Network


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

Hotspot


Dive into the research topics where Noël J.-M. Raynal is active.

Publication


Featured researches published by Noël J.-M. Raynal.


EMBO Reports | 2011

Histone deacetylase inhibitor activity in royal jelly might facilitate caste switching in bees.

Yong Kee Kim; Noël J.-M. Raynal; Vazganush Gharibyan; Ming Bo Su; Yue Yang Zhou; Jia Li; Sabrina Castellano; Gianluca Sbardella; Jean-Pierre Issa; Mark T. Bedford

Worker and queen bees are genetically indistinguishable. However, queen bees are fertile, larger and have a longer lifespan than their female worker counterparts. Differential feeding of larvae with royal jelly controls this caste switching. There is emerging evidence that the queen‐bee phenotype is driven by epigenetic mechanisms. In this study, we show that royal jelly—the secretion produced by the hypopharyngeal and mandibular glands of worker bees—has histone deacetylase inhibitor (HDACi) activity. A fatty acid, (E)‐10‐hydroxy‐2‐decenoic acid (10HDA), which accounts for up to 5% of royal jelly, harbours this HDACi activity. Furthermore, 10HDA can reactivate the expression of epigenetically silenced genes in mammalian cells. Thus, the epigenetic regulation of queen‐bee development is probably driven, in part, by HDACi activity in royal jelly.


Cancer Chemotherapy and Pharmacology | 2009

Inhibition of cytidine deaminase by zebularine enhances the antineoplastic action of 5-aza-2′-deoxycytidine

Maryse Lemaire; Louise F. Momparler; Noël J.-M. Raynal; Mark Bernstein; Richard L. Momparler

Cytidine (CR) deaminase is a key enzyme in the catabolism of cytosine nucleoside analogues, since their deamination results in a loss of their pharmacological activity. In this report we have investigated the importance of CR deaminase with respect to the antineoplastic action of inhibitors of DNA methylation, 5-aza-2′-deoxycytidine (5-AZA-CdR) and zebularine. Zebularine has a dual mechanism of action, since it can also inhibit CR deaminase. The objective of our study was to investigate the importance of zebularine as an inhibitor of CR deaminase with respect to the antineoplastic action of 5-AZA-CdR. Using an in vitro clonogenic assay, we investigated the antineoplastic action of 5-AZA-CdR and zebularine, alone and in combination on wild type 3T3 murine fibroblasts and corresponding V5 cells transduced with CR deaminase gene to express a very high level of CR deaminase activity. The V5 cells were much less sensitive to 5-AZA-CdR than the wild type 3T3 cells. The addition of zebularine significantly enhanced the antineoplastic action of 5-AZA-CdR on V5 cells, but not 3T3 cells. Enzymatic analysis on CR deaminase purified from the V5 cells showed that zebularine is a competitive inhibitor of the deamination of 5-AZA-CdR. These in vitro observations are in accord with our in vivo study in mice with L1210 leukemia, which showed that zebularine increased the antileukemic activity of 5-AZA-CdR. Pharmacokinetic analysis also showed that zebularine increased the plasma level of 5-AZA-CdR during an i.v. infusion in mice. Our results indicate that the major mechanism by which zebularine enhances the antineoplastic action of 5-AZA-CdR is by inhibition of CR deaminase. These findings provide a rationale to investigate 5-AZA-CdR in combination with zebularine in patients with advanced leukemia.


Journal of Natural Products | 2008

Antileukemic activity of genistein, a major isoflavone present in soy products.

Noël J.-M. Raynal; Louise F. Momparler; Michel Charbonneau; Richard L. Momparler

Soy has been used in traditional medicine for the treatment of various diseases, including cancer. The isoflavones present in soy have been shown in animal models to have cancer-preventing activity. However, the therapeutic effects of isoflavones against cancer are still unclear. We have evaluated the in vitro and in vivo antileukemic activity of genistein (1), a major isoflavone present in soy. We observed that it produced a dose- and time-dependent antineoplastic activity against myeloid and lymphoid leukemic cell lines. In addition, genistein treatment of the leukemic cells reactivated tumor suppressor genes that were silenced by aberrant DNA methylation. A genistein-enriched diet produced a moderate, but significant, antileukemic effect in mice. The limited extent of this in vivo response may have been due to the rapid metabolic inactivation of genistein in mice. Due to the longer half-life of genistein in humans, a soy-enriched diet has the potential to produce plasma levels of this isoflavone in the range of the concentrations used in vitro that produced an antileukemic activity.


Genome Biology | 2013

Architecture of epigenetic reprogramming following Twist1-mediated epithelial-mesenchymal transition

Gabriel G. Malouf; Joseph H. Taube; Yue Lu; Tapasree Roysarkar; Shoghag Panjarian; Marcos R. Estecio; Jaroslav Jelinek; Jumpei Yamazaki; Noël J.-M. Raynal; Hai Long; Tomomitsu Tahara; Agata A. Tinnirello; Xiu Ying Zhang; Shoudan Liang; Sendurai A. Mani; Jean-Pierre Issa

BackgroundEpithelial-mesenchymal transition (EMT) is known to impart metastasis and stemness characteristics in breast cancer. To characterize the epigenetic reprogramming following Twist1-induced EMT, we characterized the epigenetic and transcriptome landscapes using whole-genome transcriptome analysis by RNA-seq, DNA methylation by digital restriction enzyme analysis of methylation (DREAM) and histone modifications by CHIP-seq of H3K4me3 and H3K27me3 in immortalized human mammary epithelial cells relative to cells induced to undergo EMT by Twist1.ResultsEMT is accompanied by focal hypermethylation and widespread global DNA hypomethylation, predominantly within transcriptionally repressed gene bodies. At the chromatin level, the number of gene promoters marked by H3K4me3 increases by more than one fifth; H3K27me3 undergoes dynamic genomic redistribution characterized by loss at half of gene promoters and overall reduction of peak size by almost half. This is paralleled by increased phosphorylation of EZH2 at serine 21. Among genes with highly altered mRNA expression, 23.1% switch between H3K4me3 and H3K27me3 marks, and those point to the master EMT targets and regulators CDH1, PDGFRα and ESRP1. Strikingly, Twist1 increases the number of bivalent genes by more than two fold. Inhibition of the H3K27 methyltransferases EZH2 and EZH1, which form part of the Polycomb repressive complex 2 (PRC2), blocks EMT and stemness properties.ConclusionsOur findings demonstrate that the EMT program requires epigenetic remodeling by the Polycomb and Trithorax complexes leading to increased cellular plasticity. This suggests that inhibiting epigenetic remodeling and thus decrease plasticity will prevent EMT, and the associated breast cancer metastasis.


Molecular Cancer Therapeutics | 2017

Repositioning FDA-Approved Drugs in Combination with Epigenetic Drugs to Reprogram Colon Cancer Epigenome

Noël J.-M. Raynal; Elodie M. Da Costa; Justin T. Lee; Vazganush Gharibyan; Saira Ahmed; Hanghang Zhang; Takahiro Sato; Gabriel G. Malouf; Jean-Pierre Issa

Epigenetic drugs, such as DNA methylation inhibitors (DNMTi) or histone deacetylase inhibitors (HDACi), are approved in monotherapy for cancer treatment. These drugs reprogram gene expression profiles, reactivate tumor suppressor genes (TSG) producing cancer cell differentiation and apoptosis. Epigenetic drugs have been shown to synergize with other epigenetic drugs or various anticancer drugs. To discover new molecular entities that enhance epigenetic therapy, we performed a high-throughput screening using FDA-approved libraries in combination with DNMTi or HDACi. As a screening model, we used YB5 system, a human colon cancer cell line, which contains an epigenetically silenced CMV-GFP locus, mimicking TSG silencing in cancer. CMV-GFP reactivation is triggered by DNMTi or HDACi and responds synergistically to DNMTi/HDACi combination, which phenocopies TSG reactivation upon epigenetic therapy. GFP fluorescence was used as a quantitative readout for epigenetic activity. We discovered that 45 FDA-approved drugs (4% of all drugs tested) in our FDA-approved libraries enhanced DNMTi and HDACi activity, mainly belonging to anticancer and antiarrhythmic drug classes. Transcriptome analysis revealed that combination of decitabine (DNMTi) with the antiarrhythmic proscillaridin A produced profound gene expression reprogramming, which was associated with downregulation of 153 epigenetic regulators, including two known oncogenes in colon cancer (SYMD3 and KDM8). Also, we identified about 85 FDA-approved drugs that antagonized DNMTi and HDACi activity through cytotoxic mechanisms, suggesting detrimental drug interactions for patients undergoing epigenetic therapy. Overall, our drug screening identified new combinations of epigenetic and FDA-approved drugs, which can be rapidly implemented into clinical trials. Mol Cancer Ther; 16(2); 397–407. ©2016 AACR.


Epigenetics | 2015

Methylome sequencing for fibrolamellar hepatocellular carcinoma depicts distinctive features

Gabriel G. Malouf; Tomomitsu Tahara; Valérie Paradis; Monique Fabre; Catherine Guettier; Jumpei Yamazaki; Hi Long; Yue Lu; Noël J.-M. Raynal; Jaroslav Jelinek; Roger Mouawad; David Khayat; Laurence Brugières; Eric Raymond; Jean-Pierre Issa

With the goal of studying epigenetic alterations in fibrolamellar hepatocellular carcinoma (FLC) and establish an associated DNA methylation signature, we analyzed LINE-1 methylation in a cohort of FLC and performed next-generation sequencing of DNA methylation in a training set of pure-FLCs and non-cirrhotic hepatocellular carcinomas (nc-HCC). DNA methylation was correlated with gene expression. Furthermore, we established and validated an epigenetic signature differentiating pure-FLC from other HCCs. LINE-1 methylation correlated with shorter recurrence-free survival and overall survival in resected pure-FLC patients. Unsupervised clustering using CG sites located in islands distinguished pure-FLC from nc-HCC. Major DNA methylation changes occurred outside promoters, mainly in gene bodies and intergenic regions located in the vicinity of liver developmental genes (i.e., SMARCA4 and RXRA). Partially methylated domains were more prone to DNA methylation changes. Furthermore, we identified several putative tumor suppressor genes (e.g., DLEU7) and oncogenes (e.g., DUSP4). While ∼70% of identified gene promoters gaining methylation were marked by bivalent histone marks (H3K4me3/H3K27me3) in embryonic stem cells, ∼70% of those losing methylation were marked by H3K4me3. Finally, we established a pure FLC DNA methylation signature and validated it in an independent dataset. Our analysis reveals a distinct epigenetic signature of pure FLC as compared to nc-HCC, with DNA methylation changes occurring in the vicinity of liver developmental genes. These data suggest new options for targeting FLC based on cancer epigenome aberrations.


Journal of the National Cancer Institute | 2016

Hypomethylation of TET2 Target Genes Identifies a Curable Subset of Acute Myeloid Leukemia

Jumpei Yamazaki; Rodolphe Taby; Jaroslav Jelinek; Noël J.-M. Raynal; Matteo Cesaroni; Sherry Pierce; Steven M. Kornblau; Carlos E. Bueso-Ramos; Farhad Ravandi; Hagop M. Kantarjian; Jean-Pierre Issa

BACKGROUND Acute myeloid leukemia (AML) is curable in a subset of cases. The DNA methylation regulator TET2 is frequently mutated in AML, and we hypothesized that studying TET2-specific differentially methylated CpGs (tet2-DMCs) improves AML classification. METHODS We used bisulfite pyrosequencing to analyze the methylation status of four tet2-DMCs (SP140, MCCC1, EHMT1, and MTSS1) in a test group of 94 consecutive patients and a validation group of 92 consecutive patients treated with cytarabine-based chemotherapy. Data were analyzed with hierarchical clustering, Cox proportional hazards regression, and Kaplan-Meier analyses. All statistical tests were two-sided. RESULTS In the test cohort, hierarchical clustering analysis identified low levels of tet2-DMC methylation in 31 of 94 (33%) cases, and these had markedly longer overall survival (median survival 72+ vs 14 months, P = .002). Similar results were seen in the validation cohort. tet2-DMC-low status was shown to be an independent predictor of overall survival (hazard ratio = 0.29, P = .0002). In The Cancer Genome Atlas (TCGA) dataset where DNA methylation was analyzed by a different platform, tet2-DMC-low methylation was also associated with improved outcome (median survival = 55 vs 15 months, P = .0003) and was a better predictor of survival than mutations in TET2, IDH1, or IDH2, individually or combined. CONCLUSIONS Low levels of tet2-DMC methylation define a subgroup of AML that is highly curable and cannot be identified solely by genetic and cytogenetic analyses.


Drug Discovery in Cancer Epigenetics | 2016

DNA Methyltransferase Inhibitors

Noël J.-M. Raynal; Jean-Pierre Issa

Targeting epigenetic aberrations in cancer is now referred as to epigenetic therapy and targeting DNA hypermethylation specifically has been very successful in the treatment of leukemias. Two cytosine analogs inducing DNA methyltransferase inhibition, 5-azacytidine and 5-aza-2′-deoxycytidine, have been approved for the treatment of myelodysplastic syndromes (MDS) and acute myelogenous leukemia. Hypomethylating drugs induce reactivation of silenced tumor suppressor genes and cancer cell differentiation at low doses. In patients with MDS, these epigenetic drugs reduce transfusion frequency and prolong survival. Preclinical research and clinical trials are being pursued to improve their clinical efficacy and to understand the mechanisms of drug resistance. A second generation of DNA methyltransferase inhibitors were designed to address these issues and are currently in clinical trials. In this chapter, we will focus on the mechanism of action of 5-azacytidine and 5-aza-2′-deoxycytidine and highlight the historical and future perspective of DNA methyltransferase inhibitors.


Cancer Research | 2014

Abstract 380: Discovery of new epigenetic drugs among FDA-approved drug libraries

Noël J.-M. Raynal; Justin T. Lee; Youjun Wang; Judith Garriga; Gabriel G. Malouf; Sarah Naomie Dumont; Elisha J. Dettman; Vazganush Gharibyan; Saira Ahmed; Woonbok Chung; Wayne E. Childers; Magid Abou-Gharbia; Ryan A. Henry; Andrew J. Andrews; Jaroslav Jelinek; Ying Cui; Stephen B. Baylin; Donald L. Gill; Jean-Pierre Issa

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Targeting epigenetic pathways to induce tumor suppressor gene reactivation is a promising approach for cancer therapy. Epigenetic drugs produce impressive responses in some patients; however, treatment options are limited to a small number of drugs. We screened drug libraries for epigenetic activity using a live cell-based assay. We found that around 1% of US-FDA approved drugs have significant epigenetic activity detected by GFP reactivation of a DNA methylated and silenced promoter in colon cancer cells. Newly identified drugs, most prominently cardiac glycosides, induced tumor suppressor gene reactivation and showed selective anticancer apoptosis induction. These drugs did not change DNA methylation or histone acetylation globally but induced gene reactivation and cancer cell killing through calcium signaling leading to nuclear exclusion of chromatin repressors. Our data identify new epigenetic drugs that can be rapidly repurposed for cancer clinical trials and raise questions about the safety of commonly used medications. Citation Format: Noel J. Raynal, Justin T. Lee, Youjun Wang, Judith Garriga, Gabriel Malouf, Sarah Dumont, Elisha J. Dettman, Vazganush Gharibyan, Saira Ahmed, Woonbok Chung, Wayne E. Childers, Magid Abou-Gharbia, Ryan A. Henry, Andrew Andrews, Jaroslav Jelinek, Ying Cui, Stephen B. Baylin, Donald L. Gill, Jean-Pierre J. Issa. Discovery of new epigenetic drugs among FDA-approved drug libraries. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 380. doi:10.1158/1538-7445.AM2014-380


bioRxiv | 2018

Targeting MYC Overexpressing Leukemia with Cardiac Glycoside Proscillaridin Through Downregulation of Histone Acetyltransferases

Elodie M. Da Costa; Gregory Armaos; Gabrielle McInnes; Annie Beaudry; Gael Moquin-Beaudry; Virginie Bertrand-Lehouillier; Maxime Caron; Chantal Richer; Pascal St-Onge; Jeffrey R. Johnson; Nevan J. Krogan; Yuka Sai; Michael Downey; Moutih Rafei; Meaghan Boileau; Kolja Eppert; Ema Flores-Diaz; André Haman; Trang Hoang; Daniel Sinnett; Christian M. Beauséjour; Serge McGraw; Noël J.-M. Raynal

Targeting MYC oncogene remains a major therapeutic goal in cancer chemotherapy. Here, we demonstrate that proscillaridin, a cardiac glycoside approved for heart failure treatment, causing Na+/K+ pump inhibition, targets efficiently MYC overexpressing cancer cells. At clinically relevant doses, proscillaridin induced rapid downregulation of MYC protein level, and produced growth inhibition preferentially against MYC overexpressing leukemic cell lines including lymphoid and myeloid stem cell populations. Transcriptomic profile of leukemic cells after treatment showed a downregulation of gene sets involved in MYC pathways, cell replication and an upregulation of genes involved in hematopoietic differentiation. Gene expression changes were associated with an epigenetic remodeling of chromatin active marks. Proscillaridin induced a significant loss of lysine acetylation in histone H3 (at lysine 9, 14, 18 and 27). In addition, loss of lysine acetylation was observed also in non-histone proteins such as MYC itself, MYC target proteins, and a series of histone acetylation regulators. Global loss of acetylation correlated with the rapid downregulation of histone acetyltransferase proteins (such as CBP and P300) involved in histone and MYC acetylation. Overall, these results strongly support the repurposing of proscillaridin in MYC overexpressing leukemia and suggest a novel strategy to target MYC by inducing the downregulation of histone acetyltransferases involved in its stability.

Collaboration


Dive into the Noël J.-M. Raynal'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

Annie Beaudry

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Takahiro Sato

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge