Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 60(4): 580-6, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21172361

ABSTRACT

Peripheral ischemia is commonly associated with an increase in tissue ATP concentration and a decrease in tissue pH. Although in vitro data suggest that low tissue pH can affect ATP-binding affinities to P2 receptors, the mechanistic relationship between ATP and low pH on peripheral nociception has not been fully examined. This study was designed to investigate the potential role of an acidified environment on intraplantar αßmeATP-induced peripheral pain responses in rats. The mechanical allodynia (MA) produced by injection of αßmeATP was significantly increased in animals that received the drug diluted in pH 4.0 saline compared to those that received the drug diluted in pH 7.0 saline. Moreover, animals injected with αßmeATP (100 nmol) in pH 4.0 saline developed thermal hyperalgesia (TH), which did not occur in animals treated with αßmeATP diluted in pH 7.0 saline. To elucidate which receptors were involved in this pH-related facilitation of αßmeATP-induced MA and TH, rats were pretreated with PPADS (P2 antagonist), TNP-ATP (P2X antagonist), MRS2179 (P2Y1 antagonist), AMG9810 (TRPV1 antagonist) or amiloride (ASIC blocker). Both PPADS and TNP-ATP dose-dependently blocked pH-facilitated MA, while TH was significantly reduced by pre-treatment with MRS2179 or AMG9810. Moreover, amiloride injection significantly reduced low pH-induced facilitation of αßmeATP-mediated MA, but not TH. These results demonstrate that low tissue pH facilitates ATP-mediated MA via the activation of P2X receptors and ASICs, whereas TH induced by ATP under low pH conditions is mediated by the P2Y1 receptor and TRPV1, but not ASIC. Thus distinct mechanisms are responsible for the development of MA and TH under conditions of tissue acidosis and increased ATP.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Hyperalgesia/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Purinergic P2X/metabolism , Receptors, Purinergic P2Y/metabolism , Sodium Channels/metabolism , TRPV Cation Channels/metabolism , Acid Sensing Ion Channels , Adenosine Triphosphate/administration & dosage , Analysis of Variance , Animals , Hot Temperature , Hydrogen-Ion Concentration , Hyperalgesia/chemically induced , Male , Pain Measurement , Pain Perception/drug effects , Pain Perception/physiology , Physical Stimulation , Random Allocation , Rats , Rats, Sprague-Dawley
2.
Neuropharmacology ; 59(6): 460-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20600171

ABSTRACT

Our laboratory has recently demonstrated that an increase in the spinal neurosteroid, dehydroepiandrosterone sulfate (DHEAS) facilitates nociception via the activation of sigma-1 receptors and/or the allosteric inhibition GABA(A) receptors. Several lines of evidence have suggested that DHEAS positively modulates N-methyl-d-aspartate (NMDA) receptor activity within the central nervous system. Moreover, we have demonstrated that the activation of sigma-1 receptors increases NMDA receptor activity. Since NMDA receptors play a key role in the enhancement of pain perception, the present study was designed to determine whether spinally administered DHEAS modulates NMDA receptor-mediated nociceptive activity and whether this effect is mediated by sigma-1 or GABA(A) receptors. Intrathecal (i.t.) DHEAS was found to significantly potentiate i.t. NMDA-induced spontaneous pain behaviors. Subsequent immunohistochemical analysis demonstrated that i.t. DHEAS also increased protein kinase C (PKC)- and protein kinase A (PKA)-dependent phosphorylation of the NMDA receptor subunit NR1 (pNR1), which was used as a marker of NMDA receptor sensitization. The sigma-1 receptor antagonist, BD-1047, but not the GABA(A) receptor agonist, muscimol, dose-dependently suppressed DHEAS's facilitatory effect on NMDA-induced nociception and pNR1 expression. In addition, pretreatment with either a PKC or PKA blocker significantly reduced the facilitatory effect of DHEAS on NMDA-induced nociception. Conversely the GABA(A) receptor antagonist, bicuculline did not affect NMDA-induced pain behavior or pNR1 expression. The results of this study suggest that the DHEAS-induced enhancement of NMDA-mediated nociception is dependent on an increase in PKC- and PKA-dependent pNR1. Moreover, this effect of DHEAS on NMDA receptor activity is mediated by the activation of spinal sigma-1 receptors and not through the inhibition of GABA(A) receptors.


Subject(s)
Dehydroepiandrosterone Sulfate/pharmacology , N-Methylaspartate/pharmacology , Pain/metabolism , Phosphorylation/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, sigma/metabolism , Analysis of Variance , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Immunohistochemistry , Injections, Spinal , Male , Mice , Mice, Inbred ICR , Pain/chemically induced , Protein Kinase C/metabolism , Receptors, GABA-A/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism , Sigma-1 Receptor
3.
Neurosci Lett ; 477(2): 95-9, 2010 Jun 21.
Article in English | MEDLINE | ID: mdl-20417251

ABSTRACT

Our previous studies have demonstrated that intrathecal (i.t.) administration of a sigma-1 receptor agonist facilitated peripheral nociception via calcium-dependent second messenger cascades including protein kinase C (PKC). We also showed that activation of spinal sigma-1 receptors increased the phosphorylation of the NMDA receptor NR1 subunit (pNR1) in the spinal cord dorsal horn, which resulted in the potentiation of NMDA receptor function. The present study was designed to examine the effect of different PKC isoform inhibitors on sigma-1 receptor-mediated pain facilitation and increased spinal pNR1 expression in mice. The intrathecal injection of the sigma-1 receptor agonist, PRE-084 (PRE, 3nmol/5mul) increased the frequency of paw withdrawal responses to mechanical stimuli (0.6g) and the number of spinal pNR1-immunoreactive (ir) cells. Intrathecal pretreatment with inhibitors (Go6976, PKCepsilonV1-2 or PKC zetapseudosubstrate) of the PKCalpha, epsilon or zeta isoforms significantly reduced the PRE-induced pain facilitatory effect. On the other hand, the PRE-induced increase in the number of spinal pNR1-ir neurons was only blocked by inhibitors of the PKCalpha and PKCepsilon isoforms, but not the PKCzeta isoform. These findings demonstrate that the sigma-1 receptor-induced increase in spinal pNR1 expression is mediated by the PKCalpha and PKC epsilon isoforms, which in turn contribute to the pain facilitation phenomenon. Conversely, the sigma-1 receptor activation of the PKCzeta isoform appears to be involved in a pain signaling pathway that is independent of spinal pNR1 modulation.


Subject(s)
Protein Kinase C-alpha/physiology , Protein Kinase C-epsilon/physiology , Protein Kinase C/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, sigma/physiology , Spinal Cord/metabolism , Animals , Injections, Spinal , Isoenzymes/antagonists & inhibitors , Isoenzymes/physiology , Male , Mice , Mice, Inbred ICR , Neurons/metabolism , Pain/enzymology , Pain/physiopathology , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-epsilon/antagonists & inhibitors , Spinal Cord/drug effects , Sigma-1 Receptor
4.
Exp Neurol ; 224(1): 123-32, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20226782

ABSTRACT

The most common type of chronic pain following spinal cord injury (SCI) is central neuropathic pain and SCI patients typically experience mechanical allodynia and thermal hyperalgesia. The present study was designed to examine the potential role of astrocyte gap junction connectivity in the induction and maintenance of "below-level" neuropathic pain in SCI rats. We examined the effect of intrathecal treatment with carbenoxolone (CARB), a gap junction decoupler, on SCI-induced bilateral thermal hyperalgesia and mechanical allodynia during the induction phase (postoperative days 0 to 5) and the maintenance phase (days 15 to 20) following T13 spinal cord hemisection. Immunohistochemistry was performed to determine potential SCI-induced changes in spinal astrocyte activation and phosphorylation of the NMDA receptor NR1 subunit (pNR1). CARB administered during the induction period dose-dependently attenuated the development of bilateral thermal hyperalgesia and mechanical allodynia. Intrathecal CARB also significantly reduced the bilateral SCI-induced increase in GFAP-immunoreactive (ir) staining and the number of pNR1-ir cell profiles in the spinal cord dorsal horn compared to vehicle-treated rats. In contrast, CARB treatment during the maintenance phase had no effect on the established thermal hyperalgesia and mechanical allodynia nor on spinal GFAP expression or the number of pNR1-ir cell profiles. These results indicate that gap junctions play a critical role in the activation of astrocytes distant from the site of SCI and in the subsequent phosphorylation of NMDA receptors in the lumbar spinal cord. Both of these processes appear to contribute to the induction of bilateral below-level pain in SCI rats.


Subject(s)
Astrocytes/drug effects , Carbenoxolone/pharmacology , Gap Junctions/drug effects , Neuralgia/drug therapy , Recovery of Function/drug effects , Spinal Cord Injuries/complications , Analysis of Variance , Animals , Astrocytes/metabolism , Dose-Response Relationship, Drug , Glial Fibrillary Acidic Protein/metabolism , Image Processing, Computer-Assisted , Immunohistochemistry , Injections, Spinal , Male , Motor Activity/physiology , Neuralgia/etiology , Neuralgia/metabolism , Pain Measurement , Phosphorylation/physiology , Posterior Horn Cells/metabolism , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism , Thoracic Vertebrae
5.
J Pain ; 11(8): 718-27, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20338827

ABSTRACT

UNLABELLED: We have previously established a thrombus-induced ischemic pain (TIIP) model in the rat, which mimics the pathophysiology of ischemic pain in patients with peripheral arterial disease. Because ischemia commonly induces acidosis and ATP release, one of the goals of this study was to investigate the role of acid-sensing ion channels (ASICs), transient receptor potential vanilloid-1 (TRPV1) receptors, and P2X receptors in the maintenance of ischemia-induced mechanical allodynia (MA). To test this, amiloride (an ASIC blocker), AMG-9810 (a TRPV1 blocker), or PPADS (a P2Xs antagonist) was intraplantarly injected at day 3 after FeCl(2) application onto the femoral artery. Ipsilateral administration of amiloride or PPADS but not AMG-9810 dose-dependently reduced MA. However, contralateral amiloride or PPADS did not suppress contralateral MA. Interestingly, co-administration of submaximal doses of amiloride and PPADS produced a significantly prolonged suppression of MA. Furthermore, ipsilateral EGTA (a calcium chelator) or chelerythrine (a protein kinase C inhibitor) also significantly reduced MA. Collectively, these findings suggest that peripheral ASICs and P2X receptors are involved in the maintenance of TIIP, which is possibly mediated by a Ca(2+)-protein kinase C signaling mechanism. These results provide mechanistic information about peripheral ischemic nociception that may be useful for developing better therapeutic management of ischemic pain in patients with peripheral arterial disease. PERSPECTIVE: The results of the current study demonstrate that peripheral administration of an ASICs blocker or P2X antagonist significantly suppress TIIP. Co-administration of submaximal doses of ASIC and P2X antagonists produced an even greater effect. These results implicate peripheral ASICs and P2X receptors in the maintenance of thrombus-induced ischemic pain.


Subject(s)
Ischemia/metabolism , Nerve Tissue Proteins/metabolism , Pain/metabolism , Receptors, Purinergic P2/metabolism , Sodium Channels/metabolism , Acid Sensing Ion Channels , Analysis of Variance , Animals , Hot Temperature , Hyperalgesia/metabolism , Ischemia/complications , Male , Pain/etiology , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X , TRPV Cation Channels/metabolism
6.
Anesth Analg ; 110(2): 622-9, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20007733

ABSTRACT

BACKGROUND: A previous study from our laboratories showed that a significant reduction in spinal N-methyl-D-aspartate (NMDA) receptor NR1 subunit phosphorylation (pNR1) is associated with the antiallodynic effect produced by intrathecal (IT) injection of the alpha-2 adrenoceptor agonist, clonidine, in neuropathic rats. In this study, we determined whether the spontaneous pain and increased pNR1 expression induced by NMDA injection are reduced by IT injection of either clonidine or the mu-opioid receptor agonist, [D-Ala2, NMe-Phe4, Gly-ol5]-enkephalin (DAMGO). METHODS: We examined the effect of clonidine (20 microg/rat) or DAMGO (1 microg/rat) injection on IT NMDA-induced spontaneous nociceptive behavior and pNR1 expression in the spinal dorsal horn. We also determined whether the effect of clonidine is mediated by alpha-2A or alpha-2C adrenoceptors. Finally, rat spinal cords were immunohistochemically processed for double staining of pNR1 and alpha-2A or alpha-2C adrenoceptors or mu-opioid receptors. RESULTS: The NMDA-induced increase in both pNR1 expression and nociceptive behavior was significantly reduced by IT clonidine but not DAMGO. This analgesic effect of clonidine was blocked by administration of either an alpha-2A (BRL44408, 30 microg/rat) or an alpha-2C (JP-1302, 50 microg/rat) adrenoceptor antagonist. In addition, immunocytochemistry revealed that spinal pNR1 immunoreactive cells co-contain alpha-2A and alpha-2C adrenoceptors. CONCLUSIONS: These results demonstrate that the IT NMDA-induced increase in pNR1 expression and nociceptive behavior is significantly reduced by activation of alpha-2 adrenoceptors, but not mu-opioid receptors, in the spinal cord dorsal horn. Furthermore, these findings suggest that the modulation of spinal NR1 phosphorylation is linked to the effect of IT clonidine on postsynaptic neuronal activity.


Subject(s)
Analgesics/pharmacology , Behavior, Animal , N-Methylaspartate/pharmacology , Pain/drug therapy , Pain/metabolism , Posterior Horn Cells/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Opioid, mu/metabolism , Adrenergic alpha-2 Receptor Antagonists , Adrenergic alpha-Agonists/pharmacology , Animals , Clonidine/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Immunohistochemistry , Injections, Spinal , Male , Pain/chemically induced , Pain/prevention & control , Phosphorylation , Rats , Rats, Sprague-Dawley
7.
Br J Pharmacol ; 157(4): 666-73, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19422393

ABSTRACT

BACKGROUND AND PURPOSE: The neurosteroid, dehydroepiandrosterone sulphate (DHEAS) and its non-sulphated form, DHEA, are considered as crucial endogenous modulators of a number of important physiological events. Evidence suggests that DHEAS and DHEA modulate central nervous system-related functions by activating sigma-1 receptors and/or allosterically inhibiting gamma-aminobutyric acid receptor type A (GABA(A)) receptors. As both the sigma-1 receptor and the GABA(A) receptor play important roles in spinal pain transmission, the present study was designed to examine whether intrathecally injected DHEAS or DHEA affect nociceptive signalling at the spinal cord level. EXPERIMENTAL APPROACH: We first determined whether intrathecal (i.t.) DHEA or DHEAS injection was able to affect nociceptive thresholds to peripheral mechanical stimulation and subsequently examined whether this effect was mediated by sigma-1 or the GABA(A) receptors. KEY RESULTS: The i.t. DHEAS injection dose-dependently decreased the nociceptive threshold to mechanical stimulation, thus producing mechanical allodynia. Moreover, this DHEAS-induced mechanical allodynia was significantly reduced by administration of the sigma-1 receptor antagonist, BD-1047 or the GABA(A) receptor agonist, muscimol. Conversely, i.t. DHEA had no effect on mechanical sensitivity. However, when i.t. DHEA was combined with the GABA(A) receptor antagonist bicuculline, DHEA dose-dependently produced mechanical allodynia similar to that of DHEAS. This effect was blocked by BD-1047 and by muscimol. CONCLUSIONS AND IMPLICATIONS: These findings indicate that i.t. injection of DHEAS produces mechanical allodynia and that the development of this mechanical allodynia is mediated by sigma-1 and GABA(A) receptors. The findings of this study raise several interesting questions for further investigations into the mechanisms underlying neurosteroid modulation of spinal pain transmission.


Subject(s)
Analgesics/pharmacology , Dehydroepiandrosterone Sulfate/pharmacology , Pain Threshold/drug effects , Receptors, GABA-A/physiology , Receptors, sigma/physiology , Spinal Cord/physiology , Animals , Bicuculline/pharmacology , Dehydroepiandrosterone/administration & dosage , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone Sulfate/administration & dosage , Drug Interactions , Ethylenediamines/pharmacology , GABA Agonists/pharmacology , GABA-A Receptor Agonists , GABA-A Receptor Antagonists , Injections, Spinal , Male , Mice , Mice, Inbred ICR , Muscimol/pharmacology , Receptors, sigma/antagonists & inhibitors , Spinal Cord/drug effects , Sigma-1 Receptor
8.
J Pain ; 10(3): 253-63, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19010737

ABSTRACT

UNLABELLED: Although intrathecal (i.t.) administration of the alpha(2)-adrenoceptor agonist clonidine has a pronounced analgesic effect, the clinical use of clonidine is limited by its side effects. Previously, our laboratory has demonstrated that the subcutaneous injection of diluted bee venom (DBV) into an acupoint (termed apipuncture) produces significant analgesic effect in various pain animal models. The present study was designed to examine whether DBV injection into the Zusanli acupoint (ST-36) could enhance lower-dose clonidine-induced analgesic effects without the development of hypotension, bradycardia, or sedation. In the mouse formalin test, DBV injection produced a dramatic leftward shift in the dose-response curve for clonidine-induced analgesia. In a rat neuropathic pain model i.t. clonidine dose dependently suppressed chronic constriction injury (CCI)-induced mechanical allodynia and thermal hyperalgesia, and this clonidine-induced analgesic effect was significantly potentiated by apipuncture pretreatment. DBV apipuncture alone or in combination with a low dose of i.t. clonidine produced an analgesic effect similar to that of the high dose of clonidine, but without significant side effects. The analgesic effect produced by the combination of i.t. clonidine and apipuncture was completely blocked by pretreatment with an alpha(2)-adrenoceptor antagonist. These data show that DBV-apipuncture significantly enhances clonidine-induced analgesia and suggest that a combination of low dose clonidine with acupuncture therapy represents a novel strategy for pain management that could eliminates clonidine's side effects. PERSPECTIVE: This study demonstrated that intrathecal clonidine-induced analgesia is significantly enhanced when it is combined with chemical acupuncture treatment. The administration of low-dose clonidine in combination with acupuncture produced a potent analgesic effect without significant side effects and thus represents a potential novel strategy for the management of chronic pain.


Subject(s)
Acupuncture Points , Analgesics/therapeutic use , Bee Venoms/therapeutic use , Clonidine/therapeutic use , Hyperalgesia/drug therapy , Pain Measurement , Pain/drug therapy , Analgesics/adverse effects , Animals , Bee Venoms/administration & dosage , Clonidine/administration & dosage , Clonidine/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Therapy, Combination , Hyperalgesia/chemically induced , Injections, Spinal , Injections, Subcutaneous , Male , Mice , Mice, Inbred ICR , Pain/chemically induced , Pain/physiopathology , Pain Measurement/methods , Rats , Rats, Sprague-Dawley
9.
Anesthesiology ; 109(5): 879-89, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18946301

ABSTRACT

BACKGROUND: Selective blockade of spinal sigma(1) receptors (Sig-1R) suppresses nociceptive behaviors in the mouse formalin test. The current study was designed to verify whether intrathecal Sig-1R antagonists can also suppress chronic neuropathic pain. METHODS: Neuropathic pain was produced by chronic constriction injury (CCI) of the right sciatic nerve in rats. The Sig-1R antagonist BD1047 was administered intrathecally twice daily from postoperative days 0 to 5 (induction phase of neuropathic pain) or from days 15 to 20 (maintenance phase). Western blot and immunohistochemistry were performed to determine changes in Sig-1R expression and to examine the effect of BD1047 on N-methyl-D-aspartate receptor subunit 1 expression and phosphorylation in spinal cord dorsal horn from neuropathic rats. RESULTS: BD1047 administered on postoperative days 0-5 significantly attenuated CCI-induced mechanical allodynia, but not thermal hyperalgesia, and this suppression was blocked by intrathecal administration of the Sig-1R agonist PRE084. In contrast, BD1047 treatment during the maintenance phase of neuropathic pain had no effect on mechanical allodynia. Sig-1R expression significantly increased in the ipsilateral spinal cord dorsal horn from days 1 to 3 after CCI. Importantly, BD1047 (30 nmol) administered intrathecally during the induction, but not the maintenance phase, blocked the CCI-induced increase in N-methyl-D-aspartate receptor subunit 1 expression and phosphorylation. CONCLUSIONS: These results demonstrate that spinal Sig-1Rs play a critical role in both the induction of mechanical allodynia and the activation of spinal N-methyl-d-aspartate receptors in CCI rats and suggest a potential therapeutic role for the use of Sig-1R antagonists in the clinical management of neuropathic pain.


Subject(s)
Ethylenediamines/administration & dosage , Neuralgia/metabolism , Neuralgia/prevention & control , Receptors, N-Methyl-D-Aspartate/biosynthesis , Receptors, sigma/biosynthesis , Spinal Cord/metabolism , Animals , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Injections, Spinal , Male , Physical Stimulation/methods , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, sigma/antagonists & inhibitors , Spinal Cord/drug effects
10.
J Pharm Pharmacol ; 60(10): 1347-54, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18812028

ABSTRACT

Ursolic acid (UA) is pentacyclic triterpenoic acid that naturally occurs in many medicinal herbs and plants. In this study, we examined the possible suppressive effect of UA extracted from Oldenlandia diffusa on zymosan-induced acute inflammation in mice and complete Freund's adjuvant (CFA)-induced arthritis in rats. UA treatment (per oral) dose-dependently (25-200 mg kg(-1)) suppressed zymosan-induced leucocyte migration and prostaglandin E2 (PGE(2)) production in the air pouch exudates. Since the maximal effective dose of UA was 50 mg kg(-1) in the zymosan experiment, we used this dose of UA in a subsequent study using an adjuvant-induced rheumatoid arthritis model. UA treatment (50 mg kg(-1), per oral, once a day for 10 days) was started from day 12 after adjuvant injection. UA dramatically inhibited paw swelling, plasma PGE(2) production and radiological changes in the joint caused by CFA injection. Moreover, UA significantly suppressed the arthritis-induced mechanical and thermal hyperalgesia as well as the spinal Fos expression, as determined by immunohistochemistry, which was increased by CFA injection. In addition, overall anti-arthritic potency of UA was comparable with ibuprofen (100 mg kg(-1), oral) while UA did not induce significant gastric lesions as compared with the ibuprofen treatment group. These findings strongly suggest that UA is a useful suppressive compound for rheumatoid arthritis treatment with low risk of gastric problems.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Experimental/drug therapy , Inflammation/drug therapy , Triterpenes/therapeutic use , Acute Disease , Animals , Antirheumatic Agents/chemistry , Arthritis, Experimental/physiopathology , Cell Movement/drug effects , Chronic Disease , Dinoprostone/biosynthesis , Disease Models, Animal , Dose-Response Relationship, Drug , Inflammation/chemically induced , Inflammation/physiopathology , Leukocytes/cytology , Leukocytes/drug effects , Mice , Mice, Inbred ICR , Oldenlandia/chemistry , Pain/physiopathology , Pain/prevention & control , Plant Extracts/chemistry , Plant Extracts/therapeutic use , Rats , Rats, Sprague-Dawley , Triterpenes/chemistry , Zymosan , Ursolic Acid
11.
Pain ; 139(3): 520-532, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18691814

ABSTRACT

Patients with peripheral arterial disease (PAD) commonly suffer from ischemic pain associated with severe thrombosis. However, the pathophysiology of peripheral ischemic pain is not fully understood due to the lack of an adequate animal model. In this study, we developed a new rodent model of thrombus-induced ischemic pain (TIIP) to investigate the neuronal mechanisms underlying ischemic pain. Ischemia was induced by application of 20% FeCl(2) onto the surface of the femoral artery for 20min. Induction of peripheral ischemia was confirmed by measurement of the concentration of Evans blue and by increases in the ischemia-specific markers, hypoxia-inducible factor-1 alpha and vascular endothelial growth factor in the ipsilateral plantar muscles. Ischemic pain, as indicated by the presence of mechanical allodynia, developed bilaterally and peaked at days 3-9 post-FeCl(2) application and gradually decreased through day 31. Systemic heparin pretreatment dose dependently suppressed ischemic pain, suggesting that thrombosis-induced ischemia might be a key factor in TIIP. Intraplantar injection of BMS-182874, an ET(A) (endothelin-A) receptor antagonist, at day 3 selectively blocked ipsilateral pain, indicating that ET(A) receptor activity mediated TIIP. Spinal GFAP expression was significantly increased by FeCl(2) and intrathecal injection of carbenoxolone (an astrocyte gap junction decoupler) at day 3 significantly reduced TIIP, suggesting that spinal astrocyte activation plays an important role. However, the anti-inflammatory agent, ibuprofen, did not affect TIIP. In conclusion, we have developed a novel animal model of TIIP that should be useful in investigating the pathophysiological mechanisms that underlie human peripheral ischemic pain.


Subject(s)
Disease Models, Animal , Hindlimb/blood supply , Hyperalgesia/physiopathology , Ischemia/physiopathology , Thrombosis/complications , Animals , Astrocytes/chemistry , Biomarkers , Femoral Artery/drug effects , Ferrous Compounds/toxicity , Gliosis/etiology , Heparin/therapeutic use , Hot Temperature/adverse effects , Hyperalgesia/etiology , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Ischemia/etiology , Male , Oxidants/toxicity , Physical Stimulation/adverse effects , Posterior Horn Cells/physiopathology , Rats , Rats, Sprague-Dawley , Spinal Cord/pathology , Spinal Cord/physiopathology , Thrombosis/chemically induced , Thrombosis/pathology , Thrombosis/prevention & control , Touch , Vascular Endothelial Growth Factor A/analysis
12.
J Neurosci Res ; 86(16): 3644-54, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18655205

ABSTRACT

Sigma sites, originally proposed as opioid receptor subtypes, are currently thought to represent unique receptors with a specific pattern of drug selectivity, a well-established anatomical distribution and broad range of functional roles including potential involvement in nociceptive mechanisms. We have recently demonstrated that intrathecal (i.t.) treatment with a sigma-1 receptor antagonist reduced formalin-induced pain behavior. In the present study, we investigated the potential role of spinal sigma-1 receptor agonists in peripherally initiated nociception and attempted to elucidate intracellular signaling mechanisms associated with spinal cord sigma-1 receptor activation in mice. The i.t. injection of the sigma-1 receptor agonists PRE-084 (PRE) or carbetapentane (CAR) significantly decreased tail-flick latency (TFL) and increased the frequency of paw withdrawal responses to mechanical stimulation (von Frey filament, 0.6 g) as well as the amount of Fos expression in the spinal cord dorsal horn induced by noxious paw-pinch stimulation. These PRE- or CAR-induced facilitatory effects on nociception were significantly blocked by i.t. pretreatment with the sigma-1 receptor antagonist, BD-1047, the phospholipase C (PLC) inhibitor, U-73,122, the Ca(2+)-ATPase inhibitor, thapsigargin, and the protein kinase C (PKC) inhibitor, chelerythrine. Western blot analysis further revealed that i.t. PRE or CAR injection significantly increased pan-PKC as well as the PKCalpha, epsilon, and zeta isoforms in the dorsal horn. Collectively, these findings demonstrate that calcium-dependent second messenger cascades including PKC are involved in the facilitation of nociception associated with spinal sigma-1 receptor activation.


Subject(s)
Morpholines/pharmacology , Nociceptors/drug effects , Pain/chemically induced , Protein Kinase C/drug effects , Receptors, sigma/agonists , Spinal Cord/drug effects , Animals , Antitussive Agents/pharmacology , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cyclopentanes/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Ethylenediamines/pharmacology , Inflammation Mediators/metabolism , Inflammation Mediators/pharmacology , Injections, Spinal , Male , Mice , Mice, Inbred ICR , Nociceptors/metabolism , Pain/metabolism , Pain/physiopathology , Pain Measurement/drug effects , Pain Threshold/drug effects , Pain Threshold/physiology , Posterior Horn Cells/drug effects , Posterior Horn Cells/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Receptors, sigma/antagonists & inhibitors , Receptors, sigma/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Spinal Cord/metabolism , Spinal Cord/physiopathology , Sigma-1 Receptor
13.
Anesth Analg ; 107(2): 693-700, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18633054

ABSTRACT

BACKGROUND: Intrathecal (IT) administration of the alpha-2 adrenoceptor agonist, clonidine, produces significant analgesic effects. Although several mechanisms underlying clonidine-induced analgesia have been proposed, the possible interaction with N-methyl-D-aspartate (NMDA) receptors as a major antinociceptive mechanism has not been addressed. We designed the present study to determine whether clonidine or other analgesics can affect spinal NMDA receptor activation in rats with chronic constriction injury (CCI)-induced neuropathy. METHODS: Rats underwent unilateral CCI, and received IT clonidine (1, 5, 20 microg/rat), [D-Ala2, NMe-Phe4, Gly-ol5]-enkephalin (DAMGO, mu opioid receptor agonist, 1 microg/rat), gabapentin (anticonvulsant, 100 microg/rat) or vehicle 2 wks later. After drug injection, we measured the pain response to thermal or mechanical stimuli and used immunohistochemistry to evaluate spinal cord phosphorylated NMDA-receptor subunit 1 (pNR1) expression. RESULTS: Two weeks after CCI surgery, rats displayed significant mechanical allodynia and thermal hyperalgesia, and the spinal cord dorsal horn showed a significant increase in the number of pNR1 immunoreactive neurons. IT injection of clonidine (20 microg/rat), DAMGO and gabapentin potently reduced mechanical allodynia and thermal hyperalgesia. Importantly, IT clonidine, but not IT DAMGO or gabapentin, dose-dependently reduced CCI-induced pNR1 expression in all lamina of the spinal cord dorsal horn by 30 min after injection. In addition, IT injection of the alpha-2 adrenoceptor antagonist, idazoxan (40 microg/rat) 10 min before clonidine injection completely reversed clonidine's antihyperalgesic and antiallodynic effects, as well as clonidine's suppressive effect on CCI-induced NR1 phosphorylation in the spinal cord dorsal horn. CONCLUSIONS: Our data indicate that IT clonidine's antihyperalgesic/antiallodynic effect on neuropathic pain is associated with a significant reduction in spinal NMDA receptor phosphorylation and suggests a potentially novel mechanism of clonidine's action.


Subject(s)
Adrenergic alpha-Agonists/administration & dosage , Analgesics/administration & dosage , Clonidine/administration & dosage , Neuralgia/metabolism , Posterior Horn Cells/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Amines/pharmacology , Analgesics/pharmacology , Animals , Anticonvulsants/pharmacology , Clonidine/pharmacology , Cyclohexanecarboxylic Acids/pharmacology , Dose-Response Relationship, Drug , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Gabapentin , Idazoxan/pharmacology , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/antagonists & inhibitors , Spinal Cord/metabolism , gamma-Aminobutyric Acid/pharmacology
14.
Eur J Pain ; 12(5): 552-63, 2008 Jul.
Article in English | MEDLINE | ID: mdl-17933570

ABSTRACT

Phosphorylation of the N-methyl-D-aspartate (NMDA) receptor NR1 subunit (pNR1) in the spinal cord is associated with increased neuronal responsiveness, which underlies the process of central sensitization. Because of the importance of NR1 in central sensitization, the first goal of this study was to examine both time- and lamina-dependent changes in spinal NR1 and pNR1 expression in a chronic constriction injury (CCI) model of neuropathic pain. Increased excitability of capsaicin sensitive primary afferents (CSPAs), which express TRPV1 receptors, also contributes to central sensitization. Thus, we next examined whether the depletion of CSPAs with resiniferatoxin (RTX) modified the change of spinal NR1 and pNR1 expression induced by CCI. Experimental rats were euthanized at 1, 3, 7, 14, and 28 days post-CCI surgery and spinal cords processed for NR1 or pNR1 immunostaining. The number of NR1 or pNR1-immunoreactive neurons was significantly increased in all lamina (I-VI) of the ipsilateral L4/L5 dorsal horn from 1 or 7 days post-CCI, respectively. Pretreatment with RTX (0.3mg/kg, s.c. in the scruff of the neck or intraplantar) 2 days prior to CCI completely prevented induction of thermal hyperalgesia, but not mechanical allodynia in neuropathic rats. Interestingly, RTX treatment significantly attenuated the CCI-induced upregulation of NR1 and pNR1 in spinal laminae I-II and V-VI, but not laminae III-IV as compared with that of vehicle-treated CCI rats. These findings demonstrate that the increased expression of NR1 and pNR1 in spinal laminae I-II and V-VI is dependent on activation of CSPAs, which ultimately contribute to the development of thermal hyperalgesia in neuropathic rats.


Subject(s)
Afferent Pathways/physiopathology , Capsaicin/pharmacology , Diterpenes/pharmacology , Hyperalgesia/physiopathology , Posterior Horn Cells/physiology , Protein Processing, Post-Translational , Receptors, N-Methyl-D-Aspartate/metabolism , Sciatic Neuropathy/physiopathology , Spinal Cord/physiopathology , Up-Regulation/drug effects , Afferent Pathways/drug effects , Animals , Hot Temperature/adverse effects , Hyperalgesia/etiology , Ligation , Male , Perception , Phosphorylation , Pressure/adverse effects , Protein Processing, Post-Translational/drug effects , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics , TRPV Cation Channels/drug effects , TRPV Cation Channels/physiology , Time Factors
15.
Neurosci Lett ; 430(2): 163-8, 2008 Jan 10.
Article in English | MEDLINE | ID: mdl-18061346

ABSTRACT

Our recent data, obtained using a zymosan-induced inflammatory air pouch model in mice, have demonstrated that subcutaneous bee venom (BV) injection into the hind limb selectively activates the contralateral brain stem locus coeruleus (LC) and then via a descending noradrenergic pathway and subsequent adrenal medullary catecholamine release induces a potent anti-inflammatory effect. While the efferent limb of this BV-induced neuroimmune anti-inflammatory pathway is well documented, the afferent limb of this pathway is poorly understood. In particular the spinal mechanisms involved with BV activation of the LC are currently unknown. Spinal nitric oxide (NO) and its synthase (NOS) have been shown to play an important role in the transmission and amplification of neuronal information from the spinal cord to the brain stem. In the present study we evaluated whether spinal NO plays a role in BV-induced LC activation, since we have previously shown that LC activation underlies this 'BV-induced anti-inflammatory effect' (BVAI) using the mouse air pouch model. Intrathecal (i.t.) pretreatment with l-nitro arginine methyl ester (l-NAME, non-selective NOS inhibitor), hemoglobin (NO scavenger) or 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ, soluble guanylate cyclase inhibitor) abolished BVAI on zymosan-induced leukocyte migration into the air pouch. Moreover, i.t. injection of l-N-iminoethyl-lysine (l-NIL, inducible NOS inhibitor), but not 7-nitroindazole (7-NI, neuronal NOS inhibitor), also inhibited BVAI. BV injection significantly increased both the number of Fos immunoreactive neurons and tyrosine hydroxylase-Fos double labeling neurons in the contralateral LC in zymosan-induced inflamed mice. Importantly this increase in Fos expression in the LC was also completely inhibited by i.t. injection of l-NIL, but not by i.t. injection of 7-NI. Collectively these results indicate that spinal NO generated from inducible NOS is involved in the BV-induced LC activation that underlies BVAI.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Bee Venoms/therapeutic use , Locus Coeruleus/enzymology , Nitric Oxide/metabolism , Peripheral Nervous System Diseases/drug therapy , Spinal Cord/enzymology , Animals , Disease Models, Animal , Drug Interactions , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Indazoles/pharmacology , Leukocytes/drug effects , Locus Coeruleus/drug effects , Male , Mice , Mice, Inbred ICR , NG-Nitroarginine Methyl Ester/pharmacology , Neural Pathways/drug effects , Neural Pathways/enzymology , Oncogene Proteins v-fos/metabolism , Spinal Cord/drug effects , Tyrosine 3-Monooxygenase/metabolism
16.
Neuropharmacology ; 53(5): 677-86, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17765931

ABSTRACT

Previous data from our laboratories using the mouse air pouch model demonstrated that intrathecal injection of the cholinomimetic drug, neostigmine, produces a significant peripheral anti-inflammatory effect through activation of spinal muscarinic type 2 receptors. This anti-inflammatory effect is mediated by activation of sympathetic preganglionic neurons and subsequent release of adrenomedullary catecholamines. It has been established that adrenomedullary catecholamine release is controlled by sympathetic preganglionic neurons and that these neurons are modulated by GABAergic inhibitory input. To further establish the neurochemical circuitry underlying spinally mediated anti-inflammation, the present study examined whether spinal muscarinic type 2 receptors are associated with this spinal GABAergic pathway. Intrathecal injection of the M(2) receptor agonist, arecaidine but-2-ynyl ester tosylate (ABET) dose-dependently suppressed zymosan-induced leukocyte migration into the air pouch and increased Fos (neuronal activation marker) expression in sympathetic preganglionic neurons of the T7-T11 spinal cord segments (which mainly project to the adrenal medulla), but not in sympathetic preganglionic neurons of the T1-T6 or T12-L2 segments. These effects of arecaidine but-2-ynyl ester tosylate were completely blocked by intrathecal pretreatment with baclofen (a GABA(B)R agonist) but not muscimol (a GABA(A)R agonist). Intrathecal saclofen (a GABA(B)R antagonist), but not bicuculline (a GABA(A)R antagonist), significantly reduced leukocyte migration and increased Fos expression in T7-T11 sympathetic preganglionic neurons. More importantly, this intrathecal saclofen-induced anti-inflammatory effect was completely blocked by adrenalectomy or systemic pretreatment with propranonol (a beta-adrenoceptor antagonist). Collectively, these novel findings suggest that activation of spinal muscarinic type 2 receptors suppress spinal GABA(B) receptor input and that this disinhibition mechanism ultimately leads to the release of adrenal catecholamines and a subsequent reduction in peripheral inflammation.


Subject(s)
Inflammation/physiopathology , Neural Pathways/physiopathology , Receptor, Muscarinic M2/physiology , Spinal Cord/physiopathology , gamma-Aminobutyric Acid/physiology , Adrenal Cortex Hormones/pharmacology , Adrenal Glands/drug effects , Adrenal Glands/physiology , Animals , Anti-Inflammatory Agents/pharmacology , Autonomic Fibers, Preganglionic/drug effects , Baclofen/analogs & derivatives , Baclofen/pharmacology , Catecholamines/metabolism , Catecholamines/pharmacology , Cell Proliferation/drug effects , Exudates and Transudates/physiology , GABA Antagonists/pharmacology , Image Interpretation, Computer-Assisted , Immunohistochemistry , Male , Mice , Mice, Inbred ICR , Neurons/physiology , Receptor, Muscarinic M2/drug effects , Receptors, GABA-A/drug effects , Receptors, GABA-B/drug effects
17.
Neurosci Res ; 59(1): 51-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17588699

ABSTRACT

There are several reports indicating that the locus coeruleus (LC) is capable of altering immune responses. Moreover, it is well established that the LC is the major source of descending noradrenergic system. Recently we have demonstrated that subcutaneous bee venom (BV) injection dramatically suppressed peripheral inflammation through activation of sympathetic preganglionic neurons (SPNs) leading to release of adreno-medullary catecholamines. Importantly, this 'BV-induced anti-inflammatory effect' (BVAI) is also associated with an increase of the activity of LC. Based on these data, present study examined whether BV-induced LC activation increased the activity of SPNs and this pathway played a role in BVAI using a zymosan-induced inflammatory air pouch model in mice. Unilateral BV injection into left hind limb produced anti-inflammation and specifically increased Fos expression in SPNs of the T7-T11 (which mainly project to adrenal medulla), but not those of the T1-T6 or T12-L2 spinal cord. 6-Hydroxydopamine-induced unilateral lesion of the contralateral, but not ipsilateral (to the BV injection site) LC significantly blocked BVAI and BV-induced Fos expression in SPNs. Additionally, intrathecal administration of idazoxan (alpha2-adrenoceptor antagonist), blocked BVAI. These results indicate that BV-induced activation of the contralateral LC-descending noradrenergic pathway increased the activity of SPNs that project to the adrenal medulla and this pathway is necessary for BVAI.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Bee Venoms/administration & dosage , Inflammation/drug therapy , Locus Coeruleus/physiopathology , Neurons/physiology , Spinal Cord/physiopathology , Sympathetic Fibers, Postganglionic/pathology , Animals , Choline O-Acetyltransferase/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Functional Laterality , Inflammation/pathology , Injections, Spinal/methods , Locus Coeruleus/drug effects , Male , Mice , Mice, Inbred ICR , Neural Pathways/drug effects , Neural Pathways/physiopathology , Neurons/drug effects , Oncogene Proteins v-fos/metabolism , Oxidopamine/pharmacology , Spinal Cord/drug effects , Sympatholytics/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...