity , Receptors, AMPA/biosynthesis , Vesicular Glutamate Transport Protein 1/biosynthesis , Amyloid beta-Peptides/pharmacology , 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 , Hippocampus/drug effects , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Peptide Fragments/pharmacology , Receptors, AMPA/genetics , Vesicular Glutamate Transport Protein 1/genetics
8.
Cereb Cortex ; 29(9): 3982-3992, 2019 08 14.
Article in English | MEDLINE | ID: mdl-30395185

ABSTRACT

Epidemiological and experimental studies suggest that maternal immune activation (MIA) leads to developmental brain disorders, but whether the pathogenic mechanism impacts neurons already at birth is not known. We now report that MIA abolishes in mice the oxytocin-mediated delivery γ-aminobutyric acid (GABA) shift from depolarizing to hyperpolarizing in CA3 pyramidal neurons, and this is restored by the NKCC1 chloride importer antagonist bumetanide. Furthermore, MIA hippocampal pyramidal neurons at birth have a more exuberant apical arbor organization and increased apical dendritic length than age-matched controls. The frequency of spontaneous glutamatergic postsynaptic currents is also increased in MIA offspring, as well as the pairwise correlation of the synchronized firing of active cells in CA3. These alterations produced by MIA persist, since at P14-15 GABA action remains depolarizing, produces excitatory action, and network activity remains elevated with a higher frequency of spontaneous glutamatergic postsynaptic currents. Therefore, the pathogenic actions of MIA lead to important morphophysiological and network alterations in the hippocampus already at birth.


Subject(s)
CA3 Region, Hippocampal/growth & development , CA3 Region, Hippocampal/immunology , Membrane Potentials , Pregnancy/immunology , Pyramidal Cells/immunology , gamma-Aminobutyric Acid/immunology , Animals , CA3 Region, Hippocampal/drug effects , Dendrites/drug effects , Dendrites/immunology , Female , Glutamic Acid/physiology , Membrane Potentials/drug effects , Mice, Inbred C57BL , Poly I-C/administration & dosage , Pyramidal Cells/cytology , Pyramidal Cells/drug effects , Solute Carrier Family 12, Member 2/immunology
9.
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
10.
Pharmacology ; 105(7-8): 445-453, 2020.
Article in English | MEDLINE | ID: mdl-31825942

ABSTRACT

BACKGROUND: Tapentadol, a centrally acting oral analgesic, activates µ-opioid receptor and inhibits norepinephrine reuptake. Given that glutamate plays a crucial role in mediating pain, this study investigated the influence of tapentadol on spontaneous glutamatergic synaptic transmission and evoked neuronal excitability in rat hippocampal CA3 pyramidal neurons, which has been suggested to be involved in nociceptive perception. METHODS: We used electrophysiological technique to determine the effect of tapentadol on spontaneous excitatory postsynaptic currents (sEPSC), glutamate-activated currents, and neuronal excitability in CA3 pyramidal neurons in rat hippocampal slices. We also used isolated nerve terminals (synaptosomes) prepared from the rat hippocampus to examine the effect of tapentadol on glutamate release. RESULTS: Whole-cell patch clamp recordings revealed that tapentadol effectively decreased the frequencies of sEPSCs and miniature EPSCs (mEPSCs) without changing their amplitudes in hippocampal CA3 pyramidal neurons. However, glutamate-evoked inward currents were not affected by tapentadol. Further, tapentadol decreased 4-aminopyridine-induced glutamate release from hippocampal synaptosomes, and this effect was prevented by chelating the extracellular Ca2+ ions and blocking the N- and P/Q-type Ca2+ channels. In addition, burst firing induced by 4-aminopyridine and tonic repetitive firing induced by depolarizing pulses were attenuated by tapentadol. CONCLUSIONS: We conclude that tapentadol inhibits glutamatergic synaptic transmission, without modifying postsynaptic receptor sensitivity, and that this decline of excitation consequently suppresses neuronal hyperexcitability in the hippocampal CA3 area.


Subject(s)
Analgesics, Opioid/pharmacology , CA3 Region, Hippocampal/drug effects , Glutamic Acid/metabolism , Pyramidal Cells/drug effects , Tapentadol/pharmacology , Animals , CA3 Region, Hippocampal/metabolism , Excitatory Postsynaptic Potentials/drug effects , Male , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Pyramidal Cells/metabolism , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects
11.
Proc Natl Acad Sci U S A ; 114(9): 2383-2388, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28193882

ABSTRACT

Patients with early infantile epileptic encephalopathy (EIEE) experience severe seizures and cognitive impairment and are at increased risk for sudden unexpected death in epilepsy (SUDEP). EIEE13 [Online Mendelian Inheritance in Man (OMIM) # 614558] is caused by de novo missense mutations in the voltage-gated sodium channel gene SCN8A Here, we investigated the neuronal phenotype of a mouse model expressing the gain-of-function SCN8A patient mutation, p.Asn1768Asp (Nav1.6-N1768D). Our results revealed regional and neuronal subtype specificity in the effects of the N1768D mutation. Acutely dissociated hippocampal neurons from Scn8aN1768D/+ mice showed increases in persistent sodium current (INa) density in CA1 pyramidal but not bipolar neurons. In CA3, INa,P was increased in both bipolar and pyramidal neurons. Measurement of action potential (AP) firing in Scn8aN1768D/+ pyramidal neurons in brain slices revealed early afterdepolarization (EAD)-like AP waveforms in CA1 but not in CA3 hippocampal or layer II/III neocortical neurons. The maximum spike frequency evoked by depolarizing current injections in Scn8aN1768D/+ CA1, but not CA3 or neocortical, pyramidal cells was significantly reduced compared with WT. Spontaneous firing was observed in subsets of neurons in CA1 and CA3, but not in the neocortex. The EAD-like waveforms of Scn8aN1768D/+ CA1 hippocampal neurons were blocked by tetrodotoxin, riluzole, and SN-6, implicating elevated persistent INa and reverse mode Na/Ca exchange in the mechanism of hyperexcitability. Our results demonstrate that Scn8a plays a vital role in neuronal excitability and provide insight into the mechanism and future treatment of epileptogenesis in EIEE13.


Subject(s)
CA1 Region, Hippocampal/metabolism , Mutation , NAV1.6 Voltage-Gated Sodium Channel/genetics , Pyramidal Cells/metabolism , Spasms, Infantile/genetics , Action Potentials/drug effects , Amino Acid Substitution , Animals , Benzyl Compounds/pharmacology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/pathology , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/pathology , Disease Models, Animal , Gene Expression , Humans , Mice , Mice, Transgenic , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Neocortex/drug effects , Neocortex/metabolism , Neocortex/pathology , Organ Specificity , Pyramidal Cells/drug effects , Pyramidal Cells/pathology , Riluzole/pharmacology , Sodium Channel Blockers/pharmacology , Spasms, Infantile/metabolism , Spasms, Infantile/physiopathology , Tetrodotoxin/pharmacology , Thiazolidines/pharmacology
12.
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
13.
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
14.
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
15.
Int J Mol Sci ; 21(9)2020 Apr 25.
Article in English | MEDLINE | ID: mdl-32344826

ABSTRACT

OBJECTIVE: Hippocampal volume is reduced in patients with major depressive disorder (MDD) compared with healthy controls. The hippocampus is a limbic structure that has a critical role in MDD. The aim of the present study was to investigate the changes in the volume of the hippocampus and its subfields in MDD patients who responded to antidepressants and subsequently were in continuous remission. SUBJECTS AND METHODS: Eighteen patients who met the following criteria were enrolled in the present study: the DSM-IV-TR criteria for MDD, drug-naïve at least 8 weeks or more, scores on the 17-items of Hamilton Rating Scale for Depression (HAMD) of 14 points or more, and antidepressant treatment response within 8 weeks and continuous remission for at least 6 months. All participants underwent T1-weighted structural MRI and were treated with antidepressants for more than 8 weeks. We compared the volumes of the hippocampus, including its subfields, in responders at baseline to the volumes at 6 months. The volumes of the whole hippocampus and the hippocampal subfields were measured using FreeSurfer v6.0. RESULTS: The volumes of the left cornu Ammonis (CA) 3 (p = 0.016) and the granule cell layer of the dentate gyrus (GC-DG) region (p = 0.021) were significantly increased after 6 months of treatment compared with those at baseline. CONCLUSIONS: Increases in volume was observed in MDD patients who were in remission for at least 6 months.


Subject(s)
Antidepressive Agents/therapeutic use , CA3 Region, Hippocampal/pathology , Dentate Gyrus/pathology , Depressive Disorder, Major/pathology , Adult , Antidepressive Agents/pharmacology , CA3 Region, Hippocampal/diagnostic imaging , CA3 Region, Hippocampal/drug effects , Dentate Gyrus/diagnostic imaging , Dentate Gyrus/drug effects , Depressive Disorder, Major/diagnostic imaging , Depressive Disorder, Major/drug therapy , Female , Follow-Up Studies , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neuroimaging , Organ Size , Psychiatric Status Rating Scales , Remission Induction
16.
Bull Exp Biol Med ; 168(4): 419-422, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32146633

ABSTRACT

Whole-cell patch-clamp technique was employed to record chloride ionic current IGABA evoked by fast (600 msec) application of GABA to hippocampal pyramidal neurons and cerebellar Purkinje cells isolated from rat brain. GABA solution in the application pipette was either neutral (pH 7.4) or acidic (pH 7.0 or 6.0). Application of protons to neurons causes a rapid, reversible, and dose-dependent decrease in the amplitude of IGABA; the effect was more pronounced on hippocampal neurons (carrying both synaptic and extrasynaptic GABAA receptors) than in cerebellar Purkinje cells (predominantly equipped with synaptic GABAA receptors). In hippocampal neurons, pharmacological isolation of extrasynaptic component from total IGABA was performed with GABAA receptor antagonist gabazine (50 nM). The extrasynaptic component of IGABA was stronger blocked by protons than total IGABA. It was concluded that acidic medium produced more potent blocking effect on extrasynaptic GABAA receptors than on synaptic ones.


Subject(s)
Evoked Potentials/drug effects , Protons , Purkinje Cells/drug effects , Pyramidal Cells/drug effects , Receptors, GABA-A/physiology , gamma-Aminobutyric Acid/pharmacology , Animals , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/physiology , Cerebellum/cytology , Cerebellum/drug effects , Cerebellum/physiology , Dose-Response Relationship, Drug , Evoked Potentials/physiology , GABA Antagonists/pharmacology , Hydrogen-Ion Concentration , Patch-Clamp Techniques , Primary Cell Culture , Protein Subunits/physiology , Purkinje Cells/cytology , Purkinje Cells/physiology , Pyramidal Cells/cytology , Pyramidal Cells/physiology , Pyridazines/pharmacology , Rats , Rats, Wistar
17.
Bull Exp Biol Med ; 168(4): 474-478, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32146635

ABSTRACT

Peptide mimetic of nerve growth factor GK-2 in a dose of 1-2 mg/liter improves survival of cultured rat cerebellar granule neurons exposed to the cytotoxic effect of zinc ions, but has no protective effect against copper ion cytotoxicity. Experiments on cultured rat hippocampal slices demonstrated that GK-2 did not affect reactivity of pyramidal neurons and long-term potentiation in the hippocampal field CA1 and the probability of glutamate release from presynaptic terminals in the synapses of the CA3-CA1 fields. The results suggest that GK-2 does not affect the functional properties of synaptic transmission under normal conditions, but protects neurons from the toxic effects of zinc, which creates prerequisites for GK-12 use in the treatment of neurodegenerative diseases.


Subject(s)
CA1 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/drug effects , Cerebellum/drug effects , Chlorides/antagonists & inhibitors , Dipeptides/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Zinc Compounds/antagonists & inhibitors , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/physiology , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/physiology , Cerebellum/cytology , Cerebellum/physiology , Chlorides/toxicity , Copper/toxicity , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Microtomy , Neurons/cytology , Neurons/physiology , Primary Cell Culture , Rats , Synapses/drug effects , Synapses/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Zinc Compounds/toxicity
18.
Synapse ; 73(5): e22088, 2019 05.
Article in English | MEDLINE | ID: mdl-30632204

ABSTRACT

In adult female, but not male, Sprague Dawley rats, chronic immobilization stress (CIS) increases mossy fiber (MF) Leu-Enkephalin levels and redistributes delta- and mu-opioid receptors (DORs and MORs) in hippocampal CA3 pyramidal cells and GABAergic interneurons to promote excitation and learning processes following subsequent opioid exposure. Here, we demonstrate that CIS females, but not males, acquire conditioned place preference (CPP) to oxycodone and that CIS "primes" the hippocampal opioid system in females for oxycodone-associated learning. In CA3b, oxycodone-injected (Oxy) CIS females relative to saline-injected (Sal) CIS females exhibited an increase in the cytoplasmic and total densities of DORs in pyramidal cell dendrites so that they were similar to Sal- and Oxy-CIS males. Consistent with our earlier studies, Sal- and Oxy-CIS females but not CIS males had elevated DOR densities in MF-CA3 dendritic spines, which we have previously shown are important for opioid-mediated long-term potentiation. In the dentate gyrus, Oxy-CIS females had more DOR-labeled interneurons than Sal-CIS females. Moreover, Sal- and Oxy-CIS females compared to both groups of CIS males had elevated levels of DORs and MORs in GABAergic interneuron dendrites, suggesting capacity for greater synthesis or storage of these receptors in circuits important for opioid-mediated disinhibition. However, more plasmalemmal MORs were on large parvalbumin-containing dendrites of Oxy-CIS males compared to Sal-CIS males, suggesting a limited ability for increased granule cell disinhibition. These results suggest that low levels of DORs in MF-CA3 synapses and hilar GABAergic interneurons may contribute to the attenuation of oxycodone CPP in males exposed to CIS.


Subject(s)
Analgesics, Opioid/pharmacology , CA3 Region, Hippocampal/metabolism , Conditioning, Classical , Dentate Gyrus/metabolism , Oxycodone/pharmacology , Repetition Priming , Stress, Psychological/physiopathology , Animals , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/drug effects , Dendrites/metabolism , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Female , Interneurons/metabolism , Male , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Restraint, Physical , Stress, Psychological/metabolism
19.
J Clin Psychopharmacol ; 39(6): 653-657, 2019.
Article in English | MEDLINE | ID: mdl-31688386

ABSTRACT

PURPOSE/BACKGROUND: Glucocorticoids are a class of hormones that include naturally occurring cortisol and corticosterone, as well as prescription drugs commonly used to manage inflammatory, autoimmune, and allergic conditions. Adverse effects, including neuropsychiatric symptoms, are common. The hippocampus appears to be especially sensitive to the effects of glucocorticoids. However, to our knowledge, no studies to date have examined hippocampal subfields in humans receiving glucocorticoids. We examined patients on chronic glucocorticoid regimens to determine relationships between dose and duration of treatment, and hippocampal subfields, and related regions volumes. METHODS/PROCEDURES: The study included adult men and women receiving at least 5 mg daily of prednisone equivalents for at least 6 months. Volumes of brain regions were measured via magnetic resonance imaging. A multivariate general linear model was used for analysis, with brain volumes as dependent variables and age, sex, and cumulative corticosteroid exposure, as predictors. FINDINGS/RESULTS: The study population consisted of 81 adult outpatients (43 male) on corticosteroids (mean dose, 7.88 mg; mean duration, 76.75 months). Cumulative glucocorticoid exposure was negatively associated with left and right hippocampal dentate gyrus/CA3 volume. In subsequent subgroup analysis, this association held true for the age group older than the median age of 46 years but not for the younger age group. IMPLICATIONS/CONCLUSIONS: This finding is consistent with previous studies showing detrimental effects of elevated glucocorticoids on the hippocampus but further suggests that the dentate gyrus and CA3 regions are particularly vulnerable to those effects, which is consistent with animal models of chronic stress but has not been previously demonstrated in humans.


Subject(s)
CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/pathology , Dentate Gyrus/drug effects , Dentate Gyrus/pathology , Glucocorticoids/adverse effects , Neuroimaging/methods , Adult , Aged , CA3 Region, Hippocampal/diagnostic imaging , Clinical Trials as Topic , Dentate Gyrus/diagnostic imaging , Female , Glucocorticoids/administration & dosage , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Prednisone/administration & dosage , Prednisone/adverse effects , Young Adult
20.
Addict Biol ; 24(1): 51-64, 2019 01.
Article in English | MEDLINE | ID: mdl-29193459

ABSTRACT

Although chronic nicotine administration does not affect memory, its withdrawal causes massive cognitive deficits. The underlying mechanisms, however, have not been understood. We test the role of cocaine- and amphetamine-regulated transcript peptide (CART), a neuropeptide known for its procognitive properties, in this process. The mice on chronic nicotine treatment/withdrawal were subjected to novel object recognition task. The capability of the animal to discriminate between the novel and familiar objects was tested and represented as discrimination index (DI); reduction in the index suggested amnesia. Nicotine for 49 days had no effect on DI, but 8-hour withdrawal caused a significant reduction, followed by full recovery at 24-hour withdrawal timepoint. Bilateral CART infusion in dorsal hippocampus rescued deficits in DI at 8-hours, whereas CART-antibody infusion into the dorsal hippocampus attenuated the recovery at 24-hours. Commensurate changes were observed in the CART as well as CART mRNA profiles in the hippocampus. CART mRNA expression and the peptide immunoreactivity did not change significantly following chronic nicotine treatment. However, there was a significant reduction at 8-hour withdrawal, followed by a drastic increase in CART immunoreactivity as well as CART mRNA at 24-hour withdrawal, compared with 8-hour withdrawal. Distinct α7-nicotinic receptor immunoreactivity was detected on the hippocampal CART neurons, suggesting cholinergic inputs. An increase in the synaptophysin immunoreactive elements around CART cells in the dentate gyrus, cornu ammonis 3 and subiculum at 24-hour post-withdrawal timepoint suggested neuronal plasticity. CART circuit dynamics in the hippocampus seems to modulate short-term memory associated with nicotine withdrawal.


Subject(s)
Nerve Tissue Proteins/pharmacology , Nicotine/adverse effects , Nicotinic Agonists/adverse effects , Recognition, Psychology/drug effects , Substance Withdrawal Syndrome/psychology , Animals , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/metabolism , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Neuronal Plasticity , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Substance Withdrawal Syndrome/etiology , Substance Withdrawal Syndrome/genetics , Synaptophysin/drug effects , Synaptophysin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL