Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000275

ABSTRACT

In tumor cells, interleukin-6 (IL-6) signaling can lead to activation of the epidermal growth factor receptor (EGFR), which prolongs Stat3 activation. In the present experiments, we tested the hypothesis that IL-6 signaling activates EGFR signaling in peripheral and spinal nociception and examined whether EGFR localization and activation coincide with pain-related behaviors in arthritis. In vivo in anesthetized rats, spinal application of the EGFR receptor blocker gefitinib reduced the responses of spinal cord neurons to noxious joint stimulation, but only after spinal pretreatment with IL-6 and soluble IL-6 receptor. Using Western blots, we found that IL-6-induced Stat3 activation was reduced by gefitinib in microglial cells of the BV2 cell line, but not in cultured DRG neurons. Immunohistochemistry showed EGFR localization in most DRG neurons from normal rats, but significant downregulation in the acute and most painful arthritis phase. In the spinal cord of mice, EGFR was highly activated mainly in the chronic phase of inflammation, with localization in neurons. These data suggest that spinal IL-6 signaling may activate spinal EGFR signaling. Downregulation of EGFR in DRG neurons in acute arthritis may limit nociception, but pronounced delayed activation of EGFR in the spinal cord may be involved in chronic inflammatory pain.


Subject(s)
ErbB Receptors , Interleukin-6 , Sensory Receptor Cells , Spinal Cord , Animals , Female , Mice , Rats , Arthritis/metabolism , Arthritis, Experimental/metabolism , Cell Line , ErbB Receptors/metabolism , Ganglia, Spinal/metabolism , Gefitinib/pharmacology , Interleukin-6/metabolism , Receptors, Interleukin-6/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Signal Transduction , Spinal Cord/metabolism , STAT3 Transcription Factor/metabolism
2.
Br J Pharmacol ; 180(4): 441-458, 2023 02.
Article in English | MEDLINE | ID: mdl-36245399

ABSTRACT

BACKGROUND AND PURPOSE: Prostaglandin E2 is considered a major mediator of inflammatory pain, by acting on neuronal Gs protein-coupled EP2 and EP4 receptors. However, the neuronal EP3 receptor, colocalized with EP2 and EP4 receptor, is Gi protein-coupled and antagonizes the pronociceptive prostaglandin E2 effect. Here, we investigated the cellular signalling mechanisms by which the EP3 receptor reduces EP2 and EP4 receptor-evoked pronociceptive effects in sensory neurons. EXPERIMENTAL APPROACH: Experiments were performed on isolated and cultured dorsal root ganglion (DRG) neurons from wild type, phosphoinositide 3-kinase γ (PI3Kγ)-/- , and PI3Kγkinase dead (KD)/KD mice. For subtype-specific stimulations, we used specific EP2, EP3, and EP4 receptor agonists from ONO Pharmaceuticals. As a functional readout, we recorded TTX-resistant sodium currents in patch-clamp experiments. Western blots were used to investigate the activation of intracellular signalling pathways. EP4 receptor internalization was measured using immunocytochemistry. KEY RESULTS: Different pathways mediate the inhibition of EP2 and EP4 receptor-dependent pronociceptive effects by EP3 receptor stimulation. Inhibition of EP2 receptor-evoked pronociceptive effect critically depends on the kinase-independent function of the signalling protein PI3Kγ, and adenosine monophosphate activated protein kinase (AMPK) is involved. By contrast, inhibition of EP4 receptor-evoked pronociceptive effect is independent on PI3Kγ and mediated through activation of G protein-coupled receptor kinase 2 (GRK2), which enhances the internalization of the EP4 receptor after ligand binding. CONCLUSION AND IMPLICATIONS: Activation of neuronal PI3Kγ, AMPK, and GRK2 by EP3 receptor activation limits cAMP-dependent pain generation by prostaglandin E2 . These new insights hold the potential for a novel approach in pain therapy.


Subject(s)
AMP-Activated Protein Kinases , Prostaglandins , Animals , Mice , Phosphatidylinositol 3-Kinase , Phosphatidylinositol 3-Kinases , Dinoprostone/pharmacology , Dinoprostone/metabolism , Receptors, Prostaglandin E, EP4 Subtype , Receptors, Prostaglandin E, EP2 Subtype , Sensory Receptor Cells/metabolism , Pain , Analgesics , Receptors, Prostaglandin E, EP3 Subtype/metabolism
3.
J Neurochem ; 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36520021

ABSTRACT

Diseases of joints are among the most frequent causes of chronic pain. In the course of joint diseases, the peripheral and the central nociceptive system develop persistent hyperexcitability (peripheral and central sensitization). This review addresses the mechanisms of spinal sensitization evoked by arthritis. Electrophysiological recordings in anesthetized rats from spinal cord neurons with knee input in a model of acute arthritis showed that acute spinal sensitization is dependent on spinal glutamate receptors (AMPA, NMDA, and metabotropic glutamate receptors) and supported by spinal actions of neuropeptides such as neurokinins and CGRP, by prostaglandins, and by proinflammatory cytokines. In several chronic arthritis models (including immune-mediated arthritis and osteoarthritis) spinal glia activation was observed to be coincident with behavioral mechanical hyperalgesia which was attenuated or prevented by intrathecal application of minocycline, fluorocitrate, and pentoxyfylline. Some studies identified specific pathways of micro- and astroglia activation such as the purinoceptor- (P2 X7 -) cathepsin S/CX3 CR1 pathway, the mobility group box-1 protein (HMGB1), and toll-like receptor 4 (TLR4) activation, spinal NFκB/p65 activation and others. The spinal cytokines TNF, interleukin-6, interleukin-1ß, and others form a functional spinal network characterized by an interaction between neurons and glia cells which is required for spinal sensitization. Neutralization of spinal cytokines by intrathecal interventions attenuates mechanical hyperalgesia. This effect may in part result from local suppression of spinal sensitization and in part from efferent effects which attenuate the inflammatory process in the joint. In summary, arthritis evokes significant spinal hyperexcitability which is likely to contribute to the phenotype of arthritis pain in patients.

5.
J Neurochem ; 158(4): 898-911, 2021 08.
Article in English | MEDLINE | ID: mdl-34050952

ABSTRACT

Both spinal tumor necrosis factor (TNF) and interleukin-6 (IL-6) contribute to the development of "mechanical" spinal hyperexcitability in inflammatory pain states. Recently, we found that spinal sensitization by TNF was significantly reduced by blockade of spinal IL-6 signaling suggesting that IL-6 signaling is involved in spinal TNF effects. Here, we explored whether spinal interleukin-1ß (IL-1ß), also implicated in inflammatory pain, induces "mechanical" spinal hyperexcitability, and whether spinal IL-1ß effects are related to TNF and IL-6 effects. We recorded the responses of spinal cord neurons to mechanical stimulation of the knee joint in vivo and used cellular approaches on microglial and astroglial cell lines to identify interactions of IL-1ß, TNF, and IL-6. Spinal application of IL-1ß in anesthetized rats modestly enhanced responses of spinal cord neurons to innocuous and noxious mechanical joint stimulation. This effect was blocked by minocycline indicating microglia involvement, and significantly attenuated by interfering with IL-6 signaling. In the BV2 microglial cell line, IL-1ß, like TNF, enhanced the release of soluble IL-6 receptor, necessary for spinal IL-6 actions. Different to TNF, IL-1ß caused SNB-19 astrocytes to release interleukin-11. The generation of "mechanical" spinal hyperexcitability by IL-1ß was more pronounced upon spinal TNF neutralization with etanercept, suggesting that concomitant TNF limits IL-1ß effects. In BV2 cells, TNF stimulated the release of IL-1Ra, an endogenous IL-1ß antagonist. Thus, spinal IL-1ß has the potential to induce spinal hyperexcitability sharing with TNF dependency on IL-6 signaling, but TNF also limited IL-1ß effects explaining the modest effect of IL-1ß.


Subject(s)
Interleukin-1beta/pharmacology , Interleukin-6/pharmacology , Neurons/drug effects , Spinal Cord/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Interleukin-11/metabolism , Joints/innervation , Microglia/drug effects , Nociception/drug effects , Physical Stimulation , Rats , Rats, Wistar , Signal Transduction/drug effects
6.
J Neurochem ; 157(6): 1821-1837, 2021 06.
Article in English | MEDLINE | ID: mdl-32885411

ABSTRACT

Hyperalgesic priming is characterized by enhanced nociceptor sensitization by pronociceptive mediators, prototypically PGE2 . Priming has gained interest as a mechanism underlying the transition to chronic pain. Which stimuli induce priming and what cellular mechanisms are employed remains incompletely understood. In adult male rats, we present the cytokine Oncostatin M (OSM), a member of the IL-6 family, as an inducer of priming by a novel mechanism. We used a high content microscopy based approach to quantify the activation of endogenous PKA-II and ERK of thousands sensory neurons in culture. Incubation with OSM increased and prolonged ERK activation by agents that increase cAMP production such as PGE2 , forskolin, and cAMP analogs. These changes were specific to IB4/CaMKIIα positive neurons, required protein translation, and increased cAMP-to-ERK signaling. In both, control and OSM-treated neurons, cAMP/ERK signaling involved RapGEF2 and PKA but not Epac. Similar enhancement of cAMP-to-ERK signaling could be induced by GDNF, which acts mostly on IB4/CaMKIIα-positive neurons, but not by NGF, which acts mostly on IB4/CaMKIIα-negative neurons. In vitro, OSM pretreatment rendered baseline TTX-R currents ERK-dependent and switched forskolin-increased currents from partial to full ERK-dependence in small/medium sized neurons. In summary, priming induced by OSM uses a novel mechanism to enhance and prolong coupling of cAMP/PKA to ERK1/2 signaling without changing the overall pathway structure.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Hyperalgesia/metabolism , MAP Kinase Signaling System/drug effects , Oncostatin M/toxicity , Animals , Antineoplastic Agents/toxicity , Humans , Hyperalgesia/chemically induced , MAP Kinase Signaling System/physiology , Male , Mice , Rats , Rats, Sprague-Dawley
7.
Sci Rep ; 8(1): 17656, 2018 12 05.
Article in English | MEDLINE | ID: mdl-30518958

ABSTRACT

The tachykinin substance P (SP) increases neuronal excitability, participates in homeostatic control, but induces brain oedema after stroke or trauma. We asked whether SP is able to induce cortical spreading depression (CSD) which often aggravates stroke-induced pathology. In anesthetized rats we applied SP (10-5, 10-6, 10-7, or 10-8 mol/L) to a restricted cortical area and recorded CSDs there and in remote non-treated areas using microelectrodes. SP was either applied in artificial cerebrospinal fluid (ACSF), or in aqua to perform a preconditioning. Plasma extravasation in cortical grey matter was assessed with Evans Blue. Only SP dissolved in aqua induced self-regenerating CSDs. SP dissolved in ACSF did not ignite CSDs even when excitability was increased by acetate-preconditioning. Aqua alone elicited as few CSDs as the lowest concentration of SP. Local pretreatment with 250 nmol/L of a neurokinin 1 receptor antagonist prevented the SP-induced plasma extravasation, the initiation of CSDs by 10-5 mol/L SP diluted in aqua, and the initiation of CSDs by aqua alone, but did not suppress KCl-induced CSD. Thus neurokinin 1 receptor antagonists may be used to explore the involvement of SP in CSDs in clinical studies.


Subject(s)
Cortical Spreading Depression , Receptors, Neurokinin-1/metabolism , Substance P/metabolism , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Cortical Spreading Depression/drug effects , Male , Neurokinin-1 Receptor Antagonists/pharmacology , Neurotransmitter Agents/metabolism , Rats , Rats, Wistar
8.
Eur J Pharmacol ; 835: 19-30, 2018 Sep 15.
Article in English | MEDLINE | ID: mdl-30036535

ABSTRACT

Many acute and chronic inflammatory diseases, cancer and neuropathic disorders are accompanied by severe pain states reducing drastically the life quality of the patients. Biologicals which preferentially target cytokines often reduce the disease processes by influencing immune cells, tissue healing, inflammatory aspects and other typical processes of the diseases. Remarkably the effect of biologicals in pain and nociception is often neglected or insufficiently explored. However, because of the dense interaction of the nociceptive system with the surrounding peripheral or central tissue, targeting cytokines has the potential to treat pain at the same time as the other symptoms of the disease. The following review shows mainly results from animal experiments (with some parallels to human studies). It depicts where and how cytokines are involved in nociceptive processing and pain and also indicates possible target strategies. It concentrates on the excitatory cytokines IL-6, TNF-α, IL-1ß, IFN-γ and IL-17.


Subject(s)
Analgesics/pharmacology , Biological Products/pharmacology , Cytokines/metabolism , Animals , Cytokines/antagonists & inhibitors , Humans , Nociception/drug effects
9.
J Neurosci ; 36(38): 9782-91, 2016 09 21.
Article in English | MEDLINE | ID: mdl-27656018

