Skip to main content
Current Neuropharmacology logoLink to Current Neuropharmacology
. 2008 Jun;6(2):151–163. doi: 10.2174/157015908784533888

TRPV1: A Target for Next Generation Analgesics

Louis S Premkumar 1,*, Parul Sikand 1
PMCID: PMC2647151  PMID: 19305794

Abstract

Transient Receptor Potential Vanilloid 1 (TRPV1) is a Ca2+ permeant non-selective cation channel expressed in a subpopulation of primary afferent neurons. TRPV1 is activated by physical and chemical stimuli. It is critical for the detection of nociceptive and thermal inflammatory pain as revealed by the deletion of the TRPV1 gene. TRPV1 is distributed in the peripheral and central terminals of the sensory neurons and plays a role in initiating action potentials at the nerve terminals and modulating neurotransmitter release at the first sensory synapse, respectively. Distribution of TRPV1 in the nerve terminals innervating blood vessels and in parts of the CNS that are not subjected to temperature range that is required to activate TRPV1 suggests a role beyond a noxious thermal sensor. Presently, TRPV1 is being considered as a target for analgesics through evaluation of different antagonists. Here, we will discuss the distribution and the functions of TRPV1, potential use of its agonists and antagonists as analgesics and highlight the functions that are not related to nociceptive transmission that might lead to adverse effects.

Key Words: TRPV1, nociceptors, protein kinases, inflammatory mediators, pain, nociceptive ion channels.

INTRODUCTION

Pain is manifested as nociceptive (noxious stimuli), inflammatory (tissue damage) and neuropathic (neuronal lesion) pain. Noxious stimuli are transduced by peripheral nociceptors, which transmit nociceptive information to pain processing areas in the brain via the spinal cord. Sensory nerve endings express chemo-, mechano-, and thermo-sensi-tive ion channels, which include acid sensitive ion channels (ASIC), degenerin/epithelial sodium channels (DEG/ENAC), adenosine triphosphate (ATP) gated ion channels (P2X), and transient receptor potential (TRP) channels [28, 49, 116, 170]. TRP channels (TRPVanilloid, TRPAnkyrin, TRPClassical, and TRPMelastatin) are chemo-, mechano-, and thermo-sensitive [60, 123]. These receptors are sensitized by proinflammatory agents, the receptors of which are coupled to intracellular signaling pathways and mediate heightened pain perception.

TRPV1 is a transducer of noxious temperature and chemi-cal stimuli [31]. It can initiate nociceptive signaling by generating a receptor potential at the peripheral nerve endings by increasing membrane permeability to monovalent and divalent cations including Ca2+. TRPV1 is sensitized by inflammatory mediators and is responsible, in part for inflammatory pain arising from tissue injury [65, 76, 86]. TRPV1 expression is increased in neuropathic pain resulting from nerve lesion [55, 72]. Retrograde transport of nerve growth factor (NGF) released at the site of peripheral tissue injury to the DRG soma results in activation of p38 mitogen-activated protein kinase (p38 MAPK)[76]. Enhanced translation and transport of the TRPV1 protein selectively to the peripheral terminals of sensory neurons is suggested to underlie thermal hypersensitivity following tissue injury [76]. This is generally referred to as peripheral sensitization, although increased expression of TRPV1 at the central terminals of DRG neurons could contribute to central sensitization. We have recently demonstrated that PKC-mediated phosphorylation of TRPV1, expressed on the central terminals of sensory neurons, activates the receptor at body temperature resulting in enhanced glutamatergic synaptic transmission [162]. Increased neuronal activity in primary afferents could augment the activity of second order dorsal horn neurons and third order thalamic neurons. This in turn, may be interpreted as heightened pain by cortical pain sensing areas. The finding that TRPV1 knock-out mice are less susceptible to certain modalities of pain, suggests the possibility of TRPV1 antagonists as the next generation of analgesics. The selectivity of TRPV1 as a target is bolstered by the reports that TRPV1 knock-out or ablation of TRPV1 containing neurons by neonatal administration of capsaicin or resiniferatoxin (RTX) does not exhibit other obvious abnormalities [30, 135, 175]. However, it is yet to be determined how the block of TRPV1 will impact in a preexisting disease state.

DISTRIBUTION OF TRPV1

Peripheral Nervous System

A subset of primary sensory neurons with soma in dorsal root ganglia (DRG), trigeminal ganglia (TG) and nodose ganglia expresse TRPV1 [31, 67]. These are peptidergic, small to medium diameter neurons that give rise to un-myeli-nated C-fibers and thinly myelinated Aδ-fibers. TRPV1 is also expressed in neurons that are labeled for-D-galactosyl-binding lectin IB4 and express the ionotropic ATP receptor P2X3 [67, 68]. In inflammatory conditions, capsaicin insensitive neurons can express TRPV1 [8, 9]. Tissue or nerve injury may also change the expression of neurotransmitter receptors and signaling molecules in nociceptors, which may underlie chronic pain conditions [199]. Central terminals of vanilloid-sensitive neurons form synapses on the dorsal horn of the spinal cord (DRG neurons) or the spinal nucleus of the trigeminal tract (TG neurons) [105, 106, 107] transmitting nociceptive information to the CNS.

TRPV1 expression has been established in non-neuronal cells like mast cells [24], glial cells [23], bronchial epithelial cells [194], uroepithelial cells [21, 22] and keratinocytes [47, 75, 166]. The role of TRPV1 in different regions of the body is briefly discussed below.

Central Nervous System

Expression of TRPV1 in various regions of the brain has been successfully established using an array of techniques including RT-PCR, in situ hybridization and [3H]-RTX binding [2, 31, 99, 100, 119, 147, 172, 184]. Application of capsaicin resulted in an increase in the excitatory glutamatergic synaptic transmission in the nucleus tractus solitarius (NTS) [52], locus coeruleus [111], dopaminergic neurons of the substantia nigra [110] and the preoptic hypothalamic neurons [85]. In these experiments, the facilitatory effects of capsaicin on the transmission are due to activation of presynaptic TRPV1 receptors resulting in enhanced glutamate release. However, acute capsaicin application has also been reported to produce postsynaptic effects in NTS slice preparations [52], suggesting the expression of TRPV1 in second order neurons. TRPV1 activation in cortical slices also results in glutamate release [151]. In addition, iontophoretic application of capsaicin in the periaqueductal gray produced analgesia, which was inhibited by NMDA receptor blockers [132]. Interestingly, in another study, low concentrations (10 nM) of capsaicin resulted in hyperalgesia, as indicated by increased neuronal activity in the ventral medulla and decreased latency of nocifensive behavior [115]. Furthermore, peripheral application of capsaicin enhanced bursting activity in the dorsal raphe nucleus, enhanced firing rate and bursting activity of dopaminergic neurons when applied to ventral tegmental area [109], but inhibited neuronal spike activity in the cerebellum [150]. Activation of medial thalamic neurons by capsaicin enhanced the firing frequency of these neurons, which was inhibited by morphine [11].

TRPV1 expression has been demonstrated in the basal ganglia by enhanced locomotion, vasodilation and hypothermia induced by capsaicin injection [43, 44]. The physiological functions of TRPV1 in the hippocampus are unclear [7, 93]. TRPV1 activation by its agonists capsaicin, RTX and anandamide in the CA1 region of hippocampal slices resulted in the facilitation of paired pulse depression of spike potentials [7]. The enhancement of inhibitory synaptic transmission is suggested to result from the influx of Ca2+ through TRPV1, expressed on the presynaptic terminals of GABAergic neurons. However, this could not be demonstrated in ex-vivo assays performed to study the effects of TRPV1 activation on K+-evoked Ca2+ entry and release of radiolabeled GABA from rat hippocampal nerve terminals [7]. Recently, it has been demonstrated that TRPV1 knock-out mice show reduced anxiety-related behavior [112]. These animals also exhibited less freezing following auditory fear conditioning and stress sensitization as compared to their wild type littermates [112]. In addition, the knock-out mice demonstrated deficits in developing long-term potentiation in the Schaffer collateral-commissural pathway stimulation, which may underlie reduced fear and anxiety behavior [112].

Intraperitoneal administration of capsaicin and endocannabinoid uptake inhibitor AM404 produced a drop in body temperature. Pre-administration of capsazepine but not cannabinoid receptor antagonist inhibited the hypothermic effects of both capsaicin and AM404, underscoring the importance of TRPV1 in regulating body temperature [142]. Furthermore, elevated body temperature has been reported in capsaicin-desensitized animals as compared to control or TRPV1-/- mice [177]. Capsaicin-desensitized animals have been previously reported to exhibit hyperthermia in response to systemic administration of the E. coli endotoxin, lipopolysaccharide (LPS) [176]. In contrast to these findings, Iida and colleagues [74] reported normal body temperature regulation in TRPV1-/- mice and hypothermia in response to systemic LPS administration, suggesting a role of TRPV1 in temperature regulation at a peripheral site. Recently, an interesting in vivo study evaluating the efficacy of various TRPV1 antagonists in clinical development reported that antagonism of the TRPV1 receptor resulted in hyperthermia [56]. Lipophilic TRPV1 antagonists that crossed the blood-brain barrier as well as peripherally acting antagonists induced comparable hyperthermia suggesting that TRPV1 is tonically activated and involved in regulating body temperature at sites mainly in the periphery [56]. Thus, the exact mechanisms involving TRPV1 in body temperature regulation need to be further examined and hence caution should be exercised designing and testing TRPV1 antagonists.

Cardiovascular System

The heart is richly innervated by sensory and vagal nerve endings. These nerves transduce chemical and mechanical changes from the heart to the brain. Sensory nerve endings supplying the heart express TRPV1 [202] and are able to release vasoactive peptides like CGRP and SP upon activation. The endogenous activator of TRPV1 in the heart are possibly protons, as lactic acid is produced during ischemia. Lactic acidosis and extracellular acidification are important in inducing depolarization during cardiogenic sympthoexcitatory reflex (CSR) [17, 116, 134, 153, 170]. Ablation of TRPV1-expressing afferent nerve endings by RTX administration has been reported to abolish CSR mediated cardiac pain [202]. Furthermore, reactive oxygen and nitrogen species (ROS and RNS) liberated during myocardial infarction and reperfusion could activate TRPV1 present on the sensory nerve endings [154, 155]. 5-Lipoxygenase (5-LOX) metabolites of arachidonic acid have been shown to activate cardiac sensory afferents possibly by activating TRPV1 during myocardial ischemia [169].

Vasoactive peptides modulate cardiac functions in a complex manner. Capsaicin-induced neuropeptide release may induce vasodilatation, enhance cardiac blood flow and facilitate recovery from ischemia [188]. On the contrary, it has been suggested that generation of arachidonic acid metabolite, 20-hydroxyeicosatetraenoic acid (20-HETE) results in vasoconstriction via the activation of TRPV1. Destruction of c-fiber nerve endings by capsaicin, antagonism of TRPV1 using capsazepine, ruthenium red or inhibiting substance P (SP) receptor by its antagonist have been shown to prevent myogenic constriction [156].

Respiratory System

The expression of TRPV1 in the sensory nerve endings arising from the nodose ganglia is well established [119]. Neurogenic inflammation plays an important role in the pathogenesis of diseases, such as asthma and chronic obstructive pulmonary disease. TRPV1 agonists induce bronchoconstriction and airway hyperactivity, enhance mucous secretion and induce microvascular exudation [58, 88]. These effects are mediated in part by SP and calcitonin gene related peptide (CGRP) released from the nerve terminals upon TRPV1 activation. Furthermore, sensitization of TRPV1 by inflammatory mediators like prostaglandins (PGs) [108, 125], bradykinin (BK) [35, 138, 139], nerve growth factor (NGF) [35, 76, 161] and LOX metabolites of arachidonic acid [71, 73] may lead to exacerbation of asthma. Studies have shown that the sensitivity of capsaicin-induced cough reflex/response is heightened in chronic airway inflammatory diseases [58]. TRPV1 is also shown to be involved in citric acid-induced cough [95, 186], gastroeosphageal reflux-induced asthma as well as airway disorders induced following inhalation of acid fog or pollutants [62, 65, 95]. Thus, selective TRPV1 antagonists may prove promising in treating many inflammatory airway disorders.

Urinary Bladder

TRPV1 in the urinary system is associated with perception of pain and regulating the micturition reflex. Its expression in the urinary bladder, ureter, urethra and renal pelvis of rats has been established by using [3H] RTX and immunohistochemistry [1, 13, 21, 99, 135, 174, 182]. TRPV1 has been reported to co-localize with CGRP, and SP receptors in the rat urinary bladder [13, 14, 41].

TRPV1 plays an important role in enhancing bladder reflex contractions in the chronically inflamed bladder [27, 37, 48, 98] and its responses can be significantly potentiated by the activation of PKC [157]. Application of capsazepine resulted in decreased contractions in the bladder inflamed by cyclophosphamide [48]. Furthermore, in acetic acid or lipopolysaccharide (LPS)-induced inflammation of the bladder, the frequency of urinary bladder contractions was significantly enhanced in wild type TRPV1 mice but not in TRPV1 knock-out mice. The frequency of bladder reflex contractions were reported to be similar in both TRPV1+/+ and TRPV1-/- mice and it was shown that high concentrations of capsazepine had no effect on the bladder reflex activity of normal bladders [48]. In contrast to these results, Birder and colleagues [21] reported that TRPV1-/- mice exhibited high frequencies of non-voiding contractions, increased bladder volume and enhanced the frequency of low-volume urinations, suggesting that TRPV1 is involved in normal bladder functions. Conflicting data from different groups suggest that TRPV1 plays an important yet complex role in regulating bladder function.

Intravesical administration of capsaicin and RTX, a potent TRPV1 agonist results in a marked decrease in TRPV1, SP and CGRP immunoreactivity in the rat urinary bladder [13, 14], which may be as a result of nerve terminal degeneration mediated by these agents [27, 35, 37, 40, 146, 163]. This is the underlying mechanism for the successful demonstration of TRPV1 agonists in treating interstitial cystitis [99] and urinary bladder hyperreflexia [37, 45, 46]. In this regard, therapeutic potential of RTX in treating urinary incontinence is superior to capsaicin [97, 98]. Capsaicin administration is associated with rapid depolarization of the pain transmitting c-fibers, which is perceived as a burning pain. However, RTX slowly depolarizes the c-fibers resulting in depolarization block leading to minimal pain during administration [140]. Unfortunately, the high lipophilicity of RTX poses problems with efficient packaging and loss of potency after reconstitution. RTX is in clinical trials for treating urinary bladder hyperreflexia.

Gastrointestinal Tract

TRPV1 immunoreactivity has been observed in the visceral sensory afferents and on the vagal afferents that supply the stomach mucosa, musculature and enteric plexuses [10, 136, 152, 165, 197]. Visceral sensory neurons express CGRP and SP [59, 67, 148, 197]. Capsaicin-sensitive neurons in the gut serve both afferent and efferent functions [66, 105]. Transmission of gastric pain sensation to the higher centers of the brain constitutes their afferent function, whereas release of CGRP and SP in the periphery reflects their efferent function. Neurons of nodose ganglion contain high levels of TRPV1 and 70% of the vagal afferents are sensory in nature [120].

TRPV1 activation in the foregut by capsaicin produces gastric hyperemia and enhances gastric protection by increasing mucus and HCO3- secretion [65, 69, 84, 86, 87, 180, 181]. The protease-activated receptor 2 (PAR2), a Gq-coupled receptor is suggested to sensitize TRPV1 by PKC activation in the stomach, enhancing gastric protection by increasing gastric mucus secretion. This effect is TRPV1 specific as it was inhibited by capsazepine and suggested to occur as a result of CGRP and tachykinin release upon TRPV1 activation [86]. One of the major issues with conventional COX 1 and 2 inhibiting non-steroidal anti-inflammatory agents is their ability to increase gastric acidity, leading to bleeding. However, this effect has been attributed to a lack of PG synthesis and action on their receptors (IP, EP2 and EP3 in the duodenum and EP1 in the stomach) [12, 126, 178], which causes an increase in the acid secretion. PGs do not directly induce nociception, but they are able to sensitize nociceptive ion channels to bring about pain perception. In this context, it is worth noting that the same sensitizing effect could play a role in the gastric mucosa. Application of capsaicin has been shown to increase HCO3- secretion as effectively as acid-induced HCO3- secretion [6]. This effect could be specifically blocked by ablation of capsaicin sensitive neurons, capsazepine, a TRPV1 antagonist, indomethacin (COX 1 and 2 inhibitors) and N(G)-nitro-L-arginine methylester (L-NAME), a NO synthesis inhibitor. Capsaicin increased mucosal PGI2 but not PGE2 levels, corroborating this observation that only PGI2 receptor knock-out mice abolished capsaicin induced HCO3- secretion. Capsaicin-induced HCO3- secretion is contrary to the myth that food containing the hot chili pepper can worsen gastric acidity. One would expect an acid-induced effect to overlap that of a capsaicin-induced effect, given the ability of acid to activate TRPV1 potently. Interestingly, capsaicin-induced acid secretion was blocked by capsazepine, L-NAME, indomethacin, in IP receptor knock-out mice and ablation of TRPV1 containing neurons but not by the BK 2 receptor antagonist or the EP1 receptor antagonist [6]. However, the acid induced HCO3- secretion was inhibited by indothemacin, L-NAME, EP1 antagonist and sensory deafferentation but not by capsazepine [6]. Thus, acid and capsaicin enhance HCO3- secretion through common players but likely through different receptors.

TRPV1 mediated CGRP release could play a role in increasing blood supply to malignancies of the GI tract; the beneficial effect of COX inhibitors in this regard may arise from their ability to reduce TRPV1-mediated CGRP release and inhibition of PG-mediated angiogenesis. TRPV1 is associated with inducing inflammation and tissue damage in the pancreas, ileum and colon following dextran sulphate sodium-induced colitis in rats [90, 91], caerulein-induced pancreatitis [129], Clostridium difficile toxin A- and endocannabinoid-induced colitis [117, 118]. These effects of TRPV1 are associated with release of SP from the sensory afferents that activates the neurokinin receptor resulting in an activation of immune cells, mast cells and enteric cells causing hypersecretion, inflammation and finally mucosal damage [65, 117]. Patients diagnosed with active inflammatory bowel disease demonstrate a greatly increased TRPV1 immunoreactivity in colonic nerve fibers [197, 201], suggesting an important implication of the TRPV1 antagonists in treating GI distress. Recently, it has been reported that TRPV1-expressing pancreatic sensory neurons control islet inflammation and insulin resistance in type I diabetes [143]. Ablation of these TRPV1 positive sensory afferents by capsaicin application prevents the onset of diabetes and associated pancreatitis in diabetes prone non obese diabetic mice [143].

Taste and Food Intake

A TRPV1 receptor variant is attributed to be the mammalian amiloride-insensitive non-specific salt taste receptor [103]. This receptor is expressed in fungiform taste receptor cells and is a non-selective cation channel, activated by vanilloids, temperatures >38˚C, inhibited by capsazepine and is absent in TRPV1 knock-out mice [103]. Thus, developing specific salt taste suppressors or enhancers may prove beneficial in managing cardiovascular complications, such as hypertension. The activation of TRPV1 in the nodose ganglia by the endogenous feeding and body weight regulating lipid, oleoylethanolamide [3], resulted in visceral pain and reduced food intake in only wild type but not in TRPV1 knock-out mice [196]. Craving for spicy food may be as a result of capsaicin effect on the satiety centers [3]. It is interesting to note that the TRP channels that are purported to be involved in pain are also expressed in gustatory neurons and carry specific taste sensations. For example, TRPV1 carries the pungent taste associated with capsaicin and vinegar, whereas TRPA1 carries the taste associated with mustard, horse radish and wasabi.

Hearing

Capsaicin-mediated activation of the sensory nerves supplying the cochlea has been reported to increase cochlear blood flow [191,192,203]. This effect of capsaicin is attributed to the release of SP from the axons in the basilar artery, the anterior inferior cerebellar artery, the spiral modiolar artery, and the radiating arterioles of the cochlea [192]. TRPV1 immunoreactivity has been reported in the hair cells, organ of corti, vestibular and spiral ganglion cells of the cochlea [15, 203]. 5-LOX metabolites of AA activate TRPV1, TRPV1 and 5-LOX immunoreactivity has been observed in the neurons and satellite cells in adjacent sections of these ganglia [15]. TRPV1 activation in the cochlea is associated an increase in the threshold of auditory nerve compound action potentials and a reduced magnitude of cochlear microphonic and electrically-evoked otoacoustic emissions, suggesting an important modulatory role in the physiology of the inner ear [203]. In a recent study by Kitahara and colleagues [92], subcutaneous kanamycin administration in mice resulted in increased mRNA levels of TRPV1 as determined by the real-time PCR technique in the spiral and vestibular ganglia. The authors concluded that upregulated TRPV1 in these ganglia may promote neuronal survival and underlie kanamycin-induced dizziness and tinnitus as side effects. Furthermore, TRPV1 may also be implicated in hearing loss associated with inflammation in salicylates-induced tinnitus or in pathological conditions of the inner ear such as in Meniere’s disease or in migraines. Tinnitus can be equated to chronic pain conditions; both involve spontaneous neuronal firing and likely to be mediated via TRPV1.

Hair

Expression of TRPV1 on the hair follicle (mainly in the outer root sheath and hair matrix) has been established by using diaminobenzidine immunoreactivity [25]. Using cultured human scalp hair follicles, capsaicin application was shown to inhibit hair shaft elongation and induce apoptosis, suppress proliferation and enhance premature hair follicle regression. Up-regulation of endogenous hair growth inhibitors and down-regulation of endogenous hair growth promoters were also observed in cultured outer root sheath keratinocytes upon TRPV1 activation [25]. Thus, the TRPV1 receptor is an important regulator of human hair growth and its expression in hair follicles and keratinocytes suggest a role beyond nociception.

Functions of TRPV1

Noxious stimuli are transduced by peripheral nociceptors, which transmit information regarding tissue damage to pain-processing centers in the brain via the spinal cord [16]. TRPV1 in sensory nerve endings transduce various stimuli, in particular, inflammatory thermal pain as indicated by deletion of the TRPV1 gene [30, 42]. TRPV1 is involved in both afferent (sensation of pain) and efferent (neurotransmitter and neuropeptide release) functions, which are experienced with a burning sensation followed by vasodilation and sweating upon consumption of capsaicin in spicy foods. Thus, TRPV1 can mediate both inflammation and pain and this dual mode of action qualifies it to be an important player in various physiological and pathophysiological processes.

Important properties of the TRPV1 receptor include sensitization, desensitization and tachyphylaxis. Capsaicin, and certain noxious stimuli can induce a prolonged hypersensitive state and capsaicin-induced hyperalgesia is a well-characterized pain model. Following intradermal capsaicin injection in humans, pronounced hyperalgesia to heat and mechanical stimuli is experienced at the injection site, which later spreads to the surrounding tissue [172]. Desensitization is defined as a reduction in response to continued exposure to an agonist. Tachyphylaxis is defined as a reduction in response to repeated application of an agonist. Both desensitization and tachyphylaxis require extracellular Ca2+ [31], suggesting the involvement of Ca2+-dependent intracellular signaling mechanisms. Tachyphylaxis can be abolished by inhibitors of Ca2+-dependent phosphatases, suggesting the requirement for phosphorylation in channel activation [50]. Thus, Ca2+-mediated dephosphorylation may render the channel inactive leading to desensitization or tachyphylaxis. Prolonged TRPV1 activation by capsaicin may lead to its desensitization and this may account for the paradoxical use of capsaicin as an analgesic agent [171]. Interestingly, PKA-, but not PKC-mediated phosphorylation is able to reverse tachyphylaxis, suggesting distinct actions for these kinases [120, 121, 122]. Desensitization may result from Ca2+-induced modulation of TRPV1 sensitivity, and/or Ca2+ toxicity; a large and sustained Ca2+ influx via TRPV1 has been linked to neurodegeneration of the peripheral nerve terminals.

ACTIVATION CHARACTERISTICS OF TRPV1

TRPV1 is a well-characterized channel, which transduces heat in the noxious temperature range (>42oC) and is critical for nociceptive and inflammatory thermal sensation [80]. It is a Ca2+ permeant polymodal receptor activated by protons, anandamide, lipoxygenase metabolites of arachidonic acid (AA), N-arachidonyl dopamine (NADA), capsaicin (an active ingredient in hot chili peppers) and RTX, an ultrapotent agonist obtained from the cactus, Euphorbia resinifera [29]. RTX combines structural features of phorbol esters (potent activators of PKC) and vanilloid compounds. It was thought that its ability to activate PKC might be responsible for its high potency, but the concentration required to activate PKC is much higher to account for this effect [63].

Cloning and mutational analysis of TRPV1 has improved our understanding of how these receptors are activated. The TRPV1 ion channel subunit has six transmembrane-spanning segments flanked by large intracellular, N- and C-terminal domains [31, 81, 94]. Therefore, an intracellular modulator or a readily membrane-permeable agent is required to activate/block the receptor. Binding of capsaicin and RTX to TRPV1 involves amino acid residues which have been shown to reside in N- and C-cytosolic and transmembrane domains of the channel [34, 57, 81, 82]. The binding site for vanilloid compounds has been reported to lie between transmembrane domains 2 and 4. Amino acids Y511 and S512 may play a key role in capsaicin binding [78]. Other studies have implicated the importance of residues on the cytoplasmic tail in capsaicin and proton binding [78]. Amino acids in transmembrane domain 6 (N676, M677 and L678) play a significant role in capsaicin- and proton-induced activation of TRPV1 [79]. Mutation of residues at D601, E610 or E648 to neutral or basic residues resulted in a loss of proton sensitivity. In contrast, mutation of E600 resulted in increased sensitivity to vanilloids at higher temperatures [79].

A potent endogenous activator of TRPV1 has not been identified. Anandamide, NADA and lipoxygenase metabolites of AA have been shown to activate the receptor weakly. However, in the phosphorylated state, agonist and temperature sensitivity is greatly increased, in that TRPV1 could be active at normal body temperature [130, 162, 168]. Therefore, the activation mechanism could include multiple stimuli integrating synergistically to induce a maximal response.

SENSITIZATION OF TRPV1 BY PHOSPHORYLATION

Activation of Ca2+ permeant nociceptive ion channels on the peripheral and central terminals of sensory neurons leads to the synthesis and/or release of a variety of proinflammatory agents and neuropeptides, like ATP, BK, PGs, CGRP, SP and vasoactive intestinal peptide (VIP) [105, 114]. Increases in intracellular Ca2+ initiate several second messenger pathways including the activation of phospholipase A2 (PLA2), phospholipase C (PLC) and Ca2+-dependent kinases, which can lead to the generation of AA and its metabolites, release of Ca2+ from intracellular stores and phosphorylation of nociceptive ion channels, respectively.

TRPV1 is important in inflammatory thermal hyperalgesia. Recent studies have suggested that various inflammatory mediators are able to sensitize TRPV1 to chemical and physical stimuli. Inflammation results in acidic conditions, which can activate TRPV1 directly. The inflammatory mediators activate their respective G-protein coupled receptors to initiate secondary messenger pathways resulting in the activation of either PKA [20, 44, 70, 121, 131, 141], PKC [19, 32, 39, 130, 138, 172, 183], MAPK [76, 204], extracellular Ca2+/CaM-dependent kinase II (CaMKII) [83, 149] or Src kinase [77], which phosphorylate TRPV1.

Cyclic AMP-dependent protein kinase A (PKA) in primary afferents is important in mediating inflammatory hyperalgesia [144, 178, 179]. Activation of PKA by either using cAMP analogues or PGs results in hyperalgesia [178, 179]. PKA-mediated phosphorylation of TRPV1 potentiates capsaicin-mediated current and increase CGRP release in cultured DRG neurons [64, 70, 102]. Anandamide causes release of neuropeptides by activating both cannabinoid 1 (CB1) and TRPV1 receptors [5]. Activation of CB1 or CB2 receptors has been shown to increase or decrease adenylate cyclase levels, which will modulate the phosphorylation state of TRPV1. Activation of the CB1 receptors decreases Ca2+ and increases K+ conductance in the presynaptic terminals [33] that can interfere with the action of TRPV1 distributed at the central terminals of sensory neurons. Phosphorylation at S116 in the amino terminus of TRPV1 is vital in PKA-mediated regulation of TRPV1 desensitization [20]. Mutations of S116, T370, T144, and S502 are capable of reversing forskolin-induced potentiation of TRPV1 responses [20, 121, 122, 141].

Phosphorylation by PKC has been shown to sensitize TRPV1 [32, 138]. Various algesic agents like BK, ATP, trypsin and PGs are known to sensitize TRPV1 by activating PKC downstream of their G-protein coupled receptors in sensory neurons and in expressed systems. PKC-dependent phosphorylation of TRPV1 not only potentiates capsaicin- or proton-evoked responses but also reduces its temperature threshold for activation [39, 126, 168, 183]. Serine residues (S502 and S800) on TRPV1 have been identified by biochemical studies to be important in PKC-mediated effects [19, 130].

Phorbol esters can sensitize TRPV1 by phosphorylating the channel [32, 138, 193]. They have also been reported to activate the channel directly as suggested by the induction of capsazepine-sensitive inward currents in HEK cells transiently transfected with hTRPV1 [193]. PKC-mediated phos-phorylation of TRPV1 can reduce its activation threshold by partially activating the receptor such that lower concentrations of the agonist can fully activate the receptor [4, 138, 193]. In addition, PKC alters the time- and voltage-dependent properties of TRPV1. PKC-mediated phosphorylation of TRPV1 in DRG neuronal cultures has been shown to produce faster activation and slower deactivation of the channel [4]. Phosphorylation not only sensitizes the receptor and augments its response, but also promotes translocation of TRPV1 from the cytosol to the plasma membrane [124, 167, 190].

PKCε, and PKCα, calcium-independent and calcium-dependent isoforms of PKC, respectively, have been shown to phosphorylate TRPV1 [29, 32, 138, 193]. PKC plays a pivotal role in pain signaling, and the PKCε gene disruption in mice dramatically reduces thermal and inflammatory pain [89]. In mice lacking PKCγ, acute pain was preserved, but the neuropathic pain was reduced [107].

Ca2+ calmodulin-dependent protein kinase II (CaMKII) has been shown to modulate TRPV1 function and capsaicin binding [83, 149]. In contrast, calcineurin-mediated dephosphorylation causes TRPV1 desensitization [50]. Mutants of CaMKII phosphorylation sites on TRPV1 (S502 and T704) failed to elicit currents in response to application of capsaicin or RTX [83]. Thus, CaMKII and calcineurin control the subtle balance between TRPV1 phosphorylation/dephosphory-lation and its activation, deactivation and desensitization characteristics.

TRPV1 is activated by membrane-derived lipids like anandamide, oleoylethanolamide and some lipoxygenase products of arachidonic acid [3, 73, 205]. Phosphainositol 3, 4 bisphosphate (PIP2) has been suggested to tonically inhibit TRPV1 by its constitutive association with the channel. Activation of PLC by metabotropic receptors (BK2, TrkA and histamine) hydrolyzes PIP2 into diacylglycerol and inositol (1, 4, 5) trisphosphate, which relieves its tonic inhibition of the channel [35]. PIP2 regulates TRPV1 activity by interacting with amino acids between 777–820, which contains many positively charged residues [139]. This region also includes S800, the PKC phosphorylation site and overlaps with the 35 amino acid segment necessary for CaM binding. This region might thus be very important for the activation of TRPV1. BK and histamine are coupled to PLA2, activation of which, in turn, can produce AA and its metabolites leading to the modulation of TRPV1 [160].

TRPV1 AGONISTS

The possibility of using TRPV1 receptor agonists to alleviate pain is an interesting concept. Activation of TRPV1 can induce persistent depolarization of the nerve terminals causing a decrease in their ability to generate and propagate action potentials. On the other hand, TRPV1 activation causes an influx of Ca2+ that in the long-term can cause nerve terminal degeneration. Ablation of TRPV1 containing neurons by intraperitoneal injection of capsaicin or RTX has been demonstrated [127, 133, 200]. Furthermore, high concentration of capsaicin has been used to alleviate pain associated with herpes zoster, AIDS neuropathy, diabetic peripheral neuropathy, etc [80, 172]. In this strategy to avoid the pain caused by capsaicin, a prior application of lidocaine is warranted. Local application of 0.075% capsaicin ointment has been reported to have moderate to poor efficacy following eight weeks of application in neuropathic or musculoskeletal pain [113]. In itch-causing pruritis, capsaicin 0.006% has been found to be useful [104].

Intrathecal administration of capsaicin has been shown to cause a long-lasting loss of thermal sensitivity without affecting mechanical sensitivity [200]. RTX is a potent agonist of TRPV1, which exhibits unique properties that can be exploited to treat chronic pain conditions [140]. The characteristics of RTX include the following: 1. irreversible nature of activation enables the use of low concentrations (well below the toxic levels); 2. slow and sustained depolarization results in a gradual inactivation of voltage-gated Na+ channels causing a depolarization block, preventing action potential generation in the short-term and Ca2+-induced nerve terminal death in the long-term [140]. Since the nerve terminals have the ability to regenerate, long-term toxicity may not be a major concern. Intrathecal administration of RTX causes a loss of thermal sensitivity [175]. Idalora and colleagues [27] have used intrathecal administration of RTX to alleviate pain associated with osteosarcoma in dogs. We have found that selective ablation of central terminals of DRG neurons by intrathecal administration of RTX can achieve reduction of nociceptive neurotransmission. RTX in very low (nanomolar) concentrations can activate TRPV1 slowly, maximally and irreversibly leading to depolarization beyond the threshold without generating action potentials. RTX-induced sustained depolarization leads to the loss of generation of action potentials in the peripheral and/or central TRPV1 containing nociceptor nerve terminals in the short-term and ablation of these nerve terminals in the long-term. This has the potential to alleviate chronic pain conditions (S.Y. and L.S.P., unpublished results). The prevention of permanent loss of the DRG is therapeutically favorable and offers a unique approach to treat chronic pain conditions. The selectivity of RTX action on TRPV1-containing nociceptive nerve terminals may offer an advantage of a higher therapeutic index compared to other clinically available agents (toxins), such as, botulinum toxin and ω-conopeptide, which act on synaptic vesicles and on Ca2+ channels, respectively.

TRPV1 ANTAGONISTS

For decades the only antagonist available to inhibit capsaicin-mediated responses was the organo-metallic dye ruthenium red [53]. Ruthenium red is a non-competitive antagonist of TRPV1; however, based on recent studies it can be classified as a general inhibitor of TRPV channels [60, 164]. The mechanism of action is proposed to be by blocking the channel pore. Moreover, it has also been demonstrated to block other channels, in particular Ca2+ channels [53]. The well-studied TRPV1 antagonist is the synthetic vanilloid analogue, capsazepine [18, 187]. Capsazepine is a competitive antagonist of TRPV1, which is able to block both in vivo and in vitro effects of capsaicin and exhibits a relatively high selectivity for TRPV1 as compared to other TRPV channels. However, capsazepine is far from being an ideal TRPV1 antagonist as it has many disadvantages. First, it is a very weak antagonist with IC50 values of 0.2-4 μM [2, 18, 31, 101, 188, 199]. Second, it demonstrates species-dependent difference in inhibiting TRPV1. Capsazepine inhibits acid-induced activation of human TRPV1 (hTRPV1), but has no inhibitory effect on acid-induced activation of rat TRPV1 (rTRPV1) [190]; however, it can inhibit heat-mediated responses in both hTRPV1 and rTRPV1. Third, at concentrations needed to inhibit TRPV1 (~10 μM), it has been demonstrated to block amongst many receptors and channels, voltage-gated calcium channels [51] and acetylcholine receptors [101]. Furthermore, the sarcoplasmic reticulum ATPase (SERCA) inhibitor, thapsigargin, was shown to inhibit the capsaicin-induced calcium influx with an IC50 of 6.4 ± 1.9 μM as well as prevented [3H] RTX binding (IC50 = 4 ± 1.3 μM) in CHO cells expressing TRPV1 [186].

Iodination of RTX at the 5΄ position of the vanilloid moiety resulted in one of the most potent TRPV1 antagonist iodo-resiniferatoxin (iodo-RTX) [196]. Iodo-RTX has been reported to inhibit capsaicin-induced currents in Xenopus laevis oocytes expressing TRPV1 with an IC50 value of 3.9 nM. Intrathecal administration inhibited capsaicin-elicited pain responses with ED50 of 16 ng/mouse [196]. Furthermore, iodo-RTX has been demonstrated to inhibit both heat- and proton-evoked responses in sensory neurons as well as cells expressing the recombinant TRPV1 [145]. However, in an interesting study by Shimizu and colleagues [159], systemic administration of iodo-RTX in mice resulted in hypothermia, as these effects were not seen in either TRPV1 knock-out mice or RTX-pretreated animals. Iodo-RTX was also shown to enhance intracellular calcium levels and induced a small inward current in HEK293 cells stably transfected with TRPV1. A possible suggested explanation for the partial agonistic action of iodo-RTX is that it is metabolized (deiodination) to RTX in vivo. The controversy over the antagonistic vs. partial agonistic action of iodo-RTX has resulted in a loss of enthusiasm over this compound.

Since TRPV1 has become an undisputed target for a variety of painful conditions, a quest for synthesizing potent yet selective TRPV1 antagonists is unabated. A non-vanilloid TRPV1-antagonist N-(4-tertiarybutylphenyl)-4-(3-chloropyridin-2-yl)tetrahydropyrazine-1(2H)-carbox-amide (BCTC) has been synthesized. It inhibits capsaicin- and acid-induced responses in rat TRPV1 with an IC50 value in the range of 6-35 nM; unlike capsazepine, however, BCTC does block acid-mediated activation of rat TRPV1 [190]. Furthermore, orally administered BCTC (3-30 mg/kg) has also been demonstrated to inhibit both mechanical and thermal hyperalgesia associated with complete Freund’s adjuvant-induced inflammation and nerve injury [137] without affecting normal/acute nociception. BCTC is lipophilic and crosses the blood-brain barrier and may carry the risk of inducing central nervous system side effects. However, in its analgesic concentrations, BCTC has not been documented to produce any locomotor impairment usually associated with centrally acting analgesics.

The Glaxo Smith Kline compound SB-366791 is a competitive inhibitor of TRPV1 that inhibits capsaicin, acid and noxious heat-mediated activation of the receptor. It is superior to capsazepine as it blocks acid-mediated activation of rTRPV1. It has demonstrated a high selectivity in 47 different binding and electrophysiological assays [61]. Furthermore, SB-366791 does not inhibit VGCC or hyperpolarization-activated current (Ih) in cultured rodent sensory neurons. In a recent study, spontaneous and miniature excitatory synaptic currents (sEPSC and mEPSC), recorded from the substantia gelatinosa neurons of the spinal cords obtained from CFA-injected animals, were decreased in the presence of SB-366791 [96].

Abbott laboratories have in the recent years synthesized a number of promising TRPV1 antagonists. The A-425619 [1-Isoquinolin-5-yl-3-(4-trifluoromethyl-benzyl)-urea] is a com-petitive, potent (IC50= 5-9 nM) and a highly selective antagonist for the capsaicin receptor [173]. It inhibits increases in intracellular calcium influx evoked by capsaicin, NADA, anandamide or pH in HEK cells transfected with hTRPV1 as well as capsaicin-induced inward currents in cultured rat DRG neurons. Furthermore, in experiments, where TRPV1 receptors expressed in HEK cells were sensitized by a PKC activator, PDBu or low pH 6.0, capsaicin-mediated intracellular influx was inhibited by A-425619 at much lower concentrations (pA2 = 0.8 ± 0.3 and 4.3 ± 1.9 nM, respectively). Recently, a detailed study highlights the properties of two of their leading contenders for antagonizing TRPV1 receptor that are orally active. A-784168 (1-[3-(trifluoromethyl)pyri-dine-2-yl]-N-[4-(trifluoromethylsulfonyl)phenyl]-1,2,3,6-tet-rahydropyridine-4-carboxamide) and A-795614 (N-1H-indazol-4-yl-N’- [(1R)-5-piperidin-1-yl-2,3-dihydro-1H-inden-1-yl]urea) have comparable efficacy in in-vitro assays (IC50= 25 ± 2 and 14 ± 1 nM, respectively, in inhibiting capsaicin-mediated responses), but have significant differences in their ability to permeate the CNS [38]. Interestingly, in models representing chronic pain conditions, including CFA- and capsaicin-induced mechanical allodynia and osteoarthritis, both agents demonstrated equal efficacy when administered intrathecally; however, when administered orally, A-784168, which crosses the blood-brain barrier, was found to be superior than A-795614 [38]. This study clearly demonstrates that drugs which cross the blood-brain barrier are more efficient in antagonizing TRPV1 in vivo. However, it could also be argued that such highly lipophilic compounds may be associated with a greater CNS side effect profile, because of TRPV1 distribution in the brain.

Amgen has developed a piperazinylpyrimidine analog (AMG517) as a potent TRPV1 antagonist, which is in the phase 1 clinical trials. Amgen has also developed other potent compounds (AMG0347, AMG8163, AMG9810), IC50 values to antagonize capsaicin responses range between 0.5 to 86 nM [25]. Neurogen/Merck has launched NDG-8243/MK-2295 in Phase II clinical trials, the structure of which is unpublished/unreported. The latest compound to enter clinical trials is GRC-6211 from Glenmark pharmaceuticals. Other TRPV1 antagonists include JNJ-17203212 (Johnson and Johnson) has been shown to be effective in bone cancer pain and JYL1421 (Schwarz Pharma), which blocks capsaicin induced responses with an IC50 of 9.2 µM [173].

The NGF antibody is being evaluated for the treatment of chronic pain. NGF has been shown to sensitize TRPV1 by activating PLC and hydrolyzes PIP2, which tonically inhibits the channel [35, 139], or by activating PKC, PI3K [26, 35, 161, 167] and ERK [204]. Although the precise mechanism of action is debatable, it is known that NGF sensitizes TRPV1 and increases its expression and function. Therefore, blocking NGF action could down regulate TRPV1 expression and function.

CONCERNS OF TRPV1 ANTAGONISTS

TRPV1 antagonists hold a great promise in alleviating multiple modalities of pain. However, it must be borne in mind that other physiological functions that are mediated by TRPV1 channels may be affected. Of concern are the maintenance of body temperature, TRPV1-mediated SP and CGRP release and their subsequent effect on the cardiovascular functions and acute nociception. Particular efforts should be focused on changes in body temperature. In some studies, the body temperature has been shown to rise as much as 3oC. Cardiovascular consequences range from the inability to feel pain during myocardial ischemia to decreased TRPV1-mediated CGRP release that can compromise coronary circulation. CGRP is also critical for the integrity of microvascular circulation and lack of which could aggravate diabetic peripheral neuropathy and peripheral vascular disease. Gastrointestinal consequences could occur as a result of a decreased release of neuroactive substances, as well as by modulation of vagal nerve circuitry. The preferred route of oral administration of TRPV1 antagonists could have an immediate impact on the gastrointestinal functions. The main disadvantage with the existing nonselective COX inhibitors is that they increase gastric acid section that may lead to life threatening bleeding due to decreased PG synthesis. However, studies have suggested that capsaicin is capable of releasing HCO3- as efficiently as acid challenge. Therefore, blocking TRPV1 may decrease the release of HCO3-, which is essential to neutralize the acid. Recent studies show that the TRPV1 knock-out animals have deficiencies in fear conditioning and memory formation, which are serious issues to consider. RTX is in clinical trials for the treatment of urinary bladder hyperreflexia. Therefore, it should be evaluated how the block of TRPV1 could affect normal bladder functions. Furthermore, blocking TRPV1 has been shown to increase its own expression, thereby raising the possibility of rebound effects. Given the observations that TRPV1 knock-out mice have normal life span without serious abnormalities, suggest that the block of TRPV1 may not induce serious adverse effects. However, it has to be investigated how these animals react upon insult or in pathological conditions.

CONCLUDING REMARKS

Although, TRPV1 is considered to be a potential target for developing drugs to treat different modalities of pain, extensive basic science research and clinical trials have to be undertaken to uncover possible adverse effects. This task is further complicated by the advent of TRPV1 distribution in novel areas mediating unexpected functions. In particular, antagonizing the receptor may lead to cardiovascular complications as a result of decreased vasoactive peptide release. Sustained stimulation can also lead to Ca2+-dependent nerve terminal degeneration. Therefore, the consequence could be as a result of both agonistic and antagonistic actions. Finally, there is no drug that has been invented without adverse effects; therefore, the balance between the beneficial and the adverse effects should be cautiously and pragmatically considered. Using agents to selectively target nerve terminals at the periphery or at the level of the spinal cord may avoid some of the systemic adverse effects. In any case, TRPV1 is a novel target for next generation analgesics, and if proven to be successful in clinical trials, its agonists and antagonists may be added to the therapeutic armamentarium.

ACKNOWLEDGEMENT

This work was supported by a grant from National Institutes of Health (DK065742).

REFERENCES

  • 1.Acs G, Palkovits M, Blumberg PM. Comparison of [3H]resiniferatoxin binding by the vanilloid (capsaicin) receptor in dorsal root ganglia spinal cord dorsal vagal complex, sciatic and vagal nerve and urinary bladder of the rat. Life Sci. 1994;55:1017–1026. doi: 10.1016/0024-3205(94)00636-9. [DOI] [PubMed] [Google Scholar]
  • 2.Acs G, Palkovits M, Blumberg PM. Specific binding of [3H]resiniferatoxin by human and rat preoptic area, locus ceruleus, medial hypothalamus, reticular formation and ventral thalamus membrane preparations. Life Sci. 1996;59:1899–1908. doi: 10.1016/s0024-3205(96)00537-1. [DOI] [PubMed] [Google Scholar]
  • 3.Ahern GP. Activation of TRPV1 by the satiety factor oleoylethanolamide. J. Biol. Chem. 2003;278:30429–30434. doi: 10.1074/jbc.M305051200. [DOI] [PubMed] [Google Scholar]
  • 4.Ahern GP, Premkumar LS. Voltage-dependent priming of rat vanilloid receptor: effects of agonist and protein kinase C activation. J. Physiol. 2002;545:441–451. doi: 10.1113/jphysiol.2002.029561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Ahluwalia J, Urban L, Bevan S, Nagy I. Anandamide regulates neuropeptide release from capsaicin-sensitive primary sensory neurons by activating both the cannabinoid 1 receptor and the vanilloid receptor 1 in vitro. Eur. J. Neurosci. 2003;17:2611–2618. doi: 10.1046/j.1460-9568.2003.02703.x. [DOI] [PubMed] [Google Scholar]
  • 6.Aihara E, Hayashi M, Sasaki Y, Kobata A, Takeuchi K. Mechanisms underlying capsaicin-stimulated secretion in the stomach: comparison with mucosal acidification. J. Pharmacol. Exp. Ther. 2005;315:423–432. doi: 10.1124/jpet.105.087619. [DOI] [PubMed] [Google Scholar]
  • 7.Al-Hayani A, Wease KN, Ross RA, Pertwee RG, Davies SN. The endogenous cannabinoid anandamide activates vanilloid receptors in the rat hippocampal slice. Neuropharmacology. 2001;41:1000–1005. doi: 10.1016/s0028-3908(01)00145-9. [DOI] [PubMed] [Google Scholar]
  • 8.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. doi: 10.1016/s0006-8993(02)03972-0. [DOI] [PubMed] [Google Scholar]
  • 9.Amaya F, Shimosato G, Nagano M, Ueda M, Hashimoto S, Tanaka Y, Suzuki H, Tanaka M. NGF and GDNF differentially regulate TRPV1 expression that contributes to development of inflammatory thermal hyperalgesia. Eur. J. Neurosci. 2004;20:2303–2310. doi: 10.1111/j.1460-9568.2004.03701.x. [DOI] [PubMed] [Google Scholar]
  • 10.Anavi-Goffer S, McKay NG, Ashford ML, Coutts AA. Vanilloid receptor type 1-immunoreactivity is expressed by intrinsic afferent neurones in the guinea-pig myenteric plexus. Neurosci. Lett. 2002;319:53–57. doi: 10.1016/s0304-3940(01)02480-6. [DOI] [PubMed] [Google Scholar]
  • 11.Ando R, Onodera K, Shima K, Kisara K. Effects of capsaicin on spontaneous unit discharges in medial thalamic single neurons of cats. Nippon Yakurigaku Zasshi. 1977;73:955–959. doi: 10.1254/fpj.73.955. [DOI] [PubMed] [Google Scholar]
  • 12.Aoi M, Aihara E, Nakashima M, Takeuchi K. Participation of prostaglandin E receptor EP4 subtype in duodenal bicarbonate secretion in rats. Am. J. Physiol. Gastrointest. Liver Physiol. 2004;287:96–103. doi: 10.1152/ajpgi.00038.2004. [DOI] [PubMed] [Google Scholar]
  • 13.Avelino A, Cruz C, Nagy I, Cruz F. Vanilloid receptor 1 expression in the rat urinary tract. Neuroscience. 2002;109:787–798. doi: 10.1016/s0306-4522(01)00496-1. [DOI] [PubMed] [Google Scholar]
  • 14.Avelino A, Cruz F. Peptide immunoreactivity and ultrastructure of rat urinary bladder nerve fibers after topical desensitization by capsaicin or resiniferatoxin. Auton. Neurosci. 2000;86:37–46. doi: 10.1016/S1566-0702(00)00204-6. [DOI] [PubMed] [Google Scholar]
  • 15.Balaban CD, Zhou J, Li HS. Type 1 vanilloid receptor expression by mammalian inner ear ganglion cells. Hear.Res. 2003;175:165–170. doi: 10.1016/s0378-5955(02)00734-7. [DOI] [PubMed] [Google Scholar]
  • 16.Basbaum AI, Fields HL. Endogenous pain control systems: brainstem spinal pathways and endorphin circuitry. Annu. Rev. Neurosci. 1984;7:309–338. doi: 10.1146/annurev.ne.07.030184.001521. [DOI] [PubMed] [Google Scholar]
  • 17.Benson CJ, Sutherland SP. Toward an understanding of the molecules that sense myocardial ischemia. Ann. NY. Acad. Sci. 2001;940:96–109. doi: 10.1111/j.1749-6632.2001.tb03669.x. [DOI] [PubMed] [Google Scholar]
  • 18.Bevan S, Hothi S, Hughes G, James IF, Rang HP, Shah K, Walpole CS, Yeats JC. Capsazepine a competitive antagonist of the sensory neurone excitant capsaicin. Br. J. Pharmacol. 1992;107:544–552. doi: 10.1111/j.1476-5381.1992.tb12781.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Bhave G, Hu HJ, Glauner KS, Zhu W, Wang H, Brasier DJ, Oxford GS, Gereau RW. Protein kinase C phosphorylation sensitizes but does not activate the capsaicin receptor transient receptor potential vanilloid 1 (TRPV1) Proc. Natl. Acad. Sci. USA. 2003;100:12480–12485. doi: 10.1073/pnas.2032100100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Bhave G, Zhu W, Wang H, Brasier DJ, Oxford GS, Gereau RW. cAMP-dependent protein kinase regulates desensitization of the capsaicin receptor (VR1) by direct phosphorylation. Neuron. 2002;35:721–731. doi: 10.1016/s0896-6273(02)00802-4. [DOI] [PubMed] [Google Scholar]
  • 21.Birder LA, Kanai AJ, de Groat WC, Kiss S, Nealen ML, Burke NE, Dineley KE, Watkins S, Reynolds IJ, Caterina MJ. Vanilloid receptor expression suggests a sensory role for urinary bladder epithelial cells. Proc. Natl. Acad. Sci. USA. 2001;98:13396–13401. doi: 10.1073/pnas.231243698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Birder LA, Nakamura Y, Kiss S, Nealen ML, Barrick S, Kanai AJ, Wang E, Ruiz G, de Groat WC, Apodaca G, Watkins S, Caterina MJ. Altered urinary bladder function in mice lacking the vanilloid receptor TRPV1. Nat. Neurosci. 2002;5:856–860. doi: 10.1038/nn902. [DOI] [PubMed] [Google Scholar]
  • 23.Biro T, Brodie C, Modarres S, Lewin NE, Acs P, Blumberg PM. Specific vanilloid responses in C6 rat glioma cells. Brain Res. Mol. Brain Res. 998a;56:89–98. doi: 10.1016/s0169-328x(98)00033-3. [DOI] [PubMed] [Google Scholar]
  • 24.Biro T, Maurer M, Modarres S, Lewin NE, Brodie C, Acs G, Acs P, Paus R, Blumberg PM. Characterization of functional vanilloid receptors expressed by mast cells. Blood. 998b;91:1332–1340. [PubMed] [Google Scholar]
  • 25.Bodo E, Biro T, Telek A, Czifra G, Griger Z, Toth BI, Mescalchin A, Ito T, Bettermann A, Kovac L, Paus R. A hot new twist to hair biology: involvement of vanilloid receptor-1 (VR1/TRPV1) signaling in human hair growth control. Am. J. Pathol. 2005;166:985–998. doi: 10.1016/S0002-9440(10)62320-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Bonnington JK, McNaughton PA. Signalling pathways involved in the sensitisation of mouse nociceptive neurones by nerve growth factor. J. Physiol. 2003;551:433–446. doi: 10.1113/jphysiol.2003.039990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Brown DC, Iadarola MJ, Perkowski SZ, Erin H, Shofer F, Laszlo KJ, Olah Z, Mannes AJ. Physiologic and antinociceptive effects of intrathecal resiniferatoxin in a canine bone cancer model. Anesthesiology. 2005;103:1052–1059. doi: 10.1097/00000542-200511000-00020. [DOI] [PubMed] [Google Scholar]
  • 28.Burnstock G. P2X receptors in sensory neurones. Br. J. Anaesth. 2000;84:476–488. doi: 10.1093/oxfordjournals.bja.a013473. [DOI] [PubMed] [Google Scholar]
  • 29.Caterina MJ, Julius D. The vanilloid receptor a molecular gateway to the pain pathway. Ann. Rev. Neurosci. 2001;24:487–517. doi: 10.1146/annurev.neuro.24.1.487. [DOI] [PubMed] [Google Scholar]
  • 30.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. doi: 10.1126/science.288.5464.306. [DOI] [PubMed] [Google Scholar]
  • 31.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. doi: 10.1038/39807. [DOI] [PubMed] [Google Scholar]
  • 32.Cesare P, McNaughton P. A novel heat-activated current in nociceptive neurons and its sensitization by bradykinin [see comments] Proc. Natl. Acad. Sci. USA. 1996;93:15435–15439. doi: 10.1073/pnas.93.26.15435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Childers SR. Activation of G-proteins in brain by endogenous and exogenous cannabinoids. AAPSJ. 2006;8:E112–E117. doi: 10.1208/aapsj080113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chou MZ, Mtui T, Gao YD, Kohler M, Middleton RE. Resiniferatoxin binds to the capsaicin receptor (TRPV1) near the extracellular side of the S4 transmembrane domain. Biochemistry. 2004;43:2501–2511. doi: 10.1021/bi035981h. [DOI] [PubMed] [Google Scholar]
  • 35.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. doi: 10.1038/35082088. [DOI] [PubMed] [Google Scholar]
  • 36.Chung K, Schwen RJ, Coggeshall RE. Ureteral axon damage following subcutaneous administration of capsaicin in adult rats. Neurosci. Lett. 1985;53:221–226. doi: 10.1016/0304-3940(85)90189-2. [DOI] [PubMed] [Google Scholar]
  • 37.Cruz F, Guimaraes M, Silva C, Reis M. Suppression of bladder hyperreflexia by intravesical resiniferatoxin. Lancet. 1997;350:640–641. doi: 10.1016/S0140-6736(05)63330-2. [DOI] [PubMed] [Google Scholar]
  • 38.Cui M, Honore P, Zhong C, Gauvin D, Mikusa J, Hernandez G, Chandran P, Gomtsyan A, Brown B, Bayburt EK, Marsh K, Bianchi B, McDonald H, Niforatos W, Neelands TR, Moreland RB, Decker MW, Lee CH, Sullivan JP, Faltynek CR. TRPV1 receptors in the CNS play a key role in broad-spectrum analgesia of TRPV1 antagonists. J. Neurosci. 2006;26:9385–9393. doi: 10.1523/JNEUROSCI.1246-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Dai Y, Moriyama T, Higashi T, Togashi K, Kobayashi K, Yamanaka H, Tominaga M, Noguchi K. Proteinase-activated receptor 2-mediated potentiation of transient receptor potential vanilloid subfamily 1 activity reveals a mechanism for proteinase-induced inflammatory pain. J. Neurosci. 2004;24:4293–4299. doi: 10.1523/JNEUROSCI.0454-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Dasgupta P, Chandiramani VA, Beckett A, Scaravilli F, Fowler CJ. The effect of intravesical capsaicin on the suburothelial innervation in patients with detrusor hyper-reflexia. B.J.U. Int. 2000;85:238–245. doi: 10.1046/j.1464-410x.2000.00427.x. [DOI] [PubMed] [Google Scholar]
  • 41.Dattilio A, Vizzard MA. Up-regulation of protease activated receptors in bladder after cyclophosphamide induced cystitis and colocalization with capsaicin receptor (VR1) in bladder nerve fibers. J. Urol. 2005;173:635–639. doi: 10.1097/01.ju.0000143191.55468.1d. [DOI] [PubMed] [Google Scholar]
  • 42.Davis JB, Gray J, Gunthorpe MJ, Hatcher JP, Davey PT, Overend P, Harries MH, Latcham J, Clapham C, Atkinson K, Hughes SA, Rance K, Grau E, Harper AJ, Pugh PL, Rogers DC, Bingham S, Randall A, Sheardown SA. Vanilloid receptor-1 is essential for inflammatory thermal hyperalgesia. Nature. 2000;405:183–187. doi: 10.1038/35012076. [DOI] [PubMed] [Google Scholar]
  • 43.Dawbarn D, Harmar AJ, Pycock CJ. Intranigral injection of capsaicin enhances motor activity and depletes nigral 5-hydroxytryptamine but not substance P. Neuropharmacology. 1981;20:341–346. doi: 10.1016/0028-3908(81)90006-x. [DOI] [PubMed] [Google Scholar]
  • 44.De Petrocellis L, Harrison S, Bisogno T, Tognetto M, Brandi I, Smith GD, Creminon C, Davis JB, Geppetti P, Di Marzo V. The vanilloid receptor (VR1)-mediated effects of anandamide are potently enhanced by the cAMP-dependent protein kinase. J. Neurochem. 2001;77:1660–1663. doi: 10.1046/j.1471-4159.2001.00406.x. [DOI] [PubMed] [Google Scholar]
  • 45.De Ridder D, Chandiramani V, Dasgupta P, Van Poppel H, Baert L, Fowler CJ. Intravesical capsaicin as a treatment for refractory detrusor hyperreflexia: a dual center study with long-term followup. J. Urol. 1997;158:2087–2092. doi: 10.1016/s0022-5347(01)68162-x. [DOI] [PubMed] [Google Scholar]
  • 46.De Seze SM, Wiart L, de Seze MP, Soyeur L, Dosque JP, Blajezewski S, Moore N, Brochet B, Mazaux JM, Barat M, Joseph PA. Intravesical capsaicin versus resiniferatoxin for the treatment of detrusor hyperreflexia in spinal cord injured patients: a double-blind, randomized, controlled study. J. Urol. 2004;171:251–255. doi: 10.1097/01.ju.0000100385.93801.d4. [DOI] [PubMed] [Google Scholar]
  • 47.Denda M, Fuziwara S, Inoue K, Denda S, Akamatsu H, Tomitaka A, Matsunaga K. Immunoreactivity of VR1 on epidermal keratinocyte of human skin. Biochem. Biophys. Res. Commun. 2001;285:1250–1252. doi: 10.1006/bbrc.2001.5299. [DOI] [PubMed] [Google Scholar]
  • 48.Dinis P, Charrua A, Avelino A, Cruz F. Intravesical resiniferatoxin decreases spinal c-fos expression and increases bladder volume to reflex micturition in rats with chronic inflamed urinary bladders. B.J.U. Int. 2004;94:153–157. doi: 10.1111/j.1464-4096.2004.04855.x. [DOI] [PubMed] [Google Scholar]
  • 49.Ditting T, Linz P, Hilgers KF, Jung O, Geiger H, Veelken R. Putative role of epithelial sodium channels (ENaC) in the afferent limb of cardio renal reflexes in rats. Basic Res. Cardiol. 2003;98:388–400. doi: 10.1007/s00395-003-0426-7. [DOI] [PubMed] [Google Scholar]
  • 50.Docherty RJ, Yeats JC, Bevan S, Boddeke HW. Inhibition of calcineurin inhibits the desensitization of capsaicin-evoked currents in cultured dorsal root ganglion neurones from adult rats. Pflugers Arch. 1996;431:828–837. doi: 10.1007/s004240050074. [DOI] [PubMed] [Google Scholar]
  • 51.Docherty RJ, Yeats JC, Piper AS. Capsazepine block of voltage-activated calcium channels in adult rat dorsal root ganglion neurones in culture. Br. J. Pharmacol. 1997;121:1461–1467. doi: 10.1038/sj.bjp.0701272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Doyle MW, Bailey TW, Jin YH, Andresen MC. Vanilloid receptors presynaptically modulate cranial visceral afferent synaptic transmission in nucleus tractus solitarius. J. Neurosci. 2002;22:8222–8229. doi: 10.1523/JNEUROSCI.22-18-08222.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Dray A, Forbes CA, Burgess GM. Ruthenium red blocks the capsaicin-induced increase in intracellular calcium and activation of membrane currents in sensory neurones as well as the activation of peripheral nociceptors in vitro. Neurosci. Lett. 1990;110:52–59. doi: 10.1016/0304-3940(90)90786-9. [DOI] [PubMed] [Google Scholar]
  • 54.El Kouhen R, Surowy CS, Bianchi BR, Neelands TR, McDonald HA, Niforatos W, Gomtsyan A, Lee CH, Honore P, Sullivan JP, Jarvis MF, Faltynek CR. A- 425619 [1-isoquinolin-5-yl-3-(4-trifluoromethyl-benzyl)-urea], a novel and selective transient receptor potential type V1 receptor antagonist, blocks channel activation by vanilloids, heat, and acid. J. Pharmacol. Exp. Ther. 2005;314:400–409. doi: 10.1124/jpet.105.084103. [DOI] [PubMed] [Google Scholar]
  • 55.Fukuoka T, Tokunaga A, Tachibana T, Dai Y, Yamanaka H, Noguchi K. VR1, but not P2X3, increases in the spared L4 DRG in rats with L5 spinal nerve ligation. Pain. 2002;99:111–120. doi: 10.1016/s0304-3959(02)00067-2. [DOI] [PubMed] [Google Scholar]
  • 56.Gavva NR, Bannon AW, Surapaneni S, Hovland DN Jr, Lehto SG, Gore A, Juan T, Deng H, Han B, Klionsky L, Kuang R, Le A, Tamir R, Wang J, Youngblood B, Zhu D, Norman MH, Magal E, Treanor JJ, Louis JC. The vanilloid receptor TRPV1 is tonically activated in vivo and involved in body temperature regulation. J. Neurosci. 2007;27:3366–3374. doi: 10.1523/JNEUROSCI.4833-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Gavva NR, Klionsky L, Qu Y, Shi L, Tamir R, Edenson S, Zhang TJ, Viswanadhan VN, Toth A, Pearce LV, Vanderah TW, Porreca F, Blumberg PM, Lile J, Sun Y, Wild K, Louis JC, Treanor JJ. Molecular determinants of vanilloid sensitivity in TRPV1. J. Biol. Chem. 2004;279:20283–20295. doi: 10.1074/jbc.M312577200. [DOI] [PubMed] [Google Scholar]
  • 58.Geppetti P, Materazzi S, Nicoletti P. The transient receptor potential vanilloid 1: role in airway inflammation and disease. Eur. J. Pharmacol. 2006;533:207–214. doi: 10.1016/j.ejphar.2005.12.063. [DOI] [PubMed] [Google Scholar]
  • 59.Green T, Dockray GJ. Characterization of the peptidergic afferent innervation of the stomach in the rat, mouse and guinea-pig. Neuroscience. 1988;25:181–193. doi: 10.1016/0306-4522(88)90017-6. [DOI] [PubMed] [Google Scholar]
  • 60.Gunthorpe MJ, Benham CD, Randall A, Davis JB. The diversity in the vanilloid (TRPV) receptor family of ion channels. Trends Pharmacol. Sci. 2002;23:183–191. doi: 10.1016/s0165-6147(02)01999-5. [DOI] [PubMed] [Google Scholar]
  • 61.Gunthorpe MJ, Rami HK, Jerman JC, Smart D, Gill CH, Soffin EM, Luis Hannan S, Lappin SC, Egerton J, Smith GD, Worby A, Howett L, Owen D, Nasir S, Davies CH, Thompson M, Wyman PA, Randall AD, Davis JB. Identification and characterisation of SB-366791 a potent and selective vanilloid receptor (VR1/TRPV1) antagonist. Neuropharmacology. 2004;46:133–149. doi: 10.1016/s0028-3908(03)00305-8. [DOI] [PubMed] [Google Scholar]
  • 62.Harding SM. Recent clinical investigations examining the association of asthma and gastroesophageal reflux. Am. J. Med. 2003;115(Suppl. 3A):39S–44S. doi: 10.1016/s0002-9343(03)00191-8. [DOI] [PubMed] [Google Scholar]
  • 63.Harvey JS, Davis C, James IF, Burgess GM. Activation of protein kinase C by the capsaicin analogue resiniferatoxin in sensory neurones. J. Neurochem. 1995;65:1309–1317. doi: 10.1046/j.1471-4159.1995.65031309.x. [DOI] [PubMed] [Google Scholar]
  • 64.Hingtgen CM, Waite KJ, Vasko MR. Prostaglandins facilitate peptide release from rat sensory neurons by activating the adenosine 3',5'-cyclic monophosphate transduction cascade. J. Neurosci. 1995;15:5411–5419. doi: 10.1523/JNEUROSCI.15-07-05411.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Holzer P. Vanilloid receptor TRPV1 hot on the tongue and inflaming the colon. Neurogastroenterol. Motil. 2004;16:697–699. doi: 10.1111/j.1365-2982.2004.00598.x. [DOI] [PubMed] [Google Scholar]
  • 66.Holzer P, Maggi CA. Dissociation of dorsal root ganglion neurons into afferent and efferent-like neurons. Neuroscience. 1998;86:389–398. doi: 10.1016/s0306-4522(98)00047-5. [DOI] [PubMed] [Google Scholar]
  • 67.Holzer P. Capsaicin: cellular targets, mechanisms of action, and selectivity for thin sensory neurons. Pharmacol. Rev. 1991;43:143–201. [PubMed] [Google Scholar]
  • 68.Holzer P. Local effector functions of capsaicin-sensitive sensory nerve endings: involvement of tachykinins, calcitonin gene-related peptide and other neuropeptides. Neuroscience. 1988;24:739–768. doi: 10.1016/0306-4522(88)90064-4. [DOI] [PubMed] [Google Scholar]
  • 69.Horie S, Yamamoto H, Michael GJ, Uchida M, Belai A, Watanabe K, Priestley JV, Murayama T. Protective role of vanilloid receptor type 1 in HCl-induced gastric mucosal lesions in rats. Scand. J. Gastroenterol. 2004;39:303–312. doi: 10.1080/00365520310008647. [DOI] [PubMed] [Google Scholar]
  • 70.Hu HJ, Bhave G, Gereau RW. Prostaglandin and protein kinase A-dependent modulation of vanilloid receptor function by metabotropic glutamate receptor 5: potential mechanism for thermal hyperalgesia. J. Neurosci. 2002;22:7444–7452. doi: 10.1523/JNEUROSCI.22-17-07444.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Huang SM, Bisogno T, Trevisani M, Al-Hayani A, De Petrocellis L, Fezza F, Tognetto M, Petros TJ, Krey JF, Chu CJ, Miller JD, Davies SN, Geppetti P, Walker JM, Di Marzo V. An endogenous capsaicin-like substance with high potency at recombinant and native vanilloid VR1 receptors. Proc. Natl. Acad. Sci. U.S.A. 2002;99:8400–8405. doi: 10.1073/pnas.122196999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Hudson LJ, Bevan S, Wotherspoon G, Gentry C, Fox A, Winter J. VR1 protein expression increases in undamaged DRG neurons after partial nerve injury. Eur. J. Neurosci. 2001;13:2105–2114. doi: 10.1046/j.0953-816x.2001.01591.x. [DOI] [PubMed] [Google Scholar]
  • 73.Hwang SW, Cho H, Kwak J, Lee SY, Kang CJ, Jung J, Cho S, Min KH, Suh YG, Kim D, Oh U. Direct activation of capsaicin receptors by products of lipoxygenases: endogenous capsaicin-like substances. Proc. Natl. Acad. Sci. U.S.A. 2000;97:6155–6160. doi: 10.1073/pnas.97.11.6155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Iida T, Shimizu I, Nealen ML, Campbell A, Caterina M. Attenuated fever response in mice lacking TRPV1. Neurosci. Lett. 2005;378:28–33. doi: 10.1016/j.neulet.2004.12.007. [DOI] [PubMed] [Google Scholar]
  • 75.Inoue K, Koizumi S, Fuziwara S, Denda S, Denda M. Functional vanilloid receptors in cultured normal human epidermal keratinocytes. Biochem. Biophys. Res. Commun. 2002;291:124–129. doi: 10.1006/bbrc.2002.6393. [DOI] [PubMed] [Google Scholar]
  • 76.Ji RR, Samad TA, Jin SX, Schmoll R, Woolf CJ. p38 MAPK activation by NGF in primary sensory neurons after inflammation increases TRPV1 levels and maintains heat hyperalgesia. Neuron. 2002;36:57–68. doi: 10.1016/s0896-6273(02)00908-x. [DOI] [PubMed] [Google Scholar]
  • 77.Jin X, Morsy N, Winston J, Pasricha PJ, Garrett K, Akbarali HI. Modulation of TRPV1 by nonreceptor tyrosine kinase, c-Src kinase. Am. J. Physiol. Cell Physiol. 2004;287:558–563. doi: 10.1152/ajpcell.00113.2004. [DOI] [PubMed] [Google Scholar]
  • 78.Jordt SE, Julius D. Molecular basis for species-specific sensitivity to "hot" chili peppers. Cell. 2002;108:421–430. doi: 10.1016/s0092-8674(02)00637-2. [DOI] [PubMed] [Google Scholar]
  • 79.Jordt SE, Tominaga M, Julius D. Acid potentiation of the capsaicin receptor determined by a key extracellular site. Proc. Natl. Acad. Sci. USA. 2000;97:8134–8139. doi: 10.1073/pnas.100129497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Julius D, Basbaum AI. Molecular mechanisms of nociception. Nature. 2001;413:203–210. doi: 10.1038/35093019. [DOI] [PubMed] [Google Scholar]
  • 81.Jung J, Hwang SW, Kwak J, Lee SY, Kang CJ, Kim WB, Kim D, Oh U. Capsaicin binds to the intracellular domain of the capsaicin-activated ion channel. J. Neurosci. 1999;19:529–538. doi: 10.1523/JNEUROSCI.19-02-00529.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Jung J, Lee SY, Hwang SW, Cho H, Shin J, Kang YS, Kim S, Oh U. Agonist recognition sites in the cytosolic tails of vanilloid receptor 1. J. Biol. Chem. 2002;277:44448–44454. doi: 10.1074/jbc.M207103200. [DOI] [PubMed] [Google Scholar]
  • 83.Jung J, Shin JS, Lee SY, Hwang SW, Koo J, Cho H, Oh U. Phosphorylation of vanilloid receptor 1 by Ca2+/calmodulin-dependent kinase II regulates its vanilloid binding. J. Biol. Chem. 2004;279:7048–7054. doi: 10.1074/jbc.M311448200. [DOI] [PubMed] [Google Scholar]
  • 84.Kagawa S, Aoi M, Kubo Y, Kotani T, Takeuchi K. Stimulation by capsaicin of duodenal HCO3(-) secretion via afferent neurons and vanilloid receptors in rats: comparison with acid-induced HCO3(-) response. Dig. Dis. Sci. 2003;48:1850–1856. doi: 10.1023/a:1025480003388. [DOI] [PubMed] [Google Scholar]
  • 85.Karlsson U, Sundgren-Andersson AK, Johansson S, Krupp JJ. Capsaicin augments synaptic transmission in the rat medial preoptic nucleus. Brain Res. 2005;1043:1–11. doi: 10.1016/j.brainres.2004.10.064. [DOI] [PubMed] [Google Scholar]
  • 86.Kato S, Aihara E, Nakamura A, Xin H, Matsui H, Kohama K, Takeuchi K. Expression of vanilloid receptors in rat gastric epithelial cells: role in cellular protection. Biochem. Pharmacol. 2003;66:1115–1121. doi: 10.1016/s0006-2952(03)00461-1. [DOI] [PubMed] [Google Scholar]
  • 87.Kawabata A, Nishikawa H, Saitoh H, Nakaya Y, Hiramatsu K, Kubo S, Nishida M, Kawao N, Kuroda R, Sekiguchi F, Kinoshita M, Kakehi K, Arizono N, Yamagishi H, Kawai K. A protective role of protease-activated receptor 1 in rat gastric mucosa. Gastroenterology. 2004;126:208–219. doi: 10.1053/j.gastro.2003.10.071. [DOI] [PubMed] [Google Scholar]
  • 88.Keeble J, Russell F, Curtis B, Starr A, Pinter E, Brain SD. Involvement of transient receptor potential vanilloid 1 in the vascular and hyperalgesic components of joint inflammation. Arthritis Rheum. 2005;52:3248–3256. doi: 10.1002/art.21297. [DOI] [PubMed] [Google Scholar]
  • 89.Khasar SG, Lin YH, Martin A, Dadgar J, McMahon T, Wang D, Hundle B, Aley KO, Isenberg W, McCarter G, Green PG, Hodge CW, Levine JD, Messing RO. A novel nociceptor signaling pathway revealed in protein kinase C epsilon mutant mice. Neuron. 1999;24:253–260. doi: 10.1016/s0896-6273(00)80837-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Kihara N, de la Fuente SG, Fujino K, Takahashi T, Pappas TN, Mantyh CR. Vanilloid receptor-1 containing primary sensory neurones mediate dextran sulphate sodium induced colitis in rats. Gut. 2003;52:713–719. doi: 10.1136/gut.52.5.713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Kimball ES, Wallace NH, Schneider CR, D'Andrea MR, Hornby PJ. Vanilloid receptor 1 antagonists attenuate disease severity in dextran sulphate sodium-induced colitis in mice. Neurogastroenterol. Motil. 2004;16:811–818. doi: 10.1111/j.1365-2982.2004.00549.x. [DOI] [PubMed] [Google Scholar]
  • 92.Kitahara T, Li HS, Balaban CD. Changes in transient receptor potential cation channel superfamily V (TRPV) mRNA expression in the mouse inner ear ganglia after kanamycin challenge. Hear Res. 2005;201:132–144. doi: 10.1016/j.heares.2004.09.007. [DOI] [PubMed] [Google Scholar]
  • 93.Kofalvi A, Oliveira CR, Cunha RA. Lack of evidence for functional TRPV1 vanilloid receptors in rat hippocampal nerve terminals. Neurosci. Lett. 2006;403:151–156. doi: 10.1016/j.neulet.2006.04.030. [DOI] [PubMed] [Google Scholar]
  • 94.Kuzhikandathil EV, Wang H, Szabo T, Morozova N, Blumberg PM, Oxford GS. Functional analysis of capsaicin receptor (vanilloid receptor subtype 1) multimerization and agonist responsiveness using a dominant negative mutation. J. Neurosci. 2001;21:8697–8706. doi: 10.1523/JNEUROSCI.21-22-08697.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Lalloo UG, Fox AJ, Belvisi MG, Chung KF, Barnes PJ. Capsazepine inhibits cough induced by capsaicin and citric acid but not by hypertonic saline in guinea pigs. J. Appl. Physiol. 1995;79:1082–1087. doi: 10.1152/jappl.1995.79.4.1082. [DOI] [PubMed] [Google Scholar]
  • 96.Lappin SC, Randall AD, Gunthorpe MJ, Morisset V. TRPV1 antagonist, SB-366791, inhibits glutamatergic synaptic transmission in rat spinal dorsal horn following peripheral inflammation. Eur. J. Pharmacol. 2006;540:73–81. doi: 10.1016/j.ejphar.2006.04.046. [DOI] [PubMed] [Google Scholar]
  • 97.Lazzeri M, Beneforti P, Spinelli M, Zanollo A, Barbagli G, Turini D. Intravesical resiniferatoxin for the treatment of hypersensitive disorder: a randomized placebo controlled study. J. Urol. 2000;164:676–679. doi: 10.1097/00005392-200009010-00014. [DOI] [PubMed] [Google Scholar]
  • 98.Lazzeri M, Spinelli M, Beneforti P, Zanollo A, Turini D. Intravesical resiniferatoxin for the treatment of detrusor hyperreflexia refractory to capsaicin in patients with chronic spinal cord diseases. Scand. J. Urol. Nephrol. 1998;32:331–334. doi: 10.1080/003655998750015287. [DOI] [PubMed] [Google Scholar]
  • 99.Lazzeri M, Vannucchi MG, Zardo C, Spinelli M, Beneforti P, Turini D, Faussone-Pellegrini MS. Immunohistochemical evidence of vanilloid receptor 1 in normal human urinary bladder. Eur. Urol. 2004;46:792–798. doi: 10.1016/j.eururo.2004.08.007. [DOI] [PubMed] [Google Scholar]
  • 100.Liapi A, Wood JN. Extensive co-localization and heteromultimer formation of the vanilloid receptor-like protein TRPV2 and the capsaicin receptor TRPV1 in the adult rat cerebral cortex. Eur. J. Neurosci. 2005;22:825–834. doi: 10.1111/j.1460-9568.2005.04270.x. [DOI] [PubMed] [Google Scholar]
  • 101.Liu L, Simon SA. Capsazepine, a vanilloid receptor antagonist, inhibits nicotinic acetylcholine receptors in rat trigeminal ganglia. Neurosci. Lett. 1997;228:29–32. doi: 10.1016/s0304-3940(97)00358-3. [DOI] [PubMed] [Google Scholar]
  • 102.Lopshire JC, Nicol GD. The cAMP transduction cascade mediates the prostaglandin E2 enhancement of the capsaicin-elicited current in rat sensory neurons: whole-cell and single-channel studies. J. Neurosci. 1998;18:6081–92. doi: 10.1523/JNEUROSCI.18-16-06081.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Lyall V, Heck GL, Vinnikova AK, Ghosh S, Phan TH, Alam RI, Russell OF, Malik SA, Bigbee JW, DeSimone JA. The mammalian amiloride-insensitive non-specific salt taste receptor is a vanilloid receptor-1 variant. J. Physiol. 2004;558:147–159. doi: 10.1113/jphysiol.2004.065656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Lysy J, Sistiery-Ittah M, Israelit Y, Shmueli A, Strauss-Liviatan N, Mindrul V, Keret D, Goldin E. Topical capsaicin-a novel and effective treatment for idiopathic intractable pruritus ani: a randomised, placebo controlled, crossover study. Gut. 2003;52:1323–1326. doi: 10.1136/gut.52.9.1323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Maggi CA. Tachykinins and calcitonin gene-related peptide (CGRP) as co-transmitters released from peripheral endings of sensory nerves. Prog. Neurobiol. 1995;45:1–98. doi: 10.1016/0301-0082(94)e0017-b. [DOI] [PubMed] [Google Scholar]
  • 106.Maggi CA, Meli A. The sensory-efferent function of capsaicin-sensitive sensory neurons. Gen. Pharmacol. 1988;19:1–43. doi: 10.1016/0306-3623(88)90002-x. [DOI] [PubMed] [Google Scholar]
  • 107.Malmberg AB, Chen C, Tonegawa S, Basbaum AI. Preserved acute pain and reduced neuropathic pain in mice lacking PKCgamma. Science. 1997;278:279–283. doi: 10.1126/science.278.5336.279. [DOI] [PubMed] [Google Scholar]
  • 108.Malmberg AB, Yaksh TL. Capsaicin-evoked prostaglandin E2 release in spinal cord slices: relative effect of cyclooxygenase inhibitors. Eur. J. Pharmacol. 1994;271:293–299. doi: 10.1016/0014-2999(94)90786-2. [DOI] [PubMed] [Google Scholar]
  • 109.Marinelli S, Di Marzo V, Berretta N, Matias I, Maccarrone M, Bernardi G, Mercuri NB. Presynaptic facilitation of glutamatergic synapses to dopaminergic neurons of the rat substantia nigra by endogenous stimulation of vanilloid receptors. J. Neurosci. 2003;23:3136–3144. doi: 10.1523/JNEUROSCI.23-08-03136.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Marinelli S, Pascucci T, Bernardi G, Puglisi-Allegra S, Mercuri NB. Activation of TRPV1 in the VTA excites dopaminergic neurons and increases chemical- and noxious-induced dopamine release in the nucleus accumbens. Neuropsychopharmacology. 2005;30:864–870. doi: 10.1038/sj.npp.1300615. [DOI] [PubMed] [Google Scholar]
  • 111.Marinelli S, Vaughan CW, Christie MJ, Connor M. Capsaicin activation of glutamatergic synaptic transmission in the rat locus coeruleus in vitro. J. Physiol. 2002;543:531–540. doi: 10.1113/jphysiol.2002.022863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Marsch R, Foeller E, Rammes G, Bunck M, Kossl M, Holsboer F, Zieglgansberger W, Landgraf R, Lutz B, Wotjak CT. Reduced anxiety, conditioned fear, and hippocampal long-term potentiation in transient receptor potential vanilloid type 1 receptor-deficient mice. J. Neurosci. 2007;27:832–839. doi: 10.1523/JNEUROSCI.3303-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Mason L, Moore RA, Derry S, Edwards JE, McQuay HJ. Systematic review of topical capsaicin for the treatment of chronic pain. BMJ. 2004;328:991. doi: 10.1136/bmj.38042.506748.EE. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.McDonald DM, Bowden JJ, Baluk P, Bunnett NW. Neurogenic inflammation. A model for studying efferent actions of sensory nerves. Adv. Exp. Med. Biol. 1996;410:453–462. [PubMed] [Google Scholar]
  • 115.McGaraughty S, Chu KL, Bitner RS, Martino B, El KR, Han P, Nikkel AL, Burgard EC, Faltynek CR, Jarvis MF. Capsaicin infused into the PAG affects rat tail flick responses to noxious heat and alters neuronal firing in the RVM. J. Neurophysiol. 2003;90:2702–2710. doi: 10.1152/jn.00433.2003. [DOI] [PubMed] [Google Scholar]
  • 116.McQueen DS, Bond SM, Moores C, Chessell I, Humphrey PP, Dowd E. Activation of P2X receptors for adenosine triphosphate evokes cardiorespiratory reflexes in anaesthetized rats. J. Physiol. 1998;507:843–855. doi: 10.1111/j.1469-7793.1998.843bs.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.McVey DC, Schmid PC, Schmid HH, Vigna SR. Endocannabinoids induce ileitis in rats via the capsaicin receptor (VR1) J. Pharmacol. Exp. Ther. 2003;304:713–722. doi: 10.1124/jpet.102.043893. [DOI] [PubMed] [Google Scholar]
  • 118.McVey DC, Vigna SR. The capsaicin VR1 receptor mediates substance P release in toxin A-induced enteritis in rats. Peptides. 2001;22:1439–1446. doi: 10.1016/s0196-9781(01)00463-6. [DOI] [PubMed] [Google Scholar]
  • 119.Mezey E, Toth ZE, Cortright DN, Arzubi MK, Krause JE, Elde R, Guo A, Blumberg PM, Szallasi A. Distribution of mRNA for vanilloid receptor subtype 1 (VR1), and VR1-like immunoreactivity, in the central nervous system of the rat and human. Proc. Natl. Acad. Sci. U.S.A. 2000;97:3655–3660. doi: 10.1073/pnas.060496197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Michael GJ, Priestley JV. Differential expression of the mRNA for the vanilloid receptor subtype 1 in cells of the adult rat dorsal root and nodose ganglia and its downregulation by axotomy. J. Neurosci. 1999;19:1844–1854. doi: 10.1523/JNEUROSCI.19-05-01844.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Mohapatra DP, Nau C. Desensitization of capsaicin-activated currents in the vanilloid receptor TRPV1 is decreased by the cyclic AMP-dependent protein kinase pathway. J. Biol. Chem. 2003;278:50080–50090. doi: 10.1074/jbc.M306619200. [DOI] [PubMed] [Google Scholar]
  • 122.Mohapatra DP, Nau C. Regulation of Ca2+-dependent desensitization in the vanilloid receptor TRPV1 by calcineurin and cAMP-dependent protein kinase. J. Biol. Chem. 2005;280:13424–13432. doi: 10.1074/jbc.M410917200. [DOI] [PubMed] [Google Scholar]
  • 123.Montell C. Physiology, phylogeny, and functions of the TRP superfamily of cation channels. Sci. STKE. 2001;90:RE1. doi: 10.1126/stke.2001.90.re1. [DOI] [PubMed] [Google Scholar]
  • 124.Morenilla-Palao C, Planells-Cases R, Garcia-Sanz N, Ferrer-Montiel A. Regulated exocytosis contributes to protein kinase C potentiation of vanilloid receptor activity. J. Biol. Chem. 2004;279:25665–25672. doi: 10.1074/jbc.M311515200. [DOI] [PubMed] [Google Scholar]
  • 125.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. doi: 10.1186/1744-8069-1-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Moriyama T, Iida T, Kobayashi K, Higashi T, Fukuoka T, Tsumura H, Leon C, Suzuki N, Inoue K, Gachet C, Noguchi K, Tominaga M. Possible involvement of P2Y2 metabotropic receptors in ATP-induced transient receptor potential vanilloid receptor 1-mediated thermal hypersensitivity. J. Neurosci. 2003;23:6058–6062. doi: 10.1523/JNEUROSCI.23-14-06058.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Nagy JI, Emson PC, Iversen LL. A re-evaluation of the neurochemical and antinociceptive effects of intrathecal capsaicin in the rat. Brain Res. 1981;211:497–502. doi: 10.1016/0006-8993(81)90980-x. [DOI] [PubMed] [Google Scholar]
  • 128.Nakashima M, Aoi M, Aihara E, Takeuchi K. No role for prostacyclin IP receptors in duodenal. Digestion. 2004;70:16–25. doi: 10.1159/000080077. [DOI] [PubMed] [Google Scholar]
  • 129.Nathan JD, Patel AA, McVey DC, Thomas JE, Prpic V, Vigna SR, Liddle RA. Capsaicin vanilloid receptor-1 mediates substance P release in experimental pancreatitis. Am. J. Physiol. Gastrointest. Liver Physiol. 2001;281:G1322–G1328. doi: 10.1152/ajpgi.2001.281.5.G1322. [DOI] [PubMed] [Google Scholar]
  • 130.Numazaki M, Tominaga T, Toyooka H, Tominaga M. Direct phosphorylation of capsaicin receptor VR1 by protein kinase Cepsilon and identification of two target serine residues. J. Biol. Chem. 2002;277:13375–13378. doi: 10.1074/jbc.C200104200. [DOI] [PubMed] [Google Scholar]
  • 131.Olah Z, Szabo T, Karai L, Hough C, Fields RD, Caudle RM, Blumberg PM, Iadarola MJ. Ligand-induced dynamic membrane changes and cell deletion conferred by vanilloid receptor 1. J. Biol. Chem. 2001;276:11021–11030. doi: 10.1074/jbc.M008392200. [DOI] [PubMed] [Google Scholar]
  • 132.Palazzo E, de NV, Marabese I, Cuomo D, Rossi F, Berrino L, Rossi F, Maione S. Interaction between vanilloid and glutamate receptors in the central modulation of nociception. Eur. J. Pharmacol. 2002;439:69–75. doi: 10.1016/s0014-2999(02)01367-5. [DOI] [PubMed] [Google Scholar]
  • 133.Pan HL, Khan GM, Alloway KD, Chen SR. Resiniferatoxin induces paradoxical changes in thermal and mechanical sensitivities in rats mechanism of action. J. Neurosci. 2003;23:2911–2919. doi: 10.1523/JNEUROSCI.23-07-02911.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.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. doi: 10.1111/j.1469-7793.1999.0857p.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Parlani M, Conte B, Goso C, Szallasi A, Manzini S. Capsaicin-induced relaxation in the rat isolated external urethral sphincter: characterization of the vanilloid receptor and mediation by CGRP. Br. J. Pharmacol. 1993;110:989–994. doi: 10.1111/j.1476-5381.1993.tb13911.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Patterson LM, Zheng H, Ward SM, Berthoud HR. Vanilloid receptor (VR1) expression in vagal afferent neurons innervating the gastrointestinal tract. Cell Tissue Res. 2003;311:277–287. doi: 10.1007/s00441-002-0682-0. [DOI] [PubMed] [Google Scholar]
  • 137.Pomonis JD, Harrison JE, Mark L, Bristol DR, Valenzano KJ, Walker K. N-(4-Tertiarybutylphenyl)-4-(3-cholorphy-ridin-2-yl)tetrahydropyrazine -1(2H)-carbox-amide (BCTC), a novel, orally effective vanilloid receptor 1 antagonist with analgesic properties: II in vivo characterization in rat models of inflammatory and neuropathic pain. J. Pharmacol. Exp. Ther. 2003;306:387–393. doi: 10.1124/jpet.102.046268. [DOI] [PubMed] [Google Scholar]
  • 138.Premkumar LS, Ahern GP. Induction of vanilloid receptor channel activity by protein kinase C. Nature. 2000;408:985–990. doi: 10.1038/35050121. [DOI] [PubMed] [Google Scholar]
  • 139.Prescott ED, Julius D. A modular PIP2 binding site as a determinant of capsaicin receptor sensitivity. Science. 2003;300:1284–1288. doi: 10.1126/science.1083646. [DOI] [PubMed] [Google Scholar]
  • 140.Raisinghani M, Pabbidi RM, Premkumar LS. Activation of transient receptor potential vanilloid 1 (TRPV1) by resiniferatoxin. J. Physiol. 2005;567:771–786. doi: 10.1113/jphysiol.2005.087874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Rathee PK, Distler C, Obreja O, Neuhuber W, Wang GK, Wang SY, Nau C, Kress M. PKA/AKAP/VR-1 module: A common link of Gs-mediated signaling to thermal hyperalgesia. J. Neurosci. 2002;22:4740–4745. doi: 10.1523/JNEUROSCI.22-11-04740.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Rawls SM, Ding Z, Cowan A. Role of TRPV1 and cannabinoid CB1 receptors in AM 404-evoked hypothermia in rats. Pharmacol. Biochem. Behav. 2006;83:508–516. doi: 10.1016/j.pbb.2006.03.011. [DOI] [PubMed] [Google Scholar]
  • 143.Razavi R, Chan Y, Afifiyan FN, Liu XJ, Wan X, Yantha J, Tsui H, Tang L, Tsai S, Santamaria P, Driver JP, Serreze D, Salter MW, Dosch HM. TRPV1+ sensory neurons control beta cell stress and islet inflammation in autoimmune diabetes. Cell. 2006;127:1123–1135. doi: 10.1016/j.cell.2006.10.038. [DOI] [PubMed] [Google Scholar]
  • 144.Reichling DB, Levine JD. The primary afferent nociceptor as pattern generator. Pain. 1999;(6):S103–S109. doi: 10.1016/S0304-3959(99)00143-8. [DOI] [PubMed] [Google Scholar]
  • 145.Rigoni M, Trevisani M, Gazzieri D, Nadaletto R, Tognetto M, Creminon C, Davis JB, Campi B, Amadesi S, Geppetti P, Harrison S. Neurogenic responses mediated by vanilloid receptor-1 (TRPV1) are blocked by the high affinity antagonist, iodo-resiniferatoxin. Br. J. Pharmacol. 2003;138:977–985. doi: 10.1038/sj.bjp.0705110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Ritter S, Dinh TT. Capsaicin-induced neuronal degeneration in the brain and retina of preweanling rats. J. Comp. Neurol. 1990;296:447–461. doi: 10.1002/cne.902960310. [DOI] [PubMed] [Google Scholar]
  • 147.Roberts JC, Davis JB, Benham CD. [3H]Resiniferatoxin autoradiography in the CNS of wild-type and TRPV1 null mice defines TRPV1 (VR-1) protein distribution. Brain Res. 2004;995:176–183. doi: 10.1016/j.brainres.2003.10.001. [DOI] [PubMed] [Google Scholar]
  • 148.Robinson P, Okhuysen PC, Chappell CL, Weinstock JV, Lewis DE, Actor JK, White AC Jr. Substance P expression correlates with severity of diarrhea in cryptosporidiosis. J. Infect. Dis. 2003;188:290–296. doi: 10.1086/376836. [DOI] [PubMed] [Google Scholar]
  • 149.Rosenbaum T, Gordon-Shaag A, Munari M, Gordon SE. Ca2+/calmodulin modulates TRPV1 activation by capsaicin. J. Gen. Physiol. 2004;123:53–62. doi: 10.1085/jgp.200308906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Salt TE, Hill RG. The effects of microiontophoretically applied capsaicin and substance P on single neurones in the rat and cat brain. Neurosci. Lett. 1980;20:329–334. doi: 10.1016/0304-3940(80)90169-x. [DOI] [PubMed] [Google Scholar]
  • 151.Sasamura T, Kuraishi Y. Peripheral and central actions of capsaicin and VR1 receptor. Jpn. J. Pharmacol. 1999;80:275–280. doi: 10.1254/jjp.80.275. [DOI] [PubMed] [Google Scholar]
  • 152.Schicho R, Florian W, Liebmann I, Holzer P, Lippe IT. Increased expression of TRPV1 receptor in dorsal root ganglia by acid insult of the rat gastric mucosa. Eur. J. Neurosci. 2004;19:1811–1818. doi: 10.1111/j.1460-9568.2004.03290.x. [DOI] [PubMed] [Google Scholar]
  • 153.Schultz HD. The spice of life is at the root of cardiac pain. J. Physiol. 2003;551:400. doi: 10.1113/jphysiol.2003.050104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Schultz HD, Ustinova EE. Cardiac vagal afferent stimulation by free radicals during ischaemia and reperfusion. Clin. Exp. Pharmacol. Physiol. 1996;23:700–708. doi: 10.1111/j.1440-1681.1996.tb01762.x. [DOI] [PubMed] [Google Scholar]
  • 155.Schultz HD, Ustinova EE. Capsaicin receptors mediate free radical-induced activation of cardiac afferent endings. Cardiovasc. Res. 1998;38:348–355. doi: 10.1016/s0008-6363(98)00031-5. [DOI] [PubMed] [Google Scholar]
  • 156.Scotland RS, Chauhan S, Davis C, De FC, Hunt S, Kabir J, Kotsonis P, Oh U, Ahluwalia A. Vanilloid receptor TRPV1 sensory C-fibers, and vascular autoregulation: a novel mechanism involved in myogenic constriction. Circ. Res. 2004;95:1027–1034. doi: 10.1161/01.RES.0000148633.93110.24. [DOI] [PubMed] [Google Scholar]
  • 157.Sculptoreanu A, de Groat WC, Buffington CA, Birder LA. Protein kinase C contributes to abnormal capsaicin responses in DRG neurons from cats with feline interstitial cystitis. Neurosci. Lett. 2005;381:42–46. doi: 10.1016/j.neulet.2005.01.080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Seabrook GR, Sutton KG, Jarolimek W, Hollingworth GJ, Teague S, Webb J, Clark N, Boyce S, Kerby J, Ali Z, Chou M, Middleton R, Kaczorowski G, Jones AB. Functional properties of the high-affinity TRPV1 (VR1) vanilloid receptor antagonist (4-hydroxy-5-iodo-3-methoxyphenylacetate ester) iodo-resiniferatoxin. J. Pharmacol. Exp. Ther. 2002;303:1052–1060. doi: 10.1124/jpet.102.040394. [DOI] [PubMed] [Google Scholar]
  • 159.Shimizu I, Iida T, Horiuchi N, Caterina MJ. 5-Iodo-resiniferatoxin evokes hypothermia in mice and is a partial transient receptor potential vanilloid 1 agonist in vitro. J. Pharmacol. Exp. Ther. 2005;314:1378–1385. doi: 10.1124/jpet.105.084277. [DOI] [PubMed] [Google Scholar]
  • 160.Shin J, Cho H, Hwang SW, Jung J, Shin CY, Lee SY, Kim SH, Lee MG, Choi YH, Kim J, Haber NA, Reichling DB, Khasar S, Levine JD, Oh U. Bradykinin-12-lipoxygenase-VR1 signaling pathway for inflammatory hyperalgesia. Proc. Natl. Acad. Sci. USA. 2002;99:10150–10155. doi: 10.1073/pnas.152002699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Shu XQ, Mendell LM. Neurotrophins and hyperalgesia. Proc. Natl. Acad. Sci. U.S.A. 1999;96:7693–7696. doi: 10.1073/pnas.96.14.7693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Sikand P, Premkumar LS. Potentiation of glutamatergic synaptic transmission by protein kinase C mediated sensitization of TRPV1 at the first sensory synapse. J. Physiol. 2007;581:631–647. doi: 10.1113/jphysiol.2006.118620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Simone DA, Nolano M, Johnson T, Wendelschafer-Crabb G, Kennedy WR. Intradermal injection of capsaicin in humans produces degeneration and subsequent reinnervation of epidermal nerve fibers: correlation with sensory function. J. Neurosci. 1998;18:8947–8959. doi: 10.1523/JNEUROSCI.18-21-08947.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Smith GD, Gunthorpe MJ, Kelsell RE, Hayes PD, Reilly P, Facer P, Wright JE, Jerman JC, Walhin JP, Ooi L, Egerton J, Charles KJ, Smart D, Randall AD, Anand P, Davis JB. TRPV3 is a temperature-sensitive vanilloid receptor-like protein. Nature. 2002;418:186–190. doi: 10.1038/nature00894. [DOI] [PubMed] [Google Scholar]
  • 165.Snitsarev V, Whiteis CA, Abboud FM, Chapleau MW. Mechanosensory transduction of vagal and baroreceptor afferents revealed by study of isolated nodose neurons in culture. Auton. Neurosci. 2002;98:59–63. doi: 10.1016/s1566-0702(02)00033-4. [DOI] [PubMed] [Google Scholar]
  • 166.Southall MD, Li T, Gharibova LS, Pei Y, Nicol GD, Travers JB. Activation of epidermal vanilloid receptor-1 induces release of proinflammatory mediators in human keratinocytes. J. Pharmacol. Exp. Ther. 2003;304:217–222. doi: 10.1124/jpet.102.040675. [DOI] [PubMed] [Google Scholar]
  • 167.Stein A, Ufret-Vincenty CA, Hua L, Santana LF, Gordon SE. Phosphoinositide 3-kinase binds to TRPV1 and mediates NGF-stimulated TRPV1 trafficking to the plasma membrane. J. Gen. Physiol. 2006;128:509–522. doi: 10.1085/jgp.200609576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Sugiura T, Tominaga M, Katsuya H, Mizumura K. Bradykinin lowers the threshold temperature for heat activation of vanilloid receptor 1. J. Neurophysiol. 2002;88:544–548. doi: 10.1152/jn.2002.88.1.544. [DOI] [PubMed] [Google Scholar]
  • 169.Sun SY, Wang W, Schultz HD. Activation of cardiac afferents by arachidonic acid: relative contributions of metabolic pathways. Am. J. Physiol. Heart Circ. Physiol. 2001;281:H93–H104. doi: 10.1152/ajpheart.2001.281.1.H93. [DOI] [PubMed] [Google Scholar]
  • 170.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. U.S.A. 2001;98:711–716. doi: 10.1073/pnas.011404498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Szallasi A, Blumberg PM. Vanilloid receptors new insights enhance potential as a therapeutic target. Pain. 1996;68:195–208. doi: 10.1016/s0304-3959(96)03202-2. [DOI] [PubMed] [Google Scholar]
  • 172.Szallasi A, Blumberg PM. Vanilloid (Capsaicin) receptors and mechanisms. Pharmacol. Rev. 1999;51:159–212. [PubMed] [Google Scholar]
  • 173.Szallasi A, Cortright DN, Blum CA, Eid SR. The vanilloid receptor TRPV1: 10 years from channel cloning to antagonist proof-of-concept. Nat. Rev. Drug Discov. 2007;6:357–372. doi: 10.1038/nrd2280. [DOI] [PubMed] [Google Scholar]
  • 174.Szallasi A, Blumberg PM, Lundberg JM. Proton inhibition of [3H]resiniferatoxin binding to vanilloid (capsaicin) receptors in rat spinal cord. Eur. J. Pharmacol. 1995;289:181–187. doi: 10.1016/0922-4106(95)90093-4. [DOI] [PubMed] [Google Scholar]
  • 175.Szallasi A, Conte B, Goso C, Blumberg PM, Manzini S. Characterization of a peripheral vanilloid (capsaicin) receptor in the urinary bladder of the rat. Life Sci. 1993;52:L221–L226. doi: 10.1016/0024-3205(93)90051-4. [DOI] [PubMed] [Google Scholar]
  • 176.Szabo T, Olah Z, Iadarola MJ, Blumberg PM. Epidural resiniferatoxin induced prolonged regional analgesia to pain. Brain Res. 1999;840:92–98. doi: 10.1016/s0006-8993(99)01763-1. [DOI] [PubMed] [Google Scholar]
  • 177.Szekely M, Szolcsanyi J. Endotoxin fever in capsaicin treated rats. Acta Physiol. Acad. Sci. Hung. 1979;53:469–477. [PubMed] [Google Scholar]
  • 178.Szelenyi Z, Hummel Z, Szolcsanyi J, Davis JB. Daily body temperature rhythm and heat tolerance in TRPV1 knockout and capsaicin pretreated mice. Eur. J. Neurosci. 2004;19:1421–1424. doi: 10.1111/j.1460-9568.2004.03221.x. [DOI] [PubMed] [Google Scholar]
  • 179.Taiwo YO, Bjerknes LK, Goetzl EJ, Levine JD. Mediation of primary afferent peripheral hyperalgesia by the cAMP second messenger system. Neuroscienc. 1989;32:577–580. doi: 10.1016/0306-4522(89)90280-7. [DOI] [PubMed] [Google Scholar]
  • 180.Taiwo YO, Levine JD. Further confirmation of the role of adenyl cyclase and of cAMP-dependent protein kinase in primary afferent hyperalgesia. Neuroscience. 1991;44:131–135. doi: 10.1016/0306-4522(91)90255-m. [DOI] [PubMed] [Google Scholar]
  • 181.Takeuchi K, Yagi K, Kato S, Ukawa H. Roles of prostaglandin E-receptor subtypes in gastric and duodenal bicarbonate secretion in rats. Gastroenterology. 1997;113:1553–1559. doi: 10.1053/gast.1997.v113.pm9352857. [DOI] [PubMed] [Google Scholar]
  • 182.Tashima K, Nakashima M, Kagawa S, Kato S, Takeuchi K. Gastric hyperemic response induced by acid back-diffusion in rat stomachs following barrier disruption relation to vanilloid type-1 receptors. Med. Sci. Monit. 2002;8:157–163. [PubMed] [Google Scholar]
  • 183.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. doi: 10.1016/s0896-6273(00)80564-4. [DOI] [PubMed] [Google Scholar]
  • 184.Tominaga M, Wada M, Masu M. Potentiation of capsaicin receptor activity by metabotropic ATP receptors as a possible mechanism for ATP-evoked pain and hyperalgesia. Proc. Natl. Acad. Sci. U.S.A. 2001;98:6951–6956. doi: 10.1073/pnas.111025298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Toth A, Boczan J, Kedei N, Lizanecz E, Bagi Z, Papp Z, Edes I, Csiba L, Blumberg PM. Expression and distribution of vanilloid receptor 1 (TRPV1) in the adult rat brain. Brain Res. Mol. Brain Res. 2005;135:162–168. doi: 10.1016/j.molbrainres.2004.12.003. [DOI] [PubMed] [Google Scholar]
  • 186.Toth A, Kedei N, Szabo T, Wang Y, Blumberg PM. Thapsigargin binds to and inhibits the cloned vanilloid receptor-1. Biochem. Biophys. Res. Commun. 2002;293:777–782. doi: 10.1016/S0006-291X(02)00293-0. [DOI] [PubMed] [Google Scholar]
  • 187.Trevisani M, Gazzieri D, Benvenuti F, Campi B, Dinh QT, Groneberg DA, Rigoni M, Emonds-Alt X, Creminon C, Fischer A, Geppetti P, Harrison S. Ethanol causes inflammation in the airways by a neurogenic and TRPV1-dependent mechanism. J. Pharmacol. Exp. Ther. 2004;309:1167–1173. doi: 10.1124/jpet.103.064162. [DOI] [PubMed] [Google Scholar]
  • 188.Urban L, Dray A. Capsazepine, a novel capsaicin antagonist, selectively antagonises the effects of capsaicin in the mouse spinal cord in vitro. Neurosci. Lett. 1991;134:9–11. doi: 10.1016/0304-3940(91)90496-g. [DOI] [PubMed] [Google Scholar]
  • 189.Ustinova EE, Bergren D, Schultz HD. Neuropeptide depletion impairs postischemic recovery of the isolated rat heart: role of substance P. Cardiovasc. Res. 1995;30:55–63. [PubMed] [Google Scholar]
  • 190.Valenzano KJ, Grant ER, Wu G, Hachicha M, Schmid L, Tafesse L, Sun Q, Rotshteyn Y, Francis J, Limberis J, Malik S, Whittemore ER, Hodges D. N-(4-tertiarybutylphenyl)-4-(3-chloropyridin-2-yl)tetrahydropyrazine -1(2H)-carbox-amide (BCTC), a novel, orally effective vanilloid receptor 1 antagonist with analgesic properties: I in vitro characterization and pharmacokinetic properties. J. Pharmacol. Exp. Ther. 2003;306:377–386. doi: 10.1124/jpet.102.045674. [DOI] [PubMed] [Google Scholar]
  • 191.Van Buren JJ, Bhat S, Rotello R, Pauza ME, Premkumar LS. Sensitization and translocation of TRPV1 by insulin and IGF-I. Mol. Pain. 2005;1:17. doi: 10.1186/1744-8069-1-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Vass Z, Brechtelsbauer PB, Nuttall AL, Miller JM. Nitric oxide mediates capsaicin-induced increase in cochlear blood flow. Hear Res. 1996;100:114–119. doi: 10.1016/0378-5955(96)00102-5. [DOI] [PubMed] [Google Scholar]
  • 193.Vass Z, Dai CF, Steyger PS, Jancso G, Trune DR, Nuttall AL. Co-localization of the vanilloid capsaicin receptor and substance P in sensory nerve fibers innervating cochlear and vertebro-basilar arteries. Neuroscience. 2004;124:919–927. doi: 10.1016/j.neuroscience.2003.12.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Vellani V, Mapplebeck S, Moriondo A, Davis JB, McNaughton PA. Protein kinase C activation potentiates gating of the vanilloid receptor VR1 by capsaicin, protons, heat and anandamide. J. Physiol. 2001;534:813–25. doi: 10.1111/j.1469-7793.2001.00813.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Veronesi B, Oortgiesen M, Carter JD, Devlin RB. Particulate matter initiates inflammatory cytokine release by activation of capsaicin and acid receptors in a human bronchial epithelial cell line. Toxicol. Appl. Pharmacol. 1999;154:106–115. doi: 10.1006/taap.1998.8567. [DOI] [PubMed] [Google Scholar]
  • 196.Wahl P, Foged C, Tullin S, Thomsen C. Iodo-resiniferatoxin, a new potent vanilloid receptor antagonist. Mol. Pharmacol. 2001;59:9–15. doi: 10.1124/mol.59.1.9. [DOI] [PubMed] [Google Scholar]
  • 197.Wang X, Miyares RL, Ahern GP. Oleoylethanolamide excites vagal sensory neurones, induces visceral pain and reduces short-term food intake in mice via capsaicin receptor TRPV1. J. Physiol. 2005;564:541–547. doi: 10.1113/jphysiol.2004.081844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Ward SM, Bayguinov J, Won KJ, Grundy D, Berthoud HR. Distribution of the vanilloid receptor (VR1) in the gastrointestinal tract. J. Comp. Neurol. 2003;465:121–35. doi: 10.1002/cne.10801. [DOI] [PubMed] [Google Scholar]
  • 199.Wardle KA, Ranson J, Sanger GJ. Pharmacological characterization of the vanilloid receptor in the rat dorsal spinal cord. Br. J. Pharmacol. 1997;121:1012–1016. doi: 10.1038/sj.bjp.0701199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Woolf CJ, Salter MW. Neuronal plasticity: increasing the gain in pain. Science. 2000;288:1765–1769. doi: 10.1126/science.288.5472.1765. [DOI] [PubMed] [Google Scholar]
  • 201.Yaksh TL, Farb DH, Leeman SE, Jessell TM. Intrathecal capsaicin depletes substance P in the rat spinal cord and produces prolonged thermal analgesia. Science. 1979;206:481–483. doi: 10.1126/science.228392. [DOI] [PubMed] [Google Scholar]
  • 202.Yiangou Y, Facer P, Dyer NH, Chan CL, Knowles C, Williams NS, Anand P. Vanilloid receptor 1 immunoreactivity in inflamed human bowel. Lancet. 2001;357:1338–1339. doi: 10.1016/s0140-6736(00)04503-7. [DOI] [PubMed] [Google Scholar]
  • 203.Zahner MR, Li DP, Chen SR, Pan HL. Cardiac vanilloid receptor 1-expressing afferent nerves and their role in the cardiogenic sympathetic reflex in rats. J. Physiol. 2003;551:515–523. doi: 10.1113/jphysiol.2003.048207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Zheng J, Dai C, Steyger PS, Kim Y, Vass Z, Ren T, Nuttall AL. Vanilloid receptors in hearing: altered cochlear sensitivity by vanilloids and expression of TRPV1 in the organ of corti. J. Neurophysiol. 2003;90:444–455. doi: 10.1152/jn.00919.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Zhuang ZY, Xu H, Clapham DE, Ji RR. Phosphatidylinositol 3-kinase activates ERK in primary sensory neurons and mediates inflammatory heat hyperalgesia through TRPV1 sensitization. J. Neurosci. 2004;24:8300–8309. doi: 10.1523/JNEUROSCI.2893-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Zygmunt PM, Petersson J, Andersson DA, Chuang H, Sorgard M, Di Marzo V, Julius D, Hogestatt ED. Vanilloid receptors on sensory nerves mediate the vasodilator action of anandamide. Nature. 1999;400:452–457. doi: 10.1038/22761. [DOI] [PubMed] [Google Scholar]

Articles from Current Neuropharmacology are provided here courtesy of Bentham Science Publishers

RESOURCES