100 results on '"Olivier, Peulen"'
Search Results
2. Identification of myoferlin as a mitochondria-associated membranes component required for calcium signaling in PDAC cell lines
- Author
-
Sandy Anania, Martin Farnir, Raphaël Peiffer, Yasmine Boumahd, Marc Thiry, Ferman Agirman, Naima Maloujahmoum, Akeila Bellahcène, and Olivier Peulen
- Subjects
Pancreatic cancer ,Mitochondria-associated membranes ,ER-mitochondria contact sites ,Myoferlin ,Calcium signaling ,IP3R3 ,Medicine ,Cytology ,QH573-671 - Abstract
Abstract Background Pancreatic ductal adenocarcinoma is an aggressive cancer type with one of the lowest survival rates due to late diagnosis and the absence of effective treatments. A better understanding of PDAC biology will help researchers to discover the Achilles’ heel of cancer cells. In that regard, our research team investigated the function of an emerging oncoprotein known as myoferlin. Myoferlin is overexpressed in PDAC and its silencing/targeting has been shown to affect cancer cell proliferation, migration, mitochondrial dynamics and metabolism. Nevertheless, our comprehension of myoferlin functions in cells remains limited. In this study, we aimed to understand the molecular mechanism linking myoferlin silencing to mitochondrial dynamics. Methods Experiments were performed on two pancreas cancer cell lines, Panc-1 and MiaPaCa-2. Myoferlin localization on mitochondria was evaluated by immunofluorescence, proximity ligation assay, and cell fractionation. The presence of myoferlin in mitochondria-associated membranes was assessed by cell fractionation and its function in mitochondrial calcium transfer was evaluated using calcium flow experiments, proximity ligation assays, co-immunoprecipitation, and timelapse fluorescence microscopy in living cells. Results Myoferlin localization on mitochondria was investigated. Our results suggest that myoferlin is unlikely to be located on mitochondria. Instead, we identified myoferlin as a new component of mitochondria-associated membranes. Its silencing significantly reduces the mitochondrial calcium level upon stimulation, probably through myoferlin interaction with the inositol 1,4,5-triphosphate receptors 3. Conclusions For the first time, myoferlin was specifically demonstrated to be located in mitochondria-associated membranes where it participates to calcium flow. We hypothesized that this function explains our previous results on mitochondrial dynamics. This study improves our comprehension of myoferlin localization and function in cancer biology.
- Published
- 2024
- Full Text
- View/download PDF
3. Methylglyoxal: a novel upstream regulator of DNA methylation
- Author
-
Gaurav Dube, Assia Tiamiou, Martin Bizet, Yasmine Boumahd, Imène Gasmi, Rebekah Crake, Justine Bellier, Marie-Julie Nokin, Emilie Calonne, Rachel Deplus, Tom Wissocq, Olivier Peulen, Vincent Castronovo, François Fuks, and Akeila Bellahcène
- Subjects
Methylglyoxal ,DNA methylation ,Breast cancer ,Tumor suppressor genes ,Metastasis ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Aerobic glycolysis, also known as the Warburg effect, is predominantly upregulated in a variety of solid tumors, including breast cancer. We have previously reported that methylglyoxal (MG), a very reactive by-product of glycolysis, unexpectedly enhanced the metastatic potential in triple negative breast cancer (TNBC) cells. MG and MG-derived glycation products have been associated with various diseases, such as diabetes, neurodegenerative disorders, and cancer. Glyoxalase 1 (GLO1) exerts an anti-glycation defense by detoxifying MG to D-lactate. Methods Here, we used our validated model consisting of stable GLO1 depletion to induce MG stress in TNBC cells. Using genome-scale DNA methylation analysis, we report that this condition resulted in DNA hypermethylation in TNBC cells and xenografts. Results GLO1-depleted breast cancer cells showed elevated expression of DNMT3B methyltransferase and significant loss of metastasis-related tumor suppressor genes, as assessed using integrated analysis of methylome and transcriptome data. Interestingly, MG scavengers revealed to be as potent as typical DNA demethylating agents at triggering the re-expression of representative silenced genes. Importantly, we delineated an epigenomic MG signature that effectively stratified TNBC patients based on survival. Conclusion This study emphasizes the importance of MG oncometabolite, occurring downstream of the Warburg effect, as a novel epigenetic regulator and proposes MG scavengers to reverse altered patterns of gene expression in TNBC.
- Published
- 2023
- Full Text
- View/download PDF
4. Correction: Human colon cancer cells highly express myoferlin to maintain a fit mitochondrial network and escape p53-driven apoptosis
- Author
-
Gilles Rademaker, Brunella Costanza, Justine Bellier, Michael Herfs, Raphaël Peiffer, Ferman Agirman, Naïma Maloujahmoum, Yvette Habraken, Philippe Delvenne, Akeila Bellahcène, Vincent Castronovo, and Olivier Peulen
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
5. Novel XBP1s-independent function of IRE1 RNase in HIF-1α-mediated glycolysis upregulation in human macrophages upon stimulation with LPS or saturated fatty acid
- Author
-
Margaud Iovino, Megan Colonval, Chloé Wilkin, Laurent L’homme, Cédric Lassence, Manon Campas, Olivier Peulen, Pascal de Tullio, Jacques Piette, and Sylvie Legrand-Poels
- Subjects
saturated fatty acid ,macrophages ,glycolysis ,HIF-1α ,IRE1α ,inflammation ,Immunologic diseases. Allergy ,RC581-607 - Abstract
In obesity, adipose tissue infiltrating macrophages acquire a unique pro-inflammatory polarization, thereby playing a key role in the development of chronic inflammation and Type 2 diabetes. Increased saturated fatty acids (SFAs) levels have been proposed to drive this specific polarization. Accordingly, we investigated the immunometabolic reprogramming in SFA-treated human macrophages. As expected, RNA sequencing highlighted a pro-inflammatory profile but also metabolic signatures including glycolysis and hypoxia as well as a strong unfolded protein response. Glycolysis upregulation was confirmed in SFA-treated macrophages by measuring glycolytic gene expression, glucose uptake, lactate production and extracellular acidification rate. Like in LPS-stimulated macrophages, glycolysis activation in SFA-treated macrophages was dependent on HIF-1α activation and fueled the production of pro-inflammatory cytokines. SFAs and LPS both induced IRE1α endoribonuclease activity, as demonstrated by XBP1 mRNA splicing, but with different kinetics matching HIF-1α activation and the glycolytic gene expression. Interestingly, the knockdown of IRE1α and/or the pharmacological inhibition of its RNase activity prevented HIF-1α activation and significantly decreased glycolysis upregulation. Surprisingly, XBP1s appeared to be dispensable, as demonstrated by the lack of inhibiting effect of XBP1s knockdown on glycolytic genes expression, glucose uptake, lactate production and HIF-1α activation. These experiments demonstrate for the first time a key role of IRE1α in HIF-1α-mediated glycolysis upregulation in macrophages stimulated with pro-inflammatory triggers like LPS or SFAs through XBP1s-independent mechanism. IRE1 could mediate this novel function by targeting other transcripts (mRNA or pre-miRNA) through a mechanism called regulated IRE1-dependent decay or RIDD. Deciphering the underlying mechanisms of this novel IRE1 function might lead to novel therapeutic targets to curtail sterile obesity- or infection-linked inflammation.
- Published
- 2023
- Full Text
- View/download PDF
6. Paladin, overexpressed in colon cancer, is required for actin polymerisation and liver metastasis dissemination
- Author
-
Gilles Rademaker, Brunella Costanza, Sébastien Pyr dit Ruys, Raphaël Peiffer, Ferman Agirman, Naïma Maloujahmoum, Didier Vertommen, Andrei Turtoi, Akeila Bellahcène, Vincent Castronovo, and Olivier Peulen
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Introduction Colorectal cancer remains a public health issue and most colon cancer patients succumb to the development of metastases. Using a specific protocol of pressure-assisted interstitial fluid extrusion to recover soluble biomarkers, we identified paladin as a potential colon cancer liver metastases biomarker. Methods Using shRNA gene knockdown, we explored the biological function of paladin in colon cancer cells and investigated the phospho-proteome within colon cancer cells. We successively applied in vitro migration assays, in vivo metastasis models and co-immunoprecipitation experiments. Results We discovered that paladin is required for colon cancer cell migration and metastasis, and that paladin depletion altered the phospho-proteome within colon cancer cells. Data are available via ProteomeXchange with identifier PXD030803. Thanks to immunoprecipitation experiments, we demonstrated that paladin, was interacting with SSH1, a phosphatase involved in colon cancer metastasis. Finally, we showed that paladin depletion in cancer cells results in a less dynamic actin cytoskeleton. Conclusions Paladin is an undervalued protein in oncology. This study highlights for the first time that, paladin is participating in actin cytoskeleton remodelling and is required for efficient cancer cell migration.
- Published
- 2022
- Full Text
- View/download PDF
7. Myoferlin targeting triggers mitophagy and primes ferroptosis in pancreatic cancer cells
- Author
-
Gilles Rademaker, Yasmine Boumahd, Raphaël Peiffer, Sandy Anania, Tom Wissocq, Maude Liégeois, Géraldine Luis, Nor Eddine Sounni, Ferman Agirman, Naïma Maloujahmoum, Pascal De Tullio, Marc Thiry, Akeila Bellahcène, Vincent Castronovo, and Olivier Peulen
- Subjects
Myoferlin ,Ferroptosis ,Mitochondria ,Pancreas cancer ,Mitophagy ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Myoferlin, an emerging oncoprotein, has been associated with a low survival in several cancer types including pancreas ductal adenocarcinoma where it controls mitochondria structure and respiratory functions. Owing to the high susceptibility of KRAS-mutated cancer cells to iron-dependent cell death, ferroptosis, and to the high iron content in mitochondria, we investigated the relation existing between mitochondrial integrity and iron-dependent cell death. We discovered that myoferlin targeting with WJ460 pharmacological compound triggered mitophagy and ROS accumulation culminating with lipid peroxidation and apoptosis-independent cell death. WJ460 caused a reduction of the abundance of ferroptosis core regulators xc- cystine/glutamate transporter and GPX-4. Mitophagy inhibitor Mdivi1 and iron chelators inhibited the myoferlin-related ROS production and restored cell growth. Additionally, we reported a synergic effect between ferroptosis inducers, erastin and RSL3, and WJ460.
- Published
- 2022
- Full Text
- View/download PDF
8. Resistance to Gemcitabine in Pancreatic Cancer Is Connected to Methylglyoxal Stress and Heat Shock Response
- Author
-
Rebekah Crake, Imène Gasmi, Jordan Dehaye, Fanny Lardinois, Raphaël Peiffer, Naïma Maloujahmoum, Ferman Agirman, Benjamin Koopmansch, Nicky D’Haene, Oier Azurmendi Senar, Tatjana Arsenijevic, Frédéric Lambert, Olivier Peulen, Jean-Luc Van Laethem, and Akeila Bellahcène
- Subjects
oncometabolite ,methylglyoxal ,glycolysis ,therapy resistance ,gemcitabine ,metformin ,Cytology ,QH573-671 - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a fatal disease with poor prognosis. Gemcitabine is the first-line therapy for PDAC, but gemcitabine resistance is a major impediment to achieving satisfactory clinical outcomes. This study investigated whether methylglyoxal (MG), an oncometabolite spontaneously formed as a by-product of glycolysis, notably favors PDAC resistance to gemcitabine. We observed that human PDAC tumors expressing elevated levels of glycolytic enzymes together with high levels of glyoxalase 1 (GLO1), the major MG-detoxifying enzyme, present with a poor prognosis. Next, we showed that glycolysis and subsequent MG stress are triggered in PDAC cells rendered resistant to gemcitabine when compared with parental cells. In fact, acquired resistance, following short and long-term gemcitabine challenges, correlated with the upregulation of GLUT1, LDHA, GLO1, and the accumulation of MG protein adducts. We showed that MG-mediated activation of heat shock response is, at least in part, the molecular mechanism underlying survival in gemcitabine-treated PDAC cells. This novel adverse effect of gemcitabine, i.e., induction of MG stress and HSR activation, is efficiently reversed using potent MG scavengers such as metformin and aminoguanidine. We propose that the MG blockade could be exploited to resensitize resistant PDAC tumors and to improve patient outcomes using gemcitabine therapy.
- Published
- 2023
- Full Text
- View/download PDF
9. Extracellular HMGB1 blockade inhibits tumor growth through profoundly remodeling immune microenvironment and enhances checkpoint inhibitor-based immunotherapy
- Author
-
Coraline Radermecker, Pascale Hubert, Patrick Roncarati, Stephanie Demoulin, Charlotte Pilard, Marie Ancion, Celia Reynders, Thomas Lerho, Diane Bruyere, Alizee Lebeau, Margot Meunier, Marie-Julie Nokin, Elodie Hendrick, Olivier Peulen, Philippe Delvenne, and Michael Herfs
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background High-mobility group box 1 (HMGB1) is a multifunctional redox-sensitive protein involved in various intracellular (eg, chromatin remodeling, transcription, autophagy) and extracellular (inflammation, autoimmunity) processes. Regarding its role in cancer development/progression, paradoxical results exist in the literature and it is still unclear whether HMGB1 mainly acts as an oncogene or a tumor suppressor.Methods HMGB1 expression was first assessed in tissue specimens (n=359) of invasive breast, lung and cervical cancer and the two distinct staining patterns detected (nuclear vs cytoplasmic) were correlated to the secretion profile of malignant cells, patient outcomes and the presence of infiltrating immune cells within tumor microenvironment. Using several orthotopic, syngeneic mouse models of basal-like breast (4T1, 67NR and EpRas) or non-small cell lung (TC-1) cancer, the efficacy of several HMGB1 inhibitors alone and in combination with immune checkpoint blockade antibodies (anti-PD-1/PD-L1) was then investigated. Isolated from retrieved tumors, 14 immune cell (sub)populations as well as the activation status of antigen-presenting cells were extensively analyzed in each condition. Finally, the redox state of HMGB1 in tumor-extruded fluids and the influence of different forms (oxidized, reduced or disulfide) on both dendritic cell (DC) and plasmacytoid DC (pDC) activation were determined.Results Associated with an unfavorable prognosis in human patients, we clearly demonstrated that targeting extracellular HMGB1 elicits a profound remodeling of tumor immune microenvironment for efficient cancer therapy. Indeed, without affecting the global number of (CD45+) immune cells, drastic reductions of monocytic/granulocytic myeloid-derived suppressor cells (MDSC) and regulatory T lymphocytes, a higher M1/M2 ratio of macrophages as well as an increased activation of both DC and pDC were continually observed following HMGB1 inhibition. Moreover, blocking HMGB1 improved the efficacy of anti-PD-1 cancer monoimmunotherapy. We also reported that a significant fraction of HMGB1 encountered within cancer microenvironment (interstitial fluids) is oxidized and, in opposite to its reduced isoform, oxidized HMGB1 acts as a tolerogenic signal in a receptor for advanced glycation endproducts-dependent manner.Conclusion Collectively, we present evidence that extracellular HMGB1 blockade may complement first-generation cancer immunotherapies by remobilizing antitumor immune response.
- Published
- 2021
- Full Text
- View/download PDF
10. Methylglyoxal, a glycolysis metabolite, triggers metastasis through MEK/ERK/SMAD1 pathway activation in breast cancer
- Author
-
Marie-Julie Nokin, Justine Bellier, Florence Durieux, Olivier Peulen, Gilles Rademaker, Maude Gabriel, Christine Monseur, Benoit Charloteaux, Lieven Verbeke, Steven van Laere, Patrick Roncarati, Michael Herfs, Charles Lambert, Jean Scheijen, Casper Schalkwijk, Alain Colige, Jo Caers, Philippe Delvenne, Andrei Turtoi, Vincent Castronovo, and Akeila Bellahcène
- Subjects
Breast cancer ,Methylglyoxal ,SMAD1 ,Metastasis ,Carnosine ,MAPK ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Elevated aerobic glycolysis rate is a biochemical alteration associated with malignant transformation and cancer progression. This metabolic shift unavoidably generates methylglyoxal (MG), a potent inducer of dicarbonyl stress through the formation of advanced glycation end products (AGEs). We have previously shown that the silencing of glyoxalase 1 (GLO1), the main MG detoxifying enzyme, generates endogenous dicarbonyl stress resulting in enhanced growth and metastasis in vivo. However, the molecular mechanisms through which MG stress promotes metastasis development remain to be unveiled. Methods In this study, we used RNA sequencing analysis to investigate gene-expression profiling of GLO1-depleted breast cancer cells and we validated the regulated expression of selected genes of interest by RT-qPCR. Using in vitro and in vivo assays, we demonstrated the acquisition of a pro-metastatic phenotype related to dicarbonyl stress in MDA-MB-231, MDA-MB-468 and MCF7 breast cancer cellular models. Hyperactivation of MEK/ERK/SMAD1 pathway was evidenced using western blotting upon endogenous MG stress and exogenous MG treatment conditions. MEK and SMAD1 regulation of MG pro-metastatic signature genes in breast cancer cells was demonstrated by RT-qPCR. Results High-throughput transcriptome profiling of GLO1-depleted breast cancer cells highlighted a pro-metastatic signature that establishes novel connections between MG dicarbonyl stress, extracellular matrix (ECM) remodeling by neoplastic cells and enhanced cell migration. Mechanistically, we showed that these metastasis-related processes are functionally linked to MEK/ERK/SMAD1 cascade activation in breast cancer cells. We showed that sustained MEK/ERK activation in GLO1-depleted cells notably occurred through the down-regulation of the expression of dual specificity phosphatases in MG-stressed breast cancer cells. The use of carnosine and aminoguanidine, two potent MG scavengers, reversed MG stress effects in in vitro and in vivo experimental settings. Conclusions These results uncover for the first time the key role of MG dicarbonyl stress in the induction of ECM remodeling and the activation of migratory signaling pathways, both in favor of enhanced metastatic dissemination of breast cancer cells. Importantly, the efficient inhibition of mitogen-activated protein kinase (MAPK) signaling using MG scavengers further emphasizes the need to investigate their therapeutic potential across different malignancies.
- Published
- 2019
- Full Text
- View/download PDF
11. Propranolol sensitizes prostate cancer cells to glucose metabolism inhibition and prevents cancer progression
- Author
-
Laura Brohée, Olivier Peulen, Betty Nusgens, Vincent Castronovo, Marc Thiry, Alain C. Colige, and Christophe F. Deroanne
- Subjects
Medicine ,Science - Abstract
Abstract Propranolol, a widely used non-selective beta-adrenergic receptor blocker, was recently shown to display anticancer properties. Its potential to synergize with certain drugs has been also outlined. However, it is necessary to take into account all the properties of propranolol to select a drug that could be efficiently combined with. Propranolol was reported to block the late phase of autophagy. Hence, we hypothesized that in condition enhancing autophagy flux, cancer cells should be especially sensitive to propranolol. 2DG, a glycolysis inhibitor, is an anti-tumor agent having limited effect in monotherapy notably due to induction of pro-survival autophagy. Here, we report that treatment of cancer cells with propranolol in combination with the glycolysis inhibitor 2DG induced a massive accumulation of autophagosome due to autophagy blockade. The propranolol +2DG treatment efficiently prevents prostate cancer cell proliferation, induces cell apoptosis, alters mitochondrial morphology, inhibits mitochondrial bioenergetics and aggravates ER stress in vitro and also suppresses tumor growth in vivo. Our study underlines for the first time the interest to take advantage of the ability of propranolol to inhibit autophagy to design new anti-cancer therapies.
- Published
- 2018
- Full Text
- View/download PDF
12. Human papillomavirus E6/E7 oncoproteins promote radiotherapy-mediated tumor suppression by globally hijacking host DNA damage repair
- Author
-
Diane Bruyere, Patrick Roncarati, Alizee Lebeau, Thomas Lerho, Florian Poulain, Elodie Hendrick, Charlotte Pilard, Celia Reynders, Marie Ancion, Margaux Luyckx, Michael Renard, Yves Jacob, Jean-Claude Twizere, Raphael Peiffer, Olivier Peulen, Philippe Delvenne, Pascale Hubert, Alison McBride, Nicolas Gillet, Murielle Masson, and Michael Herfs
- Subjects
protein-protein interactome ,DNA damage and repair ,Medicine (miscellaneous) ,human papillomavirus ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,radiotherapy - Abstract
Rationale: Whatever the mucosa primary infected, HPV-positive cancers are traditionally associated with a favorable outcome, attributable to a high sensitivity to radiation therapy. However, the direct impact of viral E6/E7 oncoproteins on the intrinsic cellular radiosensitivity (and, globally, on host DNA repair) remains mostly speculative.Methods: Using several isogenic cell models expressing HPV16 E6 and/or E7, the effect of viral oncoproteins on global DNA damage response was first investigated by in vitro/in vivo approaches. The binary interactome of each individual HPV oncoprotein with factors involved in the various host DNA damage/repair mechanisms was then precisely mapped by Gaussia princeps luciferase complementation assay (and validated by co-immunoprecipitation). The stability/half-life of protein targets for HPV E6 and/or E7 as well as their subcellular localizations were determined. At last, the host genome integrity following E6/E7 expression and the synergy between radiotherapy and compounds targeting DNA repair were analyzed.Results: We first showed that the sole expression of one viral oncoprotein from HPV16 was able to significantly increase the sensitivity to irradiation of cells without affecting their basal viability parameters. In total, 10 novel targets (CHEK2, CLK2, CLK2/3, ERCC3, MNAT1, PER1, RMI1, RPA1, UVSSA and XRCC6) for E6 and 11 (ALKBH2, CHEK2, DNA2, DUT, ENDOV, ERCC3, PARP3, PMS1, PNKP, POLDIP2 and RBBP8) for E7 were identified. Importantly, not degraded following their interaction with E6 or E7, these proteins have been shown to be less linked to host DNA and to colocalize with HPV replication foci, denoting their crucial implication in viral life cycle. Finally, we found that E6/E7 oncoproteins globally jeopardize host genome integrity, increase the cellular sensitivity to DNA repair inhibitors and enhance their synergy with radiotherapy.Conclusion: Taken together, our findings provide a molecular insight into the direct hijacking of host DNA damage/repair responses by HPV oncoproteins, demonstrate the significant impact of this phenomenon on both intrinsic cellular radiosensitivity and host DNA integrity and suggest novel connected therapeutic vulnerabilities.
- Published
- 2023
13. The transcription factor c-Jun inhibits RBM39 to reprogram pre-mRNA splicing during genotoxic stress
- Author
-
Florence Lemaitre, Fatima Chakrama, Tina O’Grady, Olivier Peulen, Gilles Rademaker, Adeline Deward, Benoit Chabot, Jacques Piette, Alain Colige, Charles Lambert, Franck Dequiedt, and Yvette Habraken
- Subjects
Genetics - Abstract
Genotoxic agents, that are used in cancer therapy, elicit the reprogramming of the transcriptome of cancer cells. These changes reflect the cellular response to stress and underlie some of the mechanisms leading to drug resistance. Here, we profiled genome-wide changes in pre-mRNA splicing induced by cisplatin in breast cancer cells. Among the set of cisplatin-induced alternative splicing events we focused on COASY, a gene encoding a mitochondrial enzyme involved in coenzyme A biosynthesis. Treatment with cisplatin induces the production of a short isoform of COASY lacking exons 4 and 5, whose depletion impedes mitochondrial function and decreases sensitivity to cisplatin. We identified RBM39 as a major effector of the cisplatin-induced effect on COASY splicing. RBM39 also controls a genome-wide set of alternative splicing events partially overlapping with the cisplatin-mediated ones. Unexpectedly, inactivation of RBM39 in response to cisplatin involves its interaction with the AP-1 family transcription factor c-Jun that prevents RBM39 binding to pre-mRNA. Our findings therefore uncover a novel cisplatin-induced interaction between a splicing regulator and a transcription factor that has a global impact on alternative splicing and contributes to drug resistance.
- Published
- 2022
- Full Text
- View/download PDF
14. Methylglyoxal Scavengers Resensitize KRAS-Mutated Colorectal Tumors to Cetuximab
- Author
-
Justine Bellier, Marie-Julie Nokin, Maurine Caprasse, Assia Tiamiou, Arnaud Blomme, Jean L. Scheijen, Benjamin Koopmansch, Gillian M. MacKay, Barbara Chiavarina, Brunella Costanza, Gilles Rademaker, Florence Durieux, Ferman Agirman, Naïma Maloujahmoum, Pino G. Cusumano, Pierre Lovinfosse, Hing Y. Leung, Frédéric Lambert, Vincent Bours, Casper G. Schalkwijk, Roland Hustinx, Olivier Peulen, Vincent Castronovo, and Akeila Bellahcène
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: The use of cetuximab anti-epidermal growth factor receptor (anti-EGFR) antibodies has opened the era of targeted and personalized therapy in colorectal cancer (CRC). Poor response rates have been unequivocally shown in mutant KRAS and are even observed in a majority of wild-type KRAS tumors. Therefore, patient selection based on mutational profiling remains problematic. We previously identified methylglyoxal (MGO), a by-product of glycolysis, as a metabolite promoting tumor growth and metastasis. Mutant KRAS cells under MGO stress show AKT-dependent survival when compared with wild-type KRAS isogenic CRC cells. MGO induces AKT activation through phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin 2 (mTORC2) and Hsp27 regulation. Importantly, the sole induction of MGO stress in sensitive wild-type KRAS cells renders them resistant to cetuximab. MGO scavengers inhibit AKT and resensitize KRAS-mutated CRC cells to cetuximab in vivo. This study establishes a link between MGO and AKT activation and pinpoints this oncometabolite as a potential target to tackle EGFR-targeted therapy resistance in CRC. : Bellier et al. demonstrate that MGO stress is a constant feature of KRAS-mutated CRC tumors. MGO induces a key survival pathway implicated in resistance to EGFR-targeted therapy in CRC. The scavenging of this oncometabolite could be beneficial in the treatment of both wild-type and mutant KRAS CRC tumors. Keywords: methylglyoxal, colorectal cancer, KRAS mutation, EGFR-targeted therapy, Hsp27, carnosine, aminoguanidine, cetuximab, AKT signaling
- Published
- 2020
- Full Text
- View/download PDF
15. Ferlin Overview: From Membrane to Cancer Biology
- Author
-
Olivier Peulen, Gilles Rademaker, Sandy Anania, Andrei Turtoi, Akeila Bellahcène, and Vincent Castronovo
- Subjects
ferlin ,myoferlin ,dysferlin ,otoferlin ,C2 domain ,plasma membrane ,Cytology ,QH573-671 - Abstract
In mammal myocytes, endothelial cells and inner ear cells, ferlins are proteins involved in membrane processes such as fusion, recycling, endo- and exocytosis. They harbour several C2 domains allowing their interaction with phospholipids. The expression of several Ferlin genes was described as altered in several tumoural tissues. Intriguingly, beyond a simple alteration, myoferlin, otoferlin and Fer1L4 expressions were negatively correlated with patient survival in some cancer types. Therefore, it can be assumed that membrane biology is of extreme importance for cell survival and signalling, making Ferlin proteins core machinery indispensable for cancer cell adaptation to hostile environments. The evidences suggest that myoferlin, when overexpressed, enhances cancer cell proliferation, migration and metabolism by affecting various aspects of membrane biology. Targeting myoferlin using pharmacological compounds, gene transfer technology, or interfering RNA is now considered as an emerging therapeutic strategy.
- Published
- 2019
- Full Text
- View/download PDF
16. Elp3‐mediated codon‐dependent translation promotes <scp>mTORC2</scp> activation and regulates macrophage polarization
- Author
-
Dawei Chen, Ivan Nemazanyy, Olivier Peulen, Kateryna Shostak, Xinyi Xu, Seng Chuan Tang, Caroline Wathieu, Silvia Turchetto, Sylvia Tielens, Laurent Nguyen, Pierre Close, Christophe Desmet, Sebastian Klein, Alexandra Florin, Reinhard Büttner, Georgios Petrellis, Benjamin Dewals, and Alain Chariot
- Subjects
Mice ,General Immunology and Microbiology ,Macrophages ,General Neuroscience ,Animals ,Mechanistic Target of Rapamycin Complex 2 ,Macrophage Activation ,Codon ,Molecular Biology ,General Biochemistry, Genetics and Molecular Biology ,Histone Acetyltransferases ,Signal Transduction - Abstract
Macrophage polarization is a process whereby macrophages acquire distinct effector states (M1 or M2) to carry out multiple and sometimes opposite functions. We show here that translational reprogramming occurs during macrophage polarization and that this relies on the Elongator complex subunit Elp3, an enzyme that modifies the wobble uridine base U34 in cytosolic tRNAs. Elp3 expression is downregulated by classical M1-activating signals in myeloid cells, where it limits the production of pro-inflammatory cytokines via FoxO1 phosphorylation, and attenuates experimental colitis in mice. In contrast, alternative M2-activating signals upregulate Elp3 expression through a PI3K- and STAT6-dependent signaling pathway. The metabolic reprogramming linked to M2 macrophage polarization relies on Elp3 and the translation of multiple candidates, including the mitochondrial ribosome large subunit proteins Mrpl3, Mrpl13, and Mrpl47. By promoting translation of its activator Ric8b in a codon-dependent manner, Elp3 also regulates mTORC2 activation. Elp3 expression in myeloid cells further promotes Wnt-driven tumor initiation in the intestine by maintaining a pool of tumor-associated macrophages exhibiting M2 features. Collectively, our data establish a functional link between tRNA modifications, mTORC2 activation, and macrophage polarization.
- Published
- 2022
- Full Text
- View/download PDF
17. Methylglyoxal, a glycolysis side-product, induces Hsp90 glycation and YAP-mediated tumor growth and metastasis
- Author
-
Marie-Julie Nokin, Florence Durieux, Paul Peixoto, Barbara Chiavarina, Olivier Peulen, Arnaud Blomme, Andrei Turtoi, Brunella Costanza, Nicolas Smargiasso, Dominique Baiwir, Jean L Scheijen, Casper G Schalkwijk, Justine Leenders, Pascal De Tullio, Elettra Bianchi, Marc Thiry, Koji Uchida, David A Spiegel, James R Cochrane, Craig A Hutton, Edwin De Pauw, Philippe Delvenne, Dominique Belpomme, Vincent Castronovo, and Akeila Bellahcène
- Subjects
carbonyl stress ,glyoxalase 1 ,LATS1 ,breast cancer ,methylglyoxal ,YAP ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Metabolic reprogramming toward aerobic glycolysis unavoidably induces methylglyoxal (MG) formation in cancer cells. MG mediates the glycation of proteins to form advanced glycation end products (AGEs). We have recently demonstrated that MG-induced AGEs are a common feature of breast cancer. Little is known regarding the impact of MG-mediated carbonyl stress on tumor progression. Breast tumors with MG stress presented with high nuclear YAP, a key transcriptional co-activator regulating tumor growth and invasion. Elevated MG levels resulted in sustained YAP nuclear localization/activity that could be reverted using Carnosine, a scavenger for MG. MG treatment affected Hsp90 chaperone activity and decreased its binding to LATS1, a key kinase of the Hippo pathway. Cancer cells with high MG stress showed enhanced growth and metastatic potential in vivo. These findings reinforce the cumulative evidence pointing to hyperglycemia as a risk factor for cancer incidence and bring renewed interest in MG scavengers for cancer treatment.
- Published
- 2016
- Full Text
- View/download PDF
18. The E3 ligase COP1 promotes ERα signaling and suppresses EMT in breast cancer
- Author
-
Seng Chuan, Tang, Quentin, Lion, Olivier, Peulen, Philippe, Chariot, Arnaud, Lavergne, Alice, Mayer, Paula Allepuz, Fuster, Pierre, Close, Sebastian, Klein, Alexandra, Florin, Reinhard, Büttner, Ivan, Nemazanyy, Kateryna, Shostak, and Alain, Chariot
- Subjects
Cell Line, Tumor ,Ubiquitin-Protein Ligases ,Estrogen Receptor alpha ,Humans ,Breast Neoplasms ,Female ,Transfection ,Cell Proliferation ,Signal Transduction - Abstract
ERα signaling drives proliferation, survival and cancer initiation in the mammary gland. Therefore, it is critical to elucidate mechanisms by which ERα expression is regulated. We show that the tumor suppressor E3 ligase COP1 promotes the degradative polyubiquitination of the microtubule-associated protein HPIP. As such, COP1 negatively regulates estrogen-dependent AKT activation in breast cancer cells. However, COP1 also induces ERα expression and ERα-dependent gene transcription, at least through c-Jun degradation. COP1 and ERα levels are positively correlated in clinical cases of breast cancer. COP1 also supports the metabolic reprogramming by estrogens, including glycolysis. On the other hand, COP1 suppresses EMT in breast cancer cells. COP1 deficiency also contributes to Tamoxifen resistance, at least through protective autophagy. Therefore, COP1 acts as an oncogenic E3 ligase by promoting ERα signaling but also acts as a tumor suppressor candidate by preventing EMT, which reflects a dual role of COP1 in breast cancer.
- Published
- 2021
19. Methylglyoxal stress induces a major epigenetic deregulation leading to a pro-migratory phenotype in breast cancer and significant clinical relevance
- Author
-
Yasmine Boumahd, Akeila Bellahcene, Martin Bizet, Rachel Deplus, Vincent Castronovo, Marie-Julie Nokin, Gaurav Dube, Olivier Peulen, Emilie Calonne, Justine Bellier, and François Fuks
- Subjects
chemistry.chemical_compound ,Breast cancer ,chemistry ,business.industry ,Methylglyoxal ,medicine ,Cancer research ,Clinical significance ,Epigenetics ,medicine.disease ,business ,Phenotype - Published
- 2021
- Full Text
- View/download PDF
20. Treatment algorithm and prognostic factors for patients with stage I–III carcinoma of the anal canal: a 20-year multicenter study
- Author
-
Jean-Baptiste Delhorme, Franck Monnien, Michael Herfs, Laurent Martin, Lucine Vuitton, Laurence Dusserre, Celia Reynders, Jean-François Bosset, Pascale Hubert, Prudence Colpart, Anne-Sophie Woronoff, Agnès Leroux, Patrick Roncarati, Philippe Delvenne, Laurent Arnould, Elodie Hendrick, Christiane Mougin, Marie Ancion, Didier Peiffert, Alexis Lepinoy, Jean-Luc Prétet, Olivier Peulen, Alexandra Luquain, Thomas Lerho, Diane Bruyère, Séverine Valmary-Degano, Charlotte Pilard, Chloé Molimard, Marie-Christine Bone-Lepinoy, Philippe Maingon, Jean-Pierre Ghnassia, GIGA [Université Liège], Université de Liège, Service d'Oncologie Médicale [CHRU Besançon], Centre Hospitalier Régional Universitaire de Besançon (CHRU Besançon)-Université de Franche-Comté (UFC), Université Bourgogne Franche-Comté [COMUE] (UBFC)-Université Bourgogne Franche-Comté [COMUE] (UBFC), Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-Research), Centre Hospitalier Régional Universitaire de Besançon (CHRU Besançon), Carcinogénèse épithéliale : facteurs prédictifs et pronostiques - UFC (EA 3181) (CEF2P / CARCINO), Université de Franche-Comté (UFC), and Université Bourgogne Franche-Comté [COMUE] (UBFC)-Université Bourgogne Franche-Comté [COMUE] (UBFC)-Centre Hospitalier Régional Universitaire de Besançon (CHRU Besançon)
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,Pathology ,medicine.medical_specialty ,Anal Carcinoma ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Carcinoma ,Humans ,Anal cancer ,ComputingMilieux_MISCELLANEOUS ,Aged ,business.industry ,Incidence ,Incidence (epidemiology) ,Middle Aged ,Anal canal ,Anus Neoplasms ,Prognosis ,medicine.disease ,Precision medicine ,Progression-Free Survival ,3. Good health ,Regimen ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Carcinoma, Squamous Cell ,Female ,business ,Algorithms ,Chemoradiotherapy - Abstract
Despite a growing incidence in developed countries and a recent improved understanding of its pathogenesis, anal cancer management has not evolved over the past decades and drug combination used as first-line regimen still largely depends on clinician preferences. Aiming at paving the way for precision medicine, a large cohort of 372 HIV-negative patients diagnosed over a 20-year time period with locally advanced anal carcinoma was collected and carefully characterized at the clinical, demographic, histopathologic, immunologic, and virologic levels. Both the prognostic relevance of each clinicopathological parameter and the efficacy of different concurrent chemoradiation strategies were determined. Overall, the incidence of anal cancer peaked during the sixth decade (mean: 63.4) and females outnumbered males (ratio: 2.51). After completion of treatment, 95 (25.5%) patients experienced progression of persistent disease or local/distant recurrence and 102 (27.4%) died during the follow-up period (median: 53.8 months). Importantly, uni-multivariate analyses indicated that both negative HPV/p16ink4a status and aberrant p53 expression were far better predictors for reduced progression-free survival than traditional risk factors such as tumor size and nodal status. As for overall survival, the significant influences of age at diagnosis, p16ink4a status, cTNM classification as well as both CD3+ and CD4+ T-cell infiltrations within tumor microenvironment were highlighted. Cisplatin-based chemoradiotherapy was superior to both radiotherapy alone and other concurrent chemoradiation therapies in the treatment of HPV-positive tumors. Regarding their HPV-uninfected counterparts, frequent relapses were observed, whatever the treatment regimen administered. Taken together, our findings reveal that current anal cancer management and treatment have reached their limits. A dualistic classification according to HPV/p53 status should be considered with implications for therapy personalization and optimization.
- Published
- 2021
- Full Text
- View/download PDF
21. The anti-tumor effect of HDAC inhibition in a human pancreas cancer model is significantly improved by the simultaneous inhibition of cyclooxygenase 2.
- Author
-
Olivier Peulen, Arnaud Gonzalez, Paul Peixoto, Andrei Turtoi, Denis Mottet, Philippe Delvenne, and Vincent Castronovo
- Subjects
Medicine ,Science - Abstract
Pancreatic ductal adenocarcinoma is the fourth leading cause of cancer death worldwide, with no satisfactory treatment to date. In this study, we tested whether the combined inhibition of cyclooxygenase-2 (COX-2) and class I histone deacetylase (HDAC) may results in a better control of pancreatic ductal adenocarcinoma. The impact of the concomitant HDAC and COX-2 inhibition on cell growth, apoptosis and cell cycle was assessed first in vitro on human pancreas BxPC-3, PANC-1 or CFPAC-1 cells treated with chemical inhibitors (SAHA, MS-275 and celecoxib) or HDAC1/2/3/7 siRNA. To test the potential antitumoral activity of this combination in vivo, we have developed and characterized, a refined chick chorioallantoic membrane tumor model that histologically and proteomically mimics human pancreatic ductal adenocarcinoma. The combination of HDAC1/3 and COX-2 inhibition significantly impaired proliferation of BxPC-3 cells in vitro and stalled entirely the BxPC-3 cells tumor growth onto the chorioallantoic membrane in vivo. The combination was more effective than either drug used alone. Consistently, we showed that both HDAC1 and HDAC3 inhibition induced the expression of COX-2 via the NF-kB pathway. Our data demonstrate, for the first time in a Pancreatic Ductal Adenocarcinoma (PDAC) model, a significant action of HDAC and COX-2 inhibitors on cancer cell growth, which sets the basis for the development of potentially effective new combinatory therapies for pancreatic ductal adenocarcinoma patients.
- Published
- 2013
- Full Text
- View/download PDF
22. Methylglyoxal, a potent inducer of AGEs, connects between diabetes and cancer
- Author
-
Eva Lardé, Philippe Karoyan, Olivier Peulen, Justine Bellier, Marie-Julie Nokin, Vincenzo Castronovo, and Akeila Bellahcene
- Subjects
Glycation End Products, Advanced ,Endocrinology, Diabetes and Metabolism ,Bioinformatics ,Diabetes Complications ,Diabetic nephropathy ,Pathogenesis ,chemistry.chemical_compound ,Endocrinology ,Meta-Analysis as Topic ,Glycation ,Neoplasms ,Diabetes mellitus ,Internal Medicine ,medicine ,Animals ,Humans ,business.industry ,Metabolic disorder ,Methylglyoxal ,Cancer ,General Medicine ,Pyruvaldehyde ,medicine.disease ,Up-Regulation ,Oxidative Stress ,chemistry ,Hyperglycemia ,business ,Retinopathy - Abstract
Diabetes is one of the most frequent diseases throughout the world and its incidence is predicted to exponentially progress in the future. This metabolic disorder is associated with major complications such as neuropathy, retinopathy, atherosclerosis, and diabetic nephropathy, the severity of which correlates with hyperglycemia, suggesting that they are triggered by high glucose condition. Reducing sugars and reactive carbonyl species such as methylglyoxal (MGO) lead to glycation of proteins, lipids and DNA and the gradual accumulation of advanced glycation end products (AGEs) in cells and tissues. While AGEs are clearly implicated in the pathogenesis of diabetes complications, their potential involvement during malignant tumor development, progression and resistance to therapy is an emerging concept. Meta-analysis studies established that patients with diabetes are at higher risk of developing cancer and show a higher mortality rate than cancer patients free of diabetes. In this review, we highlight the potential connection between hyperglycemia-associated AGEs formation on the one hand and the recent evidence of pro-tumoral effects of MGO stress on the other hand. We also discuss the marked interest in anti-glycation compounds in view of their strategic use to treat diabetic complications but also to protect against augmented cancer risk in patients with diabetes.
- Published
- 2019
- Full Text
- View/download PDF
23. Human peroxidasin 1 promotes angiogenesis through ERK1/2, Akt, and FAK pathways
- Author
-
Michael Herfs, Mohamed Amri, Pierre Van Antwerpen, Hayfa Medfai, Paul G. Furtmüller, Christian Obinger, Benjamin Sevcnikar, Vincent Castronovo, Olivier Peulen, Vincent Nuyens, Luc Vanhamme, Alia Khalil, Karim Zouaoui Boudjeltia, Nicole Moguilevsky, Alexandre Rousseau, and Martina Paumann-Page
- Subjects
0301 basic medicine ,Physiology ,Angiogenesis ,Neovascularization, Physiologic ,Chick Embryo ,030204 cardiovascular system & hematology ,Cell Line ,Focal adhesion ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Physiology (medical) ,Animals ,Humans ,Phosphorylation ,Protein kinase B ,Cell Proliferation ,Mitogen-Activated Protein Kinase 1 ,Tube formation ,Mitogen-Activated Protein Kinase 3 ,PDGFB ,Chemistry ,Endothelial Cells ,Sciences bio-médicales et agricoles ,Cell biology ,Enzyme Activation ,030104 developmental biology ,Gene Expression Regulation ,Peroxidases ,Focal Adhesion Kinase 1 ,Signal transduction ,Cardiology and Cardiovascular Medicine ,Proto-Oncogene Proteins c-akt ,Signal Transduction ,Platelet-Derived Growth Factor Subunit B - Abstract
The term angiogenesis refers to sprouting of new blood vessels from pre-existing ones. The angiogenic process involves cell migration and tubulogenesis requiring interaction between endothelial cells and the extracellular matrix. Human peroxidasin 1 (hsPxd01) is a multidomain heme peroxidase found embedded in the basement membranes. As it promotes the stabilization of extracellular matrix, we investigated its possible role in angiogenesis both in vitro and in vivo., SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2018
- Full Text
- View/download PDF
24. Propranolol sensitizes prostate cancer cells to glucose metabolism inhibition and prevents cancer progression
- Author
-
Christophe Deroanne, Marc Thiry, Betty Nusgens, Laura Brohée, Alain Colige, Vincent Castronovo, and Olivier Peulen
- Subjects
Male ,0301 basic medicine ,Autophagosome ,Science ,Propranolol ,Article ,Mice ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Cell Line, Tumor ,Autophagy ,medicine ,Animals ,Humans ,Multidisciplinary ,Chemistry ,Prostatic Neoplasms ,Cancer ,Endoplasmic Reticulum Stress ,medicine.disease ,Xenograft Model Antitumor Assays ,Mitochondria ,Disease Models, Animal ,Glucose ,030104 developmental biology ,Apoptosis ,030220 oncology & carcinogenesis ,Cancer cell ,Disease Progression ,Unfolded protein response ,Cancer research ,Carbohydrate Metabolism ,Medicine ,Glycolysis ,medicine.drug - Abstract
Propranolol, a widely used non-selective beta-adrenergic receptor blocker, was recently shown to display anticancer properties. Its potential to synergize with certain drugs has been also outlined. However, it is necessary to take into account all the properties of propranolol to select a drug that could be efficiently combined with. Propranolol was reported to block the late phase of autophagy. Hence, we hypothesized that in condition enhancing autophagy flux, cancer cells should be especially sensitive to propranolol. 2DG, a glycolysis inhibitor, is an anti-tumor agent having limited effect in monotherapy notably due to induction of pro-survival autophagy. Here, we report that treatment of cancer cells with propranolol in combination with the glycolysis inhibitor 2DG induced a massive accumulation of autophagosome due to autophagy blockade. The propranolol +2DG treatment efficiently prevents prostate cancer cell proliferation, induces cell apoptosis, alters mitochondrial morphology, inhibits mitochondrial bioenergetics and aggravates ER stress in vitro and also suppresses tumor growth in vivo. Our study underlines for the first time the interest to take advantage of the ability of propranolol to inhibit autophagy to design new anti-cancer therapies.
- Published
- 2018
25. A dualistic model of primary anal canal adenocarcinoma with distinct cellular origins, etiologies, inflammatory microenvironments and mutational signatures: implications for personalised medicine
- Author
-
Philippe Delvenne, Frédéric Lambert, Nathalie Piazzon, Séverine Valmary-Degano, Elodie Hendrick, Vincent Bours, Aurélie Poncin, Jean-Luc Prétet, Christiane Mougin, Lucine Vuitton, Karin Segers, Olivier Peulen, David Guenat, Patrick Roncarati, Franck Monnien, Benjamin Koopmansch, Diane Bruyère, Pascale Hubert, Laurence de Leval, William Penny, Michael Herfs, Alizée Lebeau, Christopher P. Crum, and Charles M. Quick
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,Adult ,Male ,Cancer Research ,Programmed Cell Death 1 Receptor ,Adenocarcinoma/genetics ,Adenocarcinoma/pathology ,Aged ,Aged, 80 and over ,Anus Neoplasms/genetics ,Anus Neoplasms/pathology ,B7-H1 Antigen/genetics ,ErbB Receptors/genetics ,Female ,Gene Expression Regulation, Neoplastic/drug effects ,Humans ,Inflammation/genetics ,Inflammation/pathology ,Kaplan-Meier Estimate ,Lymphocytes, Tumor-Infiltrating/pathology ,Middle Aged ,Mutation ,Precision Medicine ,Prognosis ,Programmed Cell Death 1 Receptor/genetics ,Tumor Microenvironment/genetics ,Adenocarcinoma ,medicine.disease_cause ,Article ,B7-H1 Antigen ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Lymphocytes, Tumor-Infiltrating ,Tumor Microenvironment ,Medicine ,Gastrointestinal cancer ,Inflammation ,business.industry ,FOXP3 ,Anal canal ,medicine.disease ,Anus Neoplasms ,Anal canal adenocarcinoma ,3. Good health ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Anal gland ,Cancer research ,KRAS ,business - Abstract
Background Primary adenocarcinoma of the anal canal is a rare and aggressive gastrointestinal disease with unclear pathogenesis. Because of its rarity, no clear clinical practice guideline has been defined and a targeted therapeutic armamentarium has yet to be developed. The present article aimed at addressing this information gap by in-depth characterising the anal glandular neoplasms at the histologic, immunologic, genomic and epidemiologic levels. Methods In this multi-institutional study, we first examined the histological features displayed by each collected tumour (n = 74) and analysed their etiological relationship with human papillomavirus (HPV) infection. The intratumoural immune cell subsets (CD4, CD8, Foxp3), the expression of immune checkpoints (PD-1, PD-L1), the defect in mismatch repair proteins and the mutation analysis of multiple clinically relevant genes in the gastrointestinal cancer setting were also determined. Finally, the prognostic significance of each clinicopathological variable was assessed. Results Phenotypic analysis revealed two region-specific subtypes of anal canal adenocarcinoma. The significant differences in the HPV status, density of tumour-infiltrating lymphocytes, expression of immune checkpoints and mutational profile of several targetable genes further supported the separation of these latter neoplasms into two distinct entities. Importantly, anal gland/transitional-type cancers, which poorly respond to standard treatments, displayed less mutations in downstream effectors of the EGFR signalling pathway (i.e., KRAS and NRAS) and demonstrated a significantly higher expression of the immune inhibitory ligand-receptor pair PD-1/PD-L1 compared to their counterparts arising from the colorectal mucosa. Conclusions Taken together, the findings reported in the present article reveal, for the first time, that glandular neoplasms of the anal canal arise by HPV-dependent or independent pathways. These etiological differences leads to both individual immune profiles and mutational landscapes that can be targeted for therapeutic benefits.
- Published
- 2018
26. Hormetic potential of methylglyoxal, a side-product of glycolysis, in switching tumours from growth to death
- Author
-
David Spiegel, Koji Uchida, Craig A. Hutton, Justine Bellier, Akeila Bellahcene, Florence Durieux, Olivier Peulen, Marie-Julie Nokin, Vincenzo Castronovo, and James R. Cochrane
- Subjects
Glycation End Products, Advanced ,0301 basic medicine ,NF-E2-Related Factor 2 ,lcsh:Medicine ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Lactoylglutathione lyase ,Hormesis ,0302 clinical medicine ,Glycation ,Cell Line, Tumor ,Neoplasms ,Humans ,Glycolysis ,lcsh:Science ,Cell Proliferation ,Multidisciplinary ,Cell Death ,biology ,Cell growth ,Methylglyoxal ,lcsh:R ,Lactoylglutathione Lyase ,Pyruvaldehyde ,030104 developmental biology ,chemistry ,Biochemistry ,Anaerobic glycolysis ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Cancer research ,lcsh:Q ,Glyoxalase system - Abstract
Metabolic reprogramming toward aerobic glycolysis unavoidably favours methylglyoxal (MG) and advanced glycation end products (AGEs) formation in cancer cells. MG was initially considered a highly cytotoxic molecule with potential anti-cancer value. However, we have recently demonstrated that MG enhanced tumour growth and metastasis. In an attempt to understand this dual role, we explored MG-mediated dicarbonyl stress status in four breast and glioblastoma cancer cell lines in relation with their glycolytic phenotype and MG detoxifying capacity. In glycolytic cancer cells cultured in high glucose, we observed a significant increase of the conversion of MG to D-lactate through the glyoxalase system. Moreover, upon exogenous MG challenge, glycolytic cells showed elevated amounts of intracellular MG and induced de novo GLO1 detoxifying enzyme and Nrf2 expression. Thus, supporting the adaptive nature of glycolytic cancer cells to MG dicarbonyl stress when compared to non-glycolytic ones. Finally and consistent with the pro-tumoural role of MG, we showed that low doses of MG induced AGEs formation and tumour growth in vivo, both of which can be reversed using a MG scavenger. Our study represents the first demonstration of a hormetic effect of MG defined by a low-dose stimulation and a high-dose inhibition of tumour growth.
- Published
- 2017
- Full Text
- View/download PDF
27. Myoferlin Is a Yet Unknown Interactor of the Mitochondrial Dynamics' Machinery in Pancreas Cancer Cells
- Author
-
Gilles Rademaker, Raphaël Peiffer, Alexandre Hego, Ferman Agirman, Naïma Maloujahmoum, Louise Deldicque, Sandy Anania, Vincent Castronovo, Olivier Peulen, Marc Francaux, Akeila Bellahcene, Marc Thiry, and UCL - SSS/IONS/CEMO - Pôle Cellulaire et moléculaire
- Subjects
0301 basic medicine ,Cancer Research ,mitofusin ,Biology ,Mitochondrion ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,myoferlin ,pancreas cancer ,Pancreatic cancer ,medicine ,Colocalization ,Cancer ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,mitochondria ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Mitochondrial fission ,Pancreas ,Function (biology) - Abstract
Pancreas ductal adenocarcinoma is one of the deadliest cancers where surgery remains the main survival factor. Mitochondria were described to be involved in tumor aggressiveness in several cancer types including pancreas cancer. We have previously reported that myoferlin controls mitochondrial structure and function, and demonstrated that myoferlin depletion disturbs the mitochondrial dynamics culminating in a mitochondrial fission. In order to unravel the mechanism underlying this observation, we explored the myoferlin localization in pancreatic cancer cells and showed a colocalization with the mitochondrial dynamic machinery element: mitofusin. This colocalization was confirmed in several pancreas cancer cell lines and in normal cell lines as well. Moreover, in pancreas cancer cell lines, it appeared that myoferlin interacted with mitofusin. These discoveries open-up new research avenues aiming at modulating mitofusin function in pancreas cancer.
- Published
- 2020
28. Methylglyoxal Scavengers Resensitize KRAS-Mutated Colorectal Tumors to Cetuximab
- Author
-
Justine Bellier, Marie-Julie Nokin, Maurine Caprasse, Assia Tiamiou, Arnaud Blomme, Jean L. Scheijen, Benjamin Koopmansch, Gillian M. MacKay, Barbara Chiavarina, Brunella Costanza, Gilles Rademaker, Florence Durieux, Ferman Agirman, Naïma Maloujahmoum, Pino G. Cusumano, Pierre Lovinfosse, Hing Y. Leung, Frédéric Lambert, Vincent Bours, Casper G. Schalkwijk, Roland Hustinx, Olivier Peulen, Vincent Castronovo, Akeila Bellahcène, MUMC+: MA Alg Onderzoek Interne Geneeskunde (9), Interne Geneeskunde, and RS: Carim - V01 Vascular complications of diabetes and metabolic syndrome
- Subjects
Adult ,Male ,Glycosylation ,akt ,growth ,HSP27 Heat-Shock Proteins ,heat-shock-protein ,Cetuximab ,Mechanistic Target of Rapamycin Complex 2 ,Mice, SCID ,resistance ,Proto-Oncogene Proteins p21(ras) ,Phosphatidylinositol 3-Kinases ,Mice, Inbred NOD ,Stress, Physiological ,Cell Line, Tumor ,Animals ,Humans ,neoplasms ,lcsh:QH301-705.5 ,ras ,Aged ,Cell Proliferation ,Aged, 80 and over ,Carnosine ,Free Radical Scavengers ,Middle Aged ,Pyruvaldehyde ,digestive system diseases ,heat-shock-protein-27 ,targeted therapies ,Clone Cells ,Enzyme Activation ,lcsh:Biology (General) ,Mutation ,cancer cells ,hsp27 ,Colorectal Neoplasms ,metabolism ,Glycolysis ,Proto-Oncogene Proteins c-akt - Abstract
Summary: The use of cetuximab anti-epidermal growth factor receptor (anti-EGFR) antibodies has opened the era of targeted and personalized therapy in colorectal cancer (CRC). Poor response rates have been unequivocally shown in mutant KRAS and are even observed in a majority of wild-type KRAS tumors. Therefore, patient selection based on mutational profiling remains problematic. We previously identified methylglyoxal (MGO), a by-product of glycolysis, as a metabolite promoting tumor growth and metastasis. Mutant KRAS cells under MGO stress show AKT-dependent survival when compared with wild-type KRAS isogenic CRC cells. MGO induces AKT activation through phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin 2 (mTORC2) and Hsp27 regulation. Importantly, the sole induction of MGO stress in sensitive wild-type KRAS cells renders them resistant to cetuximab. MGO scavengers inhibit AKT and resensitize KRAS-mutated CRC cells to cetuximab in vivo. This study establishes a link between MGO and AKT activation and pinpoints this oncometabolite as a potential target to tackle EGFR-targeted therapy resistance in CRC. : Bellier et al. demonstrate that MGO stress is a constant feature of KRAS-mutated CRC tumors. MGO induces a key survival pathway implicated in resistance to EGFR-targeted therapy in CRC. The scavenging of this oncometabolite could be beneficial in the treatment of both wild-type and mutant KRAS CRC tumors. Keywords: methylglyoxal, colorectal cancer, KRAS mutation, EGFR-targeted therapy, Hsp27, carnosine, aminoguanidine, cetuximab, AKT signaling
- Published
- 2019
29. Development of a prototype device for near real-time surface-enhanced Raman scattering monitoring of biological samples
- Author
-
Laureen Coic, Pierre-Yves Sacre, Julie Horne, Gilles Rademaker, Olivier Peulen, Elodie Dumont, Charlotte De Bleye, Eric Ziemons, and Philippe Hubert
- Subjects
Bioanalysis ,Analyte ,Surface Properties ,Chemistry ,010401 analytical chemistry ,Proteins ,Protein Corona ,Nanotechnology ,02 engineering and technology ,Spectrum Analysis, Raman ,021001 nanoscience & nanotechnology ,01 natural sciences ,0104 chemical sciences ,Analytical Chemistry ,symbols.namesake ,Membrane ,symbols ,Sample preparation ,0210 nano-technology ,Raman spectroscopy ,Raman scattering ,Protein adsorption - Abstract
With the fast growth of bioanalytical surface-enhanced Raman scattering (SERS), analytical methods have had to adapt to the complex nature of biological samples. In particular, interfering species and protein adsorption onto the SERS substrates have been addressed by sample preparation steps, such as precipitation or extraction, and by smart SERS substrate functionalisation. These additional handling steps however result in irreversible sample alteration, which in turn prevents sample monitoring over time. A new methodology, that enables near real-time, non-invasive and non-destructive SERS monitoring of biological samples, is therefore proposed. It combines solid SERS substrates, benefitting from liquid immersion resistance for extended periods of time, with an original protein filtering device and an on-field detection by means of a handheld Raman analyser. The protein removal device aims at avoiding protein surface fouling on the SERS substrate. It consists of an ultracentrifugation membrane fixed under a cell culture insert for multi-well plates. The inside of the insert is dedicated to containing biological samples. The solid SERS substrate and a simple medium, without any protein, are placed under the insert. By carefully selecting the membrane molecular weight cutoff, selective diffusion of small analytes through the device could be achieved whereas larger proteins were retained inside the insert. Non-invasive SERS spectral acquisition was then carried out through the bottom of the multi-well plate. The diffusion of a SERS probe, 2-mercaptopyridine, and of a neurotransmitter having a less intense SERS signal, serotonin, were first successfully monitored with the device. Then, the latter was applied to distinguish between subclones of cancerous cells through differences in metabolite production. This promising methodology showed a high level of versatility, together with the capability to reduce cellular stress and contamination hazards.
- Published
- 2021
- Full Text
- View/download PDF
30. PEGylated and functionalized aliphatic polycarbonate polyplex nanoparticles for intravenous administration of HDAC5 siRNA in cancer therapy
- Author
-
Denis Mottet, Alexandra Baroni, Elodie Hendrick, Géraldine Piel, Julie Laloy, Georges Feller, George R. Dakwar, François Orange, Olivier Peulen, Antoine Frère, Philippe Dubois, Brigitte Evrard, Laetitia Mespouille, Stefaan C. De Smedt, Jérôme Diricq, Katrien Remaut, Kevin Braeckmans, Jean-Michel Dogné, and Anne Sophie Delvigne
- Subjects
intravenous administration ,Aliphatic polycarbonate ,Nanoparticle ,02 engineering and technology ,01 natural sciences ,Polyethylene Glycols ,chemistry.chemical_compound ,Mice ,DESIGN ,Morpholine ,Neoplasms ,Medicine and Health Sciences ,Organic chemistry ,General Materials Science ,Tissue Distribution ,DRUG-DELIVERY ,RNA, Small Interfering ,Guanidine ,chemistry.chemical_classification ,Polymer ,021001 nanoscience & nanotechnology ,HISTONE DEACETYLASE INHIBITORS ,SYSTEMIC GENE DELIVERY ,Protein corona ,polyethylene glycol ,Administration, Intravenous ,0210 nano-technology ,Materials science ,Polyethylene glycol ,CELLULAR UPTAKE ,010402 general chemistry ,Histone Deacetylases ,Intravenous administration ,protein corona ,Dynamic light scattering ,In vivo ,Animals ,polyplex nanoparticles ,Polycarboxylate Cement ,BARRIERS ,Isothermal titration calorimetry ,0104 chemical sciences ,RING-OPENING POLYMERIZATION ,BIOLOGICAL-FLUIDS ,chemistry ,Polyplex nanoparticles ,siRNA ,CELLS ,Biophysics ,Nanoparticles ,aliphatic polycarbonate - Abstract
Guanidine and morpholine functionalized aliphatic polycarbonate polymers are able to deliver efficiently histone deacetylase 5 (HDAC5) siRNA into the cytoplasm of cancer cells in vitro leading to a decrease of cell proliferation were previously developed. To allow these biodegradable and biocompatible polyplex nanoparticles to overcome the extracellular barriers and be effective in vivo after an intravenous injection, polyethylene glycol chains (PEG750 or PEG2000) were grafted on the polymer structure. These nanoparticles showed an average size of about 150 nm and a slightly positive -potential with complete siRNA complexation. Behavior of PEGylated and non-PEGylated polyplexes were investigated in the presence of serum, in terms of siRNA complexation (fluorescence correlation spectroscopy), size (dynamic light scattering and single-particle tracking), interaction with proteins (isothermal titration calorimetry) and cellular uptake. Surprisingly, both PEGylated and non-PEGylated formulations presented relatively good behavior in the presence of fetal bovine serum (FBS). Hemocompatibility tests showed no effect of these polyplexes on hemolysis and coagulation. In vivo biodistribution in mice was performed and showed a better siRNA accumulation at the tumor site for PEGylated polyplexes. However, cellular uptake in protein-rich conditions showed that PEGylated polyplex lost their ability to interact with biological membranes and enter into cells, showing the importance to perform in vitro investigations in physiological conditions closed to in vivo situation. In vitro, the efficiency of PEGylated nanoparticles decreases compared to non-PEGylated particles, leading to the loss of the antiproliferative effect on cancer cells.
- Published
- 2017
31. [Asporin: the protective wall against triple-negative breast cancer]
- Author
-
Akeila Bellahcene, Andrei Turtoi, P. Cusumano, Vincenzo Castronovo, Arnaud Blomme, Olivier Peulen, Université de Liège, Centre Hospitalier Universitaire de Liège (CHU-Liège), Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), and CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)
- Subjects
MESH: Extracellular Matrix Proteins ,0301 basic medicine ,MESH: Humans ,business.industry ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,MESH: Gene Expression Regulation, Neoplastic ,MESH: Triple Negative Breast Neoplasms ,General Medicine ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,030104 developmental biology ,MESH: Cell Transformation, Neoplastic ,MESH: Interleukin-1beta ,Medicine ,business ,MESH: Female ,ComputingMilieux_MISCELLANEOUS ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
International audience
- Published
- 2016
- Full Text
- View/download PDF
32. Myoferlin regulates cellular lipid metabolism and promotes metastases in triple-negative breast cancer
- Author
-
Philippe Delvenne, Sébastien Boutry, Etienne Cavalier, Masahiko Nishiyama, Olivier Peulen, Serge Goldman, Stéphanie Gofflot, Félicie Sherer, Marc Thiry, Arnaud Blomme, Robert N. Muller, Brunella Costanza, François Jouret, Vincenzo Castronovo, Touko Hirano, C. Le Goff, Ange Mouithys-Mickalad, Gilles Doumont, Andrei Turtoi, E Di Valentin, Takehiko Yokobori, Eric Lifrange, G. Van Simaeys, P. de Tullio, E. De Pauw, Akeila Bellahcene, P. Cusumano, Université de Liège, Center for Microscopy and Molecular Imaging (IBMM - CMMI), Université libre de Bruxelles (ULB), University of Mons [Belgium] (UMONS), Biothèque Wallonia-Bruxelles, Centre Hospitalier Universitaire de Liège (CHU-Liège), Mass Spectrometry Laboratory (MS LAB), Université de Liège-Centre d'Analyse des Résidus en Traces-Groupe Interdisciplinaire de Génoprotéomique Appliquée, Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), and CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)
- Subjects
0301 basic medicine ,Cancer Research ,[SDV]Life Sciences [q-bio] ,Muscle Proteins ,Triple Negative Breast Neoplasms ,Mice, SCID ,Biology ,Molecular oncology ,Oxidative Phosphorylation ,Receptor tyrosine kinase ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Growth factor receptor ,Mice, Inbred NOD ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Molecular Biology ,Triple-negative breast cancer ,Calcium-Binding Proteins ,Cytoplasmic Vesicles ,Membrane Proteins ,Cancer ,Cell cycle ,Lipid Metabolism ,medicine.disease ,3. Good health ,Cell biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Heterografts ,Female ,Glycolysis - Abstract
International audience; Myoferlin is a multiple C2-domain-containing protein that regulates membrane repair, tyrosine kinase receptor function and endocytosis in myoblasts and endothelial cells. Recently it has been reported as overexpressed in several cancers and shown to contribute to proliferation, migration and invasion of cancer cells. We have previously demonstrated that myoferlin regulates epidermal growth factor receptor activity in breast cancer. In the current study, we report a consistent overexpression of myoferlin in triple-negative breast cancer cells (TNBC) over cells originating from other breast cancer subtypes. Using a combination of proteomics, metabolomics and electron microscopy, we demonstrate that myoferlin depletion results in marked alteration of endosomal system and metabolism. Mechanistically, myoferlin depletion caused impaired vesicle traffic that led to a misbalance of saturated/unsaturated fatty acids. This provoked mitochondrial dysfunction in TNBC cells. As a consequence of the major metabolic stress, TNBC cells rapidly triggered AMP activated protein kinase-mediated metabolic reprogramming to glycolysis. This reduced their ability to balance between oxidative phosphorylation and glycolysis, rendering TNBC cells metabolically inflexible, and more sensitive to metabolic drug targeting in vitro. In line with this, our in vivo findings demonstrated a significantly reduced capacity of myoferlin-deficient TNBC cells to metastasise to lungs. The significance of this observation was further supported by clinical data, showing that TNBC patients whose tumors overexpress myoferlin have worst distant metastasis-free and overall survivals. This novel insight into myoferlin function establishes an important link between vesicle traffic, cancer metabolism and progression, offering new diagnostic and therapeutic concepts to develop treatments for TNBC patients.
- Published
- 2016
- Full Text
- View/download PDF
33. New role of osteopontin in DNA repair and impact on human glioblastoma radiosensitivity
- Author
-
Philippe Delvenne, Andrei Turtoi, Nicolas Goffart, Elettra Bianchi, Patrick Roncarati, Olivier Peulen, Marie-Julie Nokin, Akeila Bellahcene, Bernard Rogister, Natacha Leroi, Vincenzo Castronovo, Arnaud Blomme, Félix Scholtes, François Lallemand, Aurélie Henry, Yvette Habraken, Philippe Martinive, Paul Peixoto, Jérémy Lambert, Université de Liège, Centre Hospitalier Universitaire de Liège (CHU-Liège), University Medical Center [Utrecht], Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), and Herrada, Anthony
- Subjects
0301 basic medicine ,osteopontin ,DNA Repair ,MESH: Osteopontin ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,MESH: DNA Breaks, Double-Stranded ,Mice, SCID ,Radiation Tolerance ,MESH: Recombinant Proteins ,Mice ,0302 clinical medicine ,MESH: RNA, Small Interfering ,DNA damage repair ,Medicine ,MESH: Animals ,MESH: Gene Silencing ,DNA Breaks, Double-Stranded ,Osteopontin ,MESH: Mice, SCID ,Phosphorylation ,RNA, Small Interfering ,MESH: DNA Repair ,Gene knockdown ,MESH: Radiation Tolerance ,biology ,Brain Neoplasms ,MESH: Glioblastoma ,Recombinant Proteins ,radioresistance ,Oncology ,030220 oncology & carcinogenesis ,MESH: Brain Neoplasms ,[SDV.BBM.GTP] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Female ,Comet Assay ,Research Paper ,EGFRvIII ,MESH: Cell Line, Tumor ,DNA repair ,Mice, Nude ,MESH: Comet Assay ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,03 medical and health sciences ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,stomatognathic system ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Cell Line, Tumor ,Radioresistance ,Glioma ,MESH: Mice, Nude ,Animals ,Humans ,Gene Silencing ,Radiosensitivity ,Clonogenic assay ,MESH: Mice ,MESH: Humans ,MESH: Phosphorylation ,business.industry ,[SDV.NEU.NB] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,glioblastoma ,medicine.disease ,nervous system diseases ,030104 developmental biology ,Cell culture ,biology.protein ,Cancer research ,business ,MESH: Female ,MESH: Neoplasm Transplantation ,Neoplasm Transplantation - Abstract
International audience; Glioblastoma (GBM) represents the most aggressive and common solid human brain tumor. We have recently demonstrated the importance of osteopontin (OPN) in the acquisition/maintenance of stemness characters and tumorigenicity of glioma initiating cells. Consultation of publicly available TCGA database indicated that high OPN expression correlated with poor survival in GBM patients. In this study, we explored the role of OPN in GBM radioresistance using an OPN-depletion strategy in U87-MG, U87-MG vIII and U251-MG human GBM cell lines. Clonogenic experiments showed that OPN-depleted GBM cells were sensitized to irradiation. In comet assays, these cells displayed higher amounts of unrepaired DNA fragments post-irradiation when compared to control. We next evaluated the phosphorylation of key markers of DNA double-strand break repair pathway. Activating phosphorylation of H2AX, ATM and 53BP1 was significantly decreased in OPN-deficient cells. The addition of recombinant OPN prior to irradiation rescued phospho-H2AX foci formation thus establishing a new link between DNA repair and OPN expression in GBM cells. Finally, OPN knockdown improved mice survival and induced a significant reduction of heterotopic human GBM xenograft when combined with radiotherapy. This study reveals a new function of OPN in DNA damage repair process post-irradiation thus further confirming its major role in GBM aggressive disease.
- Published
- 2016
- Full Text
- View/download PDF
34. Transforming growth factor beta-induced, an extracellular matrix interacting protein, enhances glycolysis and promotes pancreatic cancer cell migration
- Author
-
Justine Leenders, Assia Tiamiou, Andrei Turtoi, Pascal de Tullio, Arnaud Blomme, Vincenzo Castronovo, Gilles Rademaker, Justine Bellier, Brunella Costanza, Olivier Peulen, Akeila Bellahcene, Philippe Delvenne, Elettra Bianchi, Université de Liège, Cancer Research UK Beatson Institute [Glasgow], Centre Hospitalier Universitaire de Liège (CHU-Liège), Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), and CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)
- Subjects
Cancer Research ,Stromal cell ,endocrine system diseases ,integrin ,extracellular matrix ,[SDV]Life Sciences [q-bio] ,Chick Embryo ,Focal adhesion ,Transforming Growth Factor beta1 ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Pancreatic cancer ,Cell Line, Tumor ,medicine ,Gene silencing ,Animals ,Humans ,Receptors, Vitronectin ,pancreas ,Gene Silencing ,Extracellular Matrix Proteins ,biology ,Chemistry ,Transforming growth factor beta ,medicine.disease ,Hypoxia-Inducible Factor 1, alpha Subunit ,Survival Analysis ,eye diseases ,digestive system diseases ,3. Good health ,Pancreatic Neoplasms ,Oncology ,030220 oncology & carcinogenesis ,Focal Adhesion Protein-Tyrosine Kinases ,Cancer research ,biology.protein ,Signal transduction ,Glycolysis ,TGFBI ,Transforming growth factor ,Carcinoma, Pancreatic Ductal ,Signal Transduction ,Subcellular Fractions - Abstract
International audience; Pancreatic ductal adenocarcinoma (PDAC) remains a deadly malignancy with no efficient therapy available up-to-date. Glycolysis is the main provider of energetic substrates to sustain cancer dissemination of PDAC. Accordingly, altering the glycolytic pathway is foreseen as a sound approach to trigger pancreatic cancer regression. Here, we show for the first time that high transforming growth factor beta-induced (TGFBI) expression in PDAC patients is associated with a poor outcome. We demonstrate that, although usually secreted by stromal cells, PDAC cells synthesize and secrete TGFBI in quantity correlated with their migratory capacity. Mechanistically, we show that TGFBI activates focal adhesion kinase signaling pathway through its binding to integrin αVβ5, leading to a significant enhancement of glycolysis and to the acquisition of an invasive phenotype. Finally, we show that TGFBI silencing significantly inhibits PDAC tumor development in a chick chorioallantoic membrane assay model. Our study highlights TGFBI as an oncogenic extracellular matrix interacting protein that bears the potential to serve as a target for new anti-PDAC therapeutic strategies.
- Published
- 2018
- Full Text
- View/download PDF
35. Myoferlin plays a key role in VEGFA secretion and impacts tumor-associated angiogenesis in human pancreas cancer
- Author
-
Philippe Delvenne, Arnaud Gonzalez, Andrei Turtoi, Vincenzo Castronovo, Mohammad Arafa, Paul Peixoto, Akeila Bellahcene, Karim Fahmy, Olivier Peulen, and Marc Thiry
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Cell type ,biology ,Angiogenesis ,Cancer ,medicine.disease ,Receptor tyrosine kinase ,3. Good health ,03 medical and health sciences ,Vascular endothelial growth factor A ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Oncology ,Internal medicine ,Pancreatic cancer ,biology.protein ,Cancer research ,medicine ,Adenocarcinoma ,Pancreas - Abstract
Pancreatic ductal adenocarcinoma is one of the most deadly forms of cancers with no satisfactory treatment to date. Recent studies have identified myoferlin, a ferlin family member, in human pancreas adenocarcinoma where its expression was associated to a bad prognosis. However, the function of myoferlin in pancreas adenocarcinoma has not been reported. In other cell types, myoferlin is involved in several key plasma membrane processes such as fusion, repair, endocytosis and tyrosine kinase receptor activity. In this study, we showed that myoferlin silencing in BxPC-3 human pancreatic cancer cells resulted in the inhibition of cell proliferation in vitro and in a significant reduction of the tumor volume in chick chorioallantoic membrane assay. In addition to be smaller, the tumors formed by the myoferlin-silenced cells showed a marked absence of functional blood vessels. We further demonstrated that this effect was due, at least in part, to an inhibition of VEGFA secretion by BxPC-3 myoferlin-silenced cells. Using immunofluorescence and electron microscopy, we linked the decreased VEGFA secretion to an impairment of VEGFA exocytosis. The clinical relevance of our results was further strengthened by a significant correlation between myoferlin expression in a series of human pancreatic malignant lesions and their angiogenic status evaluated by the determination of the blood vessel density.
- Published
- 2015
- Full Text
- View/download PDF
36. Targeting osteopontin suppresses glioblastoma stem-like cell character and tumorigenicityin vivo
- Author
-
Zofia von Marschall, Olivier Peulen, Jean-Yves Delattre, Marc Sanson, Alain Chariot, Jérôme Kroonen, Tieu-Lan Chau, Marie-Julie Nokin, Akeila Bellahcene, Aurélie Henry, Andrei Turtoi, Vincent Castronovo, Virginie Lamour, Larry W. Fisher, and Bernard Rogister
- Subjects
Homeobox protein NANOG ,0303 health sciences ,Cancer Research ,biology ,Cell growth ,CD44 ,03 medical and health sciences ,0302 clinical medicine ,stomatognathic system ,Oncology ,SOX2 ,Tumor progression ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,Cancer research ,Osteopontin ,Autocrine signalling ,PI3K/AKT/mTOR pathway ,030304 developmental biology - Abstract
Osteopontin (OPN) is a secreted protein involved in most aspects of tumor progression and metastasis development. Elevated OPN expression has been reported in multiple types of cancer including glioblastoma (GBM), the highest grade and most aggressive brain tumor. GBMs contain a subpopulation of glioma-initiating cells (GICs) implicated in progression, therapeutic resistance and recurrence. We have previously demonstrated that OPN silencing inhibited GBM cell growth in vitro and in vivo. Moreover, activation of CD44 signaling upon OPN ligation has been recently implicated in the acquisition of a stem cell phenotype by GBM cells. The present study is aimed to explore OPN autocrine function using shRNA silencing strategy in GICs enriched from GBM cell lines and a human primary GBM grown in EGF and bFGF defined medium. The removal of these growth factors and addition of serum induced a significant loss of OPN expression in GICs. We showed that OPN-silenced GICs were unable to grow as spheres and this capacity was restored by exogenous OPN. Importantly, the expression of Sox2, Oct3/4 and Nanog, key stemness transcription factors, was significantly decreased in GICs upon OPN targeting. We identified Akt/mTOR/p70S6K as the main signaling pathway triggered following OPN-mediated EGFR activation in GICs. Finally, in an orthotopic xenograft mouse model, the tumorigenic potential of U87-MG sphere cells was completely abrogated upon OPN silencing. Our demonstration of endogenous OPN major regulatory effects on GICs stemness phenotype and tumorigenicity implies a greater role than anticipated for OPN in GBM pathogenesis from initiation and progression to probable recurrence.
- Published
- 2015
- Full Text
- View/download PDF
37. Impact of the Structure of Biocompatible Aliphatic Polycarbonates on siRNA Transfection Ability
- Author
-
Laetitia Mespouille, Brigitte Evrard, Antoine Frère, Sarah Tempelaar, Philippe Dubois, Géraldine Piel, Denis Mottet, Elodie Hendrick, Michał Kawalec, Paul Peixoto, and Olivier Peulen
- Subjects
Polymers and Plastics ,Morpholino ,Polymers ,Carbonates ,Gene Expression ,Bioengineering ,Transfection ,Histone Deacetylases ,RNAi Therapeutics ,Biomaterials ,HeLa ,chemistry.chemical_compound ,Morpholine ,Materials Chemistry ,Humans ,RNA, Small Interfering ,Polycarbonate ,Guanidine ,biology ,biology.organism_classification ,Combinatorial chemistry ,chemistry ,Biochemistry ,Gene Knockdown Techniques ,visual_art ,visual_art.visual_art_medium ,RNA Interference ,Trimethylene carbonate ,HeLa Cells - Abstract
RNAi therapeutics are promising therapeutic tools that have sparked the interest of many researchers. In an effort to provide a safe alternative to PEI, we have designed a series of new guanidinium- and morpholino-functionalized biocompatible and biodegradable polycarbonate vectors. The impact of different functions (morpholino-, guanidinium-, hydrophobic groups) of the architecture (linear homopolymer to dumbbell-shape) and of the molecular weight of these copolymers on their capacity to form polyplexes and to decrease the expression of two epigenetic regulators of gene expression, HDAC7 and HDAC5, was evaluated. The use of one of these polymers combining morpholine and guanidine functions at the ratio >1 and hydrophobic trimethylene carbonate groups showed a significant decrease of mRNA and protein level in HeLa cells, similar to PEI. These results highlight the potential of polycarbonate vectors for future in vivo application as an anticancer therapy.
- Published
- 2015
- Full Text
- View/download PDF
38. Myoferlin controls mitochondrial structure and activity in pancreatic ductal adenocarcinoma, and affects tumor aggressiveness
- Author
-
Gilles, Rademaker, Vincent, Hennequière, Laura, Brohée, Marie-Julie, Nokin, Pierre, Lovinfosse, Florence, Durieux, Stéphanie, Gofflot, Justine, Bellier, Brunella, Costanza, Michael, Herfs, Raphael, Peiffer, Lucien, Bettendorff, Christophe, Deroanne, Marc, Thiry, Philippe, Delvenne, Roland, Hustinx, Akeila, Bellahcène, Vincent, Castronovo, and Olivier, Peulen
- Subjects
endocrine system diseases ,Calcium-Binding Proteins ,Membrane Proteins ,Muscle Proteins ,Adenocarcinoma ,digestive system diseases ,Oxidative Phosphorylation ,Article ,Mitochondria ,Gene Expression Regulation, Neoplastic ,Pancreatic Neoplasms ,Adenosine Triphosphate ,Cell Line, Tumor ,Autophagy ,Humans ,RNA, Small Interfering ,Energy Metabolism ,Glycolysis ,Carcinoma, Pancreatic Ductal ,Cell Proliferation - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related death. Therapeutic options remain very limited and are based on classical chemotherapies. Energy metabolism reprogramming appears as an emerging hallmark of cancer and is considered a therapeutic target with considerable potential. Myoferlin, a ferlin family member protein overexpressed in PDAC, is involved in plasma membrane biology and has a tumor-promoting function. In the continuity of our previous studies, we investigated the role of myoferlin in the context of energy metabolism in PDAC. We used selected PDAC tumor samples and PDAC cell lines together with small interfering RNA technology to study the role of myoferlin in energetic metabolism. In PDAC patients, we showed that myoferlin expression is negatively correlated with overall survival and with glycolytic activity evaluated by 18F-deoxyglucose positron emission tomography. We found out that myoferlin is more abundant in lipogenic pancreatic cancer cell lines and is required to maintain a branched mitochondrial structure and a high oxidative phosphorylation activity. The observed mitochondrial fission induced by myoferlin depletion led to a decrease of cell proliferation, ATP production, and autophagy induction, thus indicating an essential role of myoferlin for PDAC cell fitness. The metabolic phenotype switch generated by myoferlin silencing could open up a new perspective in the development of therapeutic strategies, especially in the context of energy metabolism.
- Published
- 2017
39. Triple negative tumors accumulate significantly less methylglyoxal specific adducts than other human breast cancer subtypes
- Author
-
Dominique Belpomme, Florence Durieux, Paul Peixoto, Marie-Julie Nokin, Philippe Delvenne, Andrei Turtoi, Koji Uchida, Akeila Bellahcene, Barbara Chiavarina, Elettra Bianchi, Philippe Irigaray, Vincent Castronovo, and Olivier Peulen
- Subjects
Glycation End Products, Advanced ,Triple Negative Breast Neoplasms ,Arginine ,glyoxalase 1 ,chemistry.chemical_compound ,Lactoylglutathione lyase ,Breast cancer ,breast cancer ,advanced glycation end-products ,Glycation ,Arg-pyrimidine adducts ,Cell Line, Tumor ,medicine ,methylglyoxal ,Humans ,biology ,Methylglyoxal ,Lactoylglutathione Lyase ,Cancer ,medicine.disease ,Pyruvaldehyde ,Immunohistochemistry ,Pyrimidines ,Oncology ,chemistry ,Cancer cell ,Immunology ,biology.protein ,Cancer research ,MCF-7 Cells ,Adenocarcinoma ,Female ,Research Paper - Abstract
Metabolic syndrome and type 2 diabetes are associated with increased risk of breast cancer development and progression. Methylglyoxal (MG), a glycolysis by-product, is generated through a non-enzymatic reaction from triose-phosphate intermediates. This dicarbonyl compound is highly reactive and contributes to the accumulation of advanced glycation end products. In this study, we analyzed the accumulation of Arg-pyrimidine, a MG-arginine adduct, in human breast adenocarcinoma and we observed a consistent increase of Arg-pyrimidine in cancer cells when compared with the non-tumoral counterpart. Further immunohistochemical comparative analysis of breast cancer subtypes revealed that triple negative lesions exhibited low accumulation of Arg-pyrimidine compared with other subtypes. Interestingly, the activity of glyoxalase 1 (Glo-1), an enzyme that detoxifies MG, was significantly higher in triple negative than in other subtype lesions, suggesting that these aggressive tumors are able to develop an efficient response against dicarbonyl stress. Using breast cancer cell lines, we substantiated these clinical observations by showing that, in contrast to triple positive, triple negative cells induced Glo-1 expression and activity in response to MG treatment. This is the first report that Arg-pyrimidine adduct accumulation is a consistent event in human breast cancer with a differential detection between triple negative and other breast cancer subtypes.
- Published
- 2014
40. ΔNp63 isoform-mediated β-defensin family up-regulation is associated with (lymph)angiogenesis and poor prognosis in patients with squamous cell carcinoma
- Author
-
Arnaud Gonzalez, Vincent Castronovo, Jacques Boniver, Olivier Peulen, Pascale Hubert, Sven Saussez, Patrick Roncarati, Anaelle Duray, Philippe Delvenne, Michael Herfs, Agnès Noël, Meggy Suarez-Carmona, and Charlotte Erpicum
- Subjects
Adult ,Keratinocytes ,Male ,Receptors, CCR6 ,squamous cell carcinoma ,Pathology ,medicine.medical_specialty ,beta-Defensins ,Angiogenesis ,Uterine Cervical Neoplasms ,(lymph)angiogenesis ,Biology ,Transfection ,Metastasis ,Neovascularization ,Cell Line, Tumor ,Tumor Microenvironment ,Carcinoma ,medicine ,Humans ,Genes, Tumor Suppressor ,RNA, Messenger ,Lymphangiogenesis ,RNA, Small Interfering ,Aged ,p63 ,Tumor microenvironment ,Neovascularization, Pathologic ,Chemotaxis ,Tumor Suppressor Proteins ,Endothelial Cells ,Middle Aged ,Prognosis ,medicine.disease ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Survival Rate ,Lymphatic system ,Oncology ,Head and Neck Neoplasms ,Carcinoma, Squamous Cell ,Female ,Lymph ,medicine.symptom ,defensins ,Transcription Factors ,Research Paper - Abstract
// Meggy Suarez-Carmona 1,2,* , Pascale Hubert 1,* , Arnaud Gonzalez 3 , Anaelle Duray 4 , Patrick Roncarati 1 , Charlotte Erpicum 2 , Jacques Boniver 1 , Vincent Castronovo 3 , Agnes Noel 2 , Sven Saussez 4 , Olivier Peulen 3 , Philippe Delvenne 1,** and Michael Herfs 1,** 1 Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liege, Belgium 2 Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liege, Liege, Belgium 3 Metastasis Research Laboratory, GIGA-Cancer, University of Liege, Liege, Belgium 4 Laboratory of Anatomy, Faculty of Medicine and Pharmacy, University of Mons, Mons, Belgium * These authors contributed equally to this work ** These authors share last authorship Correspondence: Michael Herfs, email: // Keywords : p63, defensins, (lymph)angiogenesis, prognosis, squamous cell carcinoma Received : January 17, 2014 Accepted : March 19, 2014 Published : March 21, 2014 Abstract Beside a role in normal development/differentiation, high p63 immunoreactivity is also frequently observed in squamous cell carcinoma (SCC). Due to the complexity of the gene, the role of each p63 isotype in tumorigenesis is still confusing. Constitutively produced or induced in inflammatory conditions, human beta-defensins (HβDs) are cationic peptides involved in host defenses against bacteria, viruses and fungi. Here, we investigated both the role of p63 proteins in the regulation of HβDs and the implication of these antimicrobial peptides in tumor (lymph)angiogenesis. Thus, in contrast to TAp63 isotypes, we observed that ΔNp63 proteins (α, β, γ) induce HβD1, 2 and 4 expression. Similar results were observed in cancer tissues and cell lines. We next demonstrated that ΔNp63-overexpressing SCC are associated with both a poor prognosis and a high tumor vascularisation and lymphangiogenesis. Moreover, we showed that HβDs exert a chemotactic activity for (lymphatic) endothelial cells in a CCR6-dependent manner. The ability of HβDs to enhance (lymph)angiogenesis in vivo was also evaluated. We observed that HβDs increase the vessel number and induce a significant increase in relative vascular area compared to negative control. Taken together, the results of this study suggest that ΔNp63-regulated HβD could promote tumor (lymph)angiogenesis in SCC microenvironment.
- Published
- 2014
- Full Text
- View/download PDF
41. Organized proteomic heterogeneity in colorectal cancer liver metastases and implications for therapies
- Author
-
Georgios Patsos, Emmanuel Di Valentin, Joan Somja, Philippe Delvenne, Arnaud Blomme, Andrei Turtoi, David Delvaux, Olivier Detry, E Mutijima, Edwin De Pauw, Olivier Peulen, Vincent Castronovo, and Delphine Debois
- Subjects
Hepatology ,Genetic heterogeneity ,Colorectal cancer ,medicine.medical_treatment ,Cancer ,Biology ,Proteomics ,medicine.disease ,Bioinformatics ,Metastasis ,Targeted therapy ,medicine ,Biomarker (medicine) ,TGFBI - Abstract
Tumor heterogeneity is a major obstacle for developing effective anticancer treatments. Recent studies have pointed to large stochastic genetic heterogeneity within cancer lesions, where no pattern seems to exist that would enable a more structured targeted therapy approach. Because to date no similar information is available at the protein (phenotype) level, we employed matrix assisted laser desorption ionization (MALDI) image-guided proteomics and explored the heterogeneity of extracellular and membrane subproteome in a unique collection of eight fresh human colorectal carcinoma (CRC) liver metastases. Monitoring the spatial distribution of over 1,000 proteins, we found unexpectedly that all liver metastasis lesions displayed a reproducible, zonally delineated pattern of functional and therapeutic biomarker heterogeneity. The peritumoral region featured elevated lipid metabolism and protein synthesis, the rim of the metastasis displayed increased cellular growth, movement, and drug metabolism, whereas the center of the lesion was characterized by elevated carbohydrate metabolism and DNA-repair activity. From the aspect of therapeutic targeting, zonal expression of known and novel biomarkers was evident, reinforcing the need to select several targets in order to achieve optimal coverage of the lesion. Finally, we highlight two novel antigens, LTBP2 and TGFBI, whose expression is a consistent feature of CRC liver metastasis. We demonstrate their in vivo antibody-based targeting and highlight their potential usefulness for clinical applications. Conclusion: The proteome heterogeneity of human CRC liver metastases has a distinct, organized pattern. This particular hallmark can now be used as part of the strategy for developing rational therapies based on multiple sets of targetable antigens. (Hepatology 2014;59:924–934)
- Published
- 2013
- Full Text
- View/download PDF
42. Methylglyoxal-Mediated Stress Correlates with High Metabolic Activity and Promotes Tumor Growth in Colorectal Cancer
- Author
-
Romain R. Dehon, Serge Goldman, Koji Uchida, Florence Durieux, Akeila Bellahcene, Félicie Sherer, Justine Bellier, Olivier Peulen, Pierre Lovinfosse, Roland Hustinx, Philippe Delvenne, Noëlla Bletard, Marie-Julie Nokin, Pieter Demetter, Barbara Chiavarina, Vincent Castronovo, Laurine Verset, Andrei Turtoi, Université de Liège, Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Université libre de Bruxelles (ULB), and University of Tokyo [Kashiwa Campus]
- Subjects
0301 basic medicine ,Colorectal cancer ,[SDV]Life Sciences [q-bio] ,Carnosine ,Informatique appliquée logiciel ,lcsh:Chemistry ,Cohort Studies ,Physico-chimie générale ,chemistry.chemical_compound ,0302 clinical medicine ,Glycation ,methylglyoxal ,Medicine ,Glycolysis ,lcsh:QH301-705.5 ,Spectroscopy ,Methylglyoxal ,Lactoylglutathione Lyase ,General Medicine ,Middle Aged ,Pyruvaldehyde ,3. Good health ,Computer Science Applications ,Biochemistry ,030220 oncology & carcinogenesis ,Colorectal Neoplasms ,Adult ,colorectal cancer ,Chimie inorganique ,Catalysis ,Article ,glyoxalase 1 ,Inorganic Chemistry ,03 medical and health sciences ,Fluorodeoxyglucose F18 ,Stress, Physiological ,Cell Line, Tumor ,Animals ,Humans ,Spectroscopie [état condense] ,Physical and Theoretical Chemistry ,MG-adducts ,18F-Fluorodeoxyglucose (18F-FDG) ,Molecular Biology ,Aged ,Cell Proliferation ,Neoplasm Staging ,business.industry ,Organic Chemistry ,Biologie moléculaire ,Cancer ,Chimie théorique ,medicine.disease ,Chimie organique ,Spectroscopie [électromagnétisme, optique, acoustique] ,030104 developmental biology ,Pyrimidines ,Glyoxalase 1 ,lcsh:Biology (General) ,lcsh:QD1-999 ,chemistry ,Anaerobic glycolysis ,Positron-Emission Tomography ,Cancer cell ,Cancer research ,Catalyses hétérogène et homogène ,business ,Chickens - Abstract
Cancer cells generally rely on aerobic glycolysis as a major source of energy. Methylglyoxal (MG), a dicarbonyl compound that is produced as a side product during glycolysis, is highly reactive and induces the formation of advanced glycation end-products that are implicated in several pathologies including cancer. All mammalian cells have an enzymatic defense against MG composed by glyoxalases GLO1 and GLO2 that converts MG to D-lactate. Colorectal cancer (CRC) is one of the most frequently occurring cancers with high morbidity and mortality. In this study, we used immunohistochemistry to examine the level of MG protein adducts, in a series of 102 CRC human tumors divided into four clinical stages. We consistently detected a high level of MG adducts and low GLO1 activity in high stage tumors compared to low stage ones suggesting a pro-tumor role for dicarbonyl stress. Accordingly, GLO1 depletion in CRC cells promoted tumor growth in vivo that was efficiently reversed using carnosine, a potent MG scavenger. Our study represents the first demonstration that MG adducts accumulation is a consistent feature of high stage CRC tumors. Our data point to MG production and detoxification levels as an important molecular link between exacerbated glycolytic activity and CRC progression., SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2016
- Full Text
- View/download PDF
43. Methylglyoxal, a glycolysis side-product, induces Hsp90 glycation and YAP-mediated tumor growth and metastasis
- Author
-
Barbara Chiavarina, Brunella Costanza, Justine Leenders, Philippe Delvenne, Elettra Bianchi, James R. Cochrane, Koji Uchida, Pascal de Tullio, Paul Peixoto, Akeila Bellahcene, Craig A. Hutton, Olivier Peulen, Dominique Baiwir, Marc Thiry, Casper G. Schalkwijk, Vincent Castronovo, Andrei Turtoi, Dominique Belpomme, Arnaud Blomme, Florence Durieux, Jean L.J.M. Scheijen, Nicolas Smargiasso, Marie-Julie Nokin, Edwin De Pauw, David Spiegel, Promovendi CD, Interne Geneeskunde, MUMC+: MA Alg Interne Geneeskunde (9), RS: CARIM - R3.01 - Vascular complications of diabetes and the metabolic syndrome, Université de Liège, Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Maastricht University [Maastricht], Cardiovascular Research Institute Maastricht (CARIM), Centre Hospitalier Universitaire de Liège (CHU-Liège), Nagoya University, Yale University [New Haven], University of Melbourne, and Association for Research and Treatments Against Cancer (ARTAC)
- Subjects
Glycation End Products, Advanced ,0301 basic medicine ,Glycosylation ,Metastasis ,chemistry.chemical_compound ,Lactoylglutathione lyase ,Glycation ,cell biology ,methylglyoxal ,Neoplasm Metastasis ,Biology (General) ,cancer biology ,General Neuroscience ,MESH: Glycation End Products, Advanced ,Methylglyoxal ,General Medicine ,Pyruvaldehyde ,MESH: Glycosylation ,Aerobiosis ,LATS1 ,3. Good health ,carbonyl stress ,MESH: Glycolysis ,Medicine ,YAP ,Glycolysis ,Research Article ,medicine.medical_specialty ,MESH: Pyruvaldehyde ,MESH: Cell Line, Tumor ,QH301-705.5 ,Science ,chicken ,Breast Neoplasms ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Biology ,MESH: Phosphoproteins ,glyoxalase 1 ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,breast cancer ,Cell Line, Tumor ,MESH: Aerobiosis ,MESH: Cell Proliferation ,Internal medicine ,MESH: HSP90 Heat-Shock Proteins ,medicine ,Humans ,HSP90 Heat-Shock Proteins ,human ,[SDV.BBM.BC]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biochemistry [q-bio.BM] ,mouse ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,MESH: Adaptor Proteins, Signal Transducing ,Hippo signaling pathway ,MESH: Humans ,General Immunology and Microbiology ,YAP-Signaling Proteins ,Phosphoproteins ,medicine.disease ,MESH: Neoplasm Metastasis ,030104 developmental biology ,Endocrinology ,chemistry ,Tumor progression ,Anaerobic glycolysis ,MESH: Protein Processing, Post-Translational ,Cancer cell ,biology.protein ,Cancer research ,Protein Processing, Post-Translational ,MESH: Breast Neoplasms ,Transcription Factors - Abstract
Metabolic reprogramming toward aerobic glycolysis unavoidably induces methylglyoxal (MG) formation in cancer cells. MG mediates the glycation of proteins to form advanced glycation end products (AGEs). We have recently demonstrated that MG-induced AGEs are a common feature of breast cancer. Little is known regarding the impact of MG-mediated carbonyl stress on tumor progression. Breast tumors with MG stress presented with high nuclear YAP, a key transcriptional co-activator regulating tumor growth and invasion. Elevated MG levels resulted in sustained YAP nuclear localization/activity that could be reverted using Carnosine, a scavenger for MG. MG treatment affected Hsp90 chaperone activity and decreased its binding to LATS1, a key kinase of the Hippo pathway. Cancer cells with high MG stress showed enhanced growth and metastatic potential in vivo. These findings reinforce the cumulative evidence pointing to hyperglycemia as a risk factor for cancer incidence and bring renewed interest in MG scavengers for cancer treatment. DOI: http://dx.doi.org/10.7554/eLife.19375.001
- Published
- 2016
- Full Text
- View/download PDF
44. Author response: Methylglyoxal, a glycolysis side-product, induces Hsp90 glycation and YAP-mediated tumor growth and metastasis
- Author
-
Jean L.J.M. Scheijen, Andrei Turtoi, James R. Cochrane, Florence Durieux, Dominique Baiwir, Paul Peixoto, Marie-Julie Nokin, Dominique Belpomme, Akeila Bellahcene, Pascal De Tullio, Edwin De Pauw, David Spiegel, Elettra Bianchi, Vincent Castronovo, Marc Thiry, Nicolas Smargiasso, Arnaud Blomme, Brunella Costanza, Craig A. Hutton, Olivier Peulen, Philippe Delvenne, Koji Uchida, Barbara Chiavarina, Casper G. Schalkwijk, and Justine Leenders
- Subjects
0301 basic medicine ,biology ,Methylglyoxal ,medicine.disease ,Hsp90 ,Metastasis ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,Glycation ,030220 oncology & carcinogenesis ,Side product ,Cancer research ,biology.protein ,medicine ,Glycolysis ,Tumor growth - Published
- 2016
- Full Text
- View/download PDF
45. Differential proteomic analysis of a human breast tumor and its matched bone metastasis identifies cell membrane and extracellular proteins associated with bone metastasis
- Author
-
Andrei Turtoi, Vincent Castronovo, Noëlla Bletard, Bruno Dumont, Edwin De Pauw, Philippe Clézardin, Olivier Peulen, Akeila Bellahcene, and Philippe Delvenne
- Subjects
Proteomics ,CA15-3 ,Cell ,Bone Neoplasms ,Breast Neoplasms ,Biology ,Biochemistry ,Bone remodeling ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,medicine ,Humans ,030304 developmental biology ,Extracellular Matrix Proteins ,0303 health sciences ,Histocytochemistry ,Membrane Proteins ,Bone metastasis ,General Chemistry ,medicine.disease ,Primary tumor ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer cell ,Immunology ,Cancer research ,Female - Abstract
The classical fate of metastasizing breast cancer cells is to seed and form secondary colonies in bones. The molecules closely associated with these processes are predominantly present at the cell surface and in the extracellular space, establishing the first contacts with the target tissue. In this study, we had the rare opportunity to analyze a bone metastatic lesion and its corresponding breast primary tumor obtained simultaneously from the same patient. Using mass spectrometry, we undertook a proteomic study on cell surface and extracellular protein-enriched material. We provide a repertoire of significantly modulated proteins, some with yet unknown roles in the bone metastatic process as well as proteins notably involved in cancer cell invasiveness and in bone metabolism. The comparison of these clinical data with those previously obtained using a human osteotropic breast cancer cell line highlighted an overlapping group of proteins. Certain differentially expressed proteins are validated in the present study using immunohistochemistry on a retrospective collection of breast tumors and matched bone metastases. Our exclusive set of selected proteins supports the setup of further investigations on both clinical samples and experimental bone metastasis models that will help to reveal the finely coordinated expression of proteins that favor the development of metastases in the bone microenvironment.
- Published
- 2012
- Full Text
- View/download PDF
46. Intestinal growth and pathology ofGiardia duodenalisassemblage subtype AI, AII, B and E in the gerbil model
- Author
-
Louis Maes, Paul Cos, Olivier Peulen, C. Van Ginneken, Ely Bénéré, and T. Van Assche
- Subjects
Giardiasis ,Male ,Genotype ,Veterinary medicine ,Virulence ,medicine.disease_cause ,Gerbil ,Microbiology ,Feces ,Intestine, Small ,medicine ,Animals ,Humans ,Giardia lamblia ,Trophozoites ,biology ,Ecology ,Pharmacology. Therapy ,Giardia ,biology.organism_classification ,Enzyme assay ,Small intestine ,Disaccharidase ,Disease Models, Animal ,Infectious Diseases ,medicine.anatomical_structure ,biology.protein ,Alkaline phosphatase ,Animal Science and Zoology ,Parasitology ,Human medicine ,Gerbillinae - Abstract
SUMMARYThis study investigated the significance of the genetic differences between assemblages A, B and E on intestinal growth and virulence. Intestinal growth and virulence were studied in 2 laboratory (AI: WB and B: GS/M-83-H7) and 6 field isolates of assemblage subtype AI, AII, B and EIII. Intestinal trophozoite burdens, body weight and faecal consistency were monitored until day 29 post-infection (p.i.), morphological (mucosal architecture and inflammation) and functional (disaccharidase and alkaline phosphatase enzyme activity) damage to the small intestine were evaluated on days 7 and 18 p.i. The assemblage subtypes AIand B were more infectious and produced higher trophozoite loads for a longer period compared to the subtypes AIIand EIII. The body weight of infected gerbils was significantly reduced compared to uninfected controls, but did not differ between the assemblage subtypes. Consistent softening of the faeces was only observed with assemblage B. Assemblage B next to assemblage subtype AIelicited relatively higher pathogenicity, characterized by more extensive damage to mucosal architecture, decreased brush-border enzyme function and infiltration of inflammatory cells. Assemblage EIIIand AIIisolates showed relatively low virulence. TheGiardiaassemblage subtypes exhibit different levels of growth and virulence in the gerbil model.
- Published
- 2012
- Full Text
- View/download PDF
47. Cellular uptake of liposomes monitored by confocal microscopy and flow cytometry
- Author
-
Géraldine Piel, Olivier Peulen, Brigitte Evrard, and Emilie Ducat
- Subjects
Liposome ,medicine.diagnostic_test ,Cell ,Pharmaceutical Science ,Biology ,Cell sorting ,In vitro ,law.invention ,Flow cytometry ,Cell biology ,medicine.anatomical_structure ,Confocal microscopy ,law ,Drug delivery ,Confocal laser scanning microscopy ,medicine ,Biophysics - Abstract
For several years, two advanced techniques, confocal laser scanning microscopy (CLSM) and flow cytometry, in particular fluorescence-activated cell sorting (FACS) have been used more and more to study the cellular uptake of liposomal drug delivery systems. These techniques provide new potential to localize carriers in cells and quantify the amount of liposomal uptake, leading to essential information on the interaction between the formulation and the target cell. A better understanding of the underlying mechanism behavior of liposomes in biological systems is essential when adapting the liposomal formulation in order to improve carrier effectiveness. The present review describes these two techniques and their use in liposomal research.
- Published
- 2011
- Full Text
- View/download PDF
48. PO-225 Dual role of methylglyoxal, a glycolysis side-product, in cancer
- Author
-
Akeila Bellahcene, Marie-Julie Nokin, David Spiegel, Koji Uchida, Vincenzo Castronovo, Justine Bellier, Florence Durieux, Craig A. Hutton, and Olivier Peulen
- Subjects
Cancer Research ,Methylglyoxal ,Cancer ,Oxidative phosphorylation ,Glutathione ,Pharmacology ,medicine.disease ,chemistry.chemical_compound ,Oncology ,chemistry ,Cell culture ,Cancer cell ,medicine ,Glycolysis ,Glyoxalase system - Abstract
Introduction Methylglyoxal (MG) is spontaneously produced during glycolysis. MG reacts with proteins, lipids and DNA, thus inducing a dicarbonyl stress. Glyoxalase 1 (GLO1) detoxifies MG into D-Lactate. High MG is notably associated with diabetes and cancer as the latter presents a deregulated energy metabolism where tumour cells use glycolysis rather than mitochondrial respiration. The impact of MG on cancer is ambiguous as some studies reported an anti-tumour and others a pro-tumour effect upon GLO1 silencing. Material and methods In this study, we aimed to evaluate the effect of MG stress on a panel of cancer cell lines: MDA-MB-231 known to be glycolytic and MCF7 cells known to be more oxidative breast cancer cells and U87-MG and U251 glioblastoma cell lines. We characterised glycolysis, MG production and detoxification under different conditions in these cell lines. U87-MG tumour growth upon MG treatment was evaluated using the chick chorioallantoic membrane tumour model (CAM). Results and discussions We observed that only MDA-MB-231 and U87-MG cell lines are able to increase their glycolysis flux upon glucose stimulation. In these conditions, both cell lines increased their MG production and detoxification via the glyoxalase system. Under exogenous MG treatment, both glycolytic cell lines showed increased expression of both NRF2 and its target gene GLO1, whereas no ROS accumulation was detected. Upon GLO1 activity inhibition, an alternative MG detoxification system, the aldo-keto reductase (AKR) enzymes, is significantly increased at least in U87-MG cells. Finally, U87-MG cells xenografted on CAM and treated with MG showed an increased growth at low MG doses while high doses reduced tumour growth. Both effects could be reversed by co-treatment with a potent MG-scavenger, L-Carnosine. Conclusion Our data demonstrate for the first time that cancer cells are not equal when facing MG stress. Their response to MG is mainly dependent upon energy metabolism and detoxification capacity (including GLO1 and AKRs). Reduced glutathione level is also an important feature to consider. Ongoing studies will help identify the molecular mecanisms underlying MG dual effect in cancer.
- Published
- 2018
- Full Text
- View/download PDF
49. PO-242 Myoferlin controls mitochondrial structure and metabolism in pancreatic ductal adenocarcinoma, and affects tumor aggressiveness
- Author
-
A. Bellhacène, Vincent Hennequière, Marc Thiry, Olivier Peulen, Gilles Rademaker, Laura Brohée, Vincenzo Castronovo, Pierre Lovinfosse, Marie-Julie Nokin, and Michael Herfs
- Subjects
Cancer Research ,Cell growth ,Cell ,Cancer ,Biology ,medicine.disease ,medicine.anatomical_structure ,Oncology ,Pancreatic cancer ,Mitophagy ,Cancer research ,medicine ,Gene silencing ,Mitochondrial fission ,Pancreas - Abstract
Introduction Pancreatic ductal adenocarcinoma (pdac) is the most common type of pancreatic cancer, and the third leading cause of cancer related death. therapeutic options remain very limited and are still based on classical chemotherapies. cell fraction can survive to the chemotherapy and is responsible for tumor relapse. it appears that these cells rely on oxydative phosphorylation (oxphos) for survival. Myoferlin, a membrane protein involved in cell fusion was recently shown by our laboratory to be overexpressed in pancreatic cancer. Material and methods We used pancreatic cancer cell lines depleted in myoferlin to assess mitochondrial function with an extracellular flux analyser. pancreas cancer samples from the institutional biobank with matched pet scan data were used to correlate myoferlin abundance and glycolysis.results and discussionsin the present study, we discovered that myoferlin was more expressed in cell lines undergoing (oxphos) than in glycolytic cell lines. in the former cell lines, we showed that myoferlin silencing reduced oxphos activity and forced cells to switch to glycolysis. the decrease in oxphos activity is associated with mitochondrial condensation and network disorganization. an increase of dynamin-related protein (drp)-1 phosphorylation in myoferlin-depleted cells led us to suggest mitochondrial fission, reducing cell proliferation, atp production and inducing autophagy and ros accumulation. electron microscopy observation revealed mitophagy, suggesting mitochondrial alterations. To confirm the clinical importance of myoferlin in pdac, we showed that low myoferlin expression was significantly correlated to high overall survival. myoferlin staining of pdac sections was negatively correlated with several 18fdg pet indices indicating that glycolytic lesions had less myoferlin. these observations are fully in accordance with our in vitro data.conclusionas the mitochondrial function was associated with cell chemoresistance, the metabolic switch induced by myoferlin silencing could open up a new perspective in the development of therapeutic strategies. among them, targeting functional domains (c2, dysf, …) of myoferlin should be a priority.
- Published
- 2018
- Full Text
- View/download PDF
50. PO-226 Dicarbonyl stress induces ECM remodelling and MAPK signalling activation in metastatic breast cancer cells
- Author
-
Benoit Charloteaux, Akeila Bellahcene, Olivier Peulen, Vincenzo Castronovo, Michael Herfs, Marie-Julie Nokin, Justine Bellier, S. Van Laere, Charles Lambert, and Gilles Rademaker
- Subjects
MAPK/ERK pathway ,Cancer Research ,Methylglyoxal ,medicine.disease ,Metastatic breast cancer ,Metastasis ,chemistry.chemical_compound ,Breast cancer ,Oncology ,chemistry ,Cancer cell ,Cancer research ,medicine ,Gene silencing ,Glycolysis - Abstract
Introduction Tumour cells use glycolysis rather than mitochondrial respiration to produce their energy. Methylglyoxal (MG), a highly reactive side-product of glycolysis, glycates proteins and nucleic acids thereby inducing dicarbonyl stress in cancer cells. In previous studies, we reported that the accumulation of MG-protein adducts is a constant feature in breast and colon tumours when compared with normal tissues. More recently, we demonstrated that the silencing of glyoxalase 1 (GLO1), the principal MG detoxifying enzyme, in human breast cancer cells generates endogenous dicarbonyl stress resulting in enhanced growth and metastasis in vivo. Material and methods We performed a high-throughput transcriptome profiling of stably GLO1-silenced MDA-MB-231 breast cancer clones in order to identify significant gene expression alterations underlying enhanced invasive and metastatic capacity of breast cancer cells upon MG stress. Results and discussions RNA-Sequencing analysis of GLO1-depleted breast cancer cells pointed to a pro-metastatic signature notably linked to ECM remodelling with the overexpression of collagens and tenascin C. GLO1-depleted cancer cells showed increased anchorage independent growth and invasion abilities, that were reverted in presence of MG scavengers thus connecting these aggressive features with MG stress. Mechanistically, we demonstrated that MG stress induces the activation of MEK/ERK pathway which signals through activated SMAD1. Conclusion Our data show for the first time that neoplasic cells under MG stress directly contribute to ECM changes and MAPK pathway activation which sustain their pro-metastatic phenotype. Based on our data, we propose that the use of MG scavengers represent a promising therapeutic strategy to block the progression of highly glycolytic tumours.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.