ABSTRACT

UNLABELLED: During peripheral inflammation, both spinal TNF-α and IL-6 are released within the spinal cord and support the generation of inflammation-evoked spinal hyperexcitability. However, whether spinal TNF-α and IL-6 act independently in parallel or in a functionally dependent manner has not been investigated. In extracellular recordings from mechanonociceptive deep dorsal horn neurons of normal rats in vivo, we found that spinal application of TNF-α increased spinal neuronal responses to mechanical stimulation of knee and ankle joints. This effect was significantly attenuated by either sgp130, which blocks IL-6 trans-signaling mediated by IL-6 and its soluble receptor IL-6R (sIL-6R); by an antibody to the IL-6 receptor; or by minocycline, which inhibits the microglia. IL-6 was localized in neurons of the spinal cord and, upon peripheral noxious stimulation in the presence of spinal TNF-α, IL-6 was released spinally. Furthermore, TNF-α recruited microglial cells to provide sIL-6R, which can form complexes with IL-6. Spinal application of IL-6 plus sIL-6R, but not of IL-6 alone, enhanced spinal hyperexcitability similar to TNF-α and the inhibition of TNF-α-induced hyperexcitability by minocycline was overcome by coadministration of sIL-6R, showing that sIL-6R is required. Neither minocycline nor the TNF-α-neutralizing compound etanercept inhibited the induction of hyperexcitability by IL-6 plus sIL-6R. Together, these data show that the induction of hyperexcitability of nociceptive deep dorsal horn neurons by TNF-α largely depends on the formation of IL-6/sIL-6R complexes that are downstream of TNF-α and requires the interactions of neurons and microglia orchestrated by TNF-α. SIGNIFICANCE STATEMENT: Both spinal TNF-α and IL-6 induce a state of spinal hyperexcitability. We present the novel finding that the full effect of TNF-α on the development of spinal hyperexcitability depends on IL-6 trans-signaling acting downstream of TNF-α. IL-6 trans-signaling requires the formation of complexes of IL-6 and soluble IL-6 receptor. Spinal TNF-α furthers the release of IL-6 from neurons in the spinal cord during peripheral noxious stimulation and recruits microglial cells to provide soluble IL-6 receptor, which can form complexes with IL-6. Therefore, a specific interaction between neurons and microglia is required for the full development of TNF-α-induced hyperexcitability of nociceptive deep horsal horn neurons.


Subject(s)
Interleukin-6/metabolism , Posterior Horn Cells/drug effects , Signal Transduction/drug effects , Spinal Cord/cytology , Tumor Necrosis Factor-alpha/pharmacology , Action Potentials/drug effects , Animals , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Cytokine Receptor gp130/metabolism , Glial Fibrillary Acidic Protein/metabolism , Male , Mice , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Minocycline/pharmacology , Physical Stimulation , Posterior Horn Cells/metabolism , Rats , Rats, Wistar , Spinal Cord/drug effects , Spinal Cord/metabolism , Time Factors , Ubiquitin Thiolesterase/metabolism
10.
Arthritis Rheumatol ; 66(3): 599-609, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24574220

ABSTRACT

OBJECTIVE: Tumor necrosis factor α (TNFα) is produced not only in peripheral tissues, but also in the spinal cord. The purpose of this study was to address the potential of peripheral and spinal TNFα to induce and maintain spinal hyperexcitability, which is a hallmark of pain states in the joints during rheumatoid arthritis and osteoarthritis. METHODS: In vivo recordings of the responses of spinal cord neurons to nociceptive knee input under normal conditions and in the presence of experimental knee joint inflammation were obtained in anesthetized rats. TNFα, etanercept, or antibodies to TNF receptors were applied to either the knee joint or the spinal cord surface. RESULTS: Injection of TNFα into the knee joint cavity increased the responses of spinal cord neurons to mechanical joint stimulation, and injection of etanercept into the knee joint reduced the inflammation-evoked spinal activity. These spinal effects closely mirrored the induction and reduction of peripheral sensitization. Responses to joint stimulation were also enhanced by spinal application of TNFα, and spinal application of either etanercept or anti-TNF receptor type I significantly attenuated the generation of inflammation-evoked spinal hyperexcitability, which is characterized by widespread pain sensitization beyond the inflamed joint. Spinally applied etanercept did not reduce established hyperexcitability in the acute kaolin/carrageenan model. In antigen-induced arthritis, etanercept decreased spinal responses on day 1, but not on day 3. CONCLUSION: While peripheral TNFα increases spinal responses to joint stimulation, spinal TNFα supports the generation of the full pattern of spinal hyperexcitability. However, established spinal hyperexcitability may be maintained by downstream mechanisms that are independent of spinal TNFα.


Subject(s)
Arthritis, Experimental/metabolism , Hyperalgesia/metabolism , Inflammation/metabolism , Knee Joint/metabolism , Neurons/physiology , Spinal Cord/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Arthritis, Experimental/pathology , Arthritis, Experimental/physiopathology , Etanercept , Hyperalgesia/pathology , Hyperalgesia/physiopathology , Immunoglobulin G/pharmacology , Inflammation/pathology , Inflammation/physiopathology , Knee Joint/pathology , Knee Joint/physiopathology , Male , Neurons/drug effects , Neurons/pathology , Nociceptors/drug effects , Nociceptors/pathology , Nociceptors/physiology , Physical Stimulation , Rats , Rats, Wistar , Receptors, Tumor Necrosis Factor , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord/physiopathology
11.
Proc Natl Acad Sci U S A ; 110(33): 13648-53, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23904482

ABSTRACT

The pain mediator prostaglandin E2 (PGE2) sensitizes nociceptive pathways through EP2 and EP4 receptors, which are coupled to Gs proteins and increase cAMP. However, PGE2 also activates EP3 receptors, and the major signaling pathway of the EP3 receptor splice variants uses inhibition of cAMP synthesis via Gi proteins. This opposite effect raises the intriguing question of whether the Gi-protein-coupled EP3 receptor may counteract the EP2 and EP4 receptor-mediated pronociceptive effects of PGE2. We found extensive localization of the EP3 receptor in primary sensory neurons and the spinal cord. The selective activation of the EP3 receptor at these sites did not sensitize nociceptive neurons in healthy animals. In contrast, it produced profound analgesia and reduced responses of peripheral and spinal nociceptive neurons to noxious stimuli but only when the joint was inflamed. In isolated dorsal root ganglion neurons, EP3 receptor activation counteracted the sensitizing effect of PGE2, and stimulation of excitatory EP receptors promoted the expression of membrane-associated inhibitory EP3 receptor. We propose, therefore, that the EP3 receptor provides endogenous pain control and that selective activation of EP3 receptors may be a unique approach to reverse inflammatory pain. Importantly, we identified the EP3 receptor in the joint nerves of patients with painful osteoarthritis.


Subject(s)
Inflammation/physiopathology , Nociception/physiology , Nociceptors/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Analysis of Variance , Animals , DNA Primers/genetics , Humans , Immunohistochemistry , Joints/physiopathology , Osteoarthritis/physiopathology , Patch-Clamp Techniques , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Rats , Rats, Inbred Lew
12.
J Cereb Blood Flow Metab ; 32(8): 1535-45, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22453631

ABSTRACT

The brainstem of the adult rat is relatively resistant to spreading depolarization (SD) but after enhancement of excitability SD can be evoked by local application of KCl. In the present experiments, we observed that the enhanced excitability even triggers prolonged periods of repetitive depolarizations (RDs), which elicit significant cardiovascular changes. In contrast to KCl-evoked SDs with amplitudes of ∼24 mV and spreading velocity of 4 mm/min, spontaneous RDs had amplitudes of 7 to 12 mV, propagated up to 30 times faster than KCl-evoked SDs, and depolarized larger brainstem areas including the contralateral side. Similarly as SD, RDs depended on glutamatergic neurotransmission and were blocked by MK-801 or by the calcium channel blocker agatoxin. They depended on sodium channels and were blocked by tetrodotoxin. Functionally, the invasion of RDs into the spinal trigeminal and other nuclei evoked bursts of action potentials, indicating that specific neuronal systems are affected. In fact, during episodes of RDs the blood pressure and the local blood flow at the surface of the brainstem and the cortex increased substantially. Brainstem RDs did not propagate into the cerebral cortex. We propose to consider brainstem RPs as a pathophysiological mechanism whose significance for brainstem disease states should be further explored.


Subject(s)
Blood Pressure/physiology , Brain Stem/physiopathology , Cortical Spreading Depression/physiology , Heart Rate/physiology , Neurons/physiology , Animals , Brain Stem/drug effects , Brain Stem/metabolism , Calcitonin Gene-Related Peptide/metabolism , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/physiology , Cortical Spreading Depression/drug effects , Heart Rate/drug effects , Male , Microelectrodes , Neurons/drug effects , Neurons/metabolism , Potassium Chloride/pharmacology , Rats , Rats, Wistar , Regional Blood Flow/drug effects , Regional Blood Flow/physiology
13.
Arthritis Rheum ; 64(7): 2233-42, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22246633

ABSTRACT

OBJECTIVE: Significant joint pain is usually widespread beyond the affected joint, which results from the sensitization of nociceptive neurons in the central nervous system (central sensitization). This study was undertaken to explore whether the proinflammatory cytokine interleukin-6 (IL-6) in the joint induces central sensitization, whether joint inflammation causes the release of IL-6 from the spinal cord, and whether spinal IL-6 contributes to central sensitization. METHODS: In anesthetized rats, electrophysiologic recordings of spinal cord neurons with sensory input from the knee joint were made. Neuronal responses to mechanical stimulation of the rat knee and leg were monitored. IL-6 and soluble IL-6 receptor (sIL-6R) were applied to the knee joint or the spinal cord. Spinal release of IL-6 was measured by enzyme-linked immunosorbent assay. Soluble gp130, which neutralizes IL-6/sIL-6R, was spinally applied during the development of joint inflammation or during established inflammation. RESULTS: A single injection of IL-6/sIL-6R into the rat knee joint as well as application of IL-6/sIL-6R to the rat spinal cord significantly increased the responses of spinal neurons to mechanical stimulation of the knee and ankle joint, i.e., induced central sensitization. Application of soluble gp130 to the rat spinal cord attenuated this effect of IL-6. The development of knee inflammation in the rat caused spinal release of IL-6. Spinal application of soluble gp130 attenuated the development of inflammation-evoked central sensitization but did not reverse it. CONCLUSION: Our findings indicate that the generation of joint pain in the rat involves not only IL-6 in the joint but also IL-6 released from the spinal cord. Spinal IL-6 contributes to central sensitization and thus promotes the widespread hyperalgesia observed in the course of joint disease.


Subject(s)
Hyperalgesia/metabolism , Interleukin-6/pharmacology , Knee Joint/drug effects , Pain/metabolism , Spinal Cord/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Animals , Arthritis, Experimental , Interleukin-6/metabolism , Knee Joint/metabolism , Male , Nociceptors/drug effects , Nociceptors/physiology , Rats , Rats, Wistar , Spinal Cord/metabolism
14.
Arthritis Res Ther ; 13(2): 210, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21542894

ABSTRACT

The peripheral nociceptor is an important target of pain therapy because many pathological conditions such as inflammation excite and sensitize peripheral nociceptors. Numerous ion channels and receptors for inflammatory mediators were identified in nociceptors that are involved in neuronal excitation and sensitization, and new targets, beyond prostaglandins and cytokines, emerged for pain therapy. This review addresses mechanisms of nociception and focuses on molecules that are currently favored as new targets in drug development or that are already targeted by new compounds at the stage of clinical trials--namely the transient receptor potential V1 receptor, nerve growth factor, and voltage-gated sodium channels--or both.


Subject(s)
Nociceptors/metabolism , Pain/physiopathology , Cytokines/metabolism , Humans , Nerve Growth Factor/metabolism , Prostaglandins/metabolism , Sodium Channels/metabolism , TRPV Cation Channels/metabolism
15.
Pain ; 152(5): 1114-1126, 2011 May.
Article in English | MEDLINE | ID: mdl-21345590

ABSTRACT

Tetrodotoxin-resistant (TTX-R) Na(+) channels play a key role in the generation of action potentials in nociceptive dorsal root ganglion (DRG) neurons and are an important target for the proinflammatory mediator prostaglandin E(2), which augments these currents. Prostaglandin D(2) (PGD(2)) is released in the tissue together with prostaglandin E(2), and it was reported to be antiinflammatory, but its effect on primary afferent neurons is unclear. In the present study we localised G(s)-protein-coupled DP1 and G(i)-protein-coupled DP2 receptors in DRG neurons, and we assessed the effect of PGD(2) on TTX-R Na(+) currents in patch-clamp recordings from small- to medium-sized cultured DRG neurons from adult rats. DP1 and DP2 receptor-like immunoreactivity was localised in the vast majority of DRG neurons. In all neurons, PGD(2) shifted conductance to more hyperpolarised potentials, depending on an action at Na(v)1.9 channels. In about one third of the neurons, PGD(2) additionally influenced Na(v)1.8 channels by facilitating conductance and by increasing maximal current amplitudes. Selective DP1 receptor activation increased the amplitude of TTX-R Na(+) currents of most neurons, but this effect was counteracted by DP2 receptor activation, which by itself had no effect. In the current-clamp mode, PGD(2) lowered the threshold for elicitation of an action potential and increased the number of action potentials per stimulus, an effect mainly depending on DP1 receptor activation. Thus, the net effect of PGD(2) on DRG neurons is pronociceptive, although the magnitude of the TTX-R Na(+) currents depends on the balance of DP1 and DP2 receptor activation.


Subject(s)
Ganglia, Spinal/cytology , Prostaglandin D2/pharmacology , Sensory Receptor Cells/drug effects , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Action Potentials/drug effects , Animals , Carbazoles/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Ganglia, Spinal/metabolism , Hydantoins/pharmacology , Male , Membrane Potentials/drug effects , Patch-Clamp Techniques , Platelet Aggregation Inhibitors/pharmacology , Prostaglandin D2/antagonists & inhibitors , Rats , Rats, Wistar , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , Sulfonamides/pharmacology
16.
Pain ; 148(1): 26-35, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19879047

ABSTRACT

Both cyclooxygenase-1 and -2 are expressed in the spinal cord, and the spinal COX product prostaglandin E(2) (PGE(2)) contributes to the generation of central sensitization upon peripheral inflammation. Vice versa spinal COX inhibition is considered an important mechanism of antihyperalgesic pain treatment. Recently, however, COX-2 was shown to be also involved in the metabolism of endocannabinoids. Because endocannabinoids can have analgesic actions it is conceivable that inhibition of spinal COX produces analgesia not only by inhibition of PG synthesis but also by inhibition of endocannabinoid breakdown. In the present study, we recorded from spinal cord neurons with input from the inflamed knee joint and we measured the spinal release of PGE(2) and the endocannabinoid 2-arachidonoyl glycerol (2-AG) in vivo, using the same stimulation procedures. COX inhibitors were applied spinally. Selective COX-1, selective COX-2 and non-selective COX inhibitors attenuated the generation of spinal hyperexcitability when applied before and during development of inflammation but, when inflammation and spinal hyperexcitability were established, only selective COX-2 inhibitors reversed spinal hyperexcitability. During established inflammation all COX inhibitors reduced release of spinal PGE(2) almost equally but only the COX-2 inhibitor prevented breakdown of 2-AG. The reversal of spinal hyperexcitability by COX-2 inhibitors was prevented or partially reversed by AM-251, an antagonist at the cannabinoid-1 receptor. We conclude that inhibition of spinal COX-2 not only reduces PG production but also endocannabinoid breakdown and provide evidence that reversal of inflammation-evoked spinal hyperexcitability by COX-2 inhibitors is more related to endocannabinoidergic mechanisms than to inhibition of spinal PG synthesis.


Subject(s)
Arachidonic Acids/metabolism , Arthritis, Experimental/enzymology , Dinoprostone/metabolism , Glycerides/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Spinal Cord/enzymology , Action Potentials/drug effects , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Disease Models, Animal , Drug Administration Routes , Endocannabinoids , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Indans/pharmacology , Indans/therapeutic use , Injections, Spinal/methods , Knee Joint/pathology , Male , Neurons/drug effects , Neurons/physiology , Pain Measurement , Physical Stimulation/methods , Piperidines/pharmacology , Piperidines/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Rats , Rats, Wistar , Spinal Cord/drug effects , Spinal Cord/pathology , Statistics, Nonparametric
17.
Exp Brain Res ; 196(1): 153-62, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19363606

ABSTRACT

Both inflammatory and degenerative diseases of joints are major causes of chronic pain. This overview addresses the clinical problem of joint pain, the nociceptive system of the joint, the mechanisms of peripheral and central sensitization during joint inflammation and long term changes during chronic joint inflammation. While the nature of inflammatory pain is obvious the nature and site of origin of osteoarthritic pain is less clear. However, in both pathological conditions mechanical hyperalgesia is the major pain problem, and indeed, both joint nociceptors and spinal nociceptive neurons with joint input show pronounced sensitization for mechanical stimulation. Molecular mechanisms of mechanical sensitization of joint nociceptors are addressed with an emphasis on cytokines, and molecular mechanisms of central sensitization include data on the role of excitatory amino acids, neuropeptides and spinal prostaglandins. The overview will also address long-term changes of pain-related behavior, response properties of neurons and receptor expression in chronic animal models of arthritis.


Subject(s)
Joints/physiopathology , Pain/physiopathology , Animals , Arthralgia/physiopathology , Arthritis/physiopathology , Brain/physiopathology , Cytokines/metabolism , Excitatory Amino Acids/metabolism , Humans , Hyperalgesia/physiopathology , Joints/immunology , Joints/innervation , Neuropeptides/metabolism , Nociceptors/physiology , Physical Stimulation , Prostaglandins/metabolism , Spinal Cord/physiopathology
18.
Neuroreport ; 17(15): 1615-8, 2006 Oct 23.
Article in English | MEDLINE | ID: mdl-17001279

ABSTRACT

In behavioral experiments, inhibition of nuclear factor-kappaB activation by systemic administration of the IkappaB kinase inhibitor S1627 has been shown to attenuate inflammatory and neuropathic pain. Here, we specifically investigated with electrophysiological recordings in anesthetized rats whether spinal application of S1627 influences hyperexcitability of dorsal horn neurons during an acute knee joint inflammation. Spinal application of S1627 before and early during development of inflammation totally prevented spinal hyperexcitability suggesting an important role of spinal nuclear factor-kappaB in this process. During established inflammation, however, S1627 did not reduce the responses of neurons to mechanical stimulation of the inflamed knee within 2.5 h after spinal administration, thus suggesting that spinal hyperexcitability is not maintained by continuous nuclear factor-kappaB activation.


Subject(s)
NF-kappa B/physiology , Posterior Horn Cells/physiology , Spinal Cord/cytology , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Arthritis, Experimental/physiopathology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Male , NF-kappa B/antagonists & inhibitors , Posterior Horn Cells/drug effects , Rats , Rats, Wistar , Time Factors
19.
J Cereb Blood Flow Metab ; 25(9): 1225-35, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15829916

ABSTRACT

Cortical spreading depression (CSD) is thought to be a neuronal mechanism that expands the penumbra zone after focal brain ischemia and that causes migraine aura. Both adrenergic agonists and antagonists significantly influence the size of the penumbra zone and decline the frequency of migraine. To study whether these compounds act by influencing CSD, we applied different drugs topically to an area of the exposed cortex of anesthetized adult rats and observed the migration of CSD-related DC potential deflections across the treated area. The adrenergic agonist norepinephrine (1 mmol/L) and the alpha(2)-agonist clonidine (0.56 mmol/L) blocked reversibly the migration of CSD. The beta-blocker propranolol (250 micromol/L to 1 mmol/L) dose-dependently diminished migration velocity or even blocked migration of CSD. The CSD blockade by the alpha(2)-antagonist yohimbine (1.75 mmol/L) was because of its action on inhibitory 5-HT(1A) receptors. None of the substances in the concentrations used had influence on regional cerebral blood flow or on systemic arterial blood pressure. The data suggest that the interference of these compounds with CSD may contribute to their beneficial therapeutic effect. The effect of beta-receptor antagonists in human migraine needs further exploration, since these drugs also work in migraine without aura.


Subject(s)
Adrenergic Antagonists/pharmacology , Adrenergic alpha-Agonists/pharmacology , Brain Damage, Chronic/prevention & control , Brain Ischemia/pathology , Cortical Spreading Depression/drug effects , Migraine Disorders/prevention & control , Migraine Disorders/physiopathology , Norepinephrine/physiology , Adrenergic beta-Antagonists/pharmacology , Animals , Blood Pressure/drug effects , Brain Damage, Chronic/etiology , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Cerebrovascular Circulation/drug effects , Clonidine/pharmacology , Drug Interactions , Electrodes, Implanted , Imidazoles/pharmacology , Indoles/pharmacology , Isoindoles , Male , Norepinephrine/pharmacology , Piperazines/pharmacology , Propranolol/pharmacology , Rats , Rats, Wistar , Serotonin 5-HT1 Receptor Antagonists
20.
J Neurosci ; 24(3): 642-51, 2004 Jan 21.
Article in English | MEDLINE | ID: mdl-14736850

ABSTRACT

Inflammatory pain is caused by sensitization of peripheral and central nociceptive neurons. Prostaglandins substantially contribute to neuronal sensitization at both sites. Prostaglandin E2 (PGE2) applied to the spinal cord causes neuronal hyperexcitability similar to peripheral inflammation. Because PGE2 can act through EP1-EP4 receptors, we addressed the role of these receptors in the spinal cord on the development of spinal hyperexcitability. Recordings were made from nociceptive dorsal horn neurons with main input from the knee joint, and responses of the neurons to noxious and innocuous stimulation of the knee, ankle, and paw were studied after spinal application of recently developed specific EP1-EP4 receptor agonists. Under normal conditions, spinal application of agonists at EP1, EP2, and EP4 receptors induced spinal hyperexcitability similar to PGE2. Interestingly, the effect of spinal EP receptor activation changed during joint inflammation. When the knee joint had been inflamed 7-11 hr before the recordings, only activation of the EP1 receptor caused additional facilitation, whereas spinal application of EP2 and EP4 receptor agonists had no effect. Additionally, an EP3alpha receptor agonist reduced responses to mechanical stimulation. The latter also attenuated spinal hyperexcitability induced by spinal PGE2. In isolated DRG neurons, the EP3alpha agonist reduced the facilitatory effect of PGE2 on TTX-resistant sodium currents. Thus pronociceptive effects of spinal PGE2 can be limited, particularly under inflammatory conditions, through activation of an inhibitory splice variant of the EP3 receptor. The latter might be an interesting target for controlling spinal hyperexcitability in inflammatory pain states.


Subject(s)
Arthritis/physiopathology , Dinoprostone/pharmacology , Knee Joint/physiopathology , Pain/physiopathology , Receptors, Prostaglandin E/metabolism , Spinal Cord/physiopathology , Animals , Arthritis/chemically induced , Carrageenan , Cell Separation , Dinoprostone/analogs & derivatives , Disease Models, Animal , Ganglia, Spinal/cytology , Kaolin , Male , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques , Physical Stimulation , Protein Isoforms/agonists , Protein Isoforms/metabolism , Rats , Rats, Wistar , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E, EP3 Subtype , Spinal Cord/drug effects , Spinal Cord/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL