Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Toxicol Appl Pharmacol ; 429: 115698, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34428447

ABSTRACT

In view of postsynaptic density 95kDA (PSD95) tethers neuronal NO synthase (nNOS) to N-methyl-d-aspartate receptor (NMDAR), the PSD95-nNOS complex represents a therapeutic target of neuropathic pain. This study therefore sought to explore the ability of PCC-0105002, a novel PSD95-nNOS small molecule inhibitor, to alter pain sensitivity in rodent neuropathic pain models. Firstly, the IC50 of PCC-0105002 for PSD95 and NOS1 binding activity was determined using an Alpha Screen assay kit. Then, we examined the effects of PCC-0105002 in the mouse formalin test and in the rat spinal nerve ligation (SNL) model, and explored the ability of PCC-0105002 to mediate analgesia and to effect motor coordination in a rota-rod test. Moreover, the mechanisms whereby PCC-0105002 mediates analgesia was explored via western blotting, Golgi staining, and co-immunoprecipitation experiments in dorsal horn. The outcomes indicated that PCC-0105002 exhibited dose-dependent attenuation of phase II pain-associated behaviors in the formalin test. The result indicated that PCC-0105002 disrupted the PSD95-nNOS interaction with IC50 of 1.408 µM. In the SNL model, PCC-0105002 suppressed mechanical allodynia, thermal hyperalgesia, and abnormal dorsal horn wide dynamic range neuron discharge. PCC-0105002 mediated an analgesic effect comparable to that of MK-801, while it was better able to enhance motor coordination as compared with MK-801. Moreover, PCC-0105002 altered signaling downstream of NMDAR and thus functionally and structurally attenuating synaptic plasticity through respective regulation of the NR2B/GluR1/CaMKIIα and Rac1/RhoA pathways. These findings suggest that the novel PSD95-nNOS inhibitor PCC-0105002 is an effective agent for alleviating neuropathic pain, and that it produces fewer motor coordination-associated side effects than do NMDAR antagonists.


Subject(s)
Aminobenzoates/therapeutic use , Analgesics/pharmacology , Disks Large Homolog 4 Protein/metabolism , Esters/therapeutic use , Motor Activity/drug effects , Neuralgia/drug therapy , Nitric Oxide Synthase Type I/metabolism , Posterior Horn Cells/drug effects , Spinal Nerves/drug effects , Aminobenzoates/pharmacology , Analgesics/toxicity , Animals , Disease Models, Animal , Esters/pharmacology , Male , Mice , Neuralgia/enzymology , Neuralgia/physiopathology , Neuronal Plasticity/drug effects , Posterior Horn Cells/enzymology , Protein Binding , Protein Interaction Domains and Motifs , Rats, Sprague-Dawley , Rotarod Performance Test , Signal Transduction , Spinal Nerves/enzymology , Spinal Nerves/physiopathology
2.
Am J Physiol Regul Integr Comp Physiol ; 314(5): R709-R715, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29384698

ABSTRACT

Intermittent spinal serotonin receptor activation elicits phrenic motor facilitation (pMF), a form of spinal respiratory motor plasticity. Episodic activation of either serotonin type 2 (5-HT2) or type 7 (5-HT7) receptors elicits pMF, although they do so via distinct cellular mechanisms known as the Q (5-HT2) and S (5-HT7) pathways to pMF. When coactivated, these pathways interact via mutual cross-talk inhibition. Although we have a rudimentary understanding of mechanisms mediating cross-talk interactions between spinal 5-HT2 subtype A (5-HT2A) and 5-HT7 receptor activation, we do not know if similar interactions exist between 5-HT2 subtype B (5-HT2B) and 5-HT7 receptors. We confirmed that either spinal 5-HT2B or 5-HT7 receptor activation alone elicits pMF and tested the hypotheses that 1) concurrent activation of both receptors suppresses pMF due to cross-talk inhibition; 2) 5-HT7 receptor inhibition of 5-HT2B receptor-induced pMF requires protein kinase A (PKA) activity; and 3) 5-HT2B receptor inhibition of 5-HT7 receptor-induced pMF requires NADPH oxidase (NOX) activity. Selective 5-HT2B and 5-HT7 receptor agonists were administered intrathecally at C4 (3 injections, 5-min intervals) to anesthetized, paralyzed, and ventilated rats. Whereas integrated phrenic nerve burst amplitude increased after selective spinal 5-HT2B or 5-HT7 receptor activation alone (i.e., pMF), pMF was no longer observed with concurrent 5-HT2B and 5-HT7 receptor agonist administration. With concurrent receptor activation, pMF was rescued by inhibiting either NOX or PKA activity, demonstrating their roles in cross-talk inhibition between these pathways to pMF. This report demonstrates cross-talk inhibition between 5-HT2B- and 5-HT7 receptor-induced pMF and that NOX and PKA activity are necessary for that cross-talk inhibition.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Diaphragm/innervation , Long-Term Potentiation , NADPH Oxidases/metabolism , Phrenic Nerve/metabolism , Receptor Cross-Talk , Receptor, Serotonin, 5-HT2B/metabolism , Receptors, Serotonin/metabolism , Spinal Nerves/enzymology , Action Potentials , Animals , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Long-Term Potentiation/drug effects , Male , NADPH Oxidases/antagonists & inhibitors , Phrenic Nerve/drug effects , Protein Kinase Inhibitors/pharmacology , Rats, Sprague-Dawley , Receptor Cross-Talk/drug effects , Receptor, Serotonin, 5-HT2B/drug effects , Receptors, Serotonin/drug effects , Respiration , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Signal Transduction , Spinal Nerves/drug effects , Time Factors
3.
Brain Res ; 1652: 62-70, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27671501

ABSTRACT

In dorsal root ganglia (DRG), satellite glial cells (SGCs) tightly ensheathe the somata of primary sensory neurons to form functional sensory units. SGCs are identified by their flattened and irregular morphology and expression of a variety of specific marker proteins. In this report, we present evidence that the 3-hydroxy-3-methylglutaryl coenzyme A synthase isoenzymes 1 and 2 (HMGCS1 and HMGCS2) are abundantly expressed in SGCs. Immunolabeling with the validated antibodies revealed that both HMGCS1 and HMGCS2 are highly colabeled with a selection of SGC markers, including GS, GFAP, Kir4.1, GLAST1, GDNF, and S100 but not with microglial cell marker Iba1, myelin sheath marker MBP, and neuronal marker ß3-tubulin or phosphorylated CaMKII. HMGCS1 but not HMGCS2 immunoreactivity in SGCs is reduced in the fifth lumbar (L5) DRGs that contain axotomized neurons following L5 spinal nerve ligation (SNL) in rats. Western blot showed that HMGCS1 protein level in axotomized L5 DRGs is reduced after SNL to 66±8% at 3 days (p<0.01, n=4 animals in each group) and 58±13% at 28 days (p<0.001, n=9 animals in each group) of its level in control samples, whereas HMGCS2 protein was comparable between injured and control DRGs. These results identify HMGCSs as the alternative markers for SGCs in DRGs. Downregulated HMGCS1 expression in DRGs after spinal nerve injury may reflect a potential role of abnormal sterol metabolism of SGCs in the nerve injured-induced neuropathic pain.


Subject(s)
Ganglia, Spinal/enzymology , Hydroxymethylglutaryl-CoA Synthase/metabolism , Neuralgia/enzymology , Peripheral Nerve Injuries/enzymology , Satellite Cells, Perineuronal/enzymology , Spinal Nerves/injuries , Animals , Disease Models, Animal , Ganglia, Spinal/injuries , Ganglia, Spinal/pathology , Hydroxymethylglutaryl-CoA Synthase/genetics , Immunoblotting , Immunohistochemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Lumbar Vertebrae , Male , Neuralgia/pathology , Peripheral Nerve Injuries/pathology , Rats, Sprague-Dawley , Satellite Cells, Perineuronal/pathology , Spinal Nerves/enzymology , Spinal Nerves/pathology
4.
Pain Med ; 17(2): 220-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26361083

ABSTRACT

OBJECTIVE: Heme oxygenase-1 (HO-1) exerts protective effects against ischemia and inflammation in the central nervous system. However, its role in neuropathic pain is still unclear. This study was undertaken to explore the distribution and possible mechanism of HO-1 in a mouse model of peripheral nerve injury. DESIGN AND METHODS: The experiment was conducted using a mouse model of L5 spinal nerve ligation (SNL). Mice received repeated intraperitoneal injection of Carbon monoxide-releasing molecule-2 (CO-RM-2), HO-1 inducer cobalt protoporphyrin IX (CoPP) or single intraspinal injection of lentivirus (LV) over-expressing HO-1. The behavior analyses were conducted. The distribution and expression of HO-1 in the spinal cord were analyzed. RESULTS: HO-1 but not HO-2 was upregulated in spinal cord microglia cells after nerve injury, and the repeated intraperitoneal administration of CORM-2 (10 mg/kg/d) or CoPP (5 mg/kg/d) both significantly reduced the mechanical allodynia and thermal hyperalgesia induced by SNL (P < 0.01). Intraspinal injection of LV-HO-1 persistently suppresses SNL-induced neuropathic pain (P < 0.01 or P < 0.05), significantly induced the spinal HO-1 protein content (P < 0.01) and inhibited the microglia activation (P < 0.01) 7 days after SNL. CONCLUSION: HO-1 upregulation could elicit potent analgesic effects against neuropathic pain, which might partly be attributed to inhibition of spinal microglia activation. HO-1 signaling pathway may present a novel strategy for the treatment of neuropathic pain.


Subject(s)
Disease Models, Animal , Heme Oxygenase-1/biosynthesis , Membrane Proteins/biosynthesis , Neuralgia/enzymology , Neuralgia/prevention & control , Pain Measurement/methods , Spinal Nerves/enzymology , Animals , Ligation/adverse effects , Male , Mice , Microglia/enzymology , Microglia/pathology , Neuralgia/pathology , Spinal Nerves/injuries , Spinal Nerves/pathology
5.
Anesth Analg ; 121(2): 545-55, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26076389

ABSTRACT

BACKGROUND: There has recently been a substantial increase in the survival of prematurely born neonates and an increase of in utero surgeries. Noxious stimulation in the newborn alters the pain response to injury in adult life. Progesterone, an effective antihyperalgesic agent in the adult, is at high concentration in the pregnant mother. Therefore, we investigated the effects of early-life progesterone on postsurgical outcomes in adult rats. METHODS: Female rat pups were administered progesterone or vehicle during the first 7 days postpartum (P1-P7). A second control group had no injections. Half of each of these groups received an incision of the hindpaw at P3 and the other half did not. At P60, all groups of these now adult rats received a second paw incision. Tactile sensitivity and thermal sensitivity were measured weekly at P14-P42 (period I), at P60 (just before the second incision), and every 2 days of P61-P70 (period II). At P67, rats were fixed by systemic paraformaldehyde perfusion and their spinal cords taken for staining and immunocytochemical analysis of activated p-p38 mitogen-activated protein kinase. RESULTS: Rats with surgery at P3 had greater tactile and thermal hyperalgesia in period I than the nonoperated rats, a difference abolished by progesterone treatment. P3 incision also resulted in long-lasting tactile and thermal hyperalgesia after the P60 incision (period II), both of which were markedly smaller in degree and faster to resolve in rats receiving early progesterone. Even in rats that were not operated on in period I, neonatal progesterone lessened the tactile hyperalgesia in period II. More spinal cells showed p-p38 staining in vehicle-treated rats as a result of the early-life incision but not in those treated with progesterone. CONCLUSIONS: These findings suggest that endogenously high progesterone in utero may have a similarly protective action and that the development of nociceptive circuitry can be strongly influenced by neonatal progesterone.


Subject(s)
Analgesics/administration & dosage , Hyperalgesia/prevention & control , Pain, Postoperative/prevention & control , Progesterone/administration & dosage , Age Factors , Animals , Animals, Newborn , Disease Models, Animal , Drug Administration Schedule , Female , Hyperalgesia/diagnosis , Hyperalgesia/enzymology , Hyperalgesia/physiopathology , Nociception/drug effects , Pain Measurement , Pain, Postoperative/diagnosis , Pain, Postoperative/enzymology , Pain, Postoperative/physiopathology , Phosphorylation , Rats, Sprague-Dawley , Sex Factors , Spinal Nerves/drug effects , Spinal Nerves/enzymology , Spinal Nerves/physiopathology , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Am J Physiol Regul Integr Comp Physiol ; 308(8): R700-7, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25673781

ABSTRACT

In many neural networks, mechanisms of compensatory plasticity respond to prolonged reductions in neural activity by increasing cellular excitability or synaptic strength. In the respiratory control system, a prolonged reduction in synaptic inputs to the phrenic motor pool elicits a TNF-α- and atypical PKC-dependent form of spinal plasticity known as inactivity-induced phrenic motor facilitation (iPMF). Although iPMF may be elicited by a prolonged reduction in respiratory neural activity, iPMF is more efficiently induced when reduced respiratory neural activity (neural apnea) occurs intermittently. Mechanisms giving rise to iPMF following intermittent neural apnea are unknown. The purpose of this study was to test the hypothesis that iPMF following intermittent reductions in respiratory neural activity requires spinal TNF-α and aPKC. Phrenic motor output was recorded in anesthetized and ventilated rats exposed to brief intermittent (5, ∼1.25 min), brief sustained (∼6.25 min), or prolonged sustained (30 min) neural apnea. iPMF was elicited following brief intermittent and prolonged sustained neural apnea, but not following brief sustained neural apnea. Unlike iPMF following prolonged neural apnea, spinal TNF-α was not required to initiate iPMF during intermittent neural apnea; however, aPKC was still required for its stabilization. These results suggest that different patterns of respiratory neural activity induce iPMF through distinct cellular mechanisms but ultimately converge on a similar downstream pathway. Understanding the diverse cellular mechanisms that give rise to inactivity-induced respiratory plasticity may lead to development of novel therapeutic strategies to treat devastating respiratory control disorders when endogenous compensatory mechanisms fail.


Subject(s)
Hypocapnia/enzymology , Neuronal Plasticity , Neurons/enzymology , Phrenic Nerve/enzymology , Protein Kinase C/metabolism , Respiratory Center/enzymology , Respiratory Muscles/innervation , Signal Transduction , Spinal Nerves/enzymology , Tumor Necrosis Factor-alpha/metabolism , Action Potentials , Animals , Disease Models, Animal , Hypercapnia/enzymology , Hypercapnia/physiopathology , Hypocapnia/blood , Hypocapnia/physiopathology , Male , Phrenic Nerve/physiopathology , Rats, Sprague-Dawley , Respiratory Center/physiopathology , Spinal Nerves/physiopathology , Time Factors
7.
Eur J Pain ; 18(5): 659-70, 2014 May.
Article in English | MEDLINE | ID: mdl-25493301

ABSTRACT

Background: Pulsed radiofrequency (PRF) has been widely used to treat chronic pain, but the effectiveness and mechanisms in preventing early neuropathic pain have not been well explored. Even fewer knowledge is available in its impact on glia-mediated nociceptive sensitization. This study aims to elucidate the modulation of PRF on nerve injury-induced pain development and activation of spinal mitogen-activated protein kinases (MAPKs). Methods: In a rat spinal nerve ligation (SNL) model, a low-volt PRF treatment was applied to the L5 dorsal root ganglion after nerve injury. Nociceptive behaviours were measured by von Frey and heat withdrawal tests at multiple time points. MAPK activations, including p-ERK and p-p38, as well as TNF-á level in the spinal dorsal horn were assessed and the cell types that expressed MAPK activation were identified by double immuno fluorescence staining.Results: We found that SNL promptly induced neuropathic pain in the affected hind limb for over 1 week as well as increased p-ERK and p-p38 in the spinal dorsal horn. PRF significantly attenuated SNL-induced mechanical allodynia and thermal hyperalgesia for 5­7 days. PRF also inhibited ERK and p38 activations, which were found majorly located within neurons and microglia, respectively. Besides, PRF significantly suppressed expression of TNF-á in the spinal dorsal horn throughout the course. Conclusions: Low-volt PRF significantly ameliorated SNL-induced acute pain. Inferentially, PRF may inhibit spinal sensitization by down-regulating spinal MAPK activations and activation-mediated cytokine release.We demonstrated that early PRF treatment in acute nerve injury helps to ameliorate neuropathic pain development.


Subject(s)
Hyperalgesia/prevention & control , Mitogen-Activated Protein Kinases/metabolism , Neuralgia/enzymology , Neuralgia/therapy , Pulsed Radiofrequency Treatment , Spinal Nerves/enzymology , Spinal Nerves/radiation effects , Acute Disease , Animals , Behavior, Animal , Disease Models, Animal , Down-Regulation/radiation effects , Enzyme Activation/radiation effects , Ganglia, Spinal/radiation effects , Immunohistochemistry , Ligation , Male , Neuroglia/radiation effects , Nociception/radiation effects , Pain Measurement , Random Allocation , Rats , Rats, Sprague-Dawley , Spinal Nerves/injuries , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Int J Clin Exp Pathol ; 5(9): 948-55, 2012.
Article in English | MEDLINE | ID: mdl-23119112

ABSTRACT

BACKGROUND: Neuropathic pain remains a prevalent and persistent clinical problem due to incomplete understanding of its pathogenesis. OBJECTIVE: The present study aimed to investigate the role of caspase-3 in the neuropathic pain in rats with chronic constriction injury (CCI). METHODS: SD rats were randomly assigned four groups (n=18 per group): sham group, normal saline group (NS group), Z-DEVD-FMK group (DEVD group) and RNA interference group (siRNA group). Z-DEVD-FMK (1 U/30 µl), siRNA targeting caspase-3 (10 µg/30 µl) and NS of equal volume were intrathecally administered once daily for 5 days starting 1 day before surgery in the DEVD, siRNA and NS group, respectively. Thermal hyperalgesia was assessed at one day before and 1, 2, 4, 5, 6, 7 and 10 days after surgery. The mRNA and protein expressions of caspase-3 were measured by real time PCR and immunofluorescence assay. Apoptosis was detected by TUNEL staining. GAP-43 expression was measured by immunofluorescence and western blot assays. RESULTS: The right paw withdrawal latency (PWL) was decreased after CCI (P<0.05). TUNEL-positive neurons and the mRNA and protein expressions of caspase-3 in the spinal cord were increased significantly. After Z-DEVD-FMK or siRNA treatment, TUNEL-positive neurons were decreased, PWLs increased (P<0.05) and the mRNA and protein expressions of caspase-3 decreased. The expression of GAP-43, a sprouting related protein, was decreased in the DEVD and siRNA group as compared to NS group (P<0.05). Up-regulation of GAP-43 following CCI was decreased following caspase-3 inhibition. Following sciatic nerve ligation, the gene expression, translation and transcription are significantly changed in the neurons which finally results in neuron apoptosis. The neuron apoptosis induce the up-regulation of GAP-43 expression leading to hyperalgesia. CONCLUSION: Caspase-3 mediated neuron apoptosis is probably responsible for the neuropathic pain in CCI rats. Inhibition of caspase-3 may serve as a treatment of neuropathic pain.


Subject(s)
Caspase 3/metabolism , Caspase Inhibitors/pharmacology , GAP-43 Protein/metabolism , Neuralgia/prevention & control , Oligopeptides/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Spinal Nerves/drug effects , Animals , Apoptosis/drug effects , Caspase 3/genetics , Caspase Inhibitors/administration & dosage , Disease Models, Animal , Down-Regulation , Fluorescent Antibody Technique , Hyperalgesia/enzymology , Hyperalgesia/genetics , Hyperalgesia/physiopathology , Hyperalgesia/prevention & control , In Situ Nick-End Labeling , Injections, Spinal , Male , Neuralgia/enzymology , Neuralgia/genetics , Neuralgia/pathology , Neuralgia/physiopathology , Oligopeptides/administration & dosage , Pain Measurement , Pain Threshold/drug effects , RNA, Messenger/metabolism , RNA, Small Interfering/administration & dosage , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Real-Time Polymerase Chain Reaction , Spinal Nerves/enzymology , Spinal Nerves/pathology , Spinal Nerves/physiopathology , Time Factors
9.
Mol Pain ; 8: 46, 2012 Jun 19.
Article in English | MEDLINE | ID: mdl-22713297

ABSTRACT

BACKGROUND: The plasma membrane Ca2+-ATPase (PMCA) is the principal means by which sensory neurons expel Ca2+ and thereby regulate the concentration of cytoplasmic Ca2+ and the processes controlled by this critical second messenger. We have previously found that painful nerve injury decreases resting cytoplasmic Ca2+ levels and activity-induced cytoplasmic Ca2+ accumulation in axotomized sensory neurons. Here we examine the contribution of PMCA after nerve injury in a rat model of neuropathic pain. RESULTS: PMCA function was isolated in dissociated sensory neurons by blocking intracellular Ca2+ sequestration with thapsigargin, and cytoplasmic Ca2+ concentration was recorded with Fura-2 fluorometry. Compared to control neurons, the rate at which depolarization-induced Ca2+ transients resolved was increased in axotomized neurons after spinal nerve ligation, indicating accelerated PMCA function. Electrophysiological recordings showed that blockade of PMCA by vanadate prolonged the action potential afterhyperpolarization, and also decreased the rate at which neurons could fire repetitively. CONCLUSION: We found that PMCA function is elevated in axotomized sensory neurons, which contributes to neuronal hyperexcitability. Accelerated PMCA function in the primary sensory neuron may contribute to the generation of neuropathic pain, and thus its modulation could provide a new pathway for peripheral treatment of post-traumatic neuropathic pain.


Subject(s)
Axotomy , Cell Membrane/enzymology , Neuralgia/enzymology , Neuralgia/pathology , Plasma Membrane Calcium-Transporting ATPases/metabolism , Sensory Receptor Cells/enzymology , Spinal Nerves/pathology , Action Potentials/drug effects , Animals , Calcium/metabolism , Cell Membrane/drug effects , Cell Size/drug effects , Enzyme Activation/drug effects , Male , Mitochondria/drug effects , Mitochondria/metabolism , Neuralgia/physiopathology , Plasma Membrane Calcium-Transporting ATPases/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/pathology , Sodium-Calcium Exchanger/metabolism , Spinal Nerves/drug effects , Spinal Nerves/enzymology , Spinal Nerves/physiopathology , Thapsigargin/pharmacology
10.
Pain ; 153(6): 1263-1273, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22482911

ABSTRACT

Injuries can induce adaptations in pain processing that result in amplification of signaling. One mechanism may be analogous to long-term potentiation and involve the atypical protein kinase C, PKMζ. The possible contribution of PKMζ-dependent and independent amplification mechanisms to experimental neuropathic pain was explored in rats with spinal nerve ligation (SNL) injury. SNL increased p-PKMζ in the rostral anterior cingulate cortex (rACC), a site that mediates, in part, the unpleasant aspects of pain. Inhibition of PKMζ within the rACC by a single administration of ζ-pseudosubstrate inhibitory peptide (ZIP) reversed SNL-induced aversiveness within 24 hours, whereas N-methyl-d-aspartate receptor blockade with MK-801 had no effects. The SNL-induced aversive state (reflecting "spontaneous" pain), was re-established in a time-dependent manner, with full recovery observed 7 days post-ZIP administration. Neither rACC ZIP nor MK-801 altered evoked responses. In contrast, spinal ZIP or MK-801, but not scrambled peptide, transiently reversed evoked hypersensitivity, but had no effect on nerve injury-induced spontaneous pain. PKMζ phosphorylation was not altered by SNL in the spinal dorsal horn. These data suggest that amplification mechanisms contribute to different aspects of neuropathic pain at different levels of the neuraxis. Thus, PKMζ-dependent amplification contributes to nerve injury-induced aversiveness within the rACC. Moreover, unlike mechanisms maintaining memory, the consequences of PKMζ inhibition within the rACC are not permanent in neuropathic pain, possibly reflecting the re-establishment of amplification mechanisms by ongoing activity of injured nerves. In the spinal cord, however, both PKMζ-dependent and independent mechanisms contribute to amplification of evoked responses, but apparently not spontaneous pain.


Subject(s)
Gyrus Cinguli/enzymology , Neuralgia/metabolism , Protein Kinase C/metabolism , Signal Transduction/physiology , Spinal Cord/enzymology , Animals , Dizocilpine Maleate/pharmacology , Male , Neuralgia/physiopathology , Peptides/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Spinal Nerves/enzymology , Spinal Nerves/injuries
11.
Bioorg Med Chem Lett ; 22(7): 2510-3, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22370270

ABSTRACT

A novel class of 1,7-disubstituted 2,3,4,5-tetrahydro-1H-benzo[b]azepine derivatives was designed, synthesized and evaluated as human nitric oxide synthase (NOS) inhibitors. Structure-activity relationship studies based on various basic amine side chains attached at the 1-position of the 2,3,4,5-tetrahydro-1H-benzo[b]azepine ring led to the identification of several potent and highly selective inhibitors (17, 18, 25, (±)-39, and (±)-40) of human neuronal NOS. The potential therapeutic application of one of these new selective nNOS inhibitors (17) was demonstrated in an in vivo spinal nerve ligation model of neuropathic pain, and various in vitro safety pharmacology studies such as the hERG K(+) channel inhibition assay and high throughput broad screen (minimal activity at 79 receptors/transporters/ion channels).


Subject(s)
Analgesics/chemical synthesis , Benzazepines/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Neuralgia/drug therapy , Nitric Oxide Synthase Type I/antagonists & inhibitors , Analgesics/administration & dosage , Analgesics/therapeutic use , Animals , Benzazepines/administration & dosage , Benzazepines/therapeutic use , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/therapeutic use , High-Throughput Screening Assays , Humans , Mice , Neuralgia/enzymology , Neuralgia/physiopathology , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Recombinant Proteins/metabolism , Spinal Nerves/drug effects , Spinal Nerves/enzymology , Spinal Nerves/physiopathology , Stereoisomerism , Structure-Activity Relationship
12.
Mol Pain ; 7: 50, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21756313

ABSTRACT

BACKGROUND: Several studies have investigated the involvement of nitric oxide (NO) in acute and chronic pain using mice lacking a single NO synthase (NOS) gene among the three isoforms: neuronal (nNOS), inducible (iNOS) and endothelial (eNOS). However, the precise role of NOS/NO in pain states remains to be determined owing to the substantial compensatory interactions among the NOS isoforms. Therefore, in this study, we used mice lacking all three NOS genes (n/i/eNOS-/-mice) and investigated the behavioral phenotypes in a series of acute and chronic pain assays. RESULTS: In a model of tissue injury-induced pain, evoked by intraplantar injection of formalin, both iNOS-/-and n/i/eNOS-/-mice exhibited attenuations of pain behaviors in the second phase compared with that in wild-type mice. In a model of neuropathic pain, nerve injury-induced behavioral and cellular responses (tactile allodynia, spinal microglial activation and Src-family kinase phosphorylation) were reduced in n/i/eNOS-/-but not iNOS-/-mice. Tactile allodynia after nerve injury was improved by acute pharmacological inhibition of all NOSs and nNOS. Furthermore, in MG-5 cells (a microglial cell-line), interferon-γ enhanced NOSs and Mac-1 mRNA expression, and the Mac-1 mRNA increase was suppressed by L-NAME co-treatment. Conversely, the NO donor, sodium nitroprusside, markedly increased mRNA expression of Mac-1, interleukin-6, toll-like receptor 4 and P2X4 receptor. CONCLUSIONS: Our results provide evidence that the NOS/NO pathway contributes to behavioral pain responses evoked by tissue injury and nerve injury. In particular, nNOS may be important for spinal microglial activation and tactile allodynia after nerve injury.


Subject(s)
Microglia/pathology , Neuralgia/enzymology , Neuralgia/pathology , Nitric Oxide Synthase/deficiency , Spinal Cord/pathology , Spinal Nerves/injuries , Spinal Nerves/pathology , Animals , Behavior, Animal/drug effects , Gene Expression Regulation/drug effects , Gene Knockout Techniques , Hyperalgesia/complications , Hyperalgesia/pathology , Inflammation/complications , Inflammation/pathology , Interferon-gamma/pharmacology , Interleukin-6/genetics , Interleukin-6/metabolism , Macrophage-1 Antigen/genetics , Macrophage-1 Antigen/metabolism , Male , Mice , Microglia/drug effects , Neuralgia/complications , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Purinergic P2X4/genetics , Receptors, Purinergic P2X4/metabolism , Spinal Cord/drug effects , Spinal Nerves/drug effects , Spinal Nerves/enzymology , Temperature , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , src-Family Kinases/metabolism
13.
J Physiol ; 589(Pt 9): 2245-60, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21486770

ABSTRACT

Spinal motoneurons are highly vulnerable in amyotrophic lateral sclerosis (ALS).Previous research using a standard animal model, the mutant superoxide dismutase-1 (SOD1)mouse, has revealed deficits in many cellular properties throughout its lifespan. The electrical properties underlying motoneuron excitability are some of the earliest to change; starting at 1 week postnatal, persistent inward currents (PICs) mediated by Na+ are upregulated and electrical conductance, a measure of cell size, increases. However, during this period these properties and many others undergo large developmental changes which have not been fully analysed.Therefore, we undertook a systematic analysis of electrical properties in more than 100 normal and mutant SOD1 motoneurons from 0 to 12 days postnatal, the neonatal to juvenile period.We compared normal mice with the most severe SOD1 model, the G93A high-expressor line. We found that the Na+ PIC and the conductance increased during development. However, mutant SOD1 motoneurons showed much greater increases than normal motoneurons; the mean Na+PIC in SOD1 motoneurons was double that of wild-type motoneurons. Additionally, in mutant SOD1 motoneurons the PIC mediated by Ca2+ increased, spike width decreased and the time course of the after-spike after-hyperpolarization shortened. These changes were advances of the normal effects of maturation. Thus, our results show that the development of normal and mutant SOD1 motoneurons follows generally similar patterns, but that the rate of development is accelerated in the mutant SOD1 motoneurons. Statistical analysis of all measured properties indicates that approximately 55% of changes attributed to the G93A SOD1 mutation can be attributed to an increased rate of maturation.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Motor Neurons/enzymology , Spinal Nerves/physiopathology , Superoxide Dismutase/metabolism , Action Potentials , Aging , Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/genetics , Analysis of Variance , Animals , Animals, Newborn , Calcium/metabolism , Disease Models, Animal , Electric Conductivity , Genotype , Humans , Kinetics , Mice , Mice, Transgenic , Mutation , Patch-Clamp Techniques , Phenotype , Sodium/metabolism , Spinal Nerves/enzymology , Superoxide Dismutase/genetics , Superoxide Dismutase-1
14.
Mol Pain ; 5: 74, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-20021638

ABSTRACT

BACKGROUND: Neuropathic pain is a complex chronic pain generated by damage to, or pathological changes in the somatosensory nervous system. Characteristic features of neuropathic pain are allodynia, hyperalgesia and spontaneous pain. Such abnormalities associated with neuropathic pain state remain to be a significant clinical problem. However, the neuronal mechanisms underlying the pathogenesis of neuropathic pain are complex and still poorly understood. Casein kinase 1 is a serine/threonine protein kinase and has been implicated in a wide range of signaling activities such as cell differentiation, proliferation, apoptosis, circadian rhythms and membrane transport. In mammals, the CK1 family consists of seven members (alpha, beta, gamma1, gamma2, gamma3, delta, and epsilon) with a highly conserved kinase domain and divergent amino- and carboxy-termini. RESULTS: Preliminary cDNA microarray analysis revealed that the expression of the casein kinase 1 epsilon (CK1epsilon) mRNA in the spinal cord of the neuropathic pain-resistant N- type Ca2+ channel deficient (Cav2.2-/-) mice was decreased by the spinal nerve injury. The same injury exerted no effects on the expression of CK1epsilon mRNA in the wild-type mice. Western blot analysis of the spinal cord identified the downregulation of CK1epsilon protein in the injured Cav2.2-/- mice, which is consistent with the data of microarray analysis. However, the expression of CK1epsilon protein was found to be up-regulated in the spinal cord of injured wild-type mice. Immunocytochemical analysis revealed that the spinal nerve injury changed the expression profiles of CK1epsilon protein in the dorsal root ganglion (DRG) and the spinal cord neurons. Both the percentage of CK1epsilon-positive neurons and the expression level of CK1epsilon protein were increased in DRG and the spinal cord of the neuropathic mice. These changes were reversed in the spinal cord of the injured Cav2.2-/- mice. Furthermore, intrathecal administration of a CK1 inhibitor IC261 produced marked anti-allodynic and anti-hyperalgesic effects on the neuropathic mice. In addition, primary afferent fiber-evoked spinal excitatory responses in the neuropathic mice were reduced by IC261. CONCLUSIONS: These results suggest that CK1epsilon plays important physiological roles in neuropathic pain signaling. Therefore CK1epsilon is a useful target for analgesic drug development.


Subject(s)
Casein Kinase 1 epsilon/metabolism , Ganglia, Spinal/enzymology , Peripheral Nervous System Diseases/enzymology , Spinal Cord/enzymology , Spinal Nerves/enzymology , Spinal Nerves/injuries , Animals , Calcium Channels, N-Type/genetics , Casein Kinase 1 epsilon/antagonists & inhibitors , Casein Kinase 1 epsilon/genetics , Disease Models, Animal , Down-Regulation/genetics , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/physiopathology , Hyperalgesia/enzymology , Hyperalgesia/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuralgia/enzymology , Neuralgia/physiopathology , Nociceptors/enzymology , Organ Culture Techniques , Peripheral Nervous System Diseases/physiopathology , Posterior Horn Cells/enzymology , RNA, Messenger/metabolism , Spinal Cord/physiopathology , Spinal Nerve Roots/enzymology , Spinal Nerve Roots/physiopathology , Spinal Nerves/physiopathology , Up-Regulation/physiology
15.
J Neurosci ; 29(12): 3766-80, 2009 Mar 25.
Article in English | MEDLINE | ID: mdl-19321773

ABSTRACT

Fatty acid amide hydrolase (FAAH) is a degradative enzyme for a group of endogenous signaling lipids that includes anandamide (AEA). AEA acts as an endocannabinoid and an endovanilloid by activating cannabinoid and vanilloid type 1 transient receptor potential (TRPV1) receptors, respectively, on dorsal root ganglion (DRG) sensory neurons. Inhibition of FAAH activity increases AEA concentrations in nervous tissue and reduces sensory hypersensitivity in animal pain models. Using immunohistochemistry, Western blotting, and reverse transcription-PCR, we demonstrate the location of the FAAH in adult rat DRG, sciatic nerve, and spinal cord. In naive rats, FAAH immunoreactivity localized to the soma of 32.7 +/- 0.8% of neurons in L4 and L5 DRG. These were small-sized (mean soma area, 395.96 +/- 5.6 mum(2)) and predominantly colabeled with peripherin and isolectin B4 markers of unmyelinated C-fiber neurons; 68% colabeled with antibodies to TRPV1 (marker of nociceptive DRG neurons), and <2% colabeled with NF200 (marker of large myelinated neurons). FAAH-IR was also present in small, NF200-negative cultured rat DRG neurons. Incubation of these cultures with the FAAH inhibitor URB597 increased AEA-evoked cobalt uptake in a capsazepine-sensitive manner. After sciatic nerve axotomy, there was a rightward shift in the cell-size distribution of FAAH-immunoreactive (IR) DRG neurons ipsilateral to injury: FAAH immunoreactivity was detected in larger-sized cells that colabeled with NF200. An ipsilateral versus contralateral increase in both the size and proportion of FAAH-IR DRG occurred after spinal nerve transection injury but not after chronic inflammation of the rat hindpaw 2 d after injection of complete Freund's adjuvant. This study reveals the location of FAAH in neural tissue involved in peripheral nociceptive transmission.


Subject(s)
Amidohydrolases/metabolism , Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Ganglia, Spinal/enzymology , Spinal Nerves/injuries , Amidohydrolases/genetics , Animals , Freund's Adjuvant , Ganglia, Spinal/cytology , Immunohistochemistry , Inflammation/chemically induced , Inflammation/enzymology , Male , Pain/enzymology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sciatic Nerve/enzymology , Sciatic Nerve/injuries , Sensory Receptor Cells/enzymology , Spinal Nerves/enzymology
16.
Nat Med ; 14(3): 331-6, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18264108

ABSTRACT

Treatment of neuropathic pain, triggered by multiple insults to the nervous system, is a clinical challenge because the underlying mechanisms of neuropathic pain development remain poorly understood. Most treatments do not differentiate between different phases of neuropathic pain pathophysiology and simply focus on blocking neurotransmission, producing transient pain relief. Here, we report that early- and late-phase neuropathic pain development in rats and mice after nerve injury require different matrix metalloproteinases (MMPs). After spinal nerve ligation, MMP-9 shows a rapid and transient upregulation in injured dorsal root ganglion (DRG) primary sensory neurons consistent with an early phase of neuropathic pain, whereas MMP-2 shows a delayed response in DRG satellite cells and spinal astrocytes consistent with a late phase of neuropathic pain. Local inhibition of MMP-9 by an intrathecal route inhibits the early phase of neuropathic pain, whereas inhibition of MMP-2 suppresses the late phase of neuropathic pain. Further, intrathecal administration of MMP-9 or MMP-2 is sufficient to produce neuropathic pain symptoms. After nerve injury, MMP-9 induces neuropathic pain through interleukin-1beta cleavage and microglial activation at early times, whereas MMP-2 maintains neuropathic pain through interleukin-1beta cleavage and astrocyte activation at later times. Inhibition of MMP may provide a novel therapeutic approach for the treatment of neuropathic pain at different phases.


Subject(s)
Matrix Metalloproteinases/metabolism , Pain/enzymology , Spinal Nerves/enzymology , Analgesics/therapeutic use , Animals , Behavior, Animal , Cytokines/metabolism , Enzyme Activation , Enzyme Inhibitors , Gene Deletion , Gene Expression Regulation , Ligation , Male , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/genetics , Mice , Microglia , Neurons/metabolism , Pain/drug therapy , Rats , Rats, Sprague-Dawley , Up-Regulation
17.
Am J Physiol Regul Integr Comp Physiol ; 294(4): R1304-11, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18272663

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) is an excitatory neuropeptide present in the rat brain stem. The extent of its localization within catecholaminergic groups and bulbospinal sympathoexcitatory neurons is not established. Using immunohistochemistry and in situ hybridization, we determined the extent of any colocalization with catecholaminergic and/or bulbospinal projections from the brain stem was determined. PACAP mRNA was found in tyrosine hydroxylase-immunoreactive (TH-ir) neurons in the C1-C3 cell groups. In the rostral ventrolateral medulla (RVLM), PACAP mRNA was found in 84% of the TH-ir neurons and 82% of bulbospinal TH-ir neurons. The functional significance of these PACAP mRNA positive bulbospinal neurons was tested by intrathecal administration of PACAP-38 in anaesthetized rats. Splanchnic sympathetic nerve activity doubled (110%) and heart rate rose significantly (19%), although blood pressure was unaffected. In addition, as previously reported, PACAP was found in the A1 cell group but not in the A5 cell group or in the locus coeruleus. The RVLM is the primary site responsible for the tonic and reflex control of blood pressure through the activity of bulbospinal presympathetic neurons, the majority of which contain TH. The results indicate 1) that pontomedullary neurons containing both TH and PACAP that project to the intermediolateral cell column originate from C1-C3 and not A5, and 2) intrathecal PACAP-38 causes a prolonged, sympathoexcitatory effect.


Subject(s)
Baroreflex , Brain Stem/metabolism , Cardiovascular System/innervation , Nerve Fibers, Unmyelinated/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Spinal Nerves/metabolism , Sympathetic Nervous System/metabolism , Animals , Blood Pressure , Brain Stem/enzymology , Denervation , Heart Rate , Injections, Spinal , Locus Coeruleus/metabolism , Male , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Unmyelinated/enzymology , Neural Pathways/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/administration & dosage , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Spinal Nerves/enzymology , Splanchnic Nerves/metabolism , Sympathetic Nervous System/enzymology , Time Factors , Tyrosine 3-Monooxygenase/metabolism
18.
J Neurochem ; 103(4): 1408-16, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17725579

ABSTRACT

Activation of P2X(3) and P2X(2/3) receptors (P2X(3)R/P2X(2/3)R), ionotropic ATP receptor subtypes, in primary sensory neurons is involved in neuropathic pain, a debilitating chronic pain that occurs after peripheral nerve injury. However, the underlying mechanisms remain unknown. We investigated the role of cytosolic phospholipase A(2) (cPLA(2)) as a downstream molecule that mediates the P2X(3)R/P2X(2/3)R-dependent neuropathic pain. We found that applying ATP to cultured dorsal root ganglion (DRG) neurons increased the level of Ser505-phosphorylated cPLA(2) and caused translocation of Ser505-phosphorylated cPLA(2) to the plasma membrane. The ATP-induced cPLA(2) activation was inhibited by a selective antagonist of P2X(3)R/P2X(2/3)R and by a selective inhibitor of cPLA(2). In the DRG in vivo, the number of cPLA(2)-activated neurons was strikingly increased after peripheral nerve injury but not after peripheral inflammation produced by complete Freund's adjuvant. Pharmacological blockade of P2X(3)R/P2X(2/3)R reversed the nerve injury-induced cPLA(2) activation in DRG neurons. Moreover, administering the cPLA(2) inhibitor near the DRG suppressed nerve injury-induced tactile allodynia, a hallmark of neuropathic pain. Our results suggest that P2X(3)R/P2X(2/3)R-dependent cPLA(2) activity in primary sensory neurons is a key event in neuropathic pain and that cPLA(2) might be a potential target for treating neuropathic pain.


Subject(s)
Neurons, Afferent/enzymology , Pain/enzymology , Phospholipases A2, Cytosolic/metabolism , Receptors, Purinergic P2/physiology , Animals , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Male , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Pain/drug therapy , Pain/metabolism , Pain Measurement/drug effects , Pain Measurement/methods , Phospholipases A2, Cytosolic/antagonists & inhibitors , Rats , Rats, Wistar , Receptors, Purinergic P2X2 , Receptors, Purinergic P2X3 , Spinal Nerves/drug effects , Spinal Nerves/enzymology , Spinal Nerves/injuries , Spinal Nerves/metabolism
19.
Anesthesiology ; 107(2): 312-21, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17667577

ABSTRACT

BACKGROUND: Current evidence indicates that p38 mitogen-activated protein kinase activation in spinal microglia contributes to the development of neuropathic pain. However, how nerve injury activates p38 in spinal microglia is incompletely unknown. Nerve injury-induced ectopic spontaneous activity is essential for the generation of neuropathic pain. The authors examined whether peripheral neural activity is necessary for p38 activation in spinal microglia. METHODS: To examine whether spinal microglia activation depends on peripheral activity in the rat spared nerve injury (SNI) model, the authors blocked conduction in the sciatic nerve before or 2 days after SNI. The block was produced by applying bupivacaine-loaded microspheres above the nerve injury site. The p38 activation was examined by p38 phosphorylation using a phosphorylated p38 antibody, and neuropathic pain-related behavior was evaluated before and after intrathecal infusion of a p38 inhibitor. RESULTS: Three days after SNI, there was a marked p38 activation in the medial two thirds of the dorsal horn, where the injured tibial and peroneal nerves terminated and where isolectin B4 staining was lost. Phosphorylated p38 was only colocalized with the microglial surface marker OX-42, indicating a microglial localization of phosphorylated p38 in the SNI model. Bupivacaine microspheres produced persistent block (loss of sensory and motor function) of the sciatic nerve for the whole period of the study (3 days). This blockade prevented but did not reverse p38 activation in spinal microglia. Intrathecal infusion of the p38 inhibitor FR167653 prevented and reversed mechanical allodynia on post-SNI day 3. CONCLUSIONS: After nerve injury, activity in the peripheral nerve is required for the induction but not the maintenance of p38 activation in spinal microglia.


Subject(s)
Microglia/drug effects , Neural Conduction/drug effects , Neuralgia/prevention & control , Sciatic Nerve/drug effects , Spinal Nerves/injuries , p38 Mitogen-Activated Protein Kinases/drug effects , Anesthetics, Local/administration & dosage , Animals , Behavior, Animal/drug effects , Blotting, Western/methods , Bupivacaine/administration & dosage , Disease Models, Animal , Enzyme Activation/drug effects , Injections, Spinal , Male , Microglia/enzymology , Microspheres , Nerve Block/methods , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Sciatic Nerve/physiopathology , Spinal Nerves/drug effects , Spinal Nerves/enzymology , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Brain Behav Immun ; 21(5): 642-51, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17174525

ABSTRACT

Accumulating evidence suggests that microglial cells in the spinal cord play an important role in the development of neuropathic pain. However, it remains largely unknown how glia interact with neurons in the spinal cord after peripheral nerve injury. Recent studies suggest that the chemokine fractalkine may mediate neural/microglial interaction via its sole receptor CX3CR1. We have examined how fractalkine activates microglia in a neuropathic pain condition produced by spinal nerve ligation (SNL). SNL induced an upregulation of CX3CR1 in spinal microglia that began on day 1, peaked on day 3, and maintained on day 10. Intrathecal injection of a neutralizing antibody against CX3CR1 suppressed not only mechanical allodynia but also the activation of p38 MAPK in spinal microglia following SNL. Conversely, intrathecal infusion of fractalkine produced a marked p38 activation and mechanical allodynia. SNL also induced a dramatic reduction of the membrane-bound fractalkine in the dorsal root ganglion, suggesting a cleavage and release of this chemokine after nerve injury. Finally, application of fractalkine to spinal slices did not produce acute facilitation of excitatory synaptic transmission in lamina II dorsal horn neurons, arguing against a direct action of fractalkine on spinal neurons. Collectively, our data suggest that (a) fractalkine cleavage (release) after nerve injury may play an important role in neural-glial interaction, and (b) microglial CX3CR1/p38 MAPK pathway is critical for the development of neuropathic pain.


Subject(s)
Chemokines, CX3C/metabolism , Membrane Proteins/metabolism , Microglia/enzymology , Neuralgia/enzymology , Receptors, Chemokine/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Analysis of Variance , Animals , CX3C Chemokine Receptor 1 , Chemokine CX3CL1 , Disease Models, Animal , Ligation , Male , Pain Threshold/physiology , Posterior Horn Cells/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Spinal Cord/cytology , Spinal Cord/enzymology , Spinal Nerves/enzymology , Spinal Nerves/injuries , Statistics, Nonparametric
SELECTION OF CITATIONS
SEARCH DETAIL
...