Network


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

Hotspot


Dive into the research topics where Claudine Rancourt is active.

Publication


Featured researches published by Claudine Rancourt.


Nature Structural & Molecular Biology | 2009

Cancer-associated regulation of alternative splicing.

Julian P. Venables; Roscoe Klinck; ChuShin Koh; Julien Gervais-Bird; Anne Bramard; Lyna Inkel; Mathieu Durand; Sonia Couture; Ulrike Froehlich; Elvy Lapointe; Jean-François Lucier; Philippe Thibault; Claudine Rancourt; Karine Tremblay; Panagiotis Prinos; Benoit Chabot; Sherif Abou Elela

Alternative splicing of pre-mRNA increases the diversity of protein functions. Here we show that about half of all active alternative splicing events in ovarian and breast tissues are changed in tumors, and many seem to be regulated by a single factor; sequence analysis revealed binding sites for the RNA binding protein FOX2 downstream of one-third of the exons skipped in cancer. High-resolution analysis of FOX2 binding sites defined the precise positions relative to alternative exons at which the protein may function as either a silencer or an enhancer. Most of the identified targets were shifted in the same direction by FOX2 depletion in cell lines as they were in breast and ovarian cancer tissues. Notably, we found expression of FOX2 itself is downregulated in ovarian cancer and its splicing is altered in breast cancer samples. These results suggest that the decreased expression of FOX2 in cancer tissues modulates splicing and controls proliferation.


Molecular Cancer | 2006

Mesothelin-MUC16 binding is a high affinity, N-glycan dependent interaction that facilitates peritoneal metastasis of ovarian tumors

Jennifer A. A. Gubbels; Jennifer A. Belisle; Masanori Onda; Claudine Rancourt; Martine Migneault; Mitchell Ho; Tapan K. Bera; Joseph P. Connor; Bangalore K. Sathyanarayana; Byungkook Lee; Ira Pastan; Manish S. Patankar

BackgroundThe mucin MUC16 and the glycosylphosphatidylinositol anchored glycoprotein mesothelin likely facilitate the peritoneal metastasis of ovarian tumors. The biochemical basis and the kinetics of the binding between these two glycoproteins are not clearly understood. Here we have addressed this deficit and provide further evidence supporting the role of the MUC16-mesothelin interaction in facilitating cell-cell binding under conditions that mimic the peritoneal environment.ResultsIn this study we utilize recombinant-Fc tagged human mesothelin to measure the binding kinetics of this glycoprotein to MUC16 expressed on the ovarian tumor cell line OVCAR-3. OVCAR-3 derived sublines that did not express MUC16 showed no affinity for mesothelin. In a flow cytometry-based assay mesothelin binds with very high affinity to the MUC16 on the OVCAR-3 cells with an apparent Kd of 5–10 nM. Maximum interaction occurs within 5 mins of incubation of the recombinant mesothelin with the OVCAR-3 cells and significant binding is observed even after 10 sec. A five-fold molar excess of soluble MUC16 was unable to completely inhibit the binding of mesothelin to the OVCAR-3 cells. Oxidation of the MUC16 glycans, removal of its N-linked oligosaccharides, and treatment of the mucin with wheat germ agglutinin and erythroagglutinating phytohemagglutinin abrogates its binding to mesothelin. These observations suggest that at least a subset of the MUC16-asscociated N-glycans is required for binding to mesothelin. We also demonstrate that MUC16 positive ovarian tumor cells exhibit increased adherence to A431 cells transfected with mesothelin (A431-Meso+). Only minimal adhesion is observed between MUC16 knockdown cells and A431-Meso+ cells. The binding between the MUC16 expressing ovarian tumor cells and the A431-Meso+ cells occurs even in the presence of ascites from patients with ovarian cancer.ConclusionThe strong binding kinetics of the mesothelin-MUC16 interaction and the cell adhesion between ovarian tumor cells and A431-Meso+ even in the presence of peritoneal fluid strongly support the importance of these two glycoproteins in the peritoneal metastasis of ovarian tumors. The demonstration that N-linked glycans are essential for mediating mesothlein-MUC16 binding may lead to novel therapeutic targets to control the spread of ovarian carcinoma.


Cancer Research | 2008

Identification of Alternative Splicing Markers for Breast Cancer

Julian P. Venables; Roscoe Klinck; Anne Bramard; Lyna Inkel; Geneviève Dufresne-Martin; ChuShin Koh; Julien Gervais-Bird; Elvy Lapointe; Ulrike Froehlich; Mathieu Durand; Daniel Gendron; Jean-Philippe Brosseau; Philippe Thibault; Jean-François Lucier; Karine Tremblay; Panagiotis Prinos; Raymund J. Wellinger; Benoit Chabot; Claudine Rancourt; Sherif Abou Elela

Breast cancer is the most common cause of cancer death among women under age 50 years, so it is imperative to identify molecular markers to improve diagnosis and prognosis of this disease. Here, we present a new approach for the identification of breast cancer markers that does not measure gene expression but instead uses the ratio of alternatively spliced mRNAs as its indicator. Using a high-throughput reverse transcription-PCR-based system for splicing annotation, we monitored the alternative splicing profiles of 600 cancer-associated genes in a panel of 21 normal and 26 cancerous breast tissues. We validated 41 alternative splicing events that significantly differed in breast tumors relative to normal breast tissues. Most cancer-specific changes in splicing that disrupt known protein domains support an increase in cell proliferation or survival consistent with a functional role for alternative splicing in cancer. In a blind screen, a classifier based on the 12 best cancer-associated splicing events correctly identified cancer tissues with 96% accuracy. Moreover, a subset of these alternative splicing events could order tissues according to histopathologic grade, and 5 markers were validated in a further blind set of 19 grade 1 and 19 grade 3 tumor samples. These results provide a simple alternative for the classification of normal and cancerous breast tumor tissues and underscore the putative role of alternative splicing in the biology of cancer.


Cancer Research | 2008

Multiple alternative splicing markers for ovarian cancer.

Roscoe Klinck; Anne Bramard; Lyna Inkel; Geneviève Dufresne-Martin; Julien Gervais-Bird; Richard Madden; Eric Paquet; ChuShin Koh; Julian P. Venables; Panagiotis Prinos; Manuela Jilaveanu-Pelmus; Raymund J. Wellinger; Claudine Rancourt; Benoit Chabot; Sherif Abou Elela

Intense efforts are currently being directed toward profiling gene expression in the hope of developing better cancer markers and identifying potential drug targets. Here, we present a sensitive new approach for the identification of cancer signatures based on direct high-throughput reverse transcription-PCR validation of alternative splicing events. This layered and integrated system for splicing annotation (LISA) fills a gap between high-throughput microarray studies and high-sensitivity individual gene investigations, and was created to monitor the splicing of 600 cancer-associated genes in 25 normal and 21 serous ovarian cancer tissues. Out of >4,700 alternative splicing events screened, the LISA identified 48 events that were significantly associated with serous ovarian tumor tissues. In a further screen directed at 39 ovarian tissues containing cancer pathologies of various origins, our ovarian cancer splicing signature successfully distinguished all normal tissues from cancer. High-volume identification of cancer-associated splice forms by the LISA paves the way for the use of alternative splicing profiling to diagnose subtypes of cancer.


BMC Cancer | 2011

Prognostic significance of IL-6 and IL-8 ascites levels in ovarian cancer patients

Denis Lane; Isabelle Matte; Claudine Rancourt; Alain Piché

BackgroundThe acellular fraction of epithelial ovarian cancer (EOC) ascites promotes de novo resistance of tumor cells and thus supports the idea that tumor cells may survive in the surrounding protective microenvironment contributing to disease recurrence. Levels of the pro-inflammatory cytokines IL-6 and IL-8 are elevated in EOC ascites suggesting that they could play a role in tumor progression.MethodsWe measured IL-6 and IL-8 levels in the ascites of 39 patients with newly diagnosed EOC. Commercially available enzyme-linked immunosorbent assay (ELISA) was used to determine IL-6 and IL-8 ascites levels. Ascites cytokine levels were correlated with clinicopathological parameters and progression-free survival.ResultsMean ascites levels for IL-6 and IL-8 were 6419 pg/ml (SEM: 1409 pg/ml) and 1408 pg/ml (SEM: 437 pg/ml) respectively. The levels of IL-6 and IL-8 in ascites were significantly lower in patients that have received prior chemotherapy before the surgery (Mann-Whitney U test, P = 0.037 for IL-6 and P = 0.008 for IL-8). Univariate analysis revealed that high IL-6 ascites levels (P = 0.021), serum CA125 levels (P = 0.04) and stage IV (P = 0.009) were significantly correlated with shorter progression-free survival. Including these variables in a multivariate analysis revealed that elevated IL-6 levels (P = 0.033) was an independent predictor of shorter progression-free survival.ConclusionElevated IL-6, but not IL-8, ascites level is an independent predictor of shorter progression-free survival.


The FASEB Journal | 1997

Adenoviral/retroviral vector chimeras: a novel strategy to achieve high-efficiency stable transduction in vivo.

Guadalupe Bilbao; Meizhen Feng; Claudine Rancourt; W H Jackson; David T. Curiel

Gene therapy to correct defective genes requires efficient gene delivery and long‐term gene expression. Realization of both goals with available vector systems has so far not been achieved. As a novel approach to solve this problem, we have developed a chimeric viral vector system that exploits favorable aspects of both adenoviral and retroviral vectors. In this schema, adenoviral vectors induce target cells to function as transient retroviral producer cells in vivo. The progeny retroviral vector particles can then effectively achieve stable transduction of neighboring cells. In this system, the nonintegrative adenoviral vector is rendered functionally integrative via the intermediate generation of an induced retroviral producer cell. Such chimeric vectors may now allow realization of the requisite goals for specific gene therapy applications.—Bilbao, G., Feng, M., Rancourt, C., Jackson, W. H., Jr., Curiel, D. T. Adenoviral/retroviral vector chimeras: a novel strategy to achieve high‐efficiency stable transduction in vivo. FASEB J. 11, 624–634 (1997)


Molecular Cancer | 2010

MUC16 provides immune protection by inhibiting synapse formation between NK and ovarian tumor cells

Jennifer A. A. Gubbels; Mildred Felder; Sachi Horibata; Jennifer A. Belisle; Arvinder Kapur; Helen Holden; Sarah Petrie; Martine Migneault; Claudine Rancourt; Joseph P. Connor; Manish S. Patankar

BackgroundCancer cells utilize a variety of mechanisms to evade immune detection and attack. Effective immune detection largely relies on the formation of an immune synapse which requires close contact between immune cells and their targets. Here, we show that MUC16, a heavily glycosylated 3-5 million Da mucin expressed on the surface of ovarian tumor cells, inhibits the formation of immune synapses between NK cells and ovarian tumor targets. Our results indicate that MUC16-mediated inhibition of immune synapse formation is an effective mechanism employed by ovarian tumors to evade immune recognition.ResultsExpression of low levels of MUC16 strongly correlated with an increased number of conjugates and activating immune synapses between ovarian tumor cells and primary naïve NK cells. MUC16-knockdown ovarian tumor cells were more susceptible to lysis by primary NK cells than MUC16 expressing controls. This increased lysis was not due to differences in the expression levels of the ligands for the activating receptors DNAM-1 and NKG2D. The NK cell leukemia cell line (NKL), which does not express KIRs but are positive for DNAM-1 and NKG2D, also conjugated and lysed MUC16-knockdown cells more efficiently than MUC16 expressing controls. Tumor cells that survived the NKL challenge expressed higher levels of MUC16 indicating selective lysis of MUC16low targets. The higher csMUC16 levels on the NKL resistant tumor cells correlated with more protection from lysis as compared to target cells that were never exposed to the effectors.ConclusionMUC16, a carrier of the tumor marker CA125, has previously been shown to facilitate ovarian tumor metastasis and inhibits NK cell mediated lysis of tumor targets. Our data now demonstrates that MUC16 expressing ovarian cancer cells are protected from recognition by NK cells. The immune protection provided by MUC16 may lead to selective survival of ovarian cancer cells that are more efficient in metastasizing within the peritoneal cavity and also at overcoming anti-tumor innate immune responses.


Molecular Cancer | 2010

Identification of Siglec-9 as the receptor for MUC16 on human NK cells, B cells, and monocytes

Jennifer A. Belisle; Sachi Horibata; Gubbels Aa Jennifer; Sarah Petrie; Arvinder Kapur; Sabine André; Hans-Joachim Gabius; Claudine Rancourt; Joseph P. Connor; James C. Paulson; Manish S. Patankar

BackgroundMUC16 is a cell surface mucin expressed at high levels by epithelial ovarian tumors. Following proteolytic cleavage, cell surface MUC16 (csMUC16) is shed in the extracellular milieu and is detected in the serum of cancer patients as the tumor marker CA125. csMUC16 acts as an adhesion molecule and facilitates peritoneal metastasis of ovarian tumors. Both sMUC16 and csMUC16 also protect cancer cells from cytotoxic responses of natural killer (NK) cells. In a previous study we demonstrated that sMUC16 binds to specific subset of NK cells. Here, we identify the csMUC16/sMUC16 binding partner expressed on immune cells.ResultsAnalysis of immune cells from the peripheral blood and peritoneal fluid of ovarian cancer patients indicates that in addition to NK cells, sMUC16 also binds to B cells and monocytes isolated from the peripheral blood and peritoneal fluid. I-type lectin, Siglec-9, is identified as the sMUC16 receptor on these immune cells. Siglec-9 is expressed on approximately 30-40% of CD16pos/CD56dim NK cells, 20-30% of B cells and >95% of monocytes. sMUC16 binds to the majority of the Siglec-9pos NK cells, B cells and monocytes. sMUC16 is released from the immune cells following neuraminidase treatment. Siglec-9 transfected Jurkat cells and monocytes isolated from healthy donors bind to ovarian tumor cells via Siglec-9-csMUC16 interaction.ConclusionsRecent studies indicate that csMUC16 can act as an anti-adhesive agent that blocks tumor-immune cell interactions. Our results demonstrate that similar to other mucins, csMUC16 can also facilitate cell adhesion by interacting with a suitable binding partner such as mesothelin or Siglec-9. Siglec-9 is an inhibitory receptor that attenuates T cell and NK cell function. sMUC16/csMUC16-Siglec-9 binding likely mediates inhibition of anti-tumor immune responses.


Gynecologic Oncology | 2011

MUC16 (CA125) regulates epithelial ovarian cancer cell growth, tumorigenesis and metastasis

Catherine Thériault; Maxime Pinard; Marina Comamala; Martine Migneault; Julie Beaudin; Isabelle Matte; Marianne Boivin; Alain Piché; Claudine Rancourt

OBJECTIVES MUC16 (CA125) protein is a high molecular weight mucin overexpressed in the majority of epithelial ovarian cancers (EOC) but not in the epithelium of normal ovaries suggesting that it might play a role in EOC pathogenesis. Here, we explored the phenotypic consequences of MUC16 knockdown and expression of its C-terminal domain with the aim of establishing a role for MUC16 in tumorigenesis. METHODS MUC16 was down-regulated by stably expressing an anti-MUC16 endoplasmic reticulum-targeted single-chain antibody which prevented MUC16 cell surface localization in NIH:OVCAR3 cells. In addition, we generated epitope tagged, N-terminal region-deleted MUC16 constructs with (MUC16TMU) and without (MUC16CTD) cytoplasmic tail deletions and stably expressed them in SKOV3 cells. RESULTS Although MUC16 knockdown did not affect the cell growth rate, knockdown cells reached a stationary growth phase after 4 days whereas control cells continued to grow for up to 7 days. Colony formation assays in soft agar demonstrated that MUC16 knockdown cells had >8-fold reduction in their ability to form colonies. Importantly, MUC16 knockdown completely prevents the formation of subcutaneous tumors in nude mice. Conversely, we show that ectopic expression of the MUC16CTD enhances SKOV3 tumor cell growth, colony formation in soft agar and enhances tumor growth and metastases in SCID mice. In addition, MUC16CTD expression increases cell motility, invasiveness, and metastatic property. Deletion of the cytoplasmic tail from the MUC16CTD completely abolished its ability to enhance tumor cell growth, cell motility and invasiveness. Furthermore, the increased invasive properties of MUC16CTD-expressing cells correlated with decreased expression of E-cadherin and increased expression of N-cadherin and vimentin. CONCLUSION These findings provide the first evidence for a critical role of MUC16 in tumor cell growth, tumorigenesis and metastases.


International Journal of Cancer | 2007

Malignant ascites protect against TRAIL-induced apoptosis by activating the PI3K/AKt pathway in human ovarian carcinoma cells

Denis Lane; Véronique Robert; Roxanne Grondin; Claudine Rancourt; Alain Piché

Ascites are commonly found in ovarian cancer patients with advanced disease and are rich in cellular components and growth‐promoting factors. The purpose of this study was to assess the effect of malignant ascites on TRAIL‐induced apoptosis. We demonstrate that malignant ascites obtained from women with advanced ovarian cancer protect tumor cells from TRAIL‐ and FasL‐induced apoptosis but not against cisplatin‐induced apoptosis. This antiapoptotic effect was consistently found among different malignant ascites while nonmalignant peritoneal fluids or conditioned medium from TRAIL‐resistant cells failed to protect tumor cells against TRAIL killing. Malignant ascites strongly inhibits TRAIL‐induced caspase‐3 activation and PARP cleavage. Furthermore, ascites activate PI3K and its downstream target Akt and increases c‐FLIPS protein levels without affecting ERK phosphorylation status. The antiapoptotic effect of malignant ascites is abrogated by the inhibition of PI3K with LY294002, by a specific inhibitor of Akt and by Akt siRNA. We further show that the pro‐survival effect of ascites can be suppressed by down‐regulation of c‐FLIPS. Our data indicate that malignant effusions protect against TRAIL‐induced apoptosis by activating the PI3K/Akt pathway. These findings demonstrate that the tumor microenvironment may contribute to the resistance of ovarian cancer cells to death receptor‐induced apoptosis.

Collaboration


Dive into the Claudine Rancourt's collaboration.

Top Co-Authors

Avatar

Isabelle Matte

Université de Sherbrooke

View shared research outputs
Top Co-Authors

Avatar

David T. Curiel

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Jennifer A. Belisle

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Joseph P. Connor

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Manish S. Patankar

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gene P. Siegal

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Minghui Wang

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Ronald D. Alvarez

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Julie Beaudin

Université de Sherbrooke

View shared research outputs
Researchain Logo
Decentralizing Knowledge