Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Neuroscience ; 168(2): 371-86, 2010 Jun 30.
Article in English | MEDLINE | ID: mdl-20394803

ABSTRACT

Social deprivation in early life disrupts emotionality and attentional processes in humans. Rearing rats in isolation reproduces some of these abnormalities, which are attenuated by daily handling. However, the neurochemical mechanisms underlying these responses remain poorly understood. We hypothesized that post-weaning social isolation alters the endocannabinoid system, a neuromodulatory system that controls emotional responding. We characterized behavioral consequences of social isolation and evaluated whether handling would reverse social isolation-induced alterations in behavioral reactivity to context and the endocannabinoid system. At weaning, pups were single or group housed and concomitantly handled or not handled daily until adulthood. Rats were tested in emotionality- and attentional-sensitive behavioral assays (open field, elevated plus maze, startle and prepulse inhibition). Cannabinoid receptor densities and endocannabinoid levels were quantified in a separate group of rats. Social isolation negatively altered behavioral responding. Socially-isolated rats that were handled showed less deficits in the open field, elevated plus maze, and prepulse inhibition tests. Social isolation produced site-specific alterations (supraoptic nucleus, ventrolateral thalamus, rostral striatum) in cannabinoid receptor densities compared to group rearing. Handling altered the endocannabinoid system in neural circuitry controlling emotional expression. Handling altered endocannabinoid content (prefrontal and piriform cortices, nucleus accumbens) and cannabinoid receptor densities (lateral globus pallidus, cingulate and piriform cortices, hippocampus) in a region-specific manner. Some effects of social isolation on the endocannabinoid system were moderated by handling. Isolates were unresponsive to handling-induced increases in cannabinoid receptor densities (caudal striatum, anterior thalamus), but were sensitive to handling-induced changes in endocannabinoid content (piriform, prefrontal cortices), compared to group-reared rats. Our findings suggest alterations in the endocannabinoid system may contribute to the abnormal isolate phenotype. Handling modifies the endocannabinoid system and behavioral reactivity to context, but surmounts only some effects of social isolation. These data implicate a pivotal role for the endocannabinoid system in stress adaptation and emotionality-related disturbances.


Subject(s)
Behavior, Animal , Cannabinoid Receptor Modulators/physiology , Endocannabinoids , Handling, Psychological , Social Isolation , Animals , Attention , Brain/metabolism , Emotions , Female , Male , Maze Learning , Rats , Rats, Sprague-Dawley , Receptors, Cannabinoid/metabolism , Reflex, Startle , Signal Transduction
2.
Br J Pharmacol ; 153(7): 1341-3, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18297102

ABSTRACT

The endocannabinoid lipid 2-arachidonoylglycerol (2-AG) is deactivated by intracellular hydrolysis catalyzed by monoacylglycerol lipase. 2-AG also serves as a substrate for oxidative metabolism catalyzed by cyclooxygenase 2 (COX-2). However, products of COX-2-mediated metabolism of endocannabinoids have not been identified in vivo. Hu and colleagues in this issue of the BJP demonstrate that COX-2 converts 2-AG into a biologically active, pro-nociceptive compound, prostaglandin E2 glycerol ester (PGE2-G). PGE2-G produces hyperalgesia in vivo and activates a rapidly acting transcription factor, nuclear factor kappa-B in vitro. These biological actions may be attributed to a unique receptor. This report of pro-nociceptive actions of an endogenous COX-2 metabolite of 2-AG that are largely opposite to known anti-nociceptive and anti-inflammatory actions of endocannabinoids has physiological relevance. These discoveries place renewed emphasis on the importance of understanding the highly interactive nature of lipid signalling pathways in the nervous system and the physiological roles of these lipid mediators in controlling homeostasis.


Subject(s)
Arachidonic Acids/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/analogs & derivatives , Dinoprostone/metabolism , Glycerides/metabolism , Animals , Dinoprostone/biosynthesis , Endocannabinoids , Homeostasis/physiology , Humans , Oxidation-Reduction , Signal Transduction
3.
Br J Pharmacol ; 153(2): 319-34, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17994113

ABSTRACT

Cannabinoids suppress behavioural responses to noxious stimulation and suppress nociceptive transmission through activation of CB1 and CB2 receptor subtypes. CB1 receptors are expressed at high levels in the central nervous system (CNS), whereas CB2 receptors are found predominantly, but not exclusively, outside the CNS. CB2 receptors are also upregulated in the CNS and dorsal root ganglia by pathological pain states. Here, we review behavioural, neurochemical and electrophysiological data, which identify cannabinoid CB2 receptors as a therapeutic target for treating pathological pain states with limited centrally, mediated side effects. The development of CB2-selective agonists (with minimal affinity for CB1) as well as mutant mice lacking CB2 receptors has provided pharmacological and genetic tools required to evaluate the effectiveness of CB2 agonists in suppressing persistent pain states. This review will examine the efficacy of cannabinoid CB2-selective agonists in suppressing acute, inflammatory and neuropathic nociception following systemic and local routes of administration. Data derived from behavioural, neurochemical and neurophysiological approaches are discussed to better understand the relationship between antinociceptive effects induced by CB2-selective agonists in behavioural studies and neural mechanisms of pain suppression. Finally, the therapeutic potential and possible limitations of CB2-based pharmacotherapies for pathological pain states induced by tissue and nerve injury are discussed.


Subject(s)
Inflammation/complications , Pain/drug therapy , Pain/etiology , Peripheral Nervous System Diseases/complications , Receptor, Cannabinoid, CB2/drug effects , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Humans , Receptor, Cannabinoid, CB2/agonists
4.
Br J Pharmacol ; 152(5): 765-77, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17572696

ABSTRACT

BACKGROUND AND PURPOSE: The ability of cannabinoids to suppress mechanical hypersensitivity (mechanical allodynia) induced by treatment with the chemotherapeutic agent vincristine was evaluated in rats. Sites of action were subsequently identified. EXPERIMENTAL APPROACH: Mechanical hypersensitivity developed over the course of ten daily injections of vincristine relative to groups receiving saline at the same times. Effects of the CB1/CB2 receptor agonist WIN55,212-2, the receptor-inactive enantiomer WIN55,212-3, the CB2-selective agonist (R,S)-AM1241, the opiate agonist morphine and vehicle on chemotherapy-induced neuropathy were evaluated. WIN55,212-2 was administered intrathecally (i.t.) or locally in the hindpaw to identify sites of action. Pharmacological specificity was established using competitive antagonists for CB1 (SR141716) or CB2 receptors (SR144528). KEY RESULTS: Systemic administration of WIN55,212-2, but not WIN55,212-3, suppressed vincristine-evoked mechanical allodynia. A leftward shift in the dose-response curve was observed following WIN55,212-2 relative to morphine treatment. The CB1 (SR141716) and CB2 (SR144528) antagonists blocked the anti-allodynic effects of WIN55,212-2. (R,S)-AM1241 suppressed vincristine-induced mechanical hypersensitivity through a CB2 mechanism. Both cannabinoid agonists suppressed vincristine-induced mechanical hypersensitivity without inducing catalepsy. Spinal sites of action are implicated in cannabinoid modulation of chemotherapy-induced neuropathy. WIN55,212-2, but not WIN55,212-3, administered i.t. suppressed vincristine-evoked mechanical hypersensitivity at doses that were inactive following local hindpaw administration. Spinal coadministration of both the CB1 and CB2 antagonists blocked the anti-allodynic effects of WIN55,212-2. CONCLUSIONS AND IMPLICATIONS: Cannabinoids suppress the maintenance of vincristine-induced mechanical allodynia through activation of CB1 and CB2 receptors. These anti-allodynic effects are mediated, at least in part, at the level of the spinal cord.


Subject(s)
Neuralgia/prevention & control , Receptor, Cannabinoid, CB1/physiology , Receptor, Cannabinoid, CB2/physiology , Vincristine/toxicity , Animals , Benzoxazines/pharmacology , Body Weight/drug effects , Camphanes/pharmacology , Cannabinoids/pharmacology , Catalepsy/chemically induced , Catalepsy/prevention & control , Dose-Response Relationship, Drug , Hindlimb , Hyperesthesia/chemically induced , Hyperesthesia/prevention & control , Injections, Intraperitoneal , Injections, Spinal , Male , Morphine/pharmacology , Morpholines/pharmacology , Naphthalenes/pharmacology , Neuralgia/chemically induced , Pain Measurement/instrumentation , Pain Measurement/methods , Pain Threshold/drug effects , Physical Stimulation , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB2/agonists , Rimonabant , Thermosensing/physiology , Vincristine/administration & dosage
5.
Br J Pharmacol ; 150(6): 673-5, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17293886

ABSTRACT

2-Arachidonoylglycerol (2-AG) is an endogenous cannabinoid (endocannabinoid) lipid whose functions remain poorly understood. Guindon and colleagues report the novel finding that exogenous application of 2-AG induces peripheral antinociceptive effects that are mediated, at least in part, by actions at peripheral cannabinoid CB(2) receptors. URB602, a recently described inhibitor of monoacylglycerol lipase, an enzyme that catalyzes 2-AG hydrolysis in vivo, also induced peripheral antinociceptive effects and enhanced the actions of 2-AG. Peripheral analgesic mechanisms represent promising therapeutic targets for suppressing pain in the absence of unwanted central nervous system side-effects (e.g. psychoactivity) associated with activation of central CB(1) receptors. The therapeutic potential of inhibitors of 2-AG deactivation for the treatment of inflammatory pain is discussed.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Enzyme Inhibitors/pharmacology , Monoacylglycerol Lipases/antagonists & inhibitors , Animals , Arachidonic Acids/metabolism , Arachidonic Acids/pharmacology , Biphenyl Compounds/pharmacology , Endocannabinoids , Glycerides/metabolism , Glycerides/pharmacology , Humans , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/drug effects , Receptor, Cannabinoid, CB2/metabolism
6.
Br J Pharmacol ; 150(2): 153-63, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17160008

ABSTRACT

BACKGROUND AND PURPOSE: Effects of locally administered agonists and antagonists for cannabinoid CB(1) and CB(2) receptors on mechanical and thermal hypersensitivity were compared after the establishment of chronic inflammation. EXPERIMENTAL APPROACH: Carrageenan was administered unilaterally to the rat hindpaw on day 1. Prophylactic efficacy of locally administered CB(1)- and CB(2)-selective agonists -arachidonyl-2-chloroethylamide (ACEA) and (R,S)-(2-iodo-5-nitro-phenyl)-[l-(l-methyl-piperidin-2-ylmethyl)-lH-ubdik-3-yl]-methanone ((R,S)-AM1241), respectively- on mechanical and thermal hypersensitivity were compared on day 2. Pharmacological specificity was evaluated using locally administered CB(1) and CB(2)-selective antagonists -N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamidehydrochloride (SR141716A) and N-[(1S)-endo-1,3,3-trimethyl bicycle [2.2.1] heptan-2-yl]-5-(4-chloro-3-methylphenyl)-1-(4-methylbenzyl)-pyrazole-3-carboxamide (SR144528), respectively. KEY RESULTS: Administration of either ACEA or AM1241 to the inflamed but not noninflamed paw suppressed the maintenance of carrageenan-evoked mechanical hyperalgesia and tactile allodynia and attenuated thermal hyperalgesia. The ACEA-induced suppression of mechanical and thermal hypersensitivity was blocked by local injection of SR141716A but not SR144528. AM1241 suppressed mechanical hypersensitivity with the reverse pharmacological specificity. The AM1241-induced suppression of thermal hyperalgesia was blocked by SR144528 and to a lesser extent by SR14176A. Co-administration of ACEA with AM1241 in the inflamed paw increased the magnitude but not the duration of thermal antihyperalgesia compared to intraplantar administration of either agonist alone. CONCLUSIONS AND IMPLICATIONS: Cannabinoids act locally through distinct CB(1) and CB(2) mechanisms to suppress mechanical hypersensitivity after the establishment of chronic inflammation, at doses that produced modest changes in thermal hyperalgesia. Additive antihyperalgesic effects were observed following prophylactic co-administration of the CB(1)- and CB(2)-selective agonists. Our results suggest that peripheral cannabinoid antihyperalgesic actions may be exploited for treatment of inflammatory pain states.


Subject(s)
Pain/drug therapy , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB2/agonists , Animals , Arachidonic Acids/pharmacology , Arachidonic Acids/therapeutic use , Cannabinoids/pharmacology , Cannabinoids/therapeutic use , Carrageenan , Chronic Disease , Drug Synergism , Hot Temperature , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Male , Pain/metabolism , Pain/physiopathology , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Touch
7.
Handb Exp Pharmacol ; (168): 509-54, 2005.
Article in English | MEDLINE | ID: mdl-16596786

ABSTRACT

A large body of literature indicates that cannabinoids suppress behavioral responses to acute and persistent noxious stimulation in animals. This review examines neuroanatomical, behavioral, and neurophysiological evidence supporting a role for cannabinoids in suppressing pain at spinal, supraspinal, and peripheral levels. Localization studies employing receptor binding and quantitative autoradiography, immunocytochemistry, and in situ hybridization are reviewed to examine the distribution of cannabinoid receptors at these levels and provide a neuroanatomical framework with which to understand the roles of endogenous cannabinoids in sensory processing. Pharmacological and transgenic approaches that have been used to study cannabinoid antinociceptive mechanisms are described. These studies provide insight into the functional roles of cannabinoid CB1 (CB1R) and CB2 (CB2R) receptor subtypes in cannabinoid antinociceptive mechanisms, as revealed in animal models of acute and persistent pain. The role of endocannabinoids and related fatty acid amides that are implicated in endogenous mechanisms for pain suppression are discussed. Human studies evaluating therapeutic potential of cannabinoid pharmacotherapies in experimental and clinical pain syndromes are evaluated. The potential of exploiting cannabinoid antinociceptive mechanisms in novel pharmacotherapies for pain is discussed.


Subject(s)
Cannabinoids/pharmacology , Pain/drug therapy , Receptor, Cannabinoid, CB1/physiology , Receptor, Cannabinoid, CB2/physiology , Animals , Cannabinoid Receptor Modulators/physiology , Cannabinoids/therapeutic use , Humans , Hyperalgesia/physiopathology , Nociceptors/physiology , Pain/physiopathology , RNA, Messenger/analysis , Receptor, Cannabinoid, CB1/analysis , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/drug effects , Spinal Cord/drug effects , Spinal Cord/physiology
8.
J Neurophysiol ; 92(6): 3562-74, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15317842

ABSTRACT

Effects of the CB2-selective cannabinoid agonist AM1241 on activity evoked in spinal wide dynamic range (WDR) neurons by transcutaneous electrical stimulation were evaluated in urethane-anesthetized rats. Recordings were obtained in both the absence and the presence of carrageenan inflammation. AM1241, administered intravenously or locally in the paw, suppressed activity evoked by transcutaneous electrical stimulation during the development of inflammation. Decreases in WDR responses resulted from a suppression of C-fiber-mediated activity and windup. Abeta- and Adelta-fiber-mediated responses were not reliably altered. The AM1241-induced suppression of electrically evoked responses was blocked by the CB2 antagonist SR144528 but not by the CB1 antagonist SR141716A. AM1241 (33 microg/kg intraplantar [i.p.l.]), administered to the carrageenan-injected paw, suppressed activity evoked in WDR neurons relative to groups receiving vehicle in the same paw or AM1241 in the opposite (noninflamed) paw. The electrophysiological effects of AM1241 (330 microg/kg intravenous [i.v.]) were greater in rats receiving i.p.l. carrageenan compared with noninflamed rats receiving an i.p.l. injection of vehicle. AM1241 failed to alter the activity of purely nonnociceptive neurons recorded in the lumbar dorsal horn. Additionally, AM1241 (330 microg/kg i.v. and i.p.l.; 33 microg/kg i.p.l.) reduced the diameter of the carrageenan-injected paw. The AM1241-induced decrease in peripheral edema was blocked by the CB2 but not by the CB1 antagonist. These data demonstrate that activation of cannabinoid CB2 receptors is sufficient to suppress neuronal activity at central levels of processing in the spinal dorsal horn. Our findings are consistent with the ability of AM1241 to normalize nociceptive thresholds and produce antinociception in inflammatory pain states.


Subject(s)
Nerve Fibers, Unmyelinated/physiology , Nociceptors/physiology , Posterior Horn Cells/physiology , Receptor, Cannabinoid, CB2/physiology , Analgesics/pharmacology , Animals , Camphanes/pharmacology , Cannabinoids , Carrageenan , Edema/chemically induced , Edema/physiopathology , Electric Stimulation , Inflammation/chemically induced , Inflammation/physiopathology , Male , Nerve Fibers, Unmyelinated/drug effects , Nociceptors/drug effects , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Rimonabant
9.
Brain Res ; 987(2): 176-85, 2003 Oct 17.
Article in English | MEDLINE | ID: mdl-14499961

ABSTRACT

The effects of neurotoxic destruction of catecholaminergic projections to the spinal cord on cannabinoid antinociception were examined in models of acute and tonic nociception. High performance liquid chromatography was used to quantify monoamine levels in sham-operated and lesioned rats. Intrathecal administration of the catecholamine neurotoxin 6-hydroxydopamine (6-OHDA) induced a selective depletion of norepinephrine (by approximately 85% of control) in rat lumbar spinal cord without altering levels of dopamine or serotonin. By contrast, brain levels of monoamines did not differ in sham-operated and lesioned rats. Pain behavior was similar in sham-operated and lesioned rats receiving vehicle in models of both acute and tonic nociception. The cannabinoid agonist WIN55,212-2 (5 or 10 mg/kg, i.p.) produced antinociception in the tail-flick test in sham-operated rats. The antinociceptive effect of WIN55,212-2 was attenuated relative to control conditions in rats depleted of spinal norepinephrine. WIN55,212-2 suppressed tonic pain behavior in the formalin test in sham-operated rats during phase 2 (15-60 min post formalin) of nociceptive responding. By contrast, in lesioned rats, WIN55,212-2 suppressed pain behavior during phase 1 (0-9.9 min) and phase 2A (10-39.9 min), but not during phase 2B (40-60 min). The cannabinoid agonist suppressed formalin-evoked Fos protein expression, a marker of neuronal activity, in the lumbar dorsal horn of sham-operated rats, but no suppression was observed in lesioned rats. The number of formalin-evoked Fos-like immunoreactive (FLI) cells was greater in lamina I and II of lesioned rats relative to sham-operated rats. These data indicate that the suppressive effect of the cannabinoid on formalin-evoked Fos protein expression in the superficial dorsal horn was attenuated following destruction of descending noradrenergic pathways. Our data are consistent with the hypothesis that cannabinoids produce antinociception, in part, by modulating descending noradrenergic systems and support a differential involvement of noradrenergic projections to the spinal cord in cannabinoid modulation of acute versus tonic nociception.


Subject(s)
Cannabinoids/pharmacology , Disease Models, Animal , Norepinephrine/metabolism , Oxidopamine/toxicity , Pain Measurement/drug effects , Analgesics/pharmacology , Animals , Benzoxazines , Male , Morpholines/pharmacology , Naphthalenes/pharmacology , Neural Pathways/drug effects , Neural Pathways/physiology , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Spinal Cord/metabolism
10.
Neuroscience ; 119(3): 747-57, 2003.
Article in English | MEDLINE | ID: mdl-12809695

ABSTRACT

Activation of cannabinoid CB(2) receptors attenuates thermal nociception in untreated animals while failing to produce centrally mediated effects such as hypothermia and catalepsy [Pain 93 (2001) 239]. The present study was conducted to test the hypothesis that activation of CB(2) in the periphery suppresses the development of inflammatory pain as well as inflammation-evoked neuronal activity at the level of the CNS. The CB(2)-selective cannabinoid agonist AM1241 (100, 330 micrograms/kg i.p.) suppressed the development of carrageenan-evoked thermal and mechanical hyperalgesia and allodynia. The AM1241-induced suppression of carrageenan-evoked behavioral sensitization was blocked by the CB(2) antagonist SR144528 but not by the CB(1) antagonist SR141716A. Intraplantar (ipl) administration of AM1241 (33 micrograms/kg ipl) suppressed hyperalgesia and allodynia following administration to the carrageenan-injected paw but was inactive following administration in the contralateral (noninflamed) paw, consistent with a local site of action. In immunocytochemical studies, AM1241 suppressed spinal Fos protein expression, a marker of neuronal activity, in the carrageenan model of inflammation. AM1241 suppressed carrageenan-evoked Fos protein expression in the superficial and neck region of the dorsal horn but not in the nucleus proprius or the ventral horn. The suppression of carrageenan-evoked Fos protein expression induced by AM1241 was blocked by coadministration of SR144528 in all spinal laminae. These data provide evidence that actions at cannabinoid CB(2) receptors are sufficient to suppress inflammation-evoked neuronal activity at rostral levels of processing in the spinal dorsal horn, consistent with the ability of AM1241 to normalize nociceptive thresholds and produce antinociception in inflammatory pain states.


Subject(s)
Analgesics/pharmacology , Cannabinoids/pharmacology , Inflammation/drug therapy , Nociceptors/drug effects , Pain/drug therapy , Posterior Horn Cells/drug effects , Receptor, Cannabinoid, CB2 , Receptors, Drug/agonists , Animals , Camphanes/pharmacology , Carrageenan/pharmacology , Disease Models, Animal , Drug Interactions/physiology , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Inflammation/metabolism , Inflammation/physiopathology , Male , Nociceptors/metabolism , Pain/metabolism , Pain/physiopathology , Pain Threshold/drug effects , Pain Threshold/physiology , Piperidines/pharmacology , Posterior Horn Cells/metabolism , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Reaction Time/physiology , Receptors, Cannabinoid , Receptors, Drug/antagonists & inhibitors , Receptors, Drug/metabolism , Rimonabant
11.
Neuroscience ; 117(3): 659-70, 2003.
Article in English | MEDLINE | ID: mdl-12617970

ABSTRACT

The present studies were conducted to test the hypothesis that systemically inactive doses of cannabinoids suppress inflammation-evoked neuronal activity in vivo via a peripheral mechanism. We examined peripheral cannabinoid modulation of spinal Fos protein expression, a marker of neuronal activity, in a rat model of inflammation. Rats received unilateral intraplantar injections of carrageenan (3%). In behavioral studies, carrageenan induced allodynia and mechanical hyperalgesia in response to stimulation with von Frey monofilaments. The cannabinoid agonist WIN55,212-2 (30 microg intraplantarly), administered concurrently with carrageenan, attenuated carrageenan-evoked allodynia and hyperalgesia relative to control conditions. In immunocytochemical studies, WIN55,212-2 suppressed the development of carrageenan-evoked Fos protein expression in the lumbar dorsal horn of the spinal cord relative to vehicle treatment. The same dose administered systemically or to the noninflamed contralateral paw failed to alter either carrageenan-evoked allodynia and hyperalgesia or carrageenan-evoked Fos protein expression, consistent with a peripheral site of action. The suppressive effects of WIN55,212-2 (30 microg intraplantarly) on carrageenan-evoked Fos protein expression and pain behavior were blocked by local administration of either the CB(2) antagonist SR144528 (30 microg intraplantarly) or the CB(1) antagonist SR141716A (100 microg intraplantarly). WIN55,212-3, the enantiomer of the active compound, also failed to suppress carrageenan-evoked Fos protein expression. These data provide direct evidence that a peripheral cannabinoid mechanism suppresses the development of inflammation-evoked neuronal activity at the level of the spinal dorsal horn and implicate a role for CB(2) and CB(1) in peripheral cannabinoid modulation of inflammatory nociception.


Subject(s)
Cannabinoids/pharmacology , Gene Expression Regulation , Pain/physiopathology , Proto-Oncogene Proteins c-fos/metabolism , Spinal Cord/drug effects , Analgesics/pharmacology , Animals , Behavior, Animal/drug effects , Benzoxazines , Camphanes/pharmacology , Cannabinoids/administration & dosage , Cannabinoids/antagonists & inhibitors , Carrageenan/pharmacology , Disease Models, Animal , Drug Administration Routes , Drug Interactions , Edema/chemically induced , Edema/prevention & control , Functional Laterality , Immunohistochemistry/methods , Inflammation/metabolism , Male , Mechanoreceptors/drug effects , Morpholines/pharmacology , Naphthalenes/pharmacology , Pain/drug therapy , Pain/metabolism , Pain Measurement/methods , Physical Stimulation , Piperidines/pharmacology , Proto-Oncogene Proteins c-fos/genetics , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Rimonabant , Spinal Cord/anatomy & histology , Spinal Cord/metabolism , Time Factors
12.
Science ; 289(5479): 628-31, 2000 Jul 28.
Article in English | MEDLINE | ID: mdl-10915627

ABSTRACT

Nociceptive neuronal circuits are formed during embryonic and postnatal times when painful stimuli are normally absent or limited. Today, medical procedures for neonates with health risks can involve tissue injury and pain for which the long-term effects are unknown. To investigate the impact of neonatal tissue injury and pain on development of nociceptive neuronal circuitry, we used an animal model of persistent hind paw peripheral inflammation. We found that, as adults, these animals exhibited spinal neuronal circuits with increased input and segmental changes in nociceptive primary afferent axons and altered responses to sensory stimulation.


Subject(s)
Neurons, Afferent/physiology , Pain , Posterior Horn Cells/physiology , Afferent Pathways , Animals , Animals, Newborn , Axons/physiology , Cell Count , Freund's Adjuvant , Ganglia, Spinal/cytology , Ganglia, Spinal/physiology , Hindlimb/innervation , Inflammation/physiopathology , Male , Neurons, Afferent/cytology , Pain Measurement , Pain Threshold , Posterior Horn Cells/cytology , Rats , Rats, Sprague-Dawley , Sciatic Nerve/cytology , Sciatic Nerve/physiology , Wheat Germ Agglutinin-Horseradish Peroxidase Conjugate
13.
Synapse ; 37(1): 71-80, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10842353

ABSTRACT

Double-label in situ hybridization was used to identify the phenotypes of striatal neurons that express mRNA for cannabinoid CB(1) receptors. Simultaneous detection of multiple mRNAs was performed by combining a (35)S-labeled ribonucleotide probe for CB(1) mRNA with digoxigenin-labeled riboprobes for striatal projection neurons (preprotachykinin A, prodynorphin, and preproenkephalin mRNAs) and interneurons (vesicular acetylcholine transporter (VAChT), choline acetyltransferase (ChAT), somatostatin, and glutamic acid decarboxylase (Mr 67,000; GAD67) mRNAs). To ascertain whether CB(1) mRNA was a marker for striatal efferents, digoxigenin-labeled probes for mRNA markers of both striatonigral (prodynorphin or preprotachykinin A mRNAs), and striatopallidal (proenkephalin mRNAs) projection neurons were combined with the (35)S-labeled probe for CB(1). A mediolateral gradient in CB(1) mRNA expression was observed at rostral and mid-striatal levels; in the same coronal sections the number of silver grains per cell ranged from below the threshold of detectability at the medial and ventral poles to saturation at the dorsolateral boundary bordered by the corpus callosum. At the caudal level examined, CB(1) mRNA was denser in the ventral sector relative to the dorsal sector. Virtually all neurons expressing mRNA markers for striatal projection neurons colocalized CB(1) mRNA. Combining a (35)S-labeled riboprobe for CB(1) with digoxigenin-labeled riboprobes for both preproenkephalin and prodynorphin confirmed localization of CB(1) mRNA to striatonigral and striatopallidal neurons expressing prodynorphin and preproenkephalin mRNAs, respectively. However, CB(1) mRNA-positive cells that failed to coexpress the other markers were also apparent. CB(1) mRNA was localized to putative GABAergic interneurons that express high levels of GAD67 mRNA. These interneurons enable functional interactions between the direct and indirect striatal output pathways. By contrast, aspiny interneurons that express preprosomatostatin mRNA and cholinergic interneurons that coexpress ChAT and VAChT mRNAs were CB(1) mRNA-negative. The present data provide direct evidence that cannabinoid receptors are synthesized in striatonigral neurons that contain dynorphin and substance P and striatopallidal neurons that contain enkephalin. By contrast, local circuit neurons in striatum that contain somatostatin or acetylcholine do not synthesize cannabinoid receptors. Published 2000 Wiley-Liss, Inc.


Subject(s)
Corpus Striatum/cytology , Interneurons/chemistry , Receptors, Drug/genetics , Animals , Arachidonic Acids/pharmacology , Calcium Channel Blockers/pharmacology , Choline O-Acetyltransferase/analysis , Choline O-Acetyltransferase/genetics , Endocannabinoids , Gene Expression/physiology , In Situ Hybridization , Interneurons/enzymology , Male , Polyunsaturated Alkamides , Protein Precursors/analysis , Protein Precursors/genetics , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, Cannabinoid , Receptors, Drug/analysis , Somatostatin/analysis , Somatostatin/genetics , Sulfur Radioisotopes
14.
Life Sci ; 65(6-7): 665-73, 1999.
Article in English | MEDLINE | ID: mdl-10462067

ABSTRACT

The discovery of cannabinoid receptors and their putative endogenous ligands raises questions as to the nature of the effects produced by cannabinoids on neural circuits that mediate pain and whether endogenous cannabinoids produced by the brain or in the periphery serve naturally to modulate pain. A sizable body of previous work showed that cannabinoid agonists suppress pain behavior in a variety of models of acute and chronic pain. However, at appropriate doses, cannabinoids also profoundly suppress motor behavior (see Sañudo-Peña et al., this volume), which complicates the interpretation of behavioral analgesia since a motor response is the endpoint of virtually all such studies. Studies conducted in this laboratory used biochemical and neurophysiological measures to determine whether cannabinoids suppress nociceptive neurotransmission. The results showed that cannabinoids suppress nociceptive neurotransmission at the level of the spinal cord and the thalamus. These effects are reversible, receptor mediated, selective for painful as opposed to nonpainful somatic stimuli, and track the behavioral analgesia both in time course and potency.


Subject(s)
Analgesia , Cannabinoids , Analgesics , Animals , Benzoxazines , Brain/metabolism , Cannabinoids/pharmacology , Humans , Morpholines/pharmacology , Naphthalenes/pharmacology , Nociceptors/drug effects , Nociceptors/physiology
15.
Neuroscience ; 92(4): 1171-5, 1999.
Article in English | MEDLINE | ID: mdl-10426476

ABSTRACT

Cannabinoids modulate nociceptive processing through central and peripheral mechanisms. The present study was conducted to evaluate axonal flow of cannabinoid receptors from the dorsal root ganglion to the periphery and to identify the putative involvement of CB1 and/or CB2 receptor subtypes. The sciatic nerve was tightly ligated to dam the flow of cannabinoid receptors to the periphery. The densities of cannabinoid receptors proximal and distal to one or two tightly constrictive ligatures was evaluated using in vitro receptor binding and high-resolution emulsion autoradiography. In both models, [3H]CP55,940 binding accumulated proximal as opposed to distal to the ligature. These data indicate that axonal transport of cannabinoid receptors to the periphery was occluded by tight constriction of the sciatic nerve. In situ hybridization histochemistry revealed that dorsal root ganglia cells synthesize CB1 but not CB2 receptor messenger RNA. By contrast, CB2 messenger RNA was highly expressed in sections of rat spleen that were processed together with the dorsal root ganglia, as previously described. These data demonstrate that neuronal cannabinoid CB1 receptors are synthesized in cells of the dorsal root ganglia and inserted on terminals in the periphery.


Subject(s)
Axonal Transport/physiology , Neurons, Afferent/physiology , Receptor, Cannabinoid, CB2 , Receptors, Drug/physiology , Animals , Autoradiography , Cyclohexanols/pharmacology , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , In Situ Hybridization , Male , Peripheral Nerves/cytology , Peripheral Nerves/physiology , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Cannabinoid , Receptors, Drug/biosynthesis , Sciatic Nerve/physiology
16.
Proc Natl Acad Sci U S A ; 96(10): 5780-5, 1999 May 11.
Article in English | MEDLINE | ID: mdl-10318961

ABSTRACT

Delta9-Tetrahydrocannabinol (Delta9-THC), the major psychoactive ingredient in preparations of Cannabis sativa (marijuana, hashish), elicits central nervous system (CNS) responses, including cognitive alterations and euphoria. These responses account for the abuse potential of cannabis, while other effects such as analgesia suggest potential medicinal applications. To study the role of the major known target of cannabinoids in the CNS, the CB1 cannabinoid receptor, we have produced a mouse strain with a disrupted CB1 gene. CB1 knockout mice appeared healthy and fertile, but they had a significantly increased mortality rate. They also displayed reduced locomotor activity, increased ring catalepsy, and hypoalgesia in hotplate and formalin tests. Delta9-THC-induced ring-catalepsy, hypomobility, and hypothermia were completely absent in CB1 mutant mice. In contrast, we still found Delta9-THC-induced analgesia in the tail-flick test and other behavioral (licking of the abdomen) and physiological (diarrhea) responses after Delta9-THC administration. Thus, most, but not all, CNS effects of Delta9-THC are mediated by the CB1 receptor.


Subject(s)
Behavior, Animal/drug effects , Dronabinol/pharmacology , Receptors, Drug/genetics , Animals , Body Temperature/drug effects , Brain/metabolism , Central Nervous System/drug effects , Cyclohexanols/metabolism , Dronabinol/analogs & derivatives , Gene Targeting/methods , Mice , Mice, Knockout , Molecular Structure , Motor Activity/drug effects , Receptors, Cannabinoid
17.
Neuroscience ; 90(3): 923-31, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10218792

ABSTRACT

In situ hybridization histochemistry was used to show the distribution of messenger RNA for central cannabinoid CB 1 receptors in dorsal root ganglia of the rat. CB1 messenger RNA was highly expressed in neuronal subpopulations of rat dorsal root ganglia. The phenotypes of neurons that express messenger RNA for CB1 were subsequently examined by combining a 35S-labeled ribonucleotide probe for CB1 messenger RNA with digoxigenin-labeled riboprobes for preprotachykinin A (substance P precursor), alpha-calcitonin gene-related peptide and preprosomatostatin (somatostatin precursor) messenger RNAs. Qualitative examination revealed expression of CBI messenger RNA predominantly in medium-and large-sized cells distributed throughout the dorsal root ganglia. The majority of neurons expressing substance P messenger RNA were CB1 messenger RNA negative and smaller in size than the CB1 messenger RNA-positive cells. Only 13% of substance P messenger RNA-positive cells expressed CB1 messenger RNA. A similar degree of co-localization was observed with alpha-calcitonin gene-related peptide: 10% of cells expressing messenger RNA for this neuropeptide were CB1 messenger RNA positive. Co-localization of CB1 and somatostatin messenger RNAs was observed in less than 0.5% of somatostatin messenger RNA-positive cells. The data suggest that subpopulations of neurons in rat dorsal root ganglia are capable of synthesizing cannabinoid receptors and inserting them on terminals in the superficial dorsal horn. These findings provide anatomical evidence for cannabinoid modulation of primary afferent transmission. Although an anatomical basis for cannabinoid-mediated suppression of release of neurogenic peptides from nociceptive primary afferents is provided, our results demonstrate that the majority of CB messenger RNA-positive neurons in the dorsal root ganglia contain transmitters and/or neuromodulators other than the neuropeptides examined herein.


Subject(s)
Ganglia, Spinal/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , Receptors, Drug/genetics , Animals , Calcitonin Gene-Related Peptide/genetics , Histocytochemistry , In Situ Hybridization , Male , Neurons/cytology , Neurons/physiology , Phenotype , Protein Precursors/genetics , Rats , Rats, Sprague-Dawley , Receptors, Cannabinoid , Somatostatin/genetics , Tachykinins/genetics , Tissue Distribution/physiology
18.
Brain Res ; 822(1-2): 17-25, 1999 Mar 20.
Article in English | MEDLINE | ID: mdl-10082879

ABSTRACT

In vitro receptor binding and quantitative autoradiography were used to assess the pre- and postsynaptic distribution of cannabinoid receptors in the cervical dorsal horn of the rat spinal cord. An extensive unilateral dorsal rhizotomy was performed across seven or eight successive spinal segments from C3 to T1 or T2. The densities of cannabinoid and mu opioid receptors in the central (C6) spinal segment were assessed 2, 4, 8, and 16 days post rhizotomy and compared with those of untreated rats. Rhizotomy induced approximately a 50% ipsilateral loss in the [3H]CP55,940 binding to spinal cannabinoid receptors that was maximal at 8 days post-rhizotomy. By comparison, the binding of [3H][d-Ala2-MePhe4, Gly-ol5]enkephalin (DAMGO) to mu receptors was depleted approximately 60% in near-adjacent sections. By contrast, changes in [3H]CP55,940 binding contralateral to the deafferentation were largely absent at all post-lesion delays. These data suggest that under conditions in which a spinal segment is completely deafferented, approximately 50% of cannabinoid receptors in the cervical (C6) dorsal horn reside presynaptically on central terminals of primary afferents. The present data provide anatomical evidence for presynaptic as well as postsynaptic localization of cannabinoid receptors in the spinal dorsal horn.


Subject(s)
Receptors, Drug/analysis , Receptors, Opioid, mu/analysis , Spinal Cord/chemistry , Analgesics/pharmacology , Analgesics, Opioid/pharmacology , Animals , Arachidonic Acids/pharmacology , Autoradiography , Calcium Channel Blockers/pharmacology , Cyclohexanols/pharmacology , Endocannabinoids , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Enkephalins/pharmacology , Functional Laterality , Male , Neurons, Afferent/chemistry , Neurons, Afferent/drug effects , Polyunsaturated Alkamides , Presynaptic Terminals/chemistry , Rats , Rats, Sprague-Dawley , Receptors, Cannabinoid , Receptors, Opioid, mu/agonists , Rhizotomy , Spinal Cord/cytology , Tritium
19.
J Neurophysiol ; 81(2): 575-83, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10036261

ABSTRACT

The effects of cannabinoid agonists on noxious heat-evoked firing of 62 spinal wide dynamic range (WDR) neurons were examined in urethan-anesthetized rats (1 cell/animal). Noxious thermal stimulation was applied with a Peltier device to the receptive fields in the ipsilateral hindpaw of isolated WDR neurons. To assess the site of action, cannabinoids were administered systemically in intact and spinally transected rats and intraventricularly. Both the aminoalkylindole cannabinoid WIN55,212-2 (125 microg/kg iv) and the bicyclic cannabinoid CP55,940 (125 microg/kg iv) suppressed noxious heat-evoked activity. Responses evoked by mild pressure in nonnociceptive neurons were not altered by CP55,940 (125 microg/kg iv), consistent with previous observations with another cannabinoid agonist, WIN55,212-2. The cannabinoid induced-suppression of noxious heat-evoked activity was blocked by pretreatment with SR141716A (1 mg/kg iv), a competitive antagonist for central cannabinoid CB1 receptors. By contrast, intravenous administration of either vehicle or the receptor-inactive enantiomer WIN55,212-3 (125 microg/kg) failed to alter noxious heat-evoked activity. The suppression of noxious heat-evoked activity induced by WIN55,212-2 in the lumbar dorsal horn of intact animals was markedly attenuated in spinal rats. Moreover, intraventricular administration of WIN55,212-2 suppressed noxious heat-evoked activity in spinal WDR neurons. By contrast, both vehicle and enantiomer were inactive. These findings suggest that cannabinoids selectively modulate the activity of nociceptive neurons in the spinal dorsal horn by actions at CB1 receptors. This modulation represents a suppression of pain neurotransmission because the inhibitory effects are selective for pain-sensitive neurons and are observed with different modalities of noxious stimulation. The data also provide converging lines of evidence for a role for descending antinociceptive mechanisms in cannabinoid modulation of spinal nociceptive processing.


Subject(s)
Cannabinoids/pharmacology , Hot Temperature , Neurons/drug effects , Neurons/physiology , Spinal Cord/drug effects , Analgesics/administration & dosage , Analgesics/pharmacology , Animals , Axotomy , Benzoxazines , Cannabinoids/administration & dosage , Cannabinoids/antagonists & inhibitors , Cyclohexanols/administration & dosage , Cyclohexanols/pharmacology , Injections, Intravenous , Injections, Intraventricular , Lumbosacral Region , Male , Mechanoreceptors/drug effects , Microinjections , Morpholines/administration & dosage , Morpholines/pharmacology , Naphthalenes/administration & dosage , Naphthalenes/pharmacology , Pain Measurement/drug effects , Piperidines/administration & dosage , Piperidines/pharmacology , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Rimonabant , Spinal Cord/physiology
20.
Zhongguo Yao Li Xue Bao ; 20(12): 1132-6, 1999 Dec.
Article in English | MEDLINE | ID: mdl-11216448

ABSTRACT

AIM: To determine whether cannabinoids suppress noxious stimulus-evoked Fos protein-like immunoreactivity (FLI) through direct actions at the spinal level. METHODS: Rats were implanted with intrathecal (ith) catheters at least one week prior to evaluation in the formalin test. Effects of the cannabinoid agonist, CP55,940 (80 micrograms ith) on formalin pain and FLI in rat spinal cord were compared with that of the prototypic narcotic analgesic, morphine (20 micrograms ith). CP55,940 suppressed pain behavior and FLI induced by intraplantar formalin. The cannabinoid suppressed Fos in the neck region of the dorsal horn and in the ventral horn, but not in the nucleus proprius. The efficacy of the cannabinoid in suppressing FLI in these laminae and pain behavior was comparable to morphine administered via the same route. However, only morphine suppressed FLI in the superficial dorsal horn relative to vehicle treatment. CONCLUSION: Cannabinoids suppress nociceptive processing, in part, through actions at the spinal level. However, morphine showed greater potency and efficacy than CP55,940 in suppressing formalin-induced FLI following spinal administration.


Subject(s)
Cannabinoids/agonists , Cyclohexanols/pharmacology , Nociceptors/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Spinal Cord/metabolism , Analgesics/pharmacology , Animals , Immunosuppressive Agents/pharmacology , Injections, Spinal , Male , Morphine/pharmacology , Pain/drug therapy , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...