Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 24(13)2023 Jul 05.
Article in English | MEDLINE | ID: mdl-37446289

ABSTRACT

Dopamine (DA) inhibits excitatory synaptic transmission in the anterior cingulate cortex (ACC), a brain region involved in the sensory and affective processing of pain. However, the DA modulation of inhibitory synaptic transmission in the ACC and its alteration of the excitatory/inhibitory (E/I) balance remains relatively understudied. Using patch-clamp recordings, we demonstrate that neither DA applied directly to the tissue slice nor complete Freund's adjuvant (CFA) injected into the hind paw significantly impacted excitatory currents (eEPSCs) in the ACC, when recorded without pharmacological isolation. However, individual neurons exhibited varied responses to DA, with some showing inhibition, potentiation, or no response. The degree of eEPSC inhibition by DA was higher in naïve slices compared to that in the CFA condition. The baseline inhibitory currents (eIPSCs) were greater in the CFA-treated slices, and DA specifically inhibited eIPSCs in the CFA-treated, but not naïve group. DA and CFA treatment did not alter the balance between excitatory and inhibitory currents. Spontaneous synaptic activity revealed that DA reduced the frequency of the excitatory currents in CFA-treated mice and decreased the amplitude of the inhibitory currents, specifically in CFA-treated mice. However, the overall synaptic drive remained similar between the naïve and CFA-treated mice. Additionally, GABAergic currents were pharmacologically isolated and found to be robustly inhibited by DA through postsynaptic D2 receptors and G-protein activity. Overall, the study suggests that CFA-induced inflammation and DA do not significantly affect the balance between excitatory and inhibitory currents in ACC neurons, but activity-dependent changes may be observed in the DA modulation of presynaptic glutamate release in the presence of inflammation.


Subject(s)
Dopamine , Gyrus Cinguli , Mice , Animals , Dopamine/pharmacology , Synaptic Transmission/physiology , Pain , Glutamic Acid/adverse effects , Inflammation/chemically induced
2.
Neuroscience ; 498: 249-259, 2022 08 21.
Article in English | MEDLINE | ID: mdl-35863681

ABSTRACT

Pain modulation of dopamine-producing nuclei is known to contribute to the affective component of chronic pain. However, pain modulation of pain-related cortical regions receiving dopaminergic inputs is understudied. The present study demonstrates that mice with chronic inflammatory injury of the hind paws develop persistent mechanical hypersensitivity and transient anxiety. Peripheral inflammation induced by injection of complete Freund's Adjuvant (CFA) induced potentiation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic receptor (AMPAR) currents with a presynaptic component in layer II/III of the ACC. After four days of inflammatory pain, the dopamine-mediated inhibition of AMPAR currents was significantly reduced in the ACC. Furthermore, dopamine enhanced presynaptic modulation of excitatory transmission, but only in mice with inflammatory pain. High-performance liquid chromatography (HPLC) analysis of dopamine tissue concentration revealed that dopamine neurotransmitter concentration in the ACC was reduced three days following CFA. Our results demonstrate that inflammatory pain induces activity-dependent changes in excitatory synaptic transmission and alters dopaminergic homeostasis in the ACC.


Subject(s)
Chronic Pain , Gyrus Cinguli , Animals , Dopamine , Freund's Adjuvant , Inflammation , Mice , Mice, Inbred C57BL , Synapses , Synaptic Transmission
3.
Neuropsychopharmacology ; 46(6): 1183-1193, 2021 05.
Article in English | MEDLINE | ID: mdl-33223518

ABSTRACT

Experiencing pain with a familiar individual can enhance one's own pain sensitivity, a process known as pain contagion. When experiencing pain with an unfamiliar individual, pain contagion is suppressed in males by activating the endocrine stress response. Here, we coupled a histological investigation with pharmacological and behavioral experiments to identify enhanced glucocorticoid receptor activity in the prelimbic subdivision of the medial prefrontal cortex as a candidate mechanism for suppressing pain contagion in stranger mice. Acute inhibition of glucocorticoid receptors in the prelimbic cortex was sufficient to elicit pain contagion in strangers, while their activation prevented pain contagion in cagemate dyads. Slice physiology recordings revealed enhanced excitatory transmission in stranger mice, an effect that was reversed by pre-treating mice with the corticosterone synthesis inhibitor metyrapone. Following removal from dyadic testing, stranger mice displayed enhanced affective-motivational pain behaviors when placed on an inescapable thermal stimulus, which were reversed by metyrapone. Together, our data suggest that the prelimbic cortex may play an integral role in modulating pain behavior within a social context and provide novel evidence towards the neural mechanism underlying the prevention of pain contagion.


Subject(s)
Prefrontal Cortex , Receptors, Glucocorticoid , Animals , Cerebral Cortex , Corticosterone , Male , Mice , Pain/drug therapy
4.
Mol Brain ; 13(1): 121, 2020 09 05.
Article in English | MEDLINE | ID: mdl-32891169

ABSTRACT

The release of dopamine (DA) into target brain areas is considered an essential event for the modulation of many physiological effects. While the anterior cingulate cortex (ACC) has been implicated in pain related behavioral processes, DA modulation of synaptic transmission within the ACC and pain related phenotypes remains unclear. Here we characterized a Crispr/Cas9 mediated somatic knockout of the D1 receptor (D1R) in all neuronal subtypes of the ACC and find reduced mechanical thresholds, without affecting locomotion and anxiety. Further, the D1R high-efficacy agonist SKF 81297 and low efficacy agonist (±)-SKF-38393 inhibit α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic receptor (AMPAR) currents in the ACC. Paradoxically, the D1R antagonists SCH-23390 and SCH 33961 when co-applied with D1R agonists produced a robust short-term synergistic depression of AMPAR currents in the ACC, demonstrating an overall inhibitory role for D1R ligands. Overall, our data indicate that absence of D1Rs in the ACC enhanced peripheral sensitivity to mechanical stimuli and D1R activation decreased glutamatergic synaptic transmission in ACC neurons.


Subject(s)
Glutamic Acid/metabolism , Gyrus Cinguli/metabolism , Receptors, Dopamine D1/metabolism , Sensory Thresholds , Synaptic Transmission , Animals , Behavior, Animal/drug effects , Benzazepines/pharmacology , CRISPR-Cas Systems/genetics , Excitatory Postsynaptic Potentials/drug effects , Gene Knockout Techniques , Gyrus Cinguli/drug effects , Gyrus Cinguli/physiopathology , Ion Channel Gating/drug effects , Male , Mice, Inbred C57BL , Pain/pathology , Pain/physiopathology , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Sensory Thresholds/drug effects , Synaptic Transmission/drug effects
5.
Neurobiol Dis ; 116: 166-178, 2018 08.
Article in English | MEDLINE | ID: mdl-29727711

ABSTRACT

Some evidence suggests that the cerebellum modulates affect via connectivities with mood-regulating corticolimbic structures, such as the prefrontal cortex and monoamine nuclei. In rats exposed to chronic unpredictable stress (CUS), we examined the neuro-behavioural effects of high frequency stimulation and surgical ablation/disconnection of the cerebellar vermis. CUS reduced sucrose preference, increased novelty-induced feeding suppression and passive coping. These depressive-like behaviours were associated with decreased cerebellar zif268 expression, indicating possible cerebellar involvement in stress pathology. These were paralleled by decreased vermal Purkinje simple and complex spiking activity and raphe serotonergic activity. Protracted (24-h) vermal stimulation reversed these behavioural deficits through serotonin-mediated mechanisms since this effect was abrogated by the serotonin-depleting agent pCPA. Vermal stimulation and disconnection lesion also enhanced serotonergic activity, but did not modify prefrontocortical pyramidal firing. This effect was likely mediated by 5-HT1A receptors (5-HT1AR). Indeed, acute vermal stimulation mimicked the effect of the 5-HT1AR agonist 8-OH-DPAT in inhibiting serotonergic activity, which was prevented by pre-treatment with the 5-HT1AR antagonist WAY100,635. These results demonstrate vermal involvement in depressive-type behaviour via its modulatory action on serotonergic neurons. They further suggest that vermal and mPFC stimulation may bestow therapeutic benefits via parallel pathways.


Subject(s)
Cerebellar Vermis/physiology , Deep Brain Stimulation/methods , Dorsal Raphe Nucleus/physiology , Prefrontal Cortex/physiology , Stress, Psychological/physiopathology , Stress, Psychological/psychology , Animals , Chronic Disease , Male , Rats , Rats, Wistar , Stress, Psychological/therapy
6.
Mol Pain ; 122016.
Article in English | MEDLINE | ID: mdl-27317578

ABSTRACT

Dopamine (DA) possesses potent neuromodulatory properties in the central nervous system. In the anterior cingulate cortex, α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPAR) are key ion channels in mediating nerve injury induced long-term potentiation (LTP) and chronic pain phenotype. In the present study, we reported the effects of DA on glutamate mediated excitatory post-synaptic currents (EPSCs) in pyramidal neurons of layer II/III of the ACC in adult mice. Bath application of DA (50 µM) caused a significant, rapid and reversible inhibition of evoked EPSCs (eEPSC). This inhibitory effect is dose-related and was absent in lower concentration of DA (5 µM). Furthermore, selective postsynaptic application of GDP-ß-S (1.6 mM) in the internal solution completely abolished the inhibitory effects of DA (50 µM). We also investigated modulation of spontaneous EPSCs (sEPSCs) and TTX sensitive, miniature EPSCs (mEPSCs) by DA. Our results indicated mixed effects of potentiation and inhibition of frequency and amplitude for sEPSCs and mEPSCs. Furthermore, high doses of SCH23390 (100 µM) and sulpiride (100 µM) revealed that, inhibition of eEPSCs is mediated by postsynaptic D2-receptors (D2R). Our finding posits a pre- and postsynaptic mode of pyramidal neuron EPSC modulation in mice ACC by DA.


Subject(s)
Dopamine/pharmacology , Gyrus Cinguli/physiology , Synaptic Transmission/drug effects , Aging/physiology , Animals , Benzazepines/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Gyrus Cinguli/drug effects , Male , Mice, Inbred C57BL , Models, Biological , Neural Inhibition/drug effects , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Receptors, Dopamine D2/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Kainic Acid/metabolism , Signal Transduction/drug effects , Sulpiride/pharmacology , Tetrodotoxin/toxicity , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
7.
Mol Pain ; 122016.
Article in English | MEDLINE | ID: mdl-27178245

ABSTRACT

The extracellular signal-regulated kinase is an important protein kinase for cortical plasticity. Long-term potentiation in the anterior cingulate cortex is believed to play important roles in chronic pain, fear, and anxiety. Previous studies of extracellular signal-regulated kinase are mainly focused on postsynaptic form of long-term potentiation (post-long-term potentiation). Little is known about the relationship between extracellular signal-regulated kinase and presynaptic long-term potentiation (pre-long-term potentiation) in cortical synapses. In this study, we examined whether pre-long-term potentiation in the anterior cingulate cortex requires the activation of presynaptic extracellular signal-regulated kinase. We found that p42/p44 mitogen-activated protein kinase inhibitors, PD98059 and U0126, suppressed the induction of pre-long-term potentiation. By contrast, these inhibitors did not affect the maintenance of pre-long-term potentiation. Using pharmacological inhibitors, we found that pre-long-term potentiation recorded for 1 h did not require transcriptional or translational processes. Our results strongly indicate that the activation of presynaptic extracellular signal-regulated kinase is required for the induction of pre-long-term potentiation, and this involvement may explain the contribution of extracellular signal-regulated kinase to mood disorders.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Gyrus Cinguli/enzymology , Gyrus Cinguli/physiology , Long-Term Potentiation , Animals , Butadienes/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Flavonoids/pharmacology , Glutamic Acid/metabolism , Gyrus Cinguli/drug effects , Long-Term Potentiation/drug effects , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Nitriles/pharmacology , Protein Biosynthesis/drug effects , Protein Kinase Inhibitors/pharmacology , Synapses/drug effects , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...