Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
bioRxiv ; 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38915638

ABSTRACT

In schizophrenia, layer 3 pyramidal neurons (L3PNs) in the dorsolateral prefrontal cortex (DLPFC) are thought to receive fewer excitatory synaptic inputs and to have lower expression levels of activity-dependent genes and of genes involved in mitochondrial energy production. In concert, these findings from previous studies suggest that DLPFC L3PNs are hypoactive in schizophrenia, disrupting the patterns of activity that are crucial for working memory, which is impaired in the illness. However, whether lower PN activity produces alterations in inhibitory and/or excitatory synaptic strength has not been tested in the primate DLPFC. Here, we decreased PN excitability in rhesus monkey DLPFC in vivo using adeno-associated viral vectors (AAVs) to produce Cre recombinase-mediated overexpression of Kir2.1 channels, a genetic silencing tool that efficiently decreases neuronal excitability. In acute slices prepared from DLPFC 7-12 weeks post-AAV microinjections, Kir2.1-overexpressing PNs had a significantly reduced excitability largely attributable to highly specific effects of the AAV-encoded Kir2.1 channels. Moreover, recordings of synaptic currents showed that Kir2.1-overexpressing DLPFC PNs had reduced strength of excitatory synapses whereas inhibitory synaptic inputs were not affected. The decrease in excitatory synaptic strength was not associated with changes in dendritic spine number, suggesting that excitatory synapse quantity was unaltered in Kir2.1-overexpressing DLPFC PNs. These findings suggest that, in schizophrenia, the excitatory synapses on hypoactive L3PNs are weaker and thus might represent a substrate for novel therapeutic interventions. Significance Statement: In schizophrenia, dorsolateral prefrontal cortex (DLPFC) pyramidal neurons (PNs) have both transcriptional and structural alterations that suggest they are hypoactive. PN hypoactivity is thought to produce synaptic alterations in schizophrenia, however the effects of lower neuronal activity on synaptic function in primate DLPFC have not been examined. Here, we used, for the first time in primate neocortex, adeno-associated viral vectors (AAVs) to reduce PN excitability with Kir2.1 channel overexpression and tested if this manipulation altered the strength of synaptic inputs onto the Kir2.1-overexpressing PNs. Recordings in DLPFC slices showed that Kir2.1 overexpression depressed excitatory (but not inhibitory), synaptic currents, suggesting that, in schizophrenia, the hypoactivity of PNs might be exacerbated by reduced strength of the excitatory synapses they receive.

2.
Cereb Cortex ; 33(12): 7754-7770, 2023 06 08.
Article in English | MEDLINE | ID: mdl-36971419

ABSTRACT

In primates, the dorsolateral prefrontal (DLPFC) and posterior parietal (PPC) cortices are key nodes in the working memory network. The working memory-related gamma oscillations induced in these areas, predominantly in layer 3, exhibit higher frequency in DLPFC. Although these regional differences in oscillation frequency are likely essential for information transfer between DLPFC and PPC, the mechanisms underlying these differences remain poorly understood. We investigated, in rhesus monkey, the DLPFC and PPC layer 3 pyramidal neuron (L3PN) properties that might regulate oscillation frequency and assessed the effects of these properties simulating oscillations in computational models. We found that GABAAR-mediated synaptic inhibition synchronizes L3PNs in both areas, but analysis of GABAAR mRNA levels and inhibitory synaptic currents suggested similar mechanisms of inhibition-mediated synchrony in DLPFC and PPC. Basal dendrite spine density and AMPAR/NMDAR mRNA levels were higher in DLPFC L3PNs, whereas excitatory synaptic currents were similar between areas. Therefore, synaptically evoked excitation might be stronger in DLPFC L3PNs due to a greater quantity of synapses in basal dendrites, a main target of recurrent excitation. Simulations in computational networks showed that oscillation frequency and power increased with increasing recurrent excitation, suggesting a mechanism by which the DLPFC-PPC differences in oscillation properties are generated.


Subject(s)
Prefrontal Cortex , Receptors, GABA-A , Animals , Prefrontal Cortex/physiology , Pyramidal Cells/physiology , Parietal Lobe , Primates
3.
Biol Psychiatry ; 94(4): 288-296, 2023 08 15.
Article in English | MEDLINE | ID: mdl-36736420

ABSTRACT

BACKGROUND: In schizophrenia, layer 3 pyramidal neurons (L3PNs) of the dorsolateral prefrontal cortex exhibit deficits in markers of excitatory synaptic inputs that are thought to disrupt the patterns of neural network activity essential for cognitive function. These deficits are usually interpreted under Irwin Feinberg's hypothesis of altered synaptic pruning, which postulates that normal periadolescent pruning, thought to preferentially eliminate weak/immature synapses, is altered in schizophrenia. However, it remains unknown whether periadolescent pruning on L3PNs in the primate dorsolateral prefrontal cortex selectively eliminates weak excitatory synapses or uniformly eliminates excitatory synapses across the full distribution of synaptic strengths. METHODS: To distinguish between these alternative models of synaptic pruning, we assessed the densities of dendritic spines, the site of most excitatory inputs to L3PNs, and the distributions of excitatory synaptic strengths in dorsolateral prefrontal cortex L3PNs from male and female monkeys across the periadolescent period of synaptic pruning. We used patch-clamp methods in acute brain slices to record miniature excitatory synaptic currents and intracellular filling with biocytin to quantify dendritic spines. RESULTS: On L3PNs, dendritic spines exhibited the expected age-related decline in density, but mean synaptic strength and the shape of synaptic strength distributions remained stable with age. CONCLUSIONS: The absence of age-related differences in mean synaptic strength and synaptic strength distributions supports the model of a uniform pattern of synaptic pruning across the full range of synaptic strengths. The implications of these findings for the pathogenesis and functional consequences of dendritic spine deficits in schizophrenia are discussed.


Subject(s)
Schizophrenia , Animals , Male , Female , Haplorhini , Pyramidal Cells/physiology , Prefrontal Cortex , Synapses/physiology , Neuronal Plasticity , Dendritic Spines/physiology
4.
Cereb Cortex ; 32(19): 4229-4242, 2022 09 19.
Article in English | MEDLINE | ID: mdl-34937087

ABSTRACT

Although music is one of human-unique traits such as language, its neural basis for cortical organization has not been well understood. In the present functional magnetic resonance imaging study, we tested an error-detection task with different types of musical error (pitch, tempo, stress, and articulation conditions) and examined three groups of secondary school students having different levels of music experience. First, we observed distinct activation patterns under these music conditions, such that specific activations under the pitch condition were consistently replicated for all tested groups in the auditory areas, as well as in the left language areas under the articulation condition. Second, music-experience-related activations were observed in multiple regions, including the right sensorimotor area under the pitch condition, as well as in the right premotor cortex under the articulation condition. Indeed, the right homologs of the language areas were specifically activated under the stress and articulation conditions. Third, activations specific to the group with the highest proficiency in music were observed under the tempo condition mostly in the right regions. These results demonstrate the existence of music-related signatures in the brain activations, including both universal and experience-related mechanisms.


Subject(s)
Music , Brain/diagnostic imaging , Brain/physiology , Brain Mapping , Humans , Language , Magnetic Resonance Imaging/methods
5.
Neurobiol Dis ; 155: 105382, 2021 07.
Article in English | MEDLINE | ID: mdl-33940180

ABSTRACT

The unique fast spiking (FS) phenotype of cortical parvalbumin-positive (PV) neurons depends on the expression of multiple subtypes of voltage-gated potassium channels (Kv). PV neurons selectively express Kcns3, the gene encoding Kv9.3 subunits, suggesting that Kcns3 expression is critical for the FS phenotype. KCNS3 expression is lower in PV neurons in the neocortex of subjects with schizophrenia, but the effects of this alteration are unclear, because Kv9.3 subunit function is poorly understood. Therefore, to assess the role of Kv9.3 subunits in PV neuron function, we combined gene expression analyses, computational modeling, and electrophysiology in acute slices from the cortex of Kcns3-deficient mice. Kcns3 mRNA levels were ~ 50% lower in cortical PV neurons from Kcns3-deficient relative to wildtype mice. While silent per se, Kv9.3 subunits are believed to amplify the Kv2.1 current in Kv2.1-Kv9.3 channel complexes. Hence, to assess the consequences of reducing Kv9.3 levels, we simulated the effects of decreasing the Kv2.1-mediated current in a computational model. The FS cell model with reduced Kv2.1 produced spike trains with irregular inter-spike intervals, or stuttering, and greater Na+ channel inactivation. As in the computational model, PV basket cells (PVBCs) from Kcns3-deficient mice displayed spike trains with strong stuttering, which depressed PVBC firing. Moreover, Kcns3 deficiency impaired the recruitment of PVBC firing at gamma frequency by stimuli mimicking synaptic input observed during cortical UP states. Our data indicate that Kv9.3 subunits are critical for PVBC physiology and suggest that KCNS3 deficiency in schizophrenia could impair PV neuron firing, possibly contributing to deficits in cortical gamma oscillations in the illness.


Subject(s)
Action Potentials/physiology , Neurons/physiology , Parvalbumins/physiology , Potassium Channels, Voltage-Gated/deficiency , Prefrontal Cortex/physiopathology , Schizophrenia/physiopathology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Culture Techniques , Potassium Channels, Voltage-Gated/genetics , Schizophrenia/genetics
6.
J Neurosci ; 39(37): 7277-7290, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31341029

ABSTRACT

In primates, working memory function depends on activity in a distributed network of cortical areas that display different patterns of delay task-related activity. These differences are correlated with, and might depend on, distinctive properties of the neurons located in each area. For example, layer 3 pyramidal neurons (L3PNs) differ significantly between primary visual and dorsolateral prefrontal (DLPFC) cortices. However, to what extent L3PNs differ between DLPFC and other association cortical areas is less clear. Hence, we compared the properties of L3PNs in monkey DLPFC versus posterior parietal cortex (PPC), a key node in the cortical working memory network. Using patch-clamp recordings and biocytin cell filling in acute brain slices, we assessed the physiology and morphology of L3PNs from monkey DLPFC and PPC. The L3PN transcriptome was studied using laser microdissection combined with DNA microarray or quantitative PCR. We found that in both DLPFC and PPC, L3PNs were divided into regular spiking (RS-L3PNs) and bursting (B-L3PNs) physiological subtypes. Whereas regional differences in single-cell excitability were modest, B-L3PNs were rare in PPC (RS-L3PN:B-L3PN, 94:6), but were abundant in DLPFC (50:50), showing greater physiological diversity. Moreover, DLPFC L3PNs display larger and more complex basal dendrites with higher dendritic spine density. Additionally, we found differential expression of hundreds of genes, suggesting a transcriptional basis for the differences in L3PN phenotype between DLPFC and PPC. These data show that the previously observed differences between DLPFC and PPC neuron activity during working memory tasks are associated with diversity in the cellular/molecular properties of L3PNs.SIGNIFICANCE STATEMENT In the human and nonhuman primate neocortex, layer 3 pyramidal neurons (L3PNs) differ significantly between dorsolateral prefrontal (DLPFC) and sensory areas. Hence, L3PN properties reflect, and may contribute to, a greater complexity of computations performed in DLPFC. However, across association cortical areas, L3PN properties are largely unexplored. We studied the physiology, dendrite morphology and transcriptome of L3PNs from macaque monkey DLPFC and posterior parietal cortex (PPC), two key nodes in the cortical working memory network. L3PNs from DLPFC had greater diversity of physiological properties and larger basal dendrites with higher spine density. Moreover, transcriptome analysis suggested a molecular basis for the differences in the physiological and morphological phenotypes of L3PNs from DLPFC and PPC.


Subject(s)
Neocortex/physiology , Parietal Lobe/physiology , Prefrontal Cortex/physiology , Pyramidal Cells/physiology , Action Potentials/physiology , Animals , Female , Laser Capture Microdissection/methods , Macaca mulatta , Male , Neocortex/cytology , Organ Culture Techniques , Parietal Lobe/cytology , Prefrontal Cortex/cytology
7.
Brain Nerve ; 70(6): 617-631, 2018 Jun.
Article in Japanese | MEDLINE | ID: mdl-29887531

ABSTRACT

Musical performance is a highly complicated task, requiring precise regulation and organization of sensorimotor system under high order cognitive functions of the human brain. The recent development of non-invasive measurements of brain structure and function, such as magnetic resonance imaging, in addition to conventional assessments, such as electrophysiology, and behavioral or cognitive tasks allows us to study short- and long-term brain plasticity associated with musical training. This article focuses on cross-sectional and longitudinal studies of musicians and indivisuals participating in music classes, reviews findings in terms of plastic and meta-plastic changes in the human brain induced by musical training over short and long period of time, and describes studies revealing transfer learning effects of musical training.


Subject(s)
Brain/physiology , Learning , Music , Neuronal Plasticity , Humans
8.
Biol Psychiatry ; 84(6): 460-470, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29523414

ABSTRACT

BACKGROUND: Testing hypotheses regarding the role of N-methyl-D-aspartate receptor (NMDAR) hypofunction in schizophrenia requires understanding the mechanisms of NMDAR regulation of prefrontal cortex (PFC) circuit function. NMDAR antagonists are thought to produce pyramidal cell (PC) disinhibition. However, inhibitory parvalbumin-positive basket cells (PVBCs) have modest NMDAR-mediated excitatory drive and thus are unlikely to participate in NMDAR antagonist-mediated disinhibition. Interestingly, recent studies demonstrated that presynaptic NMDARs enhance transmitter release at central synapses. Thus, if presynaptic NMDARs enhance gamma-aminobutyric acid release at PVBC-to-PC synapses, they could participate in NMDAR-dependent PC disinhibition. Here, we examined whether presynaptic NMDAR effects could modulate gamma-aminobutyric acid release at PVBC-to-PC synapses in mouse PFC. METHODS: Using whole-cell recordings from synaptically connected pairs in mouse PFC, we determined whether NMDA or NMDAR antagonist application affects PVBC-to-PC inhibition in a manner consistent with a presynaptic mechanism. RESULTS: NMDAR activation enhanced by ∼40% the synaptic current at PVBC-to-PC pairs. This effect was consistent with a presynaptic mechanism given that it was 1) observed with postsynaptic NMDARs blocked by intracellular MK801, 2) associated with a lower rate of transmission failures and a higher transmitter release probability, and 3) blocked by intracellular MK801 in the PVBC. NMDAR antagonist application did not affect the synaptic currents in PVBC-to-PC pairs, but it reduced the inhibitory currents elicited in PCs with simultaneous glutamate release by extracellular stimulation. CONCLUSIONS: We demonstrate that NMDAR activation enhances PVBC-to-PC inhibition in a manner consistent with presynaptic mechanisms, and we suggest that the functional impact of this presynaptic effect depends on the activity state of the PFC network.


Subject(s)
Prefrontal Cortex/cytology , Pyramidal Cells/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Synapses/metabolism , Animals , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Female , Male , Mice , Organ Culture Techniques , Parvalbumins , Patch-Clamp Techniques , Pyramidal Cells/metabolism , gamma-Aminobutyric Acid/metabolism
9.
eNeuro ; 5(5)2018.
Article in English | MEDLINE | ID: mdl-30713994

ABSTRACT

Cholinergic neuromodulation is thought to shape network activity in the PFC, and thus PFC-dependent cognitive functions. ACh may modulate the activity of parvalbumin-positive (PV+) neurons, which critically regulate cortical network function. However, the mechanisms of cholinergic regulation of PV+ neuron activity, and particularly of the basket cell (BC) versus chandelier cell (ChC) subtypes, are unclear. Using patch clamp recordings in acute slices, we examined the effects of the ACh receptor (AChR) agonist carbachol on the excitatory synaptic drive onto BCs or ChCs in layers 2 to 6 of mouse PFC. Carbachol increased the frequency and amplitude of spontaneous EPSCs (sEPSCs) recorded from PV+ BCs in layers 3-6, but not in BCs from layer 2. Moreover, carbachol did not change the sEPSCs in ChCs, which were located exclusively in layer 2. The potentiation of sEPSCs in layers 3-6 BCs was prevented by the Na+ channel blocker tetrodotoxin and was abolished by the M1-selective muscarinic AChR antagonist pirenzepine. Thus, carbachol potentiates the activity-dependent excitatory drive onto PV+ neurons via M1-muscarinic AChR activation in a cell type- and layer-specific manner. In current clamp recordings with synaptic transmission blocked, carbachol directly evoked firing in deep layer pyramidal neurons (PNs). In contrast, carbachol elicited deep layer BC firing indirectly, via glutamate-mediated synaptic drive. Our data suggest that ACh powerfully regulates PFC microcircuit function by facilitating the firing of PNs that synaptically recruit deep layer PV+ BC activity, possibly shaping the patterns of network activity that contribute to cognitive function.


Subject(s)
Parvalbumins/pharmacology , Prefrontal Cortex/drug effects , Pyramidal Cells/drug effects , Synaptic Transmission/drug effects , Animals , Animals, Newborn , Carbachol/pharmacology , Cholinergic Agents/pharmacology , Female , Glutamic Acid/pharmacology , Male , Mice , Muscarinic Antagonists/pharmacology , Parvalbumins/metabolism , Prefrontal Cortex/metabolism , Pyramidal Cells/physiology , Synapses/drug effects , Synapses/metabolism
10.
J Neurosci ; 37(19): 4883-4902, 2017 05 10.
Article in English | MEDLINE | ID: mdl-28408413

ABSTRACT

Parvalbumin-positive (PV+) neurons control the timing of pyramidal cell output in cortical neuron networks. In the prefrontal cortex (PFC), PV+ neuron activity is involved in cognitive function, suggesting that PV+ neuron maturation is critical for cognitive development. The two major PV+ neuron subtypes found in the PFC, chandelier cells (ChCs) and basket cells (BCs), are thought to play different roles in cortical circuits, but the trajectories of their physiological maturation have not been compared. Using two separate mouse lines, we found that in the mature PFC, both ChCs and BCs are abundant in superficial layer 2, but only BCs are present in deeper laminar locations. This distinctive laminar distribution was observed by postnatal day 12 (P12), when we first identified ChCs by the presence of axon cartridges. Electrophysiology analysis of excitatory synapse development, starting at P12, showed that excitatory drive remains low throughout development in ChCs, but increases rapidly before puberty in BCs, with an earlier time course in deeper-layer BCs. Consistent with a role of excitatory synaptic drive in the maturation of PV+ neuron firing properties, the fast-spiking phenotype showed different maturation trajectories between ChCs and BCs, and between superficial versus deep-layer BCs. ChC and BC maturation was nearly completed, via different trajectories, before the onset of puberty. These findings suggest that ChC and BC maturation may contribute differentially to the emergence of cognitive function, primarily during prepubertal development.SIGNIFICANCE STATEMENT Parvalbumin-positive (PV+) neurons tightly control pyramidal cell output. Thus PV+ neuron maturation in the prefrontal cortex (PFC) is crucial for cognitive development. However, the relative physiological maturation of the two major subtypes of PV+ neurons, chandelier cells (ChCs) and basket cells (BCs), has not been determined. We assessed the maturation of ChCs and BCs in different layers of the mouse PFC, and found that, from early postnatal age, ChCs and BCs differ in laminar location. Excitatory synapses and fast-spiking properties matured before the onset of puberty in both cell types, but following cell type-specific developmental trajectories. Hence, the physiological maturation of ChCs and BCs may contribute to the emergence of cognitive function differentially, and predominantly during prepubertal development.


Subject(s)
Aging/physiology , Neurogenesis/physiology , Neurons/physiology , Parvalbumins/metabolism , Prefrontal Cortex/physiology , Aging/pathology , Animals , Animals, Newborn , Female , Male , Mice , Mice, Inbred C57BL , Nerve Net/cytology , Nerve Net/physiology , Neurons/classification , Neurons/cytology , Prefrontal Cortex/cytology
11.
J Neurophysiol ; 113(6): 1850-61, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25540225

