Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 189: 108533, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33744339

ABSTRACT

Using neurokinin 1 receptor (NK1R) internalization to measure of substance P release in rat spinal cord slices, we found that it was induced by the adenylyl cyclase (AC) activator forskolin, by the protein kinase A (PKA) activators 6-Bnz-cAMP and 8-Br-cAMP, and by the activator of exchange protein activated by cAMP (Epac) 8-pCPT-2-O-Me-cAMP (CPTOMe-cAMP). Conversely, AC and PKA inhibitors decreased substance P release induced by electrical stimulation of the dorsal root. Therefore, the cAMP signaling pathway mediates substance P release in the dorsal horn. The effects of forskolin and 6-Bnz-cAMP were not additive with NMDA-induced substance P release and were decreased by the NMDA receptor blocker MK-801. In cultured dorsal horn neurons, forskolin increased NMDA-induced Ca2+ entry and the phosphorylation of the NR1 and NR2B subunits of the NMDA receptor. Therefore, cAMP-induced substance P release is mediated by the activating phosphorylation by PKA of NMDA receptors. Voltage-gated Ca2+ channels, but not by TRPV1 or TRPA1, also contributed to cAMP-induced substance P release. Activation of PKA was required for the effects of forskolin and the three cAMP analogs. Epac2 contributed to the effects of forskolin and CPTOMe-cAMP, signaling through a Raf - mitogen-activated protein kinase pathway to activate Ca2+ channels. Epac1 inhibitors induced NK1R internalization independently of substance P release. In rats with latent sensitization to pain, the effect of 6-Bnz-cAMP was unchanged, whereas the effect of forskolin was decreased due to the loss of the stimulatory effect of Epac2. Hence, substance P release induced by cAMP decreases during pain hypersensitivity.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction/physiology , Spinal Cord/metabolism , Substance P/metabolism , Animals , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/agonists , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Guanine Nucleotide Exchange Factors/agonists , Hyperalgesia/metabolism , Male , Organ Culture Techniques , Posterior Horn Cells/drug effects , Posterior Horn Cells/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Spinal Cord/drug effects
2.
Neuropharmacology ; 128: 255-268, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29042318

ABSTRACT

The interaction between NMDA receptors and µ-opioid receptors in primary afferent terminals was studied by using NMDA to induce substance P release, measured as neurokinin 1 receptor internalization. In rat spinal cord slices, the µ-opioid receptor agonists morphine, DAMGO and endomorphin-2 inhibited NMDA-induced substance P release, whereas the antagonist CTAP right-shifted the concentration response of DAMGO. In vivo, substance P release induced by intrathecal NMDA after priming with BDNF was inhibited by DAMGO. ω-Conotoxins MVIIC and GVIA inhibited about half of the NMDA-induced substance P release, showing that it was partially mediated by the opening of voltage-gated calcium (Cav) channels. In contrast, DAMGO or ω-conotoxins did not inhibit capsaicin-induced substance P release. In cultured DRG neurons, DAMGO but not ω-conotoxin inhibited NMDA-induced increases in intracellular calcium, indicating that µ-opioid receptors can inhibit NMDA receptor function by mechanisms other than inactivation of Cav channels. Moreover, DAMGO decreased the ω-conotoxin-insensitive component of the substance P release. Potent inhibition by ifenprodil showed that these NMDA receptors have the NR2B subunit. Activators of adenylyl cyclase and protein kinase A (PKA) induced substance P release and this was decreased by the NMDA receptor blocker MK-801 and by DAMGO. Conversely, inhibitors of adenylyl cyclase and PKA, but not of protein kinase C, decreased NMDA-induced substance P release. Hence, these NMDA receptors are positively modulated by the adenylyl cyclase-PKA pathway, which is inhibited by µ-opioid receptors. In conclusion, µ-opioid receptors inhibit NMDA receptor-induced substance P release through Cav channel inactivation and adenylyl cyclase inhibition.


Subject(s)
Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Opioid, mu/metabolism , Spinal Cord/metabolism , Substance P/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Analgesics, Opioid/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Cells, Cultured , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agents/pharmacology , Male , Morphine/pharmacology , Narcotic Antagonists/pharmacology , Neurons/drug effects , Neurons/metabolism , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/metabolism , Spinal Cord/cytology , Spinal Cord/drug effects , omega-Conotoxins/pharmacology
3.
J Neurosci ; 36(1): 204-21, 2016 Jan 06.
Article in English | MEDLINE | ID: mdl-26740662

ABSTRACT

Many chronic pain disorders alternate between bouts of pain and periods of remission. The latent sensitization model reproduces this in rodents by showing that the apparent recovery ("remission") from inflammatory or neuropathic pain can be reversed by opioid antagonists. Therefore, this remission represents an opioid receptor-mediated suppression of a sustained hyperalgesic state. To identify the receptors involved, we induced latent sensitization in mice and rats by injecting complete Freund's adjuvant (CFA) in the hindpaw. In WT mice, responses to mechanical stimulation returned to baseline 3 weeks after CFA. In µ-opioid receptor (MOR) knock-out (KO) mice, responses did not return to baseline but partially recovered from peak hyperalgesia. Antagonists of α2A-adrenergic and δ-opioid receptors reinstated hyperalgesia in WT mice and abolished the partial recovery from hyperalgesia in MOR KO mice. In rats, antagonists of α2A adrenergic and µ-, δ-, and κ-opioid receptors reinstated hyperalgesia during remission from CFA-induced hyperalgesia. Therefore, these four receptors suppress hyperalgesia in latent sensitization. We further demonstrated that suppression of hyperalgesia by MORs was due to their constitutive activity because of the following: (1) CFA-induced hyperalgesia was reinstated by the MOR inverse agonist naltrexone (NTX), but not by its neutral antagonist 6ß-naltrexol; (2) pro-enkephalin, pro-opiomelanocortin, and pro-dynorphin KO mice showed recovery from hyperalgesia and reinstatement by NTX; (3) there was no MOR internalization during remission; (4) MORs immunoprecipitated from the spinal cord during remission had increased Ser(375) phosphorylation; and (5) electrophysiology recordings from dorsal root ganglion neurons collected during remission showed constitutive MOR inhibition of calcium channels. SIGNIFICANCE STATEMENT: Chronic pain causes extreme suffering to millions of people, but its mechanisms remain to be unraveled. Latent sensitization is a phenomenon studied in rodents that has many key features of chronic pain: it is initiated by a variety of noxious stimuli, has indefinite duration, and pain appears in episodes that can be triggered by stress. Here, we show that, during latent sensitization, there is a sustained state of pain hypersensitivity that is continuously suppressed by the activation of µ-, δ-, and κ-opioid receptors and by adrenergic α2A receptors in the spinal cord. Furthermore, we show that the activation of µ-opioid receptors is not due to the release of endogenous opioids, but rather to its ligand-independent constitutive activity.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/administration & dosage , Hyperalgesia/prevention & control , Hyperalgesia/physiopathology , Narcotic Antagonists/administration & dosage , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Opioid/metabolism , Animals , Freund's Adjuvant , Hyperalgesia/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pain Measurement/drug effects , Rats , Rats, Sprague-Dawley , Species Specificity , Treatment Outcome
4.
Eur J Neurosci ; 39(9): 1439-54, 2014 May.
Article in English | MEDLINE | ID: mdl-24611998

ABSTRACT

NMDA receptors in primary afferent terminals can contribute to hyperalgesia by increasing neurotransmitter release. In rats and mice, we found that the ability of intrathecal NMDA to induce neurokinin 1 receptor (NK1R) internalization (a measure of substance P release) required a previous injection of BDNF. Selective knock-down of NMDA receptors in primary afferents decreased NMDA-induced NK1R internalization, confirming the presynaptic location of these receptors. The effect of BDNF was mediated by tropomyosin-related kinase B (trkB) receptors and not p75 neurotrophin receptors (p75(NTR) ), because it was not produced by proBDNF and was inhibited by the trkB antagonist ANA-12 but not by the p75(NTR) inhibitor TAT-Pep5. These effects are probably mediated through the truncated form of the trkB receptor as there is little expression of full-length trkB in dorsal root ganglion (DRG) neurons. Src family kinase inhibitors blocked the effect of BDNF, suggesting that trkB receptors promote the activation of these NMDA receptors by Src family kinase phosphorylation. Western blots of cultured DRG neurons revealed that BDNF increased Tyr(1472) phosphorylation of the NR2B subunit of the NMDA receptor, known to have a potentiating effect. Patch-clamp recordings showed that BDNF, but not proBDNF, increased NMDA receptor currents in cultured DRG neurons. NMDA-induced NK1R internalization was also enabled in a neuropathic pain model or by activating dorsal horn microglia with lipopolysaccharide. These effects were decreased by a BDNF scavenger, a trkB receptor antagonist and a Src family kinase inhibitor, indicating that BDNF released by microglia potentiates NMDA receptors in primary afferents during neuropathic pain.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Ganglia, Spinal/metabolism , Neuralgia/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Ganglia, Spinal/drug effects , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptor, trkB/metabolism , Receptors, Neurokinin-1/metabolism , Signal Transduction/drug effects , Substance P/metabolism
5.
Neuroimmunomodulation ; 19(6): 367-76, 2012.
Article in English | MEDLINE | ID: mdl-23051934

ABSTRACT

BACKGROUND/AIMS: Spinal glia activation has been proposed as one mechanism underlying visceral hyperalgesia in a rodent model of chronic stress. In order to assess the possible role of changes in circulating cytokines and in blood-spinal cord barrier (BSCB) permeability in spinal glia activation, we studied the time course of peripheral and spinal pro-inflammatory cytokines and of spinal and satellite glia markers in response to repeated water avoidance (WA) stress. METHODS: Spinal cords and dorsal root ganglion cells (DRGs) were collected from control rats, rats exposed to 1-hour WA, or 1-hour WA daily for 5 days or 1-hour WA daily for 10 days. RESULTS: We demonstrated a time-dependent change in circulating IL-1ß and spinal IL-1ß, IL-6 and TNF-α in stressed animals compared with controls. We found altered expression of the astrocyte markers GFAP and Connexin 43 in spinal and DRG samples at different time points. Finally, WA was associated with increased BSCB permeability. CONCLUSIONS: These findings confirm the concept that both peripheral and spinal immune markers are altered after chronic WA and suggest a possible link between stress-induced increase of peripheral pro-inflammatory cytokines, changes in satellite glial cells, increase in BSCB permeability and increase in spinal pro-inflammatory mediators suggesting glia activation.


Subject(s)
Cytokines/biosynthesis , Inflammation Mediators/metabolism , Neuroglia/immunology , Neuroglia/pathology , Reaction Time/immunology , Spinal Cord/immunology , Spinal Cord/metabolism , Stress, Psychological/immunology , Animals , Avoidance Learning/physiology , Biomarkers/metabolism , Chronic Disease , Inflammation Mediators/physiology , Male , Neuroglia/metabolism , Rats , Rats, Wistar , Stress, Psychological/psychology
6.
Am J Physiol Gastrointest Liver Physiol ; 301(3): G580-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21719739

ABSTRACT

Glutamate (Glu) is the primary excitatory neurotransmitter in the central nervous system and plays a critical role in the neuroplasticity of nociceptive networks. We aimed to examine the role of spinal astroglia in the modulation of glutamatergic neurotransmission in a model of chronic psychological stress-induced visceral hyperalgesia in male Wistar rats. We assessed the effect of chronic stress on different glial Glu control mechanisms in the spinal cord including N-methyl-d-aspartate receptors (NMDARs), glial Glu transporters (GLT1 and GLAST), the Glu conversion enzyme glutamine synthetase (GS), and glial fibrillary acidic protein (GFAP). We also tested the effect of pharmacological inhibition of NMDAR activation, of extracellular Glu reuptake, and of astrocyte function on visceral nociceptive response in naive and stressed rats. We observed stress-induced decreased expression of spinal GLT1, GFAP, and GS, whereas GLAST expression was upregulated. Although visceral hyperalgesia was blocked by pharmacological inhibition of spinal NMDARs, we observed no stress effects on NMDAR subunit expression or phosphorylation. The glial modulating agent propentofylline blocked stress-induced visceral hyperalgesia, and blockade of GLT1 function in control rats resulted in enhanced visceral nociceptive response. These findings provide evidence for stress-induced modulation of glia-controlled spinal Glu-ergic neurotransmission and its involvement in chronic stress-induced visceral hyperalgesia. The findings reported in this study demonstrate a unique pattern of stress-induced changes in spinal Glu signaling and metabolism associated with enhanced responses to visceral distension.


Subject(s)
Astrocytes/physiology , Glutamic Acid/physiology , Hyperalgesia/physiopathology , Spinal Cord/physiopathology , Stress, Psychological/physiopathology , Synaptic Transmission/drug effects , Amino Acid Transport System X-AG/metabolism , Animals , Dizocilpine Maleate/pharmacology , Glial Fibrillary Acidic Protein/biosynthesis , Glutamate-Ammonia Ligase/biosynthesis , Male , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Xanthines/pharmacology
7.
J Neurogenet ; 22(4): 315-31, 2008.
Article in English | MEDLINE | ID: mdl-19085272

ABSTRACT

Separate breeding colonies of C57BL/6 ("B6") mice maintained at the Jackson Laboratories ("J") and NIH ("N") have led to the emergence of two distinct substrains of C57BL/6 mice: C57BL/6J and C57BL/6N. Molecular genetic studies indicate simple sequence-length polymorphisms, single-nucleotide polymorphisms, and copy-number variants among B6 substrains that may contribute to phenotypic differences. We examined differences in motor coordination, pain sensitivity, and conditional fear in the C57BL/6J strain and three N strains: C57BL/6NCrl (Charles River), C57BL/6NTac (Taconic), and C57BL/6NHsd (Harlan Sprague Dawley). Male C57BL/6J mice demonstrated enhanced motor coordination, as measured by the rotarod assay, markedly enhanced pain sensitivity in two assays of acute thermal nociception (e.g., tail withdrawal and hot plate), and a reduced level of conditional fear. The tail withdrawal result was confirmed in a separate laboratory. We also provide a table reviewing previously reported behavioral differences among various B6 substrains and discuss the significance of environmental differences due to obtaining mice form different vendors. These data may be seen as a potential problem and as a potential opportunity. Great care must be taken when working with mice engineered by using B6 embryonic stem cell lines because control groups, backcrosses, and intercrosses could inadvertently introduce behaviorally significant polymorphic alleles or environmental confounds. On the other hand, deliberate crosses between B6 substrains may provide an opportunity to map polymorphic loci that contribute to variability in a trait on largely homogenous backgrounds, which has the potential to improve mapping resolution and aid in the selection of candidate genes.


Subject(s)
Animals, Genetically Modified/psychology , Behavior, Animal/physiology , Mice, Inbred C57BL/psychology , Models, Animal , Animals , Crosses, Genetic , Fear , Genotype , Male , Mice , Mice, Inbred C57BL/genetics , Mice, Knockout , Mice, Transgenic , Motor Activity/genetics , Pain Threshold , Phenotype
8.
Gastroenterology ; 130(6): 1729-42, 2006 May.
Article in English | MEDLINE | ID: mdl-16697737

ABSTRACT

BACKGROUND & AIMS: The neurokinin 1 receptors (NK(1)Rs) and substance P (SP) have been implicated in the stress and/or pain pathways involved in chronic pain conditions. Here we examined the participation of NK(1)Rs in sustained visceral hyperalgesia observed in rats exposed to chronic psychological stress. METHODS: Male Wistar rats were exposed to daily 1-hour water avoidance stress (WA) or sham WA for 10 consecutive days. We tested intraperitoneal or intrathecal injection of the NK(1)R antagonist SR140333 on the visceromotor reflex to colorectal distention in both groups at day 11. Real-time reverse-transcription polymerase chain reaction, Western blot, and immunohistochemistry were used to assess the expression of NK(1)Rs and/or SP in samples of colon, spinal cord, and dorsal root ganglia. RESULTS: Both intraperitoneal and intrathecal SR140333 injection diminished the enhanced visceromotor reflex to colorectal distention at day 11 in stressed rats but did not affect the response in control animals. Real-time polymerase chain reaction and Western blotting demonstrated stress-induced up-regulation of spinal NK(1)Rs. Immunohistochemistry showed an increased number of NK(1)R-expressing neurons in the laminae I of the dorsal horn in stressed rats. The expression of NK(1)Rs was decreased in colon from stressed rats compared with control. The expression of SP gene precursor in dorsal root ganglia was unchanged in stressed rats compared with controls. CONCLUSIONS: Stress-induced increased NK(1)R expression on spinal neurons and the inhibitory effect of intrathecal NK(1)R antagonist on visceral hyperalgesia support the key contribution of spinal NK(1)Rs in the molecular pathways involved in the maintenance of visceral hyperalgesia observed after chronic WA.


Subject(s)
Hyperalgesia/physiopathology , Piperidines/pharmacology , Quinuclidines/pharmacology , Receptors, Neurokinin-1/metabolism , Animals , Base Sequence , Blotting, Western , Disease Models, Animal , Electrodes, Implanted , Electromyography , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Gene Expression Regulation , Male , Molecular Sequence Data , RNA, Messenger/analysis , Rats , Rats, Wistar , Receptors, Neurokinin-1/drug effects , Reference Values , Reverse Transcriptase Polymerase Chain Reaction/methods , Sensitivity and Specificity , Stress, Physiological
9.
Am J Physiol Gastrointest Liver Physiol ; 291(2): G219-28, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16565418

ABSTRACT

N-methyl-D-aspartate (NMDA) receptors (NMDARs) on spinal afferent neurons regulate the peripheral and central release of neuropeptides involved in the development of hyperalgesia. We examined the effect of experimental colitis on the molecular and functional properties of NMDARs on these neurons. Lumbosacral dorsal root ganglia (DRG) were collected from adult rats 5 days after the induction of colitis for whole cell patch-clamp recording, Western blot analysis, and quantitative RT-PCR. Compared with neurons from control rats, those taken from animals with colitis had a threefold higher density of NMDA currents in both retrograde-labeled, colon-specific, and unlabeled DRG neurons. Increased current densities were not observed in DRG neurons taken from thoracic spinal levels. There was no significant change in NMDA or glycine affinity or in voltage-dependent Mg2+ inhibition; however, there was a 10-fold decrease in sensitivity to the NR2B subunit-selective antagonist ifenprodil. Quantitative RT-PCR and Western blot analysis indicated a 28% increase in the expression of NR2B with little or no change in the other three NR2 subunits. The addition of the Src family tyrosine kinase inhibitor PP2 (10 microM) decreased NMDAR currents in neurons from colitis but not control rats. Conversely, pretreatment of DRG neurons from control animals with 100 microM sodium orthovanadate increased NMDAR currents and decreased ifenprodil sensitivity to levels similar to those observed in neurons from animals with colitis. In conclusion, colonic inflammation upregulates the activity of NMDARs in all DRG neurons within ganglia innervating this tissue through mechanisms involving increased expression and persistent tyrosine phosphorylation.


Subject(s)
Colitis/metabolism , Disease Models, Animal , Ganglia, Spinal/metabolism , Posterior Horn Cells/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cells, Cultured , Male , Rats , Rats, Sprague-Dawley , Tissue Distribution
10.
Am J Physiol Gastrointest Liver Physiol ; 289(1): G42-53, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15746211

ABSTRACT

Chronic stress plays an important role in the development and exacerbation of symptoms in functional gastrointestinal disorders. To better understand the mechanisms underlying this relationship, we aimed to characterize changes in visceral and somatic nociception, colonic motility, anxiety-related behavior, and mucosal immune activation in rats exposed to 10 days of chronic psychological stress. Male Wistar rats were submitted daily to either 1-h water avoidance (WA) stress or sham WA for 10 consecutive days. The visceromotor response to colorectal distension, thermal somatic nociception, and behavioral responses to an open field test were measured at baseline and after chronic WA. Fecal pellets were counted after each WA stress or sham WA session as a measure of stress-induced colonic motility. Colonic samples were collected from both groups and evaluated for structural changes and neutrophil infiltration, mast cell number by immunohistochemistry, and cytokine expression by quantitative RT-PCR. Rats exposed to chronic WA (but not sham stress) developed persistent visceral hyperalgesia, whereas only transient changes in somatic nociception were observed. Chronically stressed rats also exhibited anxiety-like behaviors, enhanced fecal pellet excretion, and small but significant increases in the mast cell numbers and the expression of IL-1beta and IFN-gamma. Visceral hyperalgesia following chronic stress persisted for at least a month. Chronic psychological stress in rats results in a robust and long-lasting alteration of visceral, but not somatic nociception. Visceral hyperalgesia is associated with other behavioral manifestations of stress sensitization but was only associated with minor colonic immune activation arguing against a primary role of mucosal immune activation in the maintenance of this phenomenon.


Subject(s)
Colitis/physiopathology , Hyperalgesia/physiopathology , Stress, Psychological/physiopathology , Animals , Anxiety/immunology , Anxiety/physiopathology , Avoidance Learning , Chronic Disease , Colitis/immunology , Colitis/pathology , Colon/immunology , Colon/pathology , Colon/physiopathology , Cytokines/genetics , Disease Models, Animal , Exploratory Behavior , Feces , Gastrointestinal Motility , Hyperalgesia/immunology , Male , Nociceptors/physiology , RNA, Messenger/analysis , Rats , Rats, Wistar , Stress, Psychological/immunology , Water
11.
J Neurosci ; 24(18): 4300-12, 2004 May 05.
Article in English | MEDLINE | ID: mdl-15128844

ABSTRACT

Inflammatory proteases (mast cell tryptase and trypsins) cleave protease-activated receptor 2 (PAR2) on spinal afferent neurons and cause persistent inflammation and hyperalgesia by unknown mechanisms. We determined whether transient receptor potential vanilloid receptor 1 (TRPV1), a cation channel activated by capsaicin, protons, and noxious heat, mediates PAR2-induced hyperalgesia. PAR2 was coexpressed with TRPV1 in small- to medium-diameter neurons of the dorsal root ganglia (DRG), as determined by immunofluorescence. PAR2 agonists increased intracellular [Ca2+] ([Ca2+]i) in these neurons in culture, and PAR2-responsive neurons also responded to the TRPV1 agonist capsaicin, confirming coexpression of PAR2 and TRPV1. PAR2 agonists potentiated capsaicin-induced increases in [Ca2+]i in TRPV1-transfected human embryonic kidney (HEK) cells and DRG neurons and potentiated capsaicin-induced currents in DRG neurons. Inhibitors of phospholipase C and protein kinase C (PKC) suppressed PAR2-induced sensitization of TRPV1-mediated changes in [Ca2+]i and TRPV1 currents. Activation of PAR2 or PKC induced phosphorylation of TRPV1 in HEK cells, suggesting a direct regulation of the channel. Intraplantar injection of a PAR2 agonist caused persistent thermal hyperalgesia that was prevented by antagonism or deletion of TRPV1. Coinjection of nonhyperalgesic doses of PAR2 agonist and capsaicin induced hyperalgesia that was inhibited by deletion of TRPV1 or antagonism of PKC. PAR2 activation also potentiated capsaicin-induced release of substance P and calcitonin gene-related peptide from superfused segments of the dorsal horn of the spinal cord, where they mediate hyperalgesia. We have identified a novel mechanism by which proteases that activate PAR2 sensitize TRPV1 through PKC. Antagonism of PAR2, TRPV1, or PKC may abrogate protease-induced thermal hyperalgesia.


Subject(s)
Hyperalgesia/metabolism , Receptor, PAR-2/metabolism , Receptors, Drug/metabolism , Animals , Calcitonin Gene-Related Peptide/metabolism , Calcium/metabolism , Capsaicin/pharmacology , Cells, Cultured , Enzyme Activators/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Hyperalgesia/chemically induced , Kidney/cytology , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, PAR-2/agonists , Receptor, PAR-2/genetics , Receptors, Drug/agonists , Receptors, Drug/genetics , Spinal Cord/drug effects , Spinal Cord/metabolism , Substance P/metabolism , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism
12.
Pain ; 109(3): 443-452, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15157705

ABSTRACT

In the peripheral nervous system, N-methyl-D-aspartate receptors (NMDAR) expressed on the central and peripheral terminals of primary afferent neurons are involved in nociception. We used single cell imaging of intracellular calcium concentration ([Ca2+]i) and patch clamp techniques to characterize the functional properties of NMDARs on adult rat dorsal root ganglia (DRG) neurons in primary culture and selectively on those innervating the distal colon. In Mg2+-free extracellular solution, rapid perfusion of DRG neurons with 250 microM NMDA and 10 microM glycine caused a significant increase in [Ca2+]i, and elicited inward currents in whole cell patch clamp recordings when the holding potential was -60 mV. Both effects were reversibly inhibited by 200 microM ketamine in a use-dependent manner. The EC50 values for NMDA and glycine were 64 and 1.9 microM with Hill slope coefficients of 1.4 and 1.3, respectively. At negative potentials, extracellular Mg2+ blocked currents in a concentration- and voltage-dependent manner. The IC50 for Mg2+ at a holding potential of -100 mV was 2.0 microM. The NMDAR subtype-selective antagonist, ifenprodil, inhibited 94% of the NMDA and glycine-induced current with an IC50 of 2.6 microM. There was no evidence of multiple binding sites for ifenprodil. There was no significant difference in the NMDAR current density on DRG neurons that had innervated the colon, nor was there a difference in the EC50 for ifenprodil. These results demonstrate that functional NMDARs expressed by DRG neurons innervating both somatic and visceral tissues of adult rats are composed predominantly of NR2B subunits.


Subject(s)
Ganglia, Spinal/metabolism , Neurons, Afferent/metabolism , Nociceptors/metabolism , Pain/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cells, Cultured , Colon/innervation , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Glycine/pharmacology , Magnesium/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , N-Methylaspartate/pharmacology , Neurons, Afferent/cytology , Neurons, Afferent/drug effects , Nociceptors/cytology , Nociceptors/drug effects , Pain/physiopathology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/drug effects , Visceral Afferents/cytology , Visceral Afferents/drug effects , Visceral Afferents/metabolism
13.
J Comp Neurol ; 446(4): 325-41, 2002 May 13.
Article in English | MEDLINE | ID: mdl-11954032

ABSTRACT

N-methyl-D-aspartate (NMDA) receptors in sensory afferents participate in chronic pain by mediating peripheral and central sensitization. We studied the presence of NMDA receptor subunits in different types of primary afferents. Western blots indicated that rat dorsal root ganglia (DRG) contain NR1, NR2B, NR2C, and NR2D but not NR2A. Real-time RT-PCR showed that NR2B and NR2D were expressed at higher levels than NR2A and NR2C in DRG. Immunofluorescence with an antibody that recognized NR1 and another that recognized NR2A and NR2B showed that NR1 and NR2B colocalized in 90% of DRG neurons, including most A-fibers (identified by the presence of neurofilament 200 kDa). In contrast, an antibody recognizing NR2C and NR2D labeled only neurofilament-negative DRG profiles. This antibody stained practically all DRG cells that contained calcitonin gene-related peptide and neurokinins and those that bound isolectin B4. The percentage of cells immunoreactive for NR1, NR2A/NR2B, and NR2C/NR2D were the same in the T9, T12, L4, and L6 DRG. The intracellular distribution of the NR2 subunits was strikingly different: Whereas NR2A/NR2B immunoreactivity was found in the Golgi apparatus and occasionally at the plasma membrane, NR2C/NR2D immunoreactivity was found in the cytoplasm but not in the Golgi. The NR1 subunit was present throughout the cytoplasm and was more intense in the Golgi. These findings indicate that DRG neurons have two different NMDA receptors, one containing the NR1, NR2D, and possibly the NR2C subunits, found only in C-fibers, and the diheteromer NR1/NR2B, present in the Golgi apparatus of both A- and C-fibers.


Subject(s)
Cell Compartmentation/physiology , Ganglia, Spinal/metabolism , Nerve Fibers, Myelinated/metabolism , Nerve Fibers/metabolism , Neurons, Afferent/metabolism , Rats, Sprague-Dawley/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Antibody Specificity/immunology , Calcitonin Gene-Related Peptide/metabolism , Chronic Disease , Ganglia, Spinal/cytology , Lectins/metabolism , Male , Nerve Fibers/ultrastructure , Nerve Fibers, Myelinated/ultrastructure , Neurofilament Proteins/metabolism , Neurons, Afferent/cytology , Organelles/metabolism , Organelles/ultrastructure , Pain/metabolism , Pain/physiopathology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley/anatomy & histology , Receptors, N-Methyl-D-Aspartate/genetics , Tachykinins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...