Review Open Access
Copyright ©2014 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Anesthesiol. Mar 27, 2014; 3(1): 71-81
Published online Mar 27, 2014. doi: 10.5313/wja.v3.i1.71
Molecular mechanism of inflammatory pain
Yeu-Shiuan Su, Wei-Hsin Sun, Department of Life Sciences, National Central University, Jhongli 32054, Taiwan
Chih-Cheng Chen, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
Chih-Cheng Chen, Taiwan Mouse Clinic-National Phenotyping and Drug Testing Center, Academia Sinica, Taipei 115, Taiwan
Author contributions: Su YS and Sun WH contributed equally to this work, collected literatures, and wrote the manuscript; Chen CC designed the scope of the minireview and wrote the manuscript.
Correspondence to: Chih-Cheng Chen, PhD, Institute of Biomedical Sciences, Academia Sinica, 128 Academia Road, Section 2, Taipei 115, Taiwan. chih@ibms.sinica.edu.tw
Telephone: +886-2-26523917 Fax: +886-2-27829224
Received: June 29, 2013
Revised: September 27, 2013
Accepted: November 1, 2013
Published online: March 27, 2014

Abstract

Chronic inflammatory pain resulting from arthritis, nerve injury and tumor growth is a serious public health issue. One of the major challenges in chronic inflammatory pain research is to develop new pharmacologic treatments with long-term efficacy and few side effects. The mediators released from inflamed sites induce complex changes in peripheral and central processing by directly acting on transducer receptors located on primary sensory neurons to transmit pain signals or indirectly modulating pain signals by activating receptors coupled with G-proteins and second messengers. High local proton concentration (acidosis) is thought to be a decisive factor in inflammatory pain and other mediators such as prostaglandin, bradykinin, and serotonin enhance proton-induced pain. Proton-sensing ion channels [transient receptor potential V1 (TRPV1) and the acid-sensing ion channel (ASIC) family] are major receptors for direct excitation of nociceptive sensory neurons in response to acidosis or inflammation. G-protein-coupled receptors activated by prostaglandin, bradykinin, serotonin, and proton modulate functions of TRPV1, ASICs or other ion channels, thus leading to inflammation- or acidosis-linked hyperalgesia. Although detailed mechanisms remain unsolved, clearly different types of pain or hyperalgesia could be due to complex interactions between a distinct subset of inflammatory mediator receptors expressed in a subset of nociceptors. This review describes new directions for the development of novel therapeutic treatments in pain.

Key Words: Acid-sensing ion channel, Acidosis, G-protein-coupled receptor, Inflammation, Proton-sensing ion channel, Transient receptor potential V1

Core tip: Tissue acidosis that occurs during inflammation is central to the development and maintenance of chronic pain. Recent studies have revealed a variety of proton-sensing ion channels (e.g., acid-sensing ion channels, transient receptor potential V1) and G-protein-coupled receptors (e.g., G2 accumulation 2A, G-protein-coupled receptor 4, ovarian cancer G-protein-coupled receptor, T-cell death-associated gene 8) responsible for acid-induced pain. These cell-surface membrane proteins are promising therapeutic targets for the development of new analgesic drugs for chronic inflammatory pain.



INTRODUCTION

Cancer, nerve injury, and arthritis often cause chronic inflammatory pain[1]. Chronic pain may have a profound effect on a person’s life and society when not effectively treated. Although a variety of pharmacologic treatments are available, they are limited by unacceptable side effects or short-term efficacy. The development of long-acting pharmacologic therapies requires knowledge of how chronic inflammatory pain signals are initially interpreted and subsequently transmitted and perpetuated. This review focuses on recent findings from studies of the molecular mechanisms of inflammatory pain transmission and modulation, especially the roles of mediator-gated ion channels and G-protein-coupled receptors (GPCRs).

INFLAMMATORY PAIN

When our body senses noxious stimuli (such as a cut from a sharp knife, burn from an open flame, or contact with burning or erosive chemicals), the signal quickly activates primary sensory afferents (nociceptors) and delivers a message to the brain to elicit the pain feeling. When stimuli are absent, the painful experience disappears. The situation is called acute pain because the pain signal is transient[2]. Noxious stimuli activate transducer receptors located on medium myelinated (Aδ) and small unmyelinated (C) nociceptors to induce the receptor potential. The receptor potential activates a variety of voltage-gated ion channels to transmit pain signals to secondary nociceptors in the dorsal horn of the spinal cord, then to the brain[3].

If the tissues are damaged mechanically or by pathogen infection, autoimmune disease, or tumor growth, the sites of the damaged or infected tissues usually show inflammatory responses such as redness, swelling and heat accompanied by persistent pain; endogenous mediators released from the damaged or infected tissues increase the extravasation of the vessels and attract the immune cells, including mast cells, macrophages, neutrophils, and platelets, to the injured site for the inflammatory response[1]. The “inflammatory soup” is rich in purines, amines, cytokines, protons, ions and growth factors. These mediators can directly activate the nociceptors, evoking pain or modulating the sensitivity of the primary nociceptors, thus causing a hyperreactive reaction to stimuli. As a result, normal stimuli such as a light touch or a brush are perceived as painful (allodynia), or normally painful stimuli cause pain of greater intensity (hyperalgesia)[4]. In the periphery, inflammatory mediators bind to GPCRs to activate protein kinases A and C (PKA and PKC) to phosphorylate receptors or increase receptor expression, which enhances the sensitivity of primary nociceptors, called peripheral sensitization. Primary nociceptor-driven transmitter release activates intracellular kinases to phosphorylate receptors. This situation leads to an immediate and activity-dependent increase in the excitability and responsiveness of dorsal horn neurons, called central sensitization. Central sensitization could be sustained for some time because of transcriptional changes[2,4].

INFLAMMATORY MEDIATORS OF PAIN

The endogenous mediators, such as prostaglandin E2 (PGE2), bradykinin (BK), serotonin [5-hydroxytryptamine (5-HT)], proton, histamine, and ATP, are released from the damaged site of the tissue and immune cells to induce inflammation and nociception[2]. These mediators act on transducer receptors situated on sensory neurons to induce complex changes in peripheral and central signal processing. Although some mediators can act directly on ion channels to induce receptor potential, for the most part these chemical interactions occur through the activation of receptors coupled with G-proteins and second messengers, thus activating protein kinases. Such activated kinases phosphorylate ion channels to alter ion permeability or phosphorylate cellular proteins to increase gene expression.

Earlier studies of single mediators demonstrated that BK, PGE2, 5-HT, and proton have excitatory action on cutaneous nociceptors and induces transient pain[5-8]. More sustained effects are achieved only in a high-concentration (10-5 mol/L) combination of inflammatory mediators (BK, 5-HT, PGE2, and histamine)[9]. Steen et al[10] proposed that the combination of inflammatory mediators plays a role in sensitizing the low pH effect. The acidosis in inflamed tissues is the decisive factor for ongoing nociceptor excitation and sustained pain. However, the interaction between various mediators remains unclear.

TISSUE ACIDOSIS AND ACID-SENSING RECEPTORS

Tissue acidosis is a common phenomenon found in inflammation (reduced to pH 5.4)[11], in lesions or incisions (reduced to pH 6.5)[12], in ischemic heart or muscle (pH 5.7-7.0)[13,14], and even in malignant tumors (pH 5.8-7.4)[15]. High local proton concentrations in inflamed tissues can excite and sensitize rat skin nociceptors and can cause sustained pain in human skin[7,16,17]. As well, the combination of inflammatory mediators (BK, 5-HT, PGE2, and histamine) in acid solution (pH 6.1) can excite and sensitize rat skin nociceptors[18]. Injections of the inflammatory mediator combination in neutral solution in human skin induces dose-dependent, transient, burning pain, but the effects become more intense and prolonged when the mediator combination is in acidic solution[10]. Studies of rat dorsal root ganglion (DRG) neurons revealed that acidic solutions induced a cation conductance in a subset of neurons[19], and a proton-activated sustained current is potentiated more by the mediator combination than each mediator alone[20]. Proton-activated currents found in the sensory neurons are due to direct activation of the non-selective cation channels and indirect modulation of ion channels[21]. Proton-gated ion channels and proton-sensing GPCRs expressed on nociceptors are potential candidates responsible for acidosis-induced pain.

PROTON-GATED ION CHANNELS: ACID-SENSING ION CHANNELS

Acid-sensing ion channels (ASICs), which belong to the family of degenerin/epithelial amiloride-sensitive Na+ channels, are voltage-insensitive cationic channels activated by extracellular protons[22-25]. The ASIC family, comprising ASIC1a, ASIC1b, ASIC2a, ASIC2b, ASIC3, ASIC4 and ASIC5, is expressed in the peripheral and central nervous systems[26-28].

Among ASICs, ASIC3 is the most sensitive receptor to protons, with pH 0.5 for activation around 6.7, and is expressed in both small- and large-diameter DRG neurons[29-31]. The expression of ASIC3 in DRG is increased with hind paw inflammation in rats[32,33]. As well, ASIC3 channel activity is enhanced by several components of the inflammatory soup, such as BK, 5-HT, hypertonicity, arachidonic acid, and nitric oxide[34-38]. Thus, ASIC3 is considered a sensor of acidic and primary inflammatory pain[34]. Study of skin nerves revealed that loss of ASIC3 increases the sensitivity of mechanoreceptors to light touch but decreases that of mechanoreceptors to a noxious pinch[39,40]. Surprisingly, mice lacking the ASIC3 gene still respond to acid stimuli and have acid-induced pain or primary inflammatory pain[39,41-43]. However, inhibiting ASIC3 function with a specific peptide or small interfering RNA significantly reduces cutaneous acidic pain under normal or inflammatory conditions and postoperative pain[34,44].

Given that ASIC3 is predominantly expressed in muscle nociceptors rather than in cutaneous nociceptors[45], ASIC3 should be required for development of secondary mechanical hyperalgesia induced by acid injection in skeletal muscle or by muscle inflammation[46-48]. Although the ASIC3 requirement for development and maintenance of muscle inflammatory pain is argued, selective microRNA-targeted ASIC3 inhibits primary and secondary hyperalgesia induced by muscle inflammation[49]. Interestingly, a recent study by Lin et al[50] suggested that ASIC3-mediated muscle pain is negatively modulated by substance P via regulation of the M channel in a G-protein-independent pathway.

ASIC1a is predominantly expressed in small-diameter DRG neurons[23,51] and is less sensitive than ASIC3 with pH 0.5 for activation around 6.5[29,30]. Mice lacking ASIC1a show normal mechanical sensitivity in cutaneous afferents but enhanced mechanically evoked firing rate in gastrointestinal afferents[52,53]. In contrast to the ASIC3 role in secondary hyperalgesia, ASIC1a-deficient mice do not develop primary hyperalgesia induced by muscle inflammation, so ASIC1a and ASIC3 may play distinct roles in the development and maintenance of hyperalgesia, respectively[43]. Downregulation of ASIC1a expression in spinal dorsal horn neurons by using selective inhibitor or antisense oligonucleotides reduces complete Freund’s adjuvant (CFA)-induced thermal and mechanical hypersensitivity, which suggests that ASIC1a contributes to central sensitization in inflammatory pain[54]. A recent study provides a new view for ASIC1a and ASIC3 roles in inflammatory pain in that acidosis may induce endocytosis and maturation of macrophages through ASIC1a and ASIC3[55]. Mice lacking ASIC1a, ASIC2 and ASIC3 genes lost acid-induced transient currents, but their behavioral sensitivity to mechanical stimuli was increased, so ASICs indeed contribute cutaneous mechanosensation but in complex behavioral changes[56].

PROTON-GATED ION CHANNELS: TRPV1

Transient receptor potential/vanilloid receptor subtype 1 (TRPV1/VR1) is a 6-transmembrane domain, non-selective cation channel and activated by vanilloid, heat, capsaicin, and proton[57,58]. TRPV1 is predominantly expressed in small-diameter DRG neurons in rats and mice[57]. Disruption of the TRPV1 gene in mice reduces responses of DRG neurons to acid and thermal stimuli and eliminates carrageenan-induced thermal hyperalgesia, so TRPV1 may be involved in acid-induced pain and inflammation-induced thermal hyperalgesia[59,60]. However, surprisingly, blockage of the TRPV1 function in peripheral or spinal loci by selective antagonists inhibits mechanical hyperalgesia induced by CFA, capsaicin, or bone cancer[61-64]. Although TRPV1 participates in both mechanical allodynia and thermal hyperalgesia induced by cutaneous inflammation, it does no participate in muscle inflammation[65]. TRPV1 mediates the development of heat but not mechanical hypersensitivity after muscle inflammation[66]. With peripheral inflammation, the mRNA TRPV1 expression is increased and the channel function enhanced in DRG neurons[67-69]. Interestingly, DRG neurons with increased TRPV1 expression and function are mainly non-peptidergic rather than peptidergic neurons[69]. Since most non-peptidergic neurons project to skin targets, TRPV1 would mainly participate in cutaneous inflammatory pain[70,71]. Okun et al[72] suggested that CFA-induced ongoing pain is transient and depends on TRPV1-positive afferents but cannot be blocked by TRPV1 antagonism. TRPV1 may be responsive to noxious stimuli while nociceptors are sensitized (inflammation). Its function could be sensitized by inflammatory mediators such as BK[73,74], chemokines (CCL3)[75], 5-HT[76], PGE2[77,78], proton[79] or by protease-activated receptor 2[80,81]. A recent study suggested that TRPV1 and TRPA1 are involved in the transition of acute to chronic pain in a chronic pancreatitis model[82].

PROTON-SENSING G-PROTEIN-COUPLED RECEPTORS: OGR1 FAMILY

In 2003, Ludwig et al[83] found two GPCRs, ovarian cancer GPR 1 (OGR1) and G protein-coupled receptor 4 (GPR4), fully responsive to protons at pH 6.8 and stimulating inositol triphosphate and cAMP formation, respectively. Later, the 2 other family members, G2 accumulation (G2A) and T-cell death-associated gene 8 (TDAG8) were identified as proton receptors, with full activation at pH 6.4-6.8[84-86]. OGR1, GPR4, and G2A were previously identified as receptors for sphingosylphosphorylcholine (SPC) or lysophosphatidylcholine (LPC), but the original publications have now been retracted[87-89]. Whether OGR1, GPR4, and G2A are SPC or LPC receptors remains unclear. In addition to responding to protons, TDAG8 also responds to psychosine[85,89]. Although G2A was considered a proton-sensing receptor, Radu et al[90] suggested that G2A is less likely to be a pH sensor because it does not generate a significant response after acid stimulation. G2A shows conservation of only 1 of 5 critical histidine residues that are involved in pH-sensing of OGR1, so G2A may be less sensitive to protons[83]. Later, Obinata et al[91] found that G2A can respond to oxidized free fatty acid (9-hydroxyoctadecadienoic acid, 9-HODE). Recent studies with gene-knockout techniques have revealed the absence of some but not all pH-induced cellular effects in OGR1-, TDAG8- or GPR4-deficient mice or cells, so OGR1 family members are indeed involved in proton sensing, and the pH-dependent activities could be highly cell-type- or signaling-pathway-specific[90,92-94]. Interestingly, mice lacking G2A show some deficiencies in LPC- or acid-related cellular effects[90,95-97]. Whether G2A is a proton, LPC or fatty acid receptor remains debated.

Proton-sensing GPCRs are widely expressed in non-neuronal and neuronal tissues[98]. Approximately 75% to 82% of OGR1 family members are found in small-diameter DRG neurons responsible for nociception and 61% to 74% are present in isolectin B(4) (IB4)-positive neurons, so they may be involved in chronic pain[79,98]. Indeed, one of the members, TDAG8, showed increased expression after CFA-induced inflammation, and its activation sensitizes TRPV1 function[79]. TDAG8 is involved in CFA-induced inflammatory pain by modulating TRPV1 function. Later, knockdown of spinal TDAG8 expression was found to reduce bone cancer pain[99]. Thus, TDAG8 could have pro-nociceptive roles in the peripheral and central nervous system. Although a recent study suggested that TDAG8 is a negative regulator in inflammation because of exacerbation of arthritis induced by anti-type II collagen antibody in TDAG8-deficient mice, whether TDAG8 has an anti-nociceptive role in inflammatory pain remains unclear[100].

In endothelial cells, G2A expression blocks NF-κB activation and chemokine expression, thus inhibiting macrophage accumulation, which suggests that G2A expression may have a protective role in preventing early events of inflammation[96]. This situation could explain why G2A expression is downregulated in capsaicin- and CFA-induced inflammatory pain, so G2A could have an anti-nociceptive role in inflammatory pain[79]. GPR4 is present in endothelial cells of blood vessels, and mice lacking GPR4 show vascular abnormalities, which suggests that GPR4 has a role in vascular growth and vascular stability[93]. Vascular stability is important for leukocyte adhesion and function[101]. GPR4 antagonism attenuates acidosis-induced inflammation and modulate a wide range of inflammatory genes in endothelial cells[102].

SEROTONIN AND SEROTONIN RECEPTORS

In the periphery, serotonin (5-HT) released from platelets, mast cells, and endothelial cells into the inflamed site is pro-inflammatory and pro-nociceptive, exciting nociceptive afferents and inducing hyperalgesia[9,103-106]. In central loci, the descending pathway on serotonergic neurons from the rostral ventromedial medulla to the spinal cord has facilitatory or inhibitory effects on DRG neurons depending on the activation of 5-HT receptor subtypes[107]. Seven subgroups of serotonin receptors (5-HT1-7) have been identified, and some subtypes have more than one receptor (e.g., 5-HT1 has 5-HT1A, 5-HT1B, 5-HT1D, 5-HT1E, and 5-HT1F; and 5-HT2 has 5-HT2A, 5-HT2B, and 5-HT2C)[108]. Although Sufka et al[103] (1992) suggested that all of the 5-HT1A, 5-HT2A, and 5-HT3 subtypes participate in 5-HT-induced pain, the presence of multiple 5-HT receptors on afferent nociceptors reflects distinct pain models or mechanisms.

Taiwo et al[104] reported that only the 5-HT1A agonist mimics the 5-HT effect to induce hyperalgesia and 5-HT1A antagonists block mechanical hyperalgesia induced by 5-HT. Nevertheless, Kayser et al[109] suggested that mice lacking 5-HT1A show increased sensitivity to noxious heat but not mechanical pain stimuli. The other study of formalin testing also suggested that 5-HT1A mediates antinociception[110]. In addition to 5-HT1A, the receptors 5-HT1B, 5-HT1D and 5-HT1F also have anti-nociceptive effects in heat-evoked or formalin-induced nociceptive responses[109,110]. Later, 5-HT2B/2C but not 5-HT1A was found to mediate 5-HT-induced mechanical hyperalgesia[111]. Spinal and peripheral injection of a specific antagonist (RS127445) of 5-HT2B reduced formalin-induced flinching behavior, which suggests that 5-HT2B has a pro-nociceptive role in peripheral as well as spinal loci[112]. However, Urtikova et al[113] suggested that blockage of peripheral or spinal 5-HT2B by a specific antagonist (RS127445) could enhance hyperalgesia induced by chronic constriction nerve injury. 5-HT2B may have distinct roles in different pain models.

Ionotropic 5-HT3 is directly responsible for inflammatory pain[109,114,115]. Lack of the 5-HT3 gene in mice or blocking with the 5-HT3 antagonist granisetron elicited normal acute pain responses but reduced persistent pain responses[109,115]. Giordano et al[116] showed that 5-HT3 contributes to chemical but not thermal and mechanical nociceptive pain. 5-HT2A potentiates the effects of other inflammatory mediators[117]. In the study by Tokunaga et al[118], only the 5-HT2A agonist but not 5-HT1A and 5-HT3A agonists mimicked 5-HT-induced thermal hyperalgesia, which was blocked by the 5-HT2A antagonist ketanserin. However, Loyd et al[119] suggested that both 5-HT-induced and 5-HT-enhanced capsaicin-evoked thermal hyperalgesia require 5-HT2A and 5-HT3. Likely, 5-HT2A potentiates 5-HT3-mediated nociceptive responses to thermal stimuli. Recent studies show that 5-HT2A has a pronociceptive role in spinal nociceptive transmission and seems to be involved in both mechanical and thermal hyperalgesia in the spinal nerve ligation model[120,121].

In addition, 5-HT4 enhances the inflammatory pain signal[122]. 5-HT7 inhibits capsaicin-induced mechanical sensitivity[123]. Intrathecal injection of 5-HT2A, 5-HT3 and 5-HT4 antagonists significantly reduced spinal cord stimulation-induced long-lasting pain in rat models, with no effect by administration of 5-HT1,6,7 antagonists[124]. 5-HT2 and 5-HT7 are major receptors to potentiate TRPV1 function in inflammatory pain[76].

PROSTAGLANDIN E2

Prostaglandin E2 (PGE2), derived from an arachidonic acid by the cyclooxygenase (COX) pathway, is released from damaged cells and contributes to inflammatory pain[125]. Non-steroidal anti-inflammatory drugs are the commonly used analgesics that reduce prostaglandin synthesis by inhibiting COX-1 and COX-2[126]. Four subtypes of PGE2 receptors (EP1, EP2, EP3 and EP4) belong to GPCRs[125,127]. The roles of PGE2 receptor subtypes in pain are undefined because of inconsistent results from studies involving gene targeting techniques, but are better resolved in combined studies with pharmacological approaches[126]. PGE2-induced thermal hyperalgesia is mediated by EP1 predominantly through a PKC-dependent pathway and is due to potentiation or sensitization of TRPV1[77]. Wang and colleagues showed that PGE2-induced pain is mediated by EP3 though PKA and Epac/PKC pathways to sensitize purinergic P2X3 receptors[78,128]. Several lines of evidence also support the roles of PGE2 in modulating pain transduction. PGE2 potentiates the TRPV1 function in response to capsaicin[78]. Repeated administration of PGE2 sensitizes T-type calcium channels, thus resulting in mechanical hyperalgesia[129]. PGE2 potentiates the voltage-gated tetrodotoxin-resistant sodium channels (Nav1.5, Nav1.8 and Nav1.9) by a cAMP-PKA signaling pathway[130,131].

TRANSITION FROM ACUTE TO CHRONIC PAIN

The possible mechanisms of chronic inflammatory pain could be that continuous sensitization induced by inflammatory mediators in primary afferent nociceptors results in persistent and long-lasting pain or neuroplastic changes in primary afferent nociceptors after initiating insults lead to enhanced and prolonged sensitization of nociceptors even with low-level exposure of pro-nociceptive inflammatory mediators. The mechanisms of chronic pain and the regulation of the transition from short-term to long-lasting pain have become clearer from studies with the PGE2 priming model.

Administration of PGE2 in rat induces short-term hyperalgesia that depends on PKA activity[132]. With carrageenan pre-injection, rats display long-lasting hyperalgesia induced by PGE2, and the prolonged effect can be inhibited by PKCε blocker or attenuated by antisense oligonucleotides for PKCε[133,134]. Therefore, PKCε may be necessary to maintain hyperalgesic priming. Indeed, a highly selective PKC agonist can induce hyperalgesic priming in rat[134]. In contrast, the study by Ferrari et al[135] proposed that a transient decrease in GRK2 levels leads to increased nociceptor response to inflammatory mediators, and the reduced GRK2 levels are PKA- but not PKC-dependent. Ferrari et al[136] later proposed that the prolongation of PGE2-induced hyperalgesia is mediated by an autocrine mechanism. PGE2 activates EP receptors followed by cAMP production, which in turn activates PKA and induces hyperalgesia. The increase in intracellular cAMP level triggers the transporter to transport cAMP outside the cell. The extracellular cAMP is metabolized to AMP and adenosine, thus activating the Gi-coupled A1 adenosine receptor. The Gi pathway stimulates PKCε, which is responsible for the late phase of PGE2-induced hyperalgesia, although evidence has shown that after injury, the inflammatory mediators may release and reach the effective concentration in a different time course. Each mediator activates its own receptor subtypes, thus contributing to the development of hyperalgesia. However, which receptor is the major receptor causing the acute to chronic pain remains unclear.

ESTABLISHMENT AND MAINTENANCE OF CHRONIC PAIN: THE ROLE OF AN EXCITATORY AMINO ACID IN CENTRAL SENSITIZATION

The establishment and maintenance of chronic pain is not simply a reflection of peripheral inputs or abnormality but is also a dynamic reflection of central neuronal plasticity. Once the central sensitization occurs, painful sensations are generated even in the absence of the noxious stimulus[137]. Several lines of evidence implicate the contribution of excitatory amino acids in neuroplasticity and central sensitization in the spinal cord. Noxious stimulation or peripheral inflammation causes the release of an excitatory amino acid, glutamate, in the spinal dorsal horn[138,139]. Dorsal horn neurons that are sensitized with peripheral inflammation show increased responsiveness to the iontophoretic application of the excitatory amino acid[139,140], and such responsiveness or sensitization is reduced after the administration of glutamate receptor antagonists[141,142].

Glutamate receptors include ionotropic amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA), N-methyl-D-aspartate (NMDA), kainate receptors and metabotropic G-protein-coupled glutamate receptors (mGluRs). The contribution of ionotropic glutamate receptors to the central sensitization are considered the ability of AMPA and NMDA receptor antagonists to reduce the responsiveness of dorsal horn neurons and in producing analgesic effects[142]. Intrathecal injection of NMDA leads to hyperalgesia, which can be reversed by application of an NMDA antagonist[143]. The NMDA antagonist MK-801 reduces the hyperalgesia that develops in rats with adjuvant-induced inflammation[144] or reduces the inflammation-induced expansion of the receptive field of spinal nociceptive neurons[145].

Peripheral inflammation elevates levels of phosphorylated NMDA receptors in the spinal dorsal horn[146,147]. The sustained release of the neuropeptides (such as substance P and CGRP) and glutamate causes PKC activation and Ca2+ influxes through NMDA receptors. With Ca2+ influx, several intracellular signal pathways, including the phospholipase C-PKC pathway, phosphotidylinositol-3-kinase (PI3K) pathway, and mitogen-activated protein kinase (MAPK) pathway, are activated. Activated intracellular signaling pathways result in phosphorylation of spinal NMDA receptors, enhancing Ca2+ currents at NMDA receptors. Activated intracellular signaling pathways also phosphorylate AMPA receptors, thus increasing the density of AMPA receptors on the membrane[148]. These mechanisms create a positive feedback loop for glutamate transmission and alter the neuronal plasticity in the dorsal horn. In the formalin-induced inflammatory pain model, intrathecal injection of the MEK inhibitor PD98059 can reduce the second phase of the licking/lifting behavior and attenuate extracellular signal-regulated kinase activity, so some intracellular signaling pathways may also be involved in central sensitization[149].

CONCLUSION

At the inflamed site of the tissue, endogenous mediators (5-HT, PGE2, BK, and proton) are released from damaged cells and accumulate. Nociceptors innervating the skin, muscle and organs detect the noxious stimuli and express one or more cell-surface receptors to respond to these inflammatory mediators. The mediators can directly or indirectly alter the sensitivity of the receptors on nociceptors. ASIC3, ASIC1a and TRPV1 seem to be important transducer receptors contributing to hyperalgesia induced by inflammation. ASIC1a participates in primary mechanical hyperalgesia induced by muscle inflammation, but ASIC3 may have a predominant role in secondary mechanical hyperalgesia. TRPV1 could be responsible for mechanical and thermal hyperalgesia induced by cutaneous inflammation. Inflammatory mediators such as 5-HT, PGE2, BK, and proton sensitize TRPV1 or ASIC3 to prolong the hyperalgesia. PGE2 acts on EP1 to sensitize TRPV1 or on EP3 to sensitize P2X3. Proton and BK sensitize TRPV1 though TDAG8 and B2, respectively. 5-HT potentiates TRPV1 function, possibly through 5-HT2 and 5-HT7. Although each mediator receptor has its own dominant second-messenger signaling cascade, each could also be coupled in part to other second-messenger pathways. For short-term hyperalgesia, the cAMP-PKA pathway is dominant, but prolonged hyperalgesia is regulated by PKCε-dependent or -independent pathway.

The signal of a stimulus is triggered by a peripheral nociceptor, followed by conduction to central neurons. In acute pain, the signal is mediated by glutamate acting on AMPA and kainate subtypes of ionotropic glutamate receptors of postsynaptic neurons and generating the excitatory postsynaptic potential. If the signal is generated by intense or persistent noxious stimuli, the depolarization of the postsynaptic neurons will activate NMDA receptors. NMDA receptor activation induces Ca2+ influx, which activates intracellular signaling pathways to further enhance Ca2+ influx through NMDA receptors. NMDA receptors can also be modulated by GPCRs such as NK1, EP or mGlu receptors that are also expressed on the superficial dorsal horn of nociceptor terminals[150]. All these NMDA-receptor-mediated mechanisms contribute to central sensitization, which is important for establishing and maintaining chronic pain[151].

Footnotes

P- Reviewer: Yousef AA S- Editor: Song XX L- Editor: A E- Editor: Wang CH

References
1.  Marchand F, Perretti M, McMahon SB. Role of the immune system in chronic pain. Nat Rev Neurosci. 2005;6:521-532.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 767]  [Cited by in F6Publishing: 823]  [Article Influence: 43.3]  [Reference Citation Analysis (0)]
2.  Basbaum AI, Bautista DM, Scherrer G, Julius D. Cellular and molecular mechanisms of pain. Cell. 2009;139:267-284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2363]  [Cited by in F6Publishing: 2525]  [Article Influence: 168.3]  [Reference Citation Analysis (0)]
3.  Julius D, Basbaum AI. Molecular mechanisms of nociception. Nature. 2001;413:203-210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1718]  [Cited by in F6Publishing: 1627]  [Article Influence: 70.7]  [Reference Citation Analysis (0)]
4.  Scholz J, Woolf CJ. Can we conquer pain? Nat Neurosci. 2002;5 Suppl:1062-1067.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 593]  [Cited by in F6Publishing: 549]  [Article Influence: 25.0]  [Reference Citation Analysis (2)]
5.  Beck PW, Handwerker HO. Bradykinin and serotonin effects on various types of cutaneous nerve fibers. Pflugers Arch. 1974;347:209-222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 260]  [Cited by in F6Publishing: 284]  [Article Influence: 5.7]  [Reference Citation Analysis (1)]
6.  Schaible HG, Schmidt RF. Excitation and sensitization of fine articular afferents from cat’s knee joint by prostaglandin E2. J Physiol. 1988;403:91-104.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Steen KH, Reeh PW, Anton F, Handwerker HO. Protons selectively induce lasting excitation and sensitization to mechanical stimulation of nociceptors in rat skin, in vitro. J Neurosci. 1992;12:86-95.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Steen KH, Reeh PW, Kreysel HW. Topical acetylsalicylic, salicylic acid and indomethacin suppress pain from experimental tissue acidosis in human skin. Pain. 1995;62:339-347.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 44]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
9.  Kessler W, Kirchhoff C, Reeh PW, Handwerker HO. Excitation of cutaneous afferent nerve endings in vitro by a combination of inflammatory mediators and conditioning effect of substance P. Exp Brain Res. 1992;91:467-476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 124]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
10.  Steen KH, Steen AE, Kreysel HW, Reeh PW. Inflammatory mediators potentiate pain induced by experimental tissue acidosis. Pain. 1996;66:163-170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 90]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
11.  Dubé GR, Elagoz A, Mangat H. Acid sensing ion channels and acid nociception. Curr Pharm Des. 2009;15:1750-1766.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
12.  Woo YC, Park SS, Subieta AR, Brennan TJ. Changes in tissue pH and temperature after incision indicate acidosis may contribute to postoperative pain. Anesthesiology. 2004;101:468-475.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 132]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
13.  Jacobus WE, Taylor GJ, Hollis DP, Nunnally RL. Phosphorus nuclear magnetic resonance of perfused working rat hearts. Nature. 1977;265:756-758.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 227]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
14.  Pan HL, Longhurst JC, Eisenach JC, Chen SR. Role of protons in activation of cardiac sympathetic C-fibre afferents during ischaemia in cats. J Physiol. 1999;518:857-866.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 103]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
15.  Helmlinger G, Yuan F, Dellian M, Jain RK. Interstitial pH and pO2 gradients in solid tumors in vivo: high-resolution measurements reveal a lack of correlation. Nat Med. 1997;3:177-182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1206]  [Cited by in F6Publishing: 1110]  [Article Influence: 41.1]  [Reference Citation Analysis (0)]
16.  Steen KH, Reeh PW. Sustained graded pain and hyperalgesia from harmless experimental tissue acidosis in human skin. Neurosci Lett. 1993;154:113-116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 138]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
17.  Steen KH, Issberner U, Reeh PW. Pain due to experimental acidosis in human skin: evidence for non-adapting nociceptor excitation. Neurosci Lett. 1995;199:29-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 94]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
18.  Steen KH, Steen AE, Reeh PW. A dominant role of acid pH in inflammatory excitation and sensitization of nociceptors in rat skin, in vitro. J Neurosci. 1995;15:3982-3989.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Bevan S, Yeats J. Protons activate a cation conductance in a sub-population of rat dorsal root ganglion neurones. J Physiol. 1991;433:145-161.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Kress M, Reeh PW, Vyklicky L. An interaction of inflammatory mediators and protons in small diameter dorsal root ganglion neurons of the rat. Neurosci Lett. 1997;224:37-40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 45]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
21.  Krishtal OA, Pidoplichko VI. A receptor for protons in the nerve cell membrane. Neuroscience. 1980;5:2325-2327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 331]  [Cited by in F6Publishing: 372]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
22.  Waldmann R, Champigny G, Bassilana F, Heurteaux C, Lazdunski M. A proton-gated cation channel involved in acid-sensing. Nature. 1997;386:173-177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1033]  [Cited by in F6Publishing: 1053]  [Article Influence: 39.0]  [Reference Citation Analysis (0)]
23.  Chen CC, England S, Akopian AN, Wood JN. A sensory neuron-specific, proton-gated ion channel. Proc Natl Acad Sci USA. 1998;95:10240-10245.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 373]  [Cited by in F6Publishing: 371]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
24.  Waldmann R, Bassilana F, de Weille J, Champigny G, Heurteaux C, Lazdunski M. Molecular cloning of a non-inactivating proton-gated Na+ channel specific for sensory neurons. J Biol Chem. 1997;272:20975-20978.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 416]  [Cited by in F6Publishing: 438]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
25.  Bassilana F, Champigny G, Waldmann R, de Weille JR, Heurteaux C, Lazdunski M. The acid-sensitive ionic channel subunit ASIC and the mammalian degenerin MDEG form a heteromultimeric H+-gated Na+ channel with novel properties. J Biol Chem. 1997;272:28819-28822.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 185]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
26.  Deval E, Gasull X, Noël J, Salinas M, Baron A, Diochot S, Lingueglia E. Acid-sensing ion channels (ASICs): pharmacology and implication in pain. Pharmacol Ther. 2010;128:549-558.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 229]  [Cited by in F6Publishing: 241]  [Article Influence: 17.2]  [Reference Citation Analysis (0)]
27.  Wemmie JA, Taugher RJ, Kreple CJ. Acid-sensing ion channels in pain and disease. Nat Rev Neurosci. 2013;14:461-471.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 401]  [Cited by in F6Publishing: 432]  [Article Influence: 39.3]  [Reference Citation Analysis (1)]
28.  Wiemuth D, Sahin H, Falkenburger BH, Lefèvre CM, Wasmuth HE, Gründer S. BASIC--a bile acid-sensitive ion channel highly expressed in bile ducts. FASEB J. 2012;26:4122-4130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 33]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
29.  Sutherland SP, Benson CJ, Adelman JP, McCleskey EW. Acid-sensing ion channel 3 matches the acid-gated current in cardiac ischemia-sensing neurons. Proc Natl Acad Sci USA. 2001;98:711-716.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 234]  [Cited by in F6Publishing: 228]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
30.  Yagi J, Wenk HN, Naves LA, McCleskey EW. Sustained currents through ASIC3 ion channels at the modest pH changes that occur during myocardial ischemia. Circ Res. 2006;99:501-509.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 188]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
31.  Wu WL, Cheng CF, Sun WH, Wong CW, Chen CC. Targeting ASIC3 for pain, anxiety, and insulin resistance. Pharmacol Ther. 2012;134:127-138.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 54]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
32.  Voilley N, de Weille J, Mamet J, Lazdunski M. Nonsteroid anti-inflammatory drugs inhibit both the activity and the inflammation-induced expression of acid-sensing ion channels in nociceptors. J Neurosci. 2001;21:8026-8033.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Mamet J, Baron A, Lazdunski M, Voilley N. Proinflammatory mediators, stimulators of sensory neuron excitability via the expression of acid-sensing ion channels. J Neurosci. 2002;22:10662-10670.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Deval E, Noël J, Lay N, Alloui A, Diochot S, Friend V, Jodar M, Lazdunski M, Lingueglia E. ASIC3, a sensor of acidic and primary inflammatory pain. EMBO J. 2008;27:3047-3055.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 294]  [Cited by in F6Publishing: 310]  [Article Influence: 19.4]  [Reference Citation Analysis (0)]
35.  Allen NJ, Attwell D. Modulation of ASIC channels in rat cerebellar Purkinje neurons by ischaemia-related signals. J Physiol. 2002;543:521-529.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 124]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
36.  Smith ES, Cadiou H, McNaughton PA. Arachidonic acid potentiates acid-sensing ion channels in rat sensory neurons by a direct action. Neuroscience. 2007;145:686-698.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 100]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
37.  Cadiou H, Studer M, Jones NG, Smith ES, Ballard A, McMahon SB, McNaughton PA. Modulation of acid-sensing ion channel activity by nitric oxide. J Neurosci. 2007;27:13251-13260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 100]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
38.  Wang X, Li WG, Yu Y, Xiao X, Cheng J, Zeng WZ, Peng Z, Xi Zhu M, Xu TL. Serotonin facilitates peripheral pain sensitivity in a manner that depends on the nonproton ligand sensing domain of ASIC3 channel. J Neurosci. 2013;33:4265-4279.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 48]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
39.  Price MP, McIlwrath SL, Xie J, Cheng C, Qiao J, Tarr DE, Sluka KA, Brennan TJ, Lewin GR, Welsh MJ. The DRASIC cation channel contributes to the detection of cutaneous touch and acid stimuli in mice. Neuron. 2001;32:1071-1083.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 468]  [Cited by in F6Publishing: 452]  [Article Influence: 19.7]  [Reference Citation Analysis (0)]
40.  Chen CC, Wong CW. Neurosensory mechanotransduction through acid-sensing ion channels. J Cell Mol Med. 2013;17:337-349.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 74]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
41.  Chen CC, Zimmer A, Sun WH, Hall J, Brownstein MJ, Zimmer A. A role for ASIC3 in the modulation of high-intensity pain stimuli. Proc Natl Acad Sci USA. 2002;99:8992-8997.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 237]  [Cited by in F6Publishing: 240]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
42.  Ikeuchi M, Kolker SJ, Burnes LA, Walder RY, Sluka KA. Role of ASIC3 in the primary and secondary hyperalgesia produced by joint inflammation in mice. Pain. 2008;137:662-669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
43.  Walder RY, Rasmussen LA, Rainier JD, Light AR, Wemmie JA, Sluka KA. ASIC1 and ASIC3 play different roles in the development of Hyperalgesia after inflammatory muscle injury. J Pain. 2010;11:210-218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
44.  Deval E, Noël J, Gasull X, Delaunay A, Alloui A, Friend V, Eschalier A, Lazdunski M, Lingueglia E. Acid-sensing ion channels in postoperative pain. J Neurosci. 2011;31:6059-6066.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 130]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
45.  Molliver DC, Immke DC, Fierro L, Paré M, Rice FL, McCleskey EW. ASIC3, an acid-sensing ion channel, is expressed in metaboreceptive sensory neurons. Mol Pain. 2005;1:35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 170]  [Cited by in F6Publishing: 183]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
46.  Sluka KA, Price MP, Breese NM, Stucky CL, Wemmie JA, Welsh MJ. Chronic hyperalgesia induced by repeated acid injections in muscle is abolished by the loss of ASIC3, but not ASIC1. Pain. 2003;106:229-239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 345]  [Cited by in F6Publishing: 344]  [Article Influence: 17.2]  [Reference Citation Analysis (0)]
47.  Sluka KA, Radhakrishnan R, Benson CJ, Eshcol JO, Price MP, Babinski K, Audette KM, Yeomans DC, Wilson SP. ASIC3 in muscle mediates mechanical, but not heat, hyperalgesia associated with muscle inflammation. Pain. 2007;129:102-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 150]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
48.  Yen YT, Tu PH, Chen CJ, Lin YW, Hsieh ST, Chen CC. Role of acid-sensing ion channel 3 in sub-acute-phase inflammation. Mol Pain. 2009;5:1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 93]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
49.  Walder RY, Gautam M, Wilson SP, Benson CJ, Sluka KA. Selective targeting of ASIC3 using artificial miRNAs inhibits primary and secondary hyperalgesia after muscle inflammation. Pain. 2011;152:2348-2356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 58]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
50.  Lin CC, Chen WN, Chen CJ, Lin YW, Zimmer A, Chen CC. An antinociceptive role for substance P in acid-induced chronic muscle pain. Proc Natl Acad Sci USA. 2012;109:E76-E83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 71]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
51.  Alvarez de la Rosa D, Zhang P, Shao D, White F, Canessa CM. Functional implications of the localization and activity of acid-sensitive channels in rat peripheral nervous system. Proc Natl Acad Sci USA. 2002;99:2326-2331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 199]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
52.  Page AJ, Brierley SM, Martin CM, Price MP, Symonds E, Butler R, Wemmie JA, Blackshaw LA. Different contributions of ASIC channels 1a, 2, and 3 in gastrointestinal mechanosensory function. Gut. 2005;54:1408-1415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 217]  [Cited by in F6Publishing: 221]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
53.  Page AJ, Brierley SM, Martin CM, Martinez-Salgado C, Wemmie JA, Brennan TJ, Symonds E, Omari T, Lewin GR, Welsh MJ. The ion channel ASIC1 contributes to visceral but not cutaneous mechanoreceptor function. Gastroenterology. 2004;127:1739-1747.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 112]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
54.  Duan B, Wu LJ, Yu YQ, Ding Y, Jing L, Xu L, Chen J, Xu TL. Upregulation of acid-sensing ion channel ASIC1a in spinal dorsal horn neurons contributes to inflammatory pain hypersensitivity. J Neurosci. 2007;27:11139-11148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 124]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
55.  Kong X, Tang X, Du W, Tong J, Yan Y, Zheng F, Fang M, Gong F, Tan Z. Extracellular acidosis modulates the endocytosis and maturation of macrophages. Cell Immunol. 2013;281:44-50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 38]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
56.  Kang S, Jang JH, Price MP, Gautam M, Benson CJ, Gong H, Welsh MJ, Brennan TJ. Simultaneous disruption of mouse ASIC1a, ASIC2 and ASIC3 genes enhances cutaneous mechanosensitivity. PLoS One. 2012;7:e35225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
57.  Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D. The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature. 1997;389:816-824.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6360]  [Cited by in F6Publishing: 6414]  [Article Influence: 237.6]  [Reference Citation Analysis (0)]
58.  Tominaga M, Caterina MJ, Malmberg AB, Rosen TA, Gilbert H, Skinner K, Raumann BE, Basbaum AI, Julius D. The cloned capsaicin receptor integrates multiple pain-producing stimuli. Neuron. 1998;21:531-543.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2293]  [Cited by in F6Publishing: 2342]  [Article Influence: 90.1]  [Reference Citation Analysis (0)]
59.  Caterina MJ, Leffler A, Malmberg AB, Martin WJ, Trafton J, Petersen-Zeitz KR, Koltzenburg M, Basbaum AI, Julius D. Impaired nociception and pain sensation in mice lacking the capsaicin receptor. Science. 2000;288:306-313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2577]  [Cited by in F6Publishing: 2615]  [Article Influence: 109.0]  [Reference Citation Analysis (0)]
60.  Davis JB, Gray J, Gunthorpe MJ, Hatcher JP, Davey PT, Overend P, Harries MH, Latcham J, Clapham C, Atkinson K. Vanilloid receptor-1 is essential for inflammatory thermal hyperalgesia. Nature. 2000;405:183-187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1306]  [Cited by in F6Publishing: 1305]  [Article Influence: 54.4]  [Reference Citation Analysis (0)]
61.  Gavva NR, Tamir R, Qu Y, Klionsky L, Zhang TJ, Immke D, Wang J, Zhu D, Vanderah TW, Porreca F. AMG 9810 [(E)-3-(4-t-butylphenyl)-N-(2,3-dihydrobenzo[b][1,4] dioxin-6-yl)acrylamide], a novel vanilloid receptor 1 (TRPV1) antagonist with antihyperalgesic properties. J Pharmacol Exp Ther. 2005;313:474-484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 281]  [Cited by in F6Publishing: 314]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
62.  Ghilardi JR, Röhrich H, Lindsay TH, Sevcik MA, Schwei MJ, Kubota K, Halvorson KG, Poblete J, Chaplan SR, Dubin AE. Selective blockade of the capsaicin receptor TRPV1 attenuates bone cancer pain. J Neurosci. 2005;25:3126-3131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 271]  [Cited by in F6Publishing: 287]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
63.  Cui M, Honore P, Zhong C, Gauvin D, Mikusa J, Hernandez G, Chandran P, Gomtsyan A, Brown B, Bayburt EK. TRPV1 receptors in the CNS play a key role in broad-spectrum analgesia of TRPV1 antagonists. J Neurosci. 2006;26:9385-9393.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 228]  [Cited by in F6Publishing: 231]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
64.  Kim MS, Ryu H, Kang DW, Cho SH, Seo S, Park YS, Kim MY, Kwak EJ, Kim YS, Bhondwe RS. 2-(3-fluoro-4-methylsulfonylaminophenyl)propanamides as potent transient receptor potential vanilloid 1 (TRPV1) antagonists: structure-activity relationships of 2-amino derivatives in the N-(6-trifluoromethylpyridin-3-ylmethyl) C-region. J Med Chem. 2012;55:8392-8408.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 32]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
65.  Yu L, Yang F, Luo H, Liu FY, Han JS, Xing GG, Wan Y. The role of TRPV1 in different subtypes of dorsal root ganglion neurons in rat chronic inflammatory nociception induced by complete Freund’s adjuvant. Mol Pain. 2008;4:61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 137]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
66.  Walder RY, Radhakrishnan R, Loo L, Rasmussen LA, Mohapatra DP, Wilson SP, Sluka KA. TRPV1 is important for mechanical and heat sensitivity in uninjured animals and development of heat hypersensitivity after muscle inflammation. Pain. 2012;153:1664-1672.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 31]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
67.  Carlton SM, Coggeshall RE. Peripheral capsaicin receptors increase in the inflamed rat hindpaw: a possible mechanism for peripheral sensitization. Neurosci Lett. 2001;310:53-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 108]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
68.  Amaya F, Oh-hashi K, Naruse Y, Iijima N, Ueda M, Shimosato G, Tominaga M, Tanaka Y, Tanaka M. Local inflammation increases vanilloid receptor 1 expression within distinct subgroups of DRG neurons. Brain Res. 2003;963:190-196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 170]  [Cited by in F6Publishing: 178]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
69.  Breese NM, George AC, Pauers LE, Stucky CL. Peripheral inflammation selectively increases TRPV1 function in IB4-positive sensory neurons from adult mouse. Pain. 2005;115:37-49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 106]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
70.  Lu J, Zhou XF, Rush RA. Small primary sensory neurons innervating epidermis and viscera display differential phenotype in the adult rat. Neurosci Res. 2001;41:355-363.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 58]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
71.  Plenderleith MB, Snow PJ. The plant lectin Bandeiraea simplicifolia I-B4 identifies a subpopulation of small diameter primary sensory neurones which innervate the skin in the rat. Neurosci Lett. 1993;159:17-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 75]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
72.  Okun A, DeFelice M, Eyde N, Ren J, Mercado R, King T, Porreca F. Transient inflammation-induced ongoing pain is driven by TRPV1 sensitive afferents. Mol Pain. 2011;7:4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 65]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
73.  Chuang HH, Prescott ED, Kong H, Shields S, Jordt SE, Basbaum AI, Chao MV, Julius D. Bradykinin and nerve growth factor release the capsaicin receptor from PtdIns(4,5)P2-mediated inhibition. Nature. 2001;411:957-962.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 916]  [Cited by in F6Publishing: 940]  [Article Influence: 40.9]  [Reference Citation Analysis (0)]
74.  Premkumar LS, Ahern GP. Induction of vanilloid receptor channel activity by protein kinase C. Nature. 2000;408:985-990.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 602]  [Cited by in F6Publishing: 623]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
75.  Zhang N, Inan S, Cowan A, Sun R, Wang JM, Rogers TJ, Caterina M, Oppenheim JJ. A proinflammatory chemokine, CCL3, sensitizes the heat- and capsaicin-gated ion channel TRPV1. Proc Natl Acad Sci USA. 2005;102:4536-4541.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
76.  Ohta T, Ikemi Y, Murakami M, Imagawa T, Otsuguro K, Ito S. Potentiation of transient receptor potential V1 functions by the activation of metabotropic 5-HT receptors in rat primary sensory neurons. J Physiol. 2006;576:809-822.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 82]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
77.  Moriyama T, Higashi T, Togashi K, Iida T, Segi E, Sugimoto Y, Tominaga T, Narumiya S, Tominaga M. Sensitization of TRPV1 by EP1 and IP reveals peripheral nociceptive mechanism of prostaglandins. Mol Pain. 2005;1:3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 363]  [Cited by in F6Publishing: 395]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
78.  Wang C, Gu Y, Li GW, Huang LY. A critical role of the cAMP sensor Epac in switching protein kinase signalling in prostaglandin E2-induced potentiation of P2X3 receptor currents in inflamed rats. J Physiol. 2007;584:191-203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 79]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
79.  Chen YJ, Huang CW, Lin CS, Chang WH, Sun WH. Expression and function of proton-sensing G-protein-coupled receptors in inflammatory pain. Mol Pain. 2009;5:39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 36]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
80.  Amadesi S, Nie J, Vergnolle N, Cottrell GS, Grady EF, Trevisani M, Manni C, Geppetti P, McRoberts JA, Ennes H. Protease-activated receptor 2 sensitizes the capsaicin receptor transient receptor potential vanilloid receptor 1 to induce hyperalgesia. J Neurosci. 2004;24:4300-4312.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 299]  [Cited by in F6Publishing: 322]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
81.  Amadesi S, Cottrell GS, Divino L, Chapman K, Grady EF, Bautista F, Karanjia R, Barajas-Lopez C, Vanner S, Vergnolle N. Protease-activated receptor 2 sensitizes TRPV1 by protein kinase Cepsilon- and A-dependent mechanisms in rats and mice. J Physiol. 2006;575:555-571.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 226]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
82.  Schwartz ES, La JH, Scheff NN, Davis BM, Albers KM, Gebhart GF. TRPV1 and TRPA1 antagonists prevent the transition of acute to chronic inflammation and pain in chronic pancreatitis. J Neurosci. 2013;33:5603-5611.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 129]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
83.  Ludwig MG, Vanek M, Guerini D, Gasser JA, Jones CE, Junker U, Hofstetter H, Wolf RM, Seuwen K. Proton-sensing G-protein-coupled receptors. Nature. 2003;425:93-98.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 521]  [Cited by in F6Publishing: 518]  [Article Influence: 24.7]  [Reference Citation Analysis (0)]
84.  Murakami N, Yokomizo T, Okuno T, Shimizu T. G2A is a proton-sensing G-protein-coupled receptor antagonized by lysophosphatidylcholine. J Biol Chem. 2004;279:42484-42491.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 187]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
85.  Wang JQ, Kon J, Mogi C, Tobo M, Damirin A, Sato K, Komachi M, Malchinkhuu E, Murata N, Kimura T. TDAG8 is a proton-sensing and psychosine-sensitive G-protein-coupled receptor. J Biol Chem. 2004;279:45626-45633.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 177]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
86.  Ishii S, Kihara Y, Shimizu T. Identification of T cell death-associated gene 8 (TDAG8) as a novel acid sensing G-protein-coupled receptor. J Biol Chem. 2005;280:9083-9087.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 148]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
87.  Kabarowski JH, Zhu K, Le LQ, Witte ON, Xu Y. Lysophosphatidylcholine as a ligand for the immunoregulatory receptor G2A. Science. 2001;293:702-705.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 249]  [Cited by in F6Publishing: 246]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
88.  Xu Y, Zhu K, Hong G, Wu W, Baudhuin LM, Xiao Y, Damron DS. Sphingosylphosphorylcholine is a ligand for ovarian cancer G-protein-coupled receptor 1. Nat Cell Biol. 2000;2:261-267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 237]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
89.  Zhu K, Baudhuin LM, Hong G, Williams FS, Cristina KL, Kabarowski JH, Witte ON, Xu Y. Sphingosylphosphorylcholine and lysophosphatidylcholine are ligands for the G protein-coupled receptor GPR4. J Biol Chem. 2001;276:41325-41335.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 185]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
90.  Radu CG, Nijagal A, McLaughlin J, Wang L, Witte ON. Differential proton sensitivity of related G protein-coupled receptors T cell death-associated gene 8 and G2A expressed in immune cells. Proc Natl Acad Sci USA. 2005;102:1632-1637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 157]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
91.  Obinata H, Hattori T, Nakane S, Tatei K, Izumi T. Identification of 9-hydroxyoctadecadienoic acid and other oxidized free fatty acids as ligands of the G protein-coupled receptor G2A. J Biol Chem. 2005;280:40676-40683.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 127]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
92.  Radu CG, Cheng D, Nijagal A, Riedinger M, McLaughlin J, Yang LV, Johnson J, Witte ON. Normal immune development and glucocorticoid-induced thymocyte apoptosis in mice deficient for the T-cell death-associated gene 8 receptor. Mol Cell Biol. 2006;26:668-677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 60]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
93.  Yang LV, Radu CG, Roy M, Lee S, McLaughlin J, Teitell MA, Iruela-Arispe ML, Witte ON. Vascular abnormalities in mice deficient for the G protein-coupled receptor GPR4 that functions as a pH sensor. Mol Cell Biol. 2007;27:1334-1347.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 99]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
94.  Li H, Wang D, Singh LS, Berk M, Tan H, Zhao Z, Steinmetz R, Kirmani K, Wei G, Xu Y. Abnormalities in osteoclastogenesis and decreased tumorigenesis in mice deficient for ovarian cancer G protein-coupled receptor 1. PLoS One. 2009;4:e5705.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 65]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
95.  Le LQ, Kabarowski JH, Weng Z, Satterthwaite AB, Harvill ET, Jensen ER, Miller JF, Witte ON. Mice lacking the orphan G protein-coupled receptor G2A develop a late-onset autoimmune syndrome. Immunity. 2001;14:561-571.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 146]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
96.  Bolick DT, Whetzel AM, Skaflen M, Deem TL, Lee J, Hedrick CC. Absence of the G protein-coupled receptor G2A in mice promotes monocyte/endothelial interactions in aorta. Circ Res. 2007;100:572-580.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 33]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
97.  Yan JJ, Jung JS, Lee JE, Lee J, Huh SO, Kim HS, Jung KC, Cho JY, Nam JS, Suh HW. Therapeutic effects of lysophosphatidylcholine in experimental sepsis. Nat Med. 2004;10:161-167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 247]  [Cited by in F6Publishing: 254]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
98.  Huang CW, Tzeng JN, Chen YJ, Tsai WF, Chen CC, Sun WH. Nociceptors of dorsal root ganglion express proton-sensing G-protein-coupled receptors. Mol Cell Neurosci. 2007;36:195-210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 74]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
99.  Hang LH, Yang JP, Yin W, Wang LN, Guo F, Ji FH, Shao DH, Xu QN, Wang XY, Zuo JL. Activation of spinal TDAG8 and its downstream PKA signaling pathway contribute to bone cancer pain in rats. Eur J Neurosci. 2012;36:2107-2117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 34]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
100.  Onozawa Y, Komai T, Oda T. Activation of T cell death-associated gene 8 attenuates inflammation by negatively regulating the function of inflammatory cells. Eur J Pharmacol. 2011;654:315-319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 25]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
101.  Danese S, Papa A, Saibeni S, Repici A, Malesci A, Vecchi M. Inflammation and coagulation in inflammatory bowel disease: The clot thickens. Am J Gastroenterol. 2007;102:174-186.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Dong L, Li Z, Leffler NR, Asch AS, Chi JT, Yang LV. Acidosis activation of the proton-sensing GPR4 receptor stimulates vascular endothelial cell inflammatory responses revealed by transcriptome analysis. PLoS One. 2013;8:e61991.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 112]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
103.  Sufka KJ, Schomburg FM, Giordano J. Receptor mediation of 5-HT-induced inflammation and nociception in rats. Pharmacol Biochem Behav. 1992;41:53-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 95]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
104.  Taiwo YO, Levine JD. Serotonin is a directly-acting hyperalgesic agent in the rat. Neuroscience. 1992;48:485-490.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 168]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
105.  Ernberg M, Lundeberg T, Kopp S. Pain and allodynia/hyperalgesia induced by intramuscular injection of serotonin in patients with fibromyalgia and healthy individuals. Pain. 2000;85:31-39.  [PubMed]  [DOI]  [Cited in This Article: ]
106.  Ernberg M, Hedenberg-Magnusson B, Kurita H, Kopp S. Effects of local serotonin administration on pain and microcirculation in the human masseter muscle. J Orofac Pain. 2006;20:241-248.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Millan MJ. Descending control of pain. Prog Neurobiol. 2002;66:355-474.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2145]  [Cited by in F6Publishing: 2099]  [Article Influence: 95.4]  [Reference Citation Analysis (0)]
108.  Hoyer D, Hannon JP, Martin GR. Molecular, pharmacological and functional diversity of 5-HT receptors. Pharmacol Biochem Behav. 2002;71:533-554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1346]  [Cited by in F6Publishing: 1305]  [Article Influence: 59.3]  [Reference Citation Analysis (0)]
109.  Kayser V, Elfassi IE, Aubel B, Melfort M, Julius D, Gingrich JA, Hamon M, Bourgoin S. Mechanical, thermal and formalin-induced nociception is differentially altered in 5-HT1A-/-, 5-HT1B-/-, 5-HT2A-/-, 5-HT3A-/- and 5-HTT-/- knock-out male mice. Pain. 2007;130:235-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 123]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
110.  Granados-Soto V, Argüelles CF, Rocha-González HI, Godínez-Chaparro B, Flores-Murrieta FJ, Villalón CM. The role of peripheral 5-HT1A, 5-HT1B, 5-HT1D, 5-HT1E and 5-HT1F serotonergic receptors in the reduction of nociception in rats. Neuroscience. 2010;165:561-568.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 44]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
111.  Lin SY, Chang WJ, Lin CS, Huang CY, Wang HF, Sun WH. Serotonin receptor 5-HT2B mediates serotonin-induced mechanical hyperalgesia. J Neurosci. 2011;31:1410-1418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 59]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
112.  Cervantes-Durán C, Vidal-Cantú GC, Barragán-Iglesias P, Pineda-Farias JB, Bravo-Hernández M, Murbartián J, Granados-Soto V. Role of peripheral and spinal 5-HT2B receptors in formalin-induced nociception. Pharmacol Biochem Behav. 2012;102:30-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
113.  Urtikova N, Berson N, Van Steenwinckel J, Doly S, Truchetto J, Maroteaux L, Pohl M, Conrath M. Antinociceptive effect of peripheral serotonin 5-HT2B receptor activation on neuropathic pain. Pain. 2012;153:1320-1331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 22]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
114.  Eschalier A, Kayser V, Guilbaud G. Influence of a specific 5-HT3 antagonist on carrageenan-induced hyperalgesia in rats. Pain. 1989;36:249-255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 76]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
115.  Zeitz KP, Guy N, Malmberg AB, Dirajlal S, Martin WJ, Sun L, Bonhaus DW, Stucky CL, Julius D, Basbaum AI. The 5-HT3 subtype of serotonin receptor contributes to nociceptive processing via a novel subset of myelinated and unmyelinated nociceptors. J Neurosci. 2002;22:1010-1019.  [PubMed]  [DOI]  [Cited in This Article: ]
116.  Giordano J, Dyche J. Differential analgesic actions of serotonin 5-HT3 receptor antagonists in the mouse. Neuropharmacology. 1989;28:423-427.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 56]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
117.  Abbott FV, Hong Y, Blier P. Activation of 5-HT2A receptors potentiates pain produced by inflammatory mediators. Neuropharmacology. 1996;35:99-110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 104]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
118.  Tokunaga A, Saika M, Senba E. 5-HT2A receptor subtype is involved in the thermal hyperalgesic mechanism of serotonin in the periphery. Pain. 1998;76:349-355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 83]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
119.  Loyd DR, Chen PB, Hargreaves KM. Anti-hyperalgesic effects of anti-serotonergic compounds on serotonin- and capsaicin-evoked thermal hyperalgesia in the rat. Neuroscience. 2012;203:207-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 38]  [Reference Citation Analysis (0)]
120.  Wang D, Chen T, Gao Y, Quirion R, Hong Y. Inhibition of SNL-induced upregulation of CGRP and NPY in the spinal cord and dorsal root ganglia by the 5-HT(2A) receptor antagonist ketanserin in rats. Pharmacol Biochem Behav. 2012;101:379-386.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
121.  Rahman W, Bannister K, Bee LA, Dickenson AH. A pronociceptive role for the 5-HT2 receptor on spinal nociceptive transmission: an in vivo electrophysiological study in the rat. Brain Res. 2011;1382:29-36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 33]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
122.  Doak GJ, Sawynok J. Formalin-induced nociceptive behavior and edema: involvement of multiple peripheral 5-hydroxytryptamine receptor subtypes. Neuroscience. 1997;80:939-949.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 108]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
123.  Brenchat A, Romero L, García M, Pujol M, Burgueño J, Torrens A, Hamon M, Baeyens JM, Buschmann H, Zamanillo D. 5-HT7 receptor activation inhibits mechanical hypersensitivity secondary to capsaicin sensitization in mice. Pain. 2009;141:239-247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 109]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
124.  Song Z, Meyerson BA, Linderoth B. Spinal 5-HT receptors that contribute to the pain-relieving effects of spinal cord stimulation in a rat model of neuropathy. Pain. 2011;152:1666-1673.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 101]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
125.  Narumiya S, Sugimoto Y, Ushikubi F. Prostanoid receptors: structures, properties, and functions. Physiol Rev. 1999;79:1193-1226.  [PubMed]  [DOI]  [Cited in This Article: ]
126.  Kawabata A. Prostaglandin E2 and pain--an update. Biol Pharm Bull. 2011;34:1170-1173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 216]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
127.  Narumiya S, FitzGerald GA. Genetic and pharmacological analysis of prostanoid receptor function. J Clin Invest. 2001;108:25-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 124]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
128.  Wang C, Li GW, Huang LY. Prostaglandin E2 potentiation of P2X3 receptor mediated currents in dorsal root ganglion neurons. Mol Pain. 2007;3:22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 54]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
129.  Sekiguchi F, Aoki Y, Nakagawa M, Kanaoka D, Nishimoto Y, Tsubota-Matsunami M, Yamanaka R, Yoshida S, Kawabata A. AKAP-dependent sensitization of Ca(v) 3.2 channels via the EP(4) receptor/cAMP pathway mediates PGE(2) -induced mechanical hyperalgesia. Br J Pharmacol. 2013;168:734-745.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
130.  England S, Bevan S, Docherty RJ. PGE2 modulates the tetrodotoxin-resistant sodium current in neonatal rat dorsal root ganglion neurones via the cyclic AMP-protein kinase A cascade. J Physiol. 1996;495:429-440.  [PubMed]  [DOI]  [Cited in This Article: ]
131.  Gold MS, Levine JD, Correa AM. Modulation of TTX-R INa by PKC and PKA and their role in PGE2-induced sensitization of rat sensory neurons in vitro. J Neurosci. 1998;18:10345-10355.  [PubMed]  [DOI]  [Cited in This Article: ]
132.  Aley KO, Levine JD. Role of protein kinase A in the maintenance of inflammatory pain. J Neurosci. 1999;19:2181-2186.  [PubMed]  [DOI]  [Cited in This Article: ]
133.  Aley KO, Messing RO, Mochly-Rosen D, Levine JD. Chronic hypersensitivity for inflammatory nociceptor sensitization mediated by the epsilon isozyme of protein kinase C. J Neurosci. 2000;20:4680-4685.  [PubMed]  [DOI]  [Cited in This Article: ]
134.  Parada CA, Yeh JJ, Reichling DB, Levine JD. Transient attenuation of protein kinase Cepsilon can terminate a chronic hyperalgesic state in the rat. Neuroscience. 2003;120:219-226.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 102]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
135.  Ferrari LF, Bogen O, Alessandri-Haber N, Levine E, Gear RW, Levine JD. Transient decrease in nociceptor GRK2 expression produces long-term enhancement in inflammatory pain. Neuroscience. 2012;222:392-403.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 44]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
136.  Ferrari LF, Levine E, Levine JD. Role of a novel nociceptor autocrine mechanism in chronic pain. Eur J Neurosci. 2013;37:1705-1713.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 28]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
137.  Latremoliere A, Woolf CJ. Central sensitization: a generator of pain hypersensitivity by central neural plasticity. J Pain. 2009;10:895-926.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1935]  [Cited by in F6Publishing: 2193]  [Article Influence: 146.2]  [Reference Citation Analysis (0)]
138.  Sorkin LS, Westlund KN, Sluka KA, Dougherty PM, Willis WD. Neural changes in acute arthritis in monkeys. IV. Time-course of amino acid release into the lumbar dorsal horn. Brain Res Brain Res Rev. 1992;17:39-50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 124]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
139.  Dougherty PM, Willis WD. Enhanced responses of spinothalamic tract neurons to excitatory amino acids accompany capsaicin-induced sensitization in the monkey. J Neurosci. 1992;12:883-894.  [PubMed]  [DOI]  [Cited in This Article: ]
140.  Dougherty PM, Sluka KA, Sorkin LS, Westlund KN, Willis WD. Neural changes in acute arthritis in monkeys. I. Parallel enhancement of responses of spinothalamic tract neurons to mechanical stimulation and excitatory amino acids. Brain Res Brain Res Rev. 1992;17:1-13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 128]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
141.  Schaible HG, Grubb BD, Neugebauer V, Oppmann M. The Effects of NMDA Antagonists on Neuronal Activity in Cat Spinal Cord Evoked by Acute Inflammation in the Knee Joint. Eur J Neurosci. 1991;3:981-991.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 90]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
142.  Dougherty PM, Palecek J, Paleckova V, Sorkin LS, Willis WD. The role of NMDA and non-NMDA excitatory amino acid receptors in the excitation of primate spinothalamic tract neurons by mechanical, chemical, thermal, and electrical stimuli. J Neurosci. 1992;12:3025-3041.  [PubMed]  [DOI]  [Cited in This Article: ]
143.  Coderre TJ, Melzack R. Central neural mediators of secondary hyperalgesia following heat injury in rats: neuropeptides and excitatory amino acids. Neurosci Lett. 1991;131:71-74.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 124]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
144.  Ren K, Hylden JL, Williams GM, Ruda MA, Dubner R. The effects of a non-competitive NMDA receptor antagonist, MK-801, on behavioral hyperalgesia and dorsal horn neuronal activity in rats with unilateral inflammation. Pain. 1992;50:331-344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 332]  [Cited by in F6Publishing: 346]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
145.  Dubner R, Ruda MA. Activity-dependent neuronal plasticity following tissue injury and inflammation. Trends Neurosci. 1992;15:96-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 714]  [Cited by in F6Publishing: 746]  [Article Influence: 23.3]  [Reference Citation Analysis (0)]
146.  Guo W, Zou S, Guan Y, Ikeda T, Tal M, Dubner R, Ren K. Tyrosine phosphorylation of the NR2B subunit of the NMDA receptor in the spinal cord during the development and maintenance of inflammatory hyperalgesia. J Neurosci. 2002;22:6208-6217.  [PubMed]  [DOI]  [Cited in This Article: ]
147.  Caudle RM, Perez FM, Del Valle-Pinero AY, Iadarola MJ. Spinal cord NR1 serine phosphorylation and NR2B subunit suppression following peripheral inflammation. Mol Pain. 2005;1:25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 50]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
148.  Ji RR, Gereau RW, Malcangio M, Strichartz GR. MAP kinase and pain. Brain Res Rev. 2009;60:135-148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 714]  [Cited by in F6Publishing: 781]  [Article Influence: 48.8]  [Reference Citation Analysis (0)]
149.  Karim F, Hu HJ, Adwanikar H, Kaplan D, Gereau RW. Impaired inflammatory pain and thermal hyperalgesia in mice expressing neuron-specific dominant negative mitogen activated protein kinase kinase (MEK). Mol Pain. 2006;2:2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 39]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
150.  Mannion RJ, Costigan M, Decosterd I, Amaya F, Ma QP, Holstege JC, Ji RR, Acheson A, Lindsay RM, Wilkinson GA. Neurotrophins: peripherally and centrally acting modulators of tactile stimulus-induced inflammatory pain hypersensitivity. Proc Natl Acad Sci USA. 1999;96:9385-9390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 314]  [Cited by in F6Publishing: 323]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
151.  Stubhaug A, Breivik H, Eide PK, Kreunen M, Foss A. Mapping of punctuate hyperalgesia around a surgical incision demonstrates that ketamine is a powerful suppressor of central sensitization to pain following surgery. Acta Anaesthesiol Scand. 1997;41:1124-1132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 372]  [Cited by in F6Publishing: 381]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]