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










Publication year range
1.
J Med Chem ; 66(2): 1583-1600, 2023 01 26.
Article in English | MEDLINE | ID: mdl-36622903

ABSTRACT

Transient receptor potential ankyrin 1 (TRPA1) is a voltage-dependent, ligand-gated ion channel, and activation thereof is linked to a variety of painful conditions. Preclinical studies have demonstrated the role of TRPA1 receptors in a broad range of animal models of acute, inflammatory, and neuropathic pain. In addition, a clinical study using the TRPA1 antagonist GRC-17536 (Glenmark Pharmaceuticals) demonstrated efficacy in a subgroup of patients with painful diabetic neuropathy. Consequently, there is an increasing interest in TRPA1 inhibitors as potential analgesics. Herein, we report the identification of a fragment-like hit from a high-throughput screening (HTS) campaign and subsequent optimization to provide a novel and brain-penetrant TRPA1 inhibitor (compound 18, BAY-390), which is now being made available to the research community as an open-source in vivo probe.


Subject(s)
Neuralgia , Transient Receptor Potential Channels , Animals , Analgesics/pharmacology , Ankyrins , TRPA1 Cation Channel
2.
Sci Rep ; 11(1): 19877, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34615939

ABSTRACT

ATP-dependent P2X3 receptors play a crucial role in the sensitization of nerve fibers and pathological pain pathways. They are also involved in pathways triggering cough and may contribute to the pathophysiology of endometriosis and overactive bladder. However, despite the strong therapeutic rationale for targeting P2X3 receptors, preliminary antagonists have been hampered by off-target effects, including severe taste disturbances associated with blocking the P2X2/3 receptor heterotrimer. Here we present a P2X3 receptor antagonist, eliapixant (BAY 1817080), which is both highly potent and selective for P2X3 over other P2X subtypes in vitro, including P2X2/3. We show that eliapixant reduces inflammatory pain in relevant animal models. We also provide the first in vivo experimental evidence that P2X3 antagonism reduces neurogenic inflammation, a phenomenon hypothesised to contribute to several diseases, including endometriosis. To test whether eliapixant could help treat endometriosis, we confirmed P2X3 expression on nerve fibers innervating human endometriotic lesions. We then demonstrate that eliapixant reduces vaginal hyperalgesia in an animal model of endometriosis-associated dyspareunia, even beyond treatment cessation. Our findings indicate that P2X3 antagonism could alleviate pain, including non-menstrual pelvic pain, and modify the underlying disease pathophysiology in women with endometriosis. Eliapixant is currently under clinical development for the treatment of disorders associated with hypersensitive nerve fibers.


Subject(s)
Nerve Fibers/drug effects , Nerve Fibers/metabolism , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X3/metabolism , Somatosensory Disorders/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Disease Models, Animal , Female , Gene Expression , Humans , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Membrane Potentials/drug effects , Mice , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Rats , Receptors, Purinergic P2X3/genetics , Somatosensory Disorders/drug therapy , Somatosensory Disorders/etiology
3.
Bioorg Med Chem Lett ; 29(18): 2700-2705, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31362919

ABSTRACT

Here we report on novel and potent pyridyl-cycloalkyl-carboxylic acid inhibitors of microsomal prostaglandin E synthase-1 (PTGES). PTGES produces, as part of the prostaglandin pathway, prostaglandin E2 which is a well-known driver for pain and inflammation. This fact together with the observed upregulation of PTGES during inflammation suggests that blockade of the enzyme might provide a beneficial treatment option for inflammation related conditions such as endometriosis. Compound 5a, a close analogue of the screening hit, potently inhibited PTGES in vitro, displayed excellent PK properties in vitro and in vivo and demonstrated efficacy in a CFA-induced pain model in mice and in a rat dyspareunia endometriosis model and was therefore selected for further studies.


Subject(s)
Carboxylic Acids/pharmacology , Drug Discovery , Endometriosis/drug therapy , Enzyme Inhibitors/pharmacology , Prostaglandin-E Synthases/antagonists & inhibitors , Animals , Carboxylic Acids/chemical synthesis , Carboxylic Acids/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Endometriosis/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Female , Humans , Inflammation/drug therapy , Inflammation/metabolism , Mice , Molecular Docking Simulation , Molecular Structure , Pain/drug therapy , Pain/metabolism , Prostaglandin-E Synthases/metabolism , Rats , Structure-Activity Relationship
4.
Eur J Pharmacol ; 707(1-3): 32-40, 2013 May 05.
Article in English | MEDLINE | ID: mdl-23541724

ABSTRACT

This work aimed at establishing the relevance of using the in vivo model of cyclophosphamide (CYP)-induced bladder inflammation in rats for in vivo pharmacological studies. Specifically, we measured visceral nociception, identified key inflammatory mediators and evaluated the effects of relevant pharmacological treatments. Cystitis was induced in female rats by a single CYP injection. Sensitivity of the lower abdomen to von Frey mechanical stimulation was determined as a nociceptive parameter. Bladders were assessed for weight, wall thickness and macroscopic damage. Inflammatory mediators were quantified in bladders and urines. The effects of aspirin, ibuprofen and morphine were investigated on all these parameters. A single CYP injection increased nociceptive scores and decreased nociceptive threshold in response to mechanical stimuli between 1 and 4h post-administration. Increased bladder weight and wall thickness were associated with edema and hemorrhage. Bladder levels of IL-1ß, IL-6, MCP-1 and VCAM, and urinary levels of PGE2 were increased. In contrast, a decrease in the urinary metabolites, indoxyl sulfate and pantothenic acid, was observed. Aspirin, ibuprofen and morphine decreased CYP-induced referred visceral pain. Aspirin and ibuprofen also reversed the increased wall thickness, macroscopic damage and levels of IL-1ß, IL-6 and PGE2, and the decreased panthotenic acid levels. In contrast, morphine increased wall thickness, edema, hemorrhage, and bladder IL-6 and MCP-1 levels. This work presents a new and reliable method to evaluate visceral sensitivity in rats, and new relevant biomarkers identified in the bladder and urine to measure inflammation and pain parameters for in vivo pharmacological studies.


Subject(s)
Cyclophosphamide/toxicity , Cystitis/drug therapy , Inflammation/drug therapy , Visceral Pain/drug therapy , Analgesics, Opioid/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Biomarkers/metabolism , Cystitis/complications , Cystitis/physiopathology , Disease Models, Animal , Female , Ibuprofen/pharmacology , Inflammation/etiology , Inflammation/pathology , Inflammation Mediators/metabolism , Morphine/pharmacology , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Time Factors , Visceral Pain/etiology
5.
Eur J Pharmacol ; 667(1-3): 169-74, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21645509

ABSTRACT

Visceral hypersensitivity and an increased response to stress are two of the main symptoms of irritable bowel syndrome. Thus efforts to develop animal models of irritable bowel syndrome have centred on both of these parameters. The anticonvulsant gabapentin, which is widely used as an analgesic agent, also reduces anxiety. No data exists to our knowledge of the effects of gabapentin in animal models of co-morbid exaggerated stress response and visceral pain. Our aim was to assess the effect of gabapentin on stress and visceral hypersensitivity in two different animal models of irritable bowel syndrome. The animal models employed were the genetically susceptible Wistar Kyoto rat and the neonatally stressed maternal separation model. These animals were subjected to the open field paradigm to assess stress-induced defecation rates and colorectal distension to assess the level of visceral sensitivity. Gabapentin (30 mg/kg) prevented the stress-induced increase in faecal pellet output in the maternally separated rat, but not the Wistar Kyoto animals. On the other hand gabapentin (30 mg/kg) reduced the number of pain behaviours in response to colorectal distension in both models. These results show that whilst both models have similar responses to gabapentin in terms of visceral pain they differ in terms of their physiological response to stress. This indicates that the origin of anxiety and perhaps then visceral hypersensitivity differs in these models. Overall, these data suggest that gabapentin may be a useful treatment in disorders of co-morbid pain and an overactive stress system such as irritable bowel syndrome.


Subject(s)
Amines/pharmacology , Anxiety/drug therapy , Cyclohexanecarboxylic Acids/pharmacology , Visceral Pain/drug therapy , gamma-Aminobutyric Acid/pharmacology , Amines/therapeutic use , Animals , Anxiety/epidemiology , Comorbidity , Cyclohexanecarboxylic Acids/therapeutic use , Disease Models, Animal , Female , Gabapentin , Intestine, Large/drug effects , Male , Rats , Visceral Pain/epidemiology , gamma-Aminobutyric Acid/therapeutic use
6.
J Biol Chem ; 280(36): 31936-48, 2005 Sep 09.
Article in English | MEDLINE | ID: mdl-16027150

ABSTRACT

Tight junctions between intestinal epithelial cells prevent ingress of luminal macromolecules and bacteria and protect against inflammation and infection. During stress and inflammation, mast cells mediate increased mucosal permeability by unknown mechanisms. We hypothesized that mast cell tryptase cleaves protease-activated receptor 2 (PAR2) on colonocytes to increase paracellular permeability. Colonocytes expressed PAR2 mRNA and responded to PAR2 agonists with increased [Ca2+]i. Supernatant from degranulated mast cells increased [Ca2+]i in colonocytes, which was prevented by a tryptase inhibitor, and desensitized responses to PAR2 agonist, suggesting PAR2 cleavage. When applied to the basolateral surface of colonocytes, PAR2 agonists and mast cell supernatant decreased transepithelial resistance, increased transepithelial flux of macromolecules, and induced redistribution of tight junction ZO-1 and occludin and perijunctional F-actin. When mast cells were co-cultured with colonocytes, mast cell degranulation increased paracellular permeability of colonocytes. This was prevented by a tryptase inhibitor. We determined the role of ERK1/2 and of beta-arrestins, which recruit ERK1/2 to PAR2 in endosomes and retain ERK1/2 in the cytosol, on PAR2-mediated alterations in permeability. An ERK1/2 inhibitor abolished the effects of PAR2 agonist on permeability and redistribution of F-actin. Down-regulation of beta-arrestins with small interfering RNA inhibited PAR2-induced activation of ERK1/2 and suppressed PAR2-induced changes in permeability. Thus, mast cells signal to colonocytes in a paracrine manner by release of tryptase and activation of PAR2. PAR2 couples to beta-arrestin-dependent activation of ERK1/2, which regulates reorganization of perijunctional F-actin to increase epithelial permeability. These mechanisms may explain the increased epithelial permeability of the intestine during stress and inflammation.


Subject(s)
Arrestins/physiology , Colon/cytology , Colon/physiology , Receptor, PAR-2/physiology , Serine Endopeptidases/physiology , Tight Junctions/physiology , Actins/metabolism , Calcium/metabolism , Cell Line , Coculture Techniques , Colon/enzymology , Humans , Mast Cells/enzymology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Permeability , RNA, Small Interfering , Receptor, PAR-2/agonists , Receptor, PAR-2/genetics , Signal Transduction/physiology , Tight Junctions/enzymology , Tryptases , beta-Arrestins
7.
J Surg Res ; 117(2): 195-201, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15047123

ABSTRACT

Mechanisms of pain transduction in acute pancreatitis are poorly understood. Increased Fos expression in the spinal cord is a marker of activation of nociceptive neurons. We hypothesized that cerulein pancreatitis leads to increased Fos expression at T9 and T10, which receive sensory input from the pancreas. Rats were injected with cerulein (100 microg/kg, s.c.) or saline carrier (NS). Endpoints at 4, 6, and 10 h were serum amylase, myeloperoxidase activity (MPO), and spinal cord Fos expression (number of immunoreactive nuclei/section dorsal gray matter). Fos-like immunoreactivity (FLI) at T9-T10 was compared to internal controls (T6, T12). An average of 20 spinal cord histologic sections were evaluated per rat. Some animals were injected with the mu-opioid receptor agonist, buprenorphine (90 microg/kg, s.c.), 3 h after cerulein, and their endpoints were measured at 6 h. Analysis of variance and t tests were used for statistical analysis. Results are means +/- SEM. As expected, cerulein induced edematous pancreatitis, with a 4-fold increase in serum amylase at 6 h [cer (n = 8): 14,000 +/- 1,300 U/ml versus NS (n = 10): 3,700 +/- 300, P < 0.005)] and a 2-fold increase in MPO activity (0.25 +/- 0.05) activity units/dry wt versus 0.13 +/- 0.02, P < 0.05). Cerulein induced nearly a 2-fold increase in FLI at T9 and T10 [n = 10 (cer) and n = 13 (NS): T9, 14 +/- 1.5 versus 7.8 +/- 0.88; T10, 15 +/- 1.7 versus 8.3 +/- 0.70; P < 0.05]. Peak effects of cerulein on FLI occurred at 6 h and were greatest at T9/T10 with relative sparing of T6/T12. T6/T12 expression was similar in experimental and control groups. Buprenorphine significantly reduced both serum amylase and FLI and T9/T10. Cerulein-induced acute pancreatitis in rat increases visceral nociceptive signaling at spinal cord levels T9 and T10, with a peak at 6 h. Blockade of this effect by the mu-opioid receptor agonist buprenorphine could occur either by direct activation of central opioid receptors and/or an anti-inflammatory mechanism. FLI is a useful tool for studying the pathophysiology of pain in experimental acute pancreatitis.


Subject(s)
Nociceptors/physiopathology , Pancreatitis/physiopathology , Spinal Cord/physiopathology , Animals , Buprenorphine/pharmacology , Ceruletide , Male , Pancreatitis/chemically induced , Pancreatitis/metabolism , Proto-Oncogene Proteins c-fos/antagonists & inhibitors , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/agonists , Thoracic Vertebrae , Time Factors
8.
Can J Physiol Pharmacol ; 81(9): 920-7, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14614531

ABSTRACT

Proteinase-activated receptor-2 (PAR2) activation induces colonic inflammation by an unknown mechanism. We hypothesized that PAR2 agonists administered intracolonically in mice induce inflammation via a neurogenic mechanism. Pretreatment of mice with neurokinin-1 and calcitonin-gene-related peptide (CGRP) receptor antagonists or with capsaicin showed attenuated PAR2-agonist-induced colitis. Immunohistochemistry demonstrated a differential expression of a marker for the type-1 CGRP receptor during the time course of PAR2-agonist-induced colitis, further suggesting a role for CGRP. We conclude that PAR2-agonist-induced intestinal inflammation involves the release of neuropeptides, which by acting on their receptors cause inflammation. These results implicate PAR2 as an important mediator of intestinal neurogenic inflammation.


Subject(s)
Colitis/metabolism , Enteric Nervous System/metabolism , Receptor, PAR-2/agonists , Receptor, PAR-2/physiology , Animals , Calcitonin Gene-Related Peptide/metabolism , Capsaicin/pharmacology , Colitis/chemically induced , Colitis/physiopathology , Enteric Nervous System/drug effects , Enteric Nervous System/physiopathology , Inflammation/physiopathology , Mice , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Neuropeptides/metabolism , Oligopeptides/pharmacology , Piperidines/pharmacology , Quinuclidines/pharmacology , Receptors, Neurokinin-1/metabolism
9.
J Immunol ; 170(8): 4296-300, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12682265

ABSTRACT

Activation of colonic proteinase-activated receptor-2 (PAR-2) provokes colonic inflammation and increases mucosal permeability in mice. The mechanism of inflammation is under debate and could be neurogenic and/or the consequence of tight-junction opening with passage of exogenous pathogens into the lamina propria. The present study aimed to further characterize the inflammatory effect of PAR-2 activation by investigating: 1) the role of NO, 2) the role of afferent neurons, and 3) a possible cause and effect relationship between colonic paracellular permeability changes and mucosal inflammation. Thus, intracolonic infusion to mice of the PAR-2-activating peptide, SLIGRL, increased both myeloperoxidase (MPO) activity and damage scores indicating colonic inflammation, and enhanced colonic permeability to (51)Cr-EDTA from 2 to 4 h after its infusion. NO synthase inhibitors, L-NAME and aminoguanidine, as well as the neurotoxin capsaicin and NK1, calcitonin gene-related peptide (CGRP) receptor antagonists, SR140333 and CGRP(8-37), prevented SLIGRL-induced MPO and damage score increases and permeability. In contrast, although the tight-junction blocker, 2,4,6-triaminopyrimidine, and the myosin L chain kinase inhibitor, ML-7, prevented SLIGRL-induced increase in permeability, they did not prevent MPO and damage score increases. Taken together our data show that both NO and capsaicin-sensitive afferent neurons are involved in PAR-2-mediated colonic inflammation and paracellular permeability increase. Nevertheless, the inflammation process is not a consequence of increased permeability which results at least in part from the activation of myosin L chain kinase.


Subject(s)
Cell Membrane Permeability , Colon/pathology , Intestinal Mucosa/metabolism , Neurons, Afferent/physiology , Nitric Oxide/physiology , Receptors, Thrombin/administration & dosage , Receptors, Thrombin/physiology , Administration, Rectal , Animals , Antigens , Antigens, Surface , Azepines/administration & dosage , Azepines/pharmacology , Calcitonin Gene-Related Peptide/pharmacology , Calcitonin Gene-Related Peptide Receptor Antagonists , Cell Membrane Permeability/drug effects , Colon/drug effects , Colon/enzymology , Colon/metabolism , Denervation , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Inflammation/enzymology , Inflammation/metabolism , Inflammation/physiopathology , Injections, Intraperitoneal , Intercellular Junctions/drug effects , Intercellular Junctions/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Lectins, C-Type , Male , Mice , Myosin-Light-Chain Kinase/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily B , Naphthalenes/administration & dosage , Naphthalenes/pharmacology , Neurons, Afferent/pathology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Oligopeptides/administration & dosage , Oligopeptides/antagonists & inhibitors , Oligopeptides/pharmacology , Peptide Fragments/pharmacology , Piperidines/administration & dosage , Piperidines/pharmacology , Proteins/antagonists & inhibitors , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Quinuclidines/administration & dosage , Quinuclidines/pharmacology , Receptor, PAR-2
10.
Article in English | MEDLINE | ID: mdl-15317291

ABSTRACT

Protease-activated receptor 2 (PAR2) is the second member of a new subfamily of G-protein coupled receptors: the protease-activated receptors (PARs). At present, four different PARs have been cloned and all of them share the same basic mechanism of activation. A serine protease cleaves the extended, extracellular N-terminus of the receptor at a specific site within the protein chain to expose an N-terminal tethered ligand domain, which binds to and activates the cleaved receptor. In this manner, trypsin and mast cell beta-tryptase activate PAR2. PARs are single use receptors because proteolytic activation is irreversible and the cleaved receptors are degraded in lysosomes. Thus, PARs play important roles in emergency situations, such as trauma and inflammation. Emerging evidence indicates that PAR2 is involved in the cardiovascular, pulmonary and gastrointestinal systems, where it controls inflammation and nociception. Work with selective agonists and knockout animals suggests a contribution of PAR2 to certain inflammatory diseases. Therefore, selective antagonists or agonists of these receptors may be useful therapeutic agents for the treatment of human diseases.


Subject(s)
Receptor, PAR-2/physiology , Animals , Blood Vessels , Digestive System , Humans , Inflammation , Lung , Receptor, PAR-2/agonists , Receptor, PAR-2/metabolism , Serine Endopeptidases/metabolism , Vascular Diseases
11.
Essays Biochem ; 38: 169-83, 2002.
Article in English | MEDLINE | ID: mdl-12463169

ABSTRACT

Certain extracellular proteases, derived from the circulation and inflammatory cells, can specifically cleave and trigger protease-activated receptors (PARs), a small, but important, sub-group of the G-protein-coupled receptor super-family. Four PARs have been cloned and they all share the same basic mechanism of activation: proteases cleave at a specific site within the extracellular N-terminus to expose a new N-terminal tethered ligand domain, which binds to and thereby activates the cleaved receptor. Thrombin activates PAR1, PAR3 and PAR4, trypsin activates PAR2 and PAR4, and mast cell tryptase activates PAR2 in this manner. Activated PARs couple to signalling cascades that affect cell shape, secretion, integrin activation, metabolic responses, transcriptional responses and cell motility. PARs are 'single-use' receptors: proteolytic activation is irreversible and the cleaved receptors are degraded in lysosomes. Thus, PARs play important roles in 'emergency situations', such as trauma and inflammation. The availability of selective agonists and antagonists of protease inhibitors and of genetic models has generated evidence to suggests that proteases and their receptors play important roles in coagulation, inflammation, pain, healing and protection. Therefore, selective antagonists or agonists of these receptors may be useful therapeutic agents for the treatment of human diseases.


Subject(s)
Cell Membrane/metabolism , Receptors, Thrombin/metabolism , Receptors, Thrombin/physiology , Signal Transduction , Animals , Humans , Inflammation , Models, Biological , Protein Processing, Post-Translational , Receptor, PAR-1 , Receptor, PAR-2
12.
Am J Pathol ; 161(5): 1903-15, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12414536

ABSTRACT

Proteinase-activated receptor (PAR)-2, a G-protein-coupled receptor for trypsin and mast cell tryptase, is highly expressed in the intestine. Luminal trypsin and tryptase are elevated in the colon of inflammatory bowel disease patients. We hypothesized that luminal proteinases activate PAR-2 and induce colonic inflammation. Mice received intracolonically PAR-2 agonists (trypsin, tryptase, and a selective PAR-2-activating peptide) or control drugs (boiled enzymes, inactive peptide) and inflammatory parameters were followed at various times after this treatment. Colonic administration of PAR-2 agonists up-regulated PAR-2 expression and induced an inflammatory reaction characterized by granulocyte infiltration, increased wall thickness, tissue damage, and elevated T-helper cell type 1 cytokine. The inflammation was maximal between 4 and 6 hours and was resolved 48 hours after the intracolonic administration. PAR-2 activation also increased paracellular permeability of the colon and induced bacterial trans-location into peritoneal organs. These proinflammatory and pathophysiological changes observed in wild-type mice were not detected in PAR-2-deficient mice. Luminal proteinases activate PAR-2 in the mouse colon to induce inflammation and disrupt the integrity of the intestinal barrier. Because trypsin and tryptase are found at high levels in the colon lumen of patients with Crohn's disease or ulcerative colitis, our data may bear directly on the pathophysiology of human inflammatory bowel diseases.


Subject(s)
Colitis/etiology , Receptors, Thrombin/metabolism , Animals , Bacterial Translocation , Colitis/chemically induced , Colitis/pathology , Colon/immunology , Colon/metabolism , Colon/microbiology , Cytokines/biosynthesis , Dose-Response Relationship, Drug , Intestinal Mucosa/metabolism , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Permeability , RNA, Messenger/biosynthesis , Receptor, PAR-2 , Receptors, Thrombin/agonists , Receptors, Thrombin/genetics , Serine Endopeptidases , Trypsin , Tryptases
13.
Gastroenterology ; 122(4): 1035-47, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11910355

ABSTRACT

BACKGROUND & AIMS: PAR-2s are highly expressed throughout the gastrointestinal tract. These receptors are cleaved by trypsin and mast cell tryptase and can be activated by peptides corresponding to the tethered ligand of the receptor (SLIGRL-NH2 for rat). The aim of this study was to determine whether colonic administration of PAR-2 agonists affects visceral sensitivity to rectal distention in conscious rats. METHODS: Abdominal contractions (a criteria of visceral pain) were recorded in rats equipped with intramuscular electrodes. Rectal distention was performed at various times after intracolonic infusion of SLIGRL-NH2 and trypsin. Inflammation parameters and permeability were followed in the colon after the intracolonic injections. Fos expression at a spinal level (L4-L6) was also studied 2 hours after intracolonic injection of SLIGRL-NH2. RESULTS: Rectal distention significantly increased abdominal contractions starting at the RD volume of 0.8 mL. Intracolonic injection of SLIGRL-NH2 (200 microg/rat) and trypsin (200 U/rat), but not vehicle, LRGILS-NH2 (control peptide), boiled trypsin, or SLIGRL-NH2 injected IP, significantly increased (P < 0.05) abdominal contractions for high volumes of distention, 10- and 24-hour postinfusion. SLIGRL-NH2-induced hyperalgesia was inhibited by a NK1 receptor antagonist (SR 140333) but not by indomethacin. Intracolonic injection of SLIGRL-NH2 elevated spinal Fos expression and caused increased intestinal permeability but did not cause detectable inflammation. CONCLUSIONS: Intracolonic infusion of subinflammatory doses of PAR-2 agonists activated spinal afferent neurons and produced a delayed rectal hyperalgesia that involves changes in intestinal permeability and the activation of NK1 receptors. These results identify a possible role for proteinases and PAR-2 in the genesis of visceral hyperalgesia.


Subject(s)
Hyperalgesia/metabolism , Receptors, Thrombin/metabolism , Trypsin/pharmacology , Animals , Catheterization , Chelating Agents/pharmacokinetics , Chromium Radioisotopes , Colitis/pathology , Colon/innervation , Compliance/drug effects , Edetic Acid/pharmacokinetics , Hyperalgesia/chemically induced , Injections, Intraperitoneal , Intestinal Absorption/drug effects , Male , Neurons, Afferent/physiology , Oligopeptides/pharmacology , Prostaglandins/physiology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Receptor, PAR-2 , Receptors, Neurokinin-1/metabolism , Receptors, Thrombin/agonists , Rectum/innervation , Signal Transduction/physiology , Spinal Cord/cytology , Spinal Cord/physiology , Substance P/physiology , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...