Network


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

Hotspot


Dive into the research topics where Bernard Renaux is active.

Publication


Featured researches published by Bernard Renaux.


British Journal of Pharmacology | 2001

Glycosylation and the activation of proteinase‐activated receptor 2 (PAR2) by human mast cell tryptase

Steven J. Compton; Bernard Renaux; Suranga J. Wijesuriya; Morley D. Hollenberg

Human mast cell tryptase appears to display considerable variation in activating proteinase‐activated receptor 2 (PAR2). We found tryptase to be an inefficient activator of wild‐type rat‐PAR2 (wt‐rPAR2) and therefore decided to explore the factors that may influence tryptase activation of PAR2. Using a 20 mer peptide (P20) corresponding to the cleavage/activation sequence of wt‐rPAR2, tryptase was as efficient as trypsin in releasing the receptor‐activating sequence (SLIGRL…). However, in the presence of either human‐PAR2 or wt‐r PAR2 expressing cells, tryptase could only activate PAR2 by releasing SLIGRL from the P20 peptide, suggesting that PAR2 expressed on the cells was protected from tryptase activation. Three approaches were employed to test the hypothesis that PAR2 receptor glycosylation restricts tryptase activation. (a) pretreatment of wt‐rPAR2 expressing cells or human embryonic kidney cells (HEK293) with vibrio cholerae neuraminidase to remove oligosaccharide sialic acid, unmasked tryptase‐mediated PAR2 activation. (b) Inhibiting receptor glycosylation in HEK293 cells with tunicamycin enabled tryptase‐mediated PAR2 activation. (c) Wt‐rPAR2 devoid of the N‐terminal glycosylation sequon (PAR2T25−), but not rPAR2 devoid of the glycosylation sequon located on extracellular loop‐2 (PAR2T224A), was selectively and substantially (>30 fold) more sensitive to tryptase compared with the wt‐rPAR2. Immunocytochemistry using antisera that specifically recognized the N‐terminal precleavage sequence of PAR2 demonstrated that tryptase released the precleavage domain from PAR2T25− but not from wt‐rPAR2. Heparin : tryptase molar ratios of greater than 2 : 1 abrogated tryptase activation of PAR2T25−. Our results indicate that glycosylation of PAR2 and heparin‐inhibition of PAR2 activation by tryptase could provide novel mechanisms for regulating receptor activation by tryptase and possibly other proteases.


Bioscience Reports | 1985

The amino acid sequence of human sperm protamine P1

Don J. Mckay; Bernard Renaux; Gordon H. Dixon

Human sperm protamines have been extracted from spermatozoa pooled from several donors, converted to their S-pyridylethylated derivatives and resolved into two major components, P1 and PI, by Bio-Rex 70 chromatography. Protamine P1 was further purified by Bio-Gel P-10 chromatography and sequenced directly on a gas phase protein sequencer for 43 residues. To complete the sequence, P1 was cleaved at methionine 36 and the C-terminal tetradecapeptide was purified by h.p.i.c , and sequenced completely. The 50 residue sequence is: 10 20 30 40 ARYRC CRSQS RSRYY RQRQR SRRRR RRSCQ TRRRA MRCCR 50 PRYRP RCRRH. This sequence has a calculated molecular weight of 6674 and is homologous with four other published mammalian protamine sequences.


Journal of Biological Chemistry | 2011

Neutrophil Elastase Acts as a Biased Agonist for Proteinase-activated Receptor-2 (PAR2)

Koichiro Mihara; Hyunjae Chung; Bernard Renaux; Chang S. Lau; Daniel A. Muruve; Kathryn DeFea; Michel Bouvier; Morley D. Hollenberg

Human neutrophil proteinases (elastase, proteinase-3, and cathepsin-G) are released at sites of acute inflammation. We hypothesized that these inflammation-associated proteinases can affect cell signaling by targeting proteinase-activated receptor-2 (PAR2). The PAR family of G protein-coupled receptors is triggered by a unique mechanism involving the proteolytic unmasking of an N-terminal self-activating tethered ligand (TL). Proteinases can either activate PAR signaling by unmasking the TL sequence or disarm the receptor for subsequent enzyme activation by cleaving downstream from the TL sequence. We found that none of neutrophil elastase, cathepsin-G, and proteinase-3 can activate Gq-coupled PAR2 calcium signaling; but all of these proteinases can disarm PAR2, releasing the N-terminal TL sequence, thereby preventing Gq-coupled PAR2 signaling by trypsin. Interestingly, elastase (but neither cathepsin-G nor proteinase-3) causes a TL-independent PAR2-mediated activation of MAPK that, unlike the canonical trypsin activation, does not involve either receptor internalization or recruitment of β-arrestin. Cleavage of synthetic peptides derived from the extracellular N terminus of PAR2, downstream of the TL sequence, demonstrated distinct proteolytic sites for all three neutrophil-derived enzymes. We conclude that in inflammation, neutrophil proteinases can modulate PAR2 signaling by preventing/disarming the Gq/calcium signal pathway and, via elastase, can selectively activate the p44/42 MAPK pathway. Our data illustrate a new mode of PAR regulation that involves biased PAR2 signaling by neutrophil elastase and a disarming/silencing effect of cathepsin-G and proteinase-3.


Journal of Biological Chemistry | 2013

Neutrophil elastase and proteinase-3 trigger G-protein biased signaling through proteinase activated receptor-1 (PAR1)

Koichiro Mihara; Bernard Renaux; Mahmoud Saifeddine; Morley D. Hollenberg

Background: Proteinase-activated receptor-1 (PAR1) is a proteolytically activated G protein-coupled receptor. Neutrophil-derived enzymes might regulate PAR1 signaling. Results: Neutrophil elastase and proteinase-3 cleave and activate PAR1 signaling that is distinct from thrombin-triggered responses. Neutrophil elastase and proteinase-3 signaling through PAR1 modulates endothelial cell signaling. Conclusion: Neutrophil enzymes are Gαi-biased agonists for PAR1. Significance: Biased PAR1-activating compounds may prove of value as therapeutic agents to treat cardiovascular and inflammatory diseases. Neutrophil proteinases released at sites of inflammation can affect tissue function by either activating or disarming signal transduction mediated by proteinase-activated receptors (PARs). Because PAR1 is expressed at sites where abundant neutrophil infiltration occurs, we hypothesized that neutrophil-derived enzymes might also regulate PAR1 signaling. We report here that both neutrophil elastase and proteinase-3 cleave the human PAR1 N terminus at sites distinct from the thrombin cleavage site. This cleavage results in a disarming of thrombin-activated calcium signaling through PAR1. However, the distinct non-canonical tethered ligands unmasked by neutrophil elastase and proteinase-3, as well as synthetic peptides with sequences derived from these novel exposed tethered ligands, selectively stimulated PAR1-mediated mitogen-activated protein kinase activation. This signaling was blocked by pertussis toxin, implicating a Gαi-triggered signal pathway. We conclude that neutrophil proteinases trigger biased PAR1 signaling and we describe a novel set of tethered ligands that are distinct from the classical tethered ligand revealed by thrombin. We further demonstrate the function of this biased signaling in regulating endothelial cell barrier integrity.


Journal of Immunology | 2014

NADPH Oxidase Modifies Patterns of MHC Class II–Restricted Epitopic Repertoires through Redox Control of Antigen Processing

Euan R. O. Allan; Pankaj Tailor; Dale R. Balce; Payman Pirzadeh; Neil T. McKenna; Bernard Renaux; Amy L. Warren; Frank R. Jirik; Robin M. Yates

The chemistries within phagosomes of APCs mediate microbial destruction as well as generate peptides for presentation on MHC class II. The antimicrobial effector NADPH oxidase (NOX2), which generates superoxide within maturing phagosomes, has also been shown to regulate activities of cysteine cathepsins through modulation of the lumenal redox potential. Using real-time analyses of lumenal microenvironmental parameters, in conjunction with hydrolysis pattern assessment of phagocytosed proteins, we demonstrated that NOX2 activity not only affects levels of phagosomal proteolysis as previously shown, but also the pattern of proteolytic digestion. Additionally, it was found that NOX2 deficiency adversely affected the ability of bone marrow–derived macrophages, but not dendritic cells, to process and present the I-Ab–immunodominant peptide of the autoantigen myelin oligodendrocyte glycoprotein (MOG). Computational and experimental analyses indicated that the I-Ab binding region of the immunodominant peptide of MOG is susceptible to cleavage by the NOX2-controlled cysteine cathepsins L and S in a redox-dependent manner. Consistent with these findings, I-Ab mice that were deficient in the p47phox or gp91phox subunits of NOX2 were partially protected from MOG-induced experimental autoimmune encephalomyelitis and displayed compromised reactivation of MOG-specific CD4+ T cells in the CNS, despite eliciting a normal primary CD4+ T cell response to the inoculated MOG Ag. Taken together, this study demonstrates that the redox microenvironment within the phagosomes of APCs is a determinant in MHC class II repertoire production in a cell-specific and Ag-specific manner, which can ultimately impact susceptibility to CD4+ T cell–driven autoimmune disease processes.


Journal of Muscle Research and Cell Motility | 1994

Phosphorylation of caldesmon by smooth-muscle casein kinase II

Cindy Sutherland; Bernard Renaux; Don J. Mckay; Michael P. Walsh

SummaryA caldesmon kinase activity was partially purified from an extract of chicken gizzard smooth muscle by sequential chromatography on columns of DEAE-Sephacel, MonoQ and Superose 12. This kinase was identified as casein kinase II by Western blotting using peptide-directed antibodies raised against the α, α′ and β subunits of human casein kinase II; the smooth muscle enzyme consisted of similar subunits of Mr 43 000 (α), 39 000 (α′), and 27 000 (β). Phosphorylation of caldesmon and casein by smooth muscle casein kinase II was optimal at ∼0.1 M NaCl, did not require second messengers, and was inhibited by heparin. The kinase utilized either GTP or ATP as a substrate. Caldesmon was phosphorylated to ∼1 mol Pi mol-1 caldesmon by smooth muscle casein kinase II with a Km for caldesmon of 4.9 μM. Two-dimensional thin-layer electrophoresis indicated phosphate incorporation into both serine and threonine. All the incorporated phosphate was recovered in the N-terminal peptide (residues 1–152) generated by cleavage at cysteine 153 with 2-nitro-5-thiocyanobenzoic acid. Purification of tryptic phosphopeptides and N-terminal sequencing revealed two principal sites of phosphorylation: serine 73 and threonine 83. The following four synthetic peptides corresponding to this domain of caldesmon were examined as substrates of casein kinase II: A = RRREVNAQNSVAEEE; B = AQNSVAEEE; C = RSTDDEAA; D = SVAEEETKRSTDDE. Interestingly, only peptides C and D were phosphorylated and both only at threonine. Phosphorylation of intact caldesmon did not affect the pattern of chymotryptic digestion suggesting that it does not induce a significant conformational change in the protein substrate. Phosphorylation also had no effect on the binding of caldesmon to actin or on the caldesmon-mediated inhibition of actomyosin MgATPase activity. However, phosphorylation completely abolished the interaction of caldesmon with immobilized smooth muscle myosin. These results are consistent with the localization of the myosin-binding domain near the N-terminus of caldesmon and of the actin-binding domain near the opposite end of the elongated molecule. Casein kinase II may therefore play a role in regulating caldesmon-myosin interaction and the ability of caldesmon to cross-link actin and myosin filaments in smooth muscle.


Journal of Pharmacology and Experimental Therapeutics | 2008

Derivatized 2-Furoyl-LIGRLO-amide, a Versatile and Selective Probe for Proteinase-Activated Receptor 2. Binding and Visualization

Morley D. Hollenberg; Bernard Renaux; Eric Hyun; Steeve Houle; Nathalie Vergnolle; Mahmoud Saifeddine

The proteinase-activated receptor-2 (PAR2)-activating peptide with an N-terminal furoyl group modification, 2-furoyl-LIGRLO-NH2 (2fLI), was derivatized via its free ornithine amino group to yield [3H]propionyl-2fLI and Alexa Fluor 594-2fLI that were used as receptor probes for ligand binding assays and receptor visualization both for cultured cells in vitro and for colonic epithelial cells in vivo. The binding of the radiolabeled and fluorescent PAR2 probes was shown to be present in PAR2-transfected Kirsten normal rat kidney cells, but not in vector-alone-transfected cells, and was abolished by pretreatment of cells with saturating concentrations of receptor-selective PAR2 peptide agonists such as SLIGRL-NH2 and the parent agonist 2fLI but not by reverse-sequence peptides such as 2-furoyl-OLRGIL-NH2 that cannot activate PAR2. The relative orders of potencies for a series of PAR2 peptide agonists to compete for the binding of [3H]propionyl-2fLI (2fLI >> SLIGRL-NH2 ≅ trans-cinnamoyl-LIGRLO-NH2 > SLIGKV-NH2 > SLIGKT-NH2) mirrored qualitatively their relative potencies for PAR2-mediated calcium signaling in the same cells or for vasorelaxation in a rat aorta vascular assay. In the vascular assay, the potency of Alexa Fluor 594-2fLI was the same as 2fLI. We conclude that ornithine-derivatized 2fLI peptides are conveniently synthesized PAR2 probes that will be of value for future studies of receptor binding and visualization.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2009

Serine proteases decrease intestinal epithelial ion permeability by activation of protein kinase Cζ

Veronica Swystun; Bernard Renaux; Shoubin Wen; Michael A. Peplowski; Morley D. Hollenberg; Wallace K. MacNaughton

Epithelial permeability to ions and larger molecules in the gut is essential for fluid balance, and its dysregulation contributes to intestinal pathology. We investigated the effect of digestive serine proteases on epithelial paracellular permeability. Trypsin, chymotrypsin, and elastase elicited sustained increases in transepithelial resistance (R(TE)) in polarized monolayers of three intestinal epithelial cell lines. This effect was reflected by decreases in paracellular conductances of Na+ and Cl- and a concomitant decrease in permeability to 3,000 molecular weight dextran. The enzyme activities of the proteases were required, yet activators of known protease-activated receptors (PARs) did not reproduce the effect of these proteases on R(TE). PKCzeta isoform-specific inhibitor significantly reduced the trypsin-induced increase in R(TE) whereas PKCzeta activity was increased in cells treated with trypsin and chymotrypsin compared with control cells; this activity was reduced to control levels in the presence of PKCzeta-specific inhibitor. Ca2+ chelators and pharmacological inhibitors of cell signaling support the role for PKCzeta in the protease-induced effect. Finally, we showed that treatment with the serine proteases increased occludin immunostaining and zonula occludin-1 coimmunoprecipitation with occludin in the detergent-insoluble fraction of cell lysates, and these increases were ablated by pretreatment with PKCzeta-specific inhibitor. This finding indicates increased insertion of occludin into the cell junctional complex. These data demonstrate a role for serine proteases in the facilitation of epithelial barrier function through a mechanism that is independent of PARs and is mediated by activation of PKCzeta.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2012

Epidermal growth factor receptor transactivation is required for proteinase-activated receptor-2-induced COX-2 expression in intestinal epithelial cells

Christina Hirota; Vadim Iablokov; Michael Dicay; Bernard Renaux; Morley D. Hollenberg; Wallace K. MacNaughton

Proteinase-activated receptor (PAR)(2), a G protein-coupled receptor activated by serine proteinases, has been implicated in both intestinal inflammation and epithelial proliferation. Cyclooxygenase (COX)-2 is overexpressed in the gut during inflammation as well as in colon cancer. We hypothesized that PAR(2) drives COX-2 expression in intestinal epithelial cells. Treatment of Caco-2 colon cancer cells with the PAR(2)-activating peptide 2-furoyl-LIGRLO-NH(2) (2fLI), but not by its reverse-sequence PAR(2)-inactive peptide, for 3 h led to an increase in intracellular COX-2 protein expression accompanied by a COX-2-dependent increase in prostaglandin E(2) production. 2fLI treatment for 30 min significantly increased metalloproteinase activity in the culture supernatant. Increased epidermal growth factor receptor (EGFR) phosphorylation was observed in cell lysates following 40 min of treatment with 2fLI. The broad-spectrum metalloproteinase inhibitor marimastat inhibited both COX-2 expression and EGFR phosphorylation. The EGFR tyrosine kinase inhibitor PD153035 also abolished 2fLI-induced COX-2 expression. Although PAR(2) activation increased ERK MAPK phosphorylation, neither ERK pathway inhibitors nor a p38 MAPK inhibitor affected 2fLI-induced COX-2 expression. However, inhibition of either Src tyrosine kinase signaling by PP2, Rho kinase signaling by Y27632, or phosphatidylinositol 3 (PI3) kinase signaling by LY294002 prevented 2fLI-induced COX-2 expression. Trypsin increased COX-2 expression through PAR(2) in Caco-2 cells and in an EGFR-dependent manner in the noncancerous intestinal epithelial cell-6 cell line. In conclusion, PAR(2) activation drives COX-2 expression in Caco-2 cells via metalloproteinase-dependent EGFR transactivation and activation of Src, Rho, and PI3 kinase signaling. Our findings provide a mechanism whereby PAR(2) can participate in the progression from chronic inflammation to cancer in the intestine.


Biological Chemistry | 2008

Substrate specificity determination of mouse implantation serine proteinase and human kallikrein-related peptidase 6 by phage display*

Navneet Sharma; Katerina Oikonomopoulou; Kenichi Ito; Bernard Renaux; Eleftherios P. Diamandis; Morley D. Hollenberg; Derrick E. Rancourt

Abstract We constructed a random library of hexapeptides displayed on the surface of bacteriophage T7 to determine the substrate specificity of proteinases. The phage-displayed library was subjected to repeated rounds of biopanning with native implantation serine proteinase and recombinant human kallikrein-related peptidase 6 (KLK6) followed by selection and identification of putative substrates. For both enzymes, the results obtained demonstrate a preference for arginine and lysine at multiple positions in the recognition cleavage motif, confirming their previously reported trypsin-like substrate specificity. In the case of KLK6, there is also a pronounced presence of tryptophan within the cleaved peptide sequences, indicating its potential dual substrate specificity, acting as both a trypsin and chymotrypsin-like enzyme.

Collaboration


Dive into the Bernard Renaux'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