Network


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

Hotspot


Dive into the research topics where Josée Hamelin is active.

Publication


Featured researches published by Josée Hamelin.


Hepatology | 2008

Proprotein convertase subtilisin/kexin type 9 (PCSK9): Hepatocyte‐specific low‐density lipoprotein receptor degradation and critical role in mouse liver regeneration

Ahmed Zaid; Anna Roubtsova; Rachid Essalmani; Jadwiga Marcinkiewicz; Ann Chamberland; Josée Hamelin; Michel Tremblay; Hélène Jacques; Weijun Jin; Jean Davignon; Nabil G. Seidah; Annik Prat

The gene encoding the proprotein convertase subtilisin/kexin type 9 (PCSK9) is linked to familial hypercholesterolemia, as are those of the low‐density lipoprotein receptor (LDLR) and apolipoprotein B. PCSK9 enhances LDLR degradation, resulting in low‐density lipoprotein accumulation in plasma. To analyze the role of hepatic PCSK9, total and hepatocyte‐specific knockout mice were generated. They exhibit 42% and 27% less circulating cholesterol, respectively, showing that liver PCSK9 was responsible for two thirds of the phenotype. We also demonstrated that, in liver, PCSK9 is exclusively expressed in hepatocytes, representing the main source of circulating PCSK9. The data suggest that local but not circulating PCSK9 regulates cholesterol levels. Although transgenic mice overexpressing high levels of liver and circulating PCSK9 led to the almost complete disappearance of the hepatic LDLR, they did not recapitulate the plasma cholesterol levels observed in LDLR‐deficient mice. Single LDLR or double LDLR/PCSK9 knockout mice exhibited similar cholesterol profiles, indicating that PCSK9 regulates cholesterol homeostasis exclusively through the LDLR. Finally, the regenerating liver of PCSK9‐deficient mice exhibited necrotic lesions, which were prevented by a high‐cholesterol diet. However, lipid accumulation in hepatocytes of these mice was markedly reduced under both chow and high‐cholesterol diets, revealing that PCSK9 deficiency confers resistance to liver steatosis. Conclusion: Although PCSK9 is a target for controlling hypercholesterolemia, our data indicate that upon hepatic damage, patients lacking PCSK9 could be at risk. (HEPATOLOGY 2008;48:646–554.)


Journal of Biological Chemistry | 2006

The Proprotein Convertase (PC) PCSK9 Is Inactivated by Furin and/or PC5/6A FUNCTIONAL CONSEQUENCES OF NATURAL MUTATIONS AND POST-TRANSLATIONAL MODIFICATIONS

Suzanne Benjannet; David Rhainds; Josée Hamelin; Nasha Nassoury; Nabil G. Seidah

PCSK9 is the ninth member of the proprotein convertase (PC) family. Some of its natural mutations have been genetically associated with the development of a dominant form of familial hyper- or hypocholesterolemia. The exact mechanism of action of PCSK9 is not clear, although it is known to enhance the intracellular degradation of the low density lipoprotein (LDL) receptor in acidic compartments, likely the endosomes/lysosomes. We analyzed the post-translational modifications of PCSK9 and show that it is sulfated within its prosegment at Tyr38. We also examined the susceptibility of PCSK9 to proteolytic cleavage by the other members of the PC family. The data show that the natural gain-of-function mutations R218S, F216L, and D374Y associated with hypercholesterolemia result in total or partial loss of furin/PC5/6A processing at the motif RFHR218↓. In contrast, the loss-of-function mutations A443T and C679X lead either to the lack of trans-Golgi network/recycling endosome localization and an enhanced susceptibility to furin cleavage (A443T) or to the inability of PCSK9 to exit the endoplasmic reticulum (C679X). Furthermore, we report the presence of both native and furin-like cleaved forms of PCSK9 in circulating human plasma. Thus, we propose that PCSK9 levels are finely regulated by the basic amino acid convertases furin and PC5/6A. The latter may reduce the lifetime of this proteinase and its ability to degrade the cell-surface LDL receptor, thereby regulating the levels of circulating LDL cholesterol.


Traffic | 2007

The Cellular Trafficking of the Secretory Proprotein Convertase PCSK9 and Its Dependence on the LDLR

Nasha Nassoury; Daniel A. Blasiole; Angie T. Oler; Suzanne Benjannet; Josée Hamelin; Vivianne Poupon; Peter S. McPherson; Alan D. Attie; Annik Prat; Nabil G. Seidah

Mutations in the proprotein convertase PCSK9 gene are associated with autosomal dominant familial hyper‐ or hypocholesterolemia. These phenotypes are caused by a gain or loss of function of proprotein convertase subtilisin kexin 9 (PCSK9) to elicit the degradation of the low‐density lipoprotein receptor (LDLR) protein. Herein, we asked whether the subcellular localization of wild‐type PCSK9 or mutants of PCSK9 and the LDLR would provide insight into the mechanism of PCSK9‐dependent LDLR degradation. We show that the LDLR is the dominant partner in regulating the cellular trafficking of PCSK9. In cells lacking the LDLR, PCSK9 localized in the endoplasmic reticulum (ER). In cells expressing the LDLR, PCSK9 sorted to post‐ER compartments (i.e. endosomes in cell lines and Golgi apparatus in primary hepatocytes), where it colocalized with the LDLR. In cell lines, PCSK9 also colocalized with the LDLR at the cell surface, requiring the presence of the C‐terminal Cys/His‐rich domain of PCSK9. We provide evidence that PCSK9 promotes the degradation of the LDLR by an endocytic mechanism, as small interfering RNA‐mediated knockdown of the clathrin heavy chain reduced the functional activity of PCSK9. We also compared the subcellular localization of natural mutants of PCSK9 with that of the wild‐type enzyme in human hepatic (HuH7) cells. Whereas the mutants associated with hypercholesterolemia (S127R, F216L and R218S) localized to endosomes/lysosomes, those associated with hypocholesterolemia did not reach this compartment. We conclude that the sorting of PCSK9 to the cell surface and endosomes is required for PCSK9 to fully promote LDLR degradation and that retention in the ER prevents this activity. Mutations that affect this transport can lead to hyper‐ or hypocholesterolemia.


Journal of Lipid Research | 2010

A new method for measurement of total plasma PCSK9: clinical applications

Geneviève Dubuc; Michel J. Tremblay; Guillaume Paré; Hélène Jacques; Josée Hamelin; Suzanne Benjannet; Lucie Boulet; Jacques Genest; Lise Bernier; Nabil G. Seidah; Jean Davignon

The proprotein convertase subtilisin kexin-9 (PCSK9) circulates in plasma as mature and furin-cleaved forms. A polyclonal antibody against human PCSK9 was used to develop an ELISA that measures total plasma PCSK9 rather than only the mature form. A cross-sectional study evaluated plasma levels in normal (n = 254) and hypercholesterolemic (n = 200) subjects treated or untreated with statins or statin plus ezetimibe. In controls, mean plasma PCSK9 (89.5 ± 31.9 ng/ml) correlated positively with age, total cholesterol, LDL-cholesterol (LDL-C), triglycerides, and fasting glucose. Sequencing PCSK9 from individuals at the extremes of the normal PCSK9 distribution identified a new loss-of-function R434W variant associated with lower levels of circulating PCSK9 and LDL-C. In hypercholesterolemic subjects, PCSK9 levels were higher than in controls (99.3 ± 31.7 ng/ml, P < 0.04) and increased in proportion to the statin dose, combined or not with ezetimibe. In treated patients (n = 139), those with familial hypercholesterolemia (FH; due to LDL receptor gene mutations) had higher PCSK9 values than non-FH (147.01 ± 42.5 vs. 127.2 ± 40.8 ng/ml, P < 0.005), but LDL-C reduction correlated positively with achieved plasma PCSK9 levels to a similar extent in both subsets (r = 0.316, P < 0.02 in FH and r = 0.275, P < 0.009 in non-FH). The detection of circulating PCSK9 in both FH and non-FH subjects means that this assay could be used to monitor response to therapy in a wide range of patients.


Annals of the New York Academy of Sciences | 2006

The Subtilisin/Kexin Family of Precursor Convertases: Emphasis on PC1, PC2/7B2, POMC and the Novel Enzyme SKI-1

Nabil G. Seidah; Suzanne Benjannet; Josée Hamelin; Aida M. Mamarbachi; Ajoy Basak; Jadwiga Marcinkiewicz; Majambu Mbikay; Michel Chrétien; Mieczyslaw Marcinkiewicz

ABSTRACT: Proopiomelanocortin (POMC) is a precursor to various, bioactive peptides including ACTH, βLPH, αMSH, and βendorphin (βEND). Processing of POMC at dibasic residues is tissue‐specific and is performed by either PC1 alone (resulting in ACTH and βLPH, anterior pituitary corticotrophes) or by a combination of PC1 and PC2 (yielding αMSH and βEND, pituitary neurointermediate lobe and hypothalamus). The PC2‐specific binding protein 7B2 is intimately involved in the zymogen activation of proPC2 into PC2. Structure‐function studies of these enzymes demonstrated the presence of N‐ and C‐terminal domains, as well as specific amino acids within the catalytic segment that influence the degree of activity of each enzyme and the interaction of PC2 with 7B2. The tissue distribution, plasticity of expression, and the multiple precursors that are differentially cleaved by PC1 and/or PC2, predict a wide array of combinatorial activities of these convertases within the endocrine and neuroendocrine system. The phenotypic consequences of the absence of genetic expression of either PC1 or PC2 are now explored using knockout mice and in human patients suffering from obesity and diabetes.


Journal of Biological Chemistry | 2008

Inhibition of Chikungunya Virus Infection in Cultured Human Muscle Cells by Furin Inhibitors IMPAIRMENT OF THE MATURATION OF THE E2 SURFACE GLYCOPROTEIN

Simona Ozden; Marianne Lucas-Hourani; Pierre-Emmanuel Ceccaldi; Ajoy Basak; Menogh Valentine; Suzanne Benjannet; Josée Hamelin; Yves Jacob; Kamel Mamchaoui; Vincent Mouly; Philippe Desprès; Antoine Gessain; Gillian Butler-Browne; Michel Chrétien; Frédéric Tangy; Pierre-Olivier Vidalain; Nabil G. Seidah

Chikungunya virus (CHIKV) is a mosquito-transmitted Alphavirus that causes in humans an acute infection characterized by polyarthralgia, fever, myalgia, and headache. Since 2005 this virus has been responsible for an epidemic outbreak of unprecedented magnitude. By analogy with other alphaviruses, it is thought that cellular proteases are able to process the viral precursor protein E3E2 to produce the receptor-binding E2 protein that associates as a heterodimer with E1. Destabilization of the heterodimer by exposure to low pH allows viral fusion and infection. We show that among a large panel of proprotein convertases, membranous furin but also PC5B can process E3E2 from African CHIKV strains at the HRQRR64↓ST site, whereas a CHIKV strain of Asian origin is cleaved at RRQRR64↓SI by membranous and soluble furin, PC5A, PC5B, and PACE4 but not by PC7 or SKI-1. Using fluorogenic model peptides and recombinant convertases, we observed that the Asian strain E3E2 model peptide is cleaved most efficiently by furin and PC5A. This cleavage was also observed in CHIKV-infected cells and could be blocked by furin inhibitor decanoyl-RVKR-chloromethyl ketone. This inhibitor was compared with chloroquine for its ability to inhibit CHIKV spreading in myoblast cell cultures, a cell-type previously described as a natural target of this virus. Our results demonstrate the role of furin-like proteases in the processing of CHIKV particles and point out new approaches to inhibit this infection.


Journal of Biological Chemistry | 2008

The Regulated Cell Surface Zymogen Activation of the Proprotein Convertase PC5A Directs the Processing of Its Secretory Substrates

Gaétan Mayer; Josée Hamelin; Marie-Claude Asselin; Antonella Pasquato; Edwidge Marcinkiewicz; Meiyi Tang; Siamak Tabibzadeh; Nabil G. Seidah

The proprotein convertases are synthesized as zymogens that acquire activity upon autocatalytic removal of their NH2-terminal prosegment. Based on the convertase furin, to fold properly and gain activity, the convertases PC5A, PACE4, and PC7 are presumed to undergo two sequential prosegment cleavages in the endoplasmic reticulum and then in the trans-Golgi network. However, biochemical and immunocytochemical experiments revealed that mouse PC5A is complexed to its prosegment at the plasma membrane. This labeling is lost upon treatment with heparin and is increased by overexpressing members of the syndecan family and CD44, suggesting attachment of secreted PC5A-prosegment complex to heparan sulfate proteoglycans. Following stimulation of Y1 cells with adrenocorticotropic hormone or 8-bromo-cyclic AMP, the cell surface labeling of the prosegment of PC5A is greatly diminished, whereas the signal for mature PC5A is increased. Moreover, after stimulation, the protease activity of PC5A is enhanced, as evidenced by the cleavage of the PC5A substrates Lefty, ADAMTS-4, endothelial lipase, and PCSK9. Our data suggest a novel mechanism for PC5A activation and substrate cleavage at the cell surface, through a regulated removal of its prosegment. A similar mechanism may also apply to the convertase PACE4, thereby extending our knowledge of the molecular details of the zymogen activation and functions of these heparan sulfate proteoglycan-bound convertases.


Molecular and Cellular Biology | 2006

Deletion of the Gene Encoding Proprotein Convertase 5/6 Causes Early Embryonic Lethality in the Mouse

Rachid Essalmani; Josée Hamelin; Jadwiga Marcinkiewicz; Ann Chamberland; Majambu Mbikay; Michel Chrétien; Nabil G. Seidah; Annik Prat

ABSTRACT PC5 belongs to the proprotein convertase family and activates precursor proteins by cleavage at basic sites during their transit through the secretory pathway and/or at the cell surface. These precursors include prohormones, proreceptors, growth factors, adhesion molecules, and viral glycoproteins. The Pcsk5 gene encodes two alternatively spliced isoforms, the soluble PC5A and transmembrane PC5B. We have carefully analyzed the expression of PC5 in the mouse during development and in adulthood by in situ hybridization, as well as in mouse tissues and various cell lines by quantitative reverse transcription-PCR. The data show that adrenal cortex and intestine are the richest sources of PC5A and PC5B, respectively. To better define the specific physiological roles of PC5, we have generated a mouse Pcsk5 Δ4 -deficient allele missing exon 4 that encodes the catalytic Asp173. While Δ4/+ heterozygotes were healthy and fertile, genotyping of progeny obtained from Δ4/+ interbreeding indicated that Δ4/Δ4 embryos died between embryonic days 4.5 and 7.5. These data demonstrate that Pcsk5 is an essential gene.


Journal of Biological Chemistry | 1997

In vitro characterization of the novel proprotein convertase PC7.

Jon Scott Munzer; Ajoy Basak; Mei Zhong; Aida M. Mamarbachi; Josée Hamelin; Diane Savaria; Claude Lazure; Suzanne Benjannet; Michel Chrétien; Nabil G. Seidah

Biochemical and enzymatic characterization of the novel proprotein convertase rat PC7 (rPC7) was carried out using vaccinia virus recombinants overexpressed in mammalian BSC40 cells. Pro-PC7 is synthesized as a glycosylated zymogen (101 kDa) and processed into mature rPC7 (89 kDa) in the endoplasmic reticulum. No endogenously produced soluble forms of this membrane-anchored protein were detected. A deletion mutant (65 kDa), truncated well beyond the expected C-terminal boundary of the P-domain, produced soluble rPC7 in the culture medium. Enzymatic activity assays of rPC7 using fluorogenic peptidyl substrates indicated that the pH optimum, Ca2+ dependence, and cleavage specificity of this enzyme are largely similar to those of furin. However, with some substrates, cleavage specificity more closely resembled that of yeast kexin, suggesting differential processing of proprotein substrates by this novel convertase. We examined the rPC7- and human furin-mediated cleavage of synthetic peptides containing the processing sites of three proteins known to colocalize in situ with rPC7. Whereas both enzymes correctly processed the pro-parathyroid hormone tridecapeptide and the pro-PC4 heptadecapeptide, neither enzyme cleaved a pro-epidermal growth factor hexadecapeptide. Thus, this study establishes that rPC7 is an enzymatically functional subtilisin/kexin-like serine proteinase with a cleavage specificity resembling that of hfurin. In addition, we have demonstrated that rPC7 can correctly process peptide precursors that contain the processing sites of at least two potential physiological substrates.


FEBS Letters | 1987

Chromogranin a can act as a reversible processing enzyme inhibitor: evidence from the inhibition of the IRCM-serine protease 1 cleavage of pro-enkephalin and ACTH at pairs of basic amino acids

Nabil G. Seidah; G.N. Hendy; Josée Hamelin; J. Paquin; Claude Lazure; K.M. Metters; J. Rossier; Michel Chrétien

Bovine parathyroid chromogranin A inhibits the cleavage of Z‐Ala‐Lys‐Arg‐AMC by either trypsin or IRCM‐serine protease 1 (IRCM‐SP1), a putative novel processing enzyme originally isolated from porcine pituitary anterior and neurointermediate lobes. On larger substrates, chromogranin A is a reversible competitive inhibitor of the cleavage at pairs of basic amino acids by IRCM‐SP1. The substrates tested included pituitary ACTH and adrenal medulla pro‐enkephalin‐derived peptides such as the 8.6 kDa synenkephalincontaining precursor and peptide B. Chromogranin A is itself selectively processed by IRCM‐SP1, and ACTH was shown to compete for such cleavage. These data suggest that chromogranins as a class of acidic proteins could participate in the tissue‐specific processing of pro‐hormones.

Collaboration


Dive into the Josée Hamelin's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michel Chrétien

Ottawa Hospital Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Annik Prat

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Claude Lazure

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Majambu Mbikay

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar

Ajoy Basak

Ottawa Hospital Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge