Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Nathalie Vergnolle is active.

Publication


Featured researches published by Nathalie Vergnolle.


Nature Medicine | 2000

Agonists of proteinase-activated receptor 2 induce inflammation by a neurogenic mechanism.

Martin Steinhoff; Nathalie Vergnolle; Steven H. Young; Michele Tognetto; Silvia Amadesi; Helena S. Ennes; Marcello Trevisani; Morley D. Hollenberg; John L. Wallace; G H Caughey; Sharon E. Mitchell; Lynda M. Williams; Pierangelo Geppetti; Emeran A. Mayer; N.W. Bunnett

Trypsin and mast cell tryptase cleave proteinase-activated receptor 2 and, by unknown mechanisms, induce widespread inflammation. We found that a large proportion of primary spinal afferent neurons, which express proteinase-activated receptor 2, also contain the proinflammatory neuropeptides calcitonin gene-related peptide and substance P. Trypsin and tryptase directly signal to neurons to stimulate release of these neuropeptides, which mediate inflammatory edema induced by agonists of proteinase-activated receptor 2. This new mechanism of protease-induced neurogenic inflammation may contribute to the proinflammatory effects of mast cells in human disease. Thus, tryptase inhibitors and antagonists of proteinase-activated receptor 2 may be useful anti-inflammatory agents.


Journal of Clinical Investigation | 2007

Role for protease activity in visceral pain in irritable bowel syndrome

Nicolas Cenac; Christopher N. Andrews; M. Holzhausen; Kevin T. Chapman; Graeme S. Cottrell; Patricia Andrade-Gordon; Martin Steinhoff; Giovanni Barbara; Paul L. Beck; Nigel W. Bunnett; Keith A. Sharkey; Jose G. P. Ferraz; Eldon A. Shaffer; Nathalie Vergnolle

Mediators involved in the generation of symptoms in patients with irritable bowel syndrome (IBS) are poorly understood. Here we show that colonic biopsy samples from IBS patients release increased levels of proteolytic activity (arginine cleavage) compared to asymptomatic controls. This was dependent on the activation of NF-kappaB. In addition, increased proteolytic activity was measured in vivo, in colonic washes from IBS compared with control patients. Trypsin and tryptase expression and release were increased in colonic biopsies from IBS patients compared with control subjects. Biopsies from IBS patients (but not controls) released mediators that sensitized murine sensory neurons in culture. Sensitization was prevented by a serine protease inhibitor and was absent in neurons lacking functional protease-activated receptor-2 (PAR2). Supernatants from colonic biopsies of IBS patients, but not controls, also caused somatic and visceral hyperalgesia and allodynia in mice, when administered into the colon. These pronociceptive effects were inhibited by serine protease inhibitors and a PAR2 antagonist and were absent in PAR2-deficient mice. Our study establishes that proteases are released in IBS and that they can directly stimulate sensory neurons and generate hypersensitivity symptoms through the activation of PAR2.


Nature Medicine | 2001

Proteinase-activated receptor-2 and hyperalgesia: A novel pain pathway.

Nathalie Vergnolle; N.W. Bunnett; Keith A. Sharkey; V. Brussee; S.J. Compton; E.F. Grady; Giuseppe Cirino; Norma P. Gerard; Allan I. Basbaum; P. Andrade-Gordon; Morley D. Hollenberg; John L. Wallace

Using a combined pharmacological and gene-deletion approach, we have delineated a novel mechanism of neurokinin-1 (NK-1) receptor-dependent hyperalgesia induced by proteinase-activated receptor-2 (PAR2), a G-protein–coupled receptor expressed on nociceptive primary afferent neurons. Injections into the paw of sub-inflammatory doses of PAR2 agonists in rats and mice induced a prolonged thermal and mechanical hyperalgesia and elevated spinal Fos protein expression. This hyperalgesia was markedly diminished or absent in mice lacking the NK-1 receptor, preprotachykinin-A or PAR2 genes, or in rats treated with a centrally acting cyclooxygenase inhibitor or treated by spinal cord injection of NK-1 antagonists. Here we identify a previously unrecognized nociceptive pathway with important therapeutic implications, and our results point to a direct role for proteinases and their receptors in pain transmission.


The Journal of Neuroscience | 2004

Protease-Activated Receptor 2 Sensitizes the Capsaicin Receptor Transient Receptor Potential Vanilloid Receptor 1 to Induce Hyperalgesia

Silvia Amadesi; Jingjiang Nie; Nathalie Vergnolle; Graeme S. Cottrell; Eileen F. Grady; Marcello Trevisani; Chiara Manni; Pierangelo Geppetti; James A. McRoberts; Helena S. Ennes; John B. Davis; Emeran A. Mayer; Nigel W. Bunnett

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.


Trends in Pharmacological Sciences | 2001

Protease-activated receptors in inflammation, neuronal signaling and pain.

Nathalie Vergnolle; John L. Wallace; Nigel W. Bunnett; Morley D. Hollenberg

The ability of proteases to regulate cell function via protease-activated receptors (PARs) has led to new insights about the potential physiological functions of these enzymes. Several studies suggest that PARs play roles in both inflammation and tissue repair, depending on the cellular environment in which they act. The recent detection of PARs on peripheral and central neurons suggests that neuronal PARs might be involved not only in neurogenic inflammation and neurodegenerative processes, but also in nociception. Thus, the list of potential roles for PARs has lengthened considerably and their physiological course of action might be much broader than initially anticipated.


American Journal of Pathology | 2002

Induction of Intestinal Inflammation in Mouse by Activation of Proteinase-Activated Receptor-2

Nicolas Cenac; Anne-Marie Coelho; Cathy Nguyen; Steven J. Compton; Patricia Andrade-Gordon; Wallace K. MacNaughton; John L. Wallace; Morley D. Hollenberg; Nigel W. Bunnett; Rafael Garcia-Villar; Lionel Bueno; Nathalie Vergnolle

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 Crohns disease or ulcerative colitis, our data may bear directly on the pathophysiology of human inflammatory bowel diseases.


The Journal of Physiology | 2007

Protease‐activated receptor 2 sensitizes the transient receptor potential vanilloid 4 ion channel to cause mechanical hyperalgesia in mice

Andrew D. Grant; Graeme S. Cottrell; Silvia Amadesi; Marcello Trevisani; Paola Nicoletti; Serena Materazzi; Christophe Altier; Nicolas Cenac; Gerald W. Zamponi; Francisco Bautista-Cruz; Carlos Barajas Lopez; Elizabeth K. Joseph; Jon D. Levine; Wolfgang Liedtke; Stephen Vanner; Nathalie Vergnolle; Pierangelo Geppetti; Nigel W. Bunnett

Exacerbated sensitivity to mechanical stimuli that are normally innocuous or mildly painful (mechanical allodynia and hyperalgesia) occurs during inflammation and underlies painful diseases. Proteases that are generated during inflammation and disease cleave protease‐activated receptor 2 (PAR2) on afferent nerves to cause mechanical hyperalgesia in the skin and intestine by unknown mechanisms. We hypothesized that PAR2‐mediated mechanical hyperalgesia requires sensitization of the ion channel transient receptor potential vanilloid 4 (TRPV4). Immunoreactive TRPV4 was coexpressed by rat dorsal root ganglia (DRG) neurons with PAR2, substance P (SP) and calcitonin gene‐related peptide (CGRP), mediators of pain transmission. In PAR2‐expressing cell lines that either naturally expressed TRPV4 (bronchial epithelial cells) or that were transfected to express TRPV4 (HEK cells), pretreatment with a PAR2 agonist enhanced Ca2+ and current responses to the TRPV4 agonists phorbol ester 4α‐phorbol 12,13‐didecanoate (4αPDD) and hypotonic solutions. PAR2‐agonist similarly sensitized TRPV4 Ca2+ signals and currents in DRG neurons. Antagonists of phospholipase Cβ and protein kinases A, C and D inhibited PAR2‐induced sensitization of TRPV4 Ca2+ signals and currents. 4αPDD and hypotonic solutions stimulated SP and CGRP release from dorsal horn of rat spinal cord, and pretreatment with PAR2 agonist sensitized TRPV4‐dependent peptide release. Intraplantar injection of PAR2 agonist caused mechanical hyperalgesia in mice and sensitized pain responses to the TRPV4 agonists 4αPDD and hypotonic solutions. Deletion of TRPV4 prevented PAR2 agonist‐induced mechanical hyperalgesia and sensitization. This novel mechanism, by which PAR2 activates a second messenger to sensitize TRPV4‐dependent release of nociceptive peptides and induce mechanical hyperalgesia, may underlie inflammatory hyperalgesia in diseases where proteases are activated and released.


British Journal of Pharmacology | 1999

Characterization of the inflammatory response to proteinase‐activated receptor‐2 (PAR2)‐activating peptides in the rat paw

Nathalie Vergnolle; Morley D. Hollenberg; Keith A. Sharkey; John L. Wallace

In the present study, we have observed the development of an inflammatory reaction in the rat hindpaw, following the injection of specific agonists of PAR2 (two PAR2 activating peptides). This inflammation was characterized by oedema and granulocyte infiltration. Two selective PAR2 activating peptides, SLGRL‐NH2 and trans‐cinnamoyl‐LIGRLO‐NH2 induced significant oedema in the rat hindpaw from 1–6 h following subplantar injection. Six hours after the PAR2‐activating peptide injection, the paw tissues showed a complete disruption of tissue architecture along with an inflammatory cell infiltrate. In the inflamed paw, PAR2‐immunoreactivity was expressed on endothelial cells as well as on the infiltrating inflammatory cells. The oedema induced by the injection of the two PAR2 activating peptides was slightly reduced in rats pre‐treated with compound 48/80, but was not modified by pre‐treatment of rats with cromolyn, a mast cell stabilizer. Pre‐treatment of rats with a cyclo‐oxygenase inhibitor (indomethacin) or a nitric oxide synthase inhibitor (L‐Nω‐nitro‐L‐arginine methyl ester) had no effect on the oedema induced by the PAR2‐activating peptides. These results demonstrate that the administration of PAR2‐activating peptides into the rat paw induced an acute inflammatory response characterized by a persistent oedema (at least 6 h) and granulocyte infiltration. The PAR2‐induced inflammatory response occurred through a mechanism largely independent of mast cell activation, and of the production of prostanoids and nitric oxide.


The Journal of Physiology | 2006

Protease‐activated receptor 2 sensitizes TRPV1 by protein kinase Cɛ‐ and A‐dependent mechanisms in rats and mice

Silvia Amadesi; Graeme S. Cottrell; Lorna Divino; Kevin Chapman; Eileen F. Grady; Francisco Bautista; Rustum Karanjia; Carlos Barajas-López; Stephen Vanner; Nathalie Vergnolle; Nigel W. Bunnett

Proteases that are released during inflammation and injury cleave protease‐activated receptor 2 (PAR2) on primary afferent neurons to cause neurogenic inflammation and hyperalgesia. PAR2‐induced thermal hyperalgesia depends on sensitization of transient receptor potential vanilloid receptor 1 (TRPV1), which is gated by capsaicin, protons and noxious heat. However, the signalling mechanisms by which PAR2 sensitizes TRPV1 are not fully characterized. Using immunofluorescence and confocal microscopy, we observed that PAR2 was colocalized with protein kinase (PK) Cɛ and PKA in a subset of dorsal root ganglia neurons in rats, and that PAR2 agonists promoted translocation of PKCɛ and PKA catalytic subunits from the cytosol to the plasma membrane of cultured neurons and HEK 293 cells. Subcellular fractionation and Western blotting confirmed this redistribution of kinases, which is indicative of activation. Although PAR2 couples to phospholipase Cβ, leading to stimulation of PKC, we also observed that PAR2 agonists increased cAMP generation in neurons and HEK 293 cells, which would activate PKA. PAR2 agonists enhanced capsaicin‐stimulated increases in [Ca2+]i and whole‐cell currents in HEK 293 cells, indicating TRPV1 sensitization. The combined intraplantar injection of non‐algesic doses of PAR2 agonist and capsaicin decreased the latency of paw withdrawal to radiant heat in mice, indicative of thermal hyperalgesia. Antagonists of PKCɛ and PKA prevented sensitization of TRPV1 Ca2+ signals and currents in HEK 293 cells, and suppressed thermal hyperalgesia in mice. Thus, PAR2 activates PKCɛ and PKA in sensory neurons, and thereby sensitizes TRPV1 to cause thermal hyperalgesia. These mechanisms may underlie inflammatory pain, where multiple proteases are generated and released.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Proteinase-activated receptor 2 is an anti-inflammatory signal for colonic lamina propria lymphocytes in a mouse model of colitis.

Stefano Fiorucci; Andrea Mencarelli; Barbara Palazzetti; Eleonora Distrutti; Nathalie Vergnolle; Morley D. Hollenberg; John L. Wallace; Antonio Morelli; Giuseppe Cirino

The proteinase-activated receptor 2 (PAR-2) is a member of a family of G protein-coupled receptors for proteases. Proteases cleave PARs within the extracellular N-terminal domains to expose tethered ligands that bind to and activate the cleaved receptors. PAR-2 is highly expressed in colon in epithelial and neuronal elements. In this study we show that PAR-2 activation prevents the development and induces healing of T helper cell type 1-mediated experimental colitis induced by intrarectal administration of 2,4,6-trinitrobenzene sulfonic acid (TNBS) in mice. A role for PAR-2 in the protection against colon inflammation was explored by the use of SLIGRL-NH2, a synthetic peptide that corresponds to the mouse tethered ligand exposed after PAR-2 cleavage. TNBS-induced colitis was dose-dependently reduced by the administration of SLIGRL-NH2, whereas the scramble control peptide, LSIGRL-NH2, was uneffective. This beneficial effect was reflected by increased survival rates, improvement of macroscopic and histologic scores, decrease in mucosal content of T helper cell type 1 cytokines, protein, and mRNA, and a diminished myeloperoxidase activity. SLIGRL-NH2, but not the scramble peptide, directly inhibited IFN-γ secretion and CD44 expression on lamina propria T lymphocytes. Protection exerted by PAR-2 in TNBS-treated mice was reverted by injecting mice with a truncated form of calcitonin gene-related peptide and by sensory neurons ablation with the neurotoxin capsaicin. Collectively, these studies show that PAR-2 is an anti-inflammatory receptor in the colon and suggest that PAR-2 ligands might be effective in the treatment of inflammatory bowel diseases.

Collaboration


Dive into the Nathalie Vergnolle's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge