Network


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

Hotspot


Dive into the research topics where Gilles Doumont is active.

Publication


Featured researches published by Gilles Doumont.


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.


Oncotarget | 2016

The human box C/D snoRNAs U3 and U8 are required for pre-rRNA processing and tumorigenesis

Jean-Louis Langhendries; Emilien Nicolas; Gilles Doumont; Serge Goldman; Denis L. J. Lafontaine

Small nucleolar RNAs (snoRNAs) are emerging as a novel class of proto-oncogenes and tumor suppressors; their involvement in tumorigenesis remains unclear. The box C/D snoRNAs U3 and U8 are upregulated in breast cancers. Here we characterize the function of human U3 and U8 in ribosome biogenesis, nucleolar structure, and tumorigenesis. We show in breast (MCF-7) and lung (H1944) cancer cells that U3 and U8 are required for pre-rRNA processing reactions leading, respectively, to synthesis of the small and large ribosomal subunits. U3 or U8 depletion triggers a remarkably potent p53-dependent anti-tumor stress response involving the ribosomal proteins uL5 (RPL11) and uL18 (RPL5). Interestingly, the nucleolar structure is more sensitive to perturbations in lung cancer than in breast cancer cells. We reveal in a mouse xenograft model that the tumorigenic potential of cancer cells is reduced in the case of U3 suppression and totally abolished upon U8 depletion. Tumors derived from U3-knockdown cells displayed markedly lower metabolic volume and activity than tumors derived from aggressive control cancer cells. Unexpectedly, metabolic tracer uptake by U3-suppressed tumors appeared more heterogeneous, indicating distinctive tumor growth properties that may reflect non-conventional regulatory functions of U3 (or fragments derived from it) in mRNA metabolism.


JCI insight | 2017

Tristetraprolin expression by keratinocytes controls local and systemic inflammation

Mathieu Andrianne; Assiya Assabban; Caroline La; Denis Mogilenko; Delphine Staumont Salle; Sébastien Fleury; Gilles Doumont; Gaetan Van Simaeys; Sergei A. Nedospasov; Perry J. Blackshear; David Dombrowicz; Stanislas Goriely; Laurye Van Maele

Tristetraprolin (TTP, encoded by the Zfp36 gene) regulates the mRNA stability of several important cytokines. Due to the critical role of this RNA-binding protein in the control of inflammation, TTP deficiency leads to the spontaneous development of a complex inflammatory syndrome. So far, this phenotype has been largely attributed to dysregulated production of TNF and IL‑23 by myeloid cells, such as macrophages or DCs. Here, we generated mice with conditional deletion of TTP in keratinocytes (Zfp36fl/flK14-Cre mice, referred to herein as Zfp36ΔEP mice). Unlike DC-restricted (CD11c-Cre) or myeloid cell-restricted (LysM-Cre) TTP ablation, these mice developed exacerbated inflammation in the imiquimod-induced psoriasis model. Furthermore, Zfp36ΔEP mice progressively developed a spontaneous pathology with systemic inflammation, psoriatic-like skin lesions, and dactylitis. Finally, we provide evidence that keratinocyte-derived TNF production drives these different pathological features. In summary, these findings expand current views on the initiation of psoriasis and related arthritis by revealing the keratinocyte-intrinsic role of TTP.


Oncotarget | 2017

N-Acetylcysteine breaks resistance to trastuzumab caused by MUC4 overexpression in human HER2 positive BC-bearing nude mice monitored by 89 Zr-Trastuzumab and 18 F-FDG PET imaging

Zena Wimana; Geraldine Gebhart; Thomas Guiot; Bruno Vanderlinden; Denis Larsimont; Gilles Doumont; Gaetan Van Simaeys; Serge Goldman; Patrick Flamen; Ghanem Elias Ghanem

Trastuzumab remains an important drug in the management of human epidermal growth factor receptor 2 (HER2) overexpressing breast cancer (BC). Several studies reported resistance mechanisms to trastuzumab, including impaired HER2-accessibility caused by mucin 4 (MUC4). Previously, we demonstrated an increase of Zirconium-89-radiolabeled-trastuzumab (89Zr-Trastuzumab) accumulation when MUC4-overexpressing BC-cells were challenged with the mucolytic drug N-Acetylcysteine (NAC). Hereby, using the same approach we investigated whether tumor exposure to NAC would also enhance trastuzumab-efficacy.Dual SKBr3 (HER2+/MUC4-, sensitive to trastuzumab) and JIMT1 (HER2+/MUC4+, resistant to trastuzumab) HER2-BC-bearing-xenografts were treated with trastuzumab and NAC. Treatment was monitored by molecular imaging evaluating HER2-accessibility/activity (89Zr-Trastuzumab HER2-immunoPET) and glucose metabolism (18F-FDG-PET/CT), as well as tumor volume and the expression of key proteins.In the MUC4-positive JIMT1-tumors, the NAC-trastuzumab combination resulted in improved tumor-growth control compared to trastuzumab alone; with smaller tumor volume/weight, lower 18F-FDG uptake, lower %Ki67 and pAkt-expression. NAC reduced MUC4-expression, but did not affect HER2-expression or the trastuzumab-sensitivity of the MUC4-negative SKBr3-tumors.These findings suggest that improving HER2-accessibility by reducing MUC4-masking with the mucolytic drug NAC, results in a higher anti-tumor effect of trastuzumab. This provides a rationale for the potential benefit of this approach to possibly treat a subset of HER2-positive BC overexpressing MUC4.Trastuzumab remains an important drug in the management of human epidermal growth factor receptor 2 (HER2) overexpressing breast cancer (BC). Several studies reported resistance mechanisms to trastuzumab, including impaired HER2-accessibility caused by mucin 4 (MUC4). Previously, we demonstrated an increase of Zirconium-89-radiolabeled-trastuzumab (89Zr-Trastuzumab) accumulation when MUC4-overexpressing BC-cells were challenged with the mucolytic drug N-Acetylcysteine (NAC). Hereby, using the same approach we investigated whether tumor exposure to NAC would also enhance trastuzumab-efficacy. Dual SKBr3 (HER2+/MUC4-, sensitive to trastuzumab) and JIMT1 (HER2+/MUC4+, resistant to trastuzumab) HER2-BC-bearing-xenografts were treated with trastuzumab and NAC. Treatment was monitored by molecular imaging evaluating HER2-accessibility/activity (89Zr-Trastuzumab HER2-immunoPET) and glucose metabolism (18F-FDG-PET/CT), as well as tumor volume and the expression of key proteins. In the MUC4-positive JIMT1-tumors, the NAC-trastuzumab combination resulted in improved tumor-growth control compared to trastuzumab alone; with smaller tumor volume/weight, lower 18F-FDG uptake, lower %Ki67 and pAkt-expression. NAC reduced MUC4-expression, but did not affect HER2-expression or the trastuzumab-sensitivity of the MUC4-negative SKBr3-tumors. These findings suggest that improving HER2-accessibility by reducing MUC4-masking with the mucolytic drug NAC, results in a higher anti-tumor effect of trastuzumab. This provides a rationale for the potential benefit of this approach to possibly treat a subset of HER2-positive BC overexpressing MUC4.


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.


Bioconjugate Chemistry | 2017

Nanofitin as a New Molecular-Imaging Agent for the Diagnosis of Epidermal Growth Factor Receptor Over-Expressing Tumors

Marine Goux; Guillaume Becker; Harmony Gorré; Sylvestre Dammicco; Ariane Desselle; Dominique Egrise; Natacha Leroi; François Lallemand; Mohamed Ali Bahri; Gilles Doumont; Alain Plenevaux; Mathieu Cinier; André Luxen

Epidermal growth-factor receptor (EGFR) is involved in cell growth and proliferation and is over-expressed in malignant tissues. Although anti-EGFR-based immunotherapy became a standard of care for patients with EGFR-positive tumors, this strategy of addressing cancer tumors by targeting EGFR with monoclonal antibodies is less-developed for patient diagnostic and monitoring. Indeed, antibodies exhibit a slow blood clearance, which is detrimental for positron emission tomography (PET) imaging. New molecular probes are proposed to overcome such limitations for patient monitoring, making use of low-molecular-weight protein scaffolds as alternatives to antibodies, such as Nanofitins with better pharmacokinetic profiles. Anti-EGFR Nanofitin B10 was reformatted by genetic engineering to exhibit a unique cysteine moiety at its C-terminus, which allows the development of a fast and site-specific radiolabeling procedure with 18F-4-fluorobenzamido-N-ethylamino-maleimide (18F-FBEM). The in vivo tumor targeting and imaging profile of the anti-EGFR Cys-B10 Nanofitin was investigated in a double-tumor xenograft model by static small-animal PET at 2 h after tail-vein injection of the radiolabeled Nanofitin 18F-FBEM-Cys-B10. The image showed that the EGFR-positive tumor (A431) is clearly delineated in comparison to the EGFR-negative tumor (H520) with a significant tumor-to-background contrast. 18F-FBEM-Cys-B10 demonstrated a significantly higher retention in A431 tumors than in H520 tumors at 2.5 h post-injection with a A431-to-H520 uptake ratio of 2.53 ± 0.18 and a tumor-to-blood ratio of 4.55 ± 0.63. This study provides the first report of Nanofitin scaffold used as a targeted PET radiotracer for in vivo imaging of EGFR-positive tumor, with the anti-EGFR B10 Nanofitin used as proof-of-concept. The fast generation of specific Nanofitins via a fully in vitro selection process, together with the excellent imaging features of the Nanofitin scaffold, could facilitate the development of valuable PET-based companion diagnostics.


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

Collaboration


Dive into the Gilles Doumont's collaboration.

Top Co-Authors

Avatar

Serge Goldman

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Félicie Sherer

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

Dominique Egrise

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Simon Lacroix

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge