Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 156
Filter
1.
Nat Commun ; 10(1): 367, 2019 01 21.
Article in English | MEDLINE | ID: mdl-30664663

ABSTRACT

Opioid analgesics are powerful pain relievers; however, over time, pain control diminishes as analgesic tolerance develops. The molecular mechanisms initiating tolerance have remained unresolved to date. We have previously shown that desensitization of the µ-opioid receptor and interaction with ß-arrestins is controlled by carboxyl-terminal phosphorylation. Here we created knockin mice with a series of serine- and threonine-to-alanine mutations that render the receptor increasingly unable to recruit ß-arrestins. Desensitization is inhibited in locus coeruleus neurons of mutant mice. Opioid-induced analgesia is strongly enhanced and analgesic tolerance is greatly diminished. Surprisingly, respiratory depression, constipation, and opioid withdrawal signs are unchanged or exacerbated, indicating that ß-arrestin recruitment does not contribute to the severity of opioid side effects and, hence, predicting that G-protein-biased µ-agonists are still likely to elicit severe adverse effects. In conclusion, our findings identify carboxyl-terminal multisite phosphorylation as key step that drives acute µ-opioid receptor desensitization and long-term tolerance.


Subject(s)
Analgesics, Opioid/adverse effects , Brain/drug effects , Drug Tolerance , Pain/drug therapy , Receptors, Opioid, mu/genetics , Analgesia/methods , Analgesics, Opioid/administration & dosage , Animals , Brain/metabolism , Brain/physiopathology , Female , Fentanyl/administration & dosage , Fentanyl/adverse effects , Gene Expression , Gene Knock-In Techniques , Infusion Pumps, Implantable , Male , Mice , Mice, Transgenic , Microtomy , Morphine/administration & dosage , Morphine/adverse effects , Naloxone/administration & dosage , Naloxone/adverse effects , Pain/metabolism , Pain/physiopathology , Pain Management/methods , Phosphorylation/drug effects , Protein Binding , Receptors, Opioid, mu/metabolism , Tissue Culture Techniques , beta-Arrestins/genetics , beta-Arrestins/metabolism
3.
Br J Pharmacol ; 172(2): 492-500, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24597632

ABSTRACT

BACKGROUND AND PURPOSE: Tolerance to the behavioural effects of morphine is blunted in ß-arrestin-2 knockout mice, but opioid withdrawal is largely unaffected. The cellular mechanisms of tolerance have been studied in some neurons from ß-arrestin-2 knockouts, but tolerance and withdrawal mechanisms have not been examined at the cellular level in periaqueductal grey (PAG) neurons, which are crucial for central tolerance and withdrawal phenomena. EXPERIMENTAL APPROACH: µ-Opioid receptor (MOPr) inhibition of voltage-gated calcium channel currents (ICa ) was examined by patch-clamp recordings from acutely dissociated PAG neurons from wild-type and ß-arrestin-2 knockout mice treated chronically with morphine (CMT) or vehicle. Opioid withdrawal-induced activation of GABA transporter type 1 (GAT-1) currents was determined using perforated patch recordings from PAG neurons in brain slices. KEY RESULTS: MOPr inhibition of ICa in PAG neurons was unaffected by ß-arrestin-2 deletion. CMT impaired coupling of MOPrs to ICa in PAG neurons from wild-type mice, but this cellular tolerance was not observed in neurons from CMT ß-arrestin-2 knockouts. However, ß-arrestin-2 knockouts displayed similar opioid-withdrawal-induced activation of GAT-1 currents as wild-type PAG neurons. CONCLUSIONS AND IMPLICATIONS: In ß-arrestin-2 knockout mice, the central neurons involved in the anti-nociceptive actions of opioids also fail to develop cellular tolerance to opioids following chronic morphine. The results also provide the first cellular physiological evidence that opioid withdrawal is not disrupted by ß-arrestin-2 deletion. However, the unaffected basal sensitivity to opioids in PAG neurons provides further evidence that changes in basal MOPr sensitivity cannot account for the enhanced acute nociceptive response to morphine reported in ß-arrestin-2 knockouts. LINKED ARTICLES: This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.


Subject(s)
Arrestins/physiology , Drug Tolerance/physiology , Periaqueductal Gray/physiology , Receptors, Opioid, mu/physiology , Substance Withdrawal Syndrome/physiopathology , Analgesics, Opioid/pharmacology , Animals , Arrestins/genetics , GABA Plasma Membrane Transport Proteins/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Morphine/pharmacology , Neurons/physiology , beta-Arrestin 2 , beta-Arrestins
4.
Br J Pharmacol ; 170(2): 245-54, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23713957

ABSTRACT

BACKGROUND AND PURPOSE: Antagonists of the N-type voltage gated calcium channel (VGCC), Cav 2.2, have a potentially important role in the treatment of chronic neuropathic pain. ω-conotoxins, such MVIIA and CVID are effective in neuropathic pain models. CVID is reported to have a greater therapeutic index than MVIIA in neuropathic pain models, and it has been suggested that this is due to faster reversibility of binding, but it is not known whether this can be improved further. EXPERIMENTAL APPROACH: We examined the potency of CVID, MVIIA and two intermediate hybrids ([K10R]CVID and [R10K]MVIIA) to reverse signs of neuropathic pain in a rat nerve ligation model in parallel with production of side effects. We also examined the potency and reversibility to inhibit primary afferent synaptic neurotransmission in rat spinal cord slices. KEY RESULTS: All ω-conotoxins produced dose-dependent reduction in mechanical allodynia. They also produced side effects on the rotarod test and in a visual side-effect score. CVID displayed a marginally better therapeutic index than MVIIA. The hybrids had a lesser effect in the rotarod test than either of their parent peptides. Finally, the conotoxins all presynaptically inhibited excitatory synaptic neurotransmission into the dorsal horn and displayed recovery that was largely dependent upon the magnitude of inhibition and not the conotoxin type. CONCLUSIONS AND IMPLICATIONS: These findings indicate that CVID provides only a marginal improvement over MVIIA in a preclinical model of neuropathic pain, which appears to be unrelated to reversibility from binding. Hybrids of these conotoxins might provide viable alternative treatments.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Neuralgia/drug therapy , omega-Conotoxins/pharmacology , Analgesics, Non-Narcotic/administration & dosage , Analgesics, Non-Narcotic/toxicity , Animals , Calcium Channel Blockers/administration & dosage , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/toxicity , Disease Models, Animal , Dose-Response Relationship, Drug , Hyperalgesia/drug therapy , Male , Neuralgia/physiopathology , Peptides/administration & dosage , Peptides/chemistry , Peptides/pharmacology , Rats , Rats, Sprague-Dawley , Rotarod Performance Test , Spinal Cord/drug effects , Spinal Cord/metabolism , Synaptic Transmission/drug effects , omega-Conotoxins/administration & dosage , omega-Conotoxins/toxicity
5.
Neuropharmacology ; 62(8): 2463-71, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22410393

ABSTRACT

Opioids are effective analgesic agents but serious adverse effects such as tolerance and withdrawal contribute to opioid dependence and limit their use. Opioid withdrawal involves numerous brain regions and includes suppression of dopamine release and activation of neurons in the ventral striatum. By contrast, acute opioids increase dopamine release. Like withdrawal, acute opioids also activate neurons in the ventral striatum, suggesting that different populations of ventral striatal neurons may be activated by withdrawal and acute opioid actions. Here, immunofluorescence for the activity-related immediate-early gene, c-Fos, was examined in transgenic reporter mouse lines by confocal microscopy to study the specific populations of ventral striatal neurons activated by morphine withdrawal and acute morphine. After chronic morphine, naloxone-precipitated withdrawal strongly increased expression of c-Fos immunoreactivity, predominantly in D2-receptor (D2R) medium-sized spiny neurons (MSNs) of the nucleus accumbens (NAc) core and shell regions. By contrast, a single injection of morphine exclusively activated c-Fos immunoreactivity in D1-receptor expressing (D1R) MSNs of the core and shell of the NAc. These results reveal a striking segregation of neuronal responses occurring in the two populations of MSNs of the NAc in response to morphine withdrawal and acute morphine.


Subject(s)
Morphine/pharmacology , Neurons/drug effects , Nucleus Accumbens/drug effects , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Substance Withdrawal Syndrome/metabolism , Animals , Cell Line , Male , Mice , Mice, Transgenic , Morphine Dependence/metabolism , Naloxone/pharmacology , Neurons/metabolism , Nucleus Accumbens/metabolism , Proto-Oncogene Proteins c-fos/metabolism
6.
Neuropharmacology ; 62(7): 2202-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22306793

ABSTRACT

The large diversity of peptides from venomous creatures with high affinity for molecules involved in the development and maintenance of neuropathic pain has led to a surge in venom-derived analgesic research. Some members of the α-conotoxin family from Conus snails which specifically target subtypes of nicotinic acetylcholine receptors (nAChR) have been shown to be effective at reducing mechanical allodynia in neuropathic pain models. We sought to determine if three such peptides, Vc1.1, AuIB and MII were effective following intrathecal administration in a rat neuropathic pain model because they exhibit different affinities for the major putative pain relieving targets of α-conotoxins. Intrathecal administration of α-conotoxins, Vc1.1, AuIB and MII into neuropathic rats reduced mechanical allodynia for up to 6 h without significant side effects. In vitro patch-clamp electrophysiology of primary afferent synaptic transmission revealed the mode of action of these toxins was not via a GABA(B)-dependent mechanism, and is more likely related to their action at nAChRs containing combinations of α3, α7 or other subunits. Intrathecal nAChR subunit-selective conotoxins are therefore promising tools for the effective treatment of neuropathic pain.


Subject(s)
Conotoxins/metabolism , Neuralgia/metabolism , Protein Subunits/metabolism , Receptors, Nicotinic/metabolism , Animals , Animals, Newborn , Conotoxins/administration & dosage , Injections, Spinal , Male , Neuralgia/drug therapy , Organ Culture Techniques , Protein Subunits/agonists , Rats , Rats, Sprague-Dawley , alpha7 Nicotinic Acetylcholine Receptor
7.
Pain ; 152(2): 259-266, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20889259

ABSTRACT

α-Conotoxins that are thought to act as antagonists of nicotinic acetylcholine receptors (nAChRs) containing α3-subunits are efficacious in several preclinical models of chronic pain. Potent interactions of Vc1.1 with other targets have suggested that the pain-relieving actions of α-conotoxins might be mediated by either α9α10 nAChRs or a novel GABA(B) receptor-mediated inhibition of N-type calcium channels. Here we establish that three α-conotoxins, Vc1.1, AuIB and MII have distinct selectivity profiles for these three potential targets. Their potencies after intramuscular administration were then determined for reversal of allodynia produced by partial nerve ligation in rats. Vc1.1, which potently inhibits α9α10 nAChRs and GABA(B)/Ca(2+) channels but weakly blocks α3ß2 and α3ß4 nAChRs, produced potent, long-lasting reversal of allodynia that were prevented by pre-treatment with the GABA(B) receptor antagonist, SCH50911. α-Conotoxin AuIB, a weak α3ß4 nAChR antagonist, inhibited GABA(B)/Ca(2+) channels but did not act on α9α10 nAChRs. AuIB also produced reversal of allodynia. These findings suggest that GABA(B) receptor-dependent inhibition of N-type Ca(2+) channels can mediate the sustained anti-allodynic actions of some α-conotoxins. However, MII, a potent α3ß2 nAChR antagonist but inactive on α9α10 and α3ß4 nAChRs and GABA(B)/Ca(2+) channels, was demonstrated to have short-acting anti-allodynic action. This suggests that α3ß2 nAChRs may also contribute to reversal of allodynia. Together, these findings suggest that inhibition of α9α10 nAChR is neither necessary nor sufficient for relief of allodynia and establish that α-conotoxins selective for GABA(B) receptor-dependent inhibition of N-type Ca(2+) channels relieve allodynia, and could therefore be developed to manage chronic pain.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/metabolism , Conotoxins/pharmacology , Pain/metabolism , Pain/prevention & control , Peripheral Nervous System Diseases/complications , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/metabolism , Animals , Calcium Channel Blockers/therapeutic use , Calcium Channels, N-Type/physiology , Cells, Cultured , Conotoxins/therapeutic use , Disease Models, Animal , Female , Male , Pain/etiology , Random Allocation , Rats , Rats, Sprague-Dawley , Rats, Wistar , Sciatic Neuropathy/complications , Sciatic Neuropathy/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/pathology
8.
Br J Pharmacol ; 163(6): 1214-22, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21175570

ABSTRACT

BACKGROUND AND PURPOSE: While arachidonyl ethanolamine (anandamide) produces pharmacological effects mediated by cannabinoid CB1 receptors, it is also an agonist at the transient receptor potential vanilloid type 1 (TRPV1) ion channel. This study examined the cellular actions of anandamide in the midbrain periaqueductal grey (PAG), a region implicated in the analgesic actions of cannabinoids, and which expresses both CB1 receptors and TRPV1. EXPERIMENTAL APPROACH: In vitro whole cell patch clamp recordings of glutamatergic excitatory postsynaptic currents (EPSCs) were made from rat and mouse PAG slices. KEY RESULTS: Capsaicin (1 µM) increased the rate, but not the amplitude of miniature EPSCs in subpopulations of neurons throughout the rat and mouse PAG. Capsaicin had no effect on miniature EPSCs in PAG neurons from TRPV1 knock-out mice. In mouse PAG neurons, anandamide (30 µM) had no effect on the rate of miniature EPSCs alone, or in the presence of either the CB1 antagonist AM251 (3 µM) or the TRPV1 antagonist iodoresiniferatoxin (300 nM). Anandamide produced a decrease in miniature EPSC rate in the presence of the fatty acid amide hydrolase (FAAH) inhibitor URB597 (1 µM). By contrast, anandamide produced an increase in miniature EPSC rate in the presence of both URB597 and AM251, which was absent in TRPV1 knock-out mice. CONCLUSIONS AND IMPLICATIONS: These results suggest that the actions of anandamide within PAG are limited by enzymatic degradation by FAAH. FAAH blockade unmasks both presynaptic inhibition and excitation of glutamatergic synaptic transmission which are mediated via CB1 receptors and TRPV1 respectively.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Periaqueductal Gray/physiology , Receptor, Cannabinoid, CB1/metabolism , Synaptic Transmission/physiology , TRPV Cation Channels/metabolism , Amidohydrolases/genetics , Amidohydrolases/metabolism , Animals , Arachidonic Acids/pharmacology , Cannabinoid Receptor Modulators/pharmacology , Capsaicin/pharmacology , Endocannabinoids , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Polyunsaturated Alkamides/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/genetics , TRPV Cation Channels/genetics
9.
Obstet Gynecol Surv ; 65(11): 729-35, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21375789

ABSTRACT

UNLABELLED: Chlamydia trachomatis is the most common bacterial sexually transmitted infection in the United States. This disease disproportionately affects adolescent minority women, and untreated infection can lead to lasting reproductive tract morbidity. Recommendations for primary prevention include patient counseling to decrease risky behavior and increase barrier protection use; secondary prevention recommendations include screening and treatment of affected individuals and their sexual partners, barrier contraception use, as well as counseling to decrease behaviors that lead to reinfection. Despite these strategies, both incidence and prevalence of Chlamydia have continued to escalate in this population. Interventions to decrease chlamydial infection should encompass all facets of primary and secondary prevention as well as address the fundamental barrier to prevention-lack of perception of risk in this young age group. TARGET AUDIENCE: Obstetricians & Gynecologists, Family Physicians. LEARNING OBJECTIVES: After completion of this educational activity, the obstetrician/gynecologist should be better able to identify current screening guidelines to test for chlamydial infection in sexually active adolescents; obtain more thorough sexual histories, and understand dynamics of disproportionate disease burden in minority teens; recognize and act to decrease the high risk of reinfection in this patient population; and employ novel methods to increase STI screening.


Subject(s)
Chlamydia Infections/epidemiology , Chlamydia Infections/prevention & control , Chlamydia trachomatis , Sexually Transmitted Diseases, Bacterial/epidemiology , Sexually Transmitted Diseases, Bacterial/prevention & control , Adolescent , Adolescent Behavior , Chlamydia Infections/transmission , Contraception Behavior/psychology , Contraceptive Devices, Female , Female , Genital Diseases, Female/epidemiology , Genital Diseases, Female/microbiology , Genital Diseases, Female/prevention & control , Humans , Minority Groups , Prevalence , Sexual Behavior
10.
Mol Pharmacol ; 77(2): 139-48, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19892914

ABSTRACT

Neuronal (N)-type Ca(2+) channel-selective omega-conotoxins have emerged as potential new drugs for the treatment of chronic pain. In this study, two new omega-conotoxins, CVIE and CVIF, were discovered from a Conus catus cDNA library. Both conopeptides potently displaced (125)I-GVIA binding to rat brain membranes. In Xenopus laevis oocytes, CVIE and CVIF potently and selectively inhibited depolarization-activated Ba(2+) currents through recombinant N-type (alpha1(B-b)/alpha(2)delta1/beta(3)) Ca(2+) channels. Recovery from block increased with membrane hyperpolarization, indicating that CVIE and CVIF have a higher affinity for channels in the inactivated state. The link between inactivation and the reversibility of omega-conotoxin action was investigated by creating molecular diversity in beta subunits: N-type channels with beta(2a) subunits almost completely recovered from CVIE or CVIF block, whereas those with beta(3) subunits exhibited weak recovery, suggesting that reversibility of the omega-conotoxin block may depend on the type of beta-subunit isoform. In rat dorsal root ganglion sensory neurons, neither peptide had an effect on low-voltage-activated T-type channels but potently and selectively inhibited high voltage-activated N-type Ca(2+) channels in a voltage-dependent manner. In rat spinal cord slices, both peptides reversibly inhibited excitatory monosynaptic transmission between primary afferents and dorsal horn superficial lamina neurons. Homology models of CVIE and CVIF suggest that omega-conotoxin/voltage-gated Ca(2+) channel interaction is dominated by ionic/electrostatic interactions. In the rat partial sciatic nerve ligation model of neuropathic pain, CVIE and CVIF (1 nM) significantly reduced allodynic behavior. These N-type Ca(2+) channel-selective omega-conotoxins are therefore useful as neurophysiological tools and as potential therapeutic agents to inhibit nociceptive pain pathways.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/metabolism , Recombinant Proteins/antagonists & inhibitors , omega-Conotoxins/pharmacology , Amino Acid Sequence , Analgesics, Non-Narcotic/chemistry , Analgesics, Non-Narcotic/isolation & purification , Animals , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/isolation & purification , Calcium Channels, N-Type/physiology , Cells, Cultured , Conus Snail , Dose-Response Relationship, Drug , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Male , Molecular Sequence Data , Patch-Clamp Techniques , Protein Structure, Tertiary , Rabbits , Rats , Rats, Sprague-Dawley , Rats, Wistar , Recombinant Proteins/genetics , Xenopus laevis , omega-Conotoxins/chemistry , omega-Conotoxins/isolation & purification
11.
Br J Pharmacol ; 154(2): 384-96, 2008 May.
Article in English | MEDLINE | ID: mdl-18414400

ABSTRACT

A large range of neuroadaptations develop in response to chronic opioid exposure and these are thought to be more or less critical for expression of the major features of opioid addiction: tolerance, withdrawal and processes that may contribute to compulsive use and relapse. This review considers these adaptations at different levels of organization in the nervous system including tolerance at the mu-opioid receptor itself, cellular tolerance and withdrawal in opioid-sensitive neurons, systems tolerance and withdrawal in opioid-sensitive nerve networks, as well as synaptic plasticity in opioid sensitive nerve networks. Receptor tolerance appears to involve enhancement of mechanisms of receptor regulation, including desensitization and internalization. Adaptations causing cellular tolerance are more complex but several important processes have been identified including upregulation of cAMP/PKA and cAMP response element-binding signalling and perhaps the mitogen activated PK cascades in opioid sensitive neurons that might not only influence tolerance and withdrawal but also synaptic plasticity during cycles of intoxication and withdrawal. The potential complexity of network, or systems adaptations that interact with opioid-sensitive neurons is great but some candidate neuropeptide systems that interact with mu-opioid sensitive neurons may play a role in tolerance and withdrawal, as might activation of glial signalling. Implication of synaptic forms of learning such as long term potentiation and long term depression in opioid addiction is still in its infancy but this ultimately has the potential to identify specific synapses that contribute to compulsive use and relapse.


Subject(s)
Analgesics, Opioid/adverse effects , Behavior, Addictive/metabolism , Brain/drug effects , Drug Tolerance , Opioid-Related Disorders/metabolism , Substance Withdrawal Syndrome/metabolism , Adaptation, Physiological , Adenylyl Cyclases/metabolism , Animals , Behavior, Addictive/physiopathology , Brain/enzymology , Brain/metabolism , Brain/physiopathology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , Neural Pathways/drug effects , Neural Pathways/metabolism , Neuronal Plasticity/drug effects , Opioid-Related Disorders/physiopathology , Opioid-Related Disorders/psychology , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/metabolism , Signal Transduction/drug effects , Substance Withdrawal Syndrome/physiopathology , Substance Withdrawal Syndrome/psychology , Synaptic Transmission/drug effects
12.
Mol Pharmacol ; 72(6): 1406-10, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17804600

ABSTRACT

The synthetic alpha-conotoxin Vc1.1 is a small disulfide bonded peptide currently in development as a treatment for neuropathic pain. Unlike Vc1.1, the native post-translationally modified peptide vc1a does not act as an analgesic in vivo in rat models of neuropathic pain. It has recently been proposed that the primary target of Vc1.1 is the alpha9alpha10 nicotinic acetylcholine receptor (nAChR). We show that Vc1.1 and its post-translationally modified analogs vc1a, [P6O]Vc1.1, and [E14gamma]Vc1.1 are equally potent at inhibiting ACh-evoked currents mediated by alpha9alpha10 nAChRs. This suggests that alpha9alpha10 nAChRs are unlikely to be the molecular mechanism or therapeutic target of Vc1.1 for the treatment of neuropathic pain.


Subject(s)
Conotoxins/metabolism , Drug Delivery Systems , Pain/metabolism , Protein Subunits/metabolism , Receptors, Nicotinic/metabolism , Amino Acid Sequence , Animals , Conotoxins/therapeutic use , Drug Delivery Systems/methods , Female , Male , Molecular Sequence Data , Pain/genetics , Protein Subunits/genetics , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/genetics , Xenopus laevis
13.
Proc Natl Acad Sci U S A ; 103(45): 17030-5, 2006 Nov 07.
Article in English | MEDLINE | ID: mdl-17077153

ABSTRACT

The tetrodotoxin-resistant voltage-gated sodium channel (VGSC) Na(v)1.8 is expressed predominantly by damage-sensing primary afferent nerves and is important for the development and maintenance of persistent pain states. Here we demonstrate that muO-conotoxin MrVIB from Conus marmoreus displays substantial selectivity for Na(v)1.8 and inhibits pain behavior in models of persistent pain. In rat sensory neurons, submicromolar concentrations of MrVIB blocked tetrodotoxin-resistant current characteristic of Na(v)1.8 but not Na(v)1.9 or tetrodotoxin-sensitive VGSC currents. MrVIB blocked human Na(v)1.8 expressed in Xenopus oocytes with selectivity at least 10-fold greater than other VGSCs. In neuropathic and chronic inflammatory pain models, allodynia and hyperalgesia were both reduced by intrathecal infusion of MrVIB (0.03-3 nmol), whereas motor side effects occurred only at 30-fold higher doses. In contrast, the nonselective VGSC blocker lignocaine displayed no selectivity for allodynia and hyperalgesia versus motor side effects. The actions of MrVIB reveal that VGSC antagonists displaying selectivity toward Na(v)1.8 can alleviate chronic pain behavior with a greater therapeutic index than nonselective antagonists.


Subject(s)
Conotoxins/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Pain/drug therapy , Animals , Chronic Disease , Conotoxins/administration & dosage , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , In Vitro Techniques , Male , NAV1.8 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Oocytes/drug effects , Oocytes/metabolism , Pain/physiopathology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sodium Channel Blockers/administration & dosage , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Sodium Channels/genetics , Sodium Channels/metabolism , Tetrodotoxin/pharmacology , Xenopus laevis
14.
Neuroscience ; 137(4): 1389-96, 2006.
Article in English | MEDLINE | ID: mdl-16359817

ABSTRACT

Previous studies using c-Fos immunohistochemistry suggest that a sub-population of neurons in the midbrain periaqueductal gray region is activated during opioid withdrawal. The neurochemical identity of these cells is unknown but cellular physiological studies have implicated GABAergic neurons. The present study investigated whether GABAergic neurons are activated in the mouse periaqueductal gray during opioid withdrawal using dual-antibody immunohistochemistry for Fos and glutamic acid decarboxylase. Both chronic opioid treatment and naloxone-precipitated opioid withdrawal increased Fos expression in the periaqueductal gray, with the greatest increase being four-fold in the caudal ventrolateral subdivision following withdrawal. Neurons stained for both Fos and glutamic acid decarboxylase were greatly enhanced in all subdivisions of the periaqueductal gray following withdrawal, particularly in the lateral and ventrolateral divisions where the increase was up to 70-fold. These results suggest that activation of a subpopulation of GABAergic interneurons in the periaqueductal gray plays a role in opioid withdrawal.


Subject(s)
Genes, fos , Glutamate Decarboxylase/metabolism , Narcotics/toxicity , Neurons/physiology , Periaqueductal Gray/physiopathology , Substance Withdrawal Syndrome/physiopathology , Animals , Disease Models, Animal , Gene Expression Regulation , Mice , Naloxone/pharmacology , Neurons/enzymology
17.
Handb Exp Pharmacol ; (168): 367-83, 2005.
Article in English | MEDLINE | ID: mdl-16596781

ABSTRACT

The cannabinoid neurotransmitter system comprises cannabinoid G protein-coupled membrane receptors (CB1 and CB2), endogenous cannabinoids (endocannabinoids), as well as mechanisms for their synthesis, membrane transport and metabolism. Within the brain the marijuana constituent delta9-tetrahydrocannabinol (THC) produces its pharmacological actions by acting on cannabinoid CB1 receptors. THC modulates neuronal excitability by inhibiting synaptic transmission via presynaptic CB1-mediated mechanisms. More recently, it has been established that physiological stimulation of neurons can induce the synthesis of endocannabinoids, which also modulate synaptic transmission via cannabinoid CB1 and other receptor systems. These endogenously synthesised endocannabinoids appear to act as retrograde signalling agents, reducing synaptic inputs onto the stimulated neuron in a highly selective and restricted manner. In this review we describe the cellular mechanisms underlying retrograde endocannabinoid signalling.


Subject(s)
Cannabinoid Receptor Modulators/physiology , Endocannabinoids , Signal Transduction/physiology , Animals , Calcium/metabolism , Humans , Neuronal Plasticity , Receptor, Cannabinoid, CB1/physiology , Receptors, Metabotropic Glutamate/physiology , Receptors, Presynaptic/physiology
18.
Pain ; 111(1-2): 30-7, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15327806

ABSTRACT

This study examined the cellular actions of the anti-migraine drug sumatriptan, on neurons in the substantia gelatinosa of the spinal trigeminal nucleus pars caudalis. Sumatriptan inhibited the miniature EPSC (mEPSC) rate in a dose dependent fashion, with an EC(50) of 250 nM. Sumatriptan (3 microM) inhibited the mEPSC rate by 36%, without altering the mEPSC amplitude. This effect was partially reversed by the 5HT(1D) specific antagonist BRL15572 (10 microM). In contrast, the 5HT(1B) agonist CP93129 (10 microm) did not alter the mEPSC rate. Furthermore, sumatriptan (3 microM) decreased the amplitude of electrically evoked EPSCs (eEPSC) by 40%. After incubating the slices in ketanserin (an antagonist which shows selectivity for 5HT(1D) over 5HT(1B) receptors) sumatriptan had little effect on eEPSC amplitude. In control conditions paired stimuli resulted in paired pulse depression (PPD; the ratio eEPSC(2)/eEPSC(1)=0.7+/-0.01), whilst in the presence of sumatriptan the PPD was blocked (ratio eEPSC(2)/eEPSC(1)=0.9+/-0.1). Sumatriptan produced no post-synaptic membrane current and had no significant effect on membrane conductance over a range of membrane potentials (-60 to -130 mV). RT-PCR experiments revealed the presence of mRNA for both 5HT(1D) and 5HT(1B) receptor subtypes in the trigeminal ganglia and subnucleus caudalis. These data suggest that sumatriptan acts pre-synaptically on trigeminal primary afferent central terminals to reduce the probability of release of glutamate, and that this action is mediated through 5HT(1D) receptors.


Subject(s)
Serotonin Receptor Agonists/pharmacology , Substantia Gelatinosa/drug effects , Sumatriptan/pharmacology , Trigeminal Caudal Nucleus/drug effects , Animals , Excitatory Postsynaptic Potentials/drug effects , Patch-Clamp Techniques , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1B/genetics , Receptor, Serotonin, 5-HT1D/genetics , Substantia Gelatinosa/cytology , Substantia Gelatinosa/physiology , Trigeminal Caudal Nucleus/cytology , Trigeminal Caudal Nucleus/physiology
20.
Neuroscience ; 121(4): 959-73, 2003.
Article in English | MEDLINE | ID: mdl-14580946

ABSTRACT

The specific role of the Delta opioid receptor (DOR), in opioid-induced respiratory depression in the ventral respiratory group (VRG) is largely unknown. Here, we sought to determine (1) the relationship between DOR-immunoreactive (ir) boutons, bulbospinal and functionally identified respiratory neurons in the VRG and (2) the effects of microinjection of the selective DOR agonist, D-Pen 2,5-enkephalin (DPDPE), into different subdivisions of the VRG, on phrenic nerve discharge and mean arterial pressure. Following injections of retrograde tracer into the spinal cord or intracellular labelling of respiratory neurons, in Sprague-Dawley rats, brainstem sections were processed for retrograde or intracellular labelling and DOR-ir. Bulbospinal neurons were apposed by DOR-ir boutons regardless of whether they projected to single (cervical or thoracic ventral horn) or multiple (cervical and thoracic ventral horn) targets in the spinal cord. In the VRG, a total of 24 +/- 5% (67 +/- 13/223 +/- 49) of neurons projecting to the cervical ventral horn, and 37 +/- 3% (96 +/- 22/255 +/- 37) of neurons projecting to the thoracic ventral horn, received close appositions from DOR-ir boutons. Furthermore, DOR-ir boutons closely apposed six of seven intracellularly labelled neurons, whilst the remaining neuron itself possessed boutons that were DOR-ir. DPDPE was microinjected (10 mM, 60 nl, unilateral) into regions of respiratory field activity in the VRG of anaesthetised, vagotomised rats, and the effects on phrenic nerve discharge and mean arterial pressure were recorded. DPDPE depressed phrenic nerve amplitude, with little effect on phrenic nerve frequency in the Bötzinger complex, pre-Bötzinger complex and rVRG, the greatest effects occurring in the Bötzinger complex. The results indicate that the DOR is located on afferent inputs to respiratory neurons in the VRG. Activation of the DOR in the VRG is likely to inhibit the release of neurotransmitters from afferent inputs that modulate the pattern of activity of VRG neurons.


Subject(s)
Efferent Pathways/metabolism , Medulla Oblongata/metabolism , Periodicity , Receptors, Opioid, delta/metabolism , Respiration/drug effects , Respiratory Center/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Analgesics, Opioid/pharmacology , Animals , Biological Clocks/drug effects , Biological Clocks/physiology , Blood Pressure/drug effects , Blood Pressure/physiology , Efferent Pathways/cytology , Efferent Pathways/drug effects , Enkephalin, D-Penicillamine (2,5)-/pharmacology , Immunohistochemistry , Male , Medulla Oblongata/cytology , Medulla Oblongata/drug effects , Phrenic Nerve/drug effects , Phrenic Nerve/physiology , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/drug effects , Respiratory Center/cytology , Respiratory Center/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...