ABSTRACT

In rodent cortex GABAA receptor (GABAAR)-mediated synapses are a significant source of input onto GABA neurons, and the properties of these inputs vary among GABA neuron subtypes that differ in molecular markers and firing patterns. Some features of cortical interneurons are different between rodents and primates, but it is not known whether inhibition of GABA neurons is prominent in the primate cortex and, if so, whether these inputs show heterogeneity across GABA neuron subtypes. We thus studied GABAAR-mediated miniature synaptic events in GABAergic interneurons in layer 3 of monkey dorsolateral prefrontal cortex (DLPFC). Interneurons were identified on the basis of their firing pattern as fast spiking (FS), regular spiking (RS), burst spiking (BS), or irregular spiking (IS). Miniature synaptic events were common in all of the recorded interneurons, and the frequency of these events was highest in FS neurons. The amplitude and kinetics of miniature inhibitory postsynaptic potentials (mIPSPs) also differed between DLPFC interneuron subtypes in a manner correlated with their input resistance and membrane time constant. FS neurons had the fastest mIPSP decay times and the strongest effects of the GABAAR modulator zolpidem, suggesting that the distinctive properties of inhibitory synaptic inputs onto FS cells are in part conferred by GABAARs containing α1 subunits. Moreover, mIPSCs differed between FS and RS interneurons in a manner consistent with the mIPSP findings. These results show that in the monkey DLPFC GABAAR-mediated synaptic inputs are prominent in layer 3 interneurons and may differentially regulate the activity of different interneuron subtypes.


Subject(s)
Action Potentials , GABAergic Neurons/metabolism , Prefrontal Cortex/metabolism , Synapses/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Female , GABA-A Receptor Agonists/pharmacology , GABAergic Neurons/drug effects , GABAergic Neurons/physiology , Inhibitory Postsynaptic Potentials , Interneurons/drug effects , Interneurons/metabolism , Interneurons/physiology , Macaca mulatta , Male , Miniature Postsynaptic Potentials , Prefrontal Cortex/cytology , Prefrontal Cortex/physiology , Pyridines/pharmacology , Synapses/drug effects , Synapses/physiology , Zolpidem
12.
Cereb Cortex ; 25(11): 4076-93, 2015 Nov.
Article in English | MEDLINE | ID: mdl-24904071

ABSTRACT

Development of inhibition onto pyramidal cells may be crucial for the emergence of cortical network activity, including gamma oscillations. In primate dorsolateral prefrontal cortex (DLPFC), inhibitory synaptogenesis starts in utero and inhibitory synapse density reaches adult levels before birth. However, in DLPFC, the expression levels of γ-aminobutyric acid (GABA) synapse-related gene products changes markedly during development until young adult age, suggesting a highly protracted maturation of GABA synapse function. Therefore, we examined the development of GABA synapses by recording GABAAR-mediated inhibitory postsynaptic currents (GABAAR-IPSCs) from pyramidal cells in the DLPFC of neonatal, prepubertal, peripubertal, and adult macaque monkeys. We found that the decay of GABAAR-IPSCs, possibly including those from parvalbumin-positive GABA neurons, shortened by prepubertal age, while their amplitude increased until the peripubertal period. Interestingly, both GABAAR-mediated quantal response size, estimated by miniature GABAAR-IPSCs, and the density of GABAAR synaptic appositions, measured with immunofluorescence microscopy, were stable with age. Simulations in a computational model network with constant GABA synapse density showed that the developmental changes in GABAAR-IPSC properties had a significant impact on oscillatory activity and predicted that, whereas DLPFC circuits can generate gamma frequency oscillations by prepubertal age, mature levels of gamma band power are attained at late stages of development.


Subject(s)
Inhibitory Postsynaptic Potentials/physiology , Prefrontal Cortex/cytology , Prefrontal Cortex/growth & development , Synapses/physiology , gamma-Aminobutyric Acid/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Age Factors , Animals , Animals, Newborn , Calcium Channel Blockers/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , GABA Antagonists/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Lysine/analogs & derivatives , Lysine/metabolism , Macaca mulatta , Models, Neurological , Neurons/drug effects , Pyridazines/pharmacology , Synapses/drug effects , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology , omega-Agatoxin IVA/pharmacology
13.
J Physiol ; 591(19): 4725-48, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23818693

ABSTRACT

Cholinergic neuromodulation in neocortical networks is required for gamma oscillatory activity associated with working memory and other cognitive processes. Importantly, the cholinergic agonist carbachol (CCh) induces gamma oscillations in vitro, via mechanisms that may be shared with in vivo gamma oscillations and that are consistent with the pyramidal interneuron network gamma (PING) model. In PING oscillations, pyramidal cells (PCs), driven by asynchronous excitatory input, recruit parvalbumin-positive fast-spiking interneurons (FSNs), which then synchronize the PCs via feedback inhibition. Whereas the PING model is favoured by current data, how cholinergic neuromodulation contributes to gamma oscillation production is poorly understood. We thus studied the effects of cholinergic modulation on circuit components of the PING model in mouse medial prefrontal cortex (mPFC) brain slices. CCh depolarized and evoked action potential firing in a fraction of PCs and increased excitatory synaptic input onto FSNs. In synaptically connected pairs, CCh reduced the short-term depression at FSN-PC and PC-FSN synapses, equalizing synaptic strength during repetitive presynaptic firing while simultaneously increasing the failure probability. Interestingly, when PCs or FSNs fired in response to gamma frequency oscillatory inputs, CCh increased the firing probability per cycle. Combined with the equalization of synaptic strength, an increase by CCh in the fraction of neurons recruited per oscillation cycle may support oscillatory synchrony of similar strength during relatively long oscillation episodes such as those observed during working memory tasks, suggesting a significant functional impact of cholinergic modulation of mPFC circuit components crucial for the PING model.


Subject(s)
Action Potentials , Cholinergic Agonists/pharmacology , Cholinergic Neurons/physiology , Interneurons/physiology , Prefrontal Cortex/physiology , Pyramidal Cells/physiology , Animals , Cholinergic Neurons/drug effects , Excitatory Postsynaptic Potentials , Interneurons/drug effects , Memory, Short-Term , Mice , Nerve Net/drug effects , Nerve Net/physiology , Prefrontal Cortex/cytology , Prefrontal Cortex/drug effects , Pyramidal Cells/drug effects
14.
J Neurosci ; 31(46): 16603-10, 2011 Nov 16.
Article in English | MEDLINE | ID: mdl-22090488

ABSTRACT

The ability to detect harmful chemicals rapidly is essential for the survival of all animals. In Caenorhabditis elegans (C. elegans), repellents trigger an avoidance response, causing animals to move away from repellents. Dihydrocaffeic acid (DHCA) is a water-soluble repellent and nonflavonoid catecholic compound that can be found in plant products. Using a Xenopus laevis (X. laevis) oocyte expression system, we identified a candidate dihydrocaffeic acid receptor (DCAR), DCAR-1. DCAR-1 is a novel seven-transmembrane protein that is expressed in the ASH avoidance sensory neurons of C. elegans. dcar-1 mutant animals are defective in avoidance response to DHCA, and cell-specific expression of dcar-1 in the ASH neurons of dcar-1 mutant animals rescued the defect in avoidance response to DHCA. Our findings identify DCAR-1 as the first seven-transmembrane receptor required for avoidance of a water-soluble repellent, DHCA, in C. elegans.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caffeic Acids/pharmacology , Escape Reaction/drug effects , Receptors, G-Protein-Coupled/metabolism , 3,4-Dihydroxyphenylacetic Acid/pharmacology , Analysis of Variance , Animals , Animals, Genetically Modified , Behavior, Animal/drug effects , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Catechols/pharmacology , Cloning, Molecular/methods , Dose-Response Relationship, Drug , Escape Reaction/physiology , Hydroxybenzoates , Larva , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Potentials/drug effects , Membrane Potentials/genetics , Microinjections/methods , Models, Molecular , Mutation/genetics , Receptors, G-Protein-Coupled/genetics , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , Xenopus
15.
J Physiol ; 589(Pt 20): 4857-84, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21807615

ABSTRACT

Depolarization-induced suppression of inhibition (DSI) is a prevailing form of endocannabinoid signalling. However, several discrepancies have arisen regarding the roles played by the two major brain endocannabinoids, 2-arachidonoylglycerol (2-AG) and anandamide, in mediating DSI. Here we studied endocannabinoid signalling in the prefrontal cortex (PFC), where several components of the endocannabinoid system have been identified, but endocannabinoid signalling remains largely unexplored. In voltage clamp recordings from mouse PFC pyramidal neurons, depolarizing steps significantly suppressed IPSCs induced by application of the cholinergic agonist carbachol. DSI in PFC neurons was abolished by extra- or intracellular application of tetrahydrolipstatin (THL), an inhibitor of the 2-AG synthesis enzyme diacylglycerol lipase (DAGL). Moreover, DSI was enhanced by inhibiting 2-AG degradation, but was unaffected by inhibiting anandamide degradation. THL, however, may affect other enzymes of lipid metabolism and does not selectively target the α (DAGLα) or ß (DAGLß) isoforms of DAGL. Therefore, we studied DSI in the PFC of DAGLα(-/-) and DAGLß(-/-) mice generated via insertional mutagenesis by gene-trapping with retroviral vectors. Gene trapping strongly reduced DAGLα or DAGLß mRNA levels in a locus-specific manner. In DAGLα(-/-) mice cortical levels of 2-AG were significantly decreased and DSI was completely abolished, whereas DAGLß deficiency did not alter cortical 2-AG levels or DSI. Importantly, cortical levels of anandamide were not significantly affected in DAGLα(-/-) or DAGLß(-/-) mice. The chronic decrease of 2-AG levels in DAGLα(-/-) mice did not globally alter inhibitory transmission or the response of cannabinoid-sensitive synapses to cannabinoid receptor stimulation, although it altered some intrinsic membrane properties. Finally, we found that repetitive action potential firing of PFC pyramidal neurons suppressed synaptic inhibition in a DAGLα-dependent manner. These results show that DSI is a prominent form of endocannabinoid signalling in PFC circuits. Moreover, the close agreement between our pharmacological and genetic studies indicates that 2-AG synthesized by postsynaptic DAGLα mediates DSI in PFC neurons.


Subject(s)
Arachidonic Acids/physiology , Cannabinoid Receptor Modulators/physiology , Endocannabinoids , Glycerides/physiology , Inhibitory Postsynaptic Potentials/physiology , Lipoprotein Lipase/physiology , Neural Inhibition/physiology , Prefrontal Cortex/physiology , Animals , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Isoenzymes/antagonists & inhibitors , Isoenzymes/deficiency , Isoenzymes/physiology , Lipoprotein Lipase/antagonists & inhibitors , Lipoprotein Lipase/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Inhibition/drug effects , Patch-Clamp Techniques , Polyunsaturated Alkamides , Prefrontal Cortex/drug effects , Pyramidal Cells/drug effects , Pyramidal Cells/physiology
16.
Hippocampus ; 21(12): 1290-301, 2011 Dec.
Article in English | MEDLINE | ID: mdl-20824728

ABSTRACT

Immediate early genes (IEGs) typically are the first genetic responders to a variety of cellular activations. The IEG that encodes activity-regulated cytoskeleton-associated protein (arc/arg3.1) has attracted much interest because its mRNA is transported to and translated near activated synapses. Moreover, arc has been implicated in both long-term potentiation (LTP) and long-term depression (LTD). However, little is known about the time course of altered arc expression during LTP and LTD. Here we characterized arc mRNA levels in area CA1 of the adult rat hippocampus in vivo after LTP- and LTD-inducing stimulations that were identical, except for the temporal patterning of the stimulation pulses. We observed a persistent increase in arc mRNA level during LTP. In contrast, during LTD, arc mRNA level first was decreased and then transiently increased relative to control level. These findings demonstrate that arc mRNA is regulated differently during LTP and LTD, and they provide evidence for stimulation-induced downregulation of mRNA availability during LTD. Findings of abbreviated LTD when transcription was inhibited indicate that the prolonged maintenance of the type of N-methyl-D-aspartate receptor-dependent LTD studied here requires de novo transcription. Furthermore, lack of evidence for a LTD-associated change in the mRNA level of the IEG zif268 demonstrates that the decrease in arc mRNA during LTD is not a general genetic response. Thus, the regulation of arc expression not only differs between LTP and LTD, but also diverges from that of other IEGs implicated in activity-dependent synaptic plasticity.


Subject(s)
CA1 Region, Hippocampal/metabolism , Cytoskeletal Proteins/genetics , Gene Expression Regulation/physiology , Long-Term Potentiation/genetics , Long-Term Synaptic Depression/genetics , Nerve Tissue Proteins/genetics , RNA, Messenger/biosynthesis , 2-Amino-5-phosphonovalerate/pharmacology , Animals , CA1 Region, Hippocampal/drug effects , Cytoskeletal Proteins/biosynthesis , Early Growth Response Protein 1/biosynthesis , Early Growth Response Protein 1/genetics , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Male , Nerve Tissue Proteins/biosynthesis , Rats , Rats, Sprague-Dawley
17.
Hippocampus ; 21(10): 1093-104, 2011 Oct.
Article in English | MEDLINE | ID: mdl-20824729

ABSTRACT

Evidence shows that the serine/threonine protein phosphatase 1 (PP1) plays a critical role in synaptic plasticity and memory. Little is known about the contribution of the serine/threonine phosphatase 1 (PP2A) to synaptic plasticity. Both protein phosphatases can target the transcription factor cAMP response element binding protein (CREB), whose phosphorylation at Ser133, we previously found, was downregulated during long-term depression (LTD) of glutamatergic transmission in area CA1 of the adult hippocampus in vivo. Other work from our group showed that the activity of PP2A, as well as that of PP1, is increased after LTD induction in area CA1 in vivo. We therefore investigated here whether both protein phosphatases are necessary for LTD in area CA1, and whether they both are involved in the LTD-associated modification of CREB. We found that inhibition of either PP1 or PP2A interferes with the establishment of LTD. Furthermore, inhibition of either enzyme alone abrogated the LTD-associated dephosphorylation of CREB. Interestingly, inhibition of PP1 disrupted CREB dephosphosphorylation rapidly after LTD-inducing stimulation, whereas inhibition of PP2A did not blunt the CREB modification until a later time point. Thus, both PP1 and PP2A regulate CREB during LTD in area CA1, although possibly through different signaling pathways. Our results demonstrate that PP2A, similar to PP1, plays an essential role in the molecular events that underlie LTD at glutamatergic synapses in hippocampal area CA1 in vivo. We propose that one of the mechanisms through which these protein phosphatases may contribute to the prolonged maintenance of LTD is through the regulation of CREB.


Subject(s)
CA1 Region, Hippocampal/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Long-Term Synaptic Depression/physiology , Protein Phosphatase 1/metabolism , Protein Phosphatase 2/metabolism , Animals , CA1 Region, Hippocampal/enzymology , Electric Stimulation , Intracellular Signaling Peptides and Proteins/administration & dosage , Male , Memory , Neuronal Plasticity , Neurons/metabolism , Phosphorylation/drug effects , Polyenes/administration & dosage , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 2/antagonists & inhibitors , Pyrones/administration & dosage , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Synapses/metabolism , Time Factors , Transcription Factors/metabolism
18.
Neurosci Res ; 45(3): 335-44, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12631469

ABSTRACT

Brain ischemic insult causes glutamate release and resultant neuronal cell death. We here show that L-3,4-dihydroxyphenylalanine (DOPA) is a positive regulatory factor for glutamate release elicited by a mild brain insult using in vitro superfused rat striatal slices as a model system. Glucose deprivation for 18 min elicited release of glutamate, DOPA and dopamine (DA). Either tetrodotoxin (TTX) (1 microM) or alpha-methyl-p-tyrosine (alpha-MPT) (1 mM), a tyrosine hydroxylase inhibitor reduced markedly each of these releases. NSD-1015 (20 microM), an aromatic L-amino acid decarboxylase inhibitor restored the inhibition by alpha-MPT of glutamate and DOPA but not DA release. DOPA cyclohexyl ester (DOPA CHE) (0.3-1 microM), a competitive DOPA antagonist, concentration-dependently suppressed aglycemia-induced glutamate release, the effect which was mimicked neither by S-sulpiride nor SCH23390, a DA D(1) or D(2) receptor antagonist, respectively. Zonisamide (1-1000 microM), an anticonvulsant or YM872 (1 microM), an alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) a receptor antagonist produced no effect on aglycemia-induced glutamate release. DOPA CHE thus showed a relatively potent inhibitory action on aglycemia-induced glutamate release among several neuroprotective agents tested.


Subject(s)
Corpus Striatum/drug effects , Glucose/deficiency , Glutamic Acid/metabolism , Levodopa/analogs & derivatives , Levodopa/pharmacology , Animals , Corpus Striatum/metabolism , Dihydroxyphenylalanine/metabolism , In Vitro Techniques , Male , Neuroprotective Agents/pharmacology , Rats , Rats, Wistar
19.
Neurotoxicol Teratol ; 24(5): 629-38, 2002.
Article in English | MEDLINE | ID: mdl-12200194

ABSTRACT

DOPA seems to be a neuromodulator in striata and hippocampal CA1 and a neurotransmitter of the primary baroreceptor afferents terminating in the nucleus tractus solitarii (NTS) and baroreflex pathways in the caudal ventrolateral medulla and rostral ventrolateral medulla in the brainstem of rats. DOPA recognition sites differ from dopamine (DA) D(1) and D(2) and ionotropic glutamate receptors. Via DOPA sites, DOPA stereoselectively releases by itself neuronal glutamate from in vitro and in vivo striata. In the cultured neurons, DOPA and DA cause neuron death via autoxidation. In addition, DOPA causes autoxidation-irrelevant neuron death via glutamate release. Furthermore, DOPA released by four-vessel occlusion seems to be an upstream causal factor for glutamate release and resultant delayed neuron death by brain ischemia in striata and hippocampal CA1. Glutamate has been regarded as a neurotransmitter of baroreflex pathways. Herein, we propose a new pathway that DOPA is a neurotransmitter of the primary aortic depressor nerve and glutamate is that of secondary neurons in neuronal microcircuits of depressor sites in the NTS. DOPA seems to release unmeasurable, but functioning, endogenous glutamate from the secondary neurons via DOPA sites. A common following pathway may be ionotropic glutamate receptors-nNOS activation-NO production-baroreflex neurotransmission and delayed neuron death. However, we are concerned that DOPA therapy may accelerate neuronal degeneration process especially at progressive stages of Parkinson's disease.


Subject(s)
Brain Ischemia/metabolism , Dihydroxyphenylalanine/metabolism , Glutamic Acid/metabolism , Nerve Degeneration/metabolism , Parkinsonian Disorders/metabolism , Animals , Brain Ischemia/physiopathology , Dihydroxyphenylalanine/toxicity , Nerve Degeneration/physiopathology , Nitric Oxide/metabolism , Parkinsonian Disorders/physiopathology , Pressoreceptors/cytology , Pressoreceptors/metabolism , Rats , Solitary Nucleus/cytology , Solitary Nucleus/metabolism , Visceral Afferents/cytology , Visceral Afferents/metabolism
20.
Neurosci Res ; 43(3): 231-8, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12103441

ABSTRACT

We have proposed the hypothesis that L-3,4-dihydroxyphenylalanine (DOPA) plays a role of neurotransmitter of the primary baroreceptor afferents terminating in the nucleus tractus solitarii (NTS). In the present study, we tried to clarify whether glutamate receptors and/or nitric oxide (NO), important modulators for central cardiovascular regulation, are involved in the DOPA-induced cardiovascular responses in the nucleus. Male Wistar rats were anesthetized with urethane and artificially ventilated. Compounds or antisense oligos (17-mer) for neuronal NO synthase were microinjected into depressor sites of the unilateral nucleus. DOPA 30-300 pmol microinjected into the nucleus dose-dependently induced depressor and bradycardic responses. Prior injection of kynurenic acid (600 pmol) suppressed DOPA (300 pmol)-induced responses by approximately 80%. Prior injection of N(G)-monomethyl-L-arginine 100 nmol, a potent NO synthase inhibitor, reversibly attenuated by approximately 90% DOPA-induced responses, while the D-isomer 100 nmol produced no effect. Furthermore, prior injection of neuronal NO synthase antisense oligos (20 pmol) reversibly reduced by approximately 70% responses to DOPA. Sense or scrambled oligos produced no effect. A NO precursor L-arginine (30 nmol) induced depressor and bradycardic responses, but these responses were not affected by kynurenic acid. These results suggest important roles for glutamate receptors and NO in DOPA induced-depressor and bradycardic responses in the NTS.


Subject(s)
Bradycardia/metabolism , Dihydroxyphenylalanine/pharmacology , Kynurenic Acid/pharmacology , Nitric Oxide/biosynthesis , Receptors, Glutamate/metabolism , Solitary Nucleus/drug effects , Anesthetics, Intravenous/pharmacology , Animals , Blood Pressure/drug effects , Male , Rats , Rats, Wistar , Solitary Nucleus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...