Paul Delrée
Université libre de Bruxelles
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Paul Delrée.
The Prostate | 2012
Th. Roumeguère; Paul Delrée; P. Van Antwerpen; Sandrine Rorive; Luc Vanhamme; L. de la Kethulle de Ryhove; Didier Serteyn; Eric Wespes; M. Vanhaerverbeek; K. Zouaoui Boudjeltia
Myeloperoxidase (MPO) is a member of the peroxidase‐cyclooxygenase superfamily, which is secreted from stimulated leucocytes at inflammatory sites. It is well known that MPO catalyses oxidation reactions via the release of reactive halogenating and nitrating species and thus induces tissue damage. Several studies have already implicated MPO in the development of neoplasia. Chronic or recurrent prostatic inflammation has long been recognized as having the potential to initiate and promote the development of prostate cancer. The objective was to investigate whether MPO is present in the prostate.
Breast Cancer Research | 2014
Sébastien Toffoli; Isabelle Bar; Fadi Abdel-Sater; Paul Delrée; Pascale Hilbert; Frédéric Cavallin; Fabrice Moreau; Wim Van Criekinge; Magali Lacroix-Triki; Mario Campone; Anne-Laure Martin; Henri Roché; Jean-Pascal Machiels; Javier Carrasco; Jean-Luc Canon
IntroductionTriple Negative Breast Cancers (TNBC) represent about 12% to 20% of all breast cancers (BC) and have a worse outcome compared to other BC subtypes. TNBC often show a deficiency in DNA double-strand break repair mechanisms. This is generally related to the inactivation of a repair enzymatic complex involving BRCA1 caused either by genetic mutations, epigenetic modifications or by post-transcriptional regulations.The identification of new molecular biomarkers that would allow the rapid identification of BC presenting a BRCA1 deficiency could be useful to select patients who could benefit from PARP inhibitors, alkylating agents or platinum-based chemotherapy.MethodsGenomic DNA from 131 formalin-fixed paraffin-embedded (FFPE) tumors (luminal A and B, HER2+ and triple negative BC) with known BRCA1 mutation status or unscreened for BRCA1 mutation were analysed by array Comparative Genomic Hybridization (array CGH). One highly significant and recurrent gain in the 17q25.3 genomic region was analysed by fluorescent in situ hybridization (FISH). Expression of the genes of the 17q25.3 amplicon was studied using customized Taqman low density arrays and single Taqman assays (Applied Biosystems).ResultsWe identified by array CGH and confirmed by FISH a gain in the 17q25.3 genomic region in 90% of the BRCA1 mutated tumors. This chromosomal gain was present in only 28.6% of the BRCA1 non-mutated TNBC, 26.7% of the unscreened TNBC, 13.6% of the luminal B, 19.0% of the HER2+ and 0% of the luminal A breast cancers. The 17q25.3 gain was also detected in 50% of the TNBC with BRCA1 promoter methylation. Interestingly, BRCA1 promoter methylation was never detected in BRCA1 mutated BC. Gene expression analyses of the 17q25.3 sub-region showed a significant over-expression of 17 genes in BRCA1 mutated TNBC (n = 15) as compared to the BRCA1 non mutated TNBC (n = 13).ConclusionsIn this study, we have identified by array CGH and confirmed by FISH a recurrent gain in 17q25.3 significantly associated to BRCA1 mutated TNBC. Up-regulated genes in the 17q25.3 amplicon might represent potential therapeutic targets and warrant further investigation.
PLOS ONE | 2013
Otto Metzger-Filho; A. Catteau; Stefan Michiels; Marc Buyse; Michail Ignatiadis; Kamal S. Saini; Evandro de Azambuja; Virginie Fasolo; Sihem Naji; Jean Luc Canon; Paul Delrée; Michel Coibion; Pino Cusumano; Veronique Jossa; Jean-Pierre Kains; Denis Larsimont; Vincent Richard; Daniel Faverly; Nathalie Cornez; Peter Vuylsteke; Brigitte Vanderschueren; Hélène Peyro-Saint-Paul; Martine Piccart; Christos Sotiriou
Purpose Genomic Grade Index (GGI) is a 97-gene signature that improves histologic grade (HG) classification in invasive breast carcinoma. In this prospective study we sought to evaluate the feasibility of performing GGI in routine clinical practice and its impact on treatment recommendations. Methods Patients with pT1pT2 or operable pT3, N0-3 invasive breast carcinoma were recruited from 8 centers in Belgium. Fresh surgical samples were sent at room temperature in the MapQuant Dx™ PathKit for centralized genomic analysis. Genomic profiles were determined using Affymetrix U133 Plus 2.0 and GGI calculated using the MapQuant Dx® protocol, which defines tumors as low or high Genomic Grade (GG-1 and GG-3 respectively). Results 180 pts were recruited and 155 were eligible. The MapQuant test was performed in 142 cases and GGI was obtained in 78% of cases (n=111). Reasons for failures were 15 samples with <30% of invasive tumor cells (11%), 15 with insufficient RNA quality (10%), and 1 failed hybridization (<1%). For tumors with an available representative sample (≥ 30% inv. tumor cells) (n=127), the success rate was 87.5%. GGI reclassified 69% of the 54 HG2 tumors as GG-1 (54%) or GG-3 (46%). Changes in treatment recommendations occurred mainly in the subset of HG2 tumors reclassified into GG-3, with increased use of chemotherapy in this subset. Conclusion The use of GGI is feasible in routine clinical practice and impacts treatment decisions in early-stage breast cancer. Trial Registration ClinicalTrials.gov NCT01916837, http://clinicaltrials.gov/ct2/show/NCT01916837
PLOS ONE | 2012
Karim Zouaoui Boudjeltia; Jalil Daher; Pierre Van Antwerpen; Nicole Moguilevsky; Paul Delrée; Jean Ducobu; Martine Raes; Bassam Badran; Michel Vanhaeverbeek; Dany Brohée; Claude Remacle; Luc Vanhamme
Background Blood fluidity is maintained by a delicate balance between coagulation and fibrinolysis. The endothelial cell surface is a key player in this equilibrium and cell surface disruptions can upset the balance. We investigated the role of pericellular myeloperoxidase oxidized LDLs (Mox-LDLs) in this balance. Methods and Results We designed a technical device that enabled us to monitor fibrinolysis in real-time at the surface of an endothelial cell line (EA.hy926), and showed that Mox-LDL decreased pericellular fibrinolysis. There were no changes in fibrinolysis when EA.hy926 endothelial cells were exposed to native LDL (24 hours) at doses of 10, 50, 100 and up to 1250 µg/ml. However, treatment of EA.hy926 endothelial cells with 10 and 50 µg/ml of Mox-LDL (physiological serum concentrations) increased the lysis time by 15 and 13%, respectively (p<0.001), although this effect was not present at higher concentrations of 100 µg/ml. This effect was not correlated with any changes in PAI-1 or t-PA or PA Receptor (PAR) expression. No effect was observed at the surface of smooth muscle cells used as controls. Conclusion Our data link the current favorite hypothesis that modified LDL has a causal role in atheroma plaque formation with an old suggestion that fibrin may also play a causal role. Our data help complete the paradigm of atherosclerosis: Modified LDL locally enhances fibrin deposition (present work); fibrin deposits enhance endothelial permeability; this effect allows subendothelial accumulation of lipid and foam cells.
Oncology Letters | 2016
Didier Dequanter; Maureen Van de Velde; Isabelle Bar; Vincent Nuyens; Alexandre Rousseau; Nathalie Nagy; Luc Vanhamm; Michel Vanhaeverbeek; Dany Brohée; Paul Delrée; Karim Zouaoui Boudjeltia; Philippe Lothaire; Pierrick Uzureau
Glutathione (GSH) is the keystone of the cellular response toward oxidative stress. Elevated GSH content correlates with increased resistance to chemotherapy and radiotherapy of head and neck (HN) tumors. The purpose of the present cross-sectional study was to evaluate whether the expression of glutamate-cysteine ligase (GCL) accounts for the increased GSH availability observed in HN squamous cell carcinoma (SCC). For that purpose, the messenger (m)RNA levels of the modifier (M) and catalytic (C) subunits of GCL and its putative regulators (namely, nuclear factor erythroid 2-related factor 2, heme oxygenase-1 and nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha) were monitored in 35 surgical resections of untreated HNSCC. The localization of GCLM was evaluated using in situ hybridization and immunohistochemistry. GCLM expression was significantly increased in tumor samples, compared with normal mucosa, both at the mRNA and protein level (P=0.029), but the pathway of GCLM activation remains to be elucidated. Protein expression of GCLM was detected in the cytoplasm and nucleus. GCLM and the proliferation marker Ki-67 displayed a similar distribution, being both mainly expressed at the periphery of tumor lobules. The present study reported increased expression of GCL and the rate-limiting enzyme of GSH synthesis, within HNSCC. The nuclear localization of GCLM and the concomitant expression of Ki-67 suggested that the localization of GSH synthesis contributes to the protection against oxidative stress within hotspots of cell proliferation.
Journal of Histochemistry and Cytochemistry | 2017
Isabelle Bar; Ahmad Merhi; Fadi Abdel-Sater; Abduelhakem Ben Addi; Sara Sollennita; Jean-Luc Canon; Paul Delrée
The triple-negative breast cancer (TNBC) subtype occurs in about 15% of breast cancer and is an aggressive subtype of breast cancer with poor outcome. Furthermore, treatment of patients with TNBC is more challenging due to the heterogeneity of the disease and the absence of well-defined molecular targets. Microribonucleic acid (RNA) represents a new class of biomarkers that are frequently dysregulated in cancer. It has been described that the microRNA miR-210 is highly expressed in TNBC, and its overexpression had been linked to poor prognosis. TNBC are often infiltrated by immune cells that play a key role in cancer progression. The techniques traditionally used to analyze miR-210 expression such as next generation sequencing or quantitative real-time polymerase chain reaction (PCR) do not allow the precise identification of the cellular subtype expressing the microRNA. In this study, we have analyzed miR-210 expression by in situ hybridization in TNBC. The miR-210 signal was detected in tumor cells, but also in the tumor microenvironment, in a region positive for the pan-leucocyte marker CD45-LCA. Taken together, our results demonstrate that miR-210 is expressed in tumor cells but also in the tumor microenvironment. Our results also highlight the utility of using complementary approaches to take into account the cellular context of microRNA expression.
Pathology Research and Practice | 2016
Mounia Chidiac; Mohammad Fayyad-Kazan; Jalil Daher; Philippe Poelvoorde; Isabelle Bar; Carine Maenhaut; Paul Delrée; Bassam Badran; Luc Vanhamme
The apolipoprotein L (apoL) family has not yet been ascribed any definite patho-physiological function although the conserved BH3 protein domain suggests a role in programmed cell death. As repression of the regular apoptotic program is considered a hallmark of tumor progression, we investigated apoL expression in cancer. We show that the levels of one member of the family, apolipoprotein L1 (apoL1) is higher in papillary thyroid carcinoma compared to normal tissue. A combination of qRTPCR, immunohistochemistry and in situ hybridization allowed us to ascribe this increase to endogenous overexpression in carcinoma cells. Whether apoL1 plays an instrumental role in refraining cell death is the subject of ongoing molecular biology experiments.
Oncotarget | 2018
Isabelle Bar; Ahmad Merhi; Lionel Larbanoix; Manuel Constant; Sandy Haussy; Sophie Laurent; Jean-Luc Canon; Paul Delrée
The casein kinase 1 delta (CSNK1D) is a conserved serine/threonine protein kinase that regulates diverse cellular processes including cell cycle progression, circadian rhythm, and neurite outgrowth. Aberrant expression of CSNK1D is described in several cancer types including breast cancer, where it is amplified in about 30% of triple negative breast (TNBC). Here, we have investigated the function of CSNK1D in triple negative cancer cell migration and metastasis. By using immunohistochemistry and in situ hybridization, we found that CNSK1D is highly expressed in primary tumor cells and in tumor cells invading lymphatic nodes compared to non-metastatic tumors. In vitro, knock-down of CSNK1D expression with specific shRNAs in the breast cancer cell line MDA-MB-231 markedly inhibited cancer cell proliferation, invasion and migration and affected the expression of the tight junction proteins claudin 1, occludin and the junction adhesion molecule A. In vivo, the inactivation of CSNK1D reduced lung metastasis in MDA-MB-231 breast cancer xenografts. Altogether, our results indicate that the downregulation of CSNK1D expression inhibits the proliferation and reduces the migration and the metastasis of breast cancer cells. As numerous inhibitors of CSNK1D are currently under development, this might represent an attractive therapeutic target for the treatment of TNBC.
Archives of Biochemistry and Biophysics | 2018
Alia Khalil; Hayfa Medfai; Philippe Poelvoorde; Mohammad Fayyad Kazan; Cédric Delporte; Pierre Van Antwerpen; Yolla EL-Makhour; Patrick Biston; Paul Delrée; Bassam Badran; Luc Vanhamme; Karim Zouaoui Boudjeltia
Myeloperoxidase is a member of the mammalian peroxidase family, mainly expressed in the myeloblastic cell lineage. It is considered a major bactericidal agent as it is released in the phagosome where it catalyzes the formation of reactive oxygen species. It is also released in the extracellular spaces including blood where it is absorbed on (lipo)proteins and endothelial cell surface, interfering with endothelial function. We performed RNA sequencing on MPO-treated endothelial cells, analyzed their transcriptome and validated the profile of gene expression by individual qRT-PCR. Some of the induced genes could be grouped in several functional networks, including tubulogenesis, angiogenesis, and blood vessel morphogenesis and development as well as signal transduction pathways associated to these mechanisms. MPO treatment mimicked the effects of VEGF on several signal transduction pathways, such as Akt, ERK or FAK involved in angiogenesis. Accordingly MPO, independently of its enzymatic activity, stimulated tube formation by endothelial cells. RNA interference also pointed at a role of endogenous MPO in tubulogenesis and endothelium wound repair in vitro. These data suggest that MPO, whether from endogenous or exogenous sources, could play a role in angiogenesis and vascular repair in vivo.
Cancer Research | 2014
Ludovic Dhont; Alisson D. Stefano; Angela Szucs; Fadi Abdulsater; Isabelle Bar; Paul Delrée; Alexandra Belayew
Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Background : HLTF, beside its roles in DNA binding, transcriptional and chromatin remodeling, is involved in post-replication DNA repair through its translocase and E3 ubiquitin ligase activities. HLTF is a tumor suppressor, and hypermethylation of its promoter was observed in 30-50% of gastrointestinal cancers. Moreover, HLTF inactivation in Apc-/+ mice induced the transition from colon adenocarcinoma to a carcinoma with high chromosomal instability. The epigenetic heterogeneity of the HLTF promoter could be related to the CpG island methylator phenotype of tumor suppressor genes in colorectal cancers. In the present study we started to investigate HLTF expression in colon cancer progression in relation to the epigenetic status of its gene promoter by using pyrosequencing. Methods : The HLTF promoter contains a 653 bp CpG island with 59 CpG sites (UCSC) among which 11 CpG sites were selected for this study. Genomic DNA was extracted from (a) HeLa (cervix adenocarcinoma) and RKO (colon carcinoma) cells and (b) from formalin-fixed paraffin-embedded (FFPE) colon tumors. DNA was treated with bisulfite and amplified by PCR the HLTF promoter fragment containing the 11 CpG sites. Pyrosequencing was performed to determine the relative methylation frequency at each site (HLTF PyroMark CpG Assay, Qiagen). HLTF expression was evaluated by immunohistochemistry (IHC) on tissue sections, and by RT-PCR and immunodetection on Western blot for the cell lines. Results : There was a negative correlation between promoter methylation and HLTF expression at the RNA and protein levels in RKO and HeLa cells. The average methylation rates were 92% for RKO and 25% for HeLa cells which presented HLTF repression and expression, respectively. IHC staining for HLTF was performed in 14 FFPE colon tumors and good pyrosequencing signals was obtained for DNA extracted from 6/14 samples. Three tumor groups were defined, based on the average methylation rate in the HLTF promoter : (a) 60%. In groups (a) and (b), the tissue was still well organized and HTLF protein expression was detected by IHC in the glandular epitelium. However, in group (c), the tumors were disorganized and HLTF protein expression was low or undetectable. These preliminary results suggest a correlation between 3 criteria : HLTF immunodetection, tissue organization and low methylation of the HLTF promoter. Two colon tumors did not express HLTF and presented a hypermethylated promoter. Conclusion : We could optimize pyrosequencing methylation analysis of genomic DNA extracted from 6 FFPE colon tumors, and confirmed that higher HLTF promoter methylation was associated with its repression in correlation with worse tumor phenotypes. This sensitive technique will now be applied to larger number of samples and additional CpG sites will be assessed. Citation Format: Ludovic Dhont, Alisson Di Stefano, Angela Szucs, Fadi Abdulsater, Isabelle Bar, Paul Delree, Alexandra Belayew. Helicase-like transcription factor (HLTF) expression and gene methylation in FFPE colon tumors. [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 392. doi:10.1158/1538-7445.AM2014-392