Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Neuroscience ; 111(2): 389-97, 2002.
Article in English | MEDLINE | ID: mdl-11983324

ABSTRACT

Mechanisms underlying neuropathic pain states are poorly understood. We have compared mechanisms mediating enhanced nociception of four established models of neuropathic pain produced by very different types of insults to the peripheral nervous system: streptozotocin-induced hyperalgesia, a model of diabetic (metabolic) peripheral neuropathy, vincristine-induced hyperalgesia, a model of chemotherapeutic agent (toxic) peripheral neuropathy, and chronic constriction injury and partial nerve ligation, models of trauma-induced painful neuropathies. All four models resulted in prolonged mechanical hyperalgesia (>30% decrease in mechanical nociceptive threshold) and allodynia (detected by 10-209-mN-intensity von Frey hairs). In vincristine- and streptozotocin-induced hyperalgesia, the protein kinase A, protein kinase C and nitric oxide second messenger pathways in the periphery contributed to the hyperalgesia, while N-methyl-D-aspartate (NMDA) receptor-mediated events were not detected. None of these second messengers nor the NMDA receptor, which can contribute to peripheral sensitization of nociceptors, contributed to chronic constriction injury- and partial nerve ligation-induced hyperalgesia. In all four models the hyperalgesia was not antagonized by peripheral administration of a mu-opioid agonist.Our findings support the presence of a common abnormality in second messenger signaling in the periphery to the maintenance of two very different models of non-traumatic neuropathic pain, not shared by models of trauma-induced neuropathic pain.


Subject(s)
Nociceptors/physiology , Peripheral Nerve Injuries , Peripheral Nervous System Diseases/physiopathology , Wounds and Injuries/physiopathology , Animals , Hyperalgesia/chemically induced , Hyperalgesia/etiology , Hyperesthesia/chemically induced , Hyperesthesia/etiology , Male , Pain/physiopathology , Pain Threshold , Peripheral Nervous System Diseases/complications , Rats , Rats, Sprague-Dawley , Second Messenger Systems/physiology , Streptozocin , Vincristine , Wounds and Injuries/complications
2.
J Neurosci ; 21(17): 6933-9, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11517280

ABSTRACT

Inflammatory pain, characterized by a decrease in mechanical nociceptive threshold (hyperalgesia), arises through actions of inflammatory mediators, many of which sensitize primary afferent nociceptors via G-protein-coupled receptors. Two signaling pathways, one involving protein kinase A (PKA) and one involving the epsilon isozyme of protein kinase C (PKCepsilon), have been implicated in primary afferent nociceptor sensitization. Here we describe a third, independent pathway that involves activation of extracellular signal-regulated kinases (ERKs) 1 and 2. Epinephrine, which induces hyperalgesia by direct action at beta(2)-adrenergic receptors on primary afferent nociceptors, stimulated phosphorylation of ERK1/2 in cultured rat dorsal root ganglion cells. This was inhibited by a beta(2)-adrenergic receptor blocker and by an inhibitor of mitogen and extracellular signal-regulated kinase kinase (MEK), which phosphorylates and activates ERK1/2. Inhibitors of G(i/o)-proteins, Ras farnesyltransferases, and MEK decreased epinephrine-induced hyper-algesia. In a similar manner, phosphorylation of ERK1/2 was also decreased by these inhibitors. Local injection of dominant active MEK produced hyperalgesia that was unaffected by PKA or PKCepsilon inhibitors. Conversely, hyperalgesia produced by agents that activate PKA or PKCepsilon was unaffected by MEK inhibitors. We conclude that a Ras-MEK-ERK1/2 cascade acts independent of PKA or PKCepsilon as a novel signaling pathway for the production of inflammatory pain. This pathway may present a target for a new class of analgesic agents.


Subject(s)
Hyperalgesia/physiopathology , Mitogen-Activated Protein Kinases/metabolism , Nociceptors/physiopathology , Adrenergic beta-2 Receptor Antagonists , Animals , Cells, Cultured , Crosses, Genetic , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiopathology , Heterotrimeric GTP-Binding Proteins/antagonists & inhibitors , Heterotrimeric GTP-Binding Proteins/metabolism , Hyperalgesia/chemically induced , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Male , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Nociceptors/drug effects , Pain Measurement , Phosphorylation/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase C-epsilon , Rats , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/drug effects , ras Proteins/metabolism
3.
Eur J Neurosci ; 13(12): 2227-33, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11454025

ABSTRACT

We have evaluated the contribution of differences in second messenger signalling to sex differences in inflammatory pain and its control by sex hormones. In normal male but not female rats, epinephrine-induced mechanical hyperalgesia was antagonized by inhibitors of protein kinase Cepsilon (PKCepsilon), protein kinase A (PKA) and nitric oxide synthetase (NOS). Similarly, in PKCepsilon knockout mice, a contribution of PKCepsilon to epinephrine-dependent mechanical hyperalgesia occurred in males only. In contrast, hyperalgesia induced by prostaglandin E2, in both females and males, was dependent on PKA and NO. In both sexes, inhibitors of mitogen-activated protein kinase/extracellular-signal related kinase kinase (MEK) inhibited epinephrine hyperalgesia. In gonadectomized females, the second messenger contributions to epinephrine hyperalgesia demonstrated the pattern seen in males. Administration of oestrogen to gonadectomized females fully reconstituted the phenotype of the normal female. These data demonstrate gender differences in PKCepsilon, PKA and NO signalling in epinephrine-induced hyperalgesia which are oestrogen dependent and appear to be exerted at the level of the beta-adrenergic receptor or the G-protein to which it is coupled.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Gonadal Steroid Hormones/metabolism , Isoenzymes/metabolism , Nitric Oxide Synthase/metabolism , Pain/metabolism , Protein Kinase C/metabolism , Signal Transduction/physiology , Adrenergic Agonists/pharmacology , Animals , Central Nervous System/drug effects , Central Nervous System/metabolism , Central Nervous System/physiopathology , Cyclic AMP-Dependent Protein Kinases/drug effects , Dinoprostone/pharmacology , Enzyme Inhibitors/pharmacology , Estradiol/pharmacology , Female , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/physiopathology , Intercellular Signaling Peptides and Proteins , Isoenzymes/drug effects , Isoenzymes/genetics , MAP Kinase Kinase 1 , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Neurons/drug effects , Neurons/metabolism , Nitric Oxide Synthase/drug effects , Pain/chemically induced , Pain/physiopathology , Peptides/pharmacology , Protein Kinase C/drug effects , Protein Kinase C/genetics , Protein Kinase C-epsilon , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Sex Characteristics , Signal Transduction/drug effects , omega-N-Methylarginine/pharmacology
4.
J Pain ; 2(3): 146-50, 2001 Jun.
Article in English | MEDLINE | ID: mdl-14622824

ABSTRACT

Pain in diabetes is a common debilitating condition for which pathophysiology remains poorly understood. To evaluate the underlying mechanisms, we used intravenous injection of streptozotocin to produce rapid (24-hour) onset of diabetes (blood glucose > 300 mg/dL and urine glucose > 2,000 mg/dL with polyuria). In this model, mechanical and thermal hyperalgesia and tactile allodynia are detectable by 48 hours after streptozotocin administration in the absence of ketonuria or physical debility. Treatment with insulin attenuated hyperglycemia and prevented the development of mechanical and thermal hyperalgesia. Direct application of streptozotocin to peripheral nerve did not produce hyperalgesia. We conclude that streptozotocin can induce pain independent of a general debility or direct toxic effect of streptozotocin on peripheral nerve and that elevated blood glucose may contribute to the enhanced nociception.

5.
J Neurosci ; 20(12): 4680-5, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10844037

ABSTRACT

We have identified a mechanism, mediated by the epsilon isozyme of protein kinase C (PKCepsilon) in peripheral neurons, which may have a role in chronic inflammatory pain. Acute inflammation, produced by carrageenan injection in the rat hindpaw, produced mechanical hyperalgesia that resolved by 72 hr. However, for up to 3 weeks after carrageenan, injection of the inflammatory mediators prostaglandin E(2) or 5-hydroxytryptamine or of an adenosine A(2) agonist into the same site induced a markedly prolonged hyperalgesia (>24 hr compared with 5 hr or less in control rats not pretreated with carrageenan). A nonselective inhibitor of several PKC isozymes and a selective PKCepsilon inhibitor antagonized this prolonged hyperalgesic response equally. Acute carrageenan hyperalgesia could be inhibited by PKA or PKG antagonists. However, these antagonists did not inhibit development of the hypersensitivity to inflammatory mediators. Our findings indicate that different second messenger pathways underlie acute and prolonged inflammatory pain.


Subject(s)
Hyperalgesia/physiopathology , Isoenzymes/metabolism , Nociceptors/physiology , Pain/physiopathology , Protein Kinase C/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Carrageenan , Dinoprostone/pharmacology , Enzyme Inhibitors/pharmacology , Hindlimb , Inflammation , Male , Phenethylamines/pharmacology , Protein Kinase C-epsilon , Protein Kinase Inhibitors , Purinergic P1 Receptor Agonists , Rats , Rats, Sprague-Dawley , Serotonin/pharmacology , Time Factors
6.
J Neurosci ; 19(16): 7175-81, 1999 Aug 15.
Article in English | MEDLINE | ID: mdl-10436070

ABSTRACT

We tested the hypothesis that noxious stimuli induce pain modulation by activation of supraspinal structures. We found that intense noxious stimuli (i.e., subdermal injection of capsaicin or paw immersion in hot water) induced profound attenuation of the jaw-opening reflex in the anesthetized rat; forepaw subdermal capsaicin also elevated the mechanical hindpaw-withdrawal threshold in the awake rat. These antinociceptive effects were blocked by previous injection of either a dopamine antagonist (flupentixol) or an opioid antagonist (naloxone) into the nucleus accumbens. Additional experiments in anesthetized animals showed that the antinociceptive effect of noxious stimulation by either capsaicin (>/=100 micrograms) or hindpaw immersion in hot water (>/=45 degrees C for 4 min) correlated with the intensity of the stimulus. The maximal antinociceptive effect of capsaicin was similar in magnitude to that of a high dose of morphine (10 mg/kg) injected subcutaneously. Injection of the GABA(A)-receptor agonist muscimol, but not naloxone, into the rostroventral medulla, a major component of descending pain modulation systems, blocked capsaicin-induced antinociception. Although it is widely thought that painful stimuli may induce analgesia by activating forebrain structures, this is the first demonstration that such a mechanism exists. Furthermore, this mechanism can be engaged by naturalistic stimuli in awake animals. These observations imply that painful stimuli might under certain conditions be rewarding.


Subject(s)
Analgesia/methods , Limbic System/physiology , Medulla Oblongata/physiology , Pain/physiopathology , Reward , Animals , Electric Stimulation , Male , Neural Pathways/physiology , Rats , Rats, Sprague-Dawley
7.
J Neurosci ; 19(6): 2181-6, 1999 Mar 15.
Article in English | MEDLINE | ID: mdl-10066271

ABSTRACT

Although the initiation of inflammatory pain (hyperalgesia) has been demonstrated to require the cAMP second messenger signaling cascade, whether this mechanism and/or other mechanisms underlie the continued maintenance of the induced hyperalgesia is unknown. We report that injection of adenylyl cyclase inhibitors before but not after injection of direct-acting hyperalgesic agents (prostaglandin E2 and purine and serotonin receptor agonists) resulted in reduction in hyperalgesia, evaluated by the Randall-Selitto paw-withdrawal test. In contrast, injection of protein kinase A (PKA) inhibitors either before or after these hyperalgesic agents resulted in reduced hyperalgesia, suggesting that hyperalgesia after its activation was maintained by persistent PKA activity but not by adenylyl cyclase activity. To evaluate further the role of PKA activity in the maintenance of hyperalgesia, we injected the catalytic subunit of PKA (PKACS) that resulted in hyperalgesia similar in magnitude to that induced by the direct-acting hyperalgesic agents but much longer in duration (>48 vs 2 hr). Injection of WIPTIDE (a PKA inhibitor) at 24 hr after PKACS reduced hyperalgesia, suggesting that PKACS hyperalgesia is not independently maintained by steps downstream from PKA. In summary, our results indicate that, once established, inflammatory mediator-induced hyperalgesia is no longer maintained by adenylyl cyclase activity but rather is dependent on ongoing PKA activity. An understanding of the mechanism maintaining hyperalgesia may provide important insight into targets for the treatment of persistent pain.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Hyperalgesia/physiopathology , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenylyl Cyclase Inhibitors , Animals , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Dinoprostone/antagonists & inhibitors , Dinoprostone/pharmacology , Drug Administration Schedule , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Hindlimb/drug effects , Hyperalgesia/chemically induced , Injections, Intradermal , Male , Nociceptors/drug effects , Pain Threshold/drug effects , Phenethylamines/pharmacology , Rats , Rats, Sprague-Dawley , Serotonin Receptor Agonists/pharmacology
8.
Neuron ; 24(1): 253-60, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10677042

ABSTRACT

There is great interest in discovering new targets for pain therapy since current methods of analgesia are often only partially successful. Although protein kinase C (PKC) enhances nociceptor function, it is not known which PKC isozymes contribute. Here, we show that epinephrine-induced mechanical and thermal hyperalgesia and acetic acid-associated hyperalgesia are markedly attenuated in PKCepsilon mutant mice, but baseline nociceptive thresholds are normal. Moreover, epinephrine-, carrageenan-, and nerve growth factor- (NGF-) induced hyperalgesia in normal rats, and epinephrine-induced enhancement of tetrodotoxin-resistant Na+ current (TTX-R I(Na)) in cultured rat dorsal root ganglion (DRG) neurons, are inhibited by a PKCepsilon-selective inhibitor peptide. Our findings indicate that PKCepsilon regulates nociceptor function and suggest that PKCepsilon inhibitors could prove useful in the treatment of pain.


Subject(s)
Isoenzymes/genetics , Isoenzymes/metabolism , Mutation , Nociceptors/physiology , Protein Kinase C/genetics , Protein Kinase C/metabolism , Signal Transduction , Acetic Acid , Analgesia , Analgesics , Animals , Carrageenan , Enzyme Inhibitors , Epinephrine , Hot Temperature , Hyperalgesia/etiology , Hyperalgesia/genetics , Mice , Nerve Growth Factor , Rats , Sodium Channels/drug effects , Sodium Channels/physiology , Tetrodotoxin/pharmacology
9.
J Neurosci ; 18(17): 7008-14, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9712669

ABSTRACT

We investigated the role of nitric oxide (NO) in inflammatory hyperalgesia. Coinjection of prostaglandin E2 (PGE2) with the nitric oxide synthase (NOS) inhibitor NG-methyl-L-arginine (L-NMA) inhibited PGE2-induced hyperalgesia. L-NMA was also able to reverse that hyperalgesia. This suggests that NO contributes to the maintenance of, as well as to the induction of, PGE2-induced hyperalgesia. Consistent with the hypothesis that the NO that contributes to PGE2-induced sensitization of primary afferents is generated in the dorsal root ganglion (DRG) neurons themselves, L-NMA also inhibited the PGE2-induced increase in tetrodotoxin-resistant sodium current in patch-clamp electrophysiological studies of small diameter DRG neurons in vitro. Although NO, the product of NOS, often activates guanylyl cyclase, we found that PGE2-induced hyperalgesia was not inhibited by coinjection of 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ), a guanylyl cyclase inhibitor. We then tested whether the effect of NO depended on interaction with the adenylyl cyclase-protein kinase A (PKA) pathway, which is known to mediate PGE2-induced hyperalgesia. L-NMA inhibited hyperalgesia produced by 8-bromo-cAMP (a stable membrane permeable analog of cAMP) or by forskolin (an adenylyl cyclase activator). However, L-NMA did not inhibit hyperalgesia produced by injection of the catalytic subunit of PKA. Therefore, the contribution of NO to PGE2-induced hyperalgesia may occur in the cAMP second messenger pathway at a point before the action of PKA. We next performed experiments to test whether administration of exogenous NO precursor or donor could mimic the hyperalgesic effect of endogenous NO. Intradermal injection of either the NOS substrate L-arginine or the NO donor 3-(4-morphinolinyl)-sydnonimine hydrochloride (SIN-1) produced hyperalgesia. However, this hyperalgesia differed from PGE2-induced hyperalgesia, because it was independent of the cAMP second messenger system and blocked by the guanylyl cyclase inhibitor ODQ. Therefore, although exogenous NO induces hyperalgesia, it acts by a mechanism different from that by which endogenous NO facilitates PGE2-induced hyperalgesia. Consistent with the hypothesis that these mechanisms are distinct, we found that inhibition of PGE2-induced hyperalgesia caused by L-NMA could be reversed by a low dose of the NO donor SIN-1. The following facts suggest that this dose of SIN-1 mimics a permissive effect of basal levels of NO with regard to PGE2-induced hyperalgesia: (1) this dose of SIN-1 does not produce hyperalgesia when administered alone, and (2) the effect was not blocked by ODQ. In conclusion, we have shown that low levels of NO facilitate cAMP-dependent PGE2-induced hyperalgesia, whereas higher levels of NO produce a cGMP-dependent hyperalgesia.


Subject(s)
Dinoprostone/physiology , Hyperalgesia/physiopathology , Nitric Oxide/physiology , Nociceptors/physiology , Pain/physiopathology , Signal Transduction/physiology , Animals , Dinoprostone/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Male , Nitric Oxide Synthase/antagonists & inhibitors , Oxadiazoles/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Serotonin Receptor Agonists/pharmacology , Tetrodotoxin/pharmacology , omega-N-Methylarginine/pharmacology
10.
J Neurosci ; 17(20): 8018-23, 1997 Oct 15.
Article in English | MEDLINE | ID: mdl-9315920

ABSTRACT

The mu-opioid [D-Ala2,N-Me-Phe4,Gly-ol5]-enkephalin (DAMGO) exerts a peripheral antinociceptive effect against prostaglandin E2 (PGE2)-induced mechanical hyperalgesia in the hindpaw of the rat. Tolerance and dependence develop to this effect. We have shown previously that tolerance and dependence can be dissociated and are mediated by different second messenger systems. In the present study, we evaluated whether the same or different second messenger systems mediate the development of this peripheral opioid tolerance or dependence compared with the expression of the loss of antinociceptive effect or rebound opioid antagonist hyperalgesia (i. e., expression of tolerance and dependence). DAMGO-induced tolerance was prevented by pretreatment with the nitric oxide synthase inhibitor NG-methyl-L-arginine (NMLA) but not by the protein kinase C (PKC) inhibitor chelerythrine, the adenylyl cyclase inhibitor 2',5'-dideoxyadenosine (ddA), or the calcium chelators 3,4,5-trimethoxybenzoic acid 8-(diethylamino)-octyl ester (TMB-8) and 2-[(2-bis-[carboxymethyl]amino-5-methylphenoxy)-methyl]-6-methoxy-8-bis [carboxymethyl]aminoquinoline (Quin-2). Once established, however, expression of DAMGO tolerance was acutely reversed by TMB-8 or Quin-2 but not by chelerythrine or NMLA. In contrast, naloxone-precipitated hyperalgesia in DAMGO-tolerant paws, a measure of dependence, was blocked by pretreatment with chelerythrine but not by NMLA, ddA, TMB-8, or Quin-2. Naloxone-precipitated hyperalgesia in DAMGO-tolerant paws was acutely reversed by chelerythrine, ddA, TMB-8, or Quin-2 but not by NMLA. Taken together, these results provide the first evidence that different mechanisms mediate the development and expression of both tolerance and dependence to the peripheral antinociceptive effect of DAMGO. However, although the development of tolerance and dependence are entirely separable, the expression of tolerance and dependence shares common calcium-dependent mechanisms.


Subject(s)
Analgesics/pharmacology , Enkephalins/pharmacology , Nociceptors/drug effects , Nociceptors/physiology , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/physiology , Animals , Dinoprostone , Drug Tolerance/physiology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Male , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/agonists , Second Messenger Systems/physiology , Substance-Related Disorders/physiopathology
11.
J Neurosci ; 17(10): 3907-12, 1997 May 15.
Article in English | MEDLINE | ID: mdl-9133408

ABSTRACT

Repeated peripheral administration of the micro-opioid agonist [D-Ala2,N-Me-Phe4,gly5-ol] enkephalin (DAMGO) produces acute tolerance and dependence on its peripheral antinociceptive effect against prostaglandin E2 (PGE2)-induced mechanical hyperalgesia. In this study we evaluated the roles of protein kinase C (PKC) and nitric oxide (NO) in the development of this tolerance and dependence. Repeated administration of PKC inhibitors chelerythrine and 1-(5-isoquinolinesulfonyl)-2-methylpiperazine dihydrochloride with DAMGO did not alter the tolerance to DAMGO; however, dependence (defined as naloxone-induced withdrawal hyperalgesia) was blocked. Repeated administration of N-(n-heptyl)-5-chloro-1-naphthalenesulfonamide, a PKC activator, which alone did not produce tolerance, mimicked the dependence produced by DAMGO. Repeated administration of the NO synthase inhibitor NG-methyl-L-arginine with DAMGO blocked the development of tolerance to DAMGO but had no effect on the development of dependence. Repeated administration of L-arginine, a NO precursor, mimicked tolerance produced by repeated administration of DAMGO (i.e. , the antinociceptive effect of DAMGO was lost); however, L-arginine did not mimic dependence. These findings suggest that the development of acute tolerance and dependence on the peripheral antinociceptive effects of DAMGO have different, dissociable mechanisms. Specifically, PKC is involved in development of mu-opioid dependence, whereas the NO signaling system is involved in the development of mu-opioid tolerance.


Subject(s)
Analgesics/pharmacology , Enkephalins/pharmacology , Nociceptors/physiology , Receptors, Opioid, mu/antagonists & inhibitors , Substance-Related Disorders , Animals , Dinoprostone/pharmacology , Drug Tolerance , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Enzyme Inhibitors/pharmacology , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Male , NG-Nitroarginine Methyl Ester/pharmacology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Neurons/chemistry , Neurons/drug effects , Neurons/enzymology , Nitric Oxide/metabolism , Oxytocics/pharmacology , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Second Messenger Systems/physiology
12.
J Neurosci ; 17(2): 735-44, 1997 Jan 15.
Article in English | MEDLINE | ID: mdl-8987795

ABSTRACT

We examined the interactions among three classes of peripherally-acting antinociceptive agents (mu-opioid, alpha 2-adrenergic, and A1-adenosine) in the development of tolerance and dependence to their antinociceptive effects. Antinociception was determined by assessing the degree of inhibition of prostaglandin E2 (PGE2)-induced mechanical hyperalgesia, using the Randall-Selitto paw-withdrawal test. Tolerance developed within 4 hr to the antinociceptive effect of the alpha 2-adrenergic agonist clonidine; dependence also occurred at that time, demonstrated as a withdrawal hyperalgesia that was precipitated by the alpha 2-receptor antagonist yohimbine. These findings are similar to those reported previously for tolerance and dependence to mu and A1 peripheral antinociception (Aley et al., 1995). Furthermore, cross-tolerance and cross-withdrawal between mu, A1, and alpha 2 agonists occurred. The observations of cross-tolerance and cross-withdrawal suggest that all three receptors are located on the same primary afferent nociceptors. In addition, the observations suggest that the mechanisms of tolerance and dependence to the antinociceptive effects of mu, A1, and alpha 2 are mediated by a common mechanism. Although any of the agonists administered alone produce antinociception, we found that mu, A1, and alpha 2 receptors may not act independently to produce antinociception, but rather may require the physical presence of the other receptors to produce antinociception by any one agonist. This was suggested by the finding that clonidine (alpha 2-agonist) antinociception was blocked not only by yohimbine (alpha 2-antagonist) but also by PACPX (A1-antagonist) and by naloxone (mu-antagonist), and that DAMGO (mu-agonist) antinociception and CPA (A1-agonist) antinociception were blocked not only by naloxone (mu-antagonist) and PACPX (A1-antagonist), respectively, but also by yohimbine (alpha 2-antagonist). This cross-antagonism of antinociception occurred at the ID50 dose for each antagonist at its homologous receptor. To test the hypothesis that the physical presence of mu-opioid receptor is required not only for mu antinociception but also for alpha 2 antinociception, antisense oligodeoxynucleotides (ODNs) for the mu-opioid and alpha 2C-adrenergic receptors were administered intrathecally to reduce the expression of these receptors on primary afferent neurons. These studies demonstrated that mu-opioid ODN administration decreased not only mu-opioid but also alpha 2-adrenergic antinociception; A1 antinociception was unaffected. In contrast, alpha 2C-adrenergic ODN decreased antinociception induced by all three classes of antinociceptive agents. In conclusion, these data suggest that peripheral antinociception induced by mu, alpha 2, and A1 agonists requires the physical presence of multiple receptors. We propose that there is a mu, A1, alpha 2 receptor complex mediating antinociception in the periphery. In addition, there is cross-tolerance and cross-dependence between mu, A1, and alpha 2 antinociception, suggesting that their underlying mechanisms are related.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Analgesics/pharmacology , Clonidine/pharmacology , Dinoprostone/physiology , Hyperalgesia/physiopathology , Receptors, Adrenergic, alpha-2/physiology , Receptors, Opioid, mu/physiology , Receptors, Purinergic P1/physiology , Substance Withdrawal Syndrome/physiopathology , Substance-Related Disorders/physiopathology , Yohimbine/pharmacology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adrenergic alpha-2 Receptor Agonists , Adrenergic alpha-Agonists/therapeutic use , Adrenergic alpha-Antagonists/therapeutic use , Adrenergic alpha-Antagonists/toxicity , Analgesics/therapeutic use , Analgesics, Opioid/pharmacology , Animals , Clonidine/therapeutic use , Dinoprostone/pharmacology , Drug Interactions , Drug Tolerance , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Enkephalins/pharmacology , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Male , Models, Biological , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Oligonucleotides, Antisense/pharmacology , Purinergic P1 Receptor Agonists , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/genetics , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/genetics , Second Messenger Systems , Substance Withdrawal Syndrome/etiology , Xanthines/pharmacology , Yohimbine/therapeutic use , Yohimbine/toxicity
13.
Neuroscience ; 73(1): 259-65, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8783247

ABSTRACT

To investigate the mechanism of vincristine-induced pain in humans undergoing chemotherapy we have established a model of vincristine-induced hyperalgesia in rat. Vincristine (100 micrograms/kg) was administered daily over a period of two weeks. An acute dose-dependent decrease in mechanical nociceptive threshold and an increased response to non-noxious mechanical stimuli ("hyperalgesia") occurred after the second day of administration. Chronic lowered threshold and increased response to stimuli (determined 24 h after each injection) was first noted during the second week of vincristine administration. Responses gradually returned to baseline during the two weeks following discontinuation of treatment. Vincristine also increased sensitivity to heat stimulation. At a dose that produced hyperalgesia (100 micrograms/kg), vincristine did not cause a significant motor deficit. Peripheral administration of a mu-opioid agonist did not reduce vincristine-induced acute hyperalgesia. Hyperalgesia induced by vincristine in the rat provides a good model for the experimental study of painful peripheral neuropathies in human patients receiving vincristine as a chemotherapeutic agent.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Hyperalgesia/chemically induced , Peripheral Nervous System Diseases/chemically induced , Vincristine/toxicity , Analgesics/pharmacology , Animals , Antineoplastic Agents, Phytogenic/adverse effects , Disease Models, Animal , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Enkephalins/pharmacology , Hot Temperature , Hyperalgesia/physiopathology , Injections, Intravenous , Male , Neurons, Afferent/drug effects , Pain Threshold/drug effects , Peripheral Nervous System Diseases/physiopathology , Physical Stimulation , Psychomotor Performance/drug effects , Rats , Rats, Sprague-Dawley , Vincristine/adverse effects
14.
Neuroscience ; 71(4): 1083-90, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8684612

ABSTRACT

We report on the ability of a delayed sympathectomy after a prolonged hyperalgesia to result in a subsequent enhanced hyperalgesic response. Sympathectomy was performed one day after injection of prostaglandin E2 plus rolipram, which induces a prolonged sympathetically-maintained hyperalgesia [Aley K. O. and Levine J. D. (1995) Eur. J. Pharmac. 273, 107-112]. The duration of hyperalgesia produced by a subsequent injection of prostaglandin E2 or prostaglandin E2 plus rolipram was then assessed. Lumbar surgical sympathectomy, done on day 2 or 3, prevented prostaglandin E2 plus rolipram-induced prolonged hyperalgesia from developing when they were injected five days after surgery, similar to the previous report of the effect of prior sympathectomy to block the rolipram enhancement [Aley K. O. and Levine J. D. (1995) Eur. J. Pharmac. 273, 107-112]. Sympathectomy done five days after injection of prostaglandin E2 plus rolipram, however, paradoxically prolonged (at least 10 days) hyperalgesia induced by a subsequent prostaglandin E2 plus rolipram injection, a duration much greater than that seen after prostaglandin E2 plus rolipram in naive animals. To determine the roles of prostaglandin E2 and rolipram in the prolongation of hyperalgesic response after delayed sympathectomy, rats were treated with either prostaglandin E2 or rolipram prior to sympathectomy. Prostaglandin E2 or rolipram alone were also administered five days after the sympathectomy. It was found that sympathectomy done five days after first injecting either prostaglandin E2 or rolipram alone did not produce enhanced hyperalgesic response. These data suggest that induction of a prolonged state of mechanical hyperalgesia causes time-dependent alterations in the sympathetic control of peripheral nociceptive mechanisms such that sympathectomy can lead to enhanced hyperalgesic response. These findings may be relevant to post-sympathectomy pain, a clinical entity for which there has been no available animal models.


Subject(s)
Hyperalgesia/drug therapy , Hyperalgesia/surgery , Sympathectomy , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Dinoprostone/pharmacology , Drug Combinations , Male , Pyrrolidinones/pharmacology , Rats , Rats, Sprague-Dawley , Rolipram , Time Factors
15.
J Neurosci ; 15(12): 8031-8, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8613740

ABSTRACT

The selective mu-opioid agonist, D-Ala2,N-Me-Phe4,Gly5-ol-enkephalin (DAMGO), or the selective A1-adenosine agonist N6-cyclopentyladenosine (CPA), when coinjected intradermally with prostaglandin E2 (PGE2), dose-dependently inhibited PGE2-induced mechanical hyperalgesia in the rat hindpaw, as determined by the Randall-Selitto paw-withdrawal test. Repeated (hourly x 3) intradermal injections of DAMGO or CPA produced tolerance to the antinociceptive effect of a fourth injection 1 hr later. Furthermore, repeated (hourly x 3) intradermal injections of DAMGO produced cross-tolerance to the antinociceptive effect of CPA, and repeated (hourly x 3) intradermal injection of CPA produced cross-tolerance to the antinociceptive effect of DAMGO. The demonstration of the bidirectional cross-tolerance between the peripheral antinociceptive effects of DAMGO and CPA supports the hypothesis that both these agents produced antinociception by acting on the same cell, presumably the primary afferent nociceptor, and that the development of tolerance involves changes downstream to activation of mu-opioid and A1-adenosine receptors. The opioid antagonist naloxone, which had no effect on paw-withdrawal threshold in normal paws, produced withdrawal threshold in normal paws, produced withdrawal hyperalgesia in DAMGO-tolerant paws. Furthermore, naloxone elicited a cross-withdrawal hyperalgesia response in CPA-tolerant paws. Similarly, the A1-adenosine antagonist 1,3-dipropyl-8-(2-amino-4- chlorophenyl)-xanthine (PACPX), which had no effect on paw-withdrawal threshold in normal paws, elicited a withdrawal hyperalgesia response in CPA-tolerant paws and cross-withdrawal hyperalgesia responses in DAMGO-tolerant paws. These cross-dependence and cross-withdrawal responses suggest that the development of dependence to mu-opioid and A1-adenosine agonists involves changes in the same second messenger system downstream to both mu-opioid and A1-adenosine receptor activation.


Subject(s)
Adenosine/pharmacology , Narcotics/pharmacology , Nociceptors/drug effects , Adenosine/administration & dosage , Adenosine/analogs & derivatives , Analgesics/pharmacology , Animals , Dinoprostone , Drug Tolerance , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Enkephalins/pharmacology , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Hyperalgesia/prevention & control , Male , Morphine/pharmacology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Narcotics/administration & dosage , Rats , Rats, Sprague-Dawley , Xanthines/pharmacology
16.
Methods Find Exp Clin Pharmacol ; 13(10): 681-6, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1663195

ABSTRACT

The effect of baclofen, a GABAB agonist, has been studied in three antinociceptive tests (tail flick latency, hot plate method and acetic acid-induced writhing) in mice. In all three models, baclofen was found to elicit a dose-dependent antinociceptive effect. The observed antinociceptive effect was stereospecific, as the levo isomer of baclofen was found to be more potent than the racemic mixture. Baclofen also potentiated morphine analgesia. The antinociceptive effect of baclofen was reversed by both CGP 35348, a GABAB antagonist, and naloxone, an opioid antagonist, but not by bicuculline or picrotoxin, GABAA antagonists. However, in acetic acid-induced writhing, naloxone failed to reverse baclofen analgesia. The data suggest that the antinociceptive effect of baclofen is GABAB receptor-mediated and that there may be a GABAergic and opiate/or non-opiate interaction in eliciting the analgesic effect.


Subject(s)
Analgesia , Baclofen/pharmacology , Pain/prevention & control , Receptors, GABA-A/drug effects , Animals , Baclofen/antagonists & inhibitors , Female , GABA-A Receptor Antagonists , Male , Mice , Naloxone/pharmacology , Organophosphorus Compounds/pharmacology , Pain Measurement
17.
Indian J Exp Biol ; 29(3): 241-3, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1651904

ABSTRACT

Effects of GABAergic agents and that of electroconvulsive shock (ECS) treatment were studied on bicuculline and picrotoxin (PTX)-induced convulsions in mice. Neither acute nor chronic ECS had any significant effect on bicuculline-induced convulsions, whereas the latency for PTX-induced convulsions was delayed by both acute and chronic ECS. Baclofen treatment delayed significantly the latency for PTX-induced convulsions in animals which were subjected to both acute and chronic ECS, whereas in bicuculline-induced convulsions, it shortened the latency of convulsions 24 hr after acute ECS. Progabide delayed the bicuculline-induced convulsions except in the case of 24 hr after acute ECS and PTX-induced convulsions except in the case of animals treated chronically with ECS. Fengabine showed no significant effect on bicuculline-induced convulsions. However, on PTX-induced convulsions, the latency was delayed in animals not subjected to ECS and in those subjected to chronic ECS. The possible explanations for the alterations in the effect of GABAergic agents following electro and chemo convulsions are (i) differences in the nature of antagonism by bicuculline and PTX, (ii) alterations in receptor sensitivity or number, and (iii) alterations in the levels of endogenous neurotransmitters, the latter two resulting as a result of acute or chronic ECS.


Subject(s)
Receptors, GABA-A/drug effects , Seizures/physiopathology , Animals , Bicuculline , Electroshock , Male , Picrotoxin , Rats , Rats, Inbred Strains , Receptors, GABA-A/physiology , Seizures/etiology
18.
Indian J Exp Biol ; 28(12): 1149-59, 1990 Dec.
Article in English | MEDLINE | ID: mdl-2099331

ABSTRACT

Effect of chronic treatment with Ro 15-1788, a benzodiazepine (BZ) receptor antagonist, and its withdrawal, on the cortical and hippocampal electroencephalogram (EEG) was investigated in rats. Chronic treatment with Ro 15-1788 and its withdrawal (24 and 48 hr) were found to reduce the EEG amplitude in both cortical and hippocampal regions. This reduction in cortical and hippocampal EEG amplitude produced by chronic treatment with Ro 15-1788 and its withdrawal was reversed by gamma aminobutyric acid (GABA), pentobarbitone and picrotoxin, agents known to modulate the GABA/BZ synaptic events by acting at different sites on the complex. Baclofen a GABAB agonist and FG7142, a BZ inverse agonist were found to further reduce the EEG amplitude in the cortical and hippocampal regions of these rats, chronically treated with Ro 15-1788. Diazepam, a BZ agonist was found to have no significant effect on the alteration produced in the cortical and hippocampal EEG amplitude by chronic treatment with Ro 15-1788 or its withdrawal. It is suggested that the conformational changes produced on the GABA/BZ receptor complex by BZ receptor occupation, has a facilitatory effect on the actions of those drugs which act on the GABA/BZ receptor complex and the direction of this enhancement depended on the nature of the drug.


Subject(s)
Cerebral Cortex/drug effects , Flumazenil/pharmacology , Hippocampus/drug effects , Animals , Electroencephalography/drug effects , Flumazenil/administration & dosage , Rats , Rats, Inbred Strains
19.
Arch Int Pharmacodyn Ther ; 307: 18-31, 1990.
Article in English | MEDLINE | ID: mdl-1965677

ABSTRACT

The effect of baclofen, a GABAB-agonist, was studied on both forced swimming-induced immobility and isoprenaline-induced enhancement of forced swimming-induced immobility in mice. (+/-) Baclofen (0.5 and 1 mg/kg), and (-) baclofen (0.5, 1 and 2 mg/kg) attenuated forced swimming-induced immobility. The effect of baclofen was not reversed by bicuculline, a GABAA-antagonist. Baclofen also reduced isoprenaline-induced enhancement of forced swimming-induced immobility. On concomitant administration of a subeffective dose of baclofen with a subeffective dose of propranolol, desipramine and amitriptyline, a potentiating effect was observed. These results are corroborative of our previous finding that GABAergic agents, particularly GABAB-receptors, play a role in the modulation of despair behavior in mice and in the action of antidepressant drugs. Baclofen (5 mg/kg) did not produce any significant effect on forced swimming-induced immobility, but reduced significantly the locomotor activity of the animals. Lower doses (0.5 and 1 mg/kg) of baclofen, which reduced the forced swimming-induced immobility, did not affect the locomotor activity. At higher and lower tissue concentrations of the drug, involvement of different receptor populations is suggested.


Subject(s)
Baclofen/pharmacology , Motor Activity/drug effects , Stress, Psychological/psychology , Amitriptyline/pharmacology , Animals , Bicuculline/pharmacology , Desipramine/pharmacology , Drug Interactions , Female , Isoproterenol/pharmacology , Male , Mice , Muscle Tonus/drug effects , Postural Balance/drug effects , Propranolol/pharmacology , Rats , Rats, Inbred Strains , Receptors, GABA-A/drug effects , Reserpine/pharmacology , Stereoisomerism , Swimming
20.
Methods Find Exp Clin Pharmacol ; 11(10): 597-601, 1989 Oct.
Article in English | MEDLINE | ID: mdl-2511384

ABSTRACT

The effect of GABA agonists, namely gamma aminobutyric acid, muscimol, sodium valproate and baclofen was studied on radiant heat-induced nociception in mice. All of the drugs, with the exception of sodium valproate, enhanced the reaction time to radiant heat as effect per se. Concomitant administration of any of these agents with morphine showed a potentiation of morphine-induced analgesia. The GABA antagonists bicuculline and picrotoxin failed to reverse the antinociceptive effect; paradoxically both these agents showed antinociceptive effect per se and also enhanced the response due to morphine.


Subject(s)
Analgesics , gamma-Aminobutyric Acid/physiology , Animals , Baclofen/pharmacology , Bicuculline/pharmacology , Female , Male , Mice , Morphine/pharmacology , Muscimol/pharmacology , Pain/physiopathology , Picrotoxin/pharmacology , Rats , Rats, Inbred Strains , Reaction Time/drug effects , Sensory Thresholds/drug effects , Valproic Acid/pharmacology , gamma-Aminobutyric Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...