Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Br J Pharmacol ; 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39304979

ABSTRACT

BACKGROUND AND PURPOSE: Tetrabenazine (TBZ), used for treating hyperkinetic disorders, inhibits vesicular monoamine transporter-2 (VMAT-2), which sequesters monoamines into vesicles for exocytosis. However, our knowledge of the effect of TBZ on monoaminergic transmission is limited. Herein, we provide neurochemical evidence regarding the effect of VMAT-2 inhibition on vesicular neurotransmitter release from the prefrontal cortex (PFC) and striatum (STR) (brain regions involved in characteristic TBZ treatment side effects). The interaction between TBZ and MDMA was also assessed regarding motor behaviour in mice. EXPERIMENTAL APPROACH: Vesicular storage capacity and release of [3H]-noradrenaline ([3H]-NA), [3H]-dopamine ([3H]-DA), [3H]-serotonin ([3H]-5-HT), and [3H]-acetylcholine ([3H]-ACh) was studied in mouse PFC and STR ex vivo slice preparations using electrical field stimulation. Additionally, locomotor activity was assessed in vehicle-treated mice and compared with that of MDMA, TBZ, and co-administered animals (n = 6) using the LABORAS system. KEY RESULTS: TBZ lowered the storage capacity and inhibited the vesicular release of [3H]-NA and [3H]-DA from the PFC, and [3H]-DA and [3H]-5-HT from the STR in a concentration-dependent manner. Unlike vesamicol (vesicular ACh uptake inhibitor), TBZ failed to inhibit the vesicular release of [3H]-ACh from the PFC. When the vesicular storage of the investigated monoamines was inhibited by TBZ in the PFC and STR, MDMA induced the release of transmitters through transporter reversal; MDMA dose dependently increased locomotor activity in vivo. CONCLUSION AND IMPLICATIONS: Our observations provide neurochemical evidence explaining the mechanism of VMAT-2 inhibitors in the brain and support the involvement of dopaminergic and noradrenergic transmission in hyperkinetic movement disorders.

2.
Sci Adv ; 10(22): eado0077, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38809980

ABSTRACT

While our understanding of the nanoscale architecture of anterograde synaptic transmission is rapidly expanding, the qualitative and quantitative molecular principles underlying distinct mechanisms of retrograde synaptic communication remain elusive. We show that a particular form of tonic cannabinoid signaling is essential for setting target cell-dependent synaptic variability. It does not require the activity of the two major endocannabinoid-producing enzymes. Instead, by developing a workflow for physiological, anatomical, and molecular measurements at the same unitary synapse, we demonstrate that the nanoscale stoichiometric ratio of type 1 cannabinoid receptors (CB1Rs) to the release machinery is sufficient to predict synapse-specific release probability. Accordingly, selective decrease of extrasynaptic CB1Rs does not affect synaptic transmission, whereas in vivo exposure to the phytocannabinoid Δ9-tetrahydrocannabinol disrupts the intrasynaptic nanoscale stoichiometry and reduces synaptic variability. These findings imply that synapses leverage the nanoscale stoichiometry of presynaptic receptor coupling to the release machinery to establish synaptic strength in a target cell-dependent manner.


Subject(s)
Receptor, Cannabinoid, CB1 , Signal Transduction , Synapses , Synaptic Transmission , Animals , Synaptic Transmission/drug effects , Receptor, Cannabinoid, CB1/metabolism , Synapses/metabolism , Presynaptic Terminals/metabolism , Mice , Endocannabinoids/metabolism , Endocannabinoids/pharmacology , Dronabinol/pharmacology
3.
iScience ; 26(9): 107560, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37649698

ABSTRACT

ATP-gated P2X7 receptors (P2X7Rs) play a crucial role in brain disorders. However, how they affect normal and pathological synaptic transmission is still largely unclear. Here, by using whole-cell patch-clamp technique to record AMPA- and NMDA receptor-mediated excitatory postsynaptic currents (s/mEPSCs) in dentate gyrus granule cells (DG GCs), we revealed a modulation by P2X7Rs of presynaptic sites, especially originated from entorhinal cortex (EC)-GC path but not the mossy cell (MC)-GC path. The involvement of P2X7Rs was confirmed using a pharmacological approach. Additionally, the acute activation of P2X7Rs directly elevated calcium influx from EC-GC terminals. In postnatal phencyclidine (PCP)-induced mouse model of schizophrenia, we observed that P2X7R deficiency restored the EC-GC synapse alteration and alleviated PCP-induced symptoms. To summarize, P2X7Rs participate in the modulation of GC excitatory neurotransmission in the DG via EC-GC pathway, contributing to pathological alterations of neuronal functions leading to neurodevelopmental disorders.

4.
Neurochem Res ; 47(11): 3272-3284, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35945308

ABSTRACT

Phenylephrine (PE) is a canonical α1-adrenoceptor-selective agonist. However, unexpected effects of PE have been observed in preclinical and clinical studies, that cannot be easily explained by its actions on α1-adrenoceptors. The probability of the involvement of α2- and ß-adrenoceptors in the effect of PE has been raised. In addition, our earlier study observed that PE released noradrenaline (NA) in a [Ca2+]o-independent manner. To elucidate this issue, we have investigated the effects of PE on [3H]NA release and α1-mediated smooth muscle contractions in the mouse vas deferens (MVD) as ex vivo preparation. The release experiments were designed to assess the effects of PE at the presynaptic terminal, whereas smooth muscle isometric contractions in response to electrical field stimulation were used to measure PE effect postsynaptically. Our results show that PE at concentrations between 0.3 and 30 µM significantly enhanced the resting release of [3H]NA in a [Ca2+]o-independent manner. In addition, prazosin did not affect the release of NA evoked by PE. On the contrary, PE-evoked smooth muscle contractions were inhibited by prazosin administration indicating the α1-adrenoceptor-mediated effect. When the function of the NA transporter (NAT) was attenuated with nisoxetine, PE failed to release NA and the contractions were reduced by approximately 88%. The remaining part proved to be prazosin-sensitive. The present work supports the substantial indirect effect of PE which relays on the cytoplasmic release of NA, which might explain the reported side effects for PE.


Subject(s)
Adrenergic alpha-Antagonists , Norepinephrine , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Animals , Cytoplasm , Male , Mice , Norepinephrine/pharmacology , Phenylephrine/pharmacology , Prazosin/pharmacology , Receptors, Adrenergic, alpha-1
5.
Eur J Pharmacol ; 916: 174621, 2022 Feb 05.
Article in English | MEDLINE | ID: mdl-34965389

ABSTRACT

The histamine H3 receptor is a favourable target for the treatment of cognitive deficits. Here we report the in vitro and in vivo profile of RGH-235, a new potent, selective, and orally active H3 receptor antagonist/inverse agonist developed by Gedeon Richter Plc. Radioligand binding and functional assays were used for in vitro profiling. Procognitive efficacy was investigated in rodent cognitive tests, in models of attention deficit hyperactive disorder (ADHD) and in cognitive tests of high translational value (rat touch screen visual discrimination test, primate fixed-foreperiod visual reaction time task). Results were supported by pharmacokinetic studies, neurotransmitter release, sleep EEG and dipsogenia. RGH-235 displayed high affinity to H3 receptors (Ki = 3.0-9.2 nM, depending on species), without affinity to H1, H2 or H4 receptors and >100 other targets. RGH-235 was an inverse agonist ([35S] GTPγS binding) and antagonist (pERK1/2 ELISA), showing favourable kinetics, inhibition of the imetit-induced dipsogenia and moderate effects on sleep-wake EEG. RGH-235 stimulated neurotransmitter release both in vitro and in vivo. RGH-235 was active in spontaneously hypertensive rats (SHR), generally considered as a model of ADHD, and revealed a robust pro-cognitive profile both in rodent and primate tests (in 0.3-1 mg/kg) and in models of high translational value (e.g. in a rodent touch screen test and in non-human primates). The multiple and convergent procognitive effects of RGH-235 support the view that beneficial cognitive effects can be linked to antagonism/inverse agonism of H3 receptors.


Subject(s)
Receptors, Histamine H3 , Animals , Cognition , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Histamine/pharmacology , Histamine Agonists/metabolism , Rats , Receptors, Histamine H3/metabolism
6.
Physiol Rep ; 9(21): e15088, 2021 11.
Article in English | MEDLINE | ID: mdl-34762352

ABSTRACT

Neuronal networks cause changes in behaviorally important information processing through the vesicular release of neurotransmitters governed by the rate and timing of action potentials (APs). Herein, we provide evidence that dopamine (DA), nonquantally released from the cytoplasm, may exert similar effects in vivo. In mouse slice preparations, (+/-)-3,4-methylenedioxy-methamphetamine (MDMA, or ecstasy) and ß-phenylethylamine (ß-PEA)-induced DA release in the striatum and nucleus accumbens (NAc), two regions of the brain involved in reward-driven and social behavior and inhibited the axonal stimulation-induced release of tritiated acetylcholine ([3 H]ACh) in the striatum. The DA transporter (DAT) inhibitor (GBR-12909) prevented MDMA and ß-PEA from causing DA release. GBR-12909 could also restore some of the stimulated acetylcholine release reduced by MDMA or ß-PEA in the striatum confirming the fundamental role of DAT. In addition, hypothermia could prevent the ß-PEA-induced release in the striatum and in the NAc. Sulpiride, a D2 receptor antagonist, also prevented the inhibitory effects of MDMA or ß-PEA on stimulated ACh release, suggesting they act indirectly via binding of DA. Reflecting the neurochemical interactions in brain slices at higher system level, MDMA altered the social behavior of rats by preferentially enhancing passive social behavior. Similar to the in vitro effects, GBR-12909 treatment reversed specific elements of the MDMA-induced changes in behavior, such as passive social behavior, while left others including social play unchanged. The changes in behavior by the high level of extracellular DA-- a significant amount originating from cytoplasmic release--suggest that in addition to digital computation through synapses, the brain also uses analog communication, such as DA signaling, to mediate some elements of complex behaviors, but in a much longer time scale.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine/metabolism , Serotonin/metabolism , Signal Transduction , Social Behavior , Animals , Corpus Striatum/cytology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine D2 Receptor Antagonists/pharmacology , Male , Mice , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Nucleus Accumbens/cytology , Nucleus Accumbens/metabolism , Phenethylamines/pharmacology , Psychotropic Drugs/pharmacology , Rats , Rats, Wistar , Receptors, Dopamine D2/metabolism , Sulpiride/pharmacology
7.
Front Mol Neurosci ; 13: 566251, 2020.
Article in English | MEDLINE | ID: mdl-33262687

ABSTRACT

Background: It has been consistently reported that the deficiency of the adenosine triphosphate (ATP) sensitive purinergic receptor P2X7 (P2X7R) ameliorates symptoms in animal models of brain diseases. Objective: This study aimed to investigate the role of P2X7R in rodent models of acute and subchronic schizophrenia based on phencyclidine (PCP) delivery in animals lacking or overexpressing P2X7R, and to identify the underlying mechanisms involved. Methods: The psychotomimetic effects of acute i.p. PCP administration in C57Bl/6J wild-type, P2X7R knockout (P2rx7-/-) and overexpressing (P2X7-EGFP) young adult mice were quantified. The medial prefrontal cortex (mPFC) of P2rx7-/- and heterozygous P2X7-EGFP acutely treated animals was characterized through immunohistochemical staining. The prefrontal cortices of young adult P2rx7-/- and P2rx7tg/+ mice were examined with tritiated dopamine release experiments and the functional properties of the mPFC pyramidal neurons in layer V from P2rx7-/- mice were assessed by patch-clamp recordings. P2rx7-/- animals were subjected to a 7 days subchronic systemic PCP treatment. The animals working memory performance and PFC cytokine levels were assessed. Results: Our data strengthen the hypothesis that P2X7R modulates schizophrenia-like positive and cognitive symptoms in NMDA receptor antagonist models in a receptor expression level-dependent manner. P2X7R expression leads to higher medial PFC susceptibility to PCP-induced circuit hyperactivity. The mPFC of P2X7R knockout animals displayed distinct alterations in the neuronal activation pattern, microglial organization, specifically around hyperactive neurons, and were associated with lower intrinsic excitability of mPFC neurons. Conclusions: P2X7R expression exacerbated PCP-related effects in C57Bl/6J mice. Our findings suggest a pleiotropic role of P2X7R in the mPFC, consistent with the observed behavioral phenotype, regulating basal dopamine concentration, layer-specific neuronal activation, intrinsic excitability of neurons in the mPFC, and the interaction of microglia with hyperactive neurons. Direct measurements of P2X7R activity concerning microglial ramifications and dynamics could help to further elucidate the molecular mechanisms involved.

8.
Eur J Pharmacol ; 837: 72-80, 2018 Oct 15.
Article in English | MEDLINE | ID: mdl-30172789

ABSTRACT

The addiction-related behavioural effects of drugs of abuse are mediated by the mesocorticolimbic monoamine systems. We investigated the effects of 3,4-methylenedioxymethamphetamine (MDMA), mephedrone, ß-phenylethylamine (ß-PEA) methylphenidate (MPH) on dopamine release from mouse perfused nucleus accumbens and prefrontal cortex slices. The fractional release of [3H]-dopamine was measured at rest and in response to field stimulation. The distributions of [3H]-dopamine and its metabolites were determined using high-pressure liquid chromatography. The effect of drugs on [3H]-dopamine uptake was measured in synaptosomal P2 preparations from the frontal cortex and striatum. Similar to MDMA, mephedrone ß-PEA increased the resting release of [3H]-dopamine from the nucleus accumbens and prefrontal cortex in a [Ca2+]o-independent manner, and the stimulation-evoked release was also augmented. In contrast, MPH failed to affect the resting release but potentiated the release in response to axonal activity. Similar to dopamine transporter antagonist GBR 12909, mephedrone, MDMA and MPH biphasically inhibited the [3H]-dopamine uptake. The administration of GBR 12909 and nisoxetine, or lowering the bath temperature prevented MDMA, mephedrone and ß-PEA from enhancing the resting, cytoplasmic release of [3H]-dopamine, indicating the role of transporters in the release process. We conclude that amphetamine-like drugs of abuse and the trace amine ß-PEA excessively increase the [Ca2+]o-independent, non-vesicular release of dopamine from the cytoplasm into the extrasynaptic space and inhibit the high-affinity transporters, thereby maintaining a high ambient, non-synaptic concentration of dopamine that may tonically control the activity of neurons equipped with dopamine receptors and is likely involved in the reinforcing effects and abusive potential of amphetamines.


Subject(s)
Cytoplasm/metabolism , Dopamine Plasma Membrane Transport Proteins/physiology , Dopamine Uptake Inhibitors/pharmacology , Dopamine/metabolism , Methamphetamine/analogs & derivatives , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Phenethylamines/pharmacology , Animals , Calcium/physiology , Male , Methamphetamine/pharmacology , Mice , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism
9.
Brain Res Bull ; 131: 150-155, 2017 May.
Article in English | MEDLINE | ID: mdl-28434993

ABSTRACT

We measured the ex vivo uptake and release of [3H]noradrenaline ([3H]NA) from perfused rat spinal cord slice preparations at 1, 3 and 14days after unilateral hemisection-induced spinal cord injury (SCI) compared with control slice preparations. After surgical hemisection under anaesthesia, the rats showed characteristic signs of hemiplegia, with no movement of the ipsilateral hindlimb. After 3days, the electron microscopy images showed overall degeneration of neuronal organelles and the myelin sheath, but the synapses seemed to be intact. In ex vivo experiments, the spinal cord injury did not influence uptake but increased [3H]NA release at rest and in response to axonal stimulation. The effect of a selective noradrenaline reuptake inhibitor, nisoxetine, was studied to identify the mechanisms underlying the increase in NA release. Nisoxetine potentiated stimulation-evoked [3H]NA release from the non-injured tissue, but it gradually lost its effectiveness after injury, depending on the time (1 and 3days) elapsed after hemisection, indicating that the noradrenaline transporter binding sites of the terminals become impaired after decentralisation.


Subject(s)
Norepinephrine/metabolism , Spinal Cord Injuries/metabolism , Adrenergic Uptake Inhibitors/metabolism , Animals , Female , Fluoxetine/analogs & derivatives , Fluoxetine/metabolism , Fluoxetine/pharmacology , Neurons/metabolism , Rats , Rats, Wistar , Spinal Cord/metabolism , Spinal Cord Injuries/physiopathology
10.
Brain Res Bull ; 124: 21-6, 2016 06.
Article in English | MEDLINE | ID: mdl-26996722

ABSTRACT

Because local anesthetics are known to inhibit both sodium and potassium channels, and anesthetic properties have been attributed to the former effect, we compared their effects with those of tetrodotoxin (TTX), a selective Na(+) channel inhibitor with anesthetic activity, and 4-aminopyridine (4-AP), a selective potassium channel blocker with convulsive activity, on transmitter release during rest and in response to field (axonal) stimulation using the microvolume perfusion method and isolated prefrontal cortex and spinal cord slice preparations loaded with the radioactive transmitters [(3)H]dopamine ([(3)H]DA) and [(3)H]noradrenaline ([(3)H]NA). It is also known that local anesthetics may exert analgesic effect and, rarely, some adverse effects on the central nervous system (CNS). Neurochemical evidence demonstrated that local anesthetics administered at concentrations ranging from 0.5 to 5mM, which might have been intentionally or accidentally achieved in clinical practice (e.g., during spinal and epidural anesthesia or peripheral nerve block), led to presynaptic failures during neurochemical transmission, including inhibited transmitter release associated with axonal firing and markedly enhanced extraneuronal concentrations of transmitters due to increased resting, [Ca(2+)]o-independent release. Tetrodotoxin, a toxin with selective Na(+) channel-blocking properties, inhibited the stimulation-evoked release but failed to affect the resting release. In contrast, the potassium channel inhibitor 4-AP enhanced both the resting- and action potential-evoked transmitter releases. It is concluded that effects of local anesthetics on resting catecholamine release in the spinal cord may contribute to their action during neuropathic pain relief and spinal analgesia as well as to their side effects in the CNS.


Subject(s)
Analgesics/pharmacology , Anesthetics, Local/pharmacology , Catecholamines/metabolism , Potassium Channel Blockers/pharmacology , Prefrontal Cortex/drug effects , Spinal Cord/drug effects , 4-Aminopyridine/pharmacology , Animals , Calcium Chelating Agents/pharmacology , Drug Interactions , Egtazic Acid/pharmacology , Electric Stimulation , In Vitro Techniques , Male , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Spinal Cord/metabolism , Tritium/metabolism
11.
Cell ; 163(4): 894-906, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26544938

ABSTRACT

A deficiency in pejvakin, a protein of unknown function, causes a strikingly heterogeneous form of human deafness. Pejvakin-deficient (Pjvk(-/-)) mice also exhibit variable auditory phenotypes. Correlation between their hearing thresholds and the number of pups per cage suggest a possible harmful effect of pup vocalizations. Direct sound or electrical stimulation show that the cochlear sensory hair cells and auditory pathway neurons of Pjvk(-/-) mice and patients are exceptionally vulnerable to sound. Subcellular analysis revealed that pejvakin is associated with peroxisomes and required for their oxidative-stress-induced proliferation. Pjvk(-/-) cochleas display features of marked oxidative stress and impaired antioxidant defenses, and peroxisomes in Pjvk(-/-) hair cells show structural abnormalities after the onset of hearing. Noise exposure rapidly upregulates Pjvk cochlear transcription in wild-type mice and triggers peroxisome proliferation in hair cells and primary auditory neurons. Our results reveal that the antioxidant activity of peroxisomes protects the auditory system against noise-induced damage.


Subject(s)
Hearing Loss, Noise-Induced/metabolism , Nerve Tissue Proteins/metabolism , Peroxisomes/metabolism , Proteins/metabolism , Animals , Auditory Pathways , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Hearing Loss, Noise-Induced/pathology , Humans , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Neurons/metabolism , Oxidative Stress , Proteins/genetics
13.
Nat Commun ; 6: 6557, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25891347

ABSTRACT

The release of GABA from cholecystokinin-containing interneurons is modulated by type-1 cannabinoid receptors (CB1). Here we tested the hypothesis that the strength of CB1-mediated modulation of GABA release is related to the CB1 content of axon terminals. Basket cell boutons have on average 78% higher CB1 content than those of dendritic-layer-innervating (DLI) cells, a consequence of larger bouton surface and higher CB1 density. The CB1 antagonist AM251 caused a 54% increase in action potential-evoked [Ca(2+)] in boutons of basket cells, but not in DLI cells. However, the effect of AM251 did not correlate with CB1 immunoreactivity of individual boutons. Moreover, a CB1 agonist decreased [Ca(2+)] in a cell type- and CB1-content-independent manner. Replica immunogold labelling demonstrated the colocalization of CB1 with the Cav2.2 Ca(2+) channel subunit. Our data suggest that only a subpopulation of CB1s, within nanometre distances from their target Cav2.2 channels, are responsible for endocannabinoid-mediated modulation of GABA release.


Subject(s)
Endocannabinoids/metabolism , Presynaptic Terminals/metabolism , Receptor, Cannabinoid, CB1/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Calcium/metabolism , Female , Hippocampus/cytology , Hippocampus/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Neurons/classification , Red Fluorescent Protein
14.
Neuropharmacology ; 81: 42-54, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24486377

ABSTRACT

We investigated the mode of action of PNU-120596, a type II positive allosteric modulator of the rat α7 nicotinic acetylcholine receptor expressed by GH4C1 cells, using patch-clamp and fast solution exchange. We made two important observations: first, while PNU-120596 rapidly associated to desensitized receptors, it had at least hundredfold lower affinity to resting conformation, therefore at 10 µM concentration it dissociated from resting receptors; and second, binding of PNU-120596 slowed down dissociation of choline molecules from the receptor radically. We propose that when agonist concentration is transiently elevated in the continuous presence of the modulator (as upon the neuronal release of acetylcholine in a modulator-treated animal) these two elements together cause occurrence of a cycle of events: Binding of the modulator is limited in the absence of the agonist. When the agonist is released, it binds to the receptor, and induces desensitization, thereby enabling modulator binding. Modulator binding in turn traps the agonist within its binding site for a prolonged period of time. Once the agonist finally dissociated, the modulator can also dissociate without re-binding, and the receptor assumes its original resting conformation. In kinetic simulations this "trapped agonist cycle" mechanism did not require that the orthosteric and allosteric ligands symmetrically modify each other's affinity, only the modulator must decrease agonist accessibility, and the agonist must induce a conformation that is accessible to the modulator. This mechanism effectively prolongs and amplifies the effect of the agonist.


Subject(s)
Isoxazoles/pharmacology , Membrane Potentials/drug effects , Nicotinic Agonists/pharmacology , Phenylurea Compounds/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Acetylcholine/pharmacology , Allosteric Regulation , Animals , Biophysics , Cell Line, Tumor , Choline/pharmacology , Computer Simulation , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation , Membrane Potentials/genetics , Models, Biological , Patch-Clamp Techniques , Pituitary Neoplasms/pathology , Rats , Transfection , alpha7 Nicotinic Acetylcholine Receptor/genetics
15.
Neuropharmacology ; 81: 101-15, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24486379

ABSTRACT

The alpha7 nicotinic acetylcholine receptor (nAChR) has some peculiar kinetic properties. From the literature of α7 nAChR-mediated currents we concluded that experimentally measured kinetic properties reflected properties of the solution exchange system, rather than genuine kinetic properties of the receptors. We also concluded that all experimentally measured EC50 values for agonists must inherently be inaccurate. The aim of this study was to assess the undistorted kinetic properties of α7 nAChRs, and to construct an improved kinetic model, which can also serve as a basis of modeling the effect of the positive allosteric modulator PNU-120596, as it is described in the accompanying paper. Agonist-evoked currents were recorded from GH4C1 cells stably transfected with pCEP4/rat α7 nAChR using patch-clamp and fast solution exchange. We used two approaches to circumvent the problem of insufficient solution exchange rate: extrapolation and kinetic modeling. First, using different solution exchange rates we recorded evoked currents, and extrapolated their amplitude and kinetics to instantaneous solution exchange. Second, we constructed a kinetic model that reproduced concentration-dependence and solution exchange rate-dependence of receptors, and then we simulated receptor behavior at experimentally unattainably fast solution exchange. We also determined open probabilities during choline-evoked unmodulated and modulated currents using nonstationary fluctuation analysis. The peak open probability of 10 mM choline-evoked currents was 0.033 ± 0.006, while in the presence of choline (10 mM) and PNU-120596 (10 µM), it was increased to 0.599 ± 0.058. Our kinetic model could adequately reproduce low open probability, fast kinetics, fast recovery and solution exchange rate-dependent kinetics.


Subject(s)
Biophysical Phenomena/physiology , Membrane Potentials/physiology , alpha7 Nicotinic Acetylcholine Receptor/physiology , Acetylcholine/pharmacology , Animals , Biophysical Phenomena/drug effects , Cell Line, Tumor , Choline/pharmacology , Computer Simulation , Dose-Response Relationship, Drug , Electric Stimulation , Isoxazoles/pharmacology , Membrane Potentials/drug effects , Models, Biological , Nicotinic Agonists/pharmacology , Patch-Clamp Techniques , Phenylurea Compounds/pharmacology , Pituitary Neoplasms/pathology , Rats , Transfection
16.
J Physiol ; 591(22): 5541-53, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23981718

ABSTRACT

Using two-photon laser microscopy, high- and low-affinity dyes and patch clamp electrophysiology, we successfully measured somatic stimulation-evoked Ca(2+) transients simultaneously in the dendrites and axonal boutons of the same non-fast-spiking GABAergic interneurons in acute slice preparations obtained from hippocampal area CA1. The advantage of the acute preparation is that both neuronal connections and anatomy are maintained. Calculated as unperturbed values, the amplitudes of Ca(2+) transients and changes in [Ca(2+)]i in response to somatic single or burst stimulation were much higher in boutons (428 nM/AP) than in dendrites (49 nM/AP), leading to the conclusion that the much greater influx of Ca(2+) observed in terminals might be due to a higher density of N-type voltage-sensitive Ca(2+) channels compared to the L-type channels present in dendrites. Whereas the decay of Ca(2+) transients recorded in dendrites was primarily mono-exponential, the decay in boutons was bi-exponential, as indicated by an initial fast phase, followed by a much slower reduction in fluorescence intensity. The extrusion of Ca(2+) was much faster in boutons than in dendrites. To avoid saturation effects and the flawed conversion of fluorescence measures of [Ca(2+)]i, we assessed the limits of [Ca(2+)] measurements (which ranged between 6 and 82% of the applied dye saturation) when high- and low-affinity dyes were applied at different concentrations. When two APs were delivered at a high frequency (>3 Hz) of stimulation, the low-affinity indicators OGB-6F (KD = 3.0 µM) and OGB-5N (KD = 20 µM) were able to accurately reflect the changes in ΔF/F produced by the consecutive APs. There was no difference in the endogenous buffer capacity (κE), which can shape Ca(2+) signals, calculated in dendrites (κE = 354) or boutons (κE = 458).


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Dendrites/metabolism , GABAergic Neurons/metabolism , Hippocampus/metabolism , Interneurons/metabolism , Action Potentials/physiology , Animals , Axons/metabolism , Calcium Channels, N-Type/metabolism , Coloring Agents/metabolism , Dendrites/physiology , GABAergic Neurons/physiology , Hippocampus/physiology , Microscopy, Confocal/methods , Rats , Rats, Wistar
17.
PLoS One ; 8(6): e66547, 2013.
Article in English | MEDLINE | ID: mdl-23805233

ABSTRACT

Recent investigations have revealed that the genetic deletion of P2X7 receptors (P2rx7) results in an antidepressant phenotype in mice. However, the link between the deficiency of P2rx7 and changes in behavior has not yet been explored. In the present study, we studied the effect of genetic deletion of P2rx7 on neurochemical changes in the hippocampus that might underlie the antidepressant phenotype. P2X7 receptor deficient mice (P2rx7-/-) displayed decreased immobility in the tail suspension test (TST) and an attenuated anhedonia response in the sucrose preference test (SPT) following bacterial endotoxin (LPS) challenge. The attenuated anhedonia was reproduced through systemic treatments with P2rx7 antagonists. The activation of P2rx7 resulted in the concentration-dependent release of [(3)H]glutamate in P2rx7+/+ but not P2rx7-/- mice, and the NR2B subunit mRNA and protein was upregulated in the hippocampus of P2rx7-/- mice. The brain-derived neurotrophic factor (BDNF) expression was higher in saline but not LPS-treated P2rx7-/- mice; the P2rx7 antagonist Brilliant blue G elevated and the P2rx7 agonist benzoylbenzoyl ATP (BzATP) reduced BDNF level. This effect was dependent on the activation of NMDA and non-NMDA receptors but not on Group I metabotropic glutamate receptors (mGluR1,5). An increased 5-bromo-2-deoxyuridine (BrdU) incorporation was also observed in the dentate gyrus derived from P2rx7-/- mice. Basal level of 5-HT was increased, whereas the 5HIAA/5-HT ratio was lower in the hippocampus of P2rx7-/- mice, which accompanied the increased uptake of [(3)H]5-HT and an elevated number of [(3)H]citalopram binding sites. The LPS-induced elevation of 5-HT level was absent in P2rx7-/- mice. In conclusion there are several potential mechanisms for the antidepressant phenotype of P2rx7-/- mice, such as the absence of P2rx7-mediated glutamate release, elevated basal BDNF production, enhanced neurogenesis and increased 5-HT bioavailability in the hippocampus.


Subject(s)
Anhedonia , Dentate Gyrus/metabolism , Gene Deletion , Glutamic Acid/metabolism , Neurogenesis , Receptors, Purinergic P2X7/deficiency , Serotonin/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Dentate Gyrus/pathology , Dentate Gyrus/physiopathology , Lipopolysaccharides/toxicity , Mice , Mice, Knockout
18.
Trends Mol Med ; 19(6): 355-67, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23601906

ABSTRACT

The enzymatic activities of CD39 and CD73 play strategic roles in calibrating the duration, magnitude, and chemical nature of purinergic signals delivered to immune cells through the conversion of ADP/ATP to AMP and AMP to adenosine, respectively. This drives a shift from an ATP-driven proinflammatory environment to an anti-inflammatory milieu induced by adenosine. The CD39/CD73 pathway changes dynamically with the pathophysiological context in which it is embedded. It is becoming increasingly appreciated that altering this catabolic machinery can change the course or dictate the outcome of several pathophysiological events, such as AIDS, autoimmune diseases, infections, atherosclerosis, ischemia-reperfusion injury, and cancer, suggesting these ectoenzymes are novel therapeutic targets for managing a variety of disorders.


Subject(s)
5'-Nucleotidase/immunology , Antigens, CD/immunology , Apyrase/immunology , Immunity , Inflammation/enzymology , Animals , Humans , Inflammation/immunology
20.
Nat Methods ; 9(2): 201-8, 2012 Jan 08.
Article in English | MEDLINE | ID: mdl-22231641

ABSTRACT

The understanding of brain computations requires methods that read out neural activity on different spatial and temporal scales. Following signal propagation and integration across a neuron and recording the concerted activity of hundreds of neurons pose distinct challenges, and the design of imaging systems has been mostly focused on tackling one of the two operations. We developed a high-resolution, acousto-optic two-photon microscope with continuous three-dimensional (3D) trajectory and random-access scanning modes that reaches near-cubic-millimeter scan range and can be adapted to imaging different spatial scales. We performed 3D calcium imaging of action potential backpropagation and dendritic spike forward propagation at sub-millisecond temporal resolution in mouse brain slices. We also performed volumetric random-access scanning calcium imaging of spontaneous and visual stimulation-evoked activity in hundreds of neurons of the mouse visual cortex in vivo. These experiments demonstrate the subcellular and network-scale imaging capabilities of our system.


Subject(s)
Brain/physiology , Photons , Action Potentials , Animals , Mice , Neurons/physiology , Visual Cortex/cytology , Visual Cortex/physiology
SELECTION OF CITATIONS
SEARCH DETAIL