Network


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

Hotspot


Dive into the research topics where Etienne Lemieux is active.

Publication


Featured researches published by Etienne Lemieux.


International Journal of Cancer | 2009

Constitutively active MEK1 is sufficient to induce epithelial-to-mesenchymal transition in intestinal epithelial cells and to promote tumor invasion and metastasis.

Etienne Lemieux; Sébastien Bergeron; Véronique Durand; Claude Asselin; Caroline Saucier; Nathalie Rivard

Constitutive activation of the MAP kinase kinase MEK1 induces oncogenic transformation in intestinal epithelial cells. Loss of cell–cell adhesion followed by the dissociation of epithelial structures is a prerequisite for increased cell motility and tumor invasion. This phenotypic switch is designated epithelial‐to‐mesenchymal transition (EMT). EMT also plays an important role in determining the dissemination of tumors. However, the role of MEK1 in intestinal EMT, tumor invasion and metastasis has not been elucidated. To determine the functions of activated MEK1 in intestinal tumorigenesis, we established intestinal epithelial cell lines that overexpress wild‐type MEK1 (wtMEK) or activated MEK1 (caMEK). Our results indicate that expression of caMEK is sufficient to induce EMT as confirmed with the induction of N‐cadherin, vimentin, Snail1 and Snail2, whereas a reduction in E‐cadherin, occludin, ZO‐1 and cortical F‐actin was noted. The Snail1 and Snail2 promoter analyses revealed that Egr‐1 and Fra‐1, an AP‐1 protein, are responsible for MEK1‐induced Snail1 and Snail2 expression, respectively. Cells expressing activated MEK1 clearly acquired an invasive capacity when compared to wtMEK‐expressing cells. Zymography studies confirmed elevated levels of MMP2 and MMP9 activities in media of caMEK‐expressing cells. Importantly, cells expressing activated MEK1 induced tumors with short latency in correlation with their ability to induce experimental metastasis in vivo and to express factors known to promote colorectal cancer cell metastasis. In conclusion, our results demonstrate, for the first time, that constitutive activation of MEK1 in intestinal epithelial cells is sufficient to induce an EMT associated with tumor invasion and metastasis.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2011

Constitutive activation of the MEK/ERK pathway inhibits intestinal epithelial cell differentiation.

Etienne Lemieux; Marie-Josée Boucher; Sébastien Mongrain; François Boudreau; Claude Asselin; Nathalie Rivard

The Ras/Raf/MEK/ERK cascade regulates intestinal epithelial cell proliferation. Indeed, while barely detectable in differentiated cells of the villi, ERK1/2-activated forms are detected in the nucleus of undifferentiated human intestinal crypt cells. In addition, we and others have reported that ERKs are selectively inactivated during enterocyte differentiation. However, whether inactivation of the ERK pathway is necessary for inhibition of both proliferation and induction of differentiation of intestinal epithelial cells is unknown. Human Caco-2/15 cells, undifferentiated crypt IEC-6 cells, and differentiating Cdx3-expressing IEC-6 cells were infected with retroviruses encoding either a hemagglutinin (HA)-tagged MEK1 wild type (wtMEK) or a constitutively active S218D/S222D MEK1 mutant (caMEK). Protein and gene expression was assessed by Western blotting, semiquantitative RT-PCR, and real-time PCR. Morphology was analyzed by transmission electron microscopy. We found that 1) IEC-6/Cdx3 cells formed multicellular layers after confluence and differentiated after 30 days in culture, as assessed by increased polarization, microvilli formation, expression of differentiation markers, and ERK1/2 inhibition; 2) while activated MEK prevented neither the inhibition of ERK1/2 activities nor the differentiation process in postconfluent Caco-2/15 cells, caMEK expression prevented ERK inhibition in postconfluent IEC-6/Cdx3 cells, thus leading to maintenance of elevated ERK1/2 activities; 3) caMEK-expressing IEC-6/Cdx3 cells exhibited altered multicellular structure organization, poorly defined tight junctions, reduced number of microvilli on the apical surface, and decreased expression of the hepatocyte nuclear factor 1α transcription factor and differentiation markers, namely apolipoprotein A-4, fatty acid-binding protein, calbindin-3, mucin 2, alkaline phosphatase, and sucrase-isomaltase; and 4) increased Cdx3 phosphorylation on serine-60 (S60) in IEC-6/Cdx3 cells expressing caMEK led to decreased Cdx2 transactivation potential. These results indicate that inactivation of the ERK pathway is required to ensure the full Cdx2/3 transcriptional activity necessary for intestinal epithelial cell terminal differentiation.


Molecular Cancer | 2010

The serine protease inhibitor serpinE2 is a novel target of ERK signaling involved in human colorectal tumorigenesis

Sébastien Bergeron; Etienne Lemieux; Véronique Durand; Sébastien Cagnol; Julie C. Carrier; Jacques G. Lussier; Marie-Josée Boucher; Nathalie Rivard

BackgroundAmong the most harmful of all genetic abnormalities that appear in colorectal cancer (CRC) development are mutations of KRAS and its downstream effector BRAF as they result in abnormal extracellular signal-related kinase (ERK) signaling. In a previous report, we had shown that expression of a constitutive active mutant of MEK1 (caMEK) in normal rat intestinal epithelial cells (IECs) induced morphological transformation associated with epithelial to mesenchymal transition, growth in soft agar, invasion and metastases in nude mice. Results from microarrays comparing control to caMEK-expressing IECs identified the gene encoding for serpinE2, a serine protease inhibitor, as a potential target of activated MEK1.Results1- RT-PCR and western blot analyses confirmed the strong up-regulation of serpinE2 expression and secretion by IECs expressing oncogenic MEK, Ras or BRAF. 2- Interestingly, serpinE2 mRNA and protein were also markedly enhanced in human CRC cells exhibiting mutation in KRAS and BRAF. 3- RNAi directed against serpinE2 in caMEK-transformed rat IECs or in human CRC cell lines HCT116 and LoVo markedly decreased foci formation, anchorage-independent growth in soft agarose, cell migration and tumor formation in nude mice. 4- Treatment of CRC cell lines with U0126 markedly reduced serpinE2 mRNA levels, indicating that expression of serpinE2 is likely dependent of ERK activity. 5- Finally, Q-PCR analyses demonstrated that mRNA levels of serpinE2 were markedly increased in human adenomas in comparison to healthy adjacent tissues and in colorectal tumors, regardless of tumor stage and grade.ConclusionsOur data indicate that serpinE2 is up-regulated by oncogenic activation of Ras, BRAF and MEK1 and contributes to pro-neoplastic actions of ERK signaling in intestinal epithelial cells. Hence, serpinE2 may be a potential therapeutic target for colorectal cancer treatment.


Molecular and Cellular Biology | 2013

Epithelial Tyrosine Phosphatase SHP-2 Protects against Intestinal Inflammation in Mice

Geneviève Coulombe; Caroline Leblanc; Sébastien Cagnol; Faiza Maloum; Etienne Lemieux; Nathalie Perreault; Gen-Sheng Feng; François Boudreau; Nathalie Rivard

ABSTRACT Polymorphisms of PTPN11 encoding SHP-2 are biomarkers for ulcerative colitis (UC) susceptibility. However, their functional relevance is unknown. We thus investigated the role of epithelial SHP-2 in the control of intestinal homeostasis. Mice with an intestinal epithelial cell-specific SHP-2 deletion (SHP-2IEC-KO mice) were generated. Control and SHP-2IEC-KO mice were monitored for clinical symptoms and sacrificed for histological staining and Western blot analyses. Cytokines and chemokines, as well as intestinal permeability, were quantified. SHP-2 mRNA expression was evaluated in control and UC patients. SHP-2IEC-KO mice showed growth retardation compared to control littermates and rapidly developed severe colitis. Colon architecture was markedly altered with infiltration of immune cells, crypt abscesses, neutrophil accumulation, and reduced goblet cell numbers. Decreased expression of claudins was associated with enhanced intestinal permeability in mutant SHP-2IEC-KO mice. Inflammatory transcription factors Stat3 and NF-κB were hyperactivated early in the mutant colonic epithelium. Levels of several epithelial chemokines and cytokines were markedly enhanced in SHP-2IEC-KO mice. Of note, antibiotic treatment remarkably impaired the development of colitis in SHP-2IEC-KO mice. Finally, SHP-2 mRNA levels were significantly reduced in intestinal biopsy specimens from UC patients. Our results establish intestinal epithelial SHP-2 as a critical determinant for prevention of gut inflammation.


International Journal of Cancer | 2016

Bmp signaling in colonic mesenchyme regulates stromal microenvironment and protects from polyposis initiation

Joannie M. Allaire; Sebastien A. Roy; Camille Ouellet; Etienne Lemieux; Christine Jones; Marilène Paquet; François Boudreau; Nathalie Perreault

In the colon, myofibroblasts are primary contributors in the establishment of the microenvironment involved in tissue homeostasis. Alterations in myofibroblast functions lead to changes resulting in a toxic microenvironment nurturing tumorigenesis. Bone morphogenetic proteins (Bmps) are morphogens known to play key roles in adult gut homeostasis. Studies in genetically‐modified mice have shown that Bmp disruption in all cell layers leads to the development of gut polyposis. In contrast, our studies showed that loss of Bmp exclusively in the gastrointestinal epithelium resulted in increased epithelial proliferation without polyposis initiation, thus suggesting a key role for mesenchymal Bmp signaling in polyposis initiation. In order to identify the role of mesenchymal Bmp signaling on the microenvironment and its impact on colonic mucosa, a mouse model was generated with suppression of Bmp signaling exclusively in myofibroblasts (Bmpr1aΔMES). Bmpr1aΔMES mice exhibited increased subepithelial proliferation with changes in cellular composition leading to the development of a primed stroma with modulation of extracellular matrix proteins, immune cells and cytokines as early as 90 days of age. This microenvironmental deregulation was associated with increased polyposis initiation at one year of age. These results are the first to demonstrate that mesenchymal Bmpr1a inactivation alone is sufficient to prompt an expansion of myofibroblasts leading to the development of a reactive mesenchyme that contributes to polyposis initiation in the colon. These findings support the novel concept that inhibition of Bmp signaling in mesenchymal cells surrounding the normal epithelium leads to important changes instructing a toxic microenvironment sufficient to induce colonic polyposis.


International Journal of Oncology | 2013

Functional impact of colorectal cancer-associated mutations in the transcription factor E2F4

Marie-Christine Paquin; Caroline Leblanc; Etienne Lemieux; Benjamin Bian; Nathalie Rivard

The transcription factor E2F4 plays a critical role in cell cycle progression of normal and cancerous intestinal epithelial cells. Contrary to other E2Fs, the coding region of the E2F4 gene contains a longer spacer segment of a CAG trinucleotide repeat sequence encoding 13 consecutive serine residues, which is highly vulnerable to frameshift mutations in situations of genetic instability. Mutations in this region of the E2F4 gene have been observed in colorectal tumors with microsatellite instability. However, the effect of these changes on its function in colorectal cancer cells is currently unknown. We generated E2F4(CAG)₁₂ and E2F4(CAG)₁₄ mutants and compared their activity to the E2F4 wild-type, E2F4(CAG)₁₃. Luciferase assays with the thymidine kinase-luc reporter gene revealed that the mutants were more transcriptionally active than wild-type E2F4. The mechanism of increased activity of E2F4 was primarily related to protein stability, due to a significantly enhanced half-life of E2F4 mutants comparatively to that of wild-type E2F4. However, the association with the pocket protein p130/RBL2 did not account for this increased protein stability. Sequencing analysis of the endogenous E2F4 gene in a series of colorectal cancer cell lines showed that the microsatellite-unstable cell line SW48 exhibited a serine deletion in this gene. Accordingly, E2F4 half-life was much more elevated in SW48 cells in comparison to Caco-2/15, a microsatellite-stable cell line. Notably, in soft-agar assays, both mutants more potently increased anchorage-independent growth in comparison to wild-type E2F4. In conclusion, our data demonstrate that cancer-associated E2F4 mutations enhance the capacity of colorectal cancer cells to grow without anchorage, thereby contributing to tumor progression.


Biochimica et Biophysica Acta | 2015

Identification of GATA-4 as a novel transcriptional regulatory component of regenerating islet-derived family members.

David Lepage; Joannie Bruneau; Geneviève Brouillard; Christine Jones; Carine R. Lussier; Anthony Rémillard; Etienne Lemieux; Claude Asselin; François Boudreau

Intestinal epithelial cells are exposed to luminal bacterial threat and require adequate defense mechanisms to ensure host protection and epithelium regeneration against possible deleterious damage. Differentiated intestinal epithelial cells produce antimicrobial and regenerative components that protect against such challenges. Few intestinal specific transcription factors have been identified to control the switching from repression to activation of this class of gene. Herein, we show that gene transcription of some regenerating islet-derived (REG) family members is dependent on the transcription factor GATA-4. Silencing of GATA-4 expression in cultured intestinal epithelial cells identified Reg3β as a target gene using an unbiased approach of gene expression profiling. Co-transfection and RNA interference assays identified complex GATA-4-interactive transcriptional components required for the activation or repression of Reg3β gene activity. Conditional deletion of Gata4 in the mouse intestinal epithelium supported its regulatory role for Reg1, Reg3α, Reg3β and Reg3γ genes. Reg1 dramatic down-modulation of expression in Gata4 conditional null mice was associated with a significant decrease in intestinal epithelial cell migration. Altogether, these results identify a novel and complex role for GATA-4 in the regulation of REG family members gene expression.


Scientific Reports | 2016

Loss of mesenchymal bone morphogenetic protein signaling leads to development of reactive stroma and initiation of the gastric neoplastic cascade

Sébastien A. B. Roy; Joannie M. Allaire; Camille Ouellet; Faiza Maloum-Rami; Véronique Pomerleau; Etienne Lemieux; Jean-Philippe Babeu; Jasmin Rousseau; Marilène Paquet; Perrine Garde-Granger; François Boudreau; Nathalie Perreault

Bmps are morphogens involved in various gastric cellular functions. Studies in genetically-modified mice have shown that Bmp disruption in gastric epithelial and stromal cell compartments leads to the development of tumorigenesis. Our studies have demonstrated that abrogation of gastric epithelial Bmp signaling alone was not sufficient to recapitulate the neoplastic features associated with total gastric loss of Bmp signaling. Thus, epithelial Bmp signaling does not appear to be a key player in gastric tumorigenesis initiation. These observations suggest a greater role for stromal Bmp signaling in gastric polyposis initiation. In order to identify the specific roles played by mesenchymal Bmp signaling in gastric homeostasis, we generated a mouse model with abrogation of Bmp signaling exclusively in the gastro-intestinal mesenchyme (Bmpr1aΔMES). We were able to expose an unsuspected role for Bmp loss of signaling in leading normal gastric mesenchyme to adapt into reactive mesenchyme. An increase in the population of activated-fibroblasts, suggesting mesenchymal transdifferentiation, was observed in mutant stomach. Bmpr1aΔMES stomachs exhibited spontaneous benign polyps with presence of both intestinal metaplasia and spasmolytic-polypeptide-expressing metaplasia as early as 90 days postnatal. These results support the novel concept that loss of mesenchymal Bmp signaling cascade acts as a trigger in gastric polyposis initiation.


Cancer Research | 2012

Abstract 4169: The Wnt/β-catenin pathway is involved in KRAS-induced intestinal epithelial cell transformation

Etienne Lemieux; Nathalie Rivard

Aberrant regulation of the Wnt/β-catenin signaling pathway is one of the major causes of colorectal cancer (CRC). The APC protein is an integral part of the destruction complex that controls cytoplasmic β-catenin levels by promoting ubiquitin-mediated degradation of β-catenin. Loss-of-function mutations in APC are commonly found in CRC, leading to constitutively high levels of β-catenin which then associates with TCF4, inducing expression of target genes that are important in cell growth. The KRAS/BRAF/MEK/ERK signaling pathway is also a major transforming growth regulatory signaling pathway. Indeed, gain-of-function mutations of KRAS and BRAF are detected in up to 60% of CRCs. Although both the Wnt/β-catenin and the ERK pathways are major pathways for tumorigenesis of intestinal epithelial cells (IECs), no significant interactions between these pathways have been identified. Methods. Retrovirus encoding the HA-tagged wild-type MEK1 (wtMEK) or activated MEK1 (MEK1S218D/S222D, caMEK) or oncogenic form of KRASG12V were used to infect and transform normal IECs in culture. The transcriptional activity of β-catenin/TCF4 complex was analyzed by using the luciferase reporter systems TOPFLASH and c-myc promoter (mutated or not for TCF4 binding elements). Expression levels of Wnt/β-catenin targets axin2, lef-1 and c-myc were evaluated by qPCR and protein localisation by immunofluorescence. A dominant-negative form of TCF4 (TCF4-DN) was used to verify the contribution of β-catenin/TCF4 complex in transformation induced by oncogenic activation of KRAS or MEK1. β-catenin transcriptional activity was analyzed in human CRC cells exhibiting APC mutation, treated or not with the MEK inhibitor U0126. Results. 1- Phase-contrast microscopy revealed that KRASG12V and caMEK-expressing IECs underwent a morphological change from an epithelial morphology to an elongated morphology. Cells clearly loose cell-to-cell contacts when compared to control cells. E-cadherin and β-catenin proteins disappeared from cell-cell junctions. 2- β-catenin/TCF4 transcriptional activity as well as mRNA levels of c-myc, axin2 and lef1 were markedly increased in IECs transformed by oncogenic MEK1 or KRAS. 3- Expression of the dominant-negative form of TCF4 severely attenuated IEC transformation induced by oncogenic MEK1 or KRAS. 4- Treatment of human CRC cell lines with U0126 significantly reduced axin2, lef-1 and c-myc mRNA levels, indicating that expression of these β-catenin target genes is likely dependent on MEK/ERK activity. Accordingly, β-catenin/TCF4 transcriptional activity was inhibited following MEK inhibition by U0126 or ERK dephosphorylation by DUSP4 expression. Conclusion. Our data indicate that the oncogenic activation of the KRAS/ERK signalling pathway in IECs up-regulates β-catenin/TCF4 transcriptional activity which contributes to the tumorigenic potential of this signaling. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 4169. doi:1538-7445.AM2012-4169


Archive | 2014

Miceagainst Intestinal Inflammation in Epithelial Tyrosine Phosphatase SHP-2

Gen-Sheng Feng; François Boudreau; Nathalie Rivard; Faiza Maloum; Etienne Lemieux; Nathalie Perreault; Geneviève Coulombe; Caroline Leblanc; Sébastien Cagnol

Collaboration


Dive into the Etienne Lemieux's collaboration.

Top Co-Authors

Avatar

Nathalie Rivard

Université de Sherbrooke

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Claude Asselin

Université de Sherbrooke

View shared research outputs
Top Co-Authors

Avatar

Faiza Maloum

Université de Sherbrooke

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nathalie Perreault

Faculté de médecine – Université de Sherbrooke

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge