Network


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

Hotspot


Dive into the research topics where Félicie Sherer is active.

Publication


Featured researches published by Félicie Sherer.


Journal of Experimental Medicine | 2013

Tristetraprolin regulation of interleukin 23 mRNA stability prevents a spontaneous inflammatory disease

Céline Molle; Tong Zhang; Laure Ysebrant de Lendonck; Cyril Gueydan; Mathieu Andrianne; Félicie Sherer; Gaetan Van Simaeys; Perry J. Blackshear; Oberdan Leo; Stanislas Goriely

Tristetraprolin deficiency results in enhanced IL-23 via dysregulated mRNA decay that leads to an inflammatory syndrome characterized by cachexia, myeloid hyperplasia, dermatitis, and erosive arthritis.


International Journal of Molecular Sciences | 2017

Methylglyoxal-Mediated Stress Correlates with High Metabolic Activity and Promotes Tumor Growth in Colorectal Cancer

Barbara Chiavarina; Marie Julie M.J. Nokin; Justine Bellier; Florence Durieux; Noëlla Bletard; Félicie Sherer; Pierre Lovinfosse; Olivier Peulen; Laurine Verset; Romain R. Dehon; Pieter Demetter; Andrei Turtoi; Koji Uchida; Serge Goldman; Roland Hustinx; Philippe Delvenne; Vincent Castronovo; Akeila Bellahcene

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.


Oncogene | 2017

Myoferlin regulates cellular lipid metabolism and promotes metastases in triple-negative breast cancer.

Arnaud Blomme; Brunella Costanza; P. De Tullio; Marc Thiry; G Van Simaeys; Sébastien Boutry; Gilles Doumont; E Di Valentin; Touko Hirano; Takehiko Yokobori; Stéphanie Gofflot; Olivier Peulen; Akeila Bellahcene; Félicie Sherer; C. Le Goff; Etienne Cavalier; Ange Mouithys-Mickalad; François Jouret; P. Cusumano; Eric Lifrange; Robert N. Muller; Serge Goldman; Philippe Delvenne; E. De Pauw; Masahiko Nishiyama; Vincenzo Castronovo; Andrei Turtoi

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.


The Journal of Nuclear Medicine | 2015

PET/CT with 18F-FDG– and 18F-FBEM–Labeled Leukocytes for Metabolic Activity and Leukocyte Recruitment Monitoring in a Mouse Model of Pulmonary Fibrosis

Benjamin Bondue; Félicie Sherer; Gaetan Van Simaeys; Gilles Doumont; Dominique Egrise; Yousof Yakoub; François Huaux; Marc Parmentier; Sandrine Rorive; Sébastien Sauvage; Simon Lacroix; Olivier Vosters; Paul De Vuyst; Serge Goldman

Idiopathic pulmonary fibrosis is characterized by a progressive and irreversible respiratory failure. Validated noninvasive methods able to assess disease activity are essential for prognostic purposes as well as for the evaluation of emerging antifibrotic treatments. Methods: C57BL/6 mice were used in a murine model of pulmonary fibrosis induced by an intratracheal instillation of bleomycin (control mice were instilled with a saline solution). At different times after instillation, PET/CT with 18F-FDG– or 18F-4-fluorobenzamido-N-ethylamino-maleimide (18F-FBEM)–labeled leukocytes was performed to assess metabolic activity and leukocyte recruitment, respectively. Results: In bleomycin-treated mice, a higher metabolic activity was measured on 18F-FDG PET/CT scans from day 7 to day 24 after instillation, with a peak of activity measured at day 14. Of note, lung mean standardized uptake values correlated with bleomycin doses, histologic score of fibrosis, lung hydroxyproline content, and weight loss. Moreover, during the inflammatory phase of the model (day 7), but not the fibrotic phase (day 23), bleomycin-treated mice presented with an enhanced leukocyte recruitment as assessed by 18F-FBEM–labeled leukocyte PET/CT. Autoradiographic analysis of lung sections and CD45 immunostaining confirm the higher and early recruitment of leukocytes in bleomycin-treated mice, compared with control mice. Conclusion: 18F-FDG– and 18F-FBEM–labeled leukocyte PET/CT enable monitoring of metabolic activity and leukocyte recruitment in a mouse model of pulmonary fibrosis. Implications for preclinical evaluation of antifibrotic therapy are expected.


Contrast Media & Molecular Imaging | 2013

[18F]-FBEM, a tracer targeting cell-surface protein thiols for cell trafficking imaging.

Simon Lacroix; Dominique Egrise; Gaetan Van Simaeys; Gilles Doumont; Michel Monclus; Félicie Sherer; T Herbaux; D Leroy; Serge Goldman

We used [(18)F]-4-fluorobenzamido-N-ethylamino-maleimide ([(18)F]-FBEM) to radiolabel cells ex vivo for in vivo positron emission tomography (PET) in order to assess cell trafficking in mice. In contrast to commonly used imaging agents, [(18)F]-FBEM forms a covalent bond with thiol groups present on the cells surface. The stability of the probe in aqueous medium was tested at different pH values and cross-experiment showed that thiol-labeling efficiency was retained (at least) up to pH 9. The labeling procedure did not affect significantly the cell viability. To illustrate the procedure, PET images of living mice injected intravenously with labeled T lymphocytes were obtained. They showed the expected cell homing in the spleen that was absent in mice injected with free label.


Journal of Liver | 2015

Dynamic Molecular Imaging for Hepatic Function Assessment in Mice: Evaluation in Endotoxin-Induced and Warm Ischemia-Reperfusion Models of Acute Liver Failure

Félicie Sherer; Gaetan Van Simaeys; Jesper Kers; Qing Yuan; Gilles Doumont; Marie-Aline Laute; Cindy Peleman; Dominique Egrise; Tony Lahoutte; Véronique Flamand; Serge Goldman

Background: In hepatic transplantation, inflammatory response related to liver ischemia-reperfusion injury is an important cause of hepatocellular damage that may lead to organ dysfunction. This project aims to develop a new method of dynamic imaging for the local analysis of hepatic function using un-metabolized 99mTc-labeled mebrofenin excretion time in the bile canaliculi as a read-out. Methods: C57BL/6 female mice underwent acute liver damage induced either by endotoxin administration or by warm ischemia-reperfusion. Liver damage intensity was assessed with a 99mTc-labeled mebrofenin dynamic planar imaging protocol, together with biological parameters of liver damage — levels of blood transaminases, liver necrosis and neutrophil infiltration. The acquisition data consisted of a series of 60-frame pinhole images performed on a gamma camera. A region of interest was drawn within the hepatic area in order to measure liver activity on each frame. Excretion rate was quantified as the time necessary for the count value to reach 50% (T0.5Exc) and 20% (T0.2Exc) of the maximum liver count value. We compared biological parameters of liver damage — levels of blood transaminases, liver necrosis and neutrophil infiltration — with 99mTc-labeled mebrofenin excretion times in both models of liver damage and in control animals. Results: 99mTc-labeled mebrofenin excretion times (T0.5Exc and T0.2Exc) were significantly increased in both models of liver damage. Conclusions: We concluded that quantification of liver function is feasible in mice using dynamic planar pinhole imaging with 99mTc-mebrofenin as tracer of the hepato-biliary function. This method is particularly suited to the noninvasive evaluation of immune and pharmacological interventions aiming at a reduction of early liver insults related to ischemic-reperfusion phenomenon.


Oncogene | 2018

Murine stroma adopts a human-like metabolic phenotype in the PDX model of colorectal cancer and liver metastases

Arnaud Blomme; Gaetan Van Simaeys; Gilles Doumont; Brunella Costanza; Justine Bellier; Yukihiro Otaka; Félicie Sherer; Pierre Lovinfosse; Sébastien Boutry; Ana Palacios; Edwin De Pauw; Touko Hirano; Takehiko Yokobori; Roland Hustinx; Akeila Bellahcene; Philippe Delvenne; Olivier Detry; Serge Goldman; Masahiko Nishiyama; Vincent Castronovo; Andrei Turtoi

Cancer research is increasingly dependent of patient-derived xenograft model (PDX). However, a major point of concern regarding the PDX model remains the replacement of the human stroma with murine counterpart. In the present work we aimed at clarifying the significance of the human-to-murine stromal replacement for the fidelity of colorectal cancer (CRC) and liver metastasis (CRC-LM) PDX model. We have conducted a comparative metabolic analysis between 6 patient tumors and corresponding PDX across 4 generations. Metabolic signatures of cancer cells and stroma were measured separately by MALDI-imaging, while metabolite changes in entire tumors were quantified using mass spectrometry approach. Measurement of glucose metabolism was also conducted in vivo using [18F]-fluorodeoxyglucose (FDG) and positron emission tomography (PET). In CRC/CRC-LM PDX model, human stroma was entirely replaced at the second generation. Despite this change, MALDI-imaging demonstrated that the metabolic profiles of both stromal and cancer cells remained stable for at least four generations in comparison to the original patient material. On the tumor level, profiles of 86 water-soluble metabolites as well as 93 lipid mediators underlined the functional stability of the PDX model. In vivo PET measurement of glucose uptake (reflecting tumor glucose metabolism) supported the ex vivo observations. Our data show for the first time that CRC/CRC-LM PDX model maintains the functional stability at the metabolic level despite the early replacement of the human stroma by murine cells. The findings demonstrate that human cancer cells actively educate murine stromal cells during PDX development to adopt the human-like phenotype.


Molecular Imaging and Biology | 2015

Mucolytic Agents Can Enhance HER2 Receptor Accessibility for [(89)Zr]Trastuzumab, Improving HER2 Imaging in a Mucin-Overexpressing Breast Cancer Xenograft Mouse Model.

Zena Wimana; Geraldine Gebhart; Thomas Guiot; Bruno Vanderlinden; Renato Morandini; Gilles Doumont; Félicie Sherer; G. Van Simaeys; Serge Goldman; Ghanem Elias Ghanem; Patrick Flamen


Archive | 2016

Ischemia promotes murin colorectal liver metastases growth: In vivo documentation with preclinical imaging

Desislava Germanova; Jiri Keirsse; Pieter Demetter; Laurine Verset; Maria Antonietta Bali; Sébastien Boutry; Lionel Larbanoix; Félicie Sherer; Gaetan Van Simaeys; Gilles Doumont; Jo A. Van Ginderachter; Vincent Donckier de Donceel; Véronique Flamand


Acta Gastro-enterologica Belgica | 2015

Metabolomic, proteomic and preclinical imaging of patient-derived tumor xenografts for improving treatment of liver metastases patients

A Perez Palacios; Arnaud Blomme; Sébastien Boutry; Gilles Doumont; Félicie Sherer; G Van Simaeys; Delphine Debois; Guy Jerusalem; Joëlle Collignon; Philippe Delvenne; Olivier Detry; Edwin De Pauw; Robert N. Muller; Serge Goldman; Vincenzo Castronovo; Andrei Turtoi

Collaboration


Dive into the Félicie Sherer's collaboration.

Top Co-Authors

Avatar

Gilles Doumont

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Serge Goldman

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Gaetan Van Simaeys

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dominique Egrise

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Benjamin Bondue

Université libre de Bruxelles

View shared research outputs
Researchain Logo
Decentralizing Knowledge