Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Physiol ; 600(19): 4325-4345, 2022 10.
Article in English | MEDLINE | ID: mdl-36030507

ABSTRACT

The tachykinin peptides include substance P (SP), neurokinin A and neurokinin B, which interact with three G-protein-coupled neurokinin receptors, NK1Rs, NK2Rs and NK3Rs, respectively. Whereas high densities of NK3Rs have been detected in the basolateral amygdala (BLA), the functions of NK3Rs in this brain region have not been determined. We found that activation of NK3Rs by application of the selective agonist, senktide, persistently excited BLA principal neurons. NK3R-elicited excitation of BLA neurons was mediated by activation of a non-selective cation channel and depression of the inwardly rectifying K+ (Kir) channels. With selective channel blockers and knockout mice, we further showed that NK3R activation excited BLA neurons by depressing the G protein-activated inwardly rectifying K+ (GIRK) channels and activating TRPC4 and TRPC5 channels. The effects of NK3Rs required the functions of phospholipase Cß (PLCß), but were independent of intracellular Ca2+ release and protein kinase C. PLCß-mediated depletion of phosphatidylinositol 4,5-bisphosphate was involved in NK3R-induced excitation of BLA neurons. Microinjection of senktide into the BLA of rats augmented fear-potentiated startle (FPS) and this effect was blocked by prior injection of the selective NK3R antagonist SB 218795, suggesting that activation of NK3Rs in the BLA increased FPS. We further showed that TRPC4/5 and GIRK channels were involved in NK3R-elicited facilitation of FPS. Our results provide a cellular and molecular mechanism whereby NK3R activation excites BLA neurons and enhances FPS. KEY POINTS: Activation of NK3 receptors (NK3Rs) facilitates the excitability of principal neurons in rat basolateral amygdala (BLA). NK3R-induced excitation is mediated by inhibition of GIRK channels and activation of TRPC4/5 channels. Phospholipase Cß and depletion of phosphatidylinositol 4,5-bisphosphate are necessary for NK3R-mediated excitation of BLA principal neurons. Activation of NK3Rs in the BLA facilitates fear-potentiated startle response. GIRK channels and TRPC4/5 channels are involved in NK3R-mediated augmentation of fear-potentiated startle.


Subject(s)
Basolateral Nuclear Complex , Receptors, Neurokinin-3 , Animals , Basolateral Nuclear Complex/metabolism , Fear , Mice , Neurokinin A/metabolism , Neurokinin B/metabolism , Neurokinin B/pharmacology , Phosphatidylinositols , Phospholipases/metabolism , Protein Kinase C/metabolism , Rats , Receptors, Neurokinin-3/metabolism , Reflex, Startle , Substance P/metabolism , Substance P/pharmacology , TRPC Cation Channels/metabolism
2.
J Cell Physiol ; 237(1): 660-674, 2022 01.
Article in English | MEDLINE | ID: mdl-34287874

ABSTRACT

Arginine vasopressin (AVP) is a hormone exerting vasoconstrictive and antidiuretic action in the periphery and serves as a neuromodulator in the brain. Although the hippocampus receives vasopressinergic innervation and AVP has been shown to facilitate the excitability of CA1 pyramidal neurons, the involved ionic and signaling mechanisms have not been determined. Here we found that AVP excited CA1 pyramidal neurons by activation of V1a receptors. Functions of G proteins and phospholipase Cß (PLCß) were required for AVP-elicited excitation of CA1 pyramidal neurons, whereas intracellular Ca2+ release and protein kinase C were unnecessary. PLCß-mediated depletion of phosphatidylinositol 4,5-bisphosphate (PIP2 ) was required for AVP-elicited excitation of CA1 pyramidal neurons. AVP augmented the input resistance and increased the time constants of CA1 pyramidal neurons. AVP induced an inward current in K+ -containing intracellular solution, whereas no inward currents were observed with Cs+ -containing intracellular solution. AVP-sensitive currents showed inward rectification with a reversal potential close to the K+ reversal potential, suggesting the involvement of inwardly rectifying K+ channels. AVP-induced currents were sensitive to the micromolar concentration of Ba2+ and tertiapin-Q, whereas application of ML 133, a selective Kir2 channel blocker had no effects, suggesting that AVP excited CA1 pyramidal neurons by depressing G protein-gated inwardly rectifying K+ channels. Activation of V1a receptors in the CA1 region facilitated glutamatergic transmission onto subicular pyramidal neurons, suggesting that AVP modulates network activity in the brain. Our results may provide one of the cellular and molecular mechanisms to explain the in vivo physiological functions of AVP.


Subject(s)
Arginine Vasopressin , Pyramidal Cells , Arginine Vasopressin/metabolism , Arginine Vasopressin/pharmacology , Hippocampus/metabolism , Phospholipase C beta/metabolism , Pyramidal Cells/metabolism
3.
Neuropharmacology ; 196: 108714, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34271017

ABSTRACT

Neurotensin (NT) serves as a neuromodulator in the brain where it regulates a variety of physiological functions. Whereas the central amygdala (CeA) expresses NT peptide and NTS1 receptors and application of NT has been shown to excite CeA neurons, the underlying cellular and molecular mechanisms have not been determined. We found that activation of NTS1 receptors increased the neuronal excitability of the lateral nucleus (CeL) of CeA. Both phospholipase Cß (PLCß) and phosphatidylinositol 4,5-bisphosphate (PIP2) depletion were required, whereas intracellular Ca2+ release and PKC were unnecessary for NT-elicited excitation of CeL neurons. NT increased the input resistance and time constants of CeL neurons, suggesting that NT excites CeL neurons by decreasing a membrane conductance. Depressions of the inwardly rectifying K+ (Kir) channels including both the Kir2 subfamily and the GIRK channels were required for NT-elicited excitation of CeL neurons. Activation of NTS1 receptors in the CeL led to GABAergic inhibition of medial nucleus of CeA neurons, suggesting that NT modulates the network activity in the amygdala. Our results may provide a cellular and molecular mechanism to explain the physiological functions of NT in vivo.


Subject(s)
Action Potentials/physiology , Central Amygdaloid Nucleus/metabolism , Membrane Potentials/physiology , Neurons/metabolism , Neurotensin/metabolism , Receptors, Neurotensin/metabolism , Animals , Central Amygdaloid Nucleus/physiology , GTP-Binding Proteins/metabolism , Inhibitory Postsynaptic Potentials/physiology , Miniature Postsynaptic Potentials/physiology , Neurons/physiology , Patch-Clamp Techniques , Phospholipase C beta/metabolism , Rats , Signal Transduction
4.
J Physiol ; 599(12): 3101-3119, 2021 06.
Article in English | MEDLINE | ID: mdl-33871877

ABSTRACT

KEY POINTS: Activation of V1a vasopressin receptors facilitates neuronal excitability in the medial nucleus of central amygdala (CeM) V1a receptor activation excites about 80% CeM neurons by opening a cationic conductance and about 20% CeM neurons by suppressing an inwardly rectifying K+ (Kir) channel The cationic conductance activated by V1a receptors is identified as TRPC5 channels PLCß-mediated depletion of PIP2 is involved in V1a receptor-elicited excitation of CeM neurons Intracellular Ca2+ release and PKC are unnecessary for V1a receptor-mediated excitation of CeM neurons ABSTRACT: Arginine vasopressin (AVP) serves as a hormone in the periphery to modulate water homeostasis and a neuromodulator in the brain to regulate a diverse range of functions including anxiety, social behaviour, cognitive activities and nociception. The amygdala is an essential brain region involved in modulating defensive and appetitive behaviours, pain and alcohol use disorders. Whereas activation of V1a receptors in the medial nucleus of the central amygdala (CeM) increases neuronal excitability, the involved ionic and signalling mechanisms have not been determined. We found that activation of V1a receptors in the CeM facilitated neuronal excitability predominantly by opening TRPC5 channels, although AVP excited about one fifth of the CeM neurons via suppressing an inwardly rectifying K+ (Kir) channel. G proteins and phospholipase Cß (PLCß) were required for AVP-elicited excitation of CeM neurons, whereas intracellular Ca2+ release and the activity of protein kinase C were unnecessary. Prevention of the depletion of phosphatidylinositol 4,5-bisphosphate (PIP2 ) blocked AVP-induced excitation of CeM neurons, suggesting that PLCß-mediated depletion of PIP2 is involved in AVP-mediated excitation of CeM neurons. Our results may provide a cellular and molecular mechanism to explain the anxiogenic effects of AVP in the amygdala.


Subject(s)
Alcoholism , Central Amygdaloid Nucleus , Humans , Neurons , Phosphatidylinositol 4,5-Diphosphate , Phospholipase C beta , TRPC Cation Channels , Vasopressins
5.
Neuropharmacology ; 190: 108565, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33891950

ABSTRACT

Arginine vasopressin (AVP) is a nonapeptide that serves as a neuromodulator in the brain and a hormone in the periphery that regulates water homeostasis and vasoconstriction. The subiculum is the major output region of the hippocampus and an integral component in the networks that processes sensory and motor cues to form a cognitive map encoding spatial, contextual, and emotional information. Whereas the subiculum expresses high densities of AVP-binding sites and AVP has been shown to increase the synaptic excitability of subicular pyramidal neurons, the underlying cellular and molecular mechanisms have not been determined. We found that activation of V1a receptors increased the excitability of subicular pyramidal neurons via activation of TRPV1 channels and depression of the GIRK channels. V1a receptor-induced excitation of subicular pyramidal neurons required the function of phospholipase Cß, but was independent of intracellular Ca2+ release. Protein kinase C was responsible for AVP-mediated depression of GIRK channels, whereas degradation of phosphatidylinositol 4,5-bisphosphate was involved in V1a receptor-elicited activation of TRPV1 channels. Our results may provide one of the cellular and molecular mechanisms to explain the physiological functions of AVP in the brain.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Hippocampus/metabolism , Pyramidal Cells/metabolism , Receptors, Vasopressin/metabolism , TRPV Cation Channels/metabolism , Action Potentials , Animals , Arginine Vasopressin/pharmacology , Calcium/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Membrane Potentials , Mice , Mice, Knockout , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase C beta/metabolism , Pyramidal Cells/drug effects , Receptors, Vasopressin/agonists , TRPV Cation Channels/drug effects , TRPV Cation Channels/genetics , Vasoconstrictor Agents/pharmacology
6.
Cereb Cortex ; 31(5): 2402-2415, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33341872

ABSTRACT

Oxytocin (OXT) is a nonapeptide that serves as a neuromodulator in the brain and a hormone participating in parturition and lactation in the periphery. The subiculum is the major output region of the hippocampus and an integral component in the networks that process sensory and motor cues to form a cognitive map encoding spatial, contextual, and emotional information. Whilst the subiculum expresses the highest OXT-binding sites and is the first brain region to be activated by peripheral application of OXT, the precise actions of OXT in the subiculum have not been determined. Our results demonstrate that application of the selective OXT receptor (OXTR) agonist, [Thr4,Gly7]-oxytocin (TGOT), excited subicular neurons via activation of TRPV1 channels, and depression of K+ channels. The OXTR-mediated excitation of subicular neurons required the functions of phospholipase Cß, protein kinase C, and degradation of phosphatidylinositol 4,5-bisphosphate (PIP2). OXTR-elicited excitation of subicular neurons enhanced long-term potentiation via activation of TRPV1 channels. Our results provide a cellular and molecular mechanism to explain the physiological functions of OXT in the brain.


Subject(s)
Hippocampus/metabolism , Neurons/metabolism , Receptors, Oxytocin/metabolism , TRPV Cation Channels/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Calcium Signaling , Female , Hippocampus/cytology , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/drug effects , Oxytocin/analogs & derivatives , Oxytocin/pharmacology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase C beta/drug effects , Phospholipase C beta/metabolism , Potassium Channels/drug effects , Potassium Channels/metabolism , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Rats , Receptors, Oxytocin/agonists , Signal Transduction , TRPV Cation Channels/drug effects
7.
J Physiol ; 598(16): 3501-3520, 2020 08.
Article in English | MEDLINE | ID: mdl-32458437

ABSTRACT

KEY POINTS: Activation of oxytocin receptors (OXTRs) facilitates neuronal excitability in rat lateral nucleus of central amygdala (CeL). OXTR-induced excitation is mediated by inhibition of inwardly rectifying K+ (Kir) channels. Phospholipase Cß is necessary for OXTR-mediated excitation of CeL neurons and depression of Kir channels. OXTR-elicited depression of Kir channels and excitation of CeL neurons require the function of Ca2+ -dependent protein kinase C. ABSTRACT: Oxytocin (OXT) is a nonapeptide that exerts anxiolytic effects in the brain. The amygdala is an important structure involved in the modulation of fear and anxiety. A high density of OXT receptors (OXTRs) has been detected in the capsular (CeC) and lateral (CeL) nucleus of the central amygdala (CeA). Previous studies have demonstrated that activation of OXTRs induces remarkable increases in neuronal excitability in the CeL/C. However, the signalling and ionic mechanisms underlying OXTR-induced facilitation of neuronal excitability have not been determined. We found that activation of OXTRs in the CeL increased action potential firing frequency recorded from neurons in this region via inhibition of the inwardly rectifying K+ channels. The functions of phospholipase Cß and protein kinase C were required for OXTR-induced augmentation of neuronal excitability. Our results provide a cellular and molecular mechanism whereby activation of OXTRs exerts anxiolytic effects.


Subject(s)
Central Amygdaloid Nucleus , Action Potentials , Animals , Oxytocin , Phospholipase C beta , Protein Kinase C , Rats , Receptors, Oxytocin
8.
Neuropharmacology ; 151: 144-158, 2019 06.
Article in English | MEDLINE | ID: mdl-30998945

ABSTRACT

Nociceptin (NOP) is an endogenous opioid-like peptide that selectively activates the opioid receptor-like (ORL-1) receptors. The entorhinal cortex (EC) is closely related to temporal lobe epilepsy and expresses high densities of ORL-1 receptors. However, the functions of NOP in the EC, especially in modulating the epileptiform activity in the EC, have not been determined. We demonstrated that activation of ORL-1 receptors remarkably inhibited the epileptiform activity in entorhinal slices induced by application of picrotoxin or by deprivation of extracellular Mg2+. NOP-mediated depression of epileptiform activity was independent of synaptic transmission in the EC, but mediated by inhibition of neuronal excitability in the EC. NOP hyperpolarized entorhinal neurons via activation of K+ channels and inhibition of cation channels. Whereas application of Ba2+ at 300 µM which is effective for the inward rectifier K+ (Kir) channels slightly inhibited NOP-induced hyperpolarization, the current-voltage (I-V) curve of the net currents induced by NOP was linear without showing inward rectification. However, a role of NOP-induced inhibition of cation channels was revealed after inhibition of Kir channels by Ba2+. Furthermore, NOP-mediated augmentation of membrane currents was differently affected by application of the blockers selective for distinct subfamilies of Kir channels. Whereas SCH23390 or ML133 blocked NOP-induced augmentation of membrane currents at negative potentials, application of tertiapin-Q exerted no actions on NOP-induced alteration of membrane currents. Our results demonstrated a novel cellular and molecular mechanism whereby activation of ORL-1 receptors depresses epilepsy.


Subject(s)
Action Potentials/physiology , Entorhinal Cortex/metabolism , Potassium Channels/metabolism , Pyramidal Cells/physiology , Receptors, Opioid/metabolism , Action Potentials/drug effects , Animals , Entorhinal Cortex/drug effects , Opioid Peptides/metabolism , Potassium Channel Blockers/pharmacology , Pyramidal Cells/drug effects , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Nociceptin Receptor , Nociceptin
9.
PLoS One ; 13(1): e0189282, 2018.
Article in English | MEDLINE | ID: mdl-29324743

ABSTRACT

The dispatching of hydro-thermal system is a nonlinear programming problem with multiple constraints and high dimensions and the solution techniques of the model have been a hotspot in research. Based on the advantage of that the artificial bee colony algorithm (ABC) can efficiently solve the high-dimensional problem, an improved artificial bee colony algorithm has been proposed to solve DHTS problem in this paper. The improvements of the proposed algorithm include two aspects. On one hand, local search can be guided in efficiency by the information of the global optimal solution and its gradient in each generation. The global optimal solution improves the search efficiency of the algorithm but loses diversity, while the gradient can weaken the loss of diversity caused by the global optimal solution. On the other hand, inspired by genetic algorithm, the nectar resource which has not been updated in limit generation is transformed to a new one by using selection, crossover and mutation, which can ensure individual diversity and make full use of prior information for improving the global search ability of the algorithm. The two improvements of ABC algorithm are proved to be effective via a classical numeral example at last. Among which the genetic operator for the promotion of the ABC algorithm's performance is significant. The results are also compared with those of other state-of-the-art algorithms, the enhanced ABC algorithm has general advantages in minimum cost, average cost and maximum cost which shows its usability and effectiveness. The achievements in this paper provide a new method for solving the DHTS problems, and also offer a novel reference for the improvement of mechanism and the application of algorithms.


Subject(s)
Algorithms , Bees , Animals , Models, Economic
10.
Hippocampus ; 27(9): 971-984, 2017 09.
Article in English | MEDLINE | ID: mdl-28558129

ABSTRACT

The hippocampus is a crucial component for cognitive and emotional processing. The subiculum provides much of the output for this structure but the modulation and function of this region is surprisingly under-studied. The neuromodulator somatostatin (SST) interacts with five subtypes of SST receptors (sst1 to sst5 ) and each of these SST receptor subtypes is coupled to Gi proteins resulting in inhibition of adenylyl cyclase (AC) and decreased level of intracellular cAMP. SST modulates many physiological functions including cognition, emotion, autonomic responses and locomotion. Whereas SST has been shown to depress neuronal excitability in the subiculum, the underlying cellular and molecular mechanisms have not yet been determined. Here, we show that SST hyperpolarized two classes of subicular neurons with a calculated EC50 of 0.1 µM. Application of SST (1 µM) induced outward holding currents by primarily activating K+ channels including the G-protein-activated inwardly-rectifying potassium channels (GIRK) and KCNQ (M) channels, although inhibition of cation channels in some cells may also be implicated. SST-elicited hyperpolarization was mediated by activation of sst2 receptors and required the function of G proteins. The SST-induced hyperpolarization resulted from decreased activity of AC and reduced levels of cAMP but did not require the activity of either PKA or PKC. Inhibition of Epac2, a guanine nucleotide exchange factor, partially blocked SST-mediated hyperpolarization of subicular neurons. Furthermore, application of SST resulted in a robust depression of subicular action potential firing and the SST-induced hyperpolarization was responsible for its inhibitory action on LTP at the CA1-subicilum synapses. Our results provide a novel cellular and molecular mechanism that may explain the roles of SST in modulation of subicular function and be relevant to SST-related physiological functions.


Subject(s)
Action Potentials/drug effects , Guanine Nucleotide Exchange Factors/metabolism , Hippocampus/cytology , KCNQ Potassium Channels/metabolism , Neurons/drug effects , Somatostatin/pharmacology , Animals , Animals, Newborn , Calcium/metabolism , Cyclic AMP/metabolism , Enzyme Inhibitors/pharmacology , Guanine Nucleotide Exchange Factors/agonists , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Models, Biological , Nerve Net/drug effects , Neurons/classification , Neurotransmitter Agents/pharmacology , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology , Somatostatin/agonists , Somatostatin/antagonists & inhibitors , Tetrodotoxin/pharmacology
11.
Cereb Cortex ; 26(3): 977-90, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25405940

ABSTRACT

Neurotensin (NT) is a 13-amino acid peptide and serves as a neuromodulator in the brain. Whereas NT has been implicated in learning and memory, the underlying cellular and molecular mechanisms are ill-defined. Because the dentate gyrus receives profound innervation of fibers containing NT and expresses high density of NT receptors, we examined the effects of NT on the excitability of dentate gyrus granule cells (GCs). Our results showed that NT concentration dependently increased action potential (AP) firing frequency of the GCs by the activation of NTS1 receptors resulting in the depolarization of the GCs. NT-induced enhancement of AP firing frequency was not caused indirectly by releasing glutamate, GABA, acetylcholine, or dopamine, but due to the inhibition of TASK-3 K(+) channels. NT-mediated excitation of the GCs was G protein dependent, but independent of phospholipase C, intracellular Ca(2+) release, and protein kinase C. Immunoprecipitation experiment demonstrates that the activation of NTS1 receptors induced the association of Gαq/11 and TASK-3 channels suggesting a direct coupling of Gαq/11 to TASK-3 channels. Endogenously released NT facilitated the excitability of the GCs contributing to the induction of long-term potentiation at the perforant path-GC synapses. Our results provide a cellular mechanism that helps to explain the roles of NT in learning and memory.


Subject(s)
Dentate Gyrus/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Neurons/physiology , Potassium Channels, Tandem Pore Domain/metabolism , Potassium Channels/metabolism , Receptors, Neurotensin/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Dentate Gyrus/drug effects , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , HEK293 Cells , Humans , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurotensin/metabolism , Potassium Channels/genetics , Rats, Sprague-Dawley , Receptors, Neurotensin/genetics , Tissue Culture Techniques
12.
Hippocampus ; 25(11): 1299-313, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25740117

ABSTRACT

Whereas the ionotropic glutamate receptors are the major mediator in glutamatergic transmission, the metabotropic glutamate receptors (mGluRs) usually play a modulatory role. Whereas the entorhinal cortex (EC) is an essential structure involved in the generation and propagation of epilepsy, the roles and mechanisms of mGluRs in epilepsy in the EC have not been determined. Here, we studied the effects of activation of group II metabotropic glutamate receptors (mGluRs II) on epileptiform activity induced by picrotoxin or deprivation of extracellular Mg2+ and neuronal excitability in the medial EC. We found that activation of mGluRs II by application of the selective agonist, LY354740, exerted robust inhibition on epileptiform activity. LY354740 hyperpolarized entorhinal neurons via activation of a K+ conductance and inhibition of a Na+ -permeable channel. LY354740-induced hyperpolarization was G protein-dependent, but independent of adenylyl cyclase and protein kinase A. However, the function of Gßγ was involved in mGluRs II-mediated depression of both neuronal excitability and epileptiform activity. Our results provide a novel cellular mechanism to explain the antiepileptic effects of mGluRs II in the treatment of epilepsy.


Subject(s)
Entorhinal Cortex/metabolism , Epilepsy/metabolism , Magnesium/metabolism , Receptors, Metabotropic Glutamate/metabolism , Synaptic Potentials/physiology , Animals , Bridged Bicyclo Compounds/pharmacology , Disease Models, Animal , Entorhinal Cortex/drug effects , Epilepsy/drug therapy , Excitatory Amino Acid Agonists/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/agonists , Synaptic Potentials/drug effects
13.
J Neurosci ; 34(20): 7027-42, 2014 May 14.
Article in English | MEDLINE | ID: mdl-24828655

ABSTRACT

Neurotensin (NT) is a tridecapeptide distributed in the CNS, including the entorhinal cortex (EC), a structure that is crucial for learning and memory and undergoes the earliest pathological alterations in Alzheimer's disease (AD). Whereas NT has been implicated in modulating cognition, the cellular and molecular mechanisms by which NT modifies cognitive processes and the potential therapeutic roles of NT in AD have not been determined. Here we examined the effects of NT on neuronal excitability and spatial learning in the EC, which expresses high density of NT receptors. Brief application of NT induced persistent increases in action potential firing frequency, which could last for at least 1 h. NT-induced facilitation of neuronal excitability was mediated by downregulation of TREK-2 K(+) channels and required the functions of NTS1, phospholipase C, and protein kinase C. Microinjection of NT or NTS1 agonist, PD149163, into the EC increased spatial learning as assessed by the Barnes Maze Test. Activation of NTS1 receptors also induced persistent increases in action potential firing frequency and significantly improved the memory status in APP/PS1 mice, an animal model of AD. Our study identifies a cellular substrate underlying learning and memory and suggests that NTS1 agonists may exert beneficial actions in an animal model of AD.


Subject(s)
Alzheimer Disease/physiopathology , Entorhinal Cortex/drug effects , Maze Learning/drug effects , Neurons/drug effects , Neurotensin/pharmacology , Receptors, Neurotensin/agonists , Action Potentials/drug effects , Action Potentials/physiology , Alzheimer Disease/psychology , Animals , Disease Models, Animal , Entorhinal Cortex/physiopathology , Maze Learning/physiology , Mice , Neurons/physiology
14.
Cereb Cortex ; 24(12): 3195-208, 2014 Dec.
Article in English | MEDLINE | ID: mdl-23843440

ABSTRACT

Whereas the entorhinal cortex (EC) receives profuse dopaminergic innervations from the midbrain, the effects of dopamine (DA) on γ-Aminobutyric acid (GABA)ergic interneurons in this brain region have not been determined. We probed the actions of DA on GABAA receptor-mediated synaptic transmission in the EC. Application of DA increased the frequency, not the amplitude, of spontaneous IPSCs (sIPSCs) and miniature IPSCs (mIPSCs) recorded from entorhinal principal neurons, but slightly reduced the amplitude of the evoked IPSCs. The effects of DA were unexpectedly found to be mediated by α1 adrenoreceptors, but not by DA receptors. DA endogenously released by the application of amphetamine also increased the frequency of sIPSCs. Ca(2+) influx via T-type Ca(2+) channels was required for DA-induced facilitation of sIPSCs and mIPSCs. DA depolarized and enhanced the firing frequency of action potentials of interneurons. DA-induced depolarization was independent of extracellular Na(+) and Ca(2+) and did not require the functions of hyperpolarization-activated (Ih) channels and T-type Ca(2+) channels. DA-generated currents showed a reversal potential close to the K(+) reversal potential and inward rectification, suggesting that DA inhibits the inward rectifier K(+) channels (Kirs). Our results demonstrate that DA facilitates GABA release by activating α1 adrenoreceptors to inhibit Kirs, which further depolarize interneurons resulting in secondary Ca(2+) influx via T-type Ca(+) channels.


Subject(s)
Calcium Channels, T-Type/physiology , Dopamine/metabolism , Entorhinal Cortex/cytology , Neurons/physiology , Potassium Channels, Inwardly Rectifying/physiology , Receptors, Adrenergic, alpha-1/physiology , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Calcium Channel Blockers/pharmacology , Chelating Agents/pharmacology , Dopamine/pharmacology , Dose-Response Relationship, Drug , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Electric Stimulation , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Mibefradil/pharmacology , Neural Inhibition/drug effects , Neurons/drug effects , Neurotransmitter Agents/pharmacology , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/pharmacology
15.
Hippocampus ; 24(1): 21-31, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23966303

ABSTRACT

Bombesin and the bombesin-like peptides including neuromedin B (NMB) and gastrin-releasing peptide (GRP) are important neuromodulators in the brain. We studied their effects on GABAergic transmission and epileptiform activity in the entorhinal cortex (EC). Bath application of bombesin concentration-dependently increased both the frequency and amplitude of sIPSCs recorded from the principal neurons in the EC. Application of NMB and GRP exerted the same effects as bombesin. Bombesin had no effects on mIPSCs recorded in the presence of TTX but slightly depressed the evoked IPSCs. Omission of extracellular Ca(2+) or inclusion of voltage-gated Ca(2+) channel blockers, Cd(2+) and Ni(2+), blocked bombesin-induced increases in sIPSCs suggesting that bombesin increases GABA release via facilitating extracellular Ca(2+) influx. Bombesin induced membrane depolarization and slightly increased the input resistance of GABAergic interneurons recorded from layer III of the EC. The action potential firing frequency of the interneurons was also increased by bombesin. Bombesin-mediated depolarization of interneurons was unlikely to be mediated by the opening of a cationic conductance but due to the inhibition of inward rectifier K(+) channels. Bath application of bombesin, NMB and GRP depressed the frequency of the epileptiform activity elicited by deprivation of Mg(2+) from the extracellular solution suggesting that bombesin and the bombesin-like peptides have antiepileptic effects in the brain.


Subject(s)
Anticonvulsants/pharmacology , Bombesin/pharmacology , Entorhinal Cortex/drug effects , Neurons/drug effects , Synaptic Transmission/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Animals , Bombesin/metabolism , Entorhinal Cortex/physiology , Epilepsy/metabolism , Epilepsy/physiopathology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Neurons/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Seizures/metabolism , Seizures/physiopathology , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism
16.
Neuropharmacology ; 63(7): 1218-26, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22884625

ABSTRACT

Whereas vasopressin has been shown to enhance memory possibly by increasing long-term potentiation and direct excitation of the pyramidal neurons in the hippocampus, the effects of vasopressin on GABAergic transmission in the hippocampus remain to be determined. Here we examined the effects of vasopressin on GABAergic transmission onto CA1 pyramidal neurons and our results demonstrate that bath application of [Arg(8)]-vasopressin (AVP) dose-dependently increased the frequency of spontaneous IPSCs (sIPSCs) recorded from CA1 pyramidal neurons via activation of V(1A) receptors. Immunohistological staining and western blot further confirmed that both CA1 pyramidal neurons and interneurons expressed V(1A) receptors. Bath application of AVP altered neither the frequency nor the amplitude of miniature IPSCs in the presence of tetradotoxin and failed to change significantly the amplitude of evoked IPSCs recorded from CA1 pyramidal neurons. AVP increased the firing frequency of action potentials by depolarizing the GABAergic interneurons in the stratum radiatum of CA1 region. AVP-mediated depolarization of interneurons was mediated by inhibition of a background K(+) conductance which was insensitive to extracellular tetraethylammonium, Cs(+), 4-aminopyridine, tertiapin-Q and Ba(2+). AVP-induced depolarization of interneurons was dependent on Gα(q/11) but independent of phospholipase C, intracellular Ca(2+) release and protein kinase C. The inhibitory effects of AVP-mediated modulation of GABA release onto CA1 pyramidal neurons were overwhelmed by its strong excitation of CA1 pyramidal neurons in physiological condition but revealed when its direct excitation of the pyramidal neurons was blocked suggesting that AVP-mediated modulation of GABAergic transmission fine-tunes the excitability of CA1 pyramidal neurons.


Subject(s)
Hippocampus/metabolism , Neurons/metabolism , Receptors, Vasopressin/metabolism , Synaptic Transmission/physiology , Vasopressins/metabolism , gamma-Aminobutyric Acid/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Hippocampus/drug effects , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurons/drug effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Synapses/drug effects , Synapses/metabolism , Synaptic Transmission/drug effects , Vasopressins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...