Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
PLoS One ; 18(7): e0283299, 2023.
Article in English | MEDLINE | ID: mdl-37428799

ABSTRACT

Autism Spectrum Disorder (ASD) is a developmental disorder in which children display repetitive behavior, restricted range of interests, and atypical social interaction and communication. CUL3, coding for a Cullin family scaffold protein mediating assembly of ubiquitin ligase complexes through BTB domain substrate-recruiting adaptors, has been identified as a high-risk gene for autism. Although complete knockout of Cul3 results in embryonic lethality, Cul3 heterozygous mice have reduced CUL3 protein, demonstrate comparable body weight, and display minimal behavioral differences including decreased spatial object recognition memory. In measures of reciprocal social interaction, Cul3 heterozygous mice behaved similarly to their wild-type littermates. In area CA1 of hippocampus, reduction of Cul3 significantly increased mEPSC frequency but not amplitude nor baseline evoked synaptic transmission or paired-pulse ratio. Sholl and spine analysis data suggest there is a small yet significant difference in CA1 pyramidal neuron dendritic branching and stubby spine density. Unbiased proteomic analysis of Cul3 heterozygous brain tissue revealed dysregulation of various cytoskeletal organization proteins, among others. Overall, our results suggest that Cul3 heterozygous deletion impairs spatial object recognition memory, alters cytoskeletal organization proteins, but does not cause major hippocampal neuronal morphology, functional, or behavioral abnormalities in adult global Cul3 heterozygous mice.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Animals , Mice , Autism Spectrum Disorder/genetics , Autistic Disorder/genetics , Cullin Proteins/genetics , Cullin Proteins/metabolism , Protein Binding , Proteomics
2.
Adv Clin Exp Med ; 30(7): 727-736, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34118143

ABSTRACT

BACKGROUND: Increasing radiosensitivity of cancer cells can enhance the efficacy of cervical cancer treatment. OBJECTIVES: This study evaluated the potential roles and mechanism of baicalein in regulating the radiosensitivity of cervical cancer cells in vitro. MATERIAL AND METHODS: Real-time quantitative polymerase chain reaction (RT-qPCR) was used to assess miR-183 expression in End1/E6E7 cells, Hela cells and Hela cells irradiated with X-ray (0 Gy, 1 Gy, 3 Gy, 5 Gy, and 10 Gy). Cell Counting Kit-8 (CCK-8) method measured cell viability of Hela cells after miR-183 regulation, baicalein or RO8191 treatment. Apoptosis rates were detected using flow cytometry. Thereafter, expression of Bcl-2, Bax and caspase-3 RNA was also detected through RT-qPCR. Protein concentrations of E-cadherin, N-cadherin, Vimentin in epithelial-mesenchymal transition (EMT), phospho-JAK2/STAT3, and total Janus kinase 2/signal transducer and activator of transcription 3 STAT3 (JAK2/STAT3) were examined using enzyme-linked immunosorbent assay (ELISA) methods. RO8191, a JAK2/STAT3 activator, was used to activate the JAK2/STAT3 signaling pathway. RESULTS: The miR-183 expression was significantly lower in Hela cells compared to End1/E6E7 cells. Following upregulation of miR-183 in Hela cells, cell viability was inhibited while apoptosis was promoted. Moreover, EMT was inhibited after miR-183 over-expression. X-ray treatment markedly reduced the cell survival rate and increased miR-183 RNA expression. Baicalein treatment severely reduced the cell viability of 10-Gy X-ray-irradiated Hela cells, partially reversing the effect of miR-183, and also increased apoptosis and prevented EMT in irradiated cells. Y1007/8 in JAK2 and tyrosine (Tyr) residue 705 of STAT3 were phosphorylated, resulting in high expression of JAK2/STAT3, which was decreased by irradiation and baicalein treatment. RO8191 activated JAK2/STAT3 signaling, promoted cell viability and EMT, and inhibited cell apoptosis, while baicalein partly reversed the functions of RO8191. CONCLUSIONS: Baicalein inhibited cell viability and EMT, and induced cell apoptosis of Hela cells, through upregulating miR-183 via inactivation of the JAK2/STAT3 signaling pathway.


Subject(s)
MicroRNAs , Uterine Cervical Neoplasms , Apoptosis , Cell Proliferation , Female , Flavanones , HeLa Cells , Humans , Janus Kinase 2 , MicroRNAs/genetics , Naphthyridines , Oxadiazoles , Radiation Tolerance , STAT3 Transcription Factor , Signal Transduction , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/radiotherapy
3.
Neurobiol Learn Mem ; 167: 107132, 2020 01.
Article in English | MEDLINE | ID: mdl-31821881

ABSTRACT

Experience-dependent neuronal plasticity is a fundamental substrate of learning and memory. Intrinsic excitability is a form of neuronal plasticity that can be altered by learning and indicates the pattern of neuronal responding to external stimuli (e.g. a learning or synaptic event). Associative fear conditioning is one form of learning that alters intrinsic excitability, reflecting an experience-dependent change in neuronal function. After fear conditioning, intrinsic excitability changes are evident in brain regions that are a critical part of the fear circuit, including the amygdala, hippocampus, retrosplenial cortex, and prefrontal cortex. Some of these changes are transient and/or reversed by extinction as well as learning-specific (i.e. they are not observed in neurons from control animals). This review will explore how intrinsic neuronal excitability changes within brain structures that are critical for fear learning, and it will also discuss evidence promoting intrinsic excitability as a vital mechanism of associative fear memories. This work has raised interesting questions regarding the role of fear learning in changes of intrinsic excitability within specific subpopulations of neurons, including those that express immediate early genes and thus demonstrate experience-dependent activity, as well as in neurons classified as having a specific firing type (e.g. burst-spiking vs. regular-spiking). These findings have interesting implications for how intrinsic excitability can serve as a neural substrate of learning and memory, and suggest that intrinsic plasticity within specific subpopulations of neurons may promote consolidation of the memory trace in a flexible and efficient manner.


Subject(s)
Action Potentials , Brain/physiology , Conditioning, Classical/physiology , Fear/physiology , Memory/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Amygdala/physiology , Animals , Extinction, Psychological/physiology , Gyrus Cinguli/physiology , Hippocampus/physiology , Prefrontal Cortex/physiology
4.
PLoS Biol ; 17(10): e3000477, 2019 10.
Article in English | MEDLINE | ID: mdl-31600280

ABSTRACT

The striatum plays a fundamental role in motor learning and reward-related behaviors that are synergistically shaped by populations of D1 dopamine receptor (D1R)- and D2 dopamine receptor (D2R)-expressing medium spiny neurons (MSNs). How various neurotransmitter inputs converging on common intracellular pathways are parsed out to regulate distinct behavioral outcomes in a neuron-specific manner is poorly understood. Here, we reveal that distinct contributions of D1R-MSNs and D2R-MSNs towards reward and motor behaviors are delineated by the multifaceted signaling protein neurofibromin 1 (NF1). Using genetic mouse models, we show that NF1 in D1R-MSN modulates opioid reward, whereas loss of NF1 in D2R-MSNs delays motor learning by impeding the formation and consolidation of repetitive motor sequences. We found that motor learning deficits upon NF1 loss were associated with the disruption in dopamine signaling to cAMP in D2R-MSN. Restoration of cAMP levels pharmacologically or chemogenetically rescued the motor learning deficits seen upon NF1 loss in D2R-MSN. Our findings illustrate that multiplex signaling capabilities of MSNs are deployed at the level of intracellular pathways to achieve cell-specific control over behavioral outcomes.


Subject(s)
Corpus Striatum/physiology , Neurofibromin 1/metabolism , Neurons/physiology , Animals , Cyclic AMP/metabolism , Dopamine/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Motor Activity/physiology , Neurons/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Reward , Signal Transduction
5.
Science ; 365(6459): 1267-1273, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31416932

ABSTRACT

Opioids target the µ-opioid receptor (MOR) to produce unrivaled pain management, but their addictive properties can lead to severe abuse. We developed a whole-animal behavioral platform for unbiased discovery of genes influencing opioid responsiveness. Using forward genetics in Caenorhabditis elegans, we identified a conserved orphan receptor, GPR139, with anti-opioid activity. GPR139 is coexpressed with MOR in opioid-sensitive brain circuits, binds to MOR, and inhibits signaling to heterotrimeric guanine nucleotide-binding proteins (G proteins). Deletion of GPR139 in mice enhanced opioid-induced inhibition of neuronal firing to modulate morphine-induced analgesia, reward, and withdrawal. Thus, GPR139 could be a useful target for increasing opioid safety. These results also demonstrate the potential of C. elegans as a scalable platform for genetic discovery of G protein-coupled receptor signaling principles.


Subject(s)
Behavior, Animal , Caenorhabditis elegans/genetics , Nerve Tissue Proteins/genetics , Orphan Nuclear Receptors/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Opioid, mu/genetics , Analgesia , Animals , Animals, Genetically Modified , CRISPR-Cas Systems , Chromosome Mapping , Female , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Morphine/pharmacology , Neurons/drug effects , Signal Transduction
6.
J Biol Chem ; 294(35): 13145-13157, 2019 08 30.
Article in English | MEDLINE | ID: mdl-31311860

ABSTRACT

Stress profoundly affects physiological properties of neurons across brain circuits and thereby increases the risk for depression. However, the molecular and cellular mechanisms mediating these effects are poorly understood. In this study, we report that chronic physical restraint stress in mice decreases excitability specifically in layer 2/3 of pyramidal neurons within the prelimbic subarea of the prefrontal cortex (PFC) accompanied by the induction of depressive-like behavioral states. We found that a complex between G protein-coupled receptor (GPCR) 158 (GPR158) and regulator of G protein signaling 7 (RGS7), a regulatory GPCR signaling node recently discovered to be a key modulator of affective behaviors, plays a key role in controlling stress-induced changes in excitability in this neuronal population. Deletion of GPR158 or RGS7 enhanced excitability of layer 2/3 PFC neurons and prevented the impact of stress. Investigation of the underlying molecular mechanisms revealed that the A-type potassium channel Kv4.2 subunit is a molecular target of the GPR158-RGS7 complex. We further report that GPR158 physically associates with Kv4.2 channel and promotes its function by suppressing inhibitory modulation by cAMP-protein kinase A (PKA)-mediated phosphorylation. Taken together, our observations reveal a critical mechanism that adjusts neuronal excitability in L2/3 pyramidal neurons of the PFC and may thereby modulate the effects of stress on depression.


Subject(s)
Potassium Channels, Voltage-Gated/metabolism , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism , RGS Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Cells, Cultured , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RGS Proteins/deficiency , Receptors, G-Protein-Coupled/deficiency
7.
Neuropsychopharmacology ; 44(3): 642-653, 2019 02.
Article in English | MEDLINE | ID: mdl-30546127

ABSTRACT

Affective disorders arise from abnormal responses of the brain to prolonged exposure to challenging environmental stimuli. Recent work identified the orphan receptor GPR158 as a molecular link between chronic stress and depression. Here we reveal a non-canonical mechanism by which GPR158 exerts its effects on stress-induced depression by the complex formation with Regulator of G protein Signaling 7 (RGS7). Chronic stress promotes membrane recruitment of RGS7 via GPR158 in the medial prefrontal cortex (mPFC). The resultant complex suppresses homeostatic regulation of cAMP by inhibitory GPCRs in the region. Accordingly, RGS7 loss in mice induces an antidepressant-like phenotype and resiliency to stress, whereas its restoration within the mPFC is sufficient to rescue this phenotype in a GPR158-dependent way. These findings mechanistically link the unusual orphan receptor-RGS complex to a major stress mediator, the cAMP system and suggest new avenues for pharmacological interventions in affective disorders.


Subject(s)
Behavior, Animal/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Depression/metabolism , Homeostasis/physiology , Prefrontal Cortex/metabolism , RGS Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Stress, Psychological/metabolism , Animals , Depression/etiology , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , RGS Proteins/deficiency , Stress, Psychological/complications
8.
J Neurosci ; 38(32): 7120-7131, 2018 08 08.
Article in English | MEDLINE | ID: mdl-30006367

ABSTRACT

In the striatum, medium spiny neurons (MSNs) are heavily involved in controlling movement and reward. MSNs form two distinct populations expressing either dopamine receptor 1 (D1-MSN) or dopamine receptor 2 (D2-MSN), which differ in their projection targets and neurochemical composition. The activity of both types of MSNs is shaped by multiple neuromodulatory inputs processed by GPCRs that fundamentally impact their synaptic properties biasing behavioral outcomes. How these GPCR signaling cascades are regulated and what downstream targets they recruit in D1-MSN and D2-MSN populations are incompletely understood. In this study, we examined the cellular and molecular mechanisms underlying the action of RGS9-2, a key GPCR regulator in MSNs implicated in both movement control and actions of addictive drugs. Imaging cultured striatal neurons, we found that ablation of RGS9-2 significantly reduced calcium influx through NMDARs. Electrophysiological recordings in slices confirmed inhibition of NMDAR function in MSNs, resulting in enhanced AMPAR/NMDAR ratio. Accordingly, male mice lacking RGS9-2 displayed behavioral hypersensitivity to NMDAR blockade by MK-801 or ketamine. Recordings from genetically identified populations of striatal neurons revealed that these changes were selective to D2-MSNs. Surprisingly, we found that these postsynaptic effects resulted in remodeling of presynaptic inputs to D2-MSNs increasing the frequency of mEPSC and inhibiting paired-pulse ratio. Pharmacological dissection revealed that these adaptations were mediated by the NMDAR-dependent inhibition of retrograde endocannabinoid signaling from D2-MSNs to CB1 receptor on presynaptic terminals. Together, these data demonstrate a novel mechanism for pathway selective regulation of synaptic plasticity in MSNs controlled by GPCR signaling.SIGNIFICANCE STATEMENT This study identifies a role for a major G-protein regulator in controlling synaptic properties of striatal neurons in a pathway selective fashion.


Subject(s)
Corpus Striatum/physiology , Dopaminergic Neurons/physiology , RGS Proteins/physiology , Synaptic Transmission/physiology , Animals , Calcium Signaling , Cells, Cultured , Corpus Striatum/cytology , Dopaminergic Neurons/chemistry , Dopaminergic Neurons/classification , Dopaminergic Neurons/drug effects , Endocannabinoids/physiology , Exploratory Behavior , Female , Genes, Reporter , Glutamic Acid/metabolism , Male , Mice , Mice, Knockout , Patch-Clamp Techniques , Presynaptic Terminals/metabolism , RGS Proteins/deficiency , RGS Proteins/genetics , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/physiology , Receptors, Dopamine D2/analysis , Receptors, Dopamine D2/genetics , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology , Rotarod Performance Test , Synapses/physiology
9.
Neuroreport ; 29(10): 819-825, 2018 07 04.
Article in English | MEDLINE | ID: mdl-29672446

ABSTRACT

The ß-amyloid (Aß) deposition is one of the major pathological hallmark of Alzheimer's disease. Dysfunction in autophagy has been reported to lead to the Aß deposition. The current study aimed to investigate the effects of treadmill exercise on autophagy activity and the Aß deposition and to demonstrate whether exercise-induced reduction in the Aß deposition was associated with changes in autophagy activity. APP/PS1 transgenic mice were divided into transgenic sedentary (TG-SED, n=12) and transgenic exercise (TG-EXE, n=12) groups. Wild-type mice were also divided into sedentary (WT-SED, n=12) and exercise (WT-EXE, n=12) groups. The WT-EXE and TG-EXE mice were subjected to treadmill exercise for 12 weeks. The levels of Aß plaques and soluble forms of Aß, autophagy markers light chain 3 and P62, and lysosomal marker lysosome-associated membrane protein 1 (Lamp1) were measured in the hippocampus. Both Aß plaques and soluble forms of Aß (Aß40 and Aß42) were significantly increased in TG-SED mice compared with WT-SED mice, whereas exercise reduced Aß deposition in APP/PS1 transgenic mice. Coincidentally, TG-SED mice displayed a decrease in autophagy activity as evidenced by a significant increase in the levels of light chain 3-II and P62, as well as an accumulation of lysosome as evidenced by a significant over-expression of Lamp1. Interestingly, exercise increased autophagy activity as evidenced by a significant reduction in the levels of P62 and Lamp1 in TG-EXE mice. These findings suggest that treadmill exercise is efficient in decreasing Aß deposition by enhancing autophagy-lysosomal activity in APP/PS1 transgenic mice, demonstrating a possible approach in Alzheimer's disease prevention and treatment.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Autophagy/physiology , Hippocampus/metabolism , Physical Conditioning, Animal , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Mice, Transgenic , Physical Conditioning, Animal/methods , Plaque, Amyloid/pathology
10.
Elife ; 72018 02 08.
Article in English | MEDLINE | ID: mdl-29419376

ABSTRACT

Stress can be a motivational force for decisive action and adapting to novel environment; whereas, exposure to chronic stress contributes to the development of depression and anxiety. However, the molecular mechanisms underlying stress-responsive behaviors are not fully understood. Here, we identified the orphan receptor GPR158 as a novel regulator operating in the prefrontal cortex (PFC) that links chronic stress to depression. GPR158 is highly upregulated in the PFC of human subjects with major depressive disorder. Exposure of mice to chronic stress also increased GPR158 protein levels in the PFC in a glucocorticoid-dependent manner. Viral overexpression of GPR158 in the PFC induced depressive-like behaviors. In contrast GPR158 ablation, led to a prominent antidepressant-like phenotype and stress resiliency. We found that GPR158 exerts its effects via modulating synaptic strength altering AMPA receptor activity. Taken together, our findings identify a new player in mood regulation and introduce a pharmacological target for managing depression.


Subject(s)
Depression/physiopathology , Gene Expression Regulation , Prefrontal Cortex/physiology , Receptors, G-Protein-Coupled/metabolism , Stress, Psychological , Animals , Humans , Mice
11.
J Neurophysiol ; 119(1): 177-191, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28978762

ABSTRACT

Medial prefrontal cortex (mPFC) is critical for the expression of long-term conditioned fear. However, the neural circuits involving fear memory acquisition and retrieval are still unclear. Two subregions within mPFC that have received a lot of attention are the prelimbic (PL) and infralimbic (IL) cortices (e.g., Santini E, Quirk GJ, Porter JT. J Neurosci 28: 4028-4036, 2008; Song C, Ehlers VL, Moyer JR Jr J Neurosci 35: 13511-13524, 2015). Interestingly, PL and IL may play distinct roles during fear memory acquisition and retrieval but the underlying mechanism is poorly understood. One possibility is that the intrinsic membrane properties differ between these subregions. Thus, the current study was carried out to characterize the basic membrane properties of mPFC neurons in different layers and subregions. We found that pyramidal neurons in L2/3 were more hyperpolarized and less excitable than in L5. This was observed in both IL and PL and was associated with an enhanced h-current in L5 neurons. Within L2/3, IL neurons were more excitable than those in PL, which may be due to a lower spike threshold and higher input resistance in IL neurons. Within L5, the intrinsic excitability was comparable between neurons obtained in IL and PL. Thus, the heterogeneity in physiological properties of mPFC neurons may underlie the observed subregion-specific contribution of mPFC in cognitive function and emotional control, such as fear memory expression. NEW & NOTEWORTHY This is the first study to demonstrate that medial prefrontal cortical (mPFC) neurons are heterogeneous in both a layer- and a subregion-specific manner. Specifically, L5 neurons are more depolarized and more excitable than those neurons in L2/3, which is likely due to variations in h-current. Also, infralimbic neurons are more excitable than those of prelimbic neurons in layer 2/3, which may be due to differences in certain intrinsic properties, including input resistance and spike threshold.


Subject(s)
Prefrontal Cortex/cytology , Pyramidal Cells/physiology , Action Potentials , Animals , Fear , Male , Memory , Prefrontal Cortex/physiology , Pyramidal Cells/classification , Rats , Rats, Sprague-Dawley
12.
J Neurosci ; 35(39): 13511-24, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26424895

ABSTRACT

Neuronal activity in medial prefrontal cortex (mPFC) is critical for the formation of trace fear memory, yet the cellular mechanisms underlying these memories remain unclear. One possibility involves the modulation of intrinsic excitability within mPFC neurons that project to the basolateral complex of amygdala (BLA). The current study used a combination of retrograde labeling and in vitro whole-cell patch-clamp recordings to examine the effect of trace fear conditioning on the intrinsic excitability of layer 5 mPFC-BLA projection neurons in adult rats. Trace fear conditioning significantly enhanced the intrinsic excitability of regular spiking infralimbic (IL) projection neurons, as evidenced by an increase in the number of action potentials after current injection. These changes were also associated with a reduction in spike threshold and an increase in h current. In contrast, trace fear conditioning reduced the excitability of regular spiking prelimbic (PL) projection neurons, through a learning-related decrease of input resistance. Interestingly, the amount of conditioned freezing was (1) positively correlated with excitability of IL-BLA projection neurons after conditioning and (2) negatively correlated with excitability of PL-BLA projection neurons after extinction. Trace fear conditioning also significantly enhanced the excitability of burst spiking PL-BLA projection neurons. In both regions, conditioning-induced plasticity was learning specific (observed in conditioned but not in pseudoconditioned rats), flexible (reversed by extinction), and transient (lasted <10 d). Together, these data suggest that intrinsic plasticity within mPFC-BLA projection neurons occurs in a subregion- and cell-type-specific manner during acquisition, consolidation, and extinction of trace fear conditioning. Significance statement: Frontal lobe-related function is vital for a variety of important behaviors, some of which decline during aging. This study involves a novel combination of electrophysiological recordings from fluorescently labeled mPFC-to-amygdala projection neurons in rats with acquisition and extinction of trace fear conditioning to determine how specific neurons change during behavior. This is the first study to demonstrate that trace fear conditioning significantly alters the intrinsic excitability of mPFC-to-amygdala projection neurons in a subregion- and cell-type-specific manner, which is also transient and reversed by extinction. These data are of broad interest to the neuroscientific community, and the results will inspire additional studies investigating the cellular mechanisms underlying circuit-specific changes within the brain as a result of associative learning and memory.


Subject(s)
Amygdala/physiology , Conditioning, Classical/physiology , Fear/physiology , Limbic System/physiology , Neural Pathways/physiology , Prefrontal Cortex/physiology , Amygdala/cytology , Animals , Electrophysiological Phenomena/physiology , Extinction, Psychological , Learning/physiology , Limbic System/cytology , Male , Memory/physiology , Neural Pathways/cytology , Neuronal Plasticity/physiology , Patch-Clamp Techniques , Prefrontal Cortex/cytology , Psychomotor Performance/physiology , Rats , Rats, Inbred F344
13.
Neurobiol Learn Mem ; 105: 186-99, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23871744

ABSTRACT

"Use it or lose it" is a popular adage often associated with use-dependent enhancement of cognitive abilities. Much research has focused on understanding exactly how the brain changes as a function of experience. Such experience-dependent plasticity involves both structural and functional alterations that contribute to adaptive behaviors, such as learning and memory, as well as maladaptive behaviors, including anxiety disorders, phobias, and posttraumatic stress disorder. With the advancing age of our population, understanding how use-dependent plasticity changes across the lifespan may also help to promote healthy brain aging. A common misconception is that such experience-dependent plasticity (e.g., associative learning) is synonymous with synaptic plasticity. Other forms of plasticity also play a critical role in shaping adaptive changes within the nervous system, including intrinsic plasticity - a change in the intrinsic excitability of a neuron. Intrinsic plasticity can result from a change in the number, distribution or activity of various ion channels located throughout the neuron. Here, we review evidence that intrinsic plasticity is an important and evolutionarily conserved neural correlate of learning. Intrinsic plasticity acts as a metaplasticity mechanism by lowering the threshold for synaptic changes. Thus, learning-related intrinsic changes can facilitate future synaptic plasticity and learning. Such intrinsic changes can impact the allocation of a memory trace within a brain structure, and when compromised, can contribute to cognitive decline during the aging process. This unique role of intrinsic excitability can provide insight into how memories are formed and, more interestingly, how neurons that participate in a memory trace are selected. Most importantly, modulation of intrinsic excitability can allow for regulation of learning ability - this can prevent or provide treatment for cognitive decline not only in patients with clinical disorders but also in the aging population.


Subject(s)
Aging , Learning/physiology , Memory/physiology , Neuronal Plasticity , Neurons/physiology , Aging/physiology , Animals , Aplysia , Humans , Mice , Rats
14.
Neuroreport ; 24(6): 342-7, 2013 Apr 17.
Article in English | MEDLINE | ID: mdl-23518642

ABSTRACT

Chronic administration of D-galactose simulates the changes in natural senescence and accelerates aging in animal models and has been used in aging research. The present study was undertaken to investigate the molecular mechanisms underlying the effects of exercise on learning and memory in rats with D-galactose-induced aging. The learning and memory performance in aging rats, either after exercise or without exercise, was assessed with the Morris water maze test. The effect of treadmill exercise on the expression of amyloid-ß 42 and two key enzymes involved in processing of the ß-amyloid precursor protein, a disintegrase and metalloprotease domain 17 and ß-site amyloid precursor protein-cleaving enzyme 1, in the hippocampi of rats were monitored using real-time quantitative PCR. Moreover, oxidative stress-associated changes, including changes in superoxide dismutase activity and malondialdehyde content, in the hippocampi were assessed after exercise. Our results showed that treadmill exercise significantly improved learning and memory performance in aging rats. The behavioral changes were likely induced by repression of amyloid-ß 42 protein levels, through the upregulation of a disintegrase and metalloprotease domain 17 mRNA and downregulation of ß-site amyloid precursor protein-cleaving enzyme 1 mRNA, and a concomitant increase in superoxide dismutase activity and decrease in malondialdehyde content, in rat hippocampi. Our data suggest that exercise may be an effective therapy for alleviating learning and memory decline due to aging or the onset of neurodegenerative diseases.


Subject(s)
Aging/psychology , Amyloid/biosynthesis , Antioxidants/metabolism , Cognition/physiology , Physical Conditioning, Animal/psychology , ADAM Proteins/metabolism , ADAM17 Protein , Aging/drug effects , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Blotting, Western , Galactose , Hippocampus/metabolism , Male , Malondialdehyde/metabolism , Maze Learning/physiology , Memory Disorders/chemically induced , Memory Disorders/prevention & control , Memory Disorders/psychology , Peptide Fragments/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Running/physiology , Superoxide Dismutase/metabolism
15.
J Neurophysiol ; 107(12): 3397-408, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22442572

ABSTRACT

Experience-dependent synaptic and intrinsic plasticity are thought to be important substrates for learning-related changes in behavior. The present study combined trace fear conditioning with both extracellular and intracellular hippocampal recordings to study learning-related synaptic and intrinsic plasticity. Rats received one session of trace fear conditioning, followed by a brief conditioned stimulus (CS) test the next day. To relate behavioral performance with measures of hippocampal CA1 physiology, brain slices were prepared within 1 h of the CS test. In trace-conditioned rats, both synaptic plasticity and intrinsic excitability were significantly correlated with behavior such that better learning corresponded with enhanced long-term potentiation (LTP; r = 0.64, P < 0.05) and a smaller postburst afterhyperpolarization (AHP; r = -0.62, P < 0.05). Such correlations were not observed in pseudoconditioned rats, whose physiological data were comparable to those of poor learners and naive and chamber-exposed control rats. In addition, acquisition of trace fear conditioning did not enhance basal synaptic responses. Thus these data suggest that within the hippocampus both synaptic and intrinsic mechanisms are involved in the acquisition of trace fear conditioning.


Subject(s)
Conditioning, Psychological/physiology , Fear/physiology , Hippocampus/physiology , Neuronal Plasticity/physiology , Synapses/physiology , Animals , Long-Term Potentiation/physiology , Male , Memory/physiology , Rats , Rats, Inbred F344
16.
Neurosci Lett ; 399(1-2): 101-5, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16495001

ABSTRACT

It is widely known that hypocretins are essential for the regulation of wakefulness. Our recent reports have found that hypocretin-1 shows a direct postsynaptic excitatory effect on rat prefrontal cortex (PFC) pyramidal neurons. It remains unclear whether hypocretin-1 may interact with two classical neurotransmitter systems, glutamate and gamma-aminobutyric acid (GABA) in rat PFC. For this reason, we here investigated the modulatory actions of hypocretin-1 with these two transmitters on freshly isolated PFC pyramidal neurons using whole-cell patch-clamp recordings. We found that coadministration of hypocretin-1 and glutamate showed a synergistic effect on the recorded cells, and hypocretin-1 could excite the neurons even if GABA was present. Thus, our data suggest that there may be hypocretin-glutamate and hypocretin-GABA interactions in the PFC.


Subject(s)
Frontal Lobe/physiology , Glutamic Acid/physiology , Intracellular Signaling Peptides and Proteins/physiology , Neuropeptides/physiology , Pyramidal Cells/physiology , gamma-Aminobutyric Acid/physiology , Action Potentials , Animals , Frontal Lobe/cytology , Glutamic Acid/pharmacology , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/pharmacology , Neuropeptides/pharmacology , Orexins , Patch-Clamp Techniques , Pyramidal Cells/drug effects , Rats , Rats, Wistar , gamma-Aminobutyric Acid/pharmacology
17.
J Neurosci Res ; 82(5): 729-36, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16247802

ABSTRACT

Hypocretins are crucial for the regulation of wakefulness by the excitatory actions on multiple subcortical arousal systems. To date, there is little information about the direct postsynaptic excitatory effects of hypocretins on the neurons in prefrontal cortex (PFC), which is important for higher cognitive functions and is correlated with level of wakefulness. In this study, we tested the excitatory effects of hypocretin-1 on acutely isolated PFC pyramidal neurons of rats and studied the possible ionic mechanisms by using whole-cell patch-clamp techniques. Puff application of hypocretin-1 caused a dose-dependent excitation. Further observations that perfusion of Ca2+-free artificial cerebrospinal fluid did not influence the depolarizing effects of hypocretin-1, in conjunction with the findings that hypocretin-1 could decrease net whole-cell K+ currents, demonstrate that the excitatory effects of hypocretin-1 on PFC neurons are mediated by the inhibition of K+ currents but not Ca2+ influx. Finally, the decrease in K+ currents induced by hypocretin-1 was abolished by a protein kinase C (PKC) inhibitor (BIS II) or a phospholipase C (PLC) inhibitor (D609), suggesting that PKC and PLC appear to be involved in mediating the inhibitory effects of hypocretin-1 on K+ currents. These results indicate that hypocretin-1 exerts a postsynaptic excitatory action on PFC neurons through the inhibition of K+ currents, which probably results from activation of PKC and PLC signaling pathways.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Potassium Channels/metabolism , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism , Synaptic Transmission/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Animals, Newborn , Calcium Channels/drug effects , Calcium Channels/physiology , Calcium Signaling/drug effects , Calcium Signaling/physiology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/physiology , Excitatory Postsynaptic Potentials/drug effects , Intracellular Signaling Peptides and Proteins/pharmacology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neural Pathways/drug effects , Neural Pathways/metabolism , Neuropeptides/pharmacology , Orexins , Organ Culture Techniques , Patch-Clamp Techniques , Potassium Channels/drug effects , Prefrontal Cortex/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Pyramidal Cells/drug effects , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , Synaptic Transmission/drug effects , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism
18.
Neuroreport ; 16(13): 1529-33, 2005 Sep 08.
Article in English | MEDLINE | ID: mdl-16110284

ABSTRACT

We have investigated the direct excitatory effects of hypocretin-1 on acutely isolated prefrontal cortical pyramidal neurons and explored the signaling mechanisms of these actions. Puff application of hypocretin-1 caused an excitation in the recorded neurons. These effects of hypocretin-1 were abolished by a phospholipase C inhibitor D609, demonstrating that phospholipase C mediates the actions of hypocretin-1. A specific protein kinase C inhibitor, bisindolylmaleimide II, blocked the excitatory actions of hypocretin-1, suggesting that protein kinase C plays a key role. Finally, protein kinase A inhibitor applied intracellularly did not affect the responses. These results indicate that hypocretin-1 excites prefrontal neurons by activation of phospholipase C and protein kinase C pathways, but not protein kinase A.


Subject(s)
Intracellular Signaling Peptides and Proteins/pharmacology , Neuropeptides/pharmacology , Prefrontal Cortex/cytology , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Signal Transduction/physiology , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , In Vitro Techniques , Orexins , Patch-Clamp Techniques , Protein Kinase C/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Synapses/drug effects , Synapses/physiology , Type C Phospholipases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...