258 results on '"Michaela Fontenay"'
Search Results
2. Accelerated DNA replication fork speed due to loss of R-loops in myelodysplastic syndromes with SF3B1 mutation
- Author
-
David Rombaut, Carine Lefèvre, Tony Rached, Sabrina Bondu, Anne Letessier, Raphael M. Mangione, Batoul Farhat, Auriane Lesieur-Pasquier, Daisy Castillo-Guzman, Ismael Boussaid, Chloé Friedrich, Aurore Tourville, Magali De Carvalho, Françoise Levavasseur, Marjorie Leduc, Morgane Le Gall, Sarah Battault, Marie Temple, Alexandre Houy, Didier Bouscary, Lise Willems, Sophie Park, Sophie Raynaud, Thomas Cluzeau, Emmanuelle Clappier, Pierre Fenaux, Lionel Adès, Raphael Margueron, Michel Wassef, Samar Alsafadi, Nicolas Chapuis, Olivier Kosmider, Eric Solary, Angelos Constantinou, Marc-Henri Stern, Nathalie Droin, Benoit Palancade, Benoit Miotto, Frédéric Chédin, and Michaela Fontenay
- Subjects
Science - Abstract
Abstract Myelodysplastic syndromes (MDS) with mutated SF3B1 gene present features including a favourable outcome distinct from MDS with mutations in other splicing factor genes SRSF2 or U2AF1. Molecular bases of these divergences are poorly understood. Here we find that SF3B1-mutated MDS show reduced R-loop formation predominating in gene bodies associated with intron retention reduction, not found in U2AF1- or SRSF2-mutated MDS. Compared to erythroblasts from SRSF2- or U2AF1-mutated patients, SF3B1-mutated erythroblasts exhibit augmented DNA synthesis, accelerated replication forks, and single-stranded DNA exposure upon differentiation. Importantly, histone deacetylase inhibition using vorinostat restores R-loop formation, slows down DNA replication forks and improves SF3B1-mutated erythroblast differentiation. In conclusion, loss of R-loops with associated DNA replication stress represents a hallmark of SF3B1-mutated MDS ineffective erythropoiesis, which could be used as a therapeutic target.
- Published
- 2024
- Full Text
- View/download PDF
3. Enhanced fetal hematopoiesis in response to symptomatic SARS-CoV-2 infection during pregnancy
- Author
-
Mansour Alkobtawi, Qui Trung Ngô, Nicolas Chapuis, Romain H. Fontaine, Mira El Khoury, Matthieu Tihy, Nawa Hachem, Aude Jary, Vincent Calvez, Michaela Fontenay, Vassilis Tsatsaris, Sélim Aractingi, and Bénédicte Oulès
- Subjects
Medicine - Abstract
Abstract Background Pregnant women and their fetuses are particularly susceptible to respiratory pathogens. How they respond to SARS-CoV-2 infection is still under investigation. Methods We studied the transcriptome and phenotype of umbilical cord blood cells in pregnant women infected or not with SARS-CoV-2. Results Here we show that symptomatic maternal COVID-19 is associated with a transcriptional erythroid cell signature as compared with asymptomatic and uninfected mothers. We observe an expansion of fetal hematopoietic multipotent progenitors skewed towards erythroid differentiation that display increased clonogenicity. There was no difference in inflammatory cytokines levels in the cord blood upon maternal SARS-CoV-2 infection. Interestingly, we show an activation of hypoxia pathway in cord blood cells from symptomatic COVID-19 mothers, suggesting that maternal hypoxia may be triggering this fetal stress hematopoiesis. Conclusions Overall, these results show a fetal hematopoietic response to symptomatic COVID-19 in pregnant mothers in the absence of vertically transmitted SARS-CoV-2 infection which is likely to be a mechanism of fetal adaptation to the maternal infection and reduced oxygen supply.
- Published
- 2023
- Full Text
- View/download PDF
4. Immune‐monitoring of myelodysplastic neoplasms: Recommendations from the i4MDS consortium
- Author
-
Cristina A. Tentori, Lin P. Zhao, Benedetta Tinterri, Kathryn E. Strange, Katharina Zoldan, Konstantinos Dimopoulos, Xingmin Feng, Elena Riva, Benjamin Lim, Yannick Simoni, Vidhya Murthy, Madeline J. Hayes, Antonella Poloni, Eric Padron, Bruno A. Cardoso, Michael Cross, Susann Winter, Aida Santaolalla, Bhavisha A. Patel, Emma M. Groarke, Daniel H. Wiseman, Katy Jones, Lauren Jamieson, Charles Manogaran, Naval Daver, Laura Gallur, Wendy Ingram, P. Brent Ferrell, Katja Sockel, Nicolas Dulphy, Nicolas Chapuis, Anne S. Kubasch, Astrid M. Olsnes, Austin Kulasekararaj, Hugues De Lavellade, Wolfgang Kern, Mieke Van Hemelrijck, Dominique Bonnet, Theresia M. Westers, Sylvie Freeman, Uta Oelschlaegel, David Valcarcel, Marco G. Raddi, Kirsten Grønbæk, Michaela Fontenay, Sanam Loghavi, Valeria Santini, Antonio M. Almeida, Jonathan M. Irish, David A. Sallman, Neal S. Young, Arjan A. van deLoosdrecht, Lionel Adès, Matteo G. Della Porta, Catherine Cargo, Uwe Platzbecker, Shahram Kordasti, and i4MDS consortium
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Abstract Advancements in comprehending myelodysplastic neoplasms (MDS) have unfolded significantly in recent years, elucidating a myriad of cellular and molecular underpinnings integral to disease progression. While molecular inclusions into prognostic models have substantively advanced risk stratification, recent revelations have emphasized the pivotal role of immune dysregulation within the bone marrow milieu during MDS evolution. Nonetheless, immunotherapy for MDS has not experienced breakthroughs seen in other malignancies, partly attributable to the absence of an immune classification that could stratify patients toward optimally targeted immunotherapeutic approaches. A pivotal obstacle to establishing “immune classes” among MDS patients is the absence of validated accepted immune panels suitable for routine application in clinical laboratories. In response, we formed International Integrative Innovative Immunology for MDS (i4MDS), a consortium of multidisciplinary experts, and created the following recommendations for standardized methodologies to monitor immune responses in MDS. A central goal of i4MDS is the development of an immune score that could be incorporated into current clinical risk stratification models. This position paper first consolidates current knowledge on MDS immunology. Subsequently, in collaboration with clinical and laboratory specialists, we introduce flow cytometry panels and cytokine assays, meticulously devised for clinical laboratories, aiming to monitor the immune status of MDS patients, evaluating both immune fitness and identifying potential immune “risk factors.” By amalgamating this immunological characterization data and molecular data, we aim to enhance patient stratification, identify predictive markers for treatment responsiveness, and accelerate the development of systems immunology tools and innovative immunotherapies.
- Published
- 2024
- Full Text
- View/download PDF
5. S162: LOSS OF HEMATOPOIETIC PROGENITORS HETEROGENEITY IS AN ADVERSE PROGNOSTIC FACTOR IN MYELODYSPLASTIC SYNDROMES
- Author
-
Charles Dussiau, Thibault Comont, Camille Knosp, Ines Vergnolle, Clotilde Bravetti, Alban Canali, Amandine Houvert, Laetitia Largeaud, Christian Daveaux, Laila Zaroili, Chloe Friedrich, Ismael Boussaid, Loria Zalmai, Carole Almire, Odile Beyne-Rauzy, Lise Willems, Didier Bouscary, Olivier Gandrillon, Michaela Fontenay, Oliver Kosmider, Francois Vergez, and Nicolas Chapuis
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
6. S169: MODULATION OF THE IMMUNE LANDSCAPE IN LOWER-RISK MYELODYSPLASTIC SYNDROMES WITH IMETELSTAT-INDUCED TRANSFUSION INDEPENDENCY
- Author
-
Nicolas Chapuis, David Rombaut, Yannick Simoni, Fei Huang, and Michaela Fontenay
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
7. S163: POLYSOME INTERACTOME IN RPS14-DEFICIENT CELLS REVEALS THE INVOLVEMENT OF THE MRNA DECAY MACHINERY IN TRANSLATION SELECTIVITY.
- Author
-
Zubaidan Tuerdi, Françoise Levavasseur, Laura Falceto-Font, Malaïka Kinyua, Emilie-Fleur Gautier, Isabelle Dusanter Fourt, Michaela Fontenay, Evelyne Lauret, and Ismael Boussaid
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
8. Correction: Metabolomic analyses of COVID-19 patients unravel stage-dependent and prognostic biomarkers
- Author
-
François-Xavier Danlos, Claudia Grajeda-Iglesias, Sylvère Durand, Allan Sauvat, Mathilde Roumier, Delphine Cantin, Emeline Colomba, Julien Rohmer, Fanny Pommeret, Giulia Baciarello, Christophe Willekens, Marc Vasse, Frank Griscelli, Jean-Eudes Fahrner, Anne-Gaëlle Goubet, Agathe Dubuisson, Lisa Derosa, Nitharsshini Nirmalathasan, Delphine Bredel, Séverine Mouraud, Caroline Pradon, Annabelle Stoclin, Flore Rozenberg, Jérôme Duchemin, Georges Jourdi, Syrine Ellouze, Françoise Levavasseur, Laurence Albigès, Jean-Charles Soria, Fabrice Barlesi, Eric Solary, Fabrice André, Frédéric Pène, Félix Ackerman, Luc Mouthon, Laurence Zitvogel, Aurélien Marabelle, Jean-Marie Michot, Michaela Fontenay, and Guido Kroemer
- Subjects
Cytology ,QH573-671 - Published
- 2024
- Full Text
- View/download PDF
9. Hematopoietic differentiation is characterized by a transient peak of entropy at a single-cell level
- Author
-
Charles Dussiau, Agathe Boussaroque, Mathilde Gaillard, Clotilde Bravetti, Laila Zaroili, Camille Knosp, Chloé Friedrich, Philippe Asquier, Lise Willems, Laurent Quint, Didier Bouscary, Michaela Fontenay, Thibault Espinasse, Adriana Plesa, Pierre Sujobert, Olivier Gandrillon, and Olivier Kosmider
- Subjects
Hematopoiesis ,Single-cell RNA-seq ,Cell-to-cell variability ,Entropy ,Myelodysplastic syndromes ,Biology (General) ,QH301-705.5 - Abstract
Abstract Background Mature blood cells arise from hematopoietic stem cells in the bone marrow by a process of differentiation along one of several different lineage trajectories. This is often represented as a series of discrete steps of increasing progenitor cell commitment to a given lineage, but as for differentiation in general, whether the process is instructive or stochastic remains controversial. Here, we examine this question by analyzing single-cell transcriptomic data from human bone marrow cells, assessing cell-to-cell variability along the trajectories of hematopoietic differentiation into four different types of mature blood cells. The instructive model predicts that cells will be following the same sequence of instructions and that there will be minimal variability of gene expression between them throughout the process, while the stochastic model predicts a role for cell-to-cell variability when lineage commitments are being made. Results Applying Shannon entropy to measure cell-to-cell variability among human hematopoietic bone marrow cells at the same stage of differentiation, we observed a transient peak of gene expression variability occurring at characteristic points in all hematopoietic differentiation pathways. Strikingly, the genes whose cell-to-cell variation of expression fluctuated the most over the course of a given differentiation trajectory are pathway-specific genes, whereas genes which showed the greatest variation of mean expression are common to all pathways. Finally, we showed that the level of cell-to-cell variation is increased in the most immature compartment of hematopoiesis in myelodysplastic syndromes. Conclusions These data suggest that human hematopoietic differentiation could be better conceptualized as a dynamical stochastic process with a transient stage of cellular indetermination, and strongly support the stochastic view of differentiation. They also highlight the need to consider the role of stochastic gene expression in complex physiological processes and pathologies such as cancers, paving the way for possible noise-based therapies through epigenetic regulation.
- Published
- 2022
- Full Text
- View/download PDF
10. Pathophysiology of Myelodysplastic Syndromes
- Author
-
Michaela Fontenay, Batoul Farhat, and Ismael Boussaid
- Subjects
ineffective erythropoiesis ,mutations ,deletions ,epigenetics ,splicing ,translation ,Medicine - Abstract
Ineffective hematopoiesis is the major characteristic of early myelodysplastic syndromes. Its pathophysiology relies on a diversity of mechanisms supported by genetic events that develop in aging hematopoietic stem cells. Deletion and mutations trigger epigenetic modifications, and co-transcriptional and post-transcriptional deregulations of gene expression. Epistatic interactions between mutants may aggravate the phenotype. Amplification of minor subclones containing mutations that promote their growth and suppress the others drives the clonal evolution. Aging also participates in reprogramming the immune microenvironment towards an inflammatory state, which precedes the expansion of immunosuppressive cells such as Tregs and myeloid-derived suppressive cells that alters the anti-tumor response of effector cells. Integrating biomarkers of transcription/translation deregulation and immune contexture will help the design of personalized treatments.
- Published
- 2021
- Full Text
- View/download PDF
11. Dynamics of circulating calprotectin accurately predict the outcome of moderate COVID-19 patients
- Author
-
Nicolas Chapuis, Nusaibah Ibrahimi, Thibaut Belmondo, Claire Goulvestre, Anne-Emmanuelle Berger, Alice-Andrée Mariaggi, Muriel Andrieu, Camille Chenevier-Gobeaux, Arnaud Bayle, Lydia Campos, Cherifa Cheurfa, Richard Chocron, Jean-Luc Diehl, Benoît Doumenc, Jérôme Duchemin, Manon Duprat, Fabien François, Nicolas Gendron, Tristant Mirault, Frédéric Pène, Aurélien Philippe, Fanny Pommeret, Olivier Sanchez, David M. Smadja, Tali-Anne Szwebel, Aymeric Silvin, Florent Ginhoux, Ludovic Lacroix, Gérôme Jules-Clément, Sarobidy Rapeteramana, Colette Mavier, Laura Steller, Barbara Perniconi, Fabrice André, Damien Drubay, Michaela Fontenay, Sophie Hüe, Stéphane Paul, and Eric Solary
- Subjects
COVID-19 ,Calprotectin ,S100A8/A9 ,Biomarker ,Serial measurement ,Dynamics ,Medicine ,Medicine (General) ,R5-920 - Abstract
Summary: Background: Severe COVID-19 is associated with a high circulating level of calprotectin, the S100A8/S100A9 alarmin heterodimer. Baseline calprotectin amount measured in peripheral blood at diagnosis correlates with disease severity. The optimal use of this biomarker along COVID-19 course remains to be delineated. Methods: We focused on patients with a WHO-defined moderate COVID-19 requiring hospitalization in a medical ward. We collected plasma and serum from three independent cohorts (N = 626 patients) and measured calprotectin amount at admission. We performed longitudinal measures of calprotectin in 457 of these patients (1461 samples) and used a joint latent class mixture model in which classes were defined by age, body mass index and comorbidities to identify calprotectin trajectories predicting the risk of transfer into an intensive care unit or death. Findings: After adjustment for age, sex, body mass index and comorbidities, the predictive value of baseline calprotectin in patients with moderate COVID19 could be refined by serial monitoring of the biomarker. We discriminated three calprotectin trajectories associated with low, moderate, and high risk of poor outcome, and we designed an algorithm available as online software (https://calpla.gustaveroussy.fr:8443/) to monitor the probability of a poor outcome in individual patients with moderate COVID-19. Interpretation: These results emphasize the clinical interest of serial monitoring of calprotectin amount in the peripheral blood to anticipate the risk of poor outcomes in patients with moderate COVID-19 hospitalized in a standard care unit. Funding: The study received support (research grants) from ThermoFisher immunodiagnostics (France) and Gustave Roussy Foundation.
- Published
- 2022
- Full Text
- View/download PDF
12. Metabolomic analyses of COVID-19 patients unravel stage-dependent and prognostic biomarkers
- Author
-
François-Xavier Danlos, Claudia Grajeda-Iglesias, Sylvère Durand, Allan Sauvat, Mathilde Roumier, Delphine Cantin, Emeline Colomba, Julien Rohmer, Fanny Pommeret, Giulia Baciarello, Christophe Willekens, Marc Vasse, Frank Griscelli, Jean-Eudes Fahrner, Anne-Gaëlle Goubet, Agathe Dubuisson, Lisa Derosa, Nitharsshini Nirmalathasan, Delphine Bredel, Séverine Mouraud, Caroline Pradon, Annabelle Stoclin, Flore Rozenberg, Jérôme Duchemin, Georges Jourdi, Syrine Ellouze, Françoise Levavasseur, Laurence Albigès, Jean-Charles Soria, Fabrice Barlesi, Eric Solary, Fabrice André, Frédéric Pène, Félix Ackerman, Luc Mouthon, Laurence Zitvogel, Aurélien Marabelle, Jean-Marie Michot, Michaela Fontenay, and Guido Kroemer
- Subjects
Cytology ,QH573-671 - Abstract
Abstract The circulating metabolome provides a snapshot of the physiological state of the organism responding to pathogenic challenges. Here we report alterations in the plasma metabolome reflecting the clinical presentation of COVID-19 patients with mild (ambulatory) diseases, moderate disease (radiologically confirmed pneumonitis, hospitalization and oxygen therapy), and critical disease (in intensive care). This analysis revealed major disease- and stage-associated shifts in the metabolome, meaning that at least 77 metabolites including amino acids, lipids, polyamines and sugars, as well as their derivatives, were altered in critical COVID-19 patient’s plasma as compared to mild COVID-19 patients. Among a uniformly moderate cohort of patients who received tocilizumab, only 10 metabolites were different among individuals with a favorable evolution as compared to those who required transfer into the intensive care unit. The elevation of one single metabolite, anthranilic acid, had a poor prognostic value, correlating with the maintenance of high interleukin-10 and -18 levels. Given that products of the kynurenine pathway including anthranilic acid have immunosuppressive properties, we speculate on the therapeutic utility to inhibit the rate-limiting enzymes of this pathway including indoleamine 2,3-dioxygenase and tryptophan 2,3-dioxygenase.
- Published
- 2021
- Full Text
- View/download PDF
13. Venetoclax combination therapy induces deep AML remission with eradication of leukemic stem cells and remodeling of clonal haematopoiesis
- Author
-
Romain Vazquez, Claire Breal, Loria Zalmai, Chloe Friedrich, Carole Almire, Adrien Contejean, Sylvain Barreau, Eric Grignano, Lise Willems, Benedicte Deau-Fischer, Patricia Franchi, Marguerite Vignon, Justine Decroocq, Rudy Birsen, Lauriane Goldwirt, Sophie Kaltenbach, Lucile Couronne, Michaela Fontenay, Olivier Kosmider, Didier Bouscary, and Nicolas Chapuis
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
14. Bone marrow oxidative stress and specific antioxidant signatures in myelodysplastic syndromes
- Author
-
Frederic Picou, Christine Vignon, Christelle Debeissat, Sébastien Lachot, Olivier Kosmider, Nathalie Gallay, Amelie Foucault, Marie-Hélène Estienne, Noémie Ravalet, Marie C. Bene, Jorge Domenech, Emmanuel Gyan, Michaela Fontenay, and Olivier Herault
- Subjects
Specialties of internal medicine ,RC581-951 - Abstract
Abstract: Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal stem cell disorders with an inherent tendency for transformation in secondary acute myeloid leukemia. This study focused on the redox metabolism of bone marrow (BM) cells from 97 patients compared with 25 healthy controls. The level of reactive oxygen species (ROS) was quantified by flow cytometry in BM cell subsets as well as the expression level of 28 transcripts encoding for major enzymes involved in the antioxidant cellular response. Our results highlight increased ROS levels in BM nonlymphoid cells and especially in primitive CD34posCD38low progenitor cells. Moreover, we identified a specific antioxidant signature, dubbed “antioxidogram,” for the different MDS subgroups or secondary acute myeloblastic leukemia (sAML). Our results suggest that progression from MDS toward sAML could be characterized by 3 successive molecular steps: (1) overexpression of enzymes reducing proteic disulfide bonds (MDS with
- Published
- 2019
- Full Text
- View/download PDF
15. Dual pyroptotic biomarkers predict erythroid response in lower-risk non-del(5q) myelodysplastic syndromes treated with lenalidomide and recombinant erythropoietin
- Author
-
Chen Wang, Kathy L. McGraw, Amy F. McLemore, Rami Komrokji, Ashley A. Basiorka, Najla Al Ali, Jeffrey E. Lancet, Eric Padron, Olivier Kosmider, Michaela Fontenay, Pierre Fenaux, Alan F. List, and David A. Sallman
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2021
- Full Text
- View/download PDF
16. A Diagnostic Solution for Lupus Anticoagulant Testing in Patients Taking Direct Oral FXa Inhibitors Using DOAC Filter
- Author
-
Carine Farkh, Syrine Ellouze, Louis Gounelle, Mama Sad Houari, Jérôme Duchemin, Valérie Proulle, Michaela Fontenay, Xavier Delavenne, and Georges Jourdi
- Subjects
rivaroxaban ,lupus anticoaglant ,antiphospholipid antibodies ,direct oral anitcoagulant ,apixaban ,Medicine (General) ,R5-920 - Abstract
Background: Direct oral factor Xa (FXa) inhibitors interfere with lupus anticoagulant (LA) assays challenging antiphospholipid syndrome diagnosis in treated patients. We evaluated a new device, called DOAC Filter, and its usefulness in this setting. It is a single-use filtration cartridge in which FXa inhibitor compounds are trapped by non-covalent binding while plasma is filtered through a solid phase. Patient samples were analyzed before and after filtration: 38 rivaroxaban, 41 apixaban, and 68 none. Anticoagulant plasma concentrations were measured using specific anti-Xa assays and HPLC-MS/MS. LA testing was performed using dilute Russell Viper Venom Time (dRVVT) and Silica Clotting Time (SCT). Baseline median [min–max] concentrations were 64.8 [17.6; 311.4] for rivaroxaban and 92.1 ng/mL [37.1; 390.7] for apixaban (HPLC-MS/MS). They were significantly correlated with anti-Xa assay results (r = 0.98 and r = 0.94, respectively). dRVVT was positive in 92% rivaroxaban and 72% apixaban and SCT in 28 and 41% of samples, respectively. Post-filtration, median % of neutralization was 100% with rivaroxaban and apixaban concentrations of, respectively,
- Published
- 2021
- Full Text
- View/download PDF
17. ImmunoCluster provides a computational framework for the nonspecialist to profile high-dimensional cytometry data
- Author
-
James W Opzoomer, Jessica A Timms, Kevin Blighe, Thanos P Mourikis, Nicolas Chapuis, Richard Bekoe, Sedigeh Kareemaghay, Paola Nocerino, Benedetta Apollonio, Alan G Ramsay, Mahvash Tavassoli, Claire Harrison, Francesca Ciccarelli, Peter Parker, Michaela Fontenay, Paul R Barber, James N Arnold, and Shahram Kordasti
- Subjects
ImmunoCluster ,framework ,immunology ,computational biology ,cytometry ,immune monitoring ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
High-dimensional cytometry is an innovative tool for immune monitoring in health and disease, and it has provided novel insight into the underlying biology as well as biomarkers for a variety of diseases. However, the analysis of large multiparametric datasets usually requires specialist computational knowledge. Here, we describe ImmunoCluster (https://github.com/kordastilab/ImmunoCluster), an R package for immune profiling cellular heterogeneity in high-dimensional liquid and imaging mass cytometry, and flow cytometry data, designed to facilitate computational analysis by a nonspecialist. The analysis framework implemented within ImmunoCluster is readily scalable to millions of cells and provides a variety of visualization and analytical approaches, as well as a rich array of plotting tools that can be tailored to users’ needs. The protocol consists of three core computational stages: (1) data import and quality control; (2) dimensionality reduction and unsupervised clustering; and (3) annotation and differential testing, all contained within an R-based open-source framework.
- Published
- 2021
- Full Text
- View/download PDF
18. APR-246 induces early cell death by ferroptosis in acute myeloid leukemia
- Author
-
Rudy Birsen, Clement Larrue, Justine Decroocq, Natacha Johnson, Nathan Guiraud, Mathilde Gotanegre, Lilia Cantero-Aguilar, Eric Grignano, Tony Huynh, Michaela Fontenay, Olivier Kosmider, Patrick Mayeux, Nicolas Chapuis, Jean Emmanuel Sarry, Jerome Tamburini, and Didier Bouscary
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
APR-246 is a promising new therapeutic agent that targets p53 mutated proteins in myelodysplastic syndromes and in acute myeloid leukemia (AML). APR-246 reactivates the transcriptional activity of p53 mutants by facilitating their binding to DNA target sites. Recent studies in solid cancers have found that APR-246 can also induce p53-independent cell death. In this study, we demonstrate that AML cell death occurring early after APR-246 exposure is suppressed by iron chelators, lipophilic antioxidants and inhibitors of lipid peroxidation, and correlates with the accumulation of markers of lipid peroxidation, thus fulfilling the definition of ferroptosis, a recently described cell death process. The capacity of AML cells to detoxify lipid peroxides by increasing their cystine uptake to maintain major antioxidant molecule glutathione biosynthesis after exposure to APR-246 may be a key determinant of sensitivity to this compound. The association of APR-246 with induction of ferroptosis (either by pharmacological compounds, or genetic inactivation of SLC7A11 or GPX4) had a synergistic effect on the promotion of cell death, both in vivo and ex vivo.
- Published
- 2021
- Full Text
- View/download PDF
19. The epigenetic regulator RINF (CXXC5) maintains SMAD7 expression in human immature erythroid cells and sustains red blood cell expansion
- Author
-
Audrey Astori, Gabriel Matherat, Isabelle Munoz, Emilie-Fleur Gautier, Didier Surdez, Yaël Zermati, Frédérique Verdier, Sakina Zaidi, Vincent Feuillet, Amir Kadi, Evelyne Lauret, Olivier Delattre, Carine Lefèvre, Michaela Fontenay, Evelyne Ségal-Bendirdjian, Isabelle Dusanter-Fourt, Didier Bouscary, Olivier Hermine, Patrick Mayeux, and Frédéric Pendino
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
The gene CXXC5, encoding a retinoid-inducible nuclear factor (RINF), is located within a region at 5q31.2 commonly deleted in myelodysplastic syndrome and adult acute myeloid leukemia. RINF may act as an epigenetic regulator and has been proposed as a tumor suppressor in hematopoietic malignancies. However, functional studies in normal hematopoiesis are lacking, and its mechanism of action is unknown. Here, we evaluated the consequences of RINF silencing on cytokine-induced erythroid differentiation of human primary CD34+ progenitors. We found that RINF is expressed in immature erythroid cells and that RINF-knockdown accelerated erythropoietin-driven maturation, leading to a significant reduction (~45%) in the number of red blood cells, without affecting cell viability. The phenotype induced by RINF-silencing was dependent on tumor growth factor b (TGFb) and mediated by SMAD7, a TGFb-signaling inhibitor. RINF upregulates SMAD7 expression by direct binding to its promoter and we found a close correlation between RINF and SMAD7 mRNA levels both in CD34+ cells isolated from bone marrow of healthy donors and myelodysplastic syndrome patients with del(5q). Importantly, RINF knockdown attenuated SMAD7 expression in primary cells and ectopic SMAD7 expression was sufficient to prevent the RINF knockdown-dependent erythroid phenotype. Finally, RINF silencing affects 5’-hydroxymethylation of human erythroblasts, in agreement with its recently described role as a TET2-anchoring platform in mouse. Collectively, our data bring insight into how the epigenetic factor RINF, as a transcriptional regulator of SMAD7, may fine-tune cell sensitivity to TGFb superfamily cytokines and thus play an important role in both normal and pathological erythropoiesis.
- Published
- 2020
- Full Text
- View/download PDF
20. Integrated analyses of translatome and proteome identify the rules of translation selectivity in RPS14-deficient cells
- Author
-
Ismael Boussaid, Salomé Le Goff, Célia Floquet, Emilie-Fleur Gautier, Anna Raimbault, Pierre-Julien Viailly, Dina Al Dulaimi, Barbara Burroni, Isabelle Dusanter-Fourt, Isabelle Hatin, Patrick Mayeux, Bertrand Cosson, and Michaela Fontenay
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
In ribosomopathies, the Diamond-Blackfan anemia (DBA) or 5q- syndrome, ribosomal protein (RP) genes are affected by mutation or deletion, resulting in bone marrow erythroid hypoplasia. Unbalanced production of ribosomal subunits leading to a limited ribosome cellular content, regulates translation at the expense of the master erythroid transcription factor GATA1. In RPS14-deficient cells mimicking 5q- syndrome erythroid defects, we show that the transcript length, codon bias of the coding sequence (CDS) and 3'UTR structure are the key determinants of translation. In these cells, short transcripts with a structured 3'UTR and high CAI showed a decreased translation efficiency. Quantitative analysis of the whole proteome confirmed that the post-transcriptional changes depended on the transcript characteristics that governed the translation efficiency in conditions of low ribosome availability. In addition, proteins involved in normal erythroid differentiation share most determinants of translation selectivity. Our findings thus indicate that impaired erythroid maturation due to 5q- syndrome may proceed from a translational selectivity at the expense of the erythroid differentiation program and suggest that an interplay between the CDS and UTRs may regulate mRNA translation.
- Published
- 2020
- Full Text
- View/download PDF
21. No impact of cancer and plague-relevant FPR1 polymorphisms on COVID-19
- Author
-
Adriana Petrazzuolo, Julie Le Naour, Erika Vacchelli, Pascale Gaussem, Syrine Ellouze, Georges Jourdi, Eric Solary, Michaela Fontenay, David M. Smadja, and Guido Kroemer
- Subjects
immunogenetics ,pathogen-associated molecular patterns ,sars-cov-2 ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Formyl peptide receptor 1 (FPR1) is a pattern-recognition receptor that detects bacterial as well as endogenous danger-associated molecular patterns to trigger innate immune responses by myeloid cells. A single nucleotide polymorphism, rs867228 (allelic frequency 19–20%), in the gene coding for FPR1 accelerates the manifestation of multiple carcinomas, likely due to reduced anticancer immunosurveillance secondary to a defect in antigen presentation by dendritic cells. Another polymorphism in FPR1, rs5030880 (allelic frequency 12–13%), has been involved in the resistance to plague, correlating with the fact that FPR1 is the receptor for Yersinia pestis. Driven by the reported preclinical effects of FPR1 on lung inflammation and fibrosis, we investigated whether rs867228 or rs5030880 would affect the severity of coronavirus disease-19 (COVID-19). Data obtained on patients from two different hospitals in Paris refute the hypothesis that rs867228 or rs5030880 would affect the severity of COVID-19.
- Published
- 2020
- Full Text
- View/download PDF
22. Immune responses during COVID-19 infection
- Author
-
Cléa Melenotte, Aymeric Silvin, Anne-Gaëlle Goubet, Imran Lahmar, Agathe Dubuisson, Alimuddin Zumla, Didier Raoult, Mansouria Merad, Bertrand Gachot, Clémence Hénon, Eric Solary, Michaela Fontenay, Fabrice André, Markus Maeurer, Giuseppe Ippolito, Mauro Piacentini, Fu-Sheng Wang, Florent Ginhoux, Aurélien Marabelle, Guido Kroemer, Lisa Derosa, and Laurence Zitvogel
- Subjects
covid-19 ,sars-cov-2 ,coronavirus ,immune response ,immunity ,cellular ,humoral ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Over the past 16 years, three coronaviruses (CoVs), severe acute respiratory syndrome CoV (SARS-CoV) in 2002, Middle East respiratory syndrome CoV (MERS-CoV) in 2012 and 2015, and SARS-CoV-2 in 2020, have been causing severe and fatal human epidemics. The unpredictability of coronavirus disease-19 (COVID-19) poses a major burden on health care and economic systems across the world. This is caused by the paucity of in-depth knowledge of the risk factors for severe COVID-19, insufficient diagnostic tools for the detection of SARS-CoV-2, as well as the absence of specific and effective drug treatments. While protective humoral and cellular immune responses are usually mounted against these betacoronaviruses, immune responses to SARS-CoV2 sometimes derail towards inflammatory tissue damage, leading to rapid admissions to intensive care units. The lack of knowledge on mechanisms that tilt the balance between these two opposite outcomes poses major threats to many ongoing clinical trials dealing with immunostimulatory or immunoregulatory therapeutics. This review will discuss innate and cognate immune responses underlying protective or deleterious immune reactions against these pathogenic coronaviruses.
- Published
- 2020
- Full Text
- View/download PDF
23. Prognostic Role of Gene Mutations in Chronic Myelomonocytic Leukemia Patients Treated With Hypomethylating Agents
- Author
-
Matthieu Duchmann, Fevzi F. Yalniz, Alessandro Sanna, David Sallman, Catherine C. Coombs, Aline Renneville, Olivier Kosmider, Thorsten Braun, Uwe Platzbecker, Lise Willems, Lionel Adès, Michaela Fontenay, Raajit Rampal, Eric Padron, Nathalie Droin, Claude Preudhomme, Valeria Santini, Mrinal M. Patnaik, Pierre Fenaux, Eric Solary, and Raphael Itzykson
- Subjects
Medicine ,Medicine (General) ,R5-920 - Abstract
Somatic mutations contribute to the heterogeneous prognosis of chronic myelomonocytic leukemia (CMML). Hypomethylating agents (HMAs) are active in CMML, but analyses of small series failed to identify mutations predicting response or survival. We analyzed a retrospective multi-center cohort of 174 CMML patients treated with a median of 7 cycles of azacitidine (n = 68) or decitabine (n = 106). Sequencing data before treatment initiation were available for all patients, from Sanger (n = 68) or next generation (n = 106) sequencing. Overall response rate (ORR) was 52%, including complete response (CR) in 28 patients (17%). In multivariate analysis, ASXL1 mutations predicted a lower ORR (Odds Ratio [OR] = 0.85, p = 0.037), whereas TET2mut/ASXL1wt genotype predicted a higher CR rate (OR = 1.18, p = 0.011) independently of clinical parameters. With a median follow-up of 36.7 months, overall survival (OS) was 23.0 months. In multivariate analysis, RUNX1mut (Hazard Ratio [HR] = 2.00, p = .011), CBLmut (HR = 1.90, p = 0.03) genotypes and higher WBC (log10(WBC) HR = 2.30, p = .005) independently predicted worse OS while the TET2mut/ASXL1wt predicted better OS (HR = 0.60, p = 0.05). CMML-specific scores CPSS and GFM had limited predictive power. Our results stress the need for robust biomarkers of HMA activity in CMML and for novel treatment strategies in patients with myeloproliferative features and RUNX1 mutations. Keywords: Chronic myelomonocytic leukemia, Hypomethylating agents, Somatic mutations, Prognosis
- Published
- 2018
- Full Text
- View/download PDF
24. B-cell tumor development in Tet2-deficient mice
- Author
-
Enguerran Mouly, Hussein Ghamlouch, Veronique Della-Valle, Laurianne Scourzic, Cyril Quivoron, Damien Roos-Weil, Patrycja Pawlikowska, Véronique Saada, M'Boyba K. Diop, Cécile K. Lopez, Michaela Fontenay, Philippe Dessen, Ivo P. Touw, Thomas Mercher, Said Aoufouchi, and Olivier A. Bernard
- Subjects
Specialties of internal medicine ,RC581-951 - Abstract
Abstract: The TET2 gene encodes an α-ketoglutarate–dependent dioxygenase able to oxidize 5-methylcytosine into 5-hydroxymethylcytosine, which is a step toward active DNA demethylation. TET2 is frequently mutated in myeloid malignancies but also in B- and T-cell malignancies. TET2 somatic mutations are also identified in healthy elderly individuals with clonal hematopoiesis. Tet2-deficient mouse models showed widespread hematological differentiation abnormalities, including myeloid, T-cell, and B-cell malignancies. We show here that, similar to what is observed with constitutive Tet2-deficient mice, B-cell–specific Tet2 knockout leads to abnormalities in the B1-cell subset and a development of B-cell malignancies after long latency. Aging Tet2-deficient mice accumulate clonal CD19+ B220low immunoglobulin M+ B-cell populations with transplantable ability showing similarities to human chronic lymphocytic leukemia, including CD5 expression and sensitivity to ibrutinib-mediated B-cell receptor (BCR) signaling inhibition. Exome sequencing of Tet2−/− malignant B cells reveals C-to-T and G-to-A mutations that lie within single-stranded DNA–specific activation-induced deaminase (AID)/APOBEC (apolipoprotein B messenger RNA editing enzyme, catalytic polypeptide-like) cytidine deaminases targeted motif, as confirmed by the lack of a B-cell tumor in compound Tet2-Aicda–deficient mice. Finally, we show that Tet2 deficiency accelerates and exacerbates T-cell leukemia/lymphoma 1A–induced leukemogenesis. Together, our data establish that Tet2 deficiency predisposes to mature B-cell malignancies, which development might be attributed in part to AID-mediated accumulating mutations and BCR-mediated signaling.
- Published
- 2018
- Full Text
- View/download PDF
25. Dyserythropoiesis evaluated by the RED score and hepcidin:ferritin ratio predicts response to erythropoietin in lower-risk myelodysplastic syndromes
- Author
-
Sophie Park, Olivier Kosmider, Frédéric Maloisel, Bernard Drenou, Nicolas Chapuis, Thibaud Lefebvre, Zoubida Karim, Hervé Puy, Anne Sophie Alary, Sarah Ducamp, Frédérique Verdier, Cécile Bouilloux, Alice Rousseau, Marie-Christine Jacob, Agathe Debliquis, Agnes Charpentier, Emmanuel Gyan, Bruno Anglaret, Cecile Leyronnas, Selim Corm, Borhane Slama, Stephane Cheze, Kamel Laribi, Shanti Amé, Christian Rose, Florence Lachenal, Andrea Toma, Gian Matteo Pica, Martin Carre, Frédéric Garban, Clara Mariette, Jean-Yves Cahn, Mathieu Meunier, Olivier Herault, Pierre Fenaux, Orianne Wagner-Ballon, Valerie Bardet, Francois Dreyfus, and Michaela Fontenay
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Erythropoiesis-stimulating agents are generally the first line of treatment of anemia in patients with lower-risk myelodysplastic syndrome. We prospectively investigated the predictive value of somatic mutations, and biomarkers of ineffective erythropoiesis including the flow cytometry RED score, serum growth-differentiation factor-15, and hepcidin levels. Inclusion criteria were no prior treatment with erythropoiesis-stimulating agents, low- or intermediate-1-risk myelodysplastic syndrome according to the International Prognostic Scoring System, and a hemoglobin level 4 (P=0.05) and a hepcidin:ferritin ratio 2000 pg/mL and a hepcidin:ferritin ratio
- Published
- 2019
- Full Text
- View/download PDF
26. Enhanced Renewal of Erythroid Progenitors in Myelodysplastic Anemia by Peripheral Serotonin
- Author
-
David Sibon, Tereza Coman, Julien Rossignol, Mathilde Lamarque, Olivier Kosmider, Elisa Bayard, Guillemette Fouquet, Rachel Rignault, Selin Topçu, Pierre Bonneau, Florence Bernex, Michael Dussiot, Kathy Deroy, Laetitia Laurent, Jacques Callebert, Jean-Marie Launay, Sophie Georgin-Lavialle, Geneviève Courtois, Luc Maroteaux, Cathy Vaillancourt, Michaela Fontenay, Olivier Hermine, and Francine Côté
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Tryptophan as the precursor of several active compounds, including kynurenine and serotonin, is critical for numerous important metabolic functions. Enhanced tryptophan metabolism toward the kynurenine pathway has been associated with myelodysplastic syndromes (MDSs), which are preleukemic clonal diseases characterized by dysplastic bone marrow and cytopenias. Here, we reveal a fundamental role for tryptophan metabolized along the serotonin pathway in normal erythropoiesis and in the physiopathology of MDS-related anemia. We identify, both in human and murine erythroid progenitors, a functional cell-autonomous serotonergic network with pro-survival and proliferative functions. In vivo studies demonstrate that pharmacological increase of serotonin levels using fluoxetine, a common antidepressant, has the potential to become an important therapeutic strategy in low-risk MDS anemia refractory to erythropoietin. : Sibon et al. identify a cell-autonomous serotonergic network in human and mouse erythroid progenitors. Reduced levels of serotonin lead to decreased proliferation and survival of erythroid progenitors. Increasing serotonin’s concentration through fluoxetine, commonly used to treat depression, could be a valuable therapeutic intervention to correct myelodysplastic-syndrome-related anemia. Keywords: serotonin, Tph1, erythropoiesis, myelodysplastic syndrome, anemia, SSRI
- Published
- 2019
- Full Text
- View/download PDF
27. Does being overweight contribute to longer survival rates in myelodysplastic syndrome?
- Author
-
Eric Solary and Michaela Fontenay
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2018
- Full Text
- View/download PDF
28. Are somatic mutations predictive of response to erythropoiesis stimulating agents in lower risk myelodysplastic syndromes?
- Author
-
Olivier Kosmider, Marie Passet, Valeria Santini, Uwe Platzbecker, Valérie Andrieu, Gina Zini, Odile Beyne-Rauzy, Agnès Guerci, Erico Masala, Enrico Balleari, Ekaterina Bulycheva, François Dreyfus, Pierre Fenaux, Michaela Fontenay, and Sophie Park
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2016
- Full Text
- View/download PDF
29. Tyrosine kinase inhibitors induce down-regulation of c-Kit by targeting the ATP pocket.
- Author
-
Diane D'allard, Julie Gay, Clotilde Descarpentries, Emilie Frisan, Kevin Adam, Frederique Verdier, Célia Floquet, Patrice Dubreuil, Catherine Lacombe, Michaela Fontenay, Patrick Mayeux, and Olivier Kosmider
- Subjects
Medicine ,Science - Abstract
The stem cell factor receptor (SCF) c-Kit plays a pivotal role in regulating cell proliferation and survival in many cell types. In particular, c-Kit is required for early amplification of erythroid progenitors, while it must disappear from cell surface for the cell entering the final steps of maturation in an erythropoietin-dependent manner. We initially observed that imatinib (IM), an inhibitor targeting the tyrosine kinase activity of c-Kit concomitantly down-regulated the expression of c-Kit and accelerated the Epo-driven differentiation of erythroblasts in the absence of SCF. We investigated the mechanism by which IM or related masitinib (MA) induce c-Kit down-regulation in the human UT-7/Epo cell line. We found that the down-regulation of c-Kit in the presence of IM or MA was inhibited by a pre-incubation with methyl-β-cyclodextrin suggesting that c-Kit was internalized in the absence of ligand. By contrast to SCF, the internalization induced by TKI was independent of the E3 ubiquitin ligase c-Cbl. Furthermore, c-Kit was degraded through lysosomal, but not proteasomal pathway. In pulse-chase experiments, IM did not modulate c-Kit synthesis or maturation. Analysis of phosphotyrosine peptides in UT-7/Epo cells treated or not with IM show that IM did not modify overall tyrosine phosphorylation in these cells. Furthermore, we showed that a T670I mutation preventing the full access of IM to the ATP binding pocket, did not allow the internalization process in the presence of IM. Altogether these data show that TKI-induced internalization of c-Kit is linked to a modification of the integrity of ATP binding pocket.
- Published
- 2013
- Full Text
- View/download PDF
30. Leukemic cell xenograft in zebrafish embryo for investigating drug efficacy
- Author
-
Benoist Pruvot, Arnaud Jacquel, Nathalie Droin, Patrick Auberger, Didier Bouscary, Jérome Tamburini, Marc Muller, Michaela Fontenay, Johanna Chluba, and Eric Solary
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Zebrafish were proposed as an alternative to mammalian models to assess the efficacy and toxicity of antileukemic drugs. Due to the limited number of transgenic zebrafish leukemia models, we explored human leukemic cell xenograft in zebrafish embryos. Human leukemic cell lines and blast cells sorted from patients with acute myelogenous leukemia were injected 48 hours post-fertilization and remained in the circulation of zebrafish embryos for several days without affecting their development. Imatinib and oxaphorines did not demonstrate any toxicity on normal zebrafish embryos and decreased the leukemic burden in animals xenografted with sensitive leukemic cell lines. Two other molecules, all-trans retinoic acid and the translation inhibitor 4EGI-1, demonstrated teratogenic effects at concentrations shown to be efficient in vitro, which precluded investigation of their antileukemic activity in such models. Altogether, xenografted leukemic cells in zebrafish embryos are a pharmacologically relevant model for screening non-teratogenic drugs.
- Published
- 2011
- Full Text
- View/download PDF
31. Apoptotic pathways to death in myelodysplastic syndromes
- Author
-
Michaela Fontenay and Emmanuel Gyan
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2008
- Full Text
- View/download PDF
32. Platelet activation and coronavirus disease 2019 mortality: Insights from coagulopathy, antiplatelet therapy and inflammation
- Author
-
Aurélien Philippe, Richard Chocron, Guillaume Bonnet, Nader Yatim, Willy Sutter, Jérôme Hadjadj, Orianne Weizman, Coralie L. Guerin, Tristan Mirault, Charles Fauvel, Caroline Hauw-Berlemont, Charles-Marc Samama, Benjamin Terrier, Benjamin Planquette, Victor Waldmann, Michaela Fontenay, Olivier Sanchez, Jean-Luc Diehl, Pascale Gaussem, Ariel Cohen, Nicolas Gendron, and David M. Smadja
- Subjects
General Medicine ,Cardiology and Cardiovascular Medicine - Published
- 2023
33. Multiparameter flow cytometry in the evaluation of myelodysplasia
- Author
-
Anna Porwit, Marie C. Béné, Carolien Duetz, Sergio Matarraz, Uta Oelschlaegel, Theresia M. Westers, Orianne Wagner‐Ballon, Shahram Kordasti, Peter Valent, Frank Preijers, Canan Alhan, Frauke Bellos, Peter Bettelheim, Kate Burbury, Nicolas Chapuis, Eline Cremers, Matteo G. Della Porta, Alan Dunlop, Lisa Eidenschink‐Brodersen, Patricia Font, Michaela Fontenay, Willemijn Hobo, Robin Ireland, Ulrika Johansson, Michael R. Loken, Kiyoyuki Ogata, Alberto Orfao, Katherina Psarra, Leonie Saft, Dolores Subira, Jeroen te Marvelde, Denise A. Wells, Vincent H. J. van der Velden, Wolfgang Kern, and Arjan A. van de Loosdrecht
- Subjects
All institutes and research themes of the Radboud University Medical Center ,Histology ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,Cell Biology ,Pathology and Forensic Medicine - Abstract
Contains fulltext : 290820.pdf (Publisher’s version ) (Open Access) Multiparameter flow cytometry (MFC) is one of the essential ancillary methods in bone marrow (BM) investigation of patients with cytopenia and suspected myelodysplastic syndrome (MDS). MFC can also be applied in the follow-up of MDS patients undergoing treatment. This document summarizes recommendations from the International/European Leukemia Net Working Group for Flow Cytometry in Myelodysplastic Syndromes (ELN iMDS Flow) on the analytical issues in MFC for the diagnostic work-up of MDS. Recommendations for the analysis of several BM cell subsets such as myeloid precursors, maturing granulocytic and monocytic components and erythropoiesis are given. A core set of 17 markers identified as independently related to a cytomorphologic diagnosis of myelodysplasia is suggested as mandatory for MFC evaluation of BM in a patient with cytopenia. A myeloid precursor cell (CD34(+) CD19(-) ) count >3% should be considered immunophenotypically indicative of myelodysplasia. However, MFC results should always be evaluated as part of an integrated hematopathology work-up. Looking forward, several machine-learning-based analytical tools of interest should be applied in parallel to conventional analytical methods to investigate their usefulness in integrated diagnostics, risk stratification, and potentially even in the evaluation of response to therapy, based on MFC data. In addition, compiling large uniform datasets is desirable, as most of the machine-learning-based methods tend to perform better with larger numbers of investigated samples, especially in such a heterogeneous disease as MDS. 01 januari 2023
- Published
- 2023
34. Data from Epigenetic Control of NF-κB-Dependent FAS Gene Transcription during Progression of Myelodysplastic Syndromes
- Author
-
Michaela Fontenay, Eric Solary, Patrick Mayeux, Olivier Kosmider, Michele Goodhardt, Virginie Mariot, Diane d'Allard, Katy Billot, Blandine Benet, Catherine Humbrecht, and Sandrine Ettou
- Abstract
The death domain containing TNF receptor 6 (CD95/Fas) is a direct target for the NF-κB transcription factor and is repressed in solid tumors such as colon carcinomas. Previously, we reported that the Fas death receptor, while overexpressed in low-risk myelodysplastic syndromes (MDS), becomes undetectable on CD34+ progenitors when the disease progresses to secondary acute myeloid leukemia (AML). This study determined the interplay between NF-κB and Fas during MDS progression. We first observed that Fas was induced by TNF-α in the HL60 cell line. In these cells, p65 (RELA) was associated with the FAS promoter, and inhibition of the NF-κB pathway by an IKKα inhibitor (BAY11-7082) or lentiviral expression of a nondegradable mutant of IκBα (IκSR) blocked Fas expression. In contrast, TNF-α failed to induce Fas expression in the colon carcinoma cell line SW480, due to hypermethylation of the FAS promoter. Azacitidine rescued p65 binding on FAS promoter in vitro, and subsequently Fas expression in SW480 cells. Furthermore, inhibition of the NF-κB pathway decreased the expression of Fas in MDS CD45loCD34+ bone marrow cells. However, despite the nuclear expression of p65, Fas was often low on CD45loCD34+ AML cells. TNF-α failed to stimulate its expression, while azacitidine efficiently rescued p65 binding and Fas reexpression. Overall, these data suggest that DNA methylation at NF-κB sites is responsible for FAS gene silencing. Mol Cancer Res; 11(7); 724–35. ©2013 AACR.
- Published
- 2023
35. Supplementary Tables 1 - 4 from Epigenetic Control of NF-κB-Dependent FAS Gene Transcription during Progression of Myelodysplastic Syndromes
- Author
-
Michaela Fontenay, Eric Solary, Patrick Mayeux, Olivier Kosmider, Michele Goodhardt, Virginie Mariot, Diane d'Allard, Katy Billot, Blandine Benet, Catherine Humbrecht, and Sandrine Ettou
- Abstract
PDF file - 143K, Table S1: Clinical and biological characteristics of 53 patients with MDS/sAML. Table S2: Primers and experimental conditions for FAS and BCL2L1 quantification by RT-qPCR. Table S3: Biotinylated oligonucleotide sequences for in vitro p65 pull-down and primers for DNA methylation assay. Table S4: Primers for chromatin immunoprecipitation at FAS, B2M, RAG1 and PAX6 promoters.
- Published
- 2023
36. Supplementary Procedures, Figures 1-4 from BCL-2 and Mutant NRAS Interact Physically and Functionally in a Mouse Model of Progressive Myelodysplasia
- Author
-
Rose Ann Padua, Christine Chomienne, Irving Weissman, Ghulam J. Mufti, Marika Pla, Michaela Fontenay, Pierre Fenaux, N. Shaun B. Thomas, Azim Mohamedali, Eric Lagasse, Dean Felsher, Niclas Setterblad, Christophe Leboeuf, Annie Soulie, Murielle Reboul, Maria-Elena Noguera, Robert West, Anne Janin, Carole le Pogam, Stephanie Beurlet, Scott Kogan, and Nader Omidvar
- Abstract
Supplementary Procedures, Figures 1-4 from BCL-2 and Mutant NRAS Interact Physically and Functionally in a Mouse Model of Progressive Myelodysplasia
- Published
- 2023
37. Data from BCL-2 and Mutant NRAS Interact Physically and Functionally in a Mouse Model of Progressive Myelodysplasia
- Author
-
Rose Ann Padua, Christine Chomienne, Irving Weissman, Ghulam J. Mufti, Marika Pla, Michaela Fontenay, Pierre Fenaux, N. Shaun B. Thomas, Azim Mohamedali, Eric Lagasse, Dean Felsher, Niclas Setterblad, Christophe Leboeuf, Annie Soulie, Murielle Reboul, Maria-Elena Noguera, Robert West, Anne Janin, Carole le Pogam, Stephanie Beurlet, Scott Kogan, and Nader Omidvar
- Abstract
Myelodysplastic syndromes (MDS) are clonal stem cell hematologic disorders that evolve to acute myeloid leukemia (AML) and thus model multistep leukemogenesis. Activating RAS mutations and overexpression of BCL-2 are prognostic features of MDS/AML transformation. Using NRASD12 and BCL-2, we created two distinct models of MDS and AML, where human (h)BCL-2 is conditionally or constitutively expressed. Our novel transplantable in vivo models show that expression of hBCL-2 in a primitive compartment by mouse mammary tumor virus–long terminal repeat results in a disease resembling human MDS, whereas the myeloid MRP8 promoter induces a disease with characteristics of human AML. Expanded leukemic stem cell (Lin−/Sca-1+/c-Kit+) populations and hBCL-2 in the increased RAS-GTP complex within the expanded Sca-1+ compartment are described in both MDS/AML–like diseases. Furthermore, the oncogenic compartmentalizations provide the proapoptotic versus antiapoptotic mechanisms, by activating extracellular signal-regulated kinase and AKT signaling, in determination of the neoplastic phenotype. When hBCL-2 is switched off with doxycycline in the MDS mice, partial reversal of the phenotype was observed with persistence of bone marrow blasts and tissue infiltration as RAS recruits endogenous mouse (m)BCL-2 to remain active, thus demonstrating the role of the complex in the disease. This represents the first in vivo progression model of MDS/AML dependent on the formation of a BCL-2:RAS-GTP complex. The colocalization of BCL-2 and RAS in the bone marrow of MDS/AML patients offers targeting either oncogene as a therapeutic strategy. [Cancer Res 2007;67(24):11657–67]
- Published
- 2023
38. FOXP1 regulates oxidative stress, SIRT1 expression, and resistance to chemotherapies in acute myeloid leukemia cells
- Author
-
Francoise Levavasseur, Samia Oussous, Tuerdi Zubaidan, Olivier Kosmider, Frédéric Pendino, David Rombaut, Didier Bouscary, Michaela Fontenay, Evelyne Lauret, and Isabelle Dusanter-Fourt
- Subjects
Hematology - Abstract
Transcription factor Forkhead box P1 (FOXP1) belongs to the same protein family as the FOXOs that are well-known regulators of murine hematopoietic stem progenitor cell (HSPC) maintenance by dampening oxidative stress. FOXP1 and FOXOs can play opposite or similar roles depending on cell context; they can cross-regulate each other's expression. In a previous study, we have shown that FOXP1 contributes to normal human HSP and acute myeloid leukemia (AML) cell growth. Here we investigated the role of FOXP1 in HSPCs and AML cell oxidative stress defense in human context. FOXP1 expression level was associated with inferior survival outcome of cytogenetically normal (CN) AML patients. FOXP1 knockdown enhanced superoxide anion levels of human committed CD34+CD38+ but not stem cell-enriched CD34+CD38- HSPCs, and AML cells in vitro. It triggered enhanced NRF2 activity and increased cell oxidative stress. FOXP1 had no impact on FOXO1/3/4 expression in these cells; genetic and pharmacological inhibition of FOXOs did not change superoxide anion levels of human HSPCs and AML cells. Also, FOXP1 antioxidant activity was independent of superoxide dismutase (SOD)1-2 or catalase expression changes. Instead, FOXP1 upregulated expression of the stress sensor SIRT1 by stabilizing SIRT1 protein. FOXP1 loss sensitized AML cells to chemotherapy. Altogether, this study identified FOXP1 as a new safeguard against myeloid progenitor oxidative stress, which works independently of FOXOs but through SIRT1, and contributes to AML chemoresistance. It proposes FOXP1 expression/activity as a promising target to overcome drug-resistance of AML HSPCs.
- Published
- 2023
39. RAS activation induces synthetic lethality of MEK inhibition with mitochondrial oxidative metabolism in acute myeloid leukemia
- Author
-
Justine Decroocq, Rudy Birsen, Camille Montersino, Prasad Chaskar, Jordi Mano, Laury Poulain, Chloe Friedrich, Anne-Sophie Alary, Helene Guermouche, Ambrine Sahal, Guillemette Fouquet, Mathilde Gotanègre, Federico Simonetta, Sarah Mouche, Pierre Gestraud, Auriane Lescure, Elaine Del Nery, Claudie Bosc, Adrien Grenier, Fetta Mazed, Johanna Mondesir, Nicolas Chapuis, Liza Ho, Aicha Boughalem, Marc Lelorc’h, Camille Gobeaux, Michaela Fontenay, Christian Recher, Norbert Vey, Arnaud Guillé, Daniel Birnbaum, Olivier Hermine, Isabelle Radford-Weiss, Petros Tsantoulis, Yves Collette, Rémy Castellano, Jean-Emmanuel Sarry, Eric Pasmant, Didier Bouscary, Olivier Kosmider, Jerome Tamburini, Hôpital Cochin [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Institut Cochin (IC UM3 (UMR 8104 / U1016)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), Centre de Recherche en Cancérologie de Marseille (CRCM), Aix Marseille Université (AMU)-Institut Paoli-Calmettes, Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut Paoli-Calmettes, Fédération nationale des Centres de lutte contre le Cancer (FNCLCC), Université Paris Cité (UPCité), Interactions hôte-greffon-tumeur, ingénierie cellulaire et génique - UFC (UMR INSERM 1098) (RIGHT), Institut National de la Santé et de la Recherche Médicale (INSERM)-Etablissement français du sang [Bourgogne-Franche-Comté] (EFS BFC)-Université de Franche-Comté (UFC), Université Bourgogne Franche-Comté [COMUE] (UBFC)-Université Bourgogne Franche-Comté [COMUE] (UBFC), CHU Saint-Antoine [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Centre de Recherche Saint-Antoine (CRSA), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), Institut National de la Santé et de la Recherche Médicale (INSERM), Imagine - Institut des maladies génétiques (IHU) (Imagine - U1163), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), CHU Necker - Enfants Malades [AP-HP], Centre Hospitalier Sud Francilien, Laboratorio d'Informatica Musicale (LIM), Università degli Studi di Milano = University of Milan (UNIMI), Institut Curie [Paris], Centre de Bioinformatique (CBIO), Mines Paris - PSL (École nationale supérieure des mines de Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL), Translational Research Department, Institut Curie, BioImaging Cell and Tissue Core Facility (PICT-IBiSA), Université de Genève = University of Geneva (UNIGE), Ligue Nationale Contre le Cancer - Paris, Ligue Nationale Contre le Cancer (LNCC), Laboratoire CERBA [Saint Ouen l'Aumône], Département d’Oncologie Médicale [IPC, Marseille], Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Fédération nationale des Centres de lutte contre le Cancer (FNCLCC), Université Paris Descartes - Paris 5 (UPD5), Université Sorbonne Paris Cité (USPC), Cancer Research and Personalized Medicine - CARPEM [Paris], Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Hôpital Cochin [AP-HP], and Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Necker - Enfants Malades [AP-HP]
- Subjects
Mitogen-Activated Protein Kinase Kinases ,Cancer Research ,[SDV]Life Sciences [q-bio] ,Hematology ,Proto-Oncogene Proteins p21(ras) ,Leukemia, Myeloid, Acute ,Mice ,Oxidative Stress ,fms-Like Tyrosine Kinase 3 ,Oncology ,hemic and lymphatic diseases ,Mutation ,Animals ,Humans ,Synthetic Lethal Mutations ,neoplasms - Abstract
Despite recent advances in acute myeloid leukemia (AML) molecular characterization and targeted therapies, a majority of AML cases still lack therapeutically actionable targets. In 127 AML cases with unmet therapeutic needs, as defined by the exclusion of ELN favorable cases and of FLT3-ITD mutations, we identified 51 (40%) cases with alterations in RAS pathway genes (RAS+, mostly NF1, NRAS, KRAS, and PTPN11 genes). In 79 homogeneously treated AML patients from this cohort, RAS+ status were associated with higher white blood cell count, higher LDH, and reduced survival. In AML models of oncogenic addiction to RAS-MEK signaling, the MEK inhibitor trametinib demonstrated antileukemic activity in vitro and in vivo. However, the efficacy of trametinib was heterogeneous in ex vivo cultures of primary RAS+ AML patient specimens. From repurposing drug screens in RAS-activated AML cells, we identified pyrvinium pamoate, an anti-helminthic agent efficiently inhibiting the growth of RAS+ primary AML cells ex vivo, preferentially in trametinib-resistant PTPN11- or KRAS-mutated samples. Metabolic and genetic complementarity between trametinib and pyrvinium pamoate translated into anti-AML synergy in vitro. Moreover, this combination inhibited the propagation of RA+ AML cells in vivo in mice, indicating a potential for future clinical development of this strategy in AML.
- Published
- 2022
40. Flow cytometric analysis of myelodysplasia: Pre-analytical and technical issues—Recommendations from the European LeukemiaNet
- Author
-
Vincent H. J. van der Velden, Frank Preijers, Ulrika Johansson, Theresia M. Westers, Alan Dunlop, Anna Porwit, Marie C. Béné, Peter Valent, Jeroen te Marvelde, Orianne Wagner‐Ballon, Uta Oelschlaegel, Leonie Saft, Sharham Kordasti, Robin Ireland, Eline Cremers, Canan Alhan, Carolien Duetz, Willemijn Hobo, Nicolas Chapuis, Michaela Fontenay, Peter Bettelheim, Lisa Eidenshink‐Brodersen, Patricia Font, Michael R. Loken, Sergio Matarraz, Kiyoyuki Ogata, Alberto Orfao, Katherina Psarra, Dolores Subirá, Denise A. Wells, Matteo G. Della Porta, Kate Burbury, Frauke Bellos, Elisabeth Weiß, Wolfgang Kern, Arjan van de Loosdrecht, Hematology laboratory, Internal medicine, Hematology, VU University medical center, AII - Cancer immunology, CCA - Cancer biology and immunology, Erasmus University Medical Center [Rotterdam] (Erasmus MC), Radboudumc Nijmegen [The Netherlands], University Hospitals Bristol, Amsterdam UMC - Amsterdam University Medical Center, Royal Marsden Hospital [Surrey, UK], Lund University [Lund], Immunobiology of Human αβ and γδ T Cells and Immunotherapeutic Applications (CRCINA-ÉQUIPE 1), Centre de Recherche en Cancérologie et Immunologie Nantes-Angers (CRCINA), Université d'Angers (UA)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Université d'Angers (UA)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre hospitalier universitaire de Nantes (CHU Nantes), Centre hospitalier universitaire de Nantes (CHU Nantes), Medizinische Universität Wien = Medical University of Vienna, Institut Mondor de Recherche Biomédicale (IMRB), Institut National de la Santé et de la Recherche Médicale (INSERM)-IFR10-Université Paris-Est Créteil Val-de-Marne - Paris 12 (UPEC UP12), CHU Henri Mondor, University Hospital Carl Gustav Carus [Dresden, Germany], Technische Universität Dresden = Dresden University of Technology (TU Dresden), Karolinska Institutet [Stockholm], King‘s College London, Maastricht University [Maastricht], Hôpital Cochin [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Institut Cochin (IC UM3 (UMR 8104 / U1016)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), Ordensklinikum Linz Elisabethinen, HematoLogics, Inc. [Seattle, WA, USA], Hospital General Universitario 'Gregorio Marañón' [Madrid], Universidad de Salamanca, Instituto de Salud Carlos III [Madrid] (ISC), Metropolitan Research and Treatment Centre for Blood Disorders [Tokyo, Japan] (MRTC Japan), Evangelismos Athens General Hospital, Universidad de Guadalajara, Humanitas University [Milan] (Hunimed), University of Melbourne, Munich Leukemia Laboratory [Munich, Germany], MUMC+: MA Med Staf Artsass Interne Geneeskunde (9), RS: FHML non-thematic output, Immunology, and Bernardo, Elizabeth
- Subjects
Histology ,MASTOCYTOSIS ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,BONE-MARROW ,DIAGNOSTIC-CRITERIA ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,REFRACTORY CYTOPENIA ,CLASSIFICATION ,Pathology and Forensic Medicine ,pre-analytic issues ,03 medical and health sciences ,0302 clinical medicine ,All institutes and research themes of the Radboud University Medical Center ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,SDG 3 - Good Health and Well-being ,hemic and lymphatic diseases ,MDS ,030304 developmental biology ,0303 health sciences ,flow cytometry ,Cell Biology ,QUANTIFICATION ,LOW-GRADE ,3. Good health ,CONSENSUS STATEMENTS ,ELN ,030215 immunology ,STANDARDS - Abstract
Contains fulltext : 290818.pdf (Publisher’s version ) (Open Access) BACKGROUND: Flow cytometry (FCM) aids the diagnosis and prognostic stratification of patients with suspected or confirmed myelodysplastic syndrome (MDS). Over the past few years, significant progress has been made in the FCM field concerning technical issues (including software and hardware) and pre-analytical procedures. METHODS: Recommendations are made based on the data and expert discussions generated from 13 yearly meetings of the European LeukemiaNet international MDS Flow working group. RESULTS: We report here on the experiences and recommendations concerning (1) the optimal methods of sample processing and handling, (2) antibody panels and fluorochromes, and (3) current hardware technologies. CONCLUSIONS: These recommendations will support and facilitate the appropriate application of FCM assays in the diagnostic workup of MDS patients. Further standardization and harmonization will be required to integrate FCM in MDS diagnostic evaluations in daily practice. 01 januari 2023
- Published
- 2023
41. Clinical application of flow cytometry in patients with unexplained cytopenia and suspected myelodysplastic syndrome: A report of the European <scp>LeukemiaNet</scp> International <scp>MDS‐Flow</scp> Cytometry Working Group
- Author
-
Arjan A. van de Loosdrecht, Wolfgang Kern, Anna Porwit, Peter Valent, Sharham Kordasti, Eline Cremers, Canan Alhan, Carolien Duetz, Alan Dunlop, Willemijn Hobo, Frank Preijers, Orianne Wagner‐Ballon, Nicolas Chapuis, Michaela Fontenay, Peter Bettelheim, Lisa Eidenschink‐Brodersen, Patricia Font, Ulrika Johansson, Michael R. Loken, Jeroen G. te Marvelde, Sergio Matarraz, Kiyoyuki Ogata, Uta Oelschlaegel, Alberto Orfao, Katherina Psarra, Dolores Subirá, Denise A. Wells, Marie C. Béné, Matteo G. Della Porta, Kate Burbury, Frauke Bellos, Vincent H. J. van der Velden, Theresia M. Westers, Leonie Saft, and Robin Ireland
- Subjects
Histology ,Cell Biology ,Pathology and Forensic Medicine - Published
- 2021
42. COVID-19 is a systemic vascular hemopathy
- Author
-
Maximilian Ackermann, Gerald B. Pier, Coert Margadant, Coralie L. Guerin, David M. Smadja, Olivier Sanchez, Nicolas Gendron, Michael Laffan, Elisabeth J. M. Huijbers, Jean-Luc Diehl, Patrycja Nowak-Sliwinska, Steven J. Mentzer, Stéphanie Pons, Anna M. Randi, Arjan W. Griffioen, Michaela Fontenay, Danny Jonigk, Christian Karagiannidis, Philipp Kümpers, Julie Helms, Aurélien Philippe, Richard Chocron, David Skurnik, and Nicolas Chapuis
- Subjects
Vascular Endothelial Growth Factor A ,Cancer Research ,2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,Physiology ,Angiogenesis ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Interleukin-1beta ,Clinical Biochemistry ,MEDLINE ,Bioinformatics ,Fibrin Fibrinogen Degradation Products ,von Willebrand Factor ,Humans ,Medicine ,Myelopoiesis ,Respiratory Distress Syndrome ,Review Paper ,Neovascularization, Pathologic ,Interleukin-6 ,SARS-CoV-2 ,business.industry ,COVID-19 ,Endothelial Cells ,Membrane Proteins ,Thrombosis ,Fibroblast Growth Factor 2 ,business - Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is presenting as a systemic disease associated with vascular inflammation and endothelial injury. Severe forms of SARS-CoV-2 infection induce acute respiratory distress syndrome (ARDS) and there is still an ongoing debate on whether COVID-19 ARDS and its perfusion defect differs from ARDS induced by other causes. Beside pro-inflammatory cytokines (such as interleukin-1 β [IL-1β] or IL-6), several main pathological phenomena have been seen because of endothelial cell (EC) dysfunction: hypercoagulation reflected by fibrin degradation products called D-dimers, micro- and macrothrombosis and pathological angiogenesis. Direct endothelial infection by SARS-CoV-2 is not likely to occur and ACE-2 expression by EC is a matter of debate. Indeed, endothelial damage reported in severely ill patients with COVID-19 could be more likely secondary to infection of neighboring cells and/or a consequence of inflammation. Endotheliopathy could give rise to hypercoagulation by alteration in the levels of different factors such as von Willebrand factor. Other than thrombotic events, pathological angiogenesis is among the recent findings. Overexpression of different proangiogenic factors such as vascular endothelial growth factor (VEGF), basic fibroblast growth factor (FGF-2) or placental growth factors (PlGF) have been found in plasma or lung biopsies of COVID-19 patients. Finally, SARS-CoV-2 infection induces an emergency myelopoiesis associated to deregulated immunity and mobilization of endothelial progenitor cells, leading to features of acquired hematological malignancies or cardiovascular disease, which are discussed in this review. Altogether, this review will try to elucidate the pathophysiology of thrombotic complications, pathological angiogenesis and EC dysfunction, allowing better insight in new targets and antithrombotic protocols to better address vascular system dysfunction. Since treating SARS-CoV-2 infection and its potential long-term effects involves targeting the vascular compartment and/or mobilization of immature immune cells, we propose to define COVID-19 and its complications as a systemic vascular acquired hemopathy.
- Published
- 2021
43. Lupus Anticoagulant Single Positivity During the Acute Phase of COVID‐19 Is Not Associated With Venous Thromboembolism or In‐Hospital Mortality
- Author
-
Jeremy Boussier, Christophe Peronino, Olivier Sanchez, Benjamin Planquette, Lina Khider, Charles-Marc Samama, Frédéric Pène, Daphné Krzisch, Elise Sourdeau, Claire Goulvestre, David M. Smadja, Cherifa Cheurfa, Françoise Levasseur, Luc Darnige, Franck Pages, Tristan Mirault, Laetitia Mauge, Camille Chenevier-Gobeaux, Nicolas Gendron, Michaela Fontenay, Benjamin Debuc, Guillaume Goudot, Aurélien Philippe, Richard Chocron, Jérôme Hadjadj, Benjamin Terrier, Nadège Ochat, Jean-Luc Diehl, Tali-Anne Szwebel, Julie Brichet, Marie-Agnès Dragon-Durey, Nader Yatim, Jérôme Duchemin, Georges Jourdi, and Pascale Gaussem
- Subjects
0301 basic medicine ,Lupus anticoagulant ,Univariate analysis ,medicine.medical_specialty ,business.industry ,Immunology ,Hazard ratio ,medicine.disease ,Thrombosis ,Gastroenterology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Rheumatology ,030220 oncology & carcinogenesis ,Internal medicine ,Cohort ,Coagulopathy ,Immunology and Allergy ,Medicine ,Clinical significance ,business ,Survival analysis - Abstract
INTRODUCTION: Antiphospholipid antibodies (APA) clinical relevance in COVID-19 is controversial. We aimed to investigate the prevalence and prognostic value of conventional and non-conventional APA in COVID-19 patients. METHODS: This study was a multi-centric, prospective observational French cohort of patients hospitalized for COVID-19 suspicion. RESULTS: 249 patients were hospitalized for suspected COVID-19, including 154 with confirmed COVID-19 and 95 not confirmed. We found a significant increase in lupus anticoagulant (LA) positivity among COVID-19 positive patients (60.9% versus 23.7% in non-COVID19 patients, p
- Published
- 2021
44. Pathophysiology of Myelodysplastic Syndromes
- Author
-
Batoul Farhat, Ismael Boussaid, and Michaela Fontenay
- Subjects
Ineffective Hematopoiesis ,ineffective erythropoiesis ,epigenetics ,Effector ,deletions ,translation ,Biology ,mutations ,Somatic evolution in cancer ,splicing ,Haematopoiesis ,Immune system ,Cancer research ,Medicine ,Epigenetics ,Stem cell ,Reprogramming - Abstract
Ineffective hematopoiesis is the major characteristic of early myelodysplastic syndromes. Its pathophysiology relies on a diversity of mechanisms supported by genetic events that develop in aging hematopoietic stem cells. Deletion and mutations trigger epigenetic modifications, and co-transcriptional and post-transcriptional deregulations of gene expression. Epistatic interactions between mutants may aggravate the phenotype. Amplification of minor subclones containing mutations that promote their growth and suppress the others drives the clonal evolution. Aging also participates in reprogramming the immune microenvironment towards an inflammatory state, which precedes the expansion of immunosuppressive cells such as Tregs and myeloid-derived suppressive cells that alters the anti-tumor response of effector cells. Integrating biomarkers of transcription/translation deregulation and immune contexture will help the design of personalized treatments.
- Published
- 2021
45. Placental growth factor level in plasma predicts COVID‐19 severity and in‐hospital mortality
- Author
-
Michaela Fontenay, Cherifa Cheurfa, Bastien Rance, Olivier Bory, Jérôme Duchemin, Caroline Hauw-Berlemont, Elise Sourdeau, Richard Chocron, Maxime Gruest, Tristan Mirault, Agathe Beauvais, Nicolas Peron, Pascale Gaussem, Aurélien Philippe, Coralie L. Guerin, Guillaume Goudot, Jean Luc Diehl, Olivier Sanchez, Bertrand Hermann, Nicolas Gendron, Lina Khider, Françoise Levavasseur, Tali Anne Szwebel, Frédéric Pène, Charles Marc Samama, David M. Smadja, Benjamin Planquette, and Benjamin Terrier
- Subjects
Adult ,Vascular Endothelial Growth Factor A ,Placental growth factor ,medicine.medical_specialty ,placental growth factor ,VASCULAR BIOLOGY ,030204 cardiovascular system & hematology ,Gastroenterology ,angiogenesis ,03 medical and health sciences ,0302 clinical medicine ,COVID‐19 ,Internal medicine ,medicine ,Humans ,Hospital Mortality ,Survival analysis ,Placenta Growth Factor ,SARS-CoV-2 ,Proportional hazards model ,business.industry ,Brief Report ,COVID-19 ,Hematology ,Odds ratio ,mortality ,Confidence interval ,Vascular endothelial growth factor A ,FGF‐2 ,PlGF ,Biomarker (medicine) ,Female ,business ,Body mass index ,Biomarkers - Abstract
Background Coronavirus disease 2019 (COVID‐19) is a respiratory disease associated with vascular inflammation and endothelial injury. Objectives To correlate circulating angiogenic markers vascular endothelial growth factor A (VEGF‐A), placental growth factor (PlGF), and fibroblast growth factor 2 (FGF‐2) to in‐hospital mortality in COVID‐19 adult patients. Methods Consecutive ambulatory and hospitalized patients with COVID‐19 infection were enrolled. VEGF‐A, PlGF, and FGF‐2 were measured in each patient ≤48 h following admission. Results The study enrolled 237 patients with suspected COVID‐19: 208 patients had a positive diagnostic for COVID‐19, of whom 23 were mild outpatients and 185 patients hospitalized after admission. Levels of VEGF‐A, PlGF, and FGF‐2 significantly increase with the severity of the disease (P
- Published
- 2021
46. Dihydroartemisinin-induced ferroptosis in acute myeloid leukemia: links to iron metabolism and metallothionein
- Author
-
Eric Grignano, Lilia Cantero-Aguilar, Zubaidan Tuerdi, Thella Chabane, Romain Vazquez, Natacha Johnson, Rudy Birsen, Michaela Fontenay, Olivier Kosmider, Nicolas Chapuis, and Didier Bouscary
- Abstract
Artemisinin is an anti-malarial drug that has shown anticancer properties. Recently, ferroptosis was reported to be induced by dihydroartemisinin and linked to iron increase. In the current study, we determined the effect of artemisinin in leukemic cell lines on ferroptosis induction and iron metabolism and the cytoprotective effect triggered in leukemic cells. We found that treatment of artemisinin induces early ferroptosis by promoting ferritinophagy and subsequent iron increase. Furthermore, our study demonstrated that artemisinin activated zinc metabolism signaling, especially the upregulation of metallothionein. By inhibiting MT2A and MT1M isoforms, we showed that cytotoxic effect of artemisinin and ferroptosis induction were enhanced. Finally, we demonstrated that ferroptosis inducers effect acting on glutathione pool were highly dependent on MTs-driven antioxidant response. Taken together, our study indicated that DHA activates ferritinophagy and subsequent ferroptosis in AML and that MTs are involved in glutathione regenerating and antioxidant response.
- Published
- 2022
47. Molecular International Prognostic Scoring System for Myelodysplastic Syndromes
- Author
-
Elsa Bernard, Heinz Tuechler, Peter L. Greenberg, Robert P. Hasserjian, Juan E. Arango Ossa, Yasuhito Nannya, Sean M. Devlin, Maria Creignou, Philippe Pinel, Lily Monnier, Gunes Gundem, Juan S. Medina-Martinez, Dylan Domenico, Martin Jädersten, Ulrich Germing, Guillermo Sanz, Arjan A. van de Loosdrecht, Olivier Kosmider, Matilde Y. Follo, Felicitas Thol, Lurdes Zamora, Ronald F. Pinheiro, Andrea Pellagatti, Harold K. Elias, Detlef Haase, Christina Ganster, Lionel Ades, Magnus Tobiasson, Laura Palomo, Matteo Giovanni Della Porta, Akifumi Takaori-Kondo, Takayuki Ishikawa, Shigeru Chiba, Senji Kasahara, Yasushi Miyazaki, Agnes Viale, Kety Huberman, Pierre Fenaux, Monika Belickova, Michael R. Savona, Virginia M. Klimek, Fabio P. S. Santos, Jacqueline Boultwood, Ioannis Kotsianidis, Valeria Santini, Francesc Solé, Uwe Platzbecker, Michael Heuser, Peter Valent, Kazuma Ohyashiki, Carlo Finelli, Maria Teresa Voso, Lee-Yung Shih, Michaela Fontenay, Joop H. Jansen, José Cervera, Norbert Gattermann, Benjamin L. Ebert, Rafael Bejar, Luca Malcovati, Mario Cazzola, Seishi Ogawa, Eva Hellström-Lindberg, and Elli Papaemmanuil
- Published
- 2022
48. Abstract 6168: Implementation and adoption of a web tool to support precision diagnostic and treatment decisions for patient with myelodysplastic syndromes
- Author
-
Elsa Bernard, Juan E. Arango Ossa, Heinz Tuechler, Peter L. Greenberg, Robert P. Hasserjian, Yasuhito Nannya, Sean M. Devlin, Maria Creignou, Philippe Pinel, Lily Monier, Juan S. Medina-Martinez, Dylan Domenico, Martin Jädersten, Ulrich Germing, Guillermo Sanz, Arjan A. van de Loosdrecht, Olivier Kosmider, Matilde Y. Follo, Felicitas Thol, Lurdes Zamora, Ronald F. Pinheiro, Andrea Pellagatti, Detlef Haase, Pierre Fenaux, Monika Belickova, Michael R. Savona, Virginia M. Klimek, Fabio P. Santos, Jacqueline Boultwood, Ioannis Kotsianidis, Valeria Santini, Francesc Solé, Uwe Platzbecker, Michael Heuser, Peter Valent, Kazuma Ohyashiki, Carlo Finelli, Maria Teresa Voso, Lee-Yung Shih, Michaela Fontenay, Joop H. Jansen, José Cervera, Norbert Gattermann, Benjamin L. Ebert, Rafael Bejar, Luca Malcovati, Mario Cazzola, Seishi Ogawa, Eva Hellström-Lindberg, and Elli Papaemmanuil
- Subjects
Cancer Research ,Oncology - Abstract
Despite a detailed understanding of the genes mutated in myelodysplastic syndromes (MDS), diagnostic and treatment decisions for patients with MDS rely primarily on clinical and cytogenetic variables as considered by the Revised International Prognostic Scoring System (IPSS-R). Here we describe the recently developed Molecular IPSS (IPSS-M), a clinico-genomic risk stratification system that considers clinical, cytogenetic and genetic parameters; the implementation of a web portal to facilitate its adoption, a strategy to handle missing variables, and the worldwide utilization of the web calculator as a clinical support tool. The IPSS-M was trained on 2,957 clinically annotated diagnostic MDS samples profiled for mutations in 156 driver genes. To maximize the clinical applicability of the IPSS-M and account for missing genetic data (i.e genes missing from a sequencing panel), we implemented a strategy to calculate a risk score under three scenarios: best, worst and average. Last, we developed an online calculator as a standalone single-page web application using VueJs, and D3Js for the interactive visualizations, deployed through a CI/CD pipeline on AWS, where collection of anonymous usage analytics allows to track adoption and usability of the new proposed model. The model incorporates clinical, morphological, genetic variables informed by cytogenetics and constructed from the presence of oncogenic mutations in 31 genes. It delivers a unique risk score for each individual patient, as well as an assignment to one of six IPSS-M risk strata. Compared to the IPSS-R the IPSS-M re-stratified 46% of MDS patients. The model was validated in an external dataset of 754 MDS patients. We released an open-access IPSS-M web calculator available at https://mds-risk-model.com. By specifying the patient clinical and molecular profiles, the tool returns the patient-specific IPSS-M risk score and category, and the probability estimates over time for three clinical endpoints, i.e. leukemia free survival (LFS), overall survival, and incidence of leukemic transformation. Since its launch in June 2022, the calculator has been used by >6000 users in >75 countries, reaching a daily average of 100 users per day. Risks have been calculated for >45,000 patient profiles. 99.28% of the sessions initiated reach an IPSS-M score, suggesting that the calculator is intuitive and easy to use. We trained and validated the IPSS-M on 3,711 patients, a patient tailored risk stratification tool for patients with MDS that considers clinical, morphological and genetic variables inclusive of cytogenetics and mutations in one of 31 genes. The development of a web based tool was instrumental to the global dissemination of the model, enabling non-expert users to leverage the power of molecular biomarkers in risk stratification for patients with MDS. Citation Format: Elsa Bernard, Juan E. Arango Ossa, Heinz Tuechler, Peter L. Greenberg, Robert P. Hasserjian, Yasuhito Nannya, Sean M. Devlin, Maria Creignou, Philippe Pinel, Lily Monier, Juan S. Medina-Martinez, Dylan Domenico, Martin Jädersten, Ulrich Germing, Guillermo Sanz, Arjan A. van de Loosdrecht, Olivier Kosmider, Matilde Y. Follo, Felicitas Thol, Lurdes Zamora, Ronald F. Pinheiro, Andrea Pellagatti, Detlef Haase, Pierre Fenaux, Monika Belickova, Michael R. Savona, Virginia M. Klimek, Fabio P. Santos, Jacqueline Boultwood, Ioannis Kotsianidis, Valeria Santini, Francesc Solé, Uwe Platzbecker, Michael Heuser, Peter Valent, Kazuma Ohyashiki, Carlo Finelli, Maria Teresa Voso, Lee-Yung Shih, Michaela Fontenay, Joop H. Jansen, José Cervera, Norbert Gattermann, Benjamin L. Ebert, Rafael Bejar, Luca Malcovati, Mario Cazzola, Seishi Ogawa, Eva Hellström-Lindberg, Elli Papaemmanuil. Implementation and adoption of a web tool to support precision diagnostic and treatment decisions for patient with myelodysplastic syndromes [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6168.
- Published
- 2023
49. p53 activation during ribosome biogenesis regulates normal erythroid differentiation
- Author
-
François Morlé, Stéphane Giraudier, Eric Soler, Naomi Taylor, Charlotte Andrieu-Soler, Olivier Hermine, Patrick Mayeux, Célia Floquet, Rose Ann Padua, Frédérique Verdier, Sarah Ducamp, Michaela Fontenay, Mohammad Salma, Elisabeth M. Cramer-Borde, Ismael Boussaid, Anna Raimbault, Isabelle Hatin, Diane d'Allard, Amandine Houvert, Narla Mohandas, Boris Guyot, Emilie-Fleur Gautier, Sandrina Kinet, Marjorie Leduc, Pierre-Emmanuel Gleizes, Jean-Jacques Diaz, Salomé Le Goff, François Guillonneau, Nathalie Montel-Lehry, Centre de Recherche en Cancérologie de Lyon (UNICANCER/CRCL), Centre Léon Bérard [Lyon]-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut Cochin (IC UM3 (UMR 8104 / U1016)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), Institut de Biologie Intégrative de la Cellule (I2BC), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS), Institut de Génétique Moléculaire de Montpellier (IGMM), Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM), Plateforme protéomique 3P5 [Institut Cochin] (3P5), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Génomique, Structure et Traduction (GST), Département Biologie des Génomes (DBG), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie Intégrative de la Cellule (I2BC), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS), Laboratoire d'Excellence : Biogenèse et pathologies du globule rouge (Labex Gr-Ex), Université Sorbonne Paris Cité (USPC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), Centre de Biologie Intégrative (CBI), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Centre National de la Recherche Scientifique (CNRS), Hématopoïèse normale et pathologique : émergence, environnement et recherche translationnelle [Paris] ((UMR_S1131 / U1131)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP), Institut NeuroMyoGène (INMG), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM), Imagine - Institut des maladies génétiques (IHU) (Imagine - U1163), New York Blood Center, Hôpital Cochin [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), Institut de génétique et microbiologie [Orsay] (IGM), Université Paris-Sud - Paris 11 (UP11)-Centre National de la Recherche Scientifique (CNRS), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS), Procédés Alimentaires et Microbiologiques (PAM), Université de Bourgogne (UB)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement, ANR-18-IDEX-0001,Université de Paris,Université de Paris(2018), Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), and Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
0301 basic medicine ,[SDV]Life Sciences [q-bio] ,Immunology ,Ribosome biogenesis ,Biology ,Biochemistry ,Ribosome ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Erythroid Cells ,Downregulation and upregulation ,Transcription (biology) ,Ribosomal protein ,hemic and lymphatic diseases ,Animals ,Humans ,Erythropoiesis ,Gene ,ComputingMilieux_MISCELLANEOUS ,Organelle Biogenesis ,RNA ,Cell Differentiation ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,Cell biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Organelle biogenesis ,Tumor Suppressor Protein p53 ,Ribosomes - Abstract
The role of ribosome biogenesis in erythroid development is supported by the recognition of erythroid defects in ribosomopathies in both Diamond-Blackfan anemia and 5q− syndrome. Whether ribosome biogenesis exerts a regulatory function on normal erythroid development is still unknown. In the present study, a detailed characterization of ribosome biogenesis dynamics during human and murine erythropoiesis showed that ribosome biogenesis is abruptly interrupted by the decline in ribosomal DNA transcription and the collapse of ribosomal protein neosynthesis. Its premature arrest by the RNA Pol I inhibitor CX-5461 targeted the proliferation of immature erythroblasts. p53 was activated spontaneously or in response to CX-5461, concomitant to ribosome biogenesis arrest, and drove a transcriptional program in which genes involved in cell cycle–arrested, negative regulation of apoptosis, and DNA damage response were upregulated. RNA Pol I transcriptional stress resulted in nucleolar disruption and activation of the ATR-CHK1-p53 pathway. Our results imply that the timing of ribosome biogenesis extinction and p53 activation is crucial for erythroid development. In ribosomopathies in which ribosome availability is altered by unbalanced production of ribosomal proteins, the threshold downregulation of ribosome biogenesis could be prematurely reached and, together with pathological p53 activation, prevents a normal expansion of erythroid progenitors.
- Published
- 2021
50. DNA Replication Stress Due to Loss of R-Loops in Myelodysplastic Syndromes with SF3B1 Mutation
- Author
-
David Rombaut, Carine Lefevre, Batoul Farhat, Sabrina Bondu, Anne Letessier, Auriane Lesieur-Pasquier, Daisy Castillo-Guzman, Marjorie Leduc, Emilie-Fleur Gautier, Virginie Chesnais, Alice Rousseau, Ismael Boussaid, Sarah Battault, Alexandre Houy, Didier Bouscary, Lise Willems, Nicolas Chapuis, Sophie Park, Sophie Raynaud, Thomas Cluzeau, Emmanuelle Clappier, Pierre Fenaux, Lionel Ades, Eric Solary, Raphael Margueron, Michel Wassef, Olivier Kosmider, Samar Alsafadi, Nathalie Droin, Angelos Constantinou, Marc-Henri Stern, Benoit Miotto, Frederic Chedin, and Michaela Fontenay
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.