Network


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

Hotspot


Dive into the research topics where Nestor N. Jiménez-Vargas is active.

Publication


Featured researches published by Nestor N. Jiménez-Vargas.


Journal of Biological Chemistry | 2015

Neutrophil Elastase Activates Protease-activated Receptor-2 (PAR2) and Transient Receptor Potential Vanilloid 4 (TRPV4) to Cause Inflammation and Pain.

Peishen Zhao; TinaMarie Lieu; Nicholas Barlow; Silvia Sostegni; Silke Haerteis; Christoph Korbmacher; Wolfgang Liedtke; Nestor N. Jiménez-Vargas; Stephen Vanner; Nigel W. Bunnett

Background: Proteases cleave protease-activated receptor-2 (PAR2), which activates transient receptor potential (TRP) ion channels to cause inflammation and pain. Results: Neutrophil elastase cleaves PAR2, resulting in Gαs-mediated cAMP formation, transient receptor potential vanilloid 4 (TRPV4) activation, and sensitization of nociceptive neurons, inflammation, and pain. Conclusion: Elastase causes PAR2- and TRPV4-mediated inflammation and pain. Significance: PARs and TRP channels mediate responses to diverse proteases. Proteases that cleave protease-activated receptor-2 (PAR2) at Arg36↓Ser37 reveal a tethered ligand that binds to the cleaved receptor. PAR2 activates transient receptor potential (TRP) channels of nociceptive neurons to induce neurogenic inflammation and pain. Although proteases that cleave PAR2 at non-canonical sites can trigger distinct signaling cascades, the functional importance of the PAR2-biased agonism is uncertain. We investigated whether neutrophil elastase, a biased agonist of PAR2, causes inflammation and pain by activating PAR2 and TRP vanilloid 4 (TRPV4). Elastase cleaved human PAR2 at Ala66↓Ser67 and Ser67↓Val68. Elastase stimulated PAR2-dependent cAMP accumulation and ERK1/2 activation, but not Ca2+ mobilization, in KNRK cells. Elastase induced PAR2 coupling to Gαs but not Gαq in HEK293 cells. Although elastase did not promote recruitment of G protein-coupled receptor kinase-2 (GRK2) or β-arrestin to PAR2, consistent with its inability to promote receptor endocytosis, elastase did stimulate GRK6 recruitment. Elastase caused PAR2-dependent sensitization of TRPV4 currents in Xenopus laevis oocytes by adenylyl cyclase- and protein kinase A (PKA)-dependent mechanisms. Elastase stimulated PAR2-dependent cAMP formation and ERK1/2 phosphorylation, and a PAR2- and TRPV4-mediated influx of extracellular Ca2+ in mouse nociceptors. Adenylyl cyclase and PKA-mediated elastase-induced activation of TRPV4 and hyperexcitability of nociceptors. Intraplantar injection of elastase to mice caused edema and mechanical hyperalgesia by PAR2- and TRPV4-mediated mechanisms. Thus, the elastase-biased agonism of PAR2 causes Gαs-dependent activation of adenylyl cyclase and PKA, which activates TRPV4 and sensitizes nociceptors to cause inflammation and pain. Our results identify a novel mechanism of elastase-induced activation of TRPV4 and expand the role of PAR2 as a mediator of protease-driven inflammation and pain.


Journal of Biological Chemistry | 2016

Protein Kinase D and Gβγ Subunits Mediate Agonist-evoked Translocation of Protease-activated Receptor-2 from the Golgi Apparatus to the Plasma Membrane.

Dane D. Jensen; Peishen Zhao; Nestor N. Jiménez-Vargas; TinaMarie Lieu; Marina Gerges; Holly R. Yeatman; Meritxell Canals; Stephen Vanner; Daniel P. Poole; Nigel W. Bunnett

Agonist-evoked endocytosis of G protein-coupled receptors has been extensively studied. The mechanisms by which agonists stimulate mobilization and plasma membrane translocation of G protein-coupled receptors from intracellular stores are unexplored. Protease-activated receptor-2 (PAR2) traffics to lysosomes, and sustained protease signaling requires mobilization and plasma membrane trafficking of PAR2 from Golgi stores. We evaluated the contribution of protein kinase D (PKD) and Gβγ to this process. In HEK293 and KNRK cells, the PAR2 agonists trypsin and 2-furoyl-LIGRLO-NH2 activated PKD in the Golgi apparatus, where PKD regulates protein trafficking. PAR2 activation induced translocation of Gβγ, a PKD activator, to the Golgi apparatus, determined by bioluminescence resonance energy transfer between Gγ-Venus and giantin-Rluc8. Inhibitors of PKD (CRT0066101) and Gβγ (gallein) prevented PAR2-stimulated activation of PKD. CRT0066101, PKD1 siRNA, and gallein all inhibited recovery of PAR2-evoked Ca2+ signaling. PAR2 with a photoconvertible Kaede tag was expressed in KNRK cells to examine receptor translocation from the Golgi apparatus to the plasma membrane. Irradiation of the Golgi region (405 nm) induced green-red photo-conversion of PAR2-Kaede. Trypsin depleted PAR2-Kaede from the Golgi apparatus and repleted PAR2-Kaede at the plasma membrane. CRT0066101 inhibited PAR2-Kaede translocation to the plasma membrane. CRT0066101 also inhibited sustained protease signaling to colonocytes and nociceptive neurons that naturally express PAR2 and mediate protease-evoked inflammation and nociception. Our results reveal a major role for PKD and Gβγ in agonist-evoked mobilization of intracellular PAR2 stores that is required for sustained signaling by extracellular proteases.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Protease-activated receptor-2 in endosomes signals persistent pain of irritable bowel syndrome

Nestor N. Jiménez-Vargas; Luke Pattison; Peishen Zhao; TinaMarie Lieu; Rocco Latorre; Dane D. Jensen; Joel Castro; Luigi Aurelio; Giang T. Le; Bernard L. Flynn; Carmen Klein Herenbrink; Holly R. Yeatman; Laura E. Edgington-Mitchell; Christopher J. H. Porter; Michelle L. Halls; Meritxell Canals; Nicholas A. Veldhuis; Daniel P. Poole; Peter McLean; Gareth A. Hicks; Nicole N. Scheff; Elyssa Chen; Aditi Bhattacharya; Brian L. Schmidt; Stuart M. Brierley; Stephen Vanner; Nigel W. Bunnett

Significance Activated G protein-coupled receptors (GPCRs) internalize and can continue to signal from endosomes. The contribution of endosomal signaling to human disease is unknown. Proteases that are generated in the colon of patients with irritable bowel syndrome (IBS) can cleave protease-activated receptor-2 (PAR2) on nociceptors to cause pain. We evaluated whether PAR2 generates signals in endosomes of nociceptors that mediate persistent hyperexcitability and pain. Biopsies of colonic mucosa from IBS patients released proteases that induced PAR2 endocytosis, endosomal signaling, and persistent hyperexcitability of nociceptors. When conjugated to the transmembrane lipid cholestanol, PAR2 antagonists accumulated in endosomes and suppressed persistent hyperexcitability. The results reveal the therapeutic potential of endosomally targeted PAR2 antagonists for IBS pain, and expand the contribution of endosomal GPCR signaling to encompass processes that are relevant to disease. Once activated at the surface of cells, G protein-coupled receptors (GPCRs) redistribute to endosomes, where they can continue to signal. Whether GPCRs in endosomes generate signals that contribute to human disease is unknown. We evaluated endosomal signaling of protease-activated receptor-2 (PAR2), which has been proposed to mediate pain in patients with irritable bowel syndrome (IBS). Trypsin, elastase, and cathepsin S, which are activated in the colonic mucosa of patients with IBS and in experimental animals with colitis, caused persistent PAR2-dependent hyperexcitability of nociceptors, sensitization of colonic afferent neurons to mechanical stimuli, and somatic mechanical allodynia. Inhibitors of clathrin- and dynamin-dependent endocytosis and of mitogen-activated protein kinase kinase-1 prevented trypsin-induced hyperexcitability, sensitization, and allodynia. However, they did not affect elastase- or cathepsin S-induced hyperexcitability, sensitization, or allodynia. Trypsin stimulated endocytosis of PAR2, which signaled from endosomes to activate extracellular signal-regulated kinase. Elastase and cathepsin S did not stimulate endocytosis of PAR2, which signaled from the plasma membrane to activate adenylyl cyclase. Biopsies of colonic mucosa from IBS patients released proteases that induced persistent PAR2-dependent hyperexcitability of nociceptors, and PAR2 association with β-arrestins, which mediate endocytosis. Conjugation to cholestanol promoted delivery and retention of antagonists in endosomes containing PAR2. A cholestanol-conjugated PAR2 antagonist prevented persistent trypsin- and IBS protease-induced hyperexcitability of nociceptors. The results reveal that PAR2 signaling from endosomes underlies the persistent hyperexcitability of nociceptors that mediates chronic pain of IBS. Endosomally targeted PAR2 antagonists are potential therapies for IBS pain. GPCRs in endosomes transmit signals that contribute to human diseases.


British Journal of Pharmacology | 2018

Co-expression of μ and ∂ opioid receptors by mouse colonic nociceptors

Raquel Guerrero-Alba; Eduardo Valdez-Morales; Nestor N. Jiménez-Vargas; Romke Bron; Daniel P. Poole; David E. Reed; Joel Castro; Melissa Campaniello; Patrick A. Hughes; Stuart M. Brierley; Nigel W. Bunnett; Alan E. Lomax; Stephen Vanner

To better understand opioid signalling in visceral nociceptors, we examined the expression and selective activation of μ and δ opioid receptors by dorsal root ganglia (DRG) neurons innervating the mouse colon.


Journal of the Canadian Association of Gastroenterology | 2018

A300 DIET-MICROBIOTA INTERACTIONS UNDERLIE SYMPTOMS IN IBS

G. De Palma; David E. Reed; Marc Pigrau; Jun Lu; Sacha Sidani; Y Zhang; Yang Yu; Nestor N. Jiménez-Vargas; J Sessenwein; C D Lopez Lopez; J O Jaramillo Polanco; Elena F. Verdu; Stephen M. Collins; Alan E. Lomax; Michael Beyak; Stephen Vanner; Premysl Bercik


Journal of the Canadian Association of Gastroenterology | 2018

A20 GUT MICROBIOTA-DIET INTERACTION ALTER INTESTINAL MAST CELL NUMBERS AND DISTRIBUTION IN THE HUMANIZED IBS MOUSE MODEL

Chiko Shimbori; G. De Palma; D E Reed; Marc Pigrau; Jun Lu; Y Zhang; Yang Yu; Nestor N. Jiménez-Vargas; J Sessenwein; C D Lopez Lopez; J O Jaramillo Polanco; Elena F. Verdu; Stephen M. Collins; Alan E. Lomax; Michael Beyak; Stephen Vanner; Premysl Bercik


Journal of the Canadian Association of Gastroenterology | 2018

A25 COLONIC PROTEASES EVOKE SUSTAINED PAIN SIGNALING VIA A NOVEL ENDOSOMAL PATHWAY IN NEURONS

Nestor N. Jiménez-Vargas; Nigel W. Bunnett; Stephen Vanner


Journal of the Canadian Association of Gastroenterology | 2018

A21 FERMENTABLE CARBOHYDRATE-MICROBIOME INTERACTIONS IN A MOUSE MODEL OF IBS

G. De Palma; David E. Reed; Chiko Shimbori; Marc Pigrau; Jun Lu; Marc P. Louis-Auguste; Y Zhang; Yang Yu; Nestor N. Jiménez-Vargas; J Sessenwein; C D Lopez Lopez; J O Jaramillo Polanco; Elena F. Verdu; Stephen M. Collins; Karen Madsen; Alan E. Lomax; Michael Beyak; Stephen Vanner; Premysl Bercik


Gastroenterology | 2018

1049 - A Low Fodmap (Fermentable Carbohydrate) Diet Worsens Nociceptive Signalling Following Recovery from Acute Dss Colitis in Mice

Caroline J Tuck; Nestor N. Jiménez-Vargas; Josue O. Jaramillo Polanco; Cintya D. Lopez Lopez; Sandra Lourenssen; Alan E. Lomax; Michael Beyak; David E. Reed; Stephen Vanner


Gastroenterology | 2018

Mo1604 - Colonic Proteases Evoke Sustained Pain Signaling via a Novel Endosomal Pathway in Neurons

Nestor N. Jiménez-Vargas; Nigel W. Bunnett; Stephen Vanner

Collaboration


Dive into the Nestor N. Jiménez-Vargas'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

Jun Lu

McMaster University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge