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










Publication year range
1.
J Neurosci ; 43(10): 1668-1681, 2023 03 08.
Article in English | MEDLINE | ID: mdl-36781220

ABSTRACT

While the physical signs of opioid withdrawal are most readily observable, withdrawal insidiously drives relapse and contributes to compulsive drug use, by disrupting emotional learning circuits. How these circuits become disrupted during withdrawal is poorly understood. Because amygdala neurons mediate relapse, and are highly opioid sensitive, we hypothesized that opioid withdrawal would induce adaptations in these neurons, opening a window of disrupted emotional learning circuit function. Under normal physiological conditions, synaptic transmission between the basolateral amygdala (BLA) and the neighboring main island (Im) of GABAergic intercalated cells (ITCs) is strongly inhibited by endogenous opioids. Using patch-clamp electrophysiology in brain slices prepared from male rats, we reveal that opioid withdrawal abruptly reduces the ability of these peptides to inhibit neurotransmission, a direct consequence of a protein kinase A (PKA)-driven increase in the synaptic activity of peptidases. Reduced peptide control of neurotransmission in the amygdala shifts the excitatory/inhibitory balance of inputs onto accumbens-projecting amygdala cells involved in relapse. These findings provide novel insights into how peptidases control synaptic activity within the amygdala and presents restoration of endogenous peptide activity during withdrawal as a viable option to mitigate withdrawal-induced disruptions in emotional learning circuits and rescue the relapse behaviors exhibited during opioid withdrawal and beyond into abstinence.SIGNIFICANCE STATEMENT We find that opioid withdrawal dials down inhibitory neuropeptide activity in the amygdala. This disrupts both GABAergic and glutamatergic transmission through amygdala circuits, including reward-related outputs to the nucleus accumbens. This likely disrupts peptide-dependent emotional learning processes in the amygdala during withdrawal and may direct behavior toward compulsive drug use.


Subject(s)
Analgesics, Opioid , Substance Withdrawal Syndrome , Rats , Male , Animals , Analgesics, Opioid/pharmacology , Amygdala/physiology , Synaptic Transmission/physiology , Peptides/pharmacology , Substance Withdrawal Syndrome/metabolism , Peptide Hydrolases/metabolism
2.
J Neurosci ; 42(41): 7744-7756, 2022 10 12.
Article in English | MEDLINE | ID: mdl-36414010

ABSTRACT

The midbrain periaqueductal gray (PAG) plays a central role in pain modulation via descending pathways. Opioids and cannabinoids are thought to activate these descending pathways by relieving intrinsic GABAergic inhibition of PAG neurons which project to the rostroventromedial medulla (RVM), a process known as disinhibition. However, the PAG also receives descending extrinsic GABAergic inputs from the central nucleus of the amygdala (CeA) which are thought to inhibit PAG GABAergic interneurons. It remains unclear how opioids and cannabinoids act at these different synapses to control descending analgesic pathways. We used optogenetics, tract tracing and electrophysiology to identify the circuitry underlying opioid and cannabinoid actions within the PAG of male and female rats. It was observed that both RVM-projection and nonprojection PAG neurons received intrinsic-PAG and extrinsic-CeA synaptic inputs, which were predominantly GABAergic. Opioids acted via presynaptic µ-receptors to suppress both intrinsic and extrinsic GABAergic inputs onto all PAG neurons, although this inhibition was greater in RVM-projection neurons. By contrast, cannabinoids acted via presynaptic CB1 receptors to exclusively suppress the direct descending GABAergic input from the CeA onto RVM-projection PAG neurons. These findings indicate the CeA controls PAG output neurons which project to the RVM via parallel direct and indirect GABAergic pathways. While µ-opioids indiscriminately inhibit GABAergic inputs onto all PAG neurons, cannabinoids selectively inhibit a direct extrinsic GABAergic input from the amygdala onto PAG projection neurons. These differential actions of opioids and cannabinoids provide a flexible system to gate the descending control of analgesia from the PAG.SIGNIFICANCE STATEMENT The disinhibition hypothesis of analgesia states that opioids activate the midbrain periaqueductal gray (PAG) descending pathway by relieving the tonic inhibition of projection neurons from GABAergic interneurons. However, the PAG also receives extrinsic GABAergic inputs and is the locus of action of cannabinoid analgesics. Here, we show the relative sensitivity of GABAergic synapses to opioids and cannabinoids within the PAG depends on both the origin of presynaptic inputs and their postsynaptic targets. While opioids indiscriminately inhibit all GABAergic inputs onto all PAG neurons, cannabinoids selectively inhibit a direct extrinsic GABAergic input from the amygdala onto PAG descending projection neurons. These differential actions of opioids and cannabinoids provide a flexible system to gate PAG descending outputs.


Subject(s)
Cannabinoids , Periaqueductal Gray , Male , Female , Rats , Animals , Periaqueductal Gray/metabolism , Analgesics, Opioid/pharmacology , Analgesics, Opioid/metabolism , Cannabinoids/pharmacology , Cannabinoids/metabolism , Pain/metabolism , Medulla Oblongata/metabolism , Analgesics
3.
Int J Mol Sci ; 23(15)2022 Aug 03.
Article in English | MEDLINE | ID: mdl-35955774

ABSTRACT

(1) Background: The psychoactive and non-psychoactive constituents of cannabis, Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD), synergistically reduce allodynia in various animal models of neuropathic pain. Unfortunately, THC-containing drugs also produce substantial side-effects when administered systemically. We examined the effectiveness of targeted spinal delivery of these cannabis constituents, alone and in combination. (2) Methods: The effect of acute intrathecal drug delivery on allodynia and common cannabinoid-like side-effects was examined in a mouse chronic constriction injury (CCI) model of neuropathic pain. (3) Results: intrathecal THC and CBD produced dose-dependent reductions in mechanical and cold allodynia. In a 1:1 combination, they synergistically reduced mechanical and cold allodynia, with a two-fold increase in potency compared to their predicted additive effect. Neither THC, CBD nor combination THC:CBD produced any cannabis-like side-effects at equivalent doses. The anti-allodynic effects of THC were abolished and partly reduced by cannabinoid CB1 and CB2 receptor antagonists AM281 and AM630, respectively. The anti-allodynic effects of CBD were partly reduced by AM630. (4) Conclusions: these findings indicate that intrathecal THC and CBD, individually and in combination, could provide a safe and effective treatment for nerve injury induced neuropathic pain.


Subject(s)
Cannabidiol , Cannabinoids , Cannabis , Hallucinogens , Neuralgia , Analgesics/adverse effects , Animals , Cannabidiol/adverse effects , Cannabinoid Receptor Agonists/pharmacology , Cannabinoids/adverse effects , Disease Models, Animal , Dronabinol/adverse effects , Hallucinogens/adverse effects , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Mice , Neuralgia/drug therapy
4.
Neuropsychopharmacology ; 47(7): 1315-1330, 2022 06.
Article in English | MEDLINE | ID: mdl-35459926

ABSTRACT

Cannabinoid co-administration may enable reduced opioid doses for analgesia. This updated systematic review on the opioid-sparing effects of cannabinoids considered preclinical and clinical studies where the outcome was analgesia or opioid dose requirements. We searched Scopus, Cochrane Central Registry of Controlled Trials, Medline, and Embase (2016 onwards). Ninety-two studies met the search criteria including 15 ongoing trials. Meta-analysis of seven preclinical studies found the median effective dose (ED50) of morphine administered with delta-9-tetrahydrocannabinol was 3.5 times lower (95% CI 2.04, 6.03) than the ED50 of morphine alone. Six preclinical studies found no evidence of increased opioid abuse liability with cannabinoid administration. Of five healthy-volunteer experimental pain studies, two found increased pain, two found decreased pain and one found reduced pain bothersomeness with cannabinoid administration; three demonstrated that cannabinoid co-administration may increase opioid abuse liability. Three randomized controlled trials (RCTs) found no evidence of opioid-sparing effects of cannabinoids in acute pain. Meta-analysis of four RCTs in patients with cancer pain found no effect of cannabinoid administration on opioid dose (mean difference -3.8 mg, 95% CI -10.97, 3.37) or percentage change in pain scores (mean difference 1.84, 95% CI -2.05, 5.72); five studies found more adverse events with cannabinoids compared with placebo (risk ratio 1.13, 95% CI 1.03, 1.24). Of five controlled chronic non-cancer pain trials; one low-quality study with no control arm, and one single-dose study reported reduced pain scores with cannabinoids. Three RCTs found no treatment effect of dronabinol. Meta-analyses of observational studies found 39% reported opioid cessation (95% CI 0.15, 0.64, I2 95.5%, eight studies), and 85% reported reduction (95% CI 0.64, 0.99, I2 92.8%, seven studies). In summary, preclinical and observational studies demonstrate the potential opioid-sparing effects of cannabinoids in the context of analgesia, in contrast to higher-quality RCTs that did not provide evidence of opioid-sparing effects.


Subject(s)
Analgesia , Cannabinoids , Chronic Pain , Opioid-Related Disorders , Analgesics, Opioid , Cannabinoids/pharmacology , Cannabinoids/therapeutic use , Chronic Pain/drug therapy , Humans , Morphine/therapeutic use , Opioid-Related Disorders/drug therapy
5.
Neuropharmacology ; 197: 108736, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34343612

ABSTRACT

The endogenous cannabinoid transmitter system regulates synaptic transmission throughout the nervous system. Unlike conventional transmitters, specific stimuli induce synthesis of endocannabinoids (eCBs) in the postsynaptic neuron, and these travel backwards to modulate presynaptic inputs. In doing so, eCBs can induce short-term changes in synaptic strength and longer-term plasticity. While this eCB regulation is near ubiquitous, it displays major regional and synapse specific variations with different synapse specific forms of short-versus long-term plasticity throughout the brain. These differences are due to the plethora of pre- and postsynaptic mechanisms which have been implicated in eCB signalling, the intricacies of which are only just being realised. In this review, we shall describe the current understanding and highlight new advances in this area, with a focus on the retrograde action of eCBs at CB1 receptors (CB1Rs). This article is part of the special Issue on 'Cannabinoids'.


Subject(s)
Endocannabinoids/physiology , Neuronal Plasticity/physiology , Synapses/physiology , Animals , Cannabinoid Receptor Modulators , Endocannabinoids/pharmacology , Humans , Neuronal Plasticity/drug effects , Neurotransmitter Agents , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/physiology , Synapses/drug effects
6.
Neuropharmacology ; 189: 108529, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33741405

ABSTRACT

The psychoactive and non-psychoactive constituents of cannabis, Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD), have synergistic analgesic efficacy in animal models of neuropathic pain when injected systemically. However, the relevance of this preclinical synergy to clinical neuropathic pain studies is unclear because many of the latter use oral administration. We therefore examined the oral effectiveness of these phytocannabinoids and their interactions in a mouse chronic constriction injury (CCI) model of neuropathic pain. THC produced a dose-dependent reduction in mechanical and cold allodynia, but also induced side-effects with similar potency. CBD also reduced allodynia, albeit with lower potency than THC, but did not produce cannabinoid-like side-effects at any dose tested. Combination THC:CBD produced a dose-dependent reduction in allodynia, however, it displayed little to no synergy. Combination THC:CBD produced substantial, synergistic side-effects which increased with the proportion of CBD. These findings demonstrate that oral THC and CBD, alone and in combination, have analgesic efficacy in an animal neuropathic pain model. Unlike prior systemic injection studies, combination THC:CBD lacks analgesic synergy when delivered orally. Furthermore, both THC and combination THC:CBD display a relatively poor therapeutic window when delivered orally. This suggests that CBD provides a safer, albeit lower efficacy, oral treatment for nerve injury induced neuropathic pain than THC-containing preparations. This article is part of the special issue on 'Cannabinoids'.


Subject(s)
Analgesics, Non-Narcotic/administration & dosage , Cannabidiol/administration & dosage , Dronabinol/administration & dosage , Neuralgia/drug therapy , Neuralgia/physiopathology , Psychotropic Drugs/administration & dosage , Administration, Oral , Animals , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Neuralgia/psychology , Sciatic Neuropathy/drug therapy , Sciatic Neuropathy/physiopathology , Sciatic Neuropathy/psychology , Treatment Outcome
7.
Neuropharmacology ; 182: 108371, 2021 01.
Article in English | MEDLINE | ID: mdl-33122032

ABSTRACT

GABAA and glycine receptors mediate fast synaptic inhibitory neurotransmission. Despite studies showing that activation of cerebral glycine receptors could be a potential strategy in the treatment of epilepsy, few studies have assessed the effects of existing anticonvulsant therapies on recombinant or native glycine receptors. We, therefore, evaluated the actions of a series of anticonvulsants at recombinant human homo-oligomeric glycine receptor α1, α2 and α3 subtypes expressed in Xenopus oocytes using two-electrode voltage-clamp methods, and then assessed the most effective drug at native glycine receptors from entorhinal cortex neurons using whole-cell voltage-clamp recordings. Ganaxolone, tiagabine and zonisamide positively modulated glycine induced currents at recombinant homomeric glycine receptors. Of these, zonisamide was the most efficacious and exhibited an EC50 value ranging between 450 and 560 µM at α1, α2 and α3 subtypes. These values were not significantly different indicating a non-selective modulation of glycine receptors. Using a therapeutic concentration of zonisamide (100 µM), the potency of glycine was significantly shifted from 106 to 56 µM at α1, 185 to 112 µM at α2, and 245 to 91 µM at α3 receptors. Furthermore, zonisamide (100 µM) potentiated exogenous homomeric and heteromeric glycine mediated currents from layer II pyramidal cells of the lateral or medial entorhinal cortex. As therapeutic concentrations of zonisamide positively modulate recombinant and native glycine receptors, we propose that the anticonvulsant effects of zonisamide may, at least in part, be mediated via this action.


Subject(s)
Anticonvulsants/pharmacology , Receptors, Glycine/agonists , Receptors, Glycine/physiology , Zonisamide/pharmacology , Animals , Dose-Response Relationship, Drug , Entorhinal Cortex/drug effects , Entorhinal Cortex/physiology , Female , Glycine/pharmacology , Humans , Male , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Recombinant Proteins , Xenopus laevis
8.
Br J Pharmacol ; 177(16): 3730-3743, 2020 08.
Article in English | MEDLINE | ID: mdl-32352556

ABSTRACT

BACKGROUND AND PURPOSE: While triptans are used to treat migraine, there is evidence that they also reduce inflammation-induced pain at the spinal level. The cellular mechanisms underlying this spinal enhancement are unknown. We examined whether inflammation alters sumatriptan modulation of synaptic transmission in the rat spinal dorsal horn. EXPERIMENTAL APPROACH: Three to four days following intraplantar injection of complete Freund's adjuvant (CFA) or saline, whole cell recordings of evoked glutamatergic EPSCs were made from lumbar lamina I-II dorsal horn neurons in rat spinal slices KEY RESULTS: In 2- to 3-week-old animals, sumatriptan reduced the amplitude of evoked EPSCs and this was greater in slices from CFA, compared to saline-injected rats. In CFA-injected animals, sumatriptan increased the paired pulse ratio of evoked EPSCs and reduced the rate of spontaneous miniature EPSCs. The 5-HT1B and 5-HT1D agonists CP9 3129 and PNU109291 both inhibited evoked EPSCs in CFA but not saline-injected rats. By contrast, the 5-HT1A agonist R(+)-8-OH-DPAT inhibited evoked EPSCs in saline but not CFA-injected rats. In CFA-injected rats, the sumatriptan-induced inhibition of evoked EPSCs was reduced by the 5-HT1B and 5-HT1D antagonists NAS181 and BRL-15572. Intriguingly, the difference in sumatriptan inhibition between CFA and saline-injected animals was only observed in animals less than 4 weeks old. CONCLUSION AND IMPLICATIONS: These findings indicate that inflammation induces a developmentally regulated 5-HT1B/1D presynaptic inhibition of excitatory transmission into the rat superficial dorsal horn. Thus, triptans could potentially act as spinal analgesic agents for inflammatory pain in the juvenile setting.


Subject(s)
Sumatriptan , Synaptic Transmission , Animals , Inflammation/chemically induced , Inflammation/drug therapy , Posterior Horn Cells , Rats , Rats, Sprague-Dawley , Spinal Cord , Sumatriptan/pharmacology
9.
Br J Pharmacol ; 177(10): 2320-2332, 2020 05.
Article in English | MEDLINE | ID: mdl-31971607

ABSTRACT

BACKGROUND AND PURPOSE: The midbrain periaqueductal grey (PAG) plays a central role in modulating pain through a descending pathway that projects indirectly to the spinal cord via the rostroventral medial medulla (RVM). While opioids are potent analgesics that target the PAG, their cellular actions on descending projection neurons are unclear. EXPERIMENTAL APPROACH: Patch clamp recordings in voltage- and current-clamp mode were made from acutely prepared PAG slices from animals that received retrograde tracer injections into the RVM. KEY RESULTS: The µ-agonist DAMGO reduced GABAergic evoked inhibitory postsynaptic currents (IPSCs) in retro-labelled, RVM-projecting neurons to a greater extent than in unlabelled neurons. The κ-opioid agonist U69593 reduced evoked IPSCs to a similar extent in both neuronal groups, while the δ-opioid agonist deltorphin-II was without effect. DAMGO and U69593 both produced a reduction in the rate, but not amplitude of spontaneous miniature IPSCs and asynchronous evoked IPSCs in retro-labelled neurons. DAMGO and U69593 also suppressed glutamatergic EPSCs in retro-labelled and unlabelled neurons. The DAMGO inhibition of evoked EPSCs, however, was less than that for evoked IPSCs in retro-labelled, but not unlabelled neurons. In current clamp, DAMGO produced a depolarizing increase in evoked postsynaptic potentials in retro-labelled neurons, but directly inhibited unlabelled neurons. CONCLUSION AND IMPLICATIONS: These findings suggest that µ-opioids activate the descending analgesic pathway from the midbrain PAG by a combination of presynaptic disinhibition of RVM-projecting neurons and postsynaptic inhibition of presumptive interneurons.


Subject(s)
Analgesics, Opioid , Periaqueductal Gray , Analgesics/pharmacology , Analgesics, Opioid/pharmacology , Animals , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/metabolism , Synaptic Transmission
10.
J Neurochem ; 152(1): 92-102, 2020 01.
Article in English | MEDLINE | ID: mdl-31571215

ABSTRACT

It is thought that endogenous cannabinoids have a role in the analgesia induced by specific forms of stress. We examined if the role of endogenous cannabinoids is also dependent upon the mode of nociception, and whether this could be altered by drugs which block their enzymatic degradation. In C57BL/6 mice, restraint stress produced analgesia in the hot-plate and plantar tests, two thermal pain assays that engage distinct supraspinal and spinal nociceptive pathways. Stress-induced analgesia in the hot-plate test was abolished by pre-treatment with the opioid receptor antagonist naltrexone but was unaffected by the cannabinoid receptor antagonist 1-(2,4-Dichlorophenyl)-5-(4-iodophenyl)-4-methyl-N-4-morpholinyl-1H-pyrazole-3-carboxamide (AM281). By contrast, stress-induced analgesia in the plantar test was abolished by pre-treatment with naltrexone plus AM281, but not by either antagonist individually. Remarkably, inhibiting the breakdown of endocannabinoids, with the dual fatty acid amide hydrolase and monoacylglycerol lipase inhibitor JZL195, rescued stress-induced analgesia in the hotplate test when endogenous opioid signalling was blocked by naltrexone. Furthermore, JZL195 recruited analgesia induced by sub-threshold restraint stress in both thermal pain assays. These findings indicate the role of endocannabinoids in stress-induced analgesia differs with the type of thermal pain behaviour. However, by inhibiting their breakdown, endocannabinoids can be recruited to substitute for endogenous opioid signalling when their activity is blocked, indicating a degree of redundancy between opioid and cannabinoid systems. Together these data suggest targeting endocannabinoid breakdown could provide an alternative, or adjuvant to mainstream analgesics such as opioids.


Subject(s)
Analgesia , Endocannabinoids/physiology , Hot Temperature , Nociception/physiology , Stress, Psychological/physiopathology , Animals , Endocannabinoids/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Restraint, Physical
11.
Br J Pharmacol ; 177(2): 420-431, 2020 01.
Article in English | MEDLINE | ID: mdl-31596498

ABSTRACT

BACKGROUND AND PURPOSE: Pain is a subjective experience involving sensory discriminative and emotionally aversive components. Consistent with its role in pain processing and emotions, the amygdala modulates the aversive component of pain. The laterocapsular region of the central nucleus of the amygdala (CeLC) receives nociceptive information from the parabrachial nucleus (PB) and polymodal, including nociceptive, inputs from the basolateral nucleus of the amygdala (BLA). Opioids are strong analgesics and reduce both the sensory discriminative and the affective component of pain. However, it is unknown whether opioids regulate activity at the two nociceptive inputs to the amygdala. EXPERIMENTAL APPROACH: Using whole-cell electrophysiology, optogenetics, and immunohistochemistry, we investigated whether opioids inhibit the rat PB-CeLC and BLA-CeLC synapses. KEY RESULTS: Opioids inhibited glutamate release at the PB-CeLC and BLA-CeLC synapses. Opioid inhibition is via the µ-receptor at the PB-CeLC synapse, while at the BLA-CeLC synapse, inhibition is via µ-receptors in all neurons and via δ-receptors and κ-receptors in a subset of neurons. CONCLUSIONS AND IMPLICATIONS: Agonists of µ-receptors inhibited two of the synaptic inputs carrying nociceptive information into the laterocapsular amygdala. Therefore, µ-receptor agonists, such as morphine, will inhibit glutamate release from PB and BLA in the CeLC, and this could serve as a mechanism through which opioids reduce the affective component of pain and pain-induced associative learning. The lower than expected regulation of BLA synaptic outputs by δ-receptors does not support the proposal that opioid receptor subtypes segregate into subnuclei of brain regions.


Subject(s)
Amygdala/drug effects , Analgesics, Opioid/pharmacology , Nociception/drug effects , Nociceptive Pain/prevention & control , Pain Perception/drug effects , Synapses/drug effects , Amygdala/metabolism , Amygdala/physiopathology , Animals , Glutamic Acid/metabolism , Male , Neural Inhibition/drug effects , Nociceptive Pain/metabolism , Nociceptive Pain/physiopathology , Optogenetics , Rats, Sprague-Dawley , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Synapses/metabolism
12.
Handb Exp Pharmacol ; 254: 417, 2019.
Article in English | MEDLINE | ID: mdl-31152275

ABSTRACT

In the last paragraph of Sect. 2.1.2 on line 3 the word 'off-cells' is misspelt. It should be 'on-cells'.

13.
Handb Exp Pharmacol ; 254: 91-130, 2019.
Article in English | MEDLINE | ID: mdl-30838458

ABSTRACT

Whilst the nociceptin/orphanin FQ (N/OFQ) receptor (NOP) has similar intracellular coupling mechanisms to opioid receptors, it has distinct modulatory effects on physiological functions such as pain. These actions range from agonistic to antagonistic interactions with classical opioids within the spinal cord and brain, respectively. Understanding the electrophysiological actions of N/OFQ has been crucial in ascertaining the mechanisms by which these agonistic and antagonistic interactions occur. These similarities and differences between N/OFQ and opioids are due to the relative location of NOP versus opioid receptors on specific neuronal elements within these CNS regions. These mechanisms result in varied cellular actions including postsynaptic modulation of ion channels and presynaptic regulation of neurotransmitter release.


Subject(s)
Analgesics, Opioid/pharmacology , Opioid Peptides , Receptors, Opioid , Analgesics, Opioid/chemistry , Humans , Opioid Peptides/pharmacology , Pain , Receptors, Opioid/chemistry
14.
Br J Pharmacol ; 175(12): 2337-2347, 2018 06.
Article in English | MEDLINE | ID: mdl-29500820

ABSTRACT

BACKGROUND AND PURPOSE: Inhibitory neurotransmission plays an important role in controlling excitability within nociceptive circuits of the spinal cord dorsal horn. Loss of inhibitory signalling is thought to contribute to the development of pathological pain. Preclinical studies suggest that increasing inhibitory glycinergic signalling is a good therapeutic strategy for treating pain. One approach to increase synaptic glycine is to inhibit the activity of the glycine transporter 2 (GlyT2) on inhibitory nerve terminals. These transporters are involved in regulating glycine concentrations and recycling glycine into presynaptic terminals. Inhibiting activity of GlyT2 increases synaptic glycine, which decreases excitability in nociceptive circuits and provides analgesia in neuropathic and inflammatory pain models. EXPERIMENTAL APPROACH: We investigated the effects of reversible and irreversible GlyT2 inhibitors on inhibitory glycinergic and NMDA receptor-mediated excitatory neurotransmission in the rat dorsal horn. The effect of these drugs on synaptic signalling was determined using patch-clamp electrophysiology techniques to measure glycine- and NMDA-mediated postsynaptic currents in spinal cord slices in vitro. KEY RESULTS: We compared activity of four compounds that increase glycinergic tone with a corresponding increase in evoked glycinergic postsynaptic currents. These compounds did not deplete synaptic glycine release over time. Interestingly, none of these compounds increased glycine-mediated excitatory signalling through NMDA receptors. The results suggest that these compounds preferentially inhibit GlyT2 over G1yT1 with no potentiation of the glycine receptor and without inducing spillover from inhibitory to excitatory synapses. CONCLUSIONS AND IMPLICATIONS: GlyT2 inhibitors increase inhibitory neurotransmission in the dorsal horn and have potential as pain therapeutics. LINKED ARTICLES: This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.


Subject(s)
Glycine Agents/pharmacology , Glycine Plasma Membrane Transport Proteins/antagonists & inhibitors , Spinal Cord Dorsal Horn/drug effects , Spinal Cord/drug effects , Synapses/drug effects , Synaptic Transmission/drug effects , Animals , Glycine Agents/chemistry , Glycine Plasma Membrane Transport Proteins/metabolism , Male , Molecular Structure , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism , Spinal Cord Dorsal Horn/metabolism , Synapses/metabolism
15.
Nat Commun ; 8: 14611, 2017 03 22.
Article in English | MEDLINE | ID: mdl-28327612

ABSTRACT

Fear and emotional learning are modulated by endogenous opioids but the cellular basis for this is unknown. The intercalated cells (ITCs) gate amygdala output and thus regulate the fear response. Here we find endogenous opioids are released by synaptic stimulation to act via two distinct mechanisms within the main ITC cluster. Endogenously released opioids inhibit glutamate release through the δ-opioid receptor (DOR), an effect potentiated by a DOR-positive allosteric modulator. Postsynaptically, the opioids activate a potassium conductance through the µ-opioid receptor (MOR), suggesting for the first time that endogenously released opioids directly regulate neuronal excitability. Ultrastructural localization of endogenous ligands support these functional findings. This study demonstrates a new role for endogenously released opioids as neuromodulators engaged by synaptic activity to regulate moment-to-moment neuronal communication and excitability. These distinct actions through MOR and DOR may underlie the opposing effect of these receptor systems on anxiety and fear.


Subject(s)
Amygdala/metabolism , Anxiety/metabolism , Fear/physiology , Interneurons/metabolism , Opioid Peptides/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Glutamic Acid/metabolism , In Vitro Techniques , Male , Patch-Clamp Techniques , Potassium/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid/metabolism , Synapses/metabolism
16.
J Neurosci ; 36(2): 385-95, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-26758831

ABSTRACT

Basolateral amygdala (BLA) is critical for fear learning, and its heightened activation is widely thought to underpin a variety of anxiety disorders. Here we used chemogenetic techniques in rats to study the consequences of heightened BLA activation for fear learning and memory, and to specifically identify a mechanism linking increased activity of BLA glutamatergic neurons to aberrant fear. We expressed the excitatory hM3Dq DREADD in rat BLA glutamatergic neurons and showed that CNO acted selectively to increase their activity, depolarizing these neurons and increasing their firing rates. This chemogenetic excitation of BLA glutamatergic neurons had no effect on the acquisition of simple fear learning, regardless of whether this learning led to a weak or strong fear memory. However, in an associative blocking task, chemogenetic excitation of BLA glutamatergic neurons yielded significant learning to a blocked conditioned stimulus, which otherwise should not have been learned about. Moreover, in an overexpectation task, chemogenetic manipulation of BLA glutamatergic neurons prevented use of negative prediction error to reduce fear learning, leading to significant impairments in fear inhibition. These effects were not attributable to the chemogenetic manipulation enhancing arousal, increasing asymptotic levels of fear learning or fear memory consolidation. Instead, chemogenetic excitation of BLA glutamatergic neurons disrupted use of prediction error to regulate fear learning. SIGNIFICANCE STATEMENT: Several neuropsychiatric disorders are characterized by heightened activation of the amygdala. This heightened activation has been hypothesized to underlie increased emotional reactivity, fear over generalization, and deficits in fear inhibition. Yet the mechanisms linking heightened amygdala activation to heightened emotional learning are elusive. Here we combined chemogenetic excitation of rat basolateral amygdala glutamatergic neurons with a variety of behavioral approaches to show that, although simple fear learning is unaffected, the use of prediction error to regulate this learning is profoundly disrupted, leading to formation of inappropriate fear associations and impaired fear inhibition.


Subject(s)
Amygdala/cytology , Amygdala/physiology , Conditioning, Psychological/physiology , Fear , Neurons/physiology , Action Potentials/drug effects , Action Potentials/physiology , Amygdala/drug effects , Animals , Clozapine/analogs & derivatives , Clozapine/pharmacology , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Conditioning, Psychological/drug effects , Dependovirus/genetics , Electroshock/adverse effects , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Fear/drug effects , Glutamic Acid/metabolism , Humans , Male , Membrane Potentials/drug effects , Neurons/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Muscarinic M3/genetics , Receptors, Drug/genetics , Receptors, Drug/metabolism
17.
Sci Rep ; 5: 10934, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26055072

ABSTRACT

The acute neurotoxicity of oligomeric forms of amyloid-ß 1-42 (Aß) is implicated in the pathogenesis of Alzheimer's disease (AD). However, how these oligomers might first impair neuronal function at the onset of pathology is poorly understood. Here we have examined the underlying toxic effects caused by an increase in levels of intracellular Aß, an event that could be important during the early stages of the disease. We show that oligomerised Aß induces a rapid enhancement of AMPA receptor-mediated synaptic transmission (EPSC(A)) when applied intracellularly. This effect is dependent on postsynaptic Ca(2+) and PKA. Knockdown of GluA1, but not GluA2, prevents the effect, as does expression of a S845-phosphomutant of GluA1. Significantly, an inhibitor of Ca(2+)-permeable AMPARs (CP-AMPARs), IEM 1460, reverses the increase in the amplitude of EPSC(A). These results suggest that a primary neuronal response to intracellular Aß oligomers is the rapid synaptic insertion of CP-AMPARs.


Subject(s)
Amyloid beta-Peptides/metabolism , Hippocampus/metabolism , Receptors, AMPA/metabolism , Alzheimer Disease/metabolism , Animals , Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoplasm/metabolism , Male , Neurons/metabolism , Phosphorylation/physiology , Protein Transport/physiology , Rats , Rats, Wistar , Receptors, Glutamate/metabolism , Synapses/metabolism
18.
Nat Neurosci ; 13(10): 1216-24, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20852624

ABSTRACT

Although muscarinic acetylcholine receptors (mAChRs) and NMDA receptors (NMDARs) are important for synaptic plasticity, learning and memory, the manner in which they interact is poorly understood. We found that stimulation of muscarinic receptors, either by an agonist or by the synaptic release of acetylcholine, led to long-term depression (LTD) of NMDAR-mediated synaptic transmission. This form of LTD involved the release of Ca2+ from IP3-sensitive intracellular stores and was expressed via the internalization of NMDARs. Our results suggest that the molecular mechanism involves a dynamic interaction between the neuronal calcium sensor protein hippocalcin, the clathrin adaptor molecule AP2, the postsynaptic density enriched protein PSD-95 and NMDARs. We propose that hippocalcin binds to the SH3 region of PSD-95 under basal conditions, but it translocates to the plasma membrane on sensing Ca2+; in doing so, it causes PSD-95 to dissociate from NMDARs, permitting AP2 to bind and initiate their dynamin-dependent endocytosis.


Subject(s)
Adaptor Protein Complex 2/metabolism , Excitatory Postsynaptic Potentials/physiology , Hippocalcin/metabolism , Hippocampus/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Long-Term Synaptic Depression/physiology , Membrane Proteins/metabolism , Receptors, Muscarinic/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/physiology , Acetylcholine/metabolism , Adaptor Protein Complex 2/genetics , Animals , Biophysics/methods , Calcium/metabolism , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Disks Large Homolog 4 Protein , Drug Interactions , Electric Stimulation/methods , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/genetics , Hippocalcin/genetics , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Muscarinic Antagonists/pharmacology , Patch-Clamp Techniques , Piperidines/pharmacology , Protein Transport/physiology , Quinolones/pharmacology , RNA Interference/physiology , Rats , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...