Network


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

Hotspot


Dive into the research topics where Roxane Desjardins is active.

Publication


Featured researches published by Roxane Desjardins.


Journal of Biological Chemistry | 2009

Dissection of the Endogenous Cellular Pathways of PCSK9-induced Low Density Lipoprotein Receptor Degradation EVIDENCE FOR AN INTRACELLULAR ROUTE

Steve Poirier; Gaétan Mayer; Viviane Poupon; Peter S. McPherson; Roxane Desjardins; Kévin Ly; Marie-Claude Asselin; Robert W. Day; Franck Duclos; Mark R. Witmer; Rex A. Parker; Annik Prat; Nabil G. Seidah

Elevated levels of plasma low density lipoprotein (LDL)-cholesterol, leading to familial hypercholesterolemia, are enhanced by mutations in at least three major genes, the LDL receptor (LDLR), its ligand apolipoprotein B, and the proprotein convertase PCSK9. Single point mutations in PCSK9 are associated with either hyper- or hypocholesterolemia. Accordingly, PCSK9 is an attractive target for treatment of dyslipidemia. PCSK9 binds the epidermal growth factor domain A (EGF-A) of the LDLR and directs it to endosomes/lysosomes for destruction. Although the mechanism by which PCSK9 regulates LDLR degradation is not fully resolved, it seems to involve both intracellular and extracellular pathways. Here, we show that clathrin light chain small interfering RNAs that block intracellular trafficking from the trans-Golgi network to lysosomes rapidly increased LDLR levels within HepG2 cells in a PCSK9-dependent fashion without affecting the ability of exogenous PCSK9 to enhance LDLR degradation. In contrast, blocking the extracellular LDLR endocytosis/degradation pathway by a 4-, 6-, or 24-h incubation of cells with Dynasore or an EGF-AB peptide or by knockdown of endogenous autosomal recessive hypercholesterolemia did not significantly affect LDLR levels. The present data from HepG2 cells and mouse primary hepatocytes favor a model whereby depending on the dose and/or incubation period, endogenous PCSK9 enhances the degradation of the LDLR both extra- and intracellularly. Therefore, targeting either pathway, or both, would be an effective method to reduce PCSK9 activity in the treatment of hypercholesterolemia and coronary heart disease.


Journal of Biological Chemistry | 2008

Substrate cleavage analysis of furin and related proprotein convertases: A comparative study

Albert G. Remacle; Sergey A. Shiryaev; Eok-Soo Oh; Piotr Cieplak; Anupama Srinivasan; Ge Wei; Robert C. Liddington; Boris I. Ratnikov; Amelie Parent; Roxane Desjardins; Robert W. Day; Jeffrey W. Smith; Michal Lebl; Alex Y. Strongin

We present the data and the technology, a combination of which allows us to determine the identity of proprotein convertases (PCs) related to the processing of specific protein targets including viral and bacterial pathogens. Our results, which support and extend the data of other laboratories, are required for the design of effective inhibitors of PCs because, in general, an inhibitor design starts with a specific substrate. Seven proteinases of the human PC family cleave the multibasic motifs R-X-(R/K/X)-R↓ and, as a result, transform proproteins, including those from pathogens, into biologically active proteins and peptides. The precise cleavage preferences of PCs have not been known in sufficient detail; hence we were unable to determine the relative importance of the individual PCs in infectious diseases, thus making the design of specific inhibitors exceedingly difficult. To determine the cleavage preferences of PCs in more detail, we evaluated the relative efficiency of furin, PC2, PC4, PC5/6, PC7, and PACE4 in cleaving over 100 decapeptide sequences representing the R-X-(R/K/X)-R↓ motifs of human, bacterial, and viral proteins. Our computer analysis of the data and the follow-on cleavage analysis of the selected full-length proteins corroborated our initial results thus allowing us to determine the cleavage preferences of the PCs and to suggest which PCs are promising drug targets in infectious diseases. Our results also suggest that pathogens, including anthrax PA83 and the avian influenza A H5N1 (bird flu) hemagglutinin precursor, evolved to be as sensitive to PC proteolysis as the most sensitive normal human proteins.


Journal of Biological Chemistry | 2007

Targeting host cell furin proprotein convertases as a therapeutic strategy against bacterial toxins and viral pathogens.

Sergey A. Shiryaev; Albert G. Remacle; Boris I. Ratnikov; Nicholas A. Nelson; Alexei Y. Savinov; Ge Wei; Massimo Bottini; Michele F. Rega; Amelie Parent; Roxane Desjardins; Martin Fugère; Robert W. Day; Mojgan Sabet; Maurizio Pellecchia; Robert C. Liddington; Jeffrey W. Smith; Tomas Mustelin; Donald G. Guiney; Michal Lebl; Alex Y. Strongin

Pathogens or their toxins, including influenza virus, Pseudomonas, and anthrax toxins, require processing by host proprotein convertases (PCs) to enter host cells and to cause disease. Conversely, inhibiting PCs is likely to protect host cells from multiple furin-dependent, but otherwise unrelated, pathogens. To determine if this concept is correct, we designed specific nanomolar inhibitors of PCs modeled from the extended cleavage motif TPQRERRRKKR↓GL of the avian influenza H5N1 hemagglutinin. We then confirmed the efficacy of the inhibitory peptides in vitro against the fluorescent peptide, anthrax protective antigen (PA83), and influenza hemagglutinin substrates and also in mice in vivo against two unrelated toxins, anthrax and Pseudomonas exotoxin. Peptides with Phe/Tyr at P1′ were more selective for furin. Peptides with P1′ Thr were potent against multiple PCs. Our strategy of basing the peptide sequence on a furin cleavage motif known for an avian flu virus shows the power of starting inhibitor design with a known substrate. Our results confirm that inhibiting furin-like PCs protects the host from the distinct furin-dependent infections and lay a foundation for novel, host cell-focused therapies against acute diseases.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Disruption of the expression of the proprotein convertase PC7 reduces BDNF production and affects learning and memory in mice.

William C. Wetsel; Ramona M. Rodriguiz; Johann Guillemot; Estelle Rousselet; Rachid Essalmani; Il Hwan Kim; Jesse C. Bryant; Jadwiga Marcinkiewicz; Roxane Desjardins; Robert Day; Daniel B. Constam; Annik Prat; Nabil G. Seidah

Significance The 7-membered proprotein convertase (PC) family of basic amino acid-specific proteases is implicated in the cleavage-activation of secretory precursor proteins. Although the in vivo functions of most members have been well studied, those of the seventh member PC7 are largely unknown. Herein, we find that PC7 participates in the generation of mature BDNF both in cells and in specific brain areas such as hippocampus and amygdala. Indeed, PC7 exerts a critical role in the brain: The results of various behavioral tests in normal mice compared with those lacking PC7 expression support an in vivo role for PC7 in the regulation of certain types of cognitive performance, such as emotional memories, in part via BDNF activation. PC7 belongs to the proprotein convertase family, whose members are implicated in the cleavage of secretory precursors. The in vivo function of PC7 is unknown. Herein, we find that the precursor proBDNF is processed into mature BDNF in COS-1 cells coexpressing proBDNF with either PC7 or Furin. Conversely, the processing of proBDNF into BDNF is markedly reduced in the absence of either Furin or PC7 in mouse primary hepatocytes. In vivo we observe that BDNF and PC7 mRNAs are colocalized in mouse hippocampus and amygdala and that mature BDNF protein levels are reduced in these brain areas in PC7 KO mice but not in the hippocampus of PC1/3 KO mice. Various behavioral tests reveal that in PC7 KO mice spatial memory is intact and plasticity of responding is mildly abnormal. Episodic and emotional memories are severely impaired, but both are rescued with the tyrosine receptor kinase B agonist 7,8-dihydroxyflavone. Altogether, these results support an in vivo role for PC7 in the regulation of certain types of cognitive performance, in part via proBDNF processing. Because polymorphic variants of human PC7 are being characterized, it will be important in future studies to determine their effects on additional physiological and behavioral processes.


PLOS ONE | 2009

Autocatalytic Activation of the Furin Zymogen Requires Removal of the Emerging Enzyme's N-Terminus from the Active Site

Katarzyna Gawlik; Sergey A. Shiryaev; Wenhong Zhu; Khatereh Motamedchaboki; Roxane Desjardins; Robert Day; Albert G. Remacle; Boguslaw Stec; Alex Y. Strongin

Background Before furin can act on protein substrates, it must go through an ordered process of activation. Similar to many other proteinases, furin is synthesized as a zymogen (profurin) which becomes active only after the autocatalytic removal of its auto-inhibitory prodomain. We hypothesized that to activate profurin its prodomain had to be removed and, in addition, the emerging enzymes N-terminus had to be ejected from the catalytic cleft. Methodology/Principal Findings We constructed and analyzed the profurin mutants in which the egress of the emerging enzymes N-terminus from the catalytic cleft was restricted. Mutants were autocatalytically processed at only the primary cleavage site Arg-Thr-Lys-Arg107↓Asp108, but not at both the primary and the secondary (Arg-Gly-Val-Thr-Lys-Arg75↓Ser76) cleavage sites, yielding, as a result, the full-length prodomain and mature furins commencing from the N-terminal Asp108. These correctly processed furin mutants, however, remained self-inhibited by the constrained N-terminal sequence which continuously occupied the S′ sub-sites of the catalytic cleft and interfered with the functional activity. Further, using the in vitro cleavage of the purified prodomain and the analyses of colon carcinoma LoVo cells with the reconstituted expression of the wild-type and mutant furins, we demonstrated that a three-step autocatalytic processing including the cleavage of the prodomain at the previously unidentified Arg-Leu-Gln-Arg89↓Glu90 site, is required for the efficient activation of furin. Conclusions/Significance Collectively, our results show the restrictive role of the enzymes N-terminal region in the autocatalytic activation mechanisms. In a conceptual form, our data apply not only to profurin alone but also to a range of self-activated proteinases.


Journal of Medicinal Chemistry | 2012

The Multi-Leu Peptide Inhibitor Discriminates Between PACE4 and Furin And Exhibits Antiproliferative Effects On Prostate Cancer Cells

Christine Levesque; Martin Fugère; Anna Kwiatkowska; Frédéric Couture; Roxane Desjardins; Sophie Routhier; Philippe Moussette; Adam Prahl; Bernard Lammek; Jon R. Appel; Richard A. Houghten; François D’Anjou; Yves L. Dory; Witold Neugebauer; Robert Day

The proprotein convertases (PCs) play an important role in protein precursor activation through processing at paired basic residues. However, significant substrate cleavage redundancy has been reported between PCs. The question remains whether specific PC inhibitors can be designed. This study describes the identification of the sequence LLLLRVKR, named Multi-Leu (ML)-peptide, that displayed a 20-fold selectivity on PACE4 over furin, two enzymes with similar structural characteristics. We have previously demonstrated that PACE4 plays an important role in prostate cancer and could be a druggable target. The present study demonstrates that the ML-peptide significantly reduced the proliferation of DU145 and LNCaP prostate cancer-derived cell lines and induced G0/G1 cell cycle arrest. However, the ML-peptide must enter the cell to inhibit proliferation. It is concluded that peptide-based inhibitors can yield specific PC inhibitors and that the ML-peptide is an important lead compound that could potentially have applications in prostate cancer.


The International Journal of Biochemistry & Cell Biology | 2010

Selective and potent furin inhibitors protect cells from anthrax without significant toxicity

Albert G. Remacle; Katarzyna Gawlik; Vladislav S. Golubkov; Gregory W. Cadwell; Robert C. Liddington; Piotr Cieplak; Sherri Z. Millis; Roxane Desjardins; Sophie Routhier; Xue Wen Yuan; Witold Neugebauer; Robert Day; Alex Y. Strongin

Furin and related proprotein convertases cleave the multibasic motifs R-X-R/K/X-R in the precursor proteins and, as a result, transform the latent proproteins into biologically active proteins and peptides. Furin is present both in the intracellular secretory pathway and at the cell surface. Intracellular furin processes its multiple normal cellular targets in the Golgi and secretory vesicle compartments while cell-surface furin appears to be essential only for the processing of certain pathogenic proteins and, importantly, anthrax. To design potent, safe and selective inhibitors of furin, we evaluated the potency and selectivity of the derivatized peptidic inhibitors modeled from the extended furin cleavage sequence of avian influenza A H5N1. We determined that the N- and C-terminal modifications of the original RARRRKKRT inhibitory scaffold produced selective and potent, nanomolar range, inhibitors of furin. These inhibitors did not interfere with the normal cellular function of furin because of the likely functional redundancy existing between furin and other proprotein convertases. These furin inhibitors, however, were highly potent in blocking the furin-dependent cell-surface processing of anthrax protective antigen-83 both in vitro and cell-based assays and in vivo. We conclude that the inhibitors we have designed have a promising potential as selective anthrax inhibitors, without affecting major cell functions.


Journal of Biological Chemistry | 2012

Disruption of Proprotein Convertase 1/3 (PC1/3) Expression in Mice Causes Innate Immune Defects and Uncontrolled Cytokine Secretion

Sarah Refaie; Sandra Gagnon; Hugo Gagnon; Roxane Desjardins; François D'Anjou; Pedro D'Orléans-Juste; Xiaorong Zhu; Donald F. Steiner; Nabil G. Seidah; Claude Lazure; Michel Salzet; Robert Day

Background: PC1/3 is known for its role in neuroendocrine cells but not for its potential role in innate immunity. Results: PC1/3 knock-out mice express a dysfunctional phenotype characterized by uncontrolled cytokine secretion when challenged with lipopolysaccharide. Conclusion: PC1/3 regulates cytokine secretion in macrophages. Significance: Identifying the role of PC1/3 in macrophages will lead to a better understanding of cytokine regulation and innate immunity. The proprotein convertase 1/3 is expressed in the regulated secretory pathway of neural and endocrine cells. Its major function is in the post-translational processing and activation of precursor proteins. The PC1/3 knock-out (KO) mouse model has allowed us to elucidate its physiological functions in studies focused primarily on neuroendocrine tissues. However, PC1/3 is also expressed in cells of the immune system, mainly in macrophages. The present study explores the effects of innate immune challenge in the PC1/3 KO mouse. PC1/3 KO mice have an enlarged spleen with marked disorganization of the marginal zone and red pulp. Immunohistochemical studies using various markers demonstrate a depletion of dendritic cells in PC1/3 KO spleens. When challenged with lipopolysaccharide, PC1/3 KO mice are more susceptible to septic shock than wild-type controls or other PC KO mice, such as PC2 and PC7 null mice. Plasma levels of proinflammatory cytokines (IL-6, IL-1β, and TNF-α) were very significantly elevated in PC1/3 KO mice, consistent with a hypercytokinemia, i.e. indicative of a major systemic uncontrolled inflammatory response or cytokine storm. Peritoneal macrophages isolated from PC1/3 KO mice also demonstrate elevated cytokine secretion when treated with LPS. Electron micrographs show morphological features indicating a prolonged activation of these cells following LPS stimulation. We also present evidence that the proinflammatory Th1 pathway is dominant in the PC1/3 KO mouse model. We conclude that aside from its important role in neuroendocrine functions PC1/3 also has an important role in the regulation of the innate immune system, most likely through the regulation of cytokine secretion in macrophages.


Journal of Medicinal Chemistry | 2014

Optimization of furin inhibitors to protect against the activation of influenza hemagglutinin H5 and Shiga toxin.

Gagnon H; Sophie Beauchemin; Anna Kwiatkowska; Frédéric Couture; D'Anjou F; Christine Levesque; Dufour F; Desbiens Ar; Vaillancourt R; Bernard S; Roxane Desjardins; Malouin F; Yves L. Dory; Robert W. Day

Proprotein convertases (PCs) are crucial in the processing and entry of viral or bacterial protein precursors and confer increased infectivity of pathogens bearing a PC activation site, which results in increased symptom severity and lethality. Previously, we developed a nanomolar peptide inhibitor of PCs to prevent PC activation of infectious agents. Herein, we describe a peptidomimetic approach that increases the stability of this inhibitor for use in vivo to prevent systemic infections and cellular damage, such as that caused by influenza H5N1 and Shiga toxin. The addition of azaβ(3)-amino acids to both termini of the peptide successfully prevented influenza hemagglutinin 5 fusogenicity and Shiga toxin Vero toxicity in cell-based assays. The results from a cell-based model using stable shRNA-induced proprotein convertase knockdown indicate that only furin is the major proprotein convertase required for HA5 cleavage.


Journal of Medicinal Chemistry | 2014

Design, Synthesis, and Structure−Activity Relationship Studies of a Potent PACE4 Inhibitor

Anna Kwiatkowska; Frédéric Couture; Christine Levesque; Kévin Ly; Roxane Desjardins; Sophie Beauchemin; Adam Prahl; Bernard Lammek; Witold Neugebauer; Yves L. Dory; Robert W. Day

PACE4 plays an important role in the progression of prostate cancer and is an attractive target for the development of novel inhibitor-based tumor therapies. We previously reported the design and synthesis of a novel, potent, and relatively selective PACE4 inhibitor known as a Multi-Leu (ML) peptide. In the present work, we examined the ML peptide through detailed structure-activity relationship studies. A variety of ML-peptide analogues modified at the P8-P5 positions with leucine isomers (Nle, DLeu, and DNle) or substituted at the P1 position with arginine mimetics were tested for their inhibitory activity, specificity, stability, and antiproliferative effect. By incorporating d isomers at the P8 position or a decarboxylated arginine mimetic, we obtained analogues with an improved stability profile and excellent antiproliferative properties. The DLeu or DNle residue also has improved specificity toward PACE4, whereas specificity was reduced for a peptide modified with the arginine mimetic, such as 4-amidinobenzylamide.

Collaboration


Dive into the Roxane Desjardins's collaboration.

Top Co-Authors

Avatar

Robert Day

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kévin Ly

Université de Sherbrooke

View shared research outputs
Top Co-Authors

Avatar

Yves L. Dory

Université de Sherbrooke

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge