Network


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

Hotspot


Dive into the research topics where Michel Janicot is active.

Publication


Featured researches published by Michel Janicot.


Molecular and Cellular Biology | 2005

Disruption of Murine Mus81 Increases Genomic Instability and DNA Damage Sensitivity but Does Not Promote Tumorigenesis

Najoua Dendouga; Hui Gao; Dieder Moechars; Michel Janicot; Jorge Vialard; Clare H. McGowan

ABSTRACT The Mus81-Eme1 endonuclease is implicated in the efficient rescue of broken replication forks in Saccharomyces cerevisiae and Schizosaccharomyces pombe. We have used gene targeting to study the function of the Mus81-Eme1 endonuclease in mammalian cells. Mus81-deficient mice develop normally and are fertile. Surprisingly, embryonic fibroblasts from Mus81−/− animals fail to proliferate in vitro. This proliferation defect can be rescued by expression of the papillomavirus E6 protein that promotes degradation of p53. When grown in culture, Mus81−/− cells have elevated levels of DNA damage, acquire chromosomal aberrations, and are hypersensitive to agents that generate DNA cross-links. In contrast to the situation in yeast, murine Mus81 is not required for replication restart following camptothecin treatment. Mus81−/− mice and cells are hypersensitive to DNA cross-linking agents. Cross-link-induced double-strand break formation is normal in Mus81−/− cells, but the resolution of repair intermediates is not. The persistence of Rad51 foci in Mus81−/− cells suggests that Mus81 acts at a late step in the repair of cross-link-induced lesions. Despite these defects, Mus81−/− mice do not show increased predisposition to lymphoma or any other malignancy in the first year of life.


Molecular Cancer Research | 2005

p53-Independent Regulation of p21Waf1/Cip1 Expression and Senescence by Chk2

Cécile-Marie Aliouat-Denis; Najoua Dendouga; Ilse Van den Wyngaert; Hinrich Goehlmann; Ulf Steller; Inez Van de Weyer; Nele Van Slycken; Luc Andries; Stefan U. Kass; Walter Luyten; Michel Janicot; Jorge Vialard

The Chk2 kinase is a tumor suppressor and key component of the DNA damage checkpoint response that encompasses cell cycle arrest, apoptosis, and DNA repair. It has also been shown to have a role in replicative senescence resulting from dysfunctional telomeres. Some of these functions are at least partially exerted through activation of the p53 transcription factor. High-level expression of virally transduced Chk2 in A549 human lung carcinoma cells led to arrested proliferation, apoptosis, and senescence. These were accompanied by various molecular events, including p21Waf1/Cip1 (p21) transcriptional induction, consistent with p53 activation. However, Chk2-dependent senescence and p21 transcriptional induction also occurred in p53-defective SK-BR-3 (breast carcinoma) and HaCaT (immortalized keratinocyte) cells. Small interfering RNA–mediated knockdown of p21 in p53-defective cells expressing Chk2 resulted in a decrease in senescent cells. These results revealed a p53-independent role for Chk2 in p21 induction and senescence that may contribute to tumor suppression and genotoxic treatment outcome.


Clinical Cancer Research | 2009

JNJ-26481585, a Novel “Second-Generation” Oral Histone Deacetylase Inhibitor, Shows Broad-Spectrum Preclinical Antitumoral Activity

Janine Arts; Peter King; Ann Marien; Wim Floren; Ann Beliën; Lut Janssen; Isabelle Noëlle Constance Pilatte; Bruno Roux; Laurence Decrane; Ron Gilissen; Ian Hickson; Veronique Vreys; Eugene Cox; Kees Bol; Willem Talloen; Ilse Goris; Luc Andries; Marc Du Jardin; Michel Janicot; Martin John Page; Kristof Van Emelen; Patrick René Angibaud

Purpose: Histone deacetylase (HDAC) inhibitors have shown promising clinical activity in the treatment of hematologic malignancies, but their activity in solid tumor indications has been limited. Most HDAC inhibitors in clinical development only transiently induce histone acetylation in tumor tissue. Here, we sought to identify a second-generation class I HDAC inhibitor with prolonged pharmacodynamic response in vivo, to assess whether this results in superior antitumoral efficacy. Experimental Design: To identify novel HDAC inhibitors with superior pharmacodynamic properties, we developed a preclinical in vivo tumor model, in which tumor cells have been engineered to express fluorescent protein dependent on HDAC1 inhibition, thereby allowing noninvasive real-time evaluation of the tumor response to HDAC inhibitors. Results:In vivo pharmacodynamic analysis of 140 potent pyrimidyl-hydroxamic acid analogues resulted in the identification of JNJ-26481585. Once daily oral administration of JNJ-26481585 induced continuous histone H3 acetylation. The prolonged pharmacodynamic response translated into complete tumor growth inhibition in Ras mutant HCT116 colon carcinoma xenografts, whereas 5-fluorouracil was less active. JNJ-26481585 also fully inhibited the growth of C170HM2 colorectal liver metastases, whereas again 5-fluorouracil/Leucovorin showed modest activity. Further characterization revealed that JNJ-26481585 is a pan-HDAC inhibitor with marked potency toward HDAC1 (IC50, 0.16 nmol/L). Conclusions: The potent antitumor activity as a single agent in preclinical models combined with its favorable pharmacodynamic profile makes JNJ-26481585 a promising second-generation HDAC inhibitor. The compound is currently in clinical studies, to evaluate its potential applicability in a broad spectrum of both solid and hematologic malignancies. (Clin Cancer Res 2009;15(22):684151)


Journal of Drug Targeting | 2006

Modified poly(propylene imine) dendrimers as effective transfection agents for catalytic DNA enzymes (DNAzymes).

Frederik Tack; Annette Bakker; Sophie Maes; Nathalie Dekeyser; Monique Bruining; Cristina Elissen-Roman; Michel Janicot; Marcus E. Brewster; Henk M. Janssen; B. F. M. De Waal; P.M. Fransen; Xianwen Lou; E. W. Meijer

The major bottleneck in gene therapy remains the issue of delivery. In this work, various modified poly(propylene imine) (PPI) dendrimers are introduced as gene transfection agents. Commercially available PPI-dendrimers have been modified (i) at the exterior primary amines with acetyl groups or glycol gallate (PEG-like) groups, and (ii) at the interior tertiary amines with methyl iodide (MeI) or MeCl to produce multiple quaternized cationic sites in the core of the dendrimer. The prepared materials have been tested with respect to their binding capabilities to DNA, their toxicity in cell cultures, their in vitro transfection efficiency and their in vivo delivery possibilities. In all cases, a 33-mer oligonucleotide (DNAzyme) was used. Polyacrylamide gel electrophoresis (PAGE) studies have demonstrated strong but reversible binding, where the quarternized and higher generation dendrimer species have shown more potent binding. Typically, for the modified fourth PPI-dendrimers, binding is observed at a concentration of about 4 μM DNA and a dendrimer-DNA charge ratio of around 2:1–1:1. All the tested PPI-dendrimers display a low cellular toxicity, especially when higher serum contents are used in the culture medium. For example, most of the prepared fourth generation PPI-dendrimers are not or hardly toxic up to at least 20 μM in 20% serum. An in vitro characterization has revealed a high dendrimer-mediated intracellular uptake of the DNAzyme: all the tested fourth generation PPI-dendrimers display transfection efficiencies close to or exceeding 80%, even when the concentration of serum in the medium is increased from 10 to 40%. Finally, the potential of using modified PPI-dendrimers for in vivo gene therapy experiments is demonstrated. Injecting a G4-PEG(MeI)–ssDNA complex intravenously into Nude mice has resulted in a high nuclear uptake as confirmed by co-localization studies.


Investigational New Drugs | 2006

The Wnt-dependent signaling pathways as target in oncology drug discovery.

Nico Janssens; Michel Janicot; Tim Perera

SummaryOur current understanding of the Wnt-dependent signaling pathways is mainly based on studies performed in a number of model organisms including, Xenopus, Drosophila melanogaster, Caenorhabditis elegans and mammals. These studies clearly indicate that the Wnt-dependent signaling pathways are conserved through evolution and control many events during embryonic development. Wnt pathways have been shown to regulate cell proliferation, morphology, motility as well as cell fate. The increasing interest of the scientific community, over the last decade, in the Wnt-dependent signaling pathways is supported by the documented importance of these pathways in a broad range of physiological conditions and disease states. For instance, it has been shown that inappropriate regulation and activation of these pathways is associated with several pathological disorders including cancer, retinopathy, tetra-amelia and bone and cartilage disease such as arthritis. In addition, several components of the Wnt-dependent signaling pathways appear to play important roles in diseases such as Alzheimer’s disease, schizophrenia, bipolar disorder and in the emerging field of stem cell research. In this review, we wish to present a focused overview of the function of the Wnt-dependent signaling pathways and their role in oncogenesis and cancer development. We also want to provide information on a selection of potential drug targets within these pathways for oncology drug discovery, and summarize current data on approaches, including the development of small-molecule inhibitors, that have shown relevant effects on the Wnt-dependent signaling pathways.


Molecular Diagnosis | 2004

Housekeeping genes as internal standards in cancer research

Nico Janssens; Michel Janicot; Tim Perera; Annette Bakker

AbstractBackground: Differences in gene expression are frequently encountered in malignant tissues, and have been intensively studied as they can reflect different experimental or clinical conditions. Quantification of the often subtle changes in messenger RNA content is performed through comparison with the expression of endogenous controls. The appropriate choice of these endogenous controls (e.g. housekeeping genes) is critical for meaningful quantitative RNA analysis. The most important characteristics of housekeeping genes are that they are present in all cells and that their expression levels remain relatively constant in different experimental conditions. However, no single housekeeping gene always manifests stable expression levels under all experimental conditions. Therefore, it is necessary to characterize the suitability of various housekeeping genes to serve as internal RNA controls under particular experimental conditions where transcription effects are being tested. Aim: It was the aim of this study to determine the validity of a number of housekeeping genes for their use as internal standards in cancer research. Methods: The expression of the housekeeping genes porphobilinogen deaminase (PBGD) and mitochondrial ATP synthase 6 (mATPsy6), were compared with the expression of the more commonly used glyceraldehyde-3-phosphate dehydrogenase (GAPDH). We examined a number of cell lines and tumor versus matched normal tissue samples using real-time quantitative (RTq)-PCR. Results: Our findings suggest that in cell lines, all three of the studied housekeeping genes can be used as an internal control. When comparing tumor tissue samples with matched normal tissue samples, we validated mitochondrial ATPsy6 (mATPsy6) as the best choice for a housekeeping gene. Conclusion: Since gene expression studies are becoming increasingly important in the clinical environment, especially in cancer diagnosis and treatment, the use of an reliable housekeeping gene in these studies to normalize gene expression is essential. We conclude that a bad choice of housekeeping gene may lead to errors when interpreting experiments involving quantitation of gene expression. Our study demonstrated the usefulness of mATPsy6 as an endogenous control when comparing tumor tissue samples with normal tissue samples.


Cancer Letters | 2011

Preclinical assessment of JNJ-26854165 (Serdemetan), a novel tryptamine compound with radiosensitizing activity in vitro and in tumor xenografts.

Cyrus Chargari; Céline Leteur; Eric Angevin; Tarig Bashir; Bruno Schoentjes; Janine Arts; Michel Janicot; Jean Bourhis; Eric Deutsch

Serdemetan (JNJ-26854165) is a novel tryptamine compound with antiproliferative activity in various p53 wild-type (WT) tumor cell lines. We investigated its potential as radiosensitizer using four human cancer cell lines: H460, A549, p53-WT-HCT116, and p53-null-HCT116. Serdemetan inhibited clonogenic survival in all cell lines, but in a lower extent in p53-null-HCT116. In the combination studies, Serdemetan treatment at 0.25μM in H460 and at 5μM in A549 cells resulted in a sensitivity-enhancement ratio of 1.18 and 1.36, respectively. At 2Gy, surviving fractions were 0.72 and 0.97 for p53-WT HCT116 and p53-null cells exposed to 0.5μM of Serdemetan, respectively (p<0.05). Radiosensitization of H460 and A549 cells was associated with G2/M cell cycle arrest and with an increased expression of p53 and p21. In vivo, Serdemetan caused a greater than additive increase in tumor growth delay. The dose enhancement factor was 1.9 and 1.6 for H460 and A549 tumors, respectively. Serdemetan inhibited proliferation, capillary tube formation and migration of HMEC-1 cells. These effects were more marked concurrently with irradiation. These results in tumor and endothelial cells suggest that Serdemetan has potential as a radiosensitizer. Further investigations are warranted with regard to the molecular mechanisms underlying its actions and its dependency regarding p53 status.


Molecular Cancer Therapeutics | 2006

Real-time gene expression analysis in human xenografts for evaluation of histone deacetylase inhibitors

Ann Beliën; Stefanie De Schepper; Wim Floren; Boud Janssens; Ann Marien; Peter King; Jacky Van Dun; Luc Andries; Jan Voeten; Luc Bijnens; Michel Janicot; Janine Arts

Real-time analysis of gene expression in experimental tumor models represents a major tool to document disease biology and evaluate disease treatment. However, monitoring gene regulation in vivo still is an emerging field, and thus far it has not been linked to long-term tumor growth and disease outcome. In this report, we describe the development and validation of a fluorescence-based gene expression model driven by the promoter of the cyclin-dependent kinase inhibitor p21waf1,cip1. The latter is a key regulator of tumor cell proliferation and a major determinant in the response to many anticancer agents such as histone deacetylase inhibitors. In response to histone deacetylase inhibitors, induction of fluorescence in A2780 ovarian tumors could be monitored in living mice in a noninvasive real-time manner using whole-body imaging. Single p.o. administration of the histone deacetylase inhibitor MS-275 significantly induces tumor fluorescence in a time- and dose-dependent manner, which accurately predicted long-term antitumoral efficacy in individual mice following extended treatment. These findings illustrate that this technology allows monitoring of the biological response induced by treatment with histone deacetylase inhibitors. In addition to providing experimental pharmacokinetic/pharmacodynamic markers for investigational drugs, this model provides insight into the kinetics of in vivo regulation of transcription, which plays a key role in causing and maintaining the uncontrolled proliferation of tumor tissue. [Mol Cancer Ther 2006;5(9):2317–24]


Oncotarget | 2016

TGF-β1 and TGF-β2 abundance in liver diseases of mice and men

A Dropmann; Tatjana Dediulia; Katja Breitkopf-Heinlein; Hanna Korhonen; Michel Janicot; Sn Weber; Maria Thomas; Albrecht Piiper; Esther Bertran; Isabel Fabregat; Kerstin Abshagen; Jochen Hess; Peter Angel; Cédric Coulouarn; Steven Dooley; Nm Meindl-Beinker

TGF-β1 is a major player in chronic liver diseases promoting fibrogenesis and tumorigenesis through various mechanisms. The expression and function of TGF-β2 have not been investigated thoroughly in liver disease to date. In this paper, we provide evidence that TGF-β2 expression correlates with fibrogenesis and liver cancer development. Using quantitative realtime PCR and ELISA, we show that TGF-β2 mRNA expression and secretion increased in murine HSCs and hepatocytes over time in culture and were found in the human-derived HSC cell line LX-2. TGF-β2 stimulation of the LX-2 cells led to upregulation of the TGF-β receptors 1, 2, and 3, whereas TGF-β1 treatment did not alter or decrease their expression. In liver regeneration and fibrosis upon CCl4 challenge, the transient increase of TGF-β2 expression was accompanied by TGF-β1 and collagen expression. In bile duct ligation-induced fibrosis, TGF-β2 upregulation correlated with fibrotic markers and was more prominent than TGF-β1 expression. Accordingly, MDR2-KO mice showed significant TGF-β2 upregulation within 3 to 15 months but minor TGF-β1 expression changes. In 5 of 8 hepatocellular carcinoma (HCC)/hepatoblastoma cell lines, relatively high TGF-β2 expression and secretion were observed, with some cell lines even secreting more TGF-β2 than TGF-β1. TGF-β2 was also upregulated in tumors of TGFα/cMyc and DEN-treated mice. The analysis of publically available microarray data of 13 human HCC collectives revealed considerable upregulation of TGF-β2 as compared to normal liver. Our study demonstrates upregulation of TGF-β2 in liver disease and suggests TGF-β2 as a promising therapeutic target for tackling fibrosis and HCC.


Bioorganic & Medicinal Chemistry Letters | 2010

Identification of a series of substituted 2-piperazinyl-5-pyrimidylhydroxamic acids as potent histone deacetylase inhibitors

Patrick René Angibaud; Kristof Van Emelen; Laurence Decrane; Sven Franciscus Anna Van Brandt; Peter Ten Holte; Isabelle Noëlle Constance Pilatte; Bruno Roux; Virginie Sophie Poncelet; David Speybrouck; Laurence Queguiner; Sandrine Gaurrand; Ann Marien; Wim Floren; Lut Janssen; Marc Gustaaf Celine Verdonck; Jacky Van Dun; Jacky van Gompel; Ron Gilissen; Claire Mackie; Marc Du Jardin; Jozef Peeters; Marc Noppe; Luc Van Hijfte; Eddy Jean Edgard Freyne; Martin John Page; Michel Janicot; Janine Arts

Pursuing our efforts in designing 5-pyrimidylhydroxamic acid anti-cancer agents, we have identified a new series of potent histone deacetylase (HDAC) inhibitors. These compounds exhibit enzymatic HDAC inhibiting properties with IC(50) values in the nanomolar range and inhibit tumor cell proliferation at similar levels. Good solubility, moderate bioavailability, and promising in vivo activity in xenograft model made this series of compounds interesting starting points to design new potent HDAC inhibitors.

Collaboration


Dive into the Michel Janicot's collaboration.

Top Co-Authors

Avatar

Katja Wosikowski

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ann Marien

University of Freiburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Luc Andries

University of Freiburg

View shared research outputs
Researchain Logo
Decentralizing Knowledge