48 results on '"Loriot, Axelle"'
Search Results
2. Spatial transcriptomics profiling of gallbladder adenocarcinoma: a detailed two-case study of progression from precursor lesions to cancer
- Author
-
Pirenne, Sophie, Manzano-Núñez, Fátima, Loriot, Axelle, Cordi, Sabine, Desmet, Lieven, Aydin, Selda, Hubert, Catherine, Toffoli, Sébastien, Limaye, Nisha, Sempoux, Christine, Komuta, Mina, Gatto, Laurent, and Lemaigre, Frédéric P.
- Published
- 2024
- Full Text
- View/download PDF
3. Influence of a pro-inflammatory stimulus on the miRNA and lipid content of human dental stem cell-derived extracellular vesicles and their impact on microglial activation
- Author
-
Gratpain, Viridiane, Loriot, Axelle, Bottemanne, Pauline, d’Auria, Ludovic, Terrasi, Romano, Payen, Valéry L., van Pesch, Vincent, Muccioli, Giulio G., and des Rieux, Anne
- Published
- 2024
- Full Text
- View/download PDF
4. Gut microbiome modulates tacrolimus pharmacokinetics through the transcriptional regulation of ABCB1
- Author
-
Degraeve, Alexandra L., Haufroid, Vincent, Loriot, Axelle, Gatto, Laurent, Andries, Vanessa, Vereecke, Lars, Elens, Laure, and Bindels, Laure B.
- Published
- 2023
- Full Text
- View/download PDF
5. Specific HIF-2α (Hypoxia-Inducible Factor-2) Inhibitor PT2385 Mitigates Placental Dysfunction In Vitro and in a Rat Model of Preeclampsia (RUPP)
- Author
-
Colson, Arthur, Depoix, Christophe Louis, Lambert, Isaline, Leducq, Camille, Bedin, Marie, De Beukelaer, Michèle, Gatto, Laurent, Loriot, Axelle, de Nieuwburgh, Maureen Peers, Bouhna, Khadija, Baldin, Pamela, Hubinont, Corinne, Sonveaux, Pierre, and Debiève, Frédéric
- Published
- 2023
- Full Text
- View/download PDF
6. Therapy-induced DNA methylation inactivates MCT1 and renders tumor cells vulnerable to MCT4 inhibition
- Author
-
Vander Linden, Catherine, Corbet, Cyril, Bastien, Estelle, Martherus, Ruben, Guilbaud, Céline, Petit, Laurenne, Wauthier, Loris, Loriot, Axelle, De Smet, Charles, and Feron, Olivier
- Published
- 2021
- Full Text
- View/download PDF
7. Two Broad Categories Overlapping With Rheumatoid Arthritis Observed in Synovial Biopsies from Patients With Juvenile Idiopathic Arthritis.
- Author
-
Triaille, Clément, Tilman, Gaëlle, Baert, Charlotte A., Sokolova, Tatiana, Loriot, Axelle, Nzeusseu‐Toukap, Adrien, Meric de Bellefon, Laurent, Galant, Christine, Boulanger, Cécile, Fonseca, Joao E., Bouzin, Caroline, Durez, Patrick, Lauwerys, Bernard R., and Limaye, Nisha
- Subjects
BIOPSY ,JUVENILE idiopathic arthritis ,SYNOVIAL membranes ,MACROPHAGES ,T cells ,RHEUMATOID arthritis ,DESCRIPTIVE statistics ,RNA ,GENE expression ,IMMUNOHISTOCHEMISTRY ,HYPERTROPHY ,GENE expression profiling ,DATA analysis software ,SEQUENCE analysis ,B cells - Abstract
Objective: The objective is to characterize transcriptomic profiles and immune cell composition and distribution in juvenile idiopathic arthritis (JIA) synovial biopsies, assess for associations of these features with clinical parameters, and compare JIA and rheumatoid arthritis (RA) synovial features. Methods: RNA sequencing (RNASeq) was performed on 24 samples, with pathway analysis and inference of relative abundance of immune cell subsets based on gene expression data. Two multiplex fluorescence immunohistochemistry (IHC) panels were performed on 28 samples (including 13 on which RNASeq was performed), staining for CD206− classical and CD206+ nonclassical macrophages, and CD8+ and CD4+ T and B lymphocytes. Data were compared to a published series of early RA synovial biopsies. Results: Pathway analysis of the most variably expressed genes (n = 339) identified a B and plasma cell signature as the main driver of heterogeneity in JIA synovia, with strong overlap between JIA and RA synovitis. Multiplex IHC confirmed heterogeneity of immune cell infiltration. M1‐like macrophage–rich synovial lining was associated with greater lining hypertrophy and higher (CD45+) pan–immune cell and CD8+ T cell infiltration. Conclusion: Our study indicates significant similarities between JIA and RA synovitis. Similar to RA, JIA synovia may be broadly categorized into two groups: (1) those with an inflammatory/adaptive immune transcriptomic signature, M1‐like macrophage and CD8+ T cell infiltration, and thicker, M1‐like macrophage–rich synovial lining, and (2) those with an M2‐like macrophage transcriptomic signature, greater M2/M1‐like macrophage ratios, and thinner, M2‐like macrophage–rich synovial lining. Synovial features were not significantly associated with clinical parameters, likely because of group size and heterogeneity. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. DNA Hypomethylation Underlies Epigenetic Swapping between AGO1 and AGO1-V2 Isoforms in Tumors.
- Author
-
Fain, Jean S., Wangermez, Camille, Loriot, Axelle, Denoue, Claudia, and De Smet, Charles
- Subjects
DNA demethylation ,GENE expression ,RNA interference ,SMALL interfering RNA ,ARGONAUTE proteins - Abstract
Human tumors progress in part by accumulating epigenetic alterations, which include gains and losses of DNA methylation in different parts of the cancer cell genome. Recent work has revealed a link between these two opposite alterations by showing that DNA hypomethylation in tumors can induce the expression of transcripts that overlap downstream gene promoters and thereby induce their hypermethylation. Preliminary in silico evidence prompted us to investigate if this mechanism applies to the locus harboring AGO1, a gene that plays a central role in miRNA biogenesis and RNA interference. Inspection of public RNA-Seq datasets and RT-qPCR experiments show that an alternative transcript starting 13.4 kb upstream of AGO1 (AGO1-V2) is expressed specifically in testicular germ cells, and becomes aberrantly activated in different types of tumors, particularly in tumors of the esophagus, stomach, and lung. This expression pattern classifies AGO1-V2 into the group of "Cancer-Germline" (CG) genes. Analysis of transcriptomic and methylomic datasets provided evidence that transcriptional activation of AGO1-V2 depends on DNA demethylation of its promoter region. Western blot experiments revealed that AGO1-V2 encodes a shortened isoform of AGO1, corresponding to a truncation of 75 aa in the N-terminal domain, and which we therefore referred to as "∆NAGO1". Interestingly, significant correlations between hypomethylation/activation of AGO1-V2 and hypermethylation/repression of AGO1 were observed upon examination of tumor cell lines and tissue datasets. Overall, our study reveals the existence of a process of interdependent epigenetic alterations in the AGO1 locus, which promotes swapping between two AGO1 protein-coding mRNA isoforms in tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Transcriptional overlap links DNA hypomethylation with DNA hypermethylation at adjacent promoters in cancer
- Author
-
Fain, Jean S., Loriot, Axelle, Diacofotaki, Anna, Van Tongelen, Aurélie, and De Smet, Charles
- Published
- 2021
- Full Text
- View/download PDF
10. Dynamics and predicted drug response of a gene network linking dedifferentiation with beta-catenin dysfunction in hepatocellular carcinoma
- Author
-
Gérard, Claude, Di-Luoffo, Mickaël, Gonay, Léolo, Caruso, Stefano, Couchy, Gabrielle, Loriot, Axelle, Castven, Darko, Tao, Junyan, Konobrocka, Katarzyna, Cordi, Sabine, Monga, Satdarshan P., Hanert, Emmanuel, Marquardt, Jens U., Zucman-Rossi, Jessica, and Lemaigre, Frédéric P.
- Published
- 2019
- Full Text
- View/download PDF
11. Oncogenic roles of DNA hypomethylation through the activation of cancer-germline genes
- Author
-
Van Tongelen, Aurélie, Loriot, Axelle, and De Smet, Charles
- Published
- 2017
- Full Text
- View/download PDF
12. Disease activity drives transcriptomic heterogeneity in early untreated rheumatoid synovitis.
- Author
-
Triaille, Clément, Tilman, Gaëlle, Sokolova, Tatiana, Loriot, Axelle, Marchandise, Joelle, De Montjoye, Stéphanie, Nzeusseu-Toukap, Adrien, Méric de Bellefon, Laurent, Bouzin, Caroline, Galant, Christine, Durez, Patrick, Lauwerys, Bernard R., and Limaye, Nisha
- Published
- 2023
- Full Text
- View/download PDF
13. Axon guidance genes control hepatic artery development.
- Author
-
Gannoun, Lila, De Schrevel, Catalina, Belle, Morgane, Dauguet, Nicolas, Achouri, Younes, Loriot, Axelle, Vanderaa, Christophe, Cordi, Sabine, Alexandra Dili, Heremans, Yves, Rooman, Ilse, Leclercq, Isabelle A., Jacquemin, Patrick, Gatto, Laurent, and Lemaigre, Frédéric P.
- Subjects
HEPATIC artery ,VASCULAR smooth muscle ,EPHRIN receptors ,BILE ducts ,LIVER regeneration ,GENE silencing ,HOMEOSTASIS ,AXONS - Abstract
Earlier data on liver development demonstrated that morphogenesis of the bile duct, portal mesenchyme and hepatic artery is interdependent, yet how this interdependency is orchestrated remains unknown. Here, using 2D and 3D imaging, we first describe how portal mesenchymal cells become organised to form hepatic arteries. Next, we examined intercellular signalling active during portal area development and found that axon guidance genes are dynamically expressed in developing bile ducts and portal mesenchyme. Using tissue-specific gene inactivation in mice, we show that the repulsive guidance molecule BMP co-receptor A (RGMA)/neogenin (NEO1) receptor/ligand pair is dispensable for portal area development, but that deficient roundabout 2 (ROBO2)/ SLIT2 signalling in the portal mesenchyme causes reduced maturation of the vascular smooth muscle cells that form the tunica media of the hepatic artery. This arterial anomaly does not impact liver function in homeostatic conditions, but is associated with significant tissular damage following partial hepatectomy. In conclusion, our work identifies new players in development of the liver vasculature in health and liver regeneration. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
14. Through the discovery and characterization of a new form of pancreatitis
- Author
-
Rajput Bhatti, Memoona, Helaers, Raphaël, Loriot, Axelle, Achouri, Younes, Scheers, Isabelle, Jacquemin, P, UCL - SSS/DDUV - Institut de Duve, UCL - SSS/IREC/PEDI - Pôle de Pédiatrie, and UCL - (SLuc) Service de gastro-entérologie et hépatologie pédiatrique
- Abstract
The causes of chronic pancreatitis in children are partly different from those found in adults and remain incompletely understood. Thus, the presence of genetic mutations predisposing to pancreatitis is suspected to be a more frequent cause in children. In this context, among pediatric patients with precursor signs of pancreatitis, we identified a category of patients with symptoms typically associated with ciliopathies, a group of genetic disorders related to primary cilium dysfunction. In this work, we therefore aimed to identify mutations in ciliary genes in these patients, and to establish a link between these mutations and the development of pancreatitis. Via Whole Exome Sequencing of genomic DNA from 43 pediatric patients with idiopathic chronic pancreatitis, we first identified in three of them a mutation in the ciliary/ciliogenic genes PKHD1, HNF1β, and NPHP3. We then focused on the NPHP3 gene and generated by CRISPR/Cas9 technique two new transgenic mouse models; one replicates the NPHP3 gene mutations present in the corresponding patient (NPHP3mut1/mut2 model), while the other is a conditional inactivation of NPHP3 (NPHP3f/f model). Phenotypic analysis of these two models allowed us to conclude that loss of function of NPHP3 in pancreatic ductal cells led to inflammation and mild fibrosis associated with significant acinar atrophy and severe lipomatosis. This suggests a new form of pancreatitis in these murine models, characterized by a ciliopathic origin. In order to confirm the presence of pancreatic lipomatosis in patients with HNF1β and NPHP3 mutations, initial MRI analyses were performed and revealed a significant percentage of adipose tissue within the pancreas of these patients. Deeper analysis of mouse models, in particular to characterize the molecular mechanisms involved in the observed phenotypes, and the recruitment of a larger number of patients and control subjects, should allow us to confirm the existence of this new form of pancreatitis of ciliopathic origin. Lay abstract (English) The causes of chronic pancreatitis in children are partially different than in adults and are still incompletely understood. We found that a subset of pediatric patients with signs of early pancreatitis have clinical symptoms typically associated with ciliopathies. The objectives were to identify mutated ciliary genes in these patients and establish a link between mutations, ciliary phenotype, and pancreatitis development. Using a Whole Exome Sequencing strategy, we analyzed the genomic DNA of 43 children with idiopathic chronic pancreatitis. We generated with the CRISPR/Cas9 technique, two new transgenic mouse models to determine in vivo how ciliary gene mutations cause pancreatitis. We identified 3 patients, each with mutations in a ciliary gene: PKHD1, HNF1β and NPHP3. In both murine models, either the one reproducing the mutations found in one patient or the one with a specific deletion of NPHP3 in pancreatic ductal cells, an atypic pancreatitis characterized by the presence of lipomatosis was observed. In order to confirm the presence of pancreatic lipomatosis in patients suffering from NPHP3 or HNF1β mutations, MRI analysis was performed and revealed an important percentage of adipose tissue inside their pancreas. We validated some steps in our project with the discovery of three ciliary gene mutations in three patients. Analysis of mouse models reproducing these mutations confirms that the renal and pancreatic phenotypes found in a patient are indeed due to mutations of this gene. Lipomatosis development in the pancreas following a ciliary gene defect could be a sign of a new form of pancreatitis.
- Published
- 2022
15. Influence of cell mechanics in embryonic bile duct lumen formation: insight from quantitative modeling
- Author
-
Van Liedekerke, Paul, Gannoun, Lila, Loriot, Axelle, Lemaigre, Frédéric, Drasdo, Dirk, SImulations en Médecine, BIOtechnologie et ToXicologie de systèmes multicellulaires (SIMBIOTX ), Inria Saclay - Ile de France, Institut National de Recherche en Informatique et en Automatique (Inria)-Institut National de Recherche en Informatique et en Automatique (Inria), Modelling and Analysis for Medical and Biological Applications (MAMBA), Inria de Paris, Institut National de Recherche en Informatique et en Automatique (Inria)-Institut National de Recherche en Informatique et en Automatique (Inria)-Laboratoire Jacques-Louis Lions (LJLL (UMR_7598)), Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), Université Catholique de Louvain = Catholic University of Louvain (UCL), Leibniz Research Centre for Working Environment and Human Factors [Dortmund] (IFADO), Technische Universität Dortmund [Dortmund] (TU), and Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP)
- Subjects
[PHYS.PHYS.PHYS-BIO-PH]Physics [physics]/Physics [physics]/Biological Physics [physics.bio-ph] ,[SDV.BDD]Life Sciences [q-bio]/Development Biology - Abstract
In biological and medical literature, alternative hypotheses for initial bile duct lumen formation during embryogenesis exist, which so far remained largely untested. Guided by the quantification of morphological features and expression of genes in developing bile ducts from embryonic mouse liver, these hypotheses were sharpened and data collected that permitted to develop a high resolution individual-based computational model to test the alternative hypotheses in silico. Simulations with this model suggest that successful bile duct lumen formation primarily requires the simultaneous contribution of directed cell division of cholangiocytes, local osmotic effects generated by salt excretion in the lumen, and temporally-controlled differentiation of hepatoblasts to cholangiocytes, with apical constriction of cholangiocytes only moderately affecting luminal size.
- Published
- 2021
16. BRAF V600E Expression in Thyrocytes Causes Recruitment of Immunosuppressive STABILIN-1 Macrophages.
- Author
-
Spourquet, Catherine, Delcorte, Ophélie, Lemoine, Pascale, Dauguet, Nicolas, Loriot, Axelle, Achouri, Younes, Hollmén, Maija, Jalkanen, Sirpa, Huaux, François, Lucas, Sophie, Meerkeeck, Pierre Van, Knauf, Jeffrey A., Fagin, James A., Dessy, Chantal, Mourad, Michel, Henriet, Patrick, Tyteca, Donatienne, Marbaix, Etienne, and Pierreux, Christophe E.
- Subjects
BIOLOGICAL models ,INJECTIONS ,THYROID gland tumors ,PAPILLARY carcinoma ,ONCOGENES ,ANIMAL experimentation ,MACROPHAGES ,IMMUNOSUPPRESSION ,DOXYCYCLINE ,INTRAPERITONEAL injections ,GENE expression ,CELLULAR signal transduction ,TRANSFERASES ,EPITHELIAL cells ,MICE - Abstract
Simple Summary: Incidence of thyroid cancer, including papillary thyroid cancer, is rapidly increasing. Oncogenes, such as the BRAF
V600E , have been identified, and their effect on thyroid cancer cells have been studied in vitro and in mouse models. What is less understood is the impact of these mutations on thyroid cancer microenvironment and, in turn, the effect of changes in the microenvironment on tumor progression. We investigated the modifications in the cellular composition of thyroid cancer microenvironment using an inducible mouse model. We focused on a subpopulation of macrophages, expressing the STABILIN-1 protein, recruited in the thyroid tumor microenvironment following BRAFV600E expression. CRISPR/Cas9 genetic inactivation of Stablin-1 did not change macrophage recruitment but highlighted the immunosuppressive role of STABILIN-1-expressing macrophages. The identification of a similar subpopulation of STABILIN-1 macrophages in human thyroid diseases supports a conserved role for these macrophages and offers an opportunity for intervention. Papillary thyroid carcinoma (PTC) is the most frequent histological subtype of thyroid cancers (TC), and BRAFV600E genetic alteration is found in 60% of this endocrine cancer. This oncogene is associated with poor prognosis, resistance to radioiodine therapy, and tumor progression. Histological follow-up by anatomo-pathologists revealed that two-thirds of surgically-removed thyroids do not present malignant lesions. Thus, continued fundamental research into the molecular mechanisms of TC downstream of BRAFV600E remains central to better understanding the clinical behavior of these tumors. To study PTC, we used a mouse model in which expression of BRAFV600E was specifically switched on in thyrocytes by doxycycline administration. Upon daily intraperitoneal doxycycline injection, thyroid tissue rapidly acquired histological features mimicking human PTC. Transcriptomic analysis revealed major changes in immune signaling pathways upon BRAFV600E induction. Multiplex immunofluorescence confirmed the abundant recruitment of macrophages, among which a population of LYVE-1+/CD206+/STABILIN-1+ was dramatically increased. By genetically inactivating the gene coding for the scavenger receptor STABILIN-1, we showed an increase of CD8+ T cells in this in situ BRAFV600E -dependent TC. Lastly, we demonstrated the presence of CD206+/STABILIN-1+ macrophages in human thyroid pathologies. Altogether, we revealed the recruitment of immunosuppressive STABILIN-1 macrophages in a PTC mouse model and the interest to further study this macrophage subpopulation in human thyroid tissues. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
17. Duodenal CD8+ T resident memory cell apoptosis contributes to gut barrier dysfunction and microbial translocation in early alcohol‐associated liver disease in humans.
- Author
-
Maccioni, Luca, Loriot, Axelle, Dewulf, Joseph, Bommer, Guido, Horsmans, Yves, Lanthier, Nicolas, Leclercq, Isabelle, Schnabl, Bernd, and Stärkel, Peter
- Subjects
- *
LYSOSOMES , *LIPID metabolism , *LIVER histology , *IMMUNOLOGIC memory , *LIVER diseases , *ALCOHOLISM , *NON-alcoholic fatty liver disease , *CD8 antigen - Abstract
Summary: Background: Intestinal T cells are key in gut barrier function. Their role in early stages of alcohol‐associated liver disease (ALD) remain unknown. Aim: To explore the links between intestinal T cells, microbial translocation and ALD Methods: Patients with alcohol use disorder (AUD) following a rehabilitation programme were compared to subjects with non‐alcoholic fatty liver disease (NAFLD) and healthy controls. Clinical and laboratory data (liver stiffness, controlled attenuation parameter, AST, ALT, K18‐M65) served to identify AUD patients with isolated steatosis (minimal liver disease) or steatohepatitis/fibrosis (ALD). Serum microbial translocation markers were measured by ELISA, duodenal and plasma levels of sphingolipids by targeted LC–MS. T lymphocytes in duodenal biopsies were characterised by immunohistochemistry, flow cytometry and RNA sequencing on FACS‐sorted cells. Mechanisms for T‐cell alterations were assessed in vitro. Results: Patients with ALD, but not those with minimal liver disease, showed reduced numbers of duodenal CD8+ T resident memory (TRM) cells compared to controls or patients with NAFLD. TRM transcriptomic analysis, in vitro analyses and pharmacological inhibition of cathepsin B confirmed TRM apoptosis driven by lysosomal membrane permeabilisation and cathepsin B release into the cytosol. Altered lipid metabolism and increased duodenal and plasma sphingolipids correlated with apoptosis. Dihydroceramide dose‐dependently reduced viability of TRM. Duodenal TRM phenotypic changes, apoptosis and transcriptomic alterations correlated with increased levels of microbial translocation markers. Short‐term abstinence did not reverse TRM cell death in patients with ALD. Conclusions: Duodenal CD8+ TRM apoptosis related to functional changes in lysosomes and lipid metabolism points to impaired gut adaptive immunity specifically in patients with AUD who developed early ALD. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
18. Characterisation of a new form of pancreatitis
- Author
-
Fages, Angeline, Bhatti, Memoona Rajput, Helaers, Raphaël, Loriot, Axelle, Achouri, Younes, Fellmann, Marine, Saunier, Sophie, Viau, Amandine, Serafin, Alice, Gillion, Valentine, Godefroid, Nathalie, Jacquemin, Patrick, and Scheers, Isabelle
- Published
- 2023
- Full Text
- View/download PDF
19. Myc represses transcription through recruitment of DNA methyltransferase corepressor
- Author
-
Brenner, Carmen, Deplus, Rachel, Didelot, Céline, Loriot, Axelle, Viré, Emmanuelle, De Smet, Charles, Gutierrez, Arantxa, Danovi, Davide, Bernard, David, Boon, Thierry, Giuseppe Pelicci, Pier, Amati, Bruno, Kouzarides, Tony, de Launoit, Yvan, Di Croce, Luciano, and Fuks, François
- Published
- 2005
- Full Text
- View/download PDF
20. Transient Down-regulation of DNMT1 Methyltransferase Leads to Activation and Stable Hypomethylation of MAGE-A1 in Melanoma Cells
- Author
-
Loriot, Axelle, De Plaen, Etienne, Boon, Thierry, and De Smet, Charles
- Published
- 2006
- Full Text
- View/download PDF
21. BRAF V600E Induction in Thyrocytes Triggers Important Changes in the miRNAs Content and the Populations of Extracellular Vesicles Released in Thyroid Tumor Microenvironment.
- Author
-
Delcorte, Ophélie, Spourquet, Catherine, Lemoine, Pascale, Degosserie, Jonathan, Van Der Smissen, Patrick, Dauguet, Nicolas, Loriot, Axelle, Knauf, Jeffrey A., Gatto, Laurent, Marbaix, Etienne, Fagin, James A., and Pierreux, Christophe E.
- Subjects
EXTRACELLULAR vesicles ,THYROID gland tumors ,TUMOR microenvironment ,THYROID cancer ,NON-coding RNA ,BRAF genes - Abstract
Papillary thyroid cancer (PTC) is the most common endocrine malignancy for which diagnosis and recurrences still challenge clinicians. New perspectives to overcome these issues could come from the study of extracellular vesicle (EV) populations and content. Here, we aimed to elucidate the heterogeneity of EVs circulating in the tumor and the changes in their microRNA content during cancer progression. Using a mouse model expressing BRAF
V600E , we isolated and characterized EVs from thyroid tissue by ultracentrifugations and elucidated their microRNA content by small RNA sequencing. The cellular origin of EVs was investigated by ExoView and that of deregulated EV-microRNA by qPCR on FACS-sorted cell populations. We found that PTC released more EVs bearing epithelial and immune markers, as compared to the healthy thyroid, so that changes in EV-microRNAs abundance were mainly due to their deregulated expression in thyrocytes. Altogether, our work provides a full description of in vivo-derived EVs produced by, and within, normal and cancerous thyroid. We elucidated the global EV-microRNAs signature, the dynamic loading of microRNAs in EVs upon BRAFV600E induction, and their cellular origin. Finally, we propose that thyroid tumor-derived EV-microRNAs could support the establishment of a permissive immune microenvironment. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
22. Expression of BORIS in melanoma: Lack of association with MAGE-A1 activation
- Author
-
Kholmanskikh, Olga, Loriot, Axelle, Brasseur, Francis, De Plaen, Etienne, and De Smet, Charles
- Published
- 2008
- Full Text
- View/download PDF
23. Quantitative modeling identifies critical cell mechanics driving bile duct lumen formation.
- Author
-
Van Liedekerke, Paul, Gannoun, Lila, Loriot, Axelle, Johann, Tim, Lemaigre, Frédéric P., and Drasdo, Dirk
- Subjects
BILE ducts ,CELLULAR mechanics ,TISSUE mechanics ,INTRAHEPATIC bile ducts ,CELL division ,CELL physiology ,GALLBLADDER - Abstract
Biliary ducts collect bile from liver lobules, the smallest functional and anatomical units of liver, and carry it to the gallbladder. Disruptions in this process caused by defective embryonic development, or through ductal reaction in liver disease have a major impact on life quality and survival of patients. A deep understanding of the processes underlying bile duct lumen formation is crucial to identify intervention points to avoid or treat the appearance of defective bile ducts. Several hypotheses have been proposed to characterize the biophysical mechanisms driving initial bile duct lumen formation during embryogenesis. Here, guided by the quantification of morphological features and expression of genes in bile ducts from embryonic mouse liver, we sharpened these hypotheses and collected data to develop a high resolution individual cell-based computational model that enables to test alternative hypotheses in silico. This model permits realistic simulations of tissue and cell mechanics at sub-cellular scale. Our simulations suggest that successful bile duct lumen formation requires a simultaneous contribution of directed cell division of cholangiocytes, local osmotic effects generated by salt excretion in the lumen, and temporally-controlled differentiation of hepatoblasts to cholangiocytes, with apical constriction of cholangiocytes only moderately affecting luminal size. Author summary: The initial step in bile duct development is the formation of a biliary lumen, a process which involves several cellular mechanisms, such as cell division and polarization, and secretion of fluid. However, how these mechanisms are orchestrated in time and space is difficult to understand. Here, we built a computational model of biliary lumen formation which represents every cell and its function in detail. With the model we can simulate the effect of biophysical aspects that affect duct formation. We have tested the individual and combined effects of directed cell division, apical constriction, and osmotic effects on lumen expansion by varying the parameters that control their relative strength. Our simulations suggest that successful bile duct lumen formation requires the simultaneous contribution of directed cell division of cholangiocytes, local osmotic effects generated by salt excretion in the lumen, and temporally-controlled differentiation of hepatoblasts to cholangiocytes, with apical constriction of cholangiocytes only moderately affecting luminal size. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
24. A Mouse Model of Cholangiocarcinoma Uncovers a Role for Tensin‐4 in Tumor Progression.
- Author
-
Di‐Luoffo, Mickaël, Pirenne, Sophie, Saandi, Thoueiba, Loriot, Axelle, Gérard, Claude, Dauguet, Nicolas, Manzano‐Núñez, Fátima, Alves Souza Carvalhais, Natália, Lamoline, Florence, Cordi, Sabine, Konobrocka, Katarzyna, De Greef, Vitaline, Komuta, Mina, Halder, Georg, Jacquemin, Patrick, and Lemaigre, Frédéric P.
- Published
- 2021
- Full Text
- View/download PDF
25. Epigenetic Coactivation of MAGEA6 and CT-GABRA3 Defines Orientation of a Segmental Duplication in the Human X Chromosome
- Author
-
Fain, Jean, Van Tongelen, Aurélie, Loriot, Axelle, De Smet, Charles, and UCL - SSS/DDUV/GEPI - Epigénétique
- Subjects
MAGEA3<%2Fitalic>%22"> ,MAGEA3 MAGEA6<%2Fitalic>%22"> ,MAGEA6 Bidirectional promoter ,Cancer-germline genes ,Genome misassembly ,Segmental duplication - Abstract
The human genome harbors many duplicated segments, which sometimes show very high sequence identity. This may complicate assignment during genome assembly. One such example is in Xq28, where the arrangement of 2 recently duplicated segments varies between genome assembly versions. The duplicated segments comprise highly similar genes, including MAGEA3 and MAGEA6, which display specific expression in testicular germline cells, and also become aberrantly activated in a variety of tumors. Recently, a new gene was identified, CT-GABRA3, the transcription of which initiates inside the segmental duplication but extends far outside. According to the latest genome annotation, CT- GABRA3 starts near MAGEA3, with which it shares a bidirectional promoter. In an earlier annotation, however, the duplicated segment was positioned in the opposite orientation, and CT-GABRA3 was instead coupled with MAGEA6. To resolve this discrepancy, and based on the contention that genes connected by a bidirectional promoter are almost always co-expressed, we decided to compare the expression profiles of CT-GABRA3, MAGEA3, and MAGEA6. We found that in tumor tissues and cell lines of different origins, the expression of CT-GABRA3 was better correlated with that of MAGEA6. Moreover, in a cellular model of experimental induction with a DNA demethylation agent, activation CT-GABRA3 was associated with that of MAGEA6, but not with that of MAGEA3. Together these results support a connection between CT-GABRA3 and MAGEA6 and illustrate how promoter-sharing genes can be exploited to resolve genome assembly uncertainties.
- Published
- 2019
26. Epigenetic Coactivation of MAGEA6 and CT-GABRA3 Defines Orientation of a Segmental Duplication in the Human X Chromosome.
- Author
-
Fain, Jean S., Van Tongelen, Aurélie, Loriot, Axelle, and De Smet, Charles
- Subjects
HUMAN chromosomes ,DNA demethylation ,EPIGENETICS ,HUMAN genome ,X chromosome ,CELL lines - Abstract
The human genome harbors many duplicated segments, which sometimes show very high sequence identity. This may complicate assignment during genome assembly. One such example is in Xq28, where the arrangement of 2 recently duplicated segments varies between genome assembly versions. The duplicated segments comprise highly similar genes, including MAGEA3 and MAGEA6, which display specific expression in testicular germline cells, and also become aberrantly activated in a variety of tumors. Recently, a new gene was identified, CT-GABRA3, the transcription of which initiates inside the segmental duplication but extends far outside. According to the latest genome annotation, CT- GABRA3 starts near MAGEA3, with which it shares a bidirectional promoter. In an earlier annotation, however, the duplicated segment was positioned in the opposite orientation, and CT-GABRA3 was instead coupled with MAGEA6. To resolve this discrepancy, and based on the contention that genes connected by a bidirectional promoter are almost always co-expressed, we decided to compare the expression profiles of CT-GABRA3, MAGEA3, and MAGEA6. We found that in tumor tissues and cell lines of different origins, the expression of CT-GABRA3 was better correlated with that of MAGEA6. Moreover, in a cellular model of experimental induction with a DNA demethylation agent, activation CT-GABRA3 was associated with that of MAGEA6, but not with that of MAGEA3. Together these results support a connection between CT-GABRA3 and MAGEA6 and illustrate how promoter-sharing genes can be exploited to resolve genome assembly uncertainties. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
27. Silencing of cancer-germline genes in human preimplantation embryos: Evidence for active de novo DNA methylation in stem cells
- Author
-
Loriot, Axelle, Parvizi, Grégory K., Reister, Sven, and De Smet, Charles
- Published
- 2012
- Full Text
- View/download PDF
28. A gene expression signature identifying transient DNMT1 depletion as a causal factor of cancer-germline gene activation in melanoma.
- Author
-
Cannuyer, Julie, Van Tongelen, Aurélie, Loriot, Axelle, and De Smet, Charles
- Published
- 2015
- Full Text
- View/download PDF
29. DNA Hypomethylation and Activation of Germline-Specific Genes in Cancer.
- Author
-
De Smet, Charles and Loriot, Axelle
- Published
- 2013
- Full Text
- View/download PDF
30. A novel cancer-germline transcript carrying pro-metastatic miR-105 and TET-targeting miR-767 induced by DNA hypomethylation in tumors.
- Author
-
Loriot, Axelle, Van Tongelen, Aurélie, Blanco, Jordi, Klaessens, Simon, Cannuyer, Julie, van Baren, Nicolas, Decottignies, Anabelle, and De Smet, Charles
- Published
- 2014
- Full Text
- View/download PDF
31. Epigenetic Hierarchy within the MAGEA1 Cancer-Germline Gene: Promoter DNA Methylation Dictates Local Histone Modifications.
- Author
-
Cannuyer, Julie, Loriot, Axelle, Parvizi, Grégory K., and De Smet, Charles
- Subjects
- *
EPIGENETICS , *CANCER genes , *GERM cells , *DNA methylation , *HISTONE acetylation , *GENE expression , *DEACETYLASES - Abstract
Gene MAGEA1 belongs to a group of human germline-specific genes that rely on DNA methylation for repression in somatic tissues. Many of these genes, termed cancer-germline (CG) genes, become demethylated and activated in a wide variety of tumors, where they encode tumor-specific antigens. The process leading to DNA demethylation of CG genes in tumors remains unclear. Previous data suggested that histone acetylation might be involved. Here, we investigated the relative contribution of DNA methylation and histone acetylation in the epigenetic regulation of gene MAGEA1. We show that MAGEA1 DNA hypomethylation in expressing melanoma cells is indeed correlated with local increases in histone H3 acetylation (H3ac). However, when MAGEA1-negative cells were exposed to a histone deacetylase inhibitor (TSA), we observed only short-term activation of the gene and detected no demethylation of its promoter. As a more sensitive assay, we used a cell clone harboring a methylated MAGEA1/hph construct, which confers resistance to hygromycin upon stable re-activation. TSA induced only transient de-repression of the transgene, and did not lead to the emergence of hygromycin-resistant cells. In striking contrast, transient depletion of DNA-methyltransferase-1 in the reporter cell clone gave rise to a hygromycin-resistant population, in which the re-activated MAGEA1/hph transgene displayed not only marked DNA hypomethylation, but also significant reversal of histone marks, including gains in H3ac and H3K4me2, and losses of H3K9me2. Collectively, our results indicate that DNA methylation has a dominant role in the epigenetic hierarchy governing MAGEA1 expression. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
32. Cancer-linked satellite 2 DNA hypomethylation does not regulate Sat2 non-coding RNA expression and is initiated by heat shock pathway activation.
- Author
-
Tilman, Gaëlle, Arnoult, Nausica, Lenglez, Sandrine, Van Beneden, Amandine, Loriot, Axelle, De Smet, Charles, and Decottignies, Anabelle
- Published
- 2012
- Full Text
- View/download PDF
33. Promoter-Dependent Mechanism Leading to Selective Hypomethylation within the 5′ Region of Gene MAGE-A1 in Tumor Cells.
- Author
-
De Smet, Charles, Loriot, Axelle, and Boon, Thierry
- Subjects
- *
CANCER cells , *GERM cells , *METHYLATION , *PROMOTERS (Genetics) , *TUMORS , *DNA - Abstract
Several male germ line-specific genes, including MAGE-A1, rely on DNA methylation for their repression in normal somatic tissues. These genes become activated in many types of tumors in the course of the genomewide demethylation process which often accompanies tumorigenesis. We show that in tumor cells expressing MAGE-A1, the 5' region is significantly less methylated than the other parts of the gene. The process leading to this site-specific hypomethylation does not appear to be permanent in these tumor cells, since in vitromethylated MAGE-A1 sequences do not undergo demethylation after being stably transfected. However, in these cells there is a process that inhibits de novo methylation within the 5' region of MAGE-A1, since unmethylated MAGE-A1 transgenes undergo remethylation at all CpGs except those located within the 5' region. This local inhibition of methylation appears to depend on promoter activity. We conclude that the site-specific hypomethylation of MAGE-A1 in tumor cells relies on a transient process of demethylation followed by a persistent local inhibition of remethylation due to the presence of transcription factors. [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
34. Identification of Tissue-Specific Gene Clusters Induced by DNA Demethylation in Lung Adenocarcinoma: More Than Germline Genes.
- Author
-
Diacofotaki, Anna, Loriot, Axelle, and De Smet, Charles
- Subjects
- *
LUNG cancer & genetics , *ADENOCARCINOMA , *RESEARCH , *DNA demethylation , *GENE expression , *TISSUES , *CELL lines , *TUMOR markers , *STATISTICAL correlation , *EPIGENOMICS - Abstract
Simple Summary: Loss of DNA methylation is often observed in human tumors, but how this epigenetic alteration impacts the transcriptome of cancer cells remains largely undefined. So far, DNA hypomethylation in tumors has been associated with aberrant activation of a germline-specific gene expression program. Here, we exploited transcriptomic and methylomic datasets of lung adenocarcinoma to investigate the possibility that other gene expression programs also become ectopically activated in hypomethylated tumors. Remarkably, we found that DNA hypomethylation in lung adenocarcinoma is associated with ectopic activation of not only germline-specific genes, but also gene clusters displaying specific expression in the gastrointestinal tract, or in stratified epithelia. Interestingly, expression of genes in this latter group was of prognostic value. Together, our study brings novel insight into the transcriptomic changes associated with DNA hypomethylation in tumors, and is an incentive to explore the value of hypomethylated DNA sequences as cancer biomarkers. Genome-wide loss of DNA methylation is commonly observed in human cancers, but its impact on the tumor transcriptome remains ill-defined. Previous studies demonstrated that this epigenetic alteration causes aberrant activation of a germline-specific gene expression program. Here, we examined if DNA hypomethylation in tumors also leads to de-repression of gene clusters with other tissue specificities. To this end, we explored transcriptomic and methylomic datasets from human lung adenocarcinoma (LUAD) cell lines, normal lung, and lung alveolar type II cells, considered as the origin of LUAD. Interestingly, DNA demethylation in LUAD cell lines was associated with activation of not only germline-specific (CG) genes, but also gene clusters displaying specific expression in the gastrointestinal tract (GI), or in stratified epithelia (SE). Consistently, genes from all three clusters showed highly specific patterns of promoter methylation among normal tissues and cell types, and were generally sensitive to induction by a DNA demethylating agent. Analysis of TCGA datasets confirmed that demethylation and activation of CG, GI and SE genes also occurs in vivo in LUAD tumor tissues, in association with global genome hypomethylation. For genes of the GI cluster, we demonstrated that HNF4A is a necessary factor for transcriptional activation following promoter demethylation. Interestingly, expression of several SE genes, in particular FAM83A, correlated with both tumor grade and reduced patient survival. Together, our study uncovers novel cell-type specific gene clusters that become aberrantly activated in LUAD tumors in association with genome-wide hypomethylation. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
35. AG-205 Upregulates Enzymes Involved in Cholesterol Biosynthesis and Steroidogenesis in Human Endometrial Cells Independently of PGRMC1 and Related MAPR Proteins.
- Author
-
Thieffry, Charlotte, Van Wynendaele, Marie, Aynaci, Asena, Maja, Mauriane, Dupuis, Caroline, Loriot, Axelle, Marbaix, Etienne, and Henriet, Patrick
- Subjects
BIOSYNTHESIS ,ENDOMETRIOSIS ,CHOLESTEROL ,PROTEINS ,BREAST cancer ,ENZYMES ,PROGESTERONE receptors - Abstract
An inappropriate response to progestogens in the human endometrium can result in fertility issues and jeopardize progestin-based treatments against pathologies such as endometriosis. PGRMC1 can mediate progesterone response in the breast and ovaries but its endometrial functions remain unknown. AG-205 is an alleged PGRMC1 inhibitor but its specificity was recently questioned. We added AG-205 in the cultures of two endometrial cell lines and performed a transcriptomic comparison. AG-205 significantly increased expression of genes coding enzymes of the cholesterol biosynthetic pathway or of steroidogenesis. However, these observations were not reproduced with cells transfected with siRNA against PGRMC1 or its related proteins (MAPRs). Furthermore, AG-205 retained its ability to increase expression of selected target genes even when expression of PGRMC1 or all MAPRs was concomitantly downregulated, indicating that neither PGRMC1 nor any MAPR is required to mediate AG-205 effect. In conclusion, although AG-205 has attractive effects encouraging its use to develop therapeutic strategies, for instance against breast cancer, our study delivers two important warning messages. First, AG-205 is not specific for PGRMC1 or other MAPRs and its mechanisms of action remain unclear. Second, due to its effects on genes involved in steroidogenesis, its use may increase the risk for endometrial pathologies resulting from imbalanced hormones concentrations. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
36. DNA Methylation-Associated Repression of Cancer-Germline (ïenes in Human Embryonic and Adult Stem Cells.
- Author
-
Loriot, Axelle, Reister, Sven, Parvizi, Gregory K., Lysy, Philippe A., and De Smet, Charles
- Subjects
METHYLATION ,DNA ,STEM cells ,GENES ,TUMORS ,CANCER - Abstract
Cancer-germline (CG) genes are a particular group of germline-specific genes that rely primarily on DNA methylation for repression in somatic tissues. In a wide variety of tumors, the promoter of these genes is demethylated, and their transcription is activated. The mechanism underlying this tumor-specific activation is still unclear. It was recently suggested that CG gene expression may be a hallmark of stem cells, and that expression of these genes in several tumors may reflect the expansion of constitutively expressing cancer stem cells. To clarify this issue, we carefully evaluated the expression of several CG genes in human stem cells of embryonic and adult origin. We found no or very weak expression of CG genes in these cells. Consistently, the promoter of CG genes was highly methylated in these cells. We conclude that CG genes do not qualify as ''stemness'' genes, and propose that their activation in cancers results from a tumor-specific activation process. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
37. Identification of a novel cancer-germline transcript within the miRNA harboring GABRA3 gene : epigenetic alterations of the locus in tumors
- Author
-
Van Tongelen, Aurélie, UCL - SSS/DDUV/GEPI - Epigénétique, UCL - Faculté de pharmacie et des sciences biomédicales, De Smet, Charles, Loriot, Axelle, Lemaigre, Frédéric, Arimodo, Paola, Arnaud, Philippe, Decottignies, Anabelle, Jacquemin, Patrick, and Bommer, Guido
- Subjects
Cancer-germline genes ,DNA methylation ,Hydroxymethylation ,GABRA3 ,Epigenetics ,Hypomethylation ,Crispr/cas9 ,TET ,miRNA ,Cancer - Abstract
It is now well established that alterations in DNA methylation patterns contribute to tumor development. Both gains (DNA hypermethylation) and losses (DNA hypomethylation) of this repressive mark are observed in tumors. Our group demonstrated that DNA hypomethylation in tumors induces transcriptional activation of a defined group of genes, which normally show specific expression in the germline. These genes were grouped under the term "cancer-germline" genes (CG genes). The main goal of the laboratory is to characterize the epigenetic regulation and functional roles of CG genes. Our work focused on the GABRA3 gene locus, where we discovered the existence of two overlapping transcripts: BT-GABRA3, which is expressed specifically in brain and testis; and CT-GABRA3, starting ~250 kb upstream, which is expressed exclusively in testis. CT-GABRA3 (but not BT-GABRA3) exhibits typical features of a CG gene, as it shows promoter hypomethylation and transcriptional activation in various tumors. CT-GABRA3 carries a clustered pair of miRNAs (miR-105 and miR-767), which show concurrent expression in tumors. Interestingly, an independent group identified miR-105 as a crucial promoter of cancer metastasis, due to its ability to weaken vascular endothelial barriers following exosomal secretion. On the other hand, we demonstrated that miR-767 inhibits expression of TET1, a gene with tumor suppressive functions involved in epigenetic processes of DNA demodification. Moreover, our studies revealed that CT-GABRA3 hypomethylation/activation in tumors is correlated with hypermethylation of the downstream BT-GABRA3 promoter. The mechanism underlying this interdependent epigenetic alteration appears to involve deposition of the histone mark H3K36me3 on the entire CT-GABRA3 transcribed region, which includes the BT-GABRA3 promoter. Finally, we observed that in tumor cells where the BT-GABRA3 promoter is hypermethylated, the gene becomes sensitive to DNA demethylation, suggesting a process of epigenetic switch. Together our work revealed the existence of a novel miRNA-producing CG gene with oncogenic potential. It also uncovered a unusual mechanism of epigenetic alteration in tumors, whereby DNA hypomethylation and hypermethylation are linked via a process of transcriptional overlap. (BIFA - Sciences biomédicales et pharmaceutiques) -- UCL, 2017
- Published
- 2017
38. Tumor Microenvironment Modifications Induced by Afatinib in Squamous Cell Carcinoma of the Head and Neck: A Window-of-Opportunity Study (EORTC-90111-24111).
- Author
-
Beyaert SP, Loriot AE, Huyghe ND, Goebbels RM, Mendola A, Govaerts AS, Fortpied C, Baldin P, Licitra LF, Lalami Y, Clement PM, Machiels JH, and Schmitz S
- Abstract
Purpose: The EORTC-90111-24111 phase II window study evaluated afatinib versus no preoperative treatment in patients with primary squamous cell carcinoma of the head and neck (HNSCC). We investigated afatinib-induced tumor and microenvironment modifications by comparing pre- and posttreatment tumor biopsies., Patients and Methods: Thirty treatment-naïve patients with primary HNSCC were randomized. Twenty-five patients received afatinib for 14 days before surgery (40 mg 1×/day) and 5 patients were attributed to the control arm. Biopsies were taken at work-up and during surgery. Good quality RNA samples were used for omics analyses. The control arm was enlarged by samples coming from our previous similar window study., Results: IHC analyses of afatinib-treated tumor biopsies showed a decrease in pEGFR (P ≤ 0.05) and pERK (P ≤ 0.05); and an increase in CD3+ (P ≤ 0.01) and CD8+ (P ≤ 0.01) T-cell infiltration, and in CD3+ (P ≤ 0.05) T-cell density. RNA sequencing analyses of afatinib-treated tumor samples showed upregulation of inflammatory genes and increased expression scores of signatures predictive of response to programmed cell death protein 1 blockade (P ≤ 0.05). In posttreatment biopsies of afatinib-treated patients, two clusters were observed. Cluster 1 showed a higher expression of markers and gene sets implicated in epithelial-to-mesenchymal transition (EMT) and activation of cancer-associated fibroblasts (CAF) compared with cluster 2 and controls., Conclusions: Short-term treatment with afatinib in primary HNSCC induces CD3+ and CD8+ tumor infiltration and, in some patients, EMT and CAF activation. These results open perspectives to overcome resistance mechanisms to anti-HER therapy and to potentiate the activity of immune checkpoint inhibitors., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
39. Senescence and senotherapies in biliary atresia and biliary cirrhosis.
- Author
-
Jannone G, Bonaccorsi Riani E, de Magnée C, Tambucci R, Evraerts J, Ravau J, Baldin P, Bouzin C, Loriot A, Gatto L, Decottignies A, Najimi M, and Sokal EM
- Subjects
- Humans, Rats, Animals, Rats, Wistar, Liver metabolism, Hepatocytes metabolism, Cellular Senescence, Biliary Atresia metabolism, Liver Cirrhosis, Biliary metabolism, Liver Cirrhosis, Biliary pathology
- Abstract
Background: Premature senescence occurs in adult hepatobiliary diseases and worsens the prognosis through deleterious liver remodeling and hepatic dysfunction. Senescence might also arises in biliary atresia (BA), the first cause of pediatric liver transplantation. Since alternatives to transplantation are needed, our aim was to investigate premature senescence in BA and to assess senotherapies in a preclinical model of biliary cirrhosis., Methods: BA liver tissues were prospectively obtained at hepatoportoenterostomy (n=5) and liver transplantation (n=30) and compared to controls (n=10). Senescence was investigated through spatial whole transcriptome analysis, SA-β-gal activity, p16 and p21 expression, γ-H2AX and senescence-associated secretory phenotype (SASP). Human allogenic liver-derived progenitor cells (HALPC) or dasatinib and quercetin (D+Q) were administrated to two-month-old Wistar rats after bile duct ligation (BDL)., Results: Advanced premature senescence was evidenced in BA livers from early stage and continued to progress until liver transplantation. Senescence and SASP were predominant in cholangiocytes, but also present in surrounding hepatocytes. HALPC but not D+Q reduced the early marker of senescence p21 in BDL rats and improved biliary injury (serum γGT and Sox9 expression) and hepatocytes mass loss ( Hnf4a )., Conclusions: BA livers displayed advanced cellular senescence at diagnosis that continued to progress until liver transplantation. HALPC reduced early senescence and improved liver disease in a preclinical model of BA, providing encouraging preliminary results regarding the use of senotherapies in pediatric biliary cirrhosis.
- Published
- 2023
- Full Text
- View/download PDF
40. Dynamic Regulation of Expression of KRAS and Its Effectors Determines the Ability to Initiate Tumorigenesis in Pancreatic Acinar Cells.
- Author
-
Assi M, Achouri Y, Loriot A, Dauguet N, Dahou H, Baldan J, Libert M, Fain JS, Guerra C, Bouwens L, Barbacid M, Lemaigre FP, and Jacquemin P
- Subjects
- Acinar Cells metabolism, Animals, Apoptosis, Biomarkers, Tumor genetics, CRISPR-Cas Systems, Cell Proliferation, Disease Models, Animal, ErbB Receptors genetics, ErbB Receptors metabolism, Female, Humans, Male, Mice, Pancreatic Neoplasms etiology, Pancreatic Neoplasms metabolism, Pancreatitis etiology, Pancreatitis metabolism, Proto-Oncogene Proteins p21(ras) antagonists & inhibitors, Proto-Oncogene Proteins p21(ras) genetics, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Acinar Cells pathology, Biomarkers, Tumor metabolism, Gene Expression Regulation, Neoplastic, Mutation, Pancreatic Neoplasms pathology, Pancreatitis pathology, Proto-Oncogene Proteins p21(ras) metabolism
- Abstract
Pancreatic acinar cells are a cell type of origin for pancreatic cancer that become progressively less sensitive to tumorigenesis induced by oncogenic Kras mutations after birth. This sensitivity is increased when Kras mutations are combined with pancreatitis. Molecular mechanisms underlying these observations are still largely unknown. To identify these mechanisms, we generated the first CRISPR-edited mouse models that enable detection of wild-type and mutant KRAS proteins in vivo . Analysis of these mouse models revealed that more than 75% of adult acinar cells are devoid of detectable KRAS protein. In the 25% of acinar cells expressing KRAS protein, transcriptomic analysis highlighted a slight upregulation of the RAS and MAPK pathways. However, at the protein level, only marginal pancreatic expression of essential KRAS effectors, including C-RAF, was observed. The expression of KRAS and its effectors gradually decreased after birth. The low sensitivity of adult acinar cells to Kras mutations resulted from low expression of KRAS and its effectors and the subsequent lack of activation of RAS/MAPK pathways. Pancreatitis triggered expression of KRAS and its effectors as well as subsequent activation of downstream signaling; this induction required the activity of EGFR. Finally, expression of C-RAF in adult pancreas was required for pancreatic tumorigenesis. In conclusion, our study reveals that control of the expression of KRAS and its effectors regulates the sensitivity of acinar cells to transformation by oncogenic Kras mutations. SIGNIFICANCE: This study generates new mouse models to study regulation of KRAS during pancreatic tumorigenesis and highlights a novel mechanism through which pancreatitis sensitizes acinar cells to Kras mutations., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
41. PAR-TERRA is the main contributor to telomeric repeat-containing RNA transcripts in normal and cancer mouse cells.
- Author
-
Viceconte N, Loriot A, Lona Abreu P, Scheibe M, Fradera Sola A, Butter F, De Smet C, Azzalin CM, Arnoult N, and Decottignies A
- Subjects
- Animals, Aurora Kinases genetics, Aurora Kinases metabolism, Cell Line, Tumor, Chromosomes, Mammalian chemistry, Chromosomes, Mammalian metabolism, Computational Biology methods, Fibroblasts cytology, Fibroblasts metabolism, Gene Regulatory Networks, Granulocyte Precursor Cells cytology, Granulocyte Precursor Cells metabolism, HeLa Cells, Humans, Mice, Monocytes cytology, Monocytes metabolism, Mouse Embryonic Stem Cells cytology, Mouse Embryonic Stem Cells metabolism, Neoplasms metabolism, Neoplasms pathology, Neurons cytology, Neurons metabolism, Nuclear Proteins genetics, Nuclear Proteins metabolism, RNA, Messenger classification, RNA, Messenger metabolism, RNA-Binding Proteins classification, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, RecQ Helicases genetics, RecQ Helicases metabolism, Shelterin Complex, Telomere metabolism, Telomere-Binding Proteins genetics, Telomere-Binding Proteins metabolism, Gene Expression Regulation, Neoplastic, Neoplasms genetics, RNA, Messenger genetics, Telomere chemistry, Transcriptome
- Abstract
Telomeric repeat-containing RNA (TERRA) molecules play important roles at telomeres, from heterochromatin regulation to telomerase activity control. In human cells, TERRA is transcribed from subtelomeric promoters located on most chromosome ends and associates with telomeres. The origin of mouse TERRA molecules is, however, unclear, as transcription from the pseudoautosomal PAR locus was recently suggested to account for the vast majority of TERRA in embryonic stem cells (ESC). Here, we confirm the production of TERRA from both the chromosome 18q telomere and the PAR locus in mouse embryonic fibroblasts, ESC, and various mouse cancer and immortalized cell lines, and we identify two novel sources of TERRA on mouse chromosome 2 and X. Using various approaches, we show that PAR-TERRA molecules account for the majority of TERRA transcripts, displaying an increase of two to four orders of magnitude compared to the telomeric 18q transcript. Finally, we present a SILAC-based pull-down screen revealing a large overlap between TERRA-interacting proteins in human and mouse cells, including PRC2 complex subunits, chromatin remodeling factors, DNA replication proteins, Aurora kinases, shelterin complex subunits, Bloom helicase, Coilin, and paraspeckle proteins. Hence, despite originating from distinct genomic regions, mouse and human TERRA are likely to play similar functions in cells., (© 2021 Viceconte et al.; Published by Cold Spring Harbor Laboratory Press for the RNA Society.)
- Published
- 2021
- Full Text
- View/download PDF
42. Dnmt3a-mediated inhibition of Wnt in cardiac progenitor cells improves differentiation and remote remodeling after infarction.
- Author
-
De Pauw A, Andre E, Sekkali B, Bouzin C, Esfahani H, Barbier N, Loriot A, De Smet C, Vanhoutte L, Moniotte S, Gerber B, di Mauro V, Catalucci D, Feron O, Hilfiker-Kleiner D, and Balligand JL
- Abstract
Adult cardiac progenitor cells (CPCs) display a low capacity to differentiate into cardiomyocytes in injured hearts, strongly limiting the regenerative capacity of the mammalian myocardium. To identify new mechanisms regulating CPC differentiation, we used primary and clonally expanded Sca-1+ CPCs from murine adult hearts in homotypic culture or coculture with cardiomyocytes. Expression kinetics analysis during homotypic culture differentiation showed downregulation of Wnt target genes concomitant with increased expression of the Wnt antagonist, Wnt inhibitory factor 1 (Wif1), which is necessary to stimulate CPC differentiation. We show that the expression of the Wif1 gene is repressed by DNA methylation and regulated by the de novo DNA methyltransferase Dnmt3a. In addition, miR-29a is upregulated early during CPC differentiation and downregulates Dnmt3a expression, thereby decreasing Wif1 gene methylation and increasing the efficiency of differentiation of Sca-1+ CPCs in vitro. Extending these findings in vivo, transient silencing of Dnmt3a in CPCs subsequently injected in the border zone of infarcted mouse hearts improved CPC differentiation in situ and remote cardiac remodeling. In conclusion, miR-29a and Dnmt3a epigenetically regulate CPC differentiation through Wnt inhibition. Remote effects on cardiac remodeling support paracrine signaling beyond the local injection site, with potential therapeutic interest for cardiac repair.
- Published
- 2017
- Full Text
- View/download PDF
43. The microbiota-derived bile acid taurodeoxycholic acid improves hepatic cholesterol levels in mice with cancer cachexia.
- Author
-
Thibaut MM, Roumain M, Piron E, Gillard J, Loriot A, Neyrinck AM, Rodriguez J, Massart I, Thissen JP, Huot JR, Pin F, Bonetto A, Delzenne NM, Muccioli GG, and Bindels LB
- Subjects
- Animals, Mice, Humans, Male, Dysbiosis microbiology, Dysbiosis drug therapy, Neoplasms complications, Neoplasms metabolism, Neoplasms microbiology, Disease Models, Animal, Bacteria classification, Bacteria isolation & purification, Bacteria metabolism, Bacteria genetics, Cachexia metabolism, Cachexia drug therapy, Cachexia microbiology, Cachexia etiology, Gastrointestinal Microbiome drug effects, Bile Acids and Salts metabolism, Cholesterol metabolism, Liver metabolism, Liver drug effects
- Abstract
Alterations in bile acid profile and pathways contribute to hepatic inflammation in cancer cachexia, a syndrome worsening the prognosis of cancer patients. As the gut microbiota impinges on host metabolism through bile acids, the current study aimed to explore the functional contribution of gut microbial dysbiosis to bile acid dysmetabolism and associated disorders in cancer cachexia. Using three mouse models of cancer cachexia (the C26, MC38 and HCT116 models), we evidenced a reduction in the hepatic levels of several secondary bile acids, mainly taurodeoxycholic (TDCA). This reduction in hepatic TDCA occurred before the appearance of cachexia. Longitudinal analysis of the gut microbiota pinpointed an ASV, identified as Xylanibacter rodentium , as a bacterium potentially involved in the reduced production of TDCA. Coherently, stable isotope-based experiments highlighted a robust decrease in the microbial 7α-dehydroxylation (7α-DH) activity with no changes in the bile salt hydrolase (BSH) activity in cachectic mice. This approach also highlighted a reduced microbial 7α-hydroxysteroid dehydrogenase (7α-HSDH) and 12α-hydroxysteroid dehydrogenase (12α-HSDH) activities in these mice. The contribution of the lower production of TDCA to cancer cachexia was explored in vitro and in vivo . In vitro , TDCA prevented myotube atrophy, whereas in vivo hepatic whole transcriptome analysis revealed that TDCA administration to cachectic mice improved the unfolded protein response and cholesterol homeostasis pathways. Coherently, TDCA administration reversed hepatic cholesterol accumulation in these mice. Altogether, this work highlights the contribution of the gut microbiota to bile acid dysmetabolism and the therapeutic interest of the secondary bile acid TDCA for hepatic cholesterol homeostasis in the context of cancer cachexia. Such discovery may prove instrumental in the understanding of other metabolic diseases characterized by microbial dysbiosis. More broadly, our work demonstrates the interest and relevance of microbial activity measurements using stable isotopes, an approach currently underused in the microbiome field.
- Published
- 2025
- Full Text
- View/download PDF
44. DNA hypomethylation and activation of germline-specific genes in cancer.
- Author
-
De Smet C and Loriot A
- Subjects
- Animals, Humans, DNA Methylation, Gene Expression Regulation, Developmental, Gene Expression Regulation, Neoplastic, Germ Cells metabolism, Neoplasms genetics, Neoplasms pathology
- Abstract
DNA methylation, occurring at cytosines in CpG dinucleotides, is a potent mechanism of transcriptional repression. Proper genomic methylation -patterns become profoundly altered in cancer cells: both gains (hypermethylation) and losses (hypomethylation) of methylated sites are observed. Although DNA hypomethylation is detected in a vast majority of human tumors and affects many genomic regions, its role in tumor biology remains elusive. Surprisingly, DNA hypomethylation in cancer was found to cause the aberrant activation of only a limited group of genes. Most of these are normally expressed exclusively in germline cells and were grouped under the term "cancer-germline" (CG) genes. CG genes represent unique examples of genes that rely primarily on DNA methylation for their tissue-specific expression. They are also being exploited to uncover the mechanisms that lead to DNA hypomethylation in tumors. Moreover, as CG genes encode tumor-specific antigens, their activation in cancer highlights a direct link between epigenetic alterations and tumor immunity. As a result, clinical trials combining epigenetic drugs with anti-CG antigen vaccines are being considered.
- Published
- 2013
- Full Text
- View/download PDF
45. DNA hypomethylation in cancer: epigenetic scars of a neoplastic journey.
- Author
-
De Smet C and Loriot A
- Subjects
- Animals, Epigenesis, Genetic, Humans, Neoplasms metabolism, Neoplasms pathology, DNA Methylation, Neoplasms genetics
- Abstract
Cytosine methylation is a heritable modification of DNA in mammalian cells, and has a determinant impact on long-term gene repression and genome stability. Genomic methylation patterns, which remain generally stable in the adult, become profoundly altered in most human tumors. While discrete DNA segments become hypermethylated in cancer cells, many more sequences become hypomethylated. This review discusses our current understanding of the mechanisms that lead to DNA hypomethylation in tumors. Evidence suggests that methylation losses are not random, but rather evolve into mosaic hypomethylation patterns. It is proposed that such hypomethylation patterns result from a historical event of transient DNA demethylation, and that transcriptional regulators contribute to determining which regions escape remethylation and remain therefore unmethylated. Finally, possible stages of tumor development during which the transient DNA demethylation process may take place will be discussed.
- Published
- 2010
- Full Text
- View/download PDF
46. DNA methylation-associated repression of cancer-germline genes in human embryonic and adult stem cells.
- Author
-
Loriot A, Reister S, Parvizi GK, Lysy PA, and De Smet C
- Subjects
- Cell Line, Embryo, Mammalian cytology, Embryo, Mammalian physiology, Humans, Neoplastic Stem Cells physiology, Promoter Regions, Genetic, Reverse Transcriptase Polymerase Chain Reaction, Adult Stem Cells physiology, Antigens, Neoplasm genetics, DNA Methylation, Embryonic Stem Cells physiology, Gene Expression Regulation, Genes, Neoplasm
- Abstract
Cancer-germline (CG) genes are a particular group of germline-specific genes that rely primarily on DNA methylation for repression in somatic tissues. In a wide variety of tumors, the promoter of these genes is demethylated, and their transcription is activated. The mechanism underlying this tumor-specific activation is still unclear. It was recently suggested that CG gene expression may be a hallmark of stem cells, and that expression of these genes in several tumors may reflect the expansion of constitutively expressing cancer stem cells. To clarify this issue, we carefully evaluated the expression of several CG genes in human stem cells of embryonic and adult origin. We found no or very weak expression of CG genes in these cells. Consistently, the promoter of CG genes was highly methylated in these cells. We conclude that CG genes do not qualify as "stemness" genes, and propose that their activation in cancers results from a tumor-specific activation process.
- Published
- 2009
- Full Text
- View/download PDF
47. Mouse embryonic stem cells induce targeted DNA demethylation within human MAGE-A1 transgenes.
- Author
-
Loriot A, Sterpin C, De Backer O, and De Smet C
- Subjects
- Animals, Antigens, Neoplasm, Cell Differentiation, Cell Line, Tumor, Embryonic Stem Cells cytology, Humans, Melanoma-Specific Antigens, Mice, Neoplasm Proteins metabolism, Transfection, DNA Methylation, DNA, Neoplasm metabolism, Embryonic Stem Cells metabolism, Neoplasm Proteins genetics, Transgenes
- Abstract
Human tumor development is often associated with a DNA demethylation process. This results in the activation of germline-specific genes, such as MAGE-A1, which rely on DNA methylation for repression in somatic tissues. Here, we searched to identify a cell line possessing ongoing DNA demethylation activity targeted to MAGE-A1. We first assessed MAGE-A1-expressing human tumor cell lines, by evaluating their ability to induce demethylation of MAGE-A1 transgenes that were methylated in vitro before transfection. All cell lines lacked such activity, suggesting that MAGE-A1 hypomethylation in tumors results from a past demethylation event. We then turned to mouse embryonic stem (mES) cells, which are characterized by a high level of methylation plasticity. Interestingly, in vitro methylated MAGE-A1 transgenes became demethylated after transfection into mES cells. Demethylation was targeted to the 5'-region of MAGE-A1 and was strongly reduced at mutated MAGE-A1 transgenes exhibiting impaired promoter activity. Our results indicate that mES cells induce demethylation of MAGE-A1 and represent therefore a valuable system to study this tumor-related process.
- Published
- 2008
- Full Text
- View/download PDF
48. Five new human cancer-germline genes identified among 12 genes expressed in spermatogonia.
- Author
-
Loriot A, Boon T, and De Smet C
- Subjects
- Animals, Azacitidine pharmacology, DNA Methylation, Decitabine, Humans, Male, Mice, Neoplasms metabolism, Reverse Transcriptase Polymerase Chain Reaction, Transcription, Genetic, Tumor Cells, Cultured, X Chromosome, Azacitidine analogs & derivatives, Gene Expression Regulation, Neoplastic, Neoplasms genetics, Promoter Regions, Genetic, Spermatogonia metabolism
- Abstract
An important class of tumor-specific antigens is encoded by male germline-specific genes, such as MAGE genes, that are activated in many cancers of various histological types as a result of the demethylation of their promoter region. A number of these genes were shown to be expressed exclusively during the spermatogonia stage of spermatogenesis. A recent study reported the isolation of a new set of mouse genes that are expressed in spermatogonia but not in somatic tissues. Here, we tested the tumoral expression of the human orthologs of 12 of these genes. A remarkably high proportion, i.e., 5 of 12 genes, was found to be activated in a significant fraction of tumor samples of various histological types. Expression levels of the 5 genes, namely, NXF2, TAF2Q, FTHL17, TDRD1 and TEX15, were evaluated in normal and tumoral tissues. Except for TEX15, these genes showed sufficiently high expression levels in tumors and low background transcription in normal somatic tissues to qualify them as genes that potentially code for tumor-specific antigens. Like previously described cancer-germline genes, the 5 genes were induced in cells treated with a demethylating agent., (Copyright 2003 Wiley-Liss, Inc.)
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.