100 results on '"Franck Rapaport"'
Search Results
2. Integrative analysis identifies an older female-linked AML patient group with better risk in ECOG-ACRIN Cancer Research Group’s clinical trial E3999
- Author
-
Franck Rapaport, Kenneth Seier, Yaseswini Neelamraju, Duane Hassane, Timour Baslan, Daniel T. Gildea, Samuel Haddox, Tak Lee, H. Moses Murdock, Caroline Sheridan, Alexis Thurmond, Ling Wang, Martin Carroll, Larry D. Cripe, Hugo Fernandez, Christopher E. Mason, Elisabeth Paietta, Gail J. Roboz, Zhuoxin Sun, Martin S. Tallman, Yanming Zhang, Mithat Gönen, Ross Levine, Ari M. Melnick, Maria Kleppe, and Francine E. Garrett-Bakelman
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2022
- Full Text
- View/download PDF
3. Mutational correlates of response to hypomethylating agent therapy in acute myeloid leukemia
- Author
-
Catherine C. Coombs, David A. Sallman, Sean M. Devlin, Shweta Dixit, Abhinita Mohanty, Kristina Knapp, Najla H. Al Ali, Jeffrey E. Lancet, Alan F. List, Rami S. Komrokji, Eric Padron, Maria E. Arcila, Virginia M. Klimek, Marcel R. M. van den Brink, Martin S. Tallman, Ross L. Levine, Raajit K. Rampal, and Franck Rapaport
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2016
- Full Text
- View/download PDF
4. Genetic analysis of five children with essential thrombocytosis identified mutations in cancer-associated genes with roles in transcriptional regulation
- Author
-
Nicole Kucine, Aaron D. Viny, Raajit Rampal, Michael Berger, Nicholas Socci, Agnes Viale, James B. Bussel, Ross L. Levine, and Franck Rapaport
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2016
- Full Text
- View/download PDF
5. Determining frequent patterns of copy number alterations in cancer.
- Author
-
Franck Rapaport and Christina Leslie
- Subjects
Medicine ,Science - Abstract
Cancer progression is often driven by an accumulation of genetic changes but also accompanied by increasing genomic instability. These processes lead to a complicated landscape of copy number alterations (CNAs) within individual tumors and great diversity across tumor samples. High resolution array-based comparative genomic hybridization (aCGH) is being used to profile CNAs of ever larger tumor collections, and better computational methods for processing these data sets and identifying potential driver CNAs are needed. Typical studies of aCGH data sets take a pipeline approach, starting with segmentation of profiles, calls of gains and losses, and finally determination of frequent CNAs across samples. A drawback of pipelines is that choices at each step may produce different results, and biases are propagated forward. We present a mathematically robust new method that exploits probe-level correlations in aCGH data to discover subsets of samples that display common CNAs. Our algorithm is related to recent work on maximum-margin clustering. It does not require pre-segmentation of the data and also provides grouping of recurrent CNAs into clusters. We tested our approach on a large cohort of glioblastoma aCGH samples from The Cancer Genome Atlas and recovered almost all CNAs reported in the initial study. We also found additional significant CNAs missed by the original analysis but supported by earlier studies, and we identified significant correlations between CNAs.
- Published
- 2010
- Full Text
- View/download PDF
6. PopViz: a webserver for visualizing minor allele frequencies and damage prediction scores of human genetic variations.
- Author
-
Peng Zhang 0033, Benedetta Bigio, Franck Rapaport, Shen-Ying Zhang, Jean-Laurent Casanova, Laurent Abel, Bertrand Boisson, and Yuval Itan
- Published
- 2018
- Full Text
- View/download PDF
7. Data from Recurrent Mutations in Cyclin D3 Confer Clinical Resistance to FLT3 Inhibitors in Acute Myeloid Leukemia
- Author
-
Neil P. Shah, Ross L. Levine, Barry S. Taylor, Gideon Bollag, Brian West, Mai H. Le, Henry H. Hsu, Theodore C. Tarver, Elli Papaemmanuil, Juan S. Medina-Martinez, Matthieu Najm, Franck Rapaport, Nicholas D. Socci, Cyriac Kandoth, Evan Massi, Aaron D. Viny, and Catherine C. Smith
- Abstract
Purpose:Biomarkers of response and resistance to FLT3 tyrosine kinase inhibitors (TKI) are still emerging, and optimal clinical combinations remain unclear. The purpose of this study is to identify co-occurring mutations that influence clinical response to the novel FLT3 inhibitor pexidartinib (PLX3397).Experimental Design:We performed targeted sequencing of pretreatment blasts from 29 patients with FLT3 internal tandem duplication (ITD) mutations treated on the phase I/II trial of pexidartinib in relapsed/refractory FLT3-ITD+ acute myeloid leukemia (AML). We sequenced 37 samples from 29 patients with available material, including 8 responders and 21 non-responders treated at or above the recommended phase II dose of 3,000 mg.Results:Consistent with other studies, we identified mutations in NRAS, TP53, IDH2, and a variety of epigenetic and transcriptional regulators only in non-responders. Among the most frequently mutated genes in non-responders was Cyclin D3 (CCND3). A total of 3 individual mutations in CCND3 (Q276*, S264R, and T283A) were identified in 2 of 21 non-responders (one patient had both Q276* and S264R). No CCND3 mutations were found in pexidartinib responders. Expression of the Q276* and T283A mutations in FLT3-ITD MV4;11 cells conferred resistance to apoptosis, decreased cell-cycle arrest, and increased proliferation in the presence of pexidartinib and other FLT3 inhibitors. Inhibition of CDK4/6 activity in CCND3 mutant MV4;11 cells restored pexidartinib-induced cell-cycle arrest but not apoptosis.Conclusions:Mutations in CCND3, a gene not commonly mutated in AML, are a novel cause of clinical primary resistance to FLT3 inhibitors in AML and may have sensitivity to CDK4/6 inhibition.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Data from Recurrent Mutations in Cyclin D3 Confer Clinical Resistance to FLT3 Inhibitors in Acute Myeloid Leukemia
- Author
-
Neil P. Shah, Ross L. Levine, Barry S. Taylor, Gideon Bollag, Brian West, Mai H. Le, Henry H. Hsu, Theodore C. Tarver, Elli Papaemmanuil, Juan S. Medina-Martinez, Matthieu Najm, Franck Rapaport, Nicholas D. Socci, Cyriac Kandoth, Evan Massi, Aaron D. Viny, and Catherine C. Smith
- Abstract
Supplemental Figures
- Published
- 2023
- Full Text
- View/download PDF
9. Human IRF1 governs macrophagic IFN-γ immunity to mycobacteria
- Author
-
Jérémie Rosain, Anna-Lena Neehus, Jérémy Manry, Rui Yang, Jérémie Le Pen, Wassim Daher, Zhiyong Liu, Yi-Hao Chan, Natalia Tahuil, Özden Türel, Mathieu Bourgey, Masato Ogishi, Jean-Marc Doisne, Helena M. Izquierdo, Takayoshi Shirasaki, Tom Le Voyer, Antoine Guérin, Paul Bastard, Marcela Moncada-Vélez, Ji Eun Han, Taushif Khan, Franck Rapaport, Seon-Hui Hong, Andrew Cheung, Kathrin Haake, Barbara C. Mindt, Laura Pérez, Quentin Philippot, Danyel Lee, Peng Zhang, Darawan Rinchai, Fatima Al Ali, Manar Mahmoud Ahmad Ata, Mahbuba Rahman, Jessica N. Peel, Søren Heissel, Henrik Molina, Yasemin Kendir-Demirkol, Rasheed Bailey, Shuxiang Zhao, Jonathan Bohlen, Mathieu Mancini, Yoann Seeleuthner, Marie Roelens, Lazaro Lorenzo, Camille Soudée, María Elvira Josefina Paz, María Laura González, Mohamed Jeljeli, Jean Soulier, Serge Romana, Anne-Sophie L’Honneur, Marie Materna, Rubén Martínez-Barricarte, Mathieu Pochon, Carmen Oleaga-Quintas, Alexandre Michev, Mélanie Migaud, Romain Lévy, Marie-Alexandra Alyanakian, Flore Rozenberg, Carys A. Croft, Guillaume Vogt, Jean-François Emile, Laurent Kremer, Cindy S. Ma, Jörg H. Fritz, Stanley M. Lemon, András N. Spaan, Nicolas Manel, Laurent Abel, Margaret R. MacDonald, Stéphanie Boisson-Dupuis, Nico Marr, Stuart G. Tangye, James P. Di Santo, Qian Zhang, Shen-Ying Zhang, Charles M. Rice, Vivien Béziat, Nico Lachmann, David Langlais, Jean-Laurent Casanova, Philippe Gros, Jacinta Bustamante, Imagine - Institut des maladies génétiques (IHU) (Imagine - U1163), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), Hannover Medical School [Hannover] (MHH), St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller University [New York], Institut de Recherche en Infectiologie de Montpellier (IRIM), Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM), Yong Loo Lin School of Medicine [Singapore], Hospital del Niño Jesus, San Miguel de Tucumán, Bezmiâlem Vakıf Üniversitesi, McGill University = Université McGill [Montréal, Canada], Immunité Innée - Innate Immunity, Institut Pasteur [Paris] (IP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), Institut Curie [Paris], University of North Carolina System (UNC), Garvan Institute of medical research, The University of Sydney, CHU Necker - Enfants Malades [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Universidad de Antioquia = University of Antioquia [Medellín, Colombia], East China Normal University [Shangaï] (ECNU), Sidra Medicine [Doha, Qatar], 'Juan Pedro Garrahan' National Hospital of Pediatrics, Buenos Aires, Institut Necker Enfants-Malades (INEM - UM 111 (UMR 8253 / U1151)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), Hôpital Cochin [AP-HP], Hopital Saint-Louis [AP-HP] (AP-HP), Vanderbilt University Medical Center [Nashville], Vanderbilt University [Nashville], Institut Pasteur de Lille, Réseau International des Instituts Pasteur (RIIP), Hôpital Ambroise Paré [AP-HP], Université de Montpellier (UM), Howard Hughes Medical Institute (HHMI), The Laboratory of Human Genetics of Infectious Diseases is supported in part by grants from Inserm, Paris Cité University, the St. Giles Foundation, The Rockefeller University, the Center for Clinical and Translational Science (UL1TR001866), the National Center for Research Resources and the National Center for Advancing Sciences, the National Institutes of Health (NIH), (R01AI095983, R01AI088364, R01AI163029, and U19AI162568), the National Institute of Allergy and Infectious Diseases, the French National Research Agency (ANR) under the 'Investments for the future' program (ANR-10-IAHU-01), the Integrative Biology of Emerging Infectious Diseases Laboratory of Excellence (ANR-10-LABX-62-IBEID), the French Foundation for Medical Research (FRM) (EQU201903007798), the ANRS Nord-Sud (ANRS-COV05), ANRS (ECTZ170784-ANRS0073), GENVIR (ANR-20-CE93-003), GENMSMD (ANR-16-CE17-0005-01), AABIFNCOV (ANR-20-CO11-0001), GenMIS-C (ANR-21-COVR-0039), SUNLIVE (ANR-19-CE15-0012-01), MAFMACRO (ANR-22-CE92-0008) grants, Ecos-NORD (ECOS N°C19S01), the Fisher Center for Alzheimer's Research Foundation, the Meyer Foundation, the JPB Foundation, the European Union’s Horizon 2020 research and innovation program (824110, EASI-Genomics), the Square Foundation, Grandir–Fonds de solidarité pour l’Enfance, the Fondation du Souffle, the SCOR Corporate Foundation for Science, the French Ministry of Higher Education, Research, and Innovation (MESRI-COVID-19), and REACTing-INSERM. The Laboratory of Virology and Infectious Disease was supported in part by the NIH (R01AI091707-10 to C.M.R.). J.L.P. was supported by the Francois Wallace Monahan Postdoctoral Fellowship at The Rockefeller University and the European Molecular Biology Organization Long-Term Fellowship (ALTF 380-2018). N. Marr was supported by Sidra Medicine and the Qatar National Research Fund (NPRP9-251-3-045). The Yale Center for Mendelian Genomics (UM1HG006504) was funded by the National Human Genome Research Institute, the Yale GSP Coordinating Center (U24 HG008956), and the Yale High-Performance Computing Center (S10OD018521). This research was partly supported by Calcul Québec, Compute Canada Canadian Institutes of Health Research (CIHR) Project Grant to D. Langlais. (#168959) and a CIHR Foundation Grant (to P.G.). D. Langlais was also supported by an FRQS Chercheur-Boursier Junior 1 Award and the Calgary Foundation for Innovation John R. Evans Leaders Fund. P.G. is supported by a Distinguished James McGill Professorship award from McGill University. S.M.L is supported by the NIH: R01-AI103083 and R01-AI150095. J.R. was supported by poste d’accueil Inserm'. J.R., P.B., and T.L.V were supported by the MD-PhD program of the Imagine Institute by the Bettencourt Schueller Foundation. N.L received funding from the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation program (852178 grant), German Research Foundation,(DFG) under Germany’s Excellence Strategy—EXC 2155—project number 390874280 and REBIRTH 'Förderung aus Mitteln des Niedersächsischen Vorab'. A.-L.N. was supported by the international PhD program of the Imagine Institute, by the Bettencourt Schueller Foundation and the fin de thèse FRM program (FDT202204015102). R.Y. was supported by the Sackler Center for Biomedicine and Nutrition, the Shapiro-Silverberg Fund for the Advancement of Translational Research at the Center for Clinical and Translational Science of the Rockefeller University, and the Research Grant Program from the Immune Deficiency Foundation. D. Lee. was supported by FRMfellowship (FDM202006011282). C.S.M was supported by an Early-Mid Career Research Fellowship from the Department of Health of the New South Wales Government of Australia. S.G.T was supported by an NHMRC Leadership 3 Investigator Grant (1176665) and NHMRC grant (1113904). M.O. was supported by the David Rockefeller Graduate Program, the Funai Foundation for Information Technology, the Honjo International Scholarship Foundation, and the New York Hideyo Noguchi Memorial Society. This work was supported by grants from ANRS (ECTZ118797), Sidaction (20-2-AEQ-12822-2), and FRM (EQU202103012774) to N. Manel, and H.I. was supported by fellowships from Institut Curie, Seneca Foundation (20941/PD/18), and ANRS (ECTZ171453). A.N.S. was supported in part by the European Union’s Horizon 2020 research and innovation program (789645 Marie Sklodowska-Curie grant). Y.-H.C. is supported by an A∗STAR International Fellowship. J. Bohlen is an EMBO postdoctoral fellow. We thank the NIH Tetramer Core Facility (NTCF) for providing the 5-OP-RU-loaded MR1 tetramer, which was developed jointly with Dr. James McCluskey, Dr. Jamie Rossjohn, and Dr. David Fairlie., ANR-10-IAHU-0001,Imagine,Institut Hospitalo-Universitaire Imagine(2010), ANR-10-LABX-0062,IBEID,Integrative Biology of Emerging Infectious Diseases(2010), ANR-16-CE17-0005,GENMSMD,Dissection génétique de la Susceptibilité Mendélienne aux infections mycobactériennes chez l'homme(2016), ANR-20-CO11-0001,AABIFNCOV,Bases génétiques et immunologiques des auto-anticorps contre les interférons de type I prédisposant aux formes sévères de COVID-19.(2020), ANR-21-COVR-0039,GenMIS-C,Recherche des Déficits immunitaires innées monogéniques prédisposant au syndrome inflammatoire multisystémique chez l'enfant.(2021), ANR-19-CE15-0012,SUNLIVE,Variabilité structurale et fonctionnelle des lipides complexes chez les mycobactéries : de l'assemblage de la paroi à la physiopathologie et virulence(2019), ANR-22-CE92-0008,MAFMACRO,Genetic predisposition and the role of myeloid cells in the susceptibility to mycobacterial infection(2022), European Project: 824110,H2020-INFRAIA-2018-1,EASI-Genomics(2019), and TÜREL, ÖZDEN
- Subjects
inborn errors of immunity ,Temel Bilimler ,[SDV]Life Sciences [q-bio] ,interferon-stimulated gene ,Life Sciences ,Molecular Biology and Genetics ,Genel Biyokimya, Genetik ve Moleküler Biyoloji ,IRF1 ,General Biochemistry, Genetics and Molecular Biology ,Mycobacterium ,macrophages ,interferon-γ ,Yaşam Bilimleri ,viruses ,Cytogenetic ,Natural Sciences ,Moleküler Biyoloji ve Genetik ,Sitogenetik - Abstract
Inborn errors of human IFN-γ-dependent macrophagic immunity underlie mycobacterial diseases, whereas inborn errors of IFN-α/β-dependent intrinsic immunity underlie viral diseases. Both types of IFNs induce the transcription factor IRF1. We describe unrelated children with inherited complete IRF1 deficiency and early-onset, multiple, life-threatening diseases caused by weakly virulent mycobacteria and related intramacrophagic pathogens. These children have no history of severe viral disease, despite exposure to many viruses, including SARS-CoV-2, which is life-threatening in individuals with impaired IFN-α/β immunity. In leukocytes or fibroblasts stimulated in vitro, IRF1-dependent responses to IFN-γ are, both quantitatively and qualitatively, much stronger than those to IFN-α/β. Moreover, IRF1-deficient mononuclear phagocytes do not control mycobacteria and related pathogens normally when stimulated with IFN-γ. By contrast, IFN-α/β-dependent intrinsic immunity to nine viruses, including SARS-CoV-2, is almost normal in IRF1-deficient fibroblasts. Human IRF1 is essential for IFN-γ-dependent macrophagic immunity to mycobacteria, but largely redundant for IFN-α/β-dependent antiviral immunity.
- Published
- 2023
- Full Text
- View/download PDF
10. DiSiR: fast and robust method to identify ligand–receptor interactions at subunit level from single-cell RNA-sequencing data
- Author
-
Milad R Vahid, Andre H Kurlovs, Tommaso Andreani, Franck Augé, Reza Olfati-Saber, Emanuele de Rinaldis, Franck Rapaport, and Virginia Savova
- Subjects
Structural Biology ,Applied Mathematics ,Genetics ,Molecular Biology ,Computer Science Applications - Abstract
Most cell–cell interactions and crosstalks are mediated by ligand–receptor interactions. The advent of single-cell RNA-sequencing (scRNA-seq) techniques has enabled characterizing tissue heterogeneity at single-cell level. In the past few years, several methods have been developed to study ligand–receptor interactions at cell type level using scRNA-seq data. However, there is still no easy way to query the activity of a specific user-defined signaling pathway in a targeted way or to map the interactions of the same subunit with different ligands as part of different receptor complexes. Here, we present DiSiR, a fast and easy-to-use permutation-based software framework to investigate how individual cells are interacting with each other by analyzing signaling pathways of multi-subunit ligand-activated receptors from scRNA-seq data, not only for available curated databases of ligand–receptor interactions, but also for interactions that are not listed in these databases. We show that, when utilized to infer ligand–receptor interactions from both simulated and real datasets, DiSiR outperforms other well-known permutation-based methods, e.g. CellPhoneDB and ICELLNET. Finally, to demonstrate DiSiR’s utility in exploring data and generating biologically relevant hypotheses, we apply it to COVID lung and rheumatoid arthritis (RA) synovium scRNA-seq datasets and highlight potential differences between inflammatory pathways at cell type level for control versus disease samples.
- Published
- 2023
- Full Text
- View/download PDF
11. 1010 A single-cell transcriptomic atlas of human NK cells to guide cancer immunotherapy
- Author
-
Ryan King, Azim Amirabad, Amir Bayegan, Joachim Theilhaber, Nicole Acuff, Shannon McGrath, Xiangming Li, Franck Rapaport, Jack Pollard, and Donald Jackson
- Published
- 2022
- Full Text
- View/download PDF
12. Classification of arrayCGH data using fused SVM.
- Author
-
Franck Rapaport, Emmanuel Barillot, and Jean-Philippe Vert
- Published
- 2008
- Full Text
- View/download PDF
13. Recurrent Mutations in Cyclin D3 Confer Clinical Resistance to FLT3 Inhibitors in Acute Myeloid Leukemia
- Author
-
Elli Papaemmanuil, Mai H. Le, Catherine C. Smith, Nicholas D. Socci, Brian L. West, Franck Rapaport, Ross L. Levine, Matthieu Najm, Evan Massi, Juan S. Medina-Martinez, Neil P. Shah, Henry H. Hsu, Theodore C. Tarver, Aaron D. Viny, Gideon Bollag, Barry S. Taylor, and Cyriac Kandoth
- Subjects
Neuroblastoma RAS viral oncogene homolog ,FLT3 Internal Tandem Duplication ,Cancer Research ,Aminopyridines ,IDH2 ,Article ,hemic and lymphatic diseases ,Cell Line, Tumor ,Humans ,Medicine ,Pyrroles ,Epigenetics ,Cyclin D3 ,Protein Kinase Inhibitors ,Gene ,business.industry ,Myeloid leukemia ,Leukemia, Myeloid, Acute ,fms-Like Tyrosine Kinase 3 ,Oncology ,Drug Resistance, Neoplasm ,Mutation ,Cancer research ,business ,Tyrosine kinase - Abstract
Purpose: Biomarkers of response and resistance to FLT3 tyrosine kinase inhibitors (TKI) are still emerging, and optimal clinical combinations remain unclear. The purpose of this study is to identify co-occurring mutations that influence clinical response to the novel FLT3 inhibitor pexidartinib (PLX3397). Experimental Design: We performed targeted sequencing of pretreatment blasts from 29 patients with FLT3 internal tandem duplication (ITD) mutations treated on the phase I/II trial of pexidartinib in relapsed/refractory FLT3-ITD+ acute myeloid leukemia (AML). We sequenced 37 samples from 29 patients with available material, including 8 responders and 21 non-responders treated at or above the recommended phase II dose of 3,000 mg. Results: Consistent with other studies, we identified mutations in NRAS, TP53, IDH2, and a variety of epigenetic and transcriptional regulators only in non-responders. Among the most frequently mutated genes in non-responders was Cyclin D3 (CCND3). A total of 3 individual mutations in CCND3 (Q276*, S264R, and T283A) were identified in 2 of 21 non-responders (one patient had both Q276* and S264R). No CCND3 mutations were found in pexidartinib responders. Expression of the Q276* and T283A mutations in FLT3-ITD MV4;11 cells conferred resistance to apoptosis, decreased cell-cycle arrest, and increased proliferation in the presence of pexidartinib and other FLT3 inhibitors. Inhibition of CDK4/6 activity in CCND3 mutant MV4;11 cells restored pexidartinib-induced cell-cycle arrest but not apoptosis. Conclusions: Mutations in CCND3, a gene not commonly mutated in AML, are a novel cause of clinical primary resistance to FLT3 inhibitors in AML and may have sensitivity to CDK4/6 inhibition.
- Published
- 2021
- Full Text
- View/download PDF
14. Genomic and evolutionary portraits of disease relapse in acute myeloid leukemia
- Author
-
Richard J D'Andrea, Caroline Sheridan, Noushin Farnoud, Yaseswini Neelamraju, Agata Gruszczynska, Timour Baslan, Ross L. Levine, Donna Neuberg, Peter J. M. Valk, Martin Carroll, Franck Rapaport, Stefan Bekiranov, Tak C. Lee, Michael W. Becker, Samuel Haddox, Duane C. Hassane, Alexis Thurmond, Juan S. Medina-Martinez, Ari Melnick, Scott W. Lowe, Marc Robert de Massy, Lars Bullinger, Francine E. Garrett-Bakelman, Heardly Moses Murdock, Hematology, Rapaport, Franck, Neelamraju, Yaseswini, Baslan, Timour, Hassane, Duane, D'Andrea, Richard, and Garrett-Bakelman, Francine E
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Letter ,business.industry ,MEDLINE ,Myeloid leukemia ,Hematology ,Genomics ,Prognosis ,Evolution, Molecular ,Gene Expression Regulation, Neoplastic ,Leukemia, Myeloid, Acute ,Text mining ,SDG 3 - Good Health and Well-being ,Internal medicine ,medicine ,Biomarkers, Tumor ,Humans ,Neoplasm Recurrence, Local ,business ,DISEASE RELAPSE ,Cancer genetics - Abstract
Refereed/Peer-reviewed
- Published
- 2021
15. Inherited human IFN-γ deficiency underlies mycobacterial disease
- Author
-
Jérémie Rosain, Bernhard Fleckenstein, Chih-Yu Chi, Cheng-Lung Ku, Nico Marr, Rui Yang, Jacinta Bustamante, Flore Rozenberg, Carmen Oleaga-Quintas, Vivien Béziat, Yoann Seeleuthner, Rubén Martínez-Barricarte, Franck Rapaport, Jing-Ya Ding, Michel J. Massaad, Stéphanie Boisson-Dupuis, Raif S. Geha, Fatima Al Ali, Antoine Guérin, Laurent Abel, Taushif Khan, Gaspard Kerner, Waleed Al-Herz, Mahbuba Rahman, Jean-Laurent Casanova, Caroline Deswarte, Manon Roynard, Diane S. T. Garcia, Ahmad Al-Khabaz, and Valentine Jeanne-Julien
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Biology ,Frameshift mutation ,Interferon-gamma ,03 medical and health sciences ,symbols.namesake ,Negative selection ,0302 clinical medicine ,Immunity ,medicine ,Humans ,Gene ,Loss function ,Receptors, Interferon ,Sequence Deletion ,Genetics ,Mycobacterium Infections ,Base Sequence ,Homozygote ,Genetic Diseases, Inborn ,General Medicine ,Mycobacterium bovis ,Phenotype ,030104 developmental biology ,Cytokine ,030220 oncology & carcinogenesis ,Mendelian inheritance ,symbols ,Female ,Research Article - Abstract
Mendelian susceptibility to mycobacterial disease (MSMD) is characterized by a selective predisposition to clinical disease caused by the Bacille Calmette-Guérin (BCG) vaccine and environmental mycobacteria. The known genetic etiologies of MSMD are inborn errors of IFN-γ immunity due to mutations of 15 genes controlling the production of or response to IFN-γ. Since the first MSMD-causing mutations were reported in 1996, biallelic mutations in the genes encoding IFN-γ receptor 1 (IFN-γR1) and IFN-γR2 have been reported in many patients of diverse ancestries. Surprisingly, mutations of the gene encoding the IFN-γ cytokine itself have not been reported, raising the remote possibility that there might be other agonists of the IFN-γ receptor. We describe 2 Lebanese cousins with MSMD, living in Kuwait, who are both homozygous for a small deletion within the IFNG gene (c.354_357del), causing a frameshift that generates a premature stop codon (p.T119Ifs4*). The mutant allele is loss of expression and loss of function. We also show that the patients’ herpesvirus Saimiri–immortalized T lymphocytes did not produce IFN-γ, a phenotype that can be rescued by retrotransduction with WT IFNG cDNA. The blood T and NK lymphocytes from these patients also failed to produce and secrete detectable amounts of IFN-γ. Finally, we show that human IFNG has evolved under stronger negative selection than IFNGR1 or IFNGR2, suggesting that it is less tolerant to heterozygous deleterious mutations than IFNGR1 or IFNGR2. This may account for the rarity of patients with autosomal-recessive, complete IFN-γ deficiency relative to patients with complete IFN-γR1 and IFN-γR2 deficiencies.
- Published
- 2020
- Full Text
- View/download PDF
16. DiSiR: a software framework to identify ligand-receptor interactions at subunit level from single-cell RNA-sequencing data
- Author
-
Milad R. Vahid, Andre Kurlovs, Franck Auge, Reza Olfati-Saber, Emanuele de Rinaldis, Franck Rapaport, and Virginia Savova
- Abstract
Most of cell-cell interactions and crosstalks are mediated by ligand-receptor interactions. The advent of single-cell RNA-sequencing (scRNA-seq) techniques has enabled characterizing tissue heterogeneity at single-cell level. Over the past recent years, several methods have been developed to study ligand-receptor interactions at cell type level using scRNA-seq data. However, there is still no easy way to query the activity of a specific user-defined signaling pathway in a targeted way or map the interactions of the same subunit with different ligands as part of different receptor complexes. Here, we present DiSiR, a fast and easy-to-use permutation-based software framework to investigate how individual cells are interacting with each other by analyzing signaling pathways of multi-subunit ligand-activated receptors from scRNA-seq data, not only for available curated databases of ligand-receptor interactions, but also for interactions that are not listed in these databases. We show that, when utilized to infer melanoma disease map on a gold-standard dataset, DiSiR outperforms other well-known permutation-based methods, e.g., CellPhoneDB and ICELLNET. To demonstrate DiSiR’s utility in exploring data and generating biologically relevant hypotheses, we apply it to COVID lung and rheumatoid arthritis (RA) synovium scRNA-seq data and highlight potential differences between inflammatory pathways at cell type level for control vs. disease samples.
- Published
- 2022
- Full Text
- View/download PDF
17. Abstract 3155: Gene expression profiles reveal distinct regulatory activities of transcription factors GATA1 and TAL1 upon AML relapse
- Author
-
Zhenjia Wang, Yaseswini Neelamraju, Cem Meydan, Nicholas Dunham, Jorge Gandara, Tak Lee, Subhash Prajapati, Franck Rapaport, Caroline Sheridan, Paul Zumbo, Michael Becker, Lars Bullinger, Martin Carroll, Richard D’Andrea, Richard Dillon, Ross Levine, Christopher E. Mason, Ari Melnick, Donna Neuberg, Stefan Bekiranov, Chongzhi Zang, and Francine E. Garrett-Bakelman
- Subjects
Cancer Research ,Oncology - Abstract
The purpose of this study is to identify key regulatory pathways that potentially drive abnormal gene expression program in relapsed Acute Myeloid Leukemia (AML) patients, by integrative computational analyses on multi-omics molecular profiles. Relapsed AML remains a clinical challenge. Epigenetic heterogeneity may contribute to transcriptional dysregulation and disease progression in AML. However, what specific transcriptional programs and potential regulatory mechanisms contribute to disease relapse are not yet well understood. To characterize the global transcriptional landscapes in relapsed AML, we integrated genomics data from two cohorts of matched diagnosis and relapse patient specimens. We identified 5,416 differentially expressed genes (DEGs) between diagnosis and relapse in Cohort I. Unsupervised clustering yielded three distinct DEG groups: group A, B and C genes that were predominantly (88%) down-regulated, divergently regulated, or predominantly (65%) up-regulated, respectively, upon relapse. The expression pattern of all DEGs separated the patients into two clusters, most robustly by Group B genes. Interestingly, the majority of DEGs did not associate with changes in gene promoter methylation. Similar patterns were observed in Cohort II. We used Binding Analysis for Regulation of Transcription (BART) to identify transcriptional regulators (TRs) that potentially regulated the DEGs not associated with DNA methylation changes, and assessed the differential expression of identified TRs during disease progression. PU.1 was identified as a potential TR for Group A genes and was down-regulated upon relapse. GATA1 and TAL1 were identified as regulating Group B genes and were up-regulated in patient cluster1 and down-regulated in cluster2, consistent with the expression pattern of Group B genes. RBBP5 was a top predicted TR for Group C genes and was up-regulated upon relapse. We next validated the potential functionality of those predicted factors. In NSG mice transplanted with a human AML specimen, TAL1 and GATA1 were downregulated in AML cells collected four weeks after chemotherapy treatment, and were inferred as TRs for the down-regulated genes, similar to the patient data. PU.1 was inferred as regulating the up-regulated genes. Furthermore, we found that the level of differential expression of TAL1, GATA1, and PU.1 in each patient specimen associated with the correlation of DEG profiles between the patient specimen and TR perturbation in human-derived hematopoietic cell lines. Our results support the possibility that in some AML patients, TRs with roles in hematopoiesis and leukemia might contribute to disease relapse. Further mechanistic studies deciphering the molecular and phenotypic events facilitated by these TRs will yield significant insight into disease biology and possible therapeutic targeting approaches in relapsed AML. Citation Format: Zhenjia Wang, Yaseswini Neelamraju, Cem Meydan, Nicholas Dunham, Jorge Gandara, Tak Lee, Subhash Prajapati, Franck Rapaport, Caroline Sheridan, Paul Zumbo, Michael Becker, Lars Bullinger, Martin Carroll, Richard D’Andrea, Richard Dillon, Ross Levine, Christopher E. Mason, Ari Melnick, Donna Neuberg, Stefan Bekiranov, Chongzhi Zang, Francine E. Garrett-Bakelman. Gene expression profiles reveal distinct regulatory activities of transcription factors GATA1 and TAL1 upon AML relapse [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 3155.
- Published
- 2023
- Full Text
- View/download PDF
18. Biochemically deleterious human NFKB1 variants underlie an autosomal dominant form of common variable immunodeficiency
- Author
-
Peng Zhang, Katharina Thoma, Bingnan Lyu, Gulbu Uzel, Juan Li, Boualem Hammadi, András N Spaan, Jamila El Baghdadi, Alexandra F. Freeman, Charlotte Cunningham-Rundles, Yu Zhang, Jean-Laurent Casanova, Claire Fieschi, Franck Rapaport, Jérémie Rosain, Vivien Béziat, Simon J. Pelham, Wei-Te Lei, Anne Puel, Maya Chrabieh, Helen C. Su, V. Koneti Rao, David Hum, Stéphanie Boisson-Dupuis, Jacinta Bustamante, Mélanie Migaud, Anne-Sophie Korganow, Qian Zhang, Aurélie Cobat, Steven M. Holland, Laurent Abel, Vishukumar Aimanianda, Bertrand Boisson, Manfred Fliegauf, Benedetta Bigio, Bodo Grimbacher, Yoann Seeleuthner, Takaki Asano, Carol J Saunders, Shen-Ying Zhang, and Emmanuelle Jouanguy
- Subjects
Transcriptional Activation ,P50 ,Immunology ,Mutant ,Population ,Haploinsufficiency ,Biology ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Immunology and Allergy ,education ,Loss function ,030304 developmental biology ,Dominance (genetics) ,Genetics ,0303 health sciences ,education.field_of_study ,Common variable immunodeficiency ,HEK 293 cells ,NF-kappa B ,NF-kappa B p50 Subunit ,medicine.disease ,Common Variable Immunodeficiency ,HEK293 Cells ,Phenotype ,COS Cells ,030215 immunology - Abstract
Autosomal dominant (AD) NFKB1 deficiency is thought to be the most common genetic etiology of common variable immunodeficiency (CVID). However, the causal link between NFKB1 variants and CVID has not been demonstrated experimentally and genetically, as there has been insufficient biochemical characterization and enrichment analysis. We show that the cotransfection of NFKB1-deficient HEK293T cells (lacking both p105 and its cleaved form p50) with a κB reporter, NFKB1/p105, and a homodimerization-defective RELA/p65 mutant results in p50:p65 heterodimer–dependent and p65:p65 homodimer–independent transcriptional activation. We found that 59 of the 90 variants in patients with CVID or related conditions were loss of function or hypomorphic. By contrast, 258 of 260 variants in the general population or patients with unrelated conditions were neutral. None of the deleterious variants displayed negative dominance. The enrichment in deleterious NFKB1 variants of patients with CVID was selective and highly significant (P = 2.78 × 10−15). NFKB1 variants disrupting NFKB1/p50 transcriptional activity thus underlie AD CVID by haploinsufficiency, whereas neutral variants in this assay should not be considered causal.
- Published
- 2021
- Full Text
- View/download PDF
19. Inherited human c-Rel deficiency disrupts myeloid and lymphoid immunity to multiple infectious agents
- Author
-
Rui Yang, Rubén Martínez-Barricarte, Janet Markle, Franck Rapaport, Stuart G. Tangye, Mathieu Bourgey, David Langlais, Anne Puel, Aziz Belkadi, Masato Ogishi, Simon J. Pelham, Jean-Laurent Casanova, Bertrand Boisson, Cindy S. Ma, Vivien Béziat, Eman Abou Moussa, Jérémie Rosain, Laurent Abel, Tomi Lazarov, Serkan Belkaya, Fatima Ailal, Ibtihal Benhsaien, Coralie Briand, Alessandro Plebani, Romain Lévy, Frederic Geissmann, Caroline Deswarte, Andrea Guennoun, Luis R. Saraiva, Tanwir Habib, Mehdi Emam, Vassilios Lougaris, Philippe Gros, Yu Jerry Zhou, Ahmed Aziz Bousfiha, Geetha Rao, Kathryn Payne, Sylvain Breton, Ai Ing Lim, Kang Liu, Kunihiko Moriya, Danielle T. Avery, Bénédicte Neven, Nico Marr, Jacinta Bustamante, Mélanie Migaud, and James P. Di Santo
- Subjects
Myeloid ,Primary Immunodeficiency Diseases ,Naive B cell ,Adaptive Immunity ,Biology ,Lymphocyte Activation ,Consanguinity ,Immunity ,medicine ,Humans ,Protein Isoforms ,Myeloid Cells ,Lymphocytes ,Child ,B cell ,CD86 ,Host Microbial Interactions ,Homozygote ,Hematopoietic Stem Cell Transplantation ,General Medicine ,Acquired immune system ,Immunity, Innate ,Proto-Oncogene Proteins c-rel ,medicine.anatomical_structure ,Mutation ,Immunology ,Female ,Genes, rel ,REL ,CD8 ,Research Article - Abstract
We studied a child with severe viral, bacterial, fungal, and parasitic diseases, who was homozygous for a loss-of-function mutation of REL, encoding c-Rel, which is selectively expressed in lymphoid and myeloid cells. The patient had low frequencies of NK, effector memory cells reexpressing CD45RA (Temra) CD8(+) T cells, memory CD4(+) T cells, including Th1 and Th1*, Tregs, and memory B cells, whereas the counts and proportions of other leukocyte subsets were normal. Functional deficits of myeloid cells included the abolition of IL-12 and IL-23 production by conventional DC1s (cDC1s) and monocytes, but not cDC2s. c-Rel was also required for induction of CD86 expression on, and thus antigen-presenting cell function of, cDCs. Functional deficits of lymphoid cells included reduced IL-2 production by naive T cells, correlating with low proliferation and survival rates and poor production of Th1, Th2, and Th17 cytokines by memory CD4(+) T cells. In naive CD4(+) T cells, c-Rel is dispensable for early IL2 induction but contributes to later phases of IL2 expression. The patient’s naive B cells displayed impaired MYC and BCL2L1 induction, compromising B cell survival and proliferation and preventing their differentiation into Ig-secreting plasmablasts. Inherited c-Rel deficiency disrupts the development and function of multiple myeloid and lymphoid cells, compromising innate and adaptive immunity to multiple infectious agents.
- Published
- 2021
- Full Text
- View/download PDF
20. Fatal Cytomegalovirus Infection in an Adult with Inherited NOS2 Deficiency
- Author
-
Fatima Al Ali, Laurent Abel, Jean-François Emile, Davood Mansouri, Mahbuba Rahman, Franck Rapaport, Stéphanie Boisson-Dupuis, Nicholas Hernandez, Taushif Khan, Anna-Lena Neehus, David Hum, Benedetta Bigio, Vivien Béziat, Nico Marr, Jean-Laurent Casanova, Scott Drutman, Jacinta Bustamante, Emmanuelle Jouanguy, Majid Marjani, Robert Fisch, Ruslana Bryk, Nahal Mansouri, Serkan Belkaya, Lazaro Lorenzo-Diaz, Carl Nathan, and Seyed Alireza Mahdaviani
- Subjects
Male ,Genotype ,Congenital cytomegalovirus infection ,Nitric Oxide Synthase Type II ,Severe disease ,macromolecular substances ,030204 cardiovascular system & hematology ,Nitric Oxide ,Article ,03 medical and health sciences ,Fatal Outcome ,0302 clinical medicine ,Loss of Function Mutation ,parasitic diseases ,Exome Sequencing ,medicine ,Humans ,030212 general & internal medicine ,Frameshift Mutation ,business.industry ,Homozygote ,fungi ,food and beverages ,virus diseases ,General Medicine ,Middle Aged ,medicine.disease ,Pedigree ,Multiple infections ,Cytomegalovirus infection ,Cytomegalovirus Infections ,Immunology ,Female ,business - Abstract
BACKGROUND: Cytomegalovirus (CMV) can cause severe disease in children and adults with a variety of inherited or acquired T-cell immunodeficiencies, who are prone to multiple infections. It can also rarely cause disease in otherwise healthy persons. The pathogenesis of idiopathic CMV disease is unknown. Inbred mice that lack the gene encoding nitric oxide synthase 2 (Nos2) are susceptible to the related murine CMV infection. METHODS: We studied a previously healthy 51-year-old man from Iran who after acute CMV infection had an onset of progressive CMV disease that led to his death 29 months later. We hypothesized that the patient may have had a novel type of inborn error of immunity. Thus, we performed whole-exome sequencing and tested candidate mutant alleles experimentally. RESULTS: We found a homozygous frameshift mutation in NOS2 encoding a truncated NOS2 protein that did not produce nitric oxide, which determined that the patient had autosomal recessive NOS2 deficiency. Moreover, all NOS2 variants that we found in homozygosity in public databases encoded functional proteins, as did all other variants with an allele frequency greater than 0.001. CONCLUSIONS: These findings suggest that inherited NOS2 deficiency was clinically silent in this patient until lethal infection with CMV. Moreover, NOS2 appeared to be redundant for control of other pathogens in this patient. (Funded by the National Center for Advancing Translational Sciences and others.)
- Published
- 2020
- Full Text
- View/download PDF
21. Human STAT3 variants underlie autosomal dominant hyper-IgE syndrome by negative dominance
- Author
-
Capucine Picard, Joëlle Khourieh, Jane Peake, Antoine Guérin, Bertrand Boisson, Aziz Bousfiha, Jamila El Baghdadi, David Hum, Takaki Asano, Jean-Laurent Casanova, Andrew Williams, Simon J. Pelham, Stéphanie Boisson-Dupuis, Peng Zhang, Maya Chrabieh, Luke Droney, Wei-Te Lei, Ilham Fadil, Ji Eun Han, András N Spaan, Qian Zhang, Nevin Hatipoğlu, Franck Rapaport, Anne Puel, Tanwir Habib, Nico Marr, Fatih Celmeli, Vivien Béziat, Joseph Mackie, Biman Saikia, Stuart G. Tangye, Laurent Abel, Tayfun Ozcelik, Juan Li, Sudhir Gupta, Luckshman Ganeshanandan, and Özçelik, Tayfun
- Subjects
Male ,Infectious disease and host defense ,Dominant negative ,Innate immunity and inflammation ,0302 clinical medicine ,Immunology and Allergy ,10. No inequality ,STAT3 ,Child ,Frameshift Mutation ,Dominance (genetics) ,Genes, Dominant ,Translation reinitiation ,Genetics ,0303 health sciences ,education.field_of_study ,Middle Aged ,3. Good health ,Pedigree ,Codon, Nonsense ,030220 oncology & carcinogenesis ,Child, Preschool ,Autosomal dominant hyper-IgE syndrome ,Female ,Haploinsufficiency ,Job Syndrome ,Gene isoform ,Adult ,STAT3 Transcription Factor ,Adolescent ,Immunology ,Population ,Innate Immunity and Inflammation ,Biology ,Article ,Infectious Disease and Host Defense ,Evolution, Molecular ,03 medical and health sciences ,Immunodeficiency ,Humans ,Family ,RNA, Messenger ,education ,Alleles ,030304 developmental biology ,Human disease genetics ,Infant, Newborn ,Infant ,Alternative Splicing ,Genetics, Population ,HEK293 Cells ,Protein Biosynthesis ,Mutation ,biology.protein ,Human Disease Genetics - Abstract
Most patients with autosomal dominant hyper-IgE syndrome (AD-HIES) are heterozygous for rare STAT3 variants. The mechanism of dominance was recently questioned. The authors show that both in-frame and out-of-frame STAT3 variants underlie AD-HIES by negative dominance and not haploinsufficiency., Most patients with autosomal dominant hyper-IgE syndrome (AD-HIES) carry rare heterozygous STAT3 variants. Only six of the 135 in-frame variants reported have been experimentally shown to be dominant negative (DN), and it has been recently suggested that eight out-of-frame variants operate by haploinsufficiency. We experimentally tested these 143 variants, 7 novel out-of-frame variants found in HIES patients, and other STAT3 variants from the general population. Strikingly, all 15 out-of-frame variants were DN via their encoded (1) truncated proteins, (2) neoproteins generated from a translation reinitiation codon, and (3) isoforms from alternative transcripts or a combination thereof. Moreover, 128 of the 135 in-frame variants (95%) were also DN. The patients carrying the seven non-DN STAT3 in-frame variants have not been studied for other genetic etiologies. Finally, none of the variants from the general population tested, including an out-of-frame variant, were DN. Overall, our findings show that heterozygous STAT3 variants, whether in or out of frame, underlie AD-HIES through negative dominance rather than haploinsufficiency., Graphical Abstract
- Published
- 2021
22. Negative selection on human genes underlying inborn errors depends on disease outcome and both the mode and mechanism of inheritance
- Author
-
Lluis Quintana-Murci, Etienne Patin, Jean-Laurent Casanova, Laurent Abel, Anne Puel, Jérémie Rosain, Shen-Ying Zhang, Vivien Béziat, Joseph G. Gleeson, Anne Gregor, Emmanuelle Jouanguy, Franck Rapaport, Stéphanie Boisson-Dupuis, Qian Zhang, Bertrand Boisson, Jacinta Bustamante, St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller University [New York], Imagine - Institut des maladies génétiques (IHU) (Imagine - U1163), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP), Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Center for the Study of Primary Immunodeficiencies [Paris], Université Paris Descartes - Paris 5 (UPD5)-CHU Necker - Enfants Malades [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Howard Hughes Medical Institute [La Jolla, CA, USA], Rady Children’s Institute of Genomic Medicine [La Jolla, CA, USA], University of California, Laboratory for Pediatric Brain Disease, Génétique Evolutive Humaine - Human Evolutionary Genetics, Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Collège de France (CdF (institution)), Howard Hughes Medical Institute [New York] (HHMI), Howard Hughes Medical Institute (HHMI)-Rockefeller University [New York]-Columbia University Irving Medical Center (CUIMC)-New York University School of Medicine, NYU System (NYU)-NYU System (NYU), This work was supported in part by the Rockefeller University, the St. Giles Foundation, Institut National de la Santé et de la Recherche Médicale, Paris Descartes University, the National Center for Research Resources and the National Center for Advancing Sciences, NIH Clinical and Translational Science Award program (Grant UL1TR001866), the Integrative Biology of Emerging Infectious Diseases Laboratory of Excellence (Grant ANR-10-LABX-62-IBEID), and the French National Research Agency (ANR) (Grants ANR-10-IAHU-01, ANR-15-CE17-0014). The laboratory of L.Q.-M. and E.P. is supported by the Institut Pasteur, the Collège de France, the Investissement d’Avenir program, Laboratoires d’Excellence 'Milieu Intérieur' (Grant ANR-10-LABX-69-01), Fondation pour la Recherche Médicale (Grant FRM DEQ20180339214), Fondation Allianz–Institut de France, and Fondation de France., ANR-10-LABX-0062,IBEID,Integrative Biology of Emerging Infectious Diseases(2010), ANR-10-IAHU-0001,Imagine,Institut Hospitalo-Universitaire Imagine(2010), ANR-15-CE17-0014,ProgLegio,Biomarqueurs bactériens et humains d'intérêt pronostic pour les légionelloses sévères(2015), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), University of California (UC), Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), Collège de France - Chaire Génomique humaine et évolution, Howard Hughes Medical Institute (HHMI)-New York University School of Medicine, NYU System (NYU)-NYU System (NYU)-Rockefeller University [New York]-Columbia University Irving Medical Center (CUIMC), and Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Candidate gene ,Population ,Genes, Recessive ,Biology ,03 medical and health sciences ,Negative selection ,0302 clinical medicine ,Humans ,Selection, Genetic ,education ,Selection (genetic algorithm) ,Loss function ,Genes, Dominant ,030304 developmental biology ,Genetics ,0303 health sciences ,education.field_of_study ,Multidisciplinary ,Mechanism (biology) ,Immunity ,Inheritance (genetic algorithm) ,Genetic Variation ,Biological Sciences ,Penetrance ,3. Good health ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,Metabolism, Inborn Errors ,030217 neurology & neurosurgery - Abstract
International audience; Genetic variants underlying life-threatening diseases, being unlikely to be transmitted to the next generation, are gradually and selectively eliminated from the population through negative selection. We study the determinants of this evolutionary process in human genes underlying monogenic diseases by comparing various negative selection scores and an integrative approach, CoNeS, at 366 loci underlying inborn errors of immunity (IEI). We find that genes underlying autosomal dominant (AD) or X-linked IEI have stronger negative selection scores than those underlying autosomal recessive (AR) IEI, whose scores are not different from those of genes not known to be disease causing. Nevertheless, genes underlying AR IEI that are lethal before reproductive maturity with complete penetrance have stronger negative selection scores than other genes underlying AR IEI. We also show that genes underlying AD IEI by loss of function have stronger negative selection scores than genes underlying AD IEI by gain of function, while genes underlying AD IEI by haploinsufficiency are under stronger negative selection than other genes underlying AD IEI. These results are replicated in 1,140 genes underlying inborn errors of neurodevelopment. Finally, we propose a supervised classifier, SCoNeS, which predicts better than state-of-the-art approaches whether a gene is more likely to underlie an AD or AR disease. The clinical outcomes of monogenic inborn errors, together with their mode and mechanisms of inheritance, determine the levels of negative selection at their corresponding loci. Integrating scores of negative selection may facilitate the prioritization of candidate genes and variants in patients suspected to carry an inborn error.
- Published
- 2021
- Full Text
- View/download PDF
23. Inherited GATA2 deficiency is dominant by haploinsufficiency and displays incomplete clinical penetrance
- Author
-
Rubén Martínez-Barricarte, Guillaume Vogt, Gabriela López-Herrera, Lidia Branco, Laurent Abel, Patricia Mariá O.Farrill Romanillos, Anna-Lena Neehus, Manon Roynard, Bengü Gerçeker, Júlia Vasconcelos, José Luis Franco Restrepo, Franck Rapaport, Sairaj Munavar Sajjath, Caroline Deswarte, Jean Donadieu, Christine Bellanné-Chantelot, Fatma Omur Ardeniz, Antoine Guérin, Antonio Condino-Neto, Noé Ramirez Alejo, A. S. Brunel, Caroline Thomas, Claire Lozano, Alexis Cuffel, Laura Berrón-Ruiz, Rebeca Pérez de Diego, Carmen Oleaga-Quintas, Kang Liu, Elise Launay, Mónica Martínez-Gallo, Vanesa Cunill Monjo, Marlène Pasquet, Laura Marques, D.B. Lew, Claire Fieschi, Fethi Mellouli, Tom Le Voyer, Carlos Rodríguez-Gallego, Luiz Fernando Job Jobim, Nora Hilda Segura Méndez, Eric Jeziorski, Yu Jerry Zhou, Andrés Augusto Arias, Stéphanie Boisson-Dupuis, Marie-Gabrielle Vigué, Anne Puel, Stéphane Marot, Aurélie Cobat, Kacy A Ramirez, Nuria Fernández-Hidalgo, Jacinta Bustamante, Jérémie Rosain, Lazaro Lorenzo-Diaz, Mariana Jobim, Regis A. Campos, Torsten Witte, Roger Colobran, Jean-Laurent Casanova, Marcela Moncada-Vélez, Edgar Borges de Oliveira-Júnior, and Ege Üniversitesi
- Subjects
Adult ,Male ,Adolescent ,Genotype ,GATA2 Deficiency ,mycobacteria ,DNA Mutational Analysis ,Immunology ,Penetrance ,Context (language use) ,Locus (genetics) ,Kaplan-Meier Estimate ,Monocytopenia ,Asymptomatic ,Article ,Cell Line ,Young Adult ,Databases, Genetic ,Outcome Assessment, Health Care ,Exome Sequencing ,medicine ,GATA2 ,Humans ,Immunology and Allergy ,Genetic Predisposition to Disease ,Child ,Alleles ,Genetic Association Studies ,Germ-Line Mutation ,Genes, Dominant ,Mycobacterium Infections ,Primary immunodeficiency ,FENÓTIPOS ,business.industry ,Middle Aged ,medicine.disease ,Hematologic Diseases ,Pedigree ,haploinsufficiency ,Phenotype ,tuberculosis ,Female ,medicine.symptom ,business ,Haploinsufficiency - Abstract
Purpose Germline heterozygous mutations of GATA2 underlie a variety of hematological and clinical phenotypes. The genetic, immunological, and clinical features of GATA2-deficient patients with mycobacterial diseases in the familial context remain largely unknown. Methods We enrolled 15 GATA2 index cases referred for mycobacterial disease. We describe their genetic and clinical features including their relatives. Results We identified 12 heterozygous GATA2 mutations, two of which had not been reported. Eight of these mutations were loss-of-function, and four were hypomorphic. None was dominant-negative in vitro, and the GATA2 locus was found to be subject to purifying selection, strongly suggesting a mechanism of haploinsufficiency. Three relatives of index cases had mycobacterial disease and were also heterozygous, resulting in 18 patients in total. Mycobacterial infection was the first clinical manifestation in 11 patients, at a mean age of 22.5 years (range: 12 to 42 years). Most patients also suffered from other infections, monocytopenia, or myelodysplasia. Strikingly, the clinical penetrance was incomplete (32.9% by age 40 years), as 16 heterozygous relatives aged between 6 and 78 years, including 4 older than 60 years, were completely asymptomatic. Conclusion Clinical penetrance for mycobacterial disease was found to be similar to other GATA2 deficiency-related manifestations. These observations suggest that other mechanisms contribute to the phenotypic expression of GATA2 deficiency. A diagnosis of autosomal dominant GATA2 deficiency should be considered in patients with mycobacterial infections and/or other GATA2 deficiency-related phenotypes at any age in life. Moreover, all direct relatives should be genotyped at the GATA2 locus., INSERM, University of Paris; Rockefeller University; St. Giles Foundation; National Institute of Allergy and Infectious Diseases (NIAID) of the National Institutes of Health (NIH)United States Department of Health & Human ServicesNational Institutes of Health (NIH) - USANIH National Institute of Allergy & Infectious Diseases (NIAID) [R37AI095983]; French National Research Agency (ANR) under the "Investments for the future" programFrench National Research Agency (ANR) [ANR-10-IAHU-01, ANR13-ISV3-0001-01, ANR-16-CE17-0005-01]; ECOS-NORD [C19S01-63407, SRC2017]; ANRHGDIFDFrench National Research Agency (ANR) [ANR-14-CE15-006-01]; ANR-IFNGPHOX [ANR-13-ISV3-0001-01]; GENMSMD [ANR-16-CE17-0005-01]; Fundacao de Amparo a Pesquisa do Estado de Sao Paulo (FAPESP)Fundacao de Amparo a Pesquisa do Estado de Sao Paulo (FAPESP) [2012/11757-2, 2010/51814-0, 2012/51094-2]; Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPQ)National Council for Scientific and Technological Development (CNPq) [303809/2010-8]; Instituto de Salud Carlos IIIInstituto de Salud Carlos IIIEuropean Commission [PI11/01086, PI14/00405]; European Regional Development Fund (ERDF)European Commission; Colombia-France (ECOS-NORD/COLCIENCIAS/MEN/ICETEX) [619-2013]; Diana Garcia de Olarte foundation PID; ColcienciasDepartamento Administrativo de Ciencia, Tecnologia e Innovacion Colciencias [713-2016, 111574455633]; "Poste d'accueil" INSERMInstitut National de la Sante et de la Recherche Medicale (Inserm); Imagine Institute, The Laboratory of Human Genetics of Infectious Diseases is supported in part by institutional grants from INSERM, University of Paris, The Rockefeller University and the St. Giles Foundation, the National Institute of Allergy and Infectious Diseases (NIAID) of the National Institutes of Health (NIH) (R37AI095983), and grants from the French National Research Agency (ANR) under the "Investments for the future" program (ANR-10-IAHU-01) and IFNGPHOX (ANR13-ISV3-0001-01 for JB and ACN), GENMSMD (ANR-16-CE17-0005-01 for JB) grants, ECOS-NORD (C19S01-63407 for JB and JFR), and SRC2017 (for JB). CO-Q is supported by ANRHGDIFD (ANR-14-CE15-006-01). AG was supported by the ANR-IFNGPHOX (ANR-13-ISV3-0001-01), GENMSMD (ANR-16-CE17-0005-01), and the Imagine Institute. AC-N and EBO-J are supported by Fundacao de Amparo a Pesquisa do Estado de Sao Paulo (FAPESP grants 2012/11757-2, 2010/51814-0, and 2012/51094-2) and Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPQ grant 303809/2010-8). MMG and RC are supported by Instituto de Salud Carlos III, grants PI11/01086 and PI14/00405, co-financed by the European Regional Development Fund (ERDF). JFR and AAA are supported by Colombia-France (ECOS-NORD/COLCIENCIAS/MEN/ICETEX; 619-2013, Diana Garcia de Olarte foundation PID and Colciencias grant 713-2016 #111574455633). JR was supported by "Poste d'accueil" INSERM and Imagine Institute.
- Published
- 2021
24. Inherited PD-1 deficiency underlies tuberculosis and autoimmunity in a child
- Author
-
Phillip Wong, Peng Zhang, Geetha Rao, Gaspard Kerner, Bertrand Boisson, Figen Dogu, Matthew Adamow, Taushif Khan, Ilhan Tezcan, Simon J. Pelham, Samuel C. Williams, Cindy S. Ma, Caner Aytekin, Deniz Cagdas, Mahbuba Rahman, Tasuku Honjo, Masato Ogishi, Jean-François Emile, Franck Rapaport, Jérémie Rosain, Conor Gruber, Leena Kainulainen, Nico Marr, Garrett Allington, Ferda O. Hosnut, Scott Drutman, Jedd D. Wolchok, David Langlais, Yuka Nakajima, Matthew D. Hellmann, Laurent Abel, Mathieu Bourgey, Aydan Ikinciogullari, Philippe Gros, Jacinta Bustamante, Wei-Te Lei, Stuart G. Tangye, Jean-Laurent Casanova, Flore Rozenberg, Richard P. Lifton, Fatima Al Ali, Michael S. Glickman, Maya Chrabieh, Stéphanie Boisson-Dupuis, Luigi D. Notarangelo, V. Koneti Rao, Ottavia M. Delmonte, Margaret K. Callahan, Vivien Béziat, Silvia Vilarinho, Dusan Bogunovic, András N Spaan, Tomonori Yaguchi, Kenji Chamoto, and Rui Yang
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,Male ,STAT3 Transcription Factor ,Tuberculosis ,Programmed Cell Death 1 Receptor ,Hepatosplenomegaly ,Autoimmunity ,CD8-Positive T-Lymphocytes ,medicine.disease_cause ,Interleukin-23 ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Immunity ,RAR-related orphan receptor gamma ,Interferon ,Neoplasms ,Medicine ,Humans ,Child ,Immune Checkpoint Inhibitors ,Intraepithelial Lymphocytes ,business.industry ,Interleukin-6 ,Interleukin ,General Medicine ,Mycobacterium tuberculosis ,Nuclear Receptor Subfamily 1, Group F, Member 3 ,medicine.disease ,CD56 Antigen ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunology ,Immunotherapy ,medicine.symptom ,business ,medicine.drug - Abstract
The pathophysiology of adverse events following programmed cell death protein 1 (PD-1) blockade, including tuberculosis (TB) and autoimmunity, remains poorly characterized. We studied a patient with inherited PD-1 deficiency and TB who died of pulmonary autoimmunity. The patient’s leukocytes did not express PD-1 or respond to PD-1-mediated suppression. The patient’s lymphocytes produced only small amounts of interferon (IFN)-γ upon mycobacterial stimuli, similarly to patients with inborn errors of IFN-γ production who are vulnerable to TB. This phenotype resulted from a combined depletion of Vδ2+ γδ T, mucosal-associated invariant T and CD56bright natural killer lymphocytes and dysfunction of other T lymphocyte subsets. Moreover, the patient displayed hepatosplenomegaly and an expansion of total, activated and RORγT+ CD4−CD8− double-negative αβ T cells, similar to patients with STAT3 gain-of-function mutations who display lymphoproliferative autoimmunity. This phenotype resulted from excessive amounts of STAT3-activating cytokines interleukin (IL)-6 and IL-23 produced by activated T lymphocytes and monocytes, and the STAT3-dependent expression of RORγT by activated T lymphocytes. Our work highlights the indispensable role of human PD-1 in governing both antimycobacterial immunity and self-tolerance, while identifying potentially actionable molecular targets for the diagnostic and therapeutic management of TB and autoimmunity in patients on PD-1 blockade. Dysregulated immune features in a patient with a homozygous loss-of-function mutation in PDCD1 suggest that IL-6, IL-23, STAT3 and RORγT might be potential targets for treatment of PD-1 blockade-induced autoimmunity.
- Published
- 2020
25. Common homozygosity for predicted loss-of-function variants reveals both redundant and advantageous effects of dispensable human genes
- Author
-
Lluis Quintana-Murci, Antoine Favier, Franck Rapaport, David Neil Cooper, Peter D. Stenson, Etienne Patin, Laurent Abel, Jean-Laurent Casanova, Yufei Luo, Marie Lopez, Antonio Rausell, Yoann Seeleuthner, The Clinical Bioinformatics laboratory (Equipe Inserm U1163), Imagine - Institut des maladies génétiques (IHU) (Imagine - U1163), 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)-Université de Paris (UP), Génétique Evolutive Humaine - Human Evolutionary Genetics, Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Human genetics of infectious diseases: Complex predisposition (Equipe Inserm U1163), St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller University [New York], Institute of Medical Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK, Howard Hughes Medical Institute (HHMI), Chaire Génomique humaine et évolution, Collège de France (CdF (institution)), The Laboratory of Clinical Bioinformatics was supported by the French National Research Agency (ANR)'Investissements d’Avenir'Program (Grant ANR-10-IAHU-01) and Christian Dior Couture, Dior. The Laboratory of Human Geneticsof Infectious Diseases was supported, in part, by grants from ANR under the'Investissements d’Avenir'Program (Grant ANR-10-IAHU-01), the Fondation pourla Recherche Médicale (Equipe FRM EQU201903007798), the St. Giles Foundation,and the Rockefeller University. The laboratory of L.Q.-M. is supported by theInstitut Pasteur, the Collège de France, the French Government’s Investissementsd’Avenir program, Laboratoires d’Excellence'Integrative Biology of Emerging Infectious Diseases'(Grant ANR-10-LABX-62-IBEID) and'Milieu Intérieur'(GrantANR-10-LABX-69-22701), and the Fondation pour la Recherche Médicale (EquipeFRM DEQ20180339214). D.N.C. and P.D.S. acknowledge the financial supportof Qiagen Inc. through a license agreement with Cardiff University., ANR-10-IAHU-0001,Imagine,Institut Hospitalo-Universitaire Imagine(2010), ANR-10-LABX-0062,IBEID,Integrative Biology of Emerging Infectious Diseases(2010), ANR-10-LABX-0069,MILIEU INTERIEUR,GENETIC & ENVIRONMENTAL CONTROL OF IMMUNE PHENOTYPE VARIANCE: ESTABLISHING A PATH TOWARDS PERSONALIZED MEDICINE(2010), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Cardiff University, Collège de France - Chaire Génomique humaine et évolution, PATIN, Etienne, Instituts Hospitalo-Universitaires - Institut Hospitalo-Universitaire Imagine - - Imagine2010 - ANR-10-IAHU-0001 - IAHU - VALID, Integrative Biology of Emerging Infectious Diseases - - IBEID2010 - ANR-10-LABX-0062 - LABX - VALID, and Laboratoires d'excellence - GENETIC & ENVIRONMENTAL CONTROL OF IMMUNE PHENOTYPE VARIANCE: ESTABLISHING A PATH TOWARDS PERSONALIZED MEDICINE - - MILIEU INTERIEUR2010 - ANR-10-LABX-0069 - LABX - VALID
- Subjects
Linkage disequilibrium ,Pseudogene ,Population ,MESH: Human Genetics ,[SDV.GEN.GH] Life Sciences [q-bio]/Genetics/Human genetics ,Biology ,03 medical and health sciences ,Negative selection ,0302 clinical medicine ,MESH: Fucosyltransferases ,Loss of Function Mutation ,positive selection ,Apolipoproteins L ,Humans ,MESH: Proteins ,MESH: Genetic Variation ,Allele ,education ,Gene ,Loss function ,Alleles ,030304 developmental biology ,Genetics ,0303 health sciences ,education.field_of_study ,Multidisciplinary ,Sex Chromosomes ,MESH: Humans ,redundancy ,MESH: Alleles ,MESH: Apolipoproteins L ,Homozygote ,Genetic Variation ,Proteins ,negative selection ,MESH: Sex Chromosomes ,Human Genetics ,MESH: Loss of Function Mutation ,Biological Sciences ,Fucosyltransferases ,Phenotype ,loss of function ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,Human genome ,pseudogenization ,030217 neurology & neurosurgery ,MESH: Homozygote - Abstract
Humans homozygous or hemizygous for variants predicted to cause a loss of function of the corresponding protein do not necessarily present with overt clinical phenotypes. However, the set of effectively dispensable genes in the human genome has not yet been fully characterized. We report here 190 autosomal genes with 207 predicted loss-of-function variants, for which the frequency of homozygous individuals exceeds 1% in at least one human population from five major ancestry groups. No such genes were identified on the X and Y chromosomes. Manual curation revealed that 28 variants (15%) had been misannotated as loss-of-function, mainly due to linkage disequilibrium with different compensatory variants. Of the 179 remaining variants in 166 genes (0.82% of 20,232 genes), only 11 alleles in 11 genes had previously been confirmed experimentally to be loss-of-function. The set of 166 dispensable genes was enriched in olfactory receptor genes (41 genes), but depleted of genes expressed in a wide range of organs and in leukocytes. The 125 dispensable non-olfactory receptor genes displayed a relaxation of selective constraints both between species and within humans, consistent with greater redundancy. In total, 62 of these 125 genes were found to be dispensable in at least three human populations, suggesting possible evolution toward pseudogenes. Out of the 179 common loss-of-function variants, 72 could be tested for two neutrality selection statistics, and eight displayed robust signals of positive selection. These variants included the knownFUT2mutant allele conferring resistance to intestinal viruses and anAPOL3variant involved in resistance to parasitic infections. Finally, the 41 dispensable olfactory receptor genes also displayed a strong relaxation of selective constraints similar to that observed for the 341 non-dispensable olfactory receptor genes. Overall, the identification of 166 genes for which a sizeable proportion of humans are homozygous for predicted loss-of-function alleles reveals both redundancies and advantages of such deficiencies for human survival.Significance statementHuman genes homozygous for seemingly loss of function (LoF) variants are increasingly reported in a sizeable proportion of individuals without overt clinical phenotypes. Here, we found 166 genes with 179 predicted LoF variants for which the frequency of homozygous individuals exceeds 1% in at least one of the populations present in databases ExAC and gnomAD. This set of putatively dispensable genes showed relaxation of selective constraints suggesting that a large number of these genes are undergoing pseudogenization. Eight of the common LoF variants displayed robust signals of positive selection including two variants located in genes involved in resistance to infectious diseases. The identification of dispensable genes will allow identifying functions that are, at least nowadays, redundant, or possibly advantageous, for human survival.
- Published
- 2020
- Full Text
- View/download PDF
26. Negative selection on human genes causing severe inborn errors depends on disease outcome and both the mode and mechanism of inheritance
- Author
-
Bertrand Boisson, Lluis Quintana-Murci, Jacinta Bustamante, Anne Gregor, Emmanuelle Jouanguy, Stéphanie Boisson-Dupuis, Anne Puel, Franck Rapaport, Jérémie Rosain, Vivien Béziat, Joseph G. Gleeson, Shen-Ying Zhang, Qian Zhang, Jean-Laurent Casanova, Etienne Patin, and Laurent Abel
- Subjects
Genetics ,0303 health sciences ,education.field_of_study ,Candidate gene ,Mechanism (biology) ,Population ,Inheritance (genetic algorithm) ,Biology ,Penetrance ,03 medical and health sciences ,Negative selection ,0302 clinical medicine ,education ,Haploinsufficiency ,030217 neurology & neurosurgery ,Selection (genetic algorithm) ,030304 developmental biology - Abstract
BackgroundGenetic variants underlying severe diseases are less likely to be transmitted to the next generation, and are thus gradually and selectively eliminated from the population through negative selection. Here, we study the determinants of this evolutionary process in genes underlying severe diseases in humans.ResultsWe propose a novel approach, CoNeS, integrating known negative selection scores through principal component projection. We compare evidence for negative selection at 319 genes underlying inborn errors of immunity (IEI), which are life-threatening monogenic disorders. We find that genes underlying autosomal dominant (AD) or X-linked IEI are under stronger negative selection than those underlying autosomal recessive (AR) IEI, which are under no stronger selection than genes not known to be disease-causing. However, we find that genes with mutations causing AR IEI that are lethal before reproductive maturity and that display complete penetrance are under stronger negative selection than other genes underlying AR IEI. We also find that genes underlying AD IEI by haploinsufficiency are under stronger negative selection than other genes underlying AD IEI. Finally, we replicate these results in a study of 1,140 genes causing inborn errors of neurodevelopment.ConclusionsThese findings collectively show that the clinical outcomes of inborn errors, together with the mode and mechanism of inheritance of these errors, determine the strength of negative selection acting on severe disease-causing genes. These findings suggest that estimating the intensity of negative selection with CoNeS may facilitate the selection of candidate genes in patients suspected to carry an inborn error.
- Published
- 2020
- Full Text
- View/download PDF
27. Classification of microarray data using gene networks.
- Author
-
Franck Rapaport, Andrei Yu. Zinovyev, Marie Dutreix, Emmanuel Barillot, and Jean-Philippe Vert
- Published
- 2007
- Full Text
- View/download PDF
28. JAK2/IDH-mutant–driven myeloproliferative neoplasm is sensitive to combined targeted inhibition
- Author
-
Katharine E. Yen, Julie Teruya-Feldstein, Anna Sophia McKenney, Amritha Varshini Hanasoge Somasundara, Kwok-Kin Wong, April Chiu, Ross L. Levine, Elizaveta Freinkman, Craig B. Thompson, Efthymia Papalexi, Raj Nagaraja, Matthew G. Vander Heiden, Kaitlyn Shank, Mya Steadman, Barbara Spitzer, Jihae Ahn, Andrew M. Intlekofer, Allison N. Lau, Alan H. Shih, Esra A. Akbay, Franck Rapaport, Minal Patel, Raajit K. Rampal, Broad Institute of MIT and Harvard, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Department of Mechanical Engineering, Sloan School of Management, Koch Institute for Integrative Cancer Research at MIT, Lau, Allison N., Shihadeh, Alan, Vander Heiden, Matthew G., Thompson, Craig B., and Levine, Ross L
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Mutant ,Antineoplastic Agents ,Mice, Transgenic ,Biology ,IDH2 ,Epigenesis, Genetic ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Leukemias ,medicine ,Animals ,Humans ,Progenitor cell ,Myeloproliferative neoplasm ,Aged ,Myeloproliferative Disorders ,Hematology ,Gene Expression Profiling ,Stem Cells ,Bone marrow failure ,food and beverages ,Myeloid leukemia ,General Medicine ,Janus Kinase 2 ,Middle Aged ,medicine.disease ,Isocitrate Dehydrogenase ,Mice, Mutant Strains ,3. Good health ,Gene Expression Regulation, Neoplastic ,Phenotype ,030104 developmental biology ,Isocitrate dehydrogenase ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Disease Progression ,Cancer research ,Female ,Drug therapy ,Stem cell ,Corrigendum ,Research Article - Abstract
Patients with myeloproliferative neoplasms (MPNs) frequently progress to bone marrow failure or acute myeloid leukemia (AML), and mutations in epigenetic regulators such as the metabolic enzyme isocitrate dehydrogenase (IDH) are associated with poor outcomes. Here, we showed that combined expression of Jak2V617Fand mutant IDH1R132Hor Idh2R140Q induces MPN progression, alters stem/progenitor cell function, and impairs differentiation in mice. Jak2V617FIdh2R140Q–mutant MPNs were sensitive to small-molecule inhibition of IDH. Combined inhibition of JAK2 and IDH2 normalized the stem and progenitor cell compartments in the murine model and reduced disease burden to a greater extent than was seen with JAK inhibition alone. In addition, combined JAK2 and IDH2 inhibitor treatment also reversed aberrant gene expression in MPN stem cells and reversed the metabolite perturbations induced by concurrent JAK2 and IDH2 mutations. Combined JAK2 and IDH2 inhibitor therapy also showed cooperative efficacy in cells from MPN patients with both JAK2mutand IDH2mutmutations. Taken together, these data suggest that combined JAK and IDH inhibition May offer a therapeutic advantage in this high-risk MPN subtype., Damon Runyon Cancer Research Foundation (DRG-2241-15), Howard Hughes Medical Institute (Faculty Scholars Award), Stand Up To Cancer, National Cancer Institute (U.S.) (P50CA165962), National Cancer Institute (U.S.) (P30CA14051), Koch Institute for Integrative Cancer Research ( Dana-Farber Harvard Cancer Center Bridge Project), Leukemia & Lymphoma Society of America. Specialized Center of Research (SCOR) Program, National Institutes of Health (U.S.) (grant U54OD020355-01), National Institutes of Health (U.S.) (grant NCI R01CA172636), National Institutes of Health (U.S.) (grant R35CA197594), National Cancer Institute (U.S.) (Cancer Center Support Grant (P30 CA008747).
- Published
- 2018
- Full Text
- View/download PDF
29. Targeted genomic analysis of cutaneous T cell lymphomas identifies a subset with aggressive clinicopathological features
- Author
-
Christiane Querfeld, Patricia L. Myskowski, Ahmet Dogan, Marina P. Siakantaris, Franck Rapaport, Alison J. Moskowitz, Kimon V. Argyropoulos, Abhinita Mohanty, Patrizia Mondello, Melissa Pulitzer, Natasha Galasso, Peter C. Louis, Marcel R.M. van den Brink, Steven M. Horwitz, Francesco Maura, and M. Lia Palomba
- Subjects
Male ,Skin Neoplasms ,T cell ,MEDLINE ,Translational research ,Computational biology ,Biology ,lcsh:RC254-282 ,Text mining ,Correspondence ,medicine ,Cancer genomics ,Humans ,Aged ,Extramural ,business.industry ,Hematology ,Genomics ,Middle Aged ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Lymphoma, T-Cell, Cutaneous ,medicine.anatomical_structure ,Oncology ,Clinicopathological features ,Female ,business - Published
- 2020
30. Dual Targeting of Oncogenic Activation and Inflammatory Signaling Increases Therapeutic Efficacy in Myeloproliferative Neoplasms
- Author
-
Noushin Farnoud, Lihua Zou, Bradley E. Bernstein, Peter van Galen, Lauren Dong, Franck Rapaport, Keith B. Cordner, Jun Qi, Matthew Witkin, Richard Koche, Sofie De Groote, Erin McGovern, Juan Medina, Corinne E. Hill, James E. Bradner, Maria Kleppe, Ross L. Levine, Jaime M. Reyes, Efthymia Papalexi, Justin M. Roberts, Raajit K. Rampal, Matthew D. Keller, Julie Teruya-Feldstein, and Amritha Varshini Hanasoge Somasundara
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Mice, Transgenic ,Article ,Proinflammatory cytokine ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Neoplasms ,medicine ,Animals ,Progenitor cell ,Myelofibrosis ,Enhancer ,Protein Kinase Inhibitors ,Regulation of gene expression ,Inflammation ,Myeloproliferative Disorders ,business.industry ,NF-kappa B ,food and beverages ,NF-κB ,Cell Biology ,Janus Kinase 2 ,medicine.disease ,Bromodomain ,030104 developmental biology ,Cytokine ,chemistry ,Gene Expression Regulation ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Cytokines ,business ,Signal Transduction - Abstract
Summary Genetic and functional studies underscore the central role of JAK/STAT signaling in myeloproliferative neoplasms (MPNs). However, the mechanisms that mediate transformation in MPNs are not fully delineated, and clinically utilized JAK inhibitors have limited ability to reduce disease burden or reverse myelofibrosis. Here we show that MPN progenitor cells are characterized by marked alterations in gene regulation through differential enhancer utilization, and identify nuclear factor κB (NF-κB) signaling as a key pathway activated in malignant and non-malignant cells in MPN. Inhibition of BET bromodomain proteins attenuated NF-κB signaling and reduced cytokine production in vivo . Most importantly, combined JAK/BET inhibition resulted in a marked reduction in the serum levels of inflammatory cytokines, reduced disease burden, and reversed bone marrow fibrosis in vivo .
- Published
- 2018
- Full Text
- View/download PDF
31. Human SNORA31 variations impair cortical neuron-intrinsic immunity to HSV-1 and underlie herpes simplex encephalitis
- Author
-
Zobaida Alsum, Soraya Boucherit, Lluis Quintana-Murci, Peng Zhang, Luigi D. Notarangelo, Joseph A. Church, Daxing Gao, Rabih Halwani, Lazaro Lorenzo, Thomas M. Carlile, Maria F. Rojas-Duran, Ahmed Aziz Bousfiha, Yuval Itan, Laurent Abel, Flore Rozenberg, Fabien G. Lafaille, Wendy V. Gilbert, Jean-Laurent Casanova, Vimel Rattina, Marc Tessier-Lavigne, Lorenz Studer, Trine H. Mogensen, Bastian Zimmer, Benoit Henry, Søren R. Paludan, Shen-Ying Zhang, Franck Rapaport, Gregory A. Smith, Saleh Al-Muhsen, Michael J. Ciancanelli, Gaspard Kerner, Jessica L. McAlpine, Kerry Dobbs, Madalina E. Carter-Timofte, Laura Marques, Oliver Harschnitz, Osefame Ewaleifoh, Mary Hasek, Dominik Paquet, Marc Tardieu, Naima Amenzoui, Yoon Seung Lee, Dylan Kwart, Rockefeller University [New York], Memorial Sloane Kettering Cancer Center [New York], Laboratory of Human Genetics of Infectious Diseases (Necker Branch - INSERM U1163), Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Necker - Enfants Malades [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Imagine - Institut des maladies génétiques (IMAGINE - U1163), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Northwestern University Feinberg School of Medicine, Aarhus University Hospital, National Institutes of Health [Bethesda] (NIH), Yale University [New Haven], Service de Virologie [CHU Cochin], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Cochin [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), University of Sharjah, CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Ibn Rochd University Hospital of Casablanca, University Hassan II of Casablanca, Morocco., King Saud University [Riyadh] (KSU), Centro Hospitalar do Porto, University of Southern California (USC), Service de neurologie pédiatrique et maladies métaboliques, Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Robert Debré-Université Paris Diderot - Paris 7 (UPD7), Génétique Evolutive Humaine - Human Evolutionary Genetics, Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Stanford University, This work was funded in part by the National Center for Advancing Translational Sciences, National Institutes of Health (NIH), Clinical and Translational Science Award program (grant nos. UL1TR000043 and UL1TR001866), NIH grants (nos. R01AI088364 to J.L.C. and S.Y.Z., R01NS072381 to J.-L.C. and S.-Y.Z. and R01GM101316 to W.G.), a grant from the Integrative Biology of Emerging Infectious Diseases Laboratory of Excellence (no. ANR-10-LABX-62-IBEID to L.A.) and the French National Research Agency (ANR) under the ‘Investments for the future’ program (no. ANR-10-IAHU-01 to L.A.), the ANR grant IEIHSEER (no. ANR-14-CE14-0008-01 to S.-Y.Z.), the Lundbeck Foundation (grant no. R268-2016-3927 to S.R.P.), the Rockefeller University, INSERM, Paris Descartes University and the St Giles Foundation., ANR-10-LABX-0062,IBEID,Integrative Biology of Emerging Infectious Diseases(2010), ANR-14-CE14-0008,IEIHSEER,L'encéphalite Herpétique de l'enfant résulte de déficits héréditaires d'immunité contre l'HSV-1: une exception ou une règle?(2014), ANR-10-IAHU-0001,Imagine,Institut Hospitalo-Universitaire Imagine(2010), Hôpital Cochin [AP-HP], Service des maladies infectieuses et tropicales [CHU Pitié-Salpêtrière], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), University of Southern California [Los Angeles, CA, USA], Université Paris Diderot - Paris 7 (UPD7)-Hôpital Robert Debré-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), and Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Adult ,Central Nervous System ,Male ,0301 basic medicine ,Intrinsic immunity ,viruses ,Central nervous system ,Herpesvirus 1, Human ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Interferon ,[SDV.MHEP.MI]Life Sciences [q-bio]/Human health and pathology/Infectious diseases ,medicine ,Humans ,RNA, Small Nucleolar ,Genetic Predisposition to Disease ,Small nucleolar RNA ,Neurons ,Immunity ,Infant ,RNA ,General Medicine ,Middle Aged ,3. Good health ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,Child, Preschool ,030220 oncology & carcinogenesis ,Forebrain ,Metagenome ,Female ,Encephalitis, Herpes Simplex ,Small nuclear RNA ,medicine.drug - Abstract
International audience; Herpes simplex virus-1 (HSV-1) encephalitis (HSE) is typically sporadic. Inborn errors of TLR3- and DBR1-mediated central nervous system cell-intrinsic immunity can account for forebrain and brainstem HSE, respectively. We report five unrelated patients with forebrain HSE, each heterozygous for one of four rare variants of SNORA31, encoding a small nucleolar RNA of the H/ACA class that are predicted to direct the isomerization of uridine residues to pseudouridine in small nuclear RNA and ribosomal RNA. We show that CRISPR/Cas9-introduced bi- and monoallelic SNORA31 deletions render human pluripotent stem cell (hPSC)-derived cortical neurons susceptible to HSV-1. Accordingly, SNORA31-mutated patient hPSC-derived cortical neurons are susceptible to HSV-1, like those from TLR3- or STAT1-deficient patients. Exogenous interferon (IFN)-β renders SNORA31- and TLR3- but not STAT1-mutated neurons resistant to HSV-1. Finally, transcriptome analysis of SNORA31-mutated neurons revealed normal responses to TLR3 and IFN-α/β stimulation but abnormal responses to HSV-1. Human SNORA31 thus controls central nervous system neuron-intrinsic immunity to HSV-1 by a distinctive mechanism.
- Published
- 2019
- Full Text
- View/download PDF
32. Chronic mucocutaneous candidiasis and connective tissue disorder in humans with impaired JNK1-dependent responses to IL-17A/F and TGF-β
- Author
-
Marco Ritelli, Harry C. Dietz, Danielle T. Avery, Bertrand Boisson, Soraya Boucherit, Lucie Grodecká, Stuart G. Tangye, Romain Lévy, Kathryn Payne, Tomáš Freiberger, Sophie Cypowyj, Juan Li, Valérie Cormier-Daire, Nicoletta Zoppi, Laurent Abel, Geetha Rao, Vivien Béziat, Andrea Guennoun, Benedetta Bigio, Maya Chrabieh, Salim Bougarn, Marina Colombi, Lei Shang, Emilie Corvilain, Yuval Itan, Anne Puel, Franck Rapaport, Nico Marr, Fransiska Malfait, Delfien Syx, Mélanie Migaud, Tanwir Habib, Sabri Boughorbel, Jean-Laurent Casanova, and Cindy S. Ma
- Subjects
Male ,0301 basic medicine ,Connective Tissue Disorder ,MAPK8 ,Immunology ,Mucocutaneous zone ,Connective tissue ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,Transforming Growth Factor beta ,medicine ,Humans ,Mitogen-Activated Protein Kinase 8 ,Chronic mucocutaneous candidiasis ,Connective Tissue Diseases ,Alleles ,Cells, Cultured ,business.industry ,Candidiasis, Chronic Mucocutaneous ,Interleukin-17 ,General Medicine ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Female ,Signal transduction ,Haploinsufficiency ,business - Abstract
Genetic etiologies of chronic mucocutaneous candidiasis (CMC) disrupt human IL-17A/F-dependent immunity at mucosal surfaces, whereas those of connective tissue disorders (CTDs) often impair the TGF-β-dependent homeostasis of connective tissues. The signaling pathways involved are incompletely understood. We report a three-generation family with an autosomal dominant (AD) combination of CMC and a previously undescribed form of CTD that clinically overlaps with Ehlers-Danlos syndrome (EDS). The patients are heterozygous for a private splice-site variant of MAPK8, the gene encoding c-Jun N-terminal kinase 1 (JNK1), a component of the MAPK signaling pathway. This variant is loss-of-expression and loss-of-function in the patients' fibroblasts, which display AD JNK1 deficiency by haploinsufficiency. These cells have impaired, but not abolished, responses to IL-17A and IL-17F. Moreover, the development of the patients' TH17 cells was impaired ex vivo and in vitro, probably due to the involvement of JNK1 in the TGF-β-responsive pathway and further accounting for the patients' CMC. Consistently, the patients' fibroblasts displayed impaired JNK1- and c-Jun/ATF-2-dependent induction of key extracellular matrix (ECM) components and regulators, but not of EDS-causing gene products, in response to TGF-β. Furthermore, they displayed a transcriptional pattern in response to TGF-β different from that of fibroblasts from patients with Loeys-Dietz syndrome caused by mutations of TGFBR2 or SMAD3, further accounting for the patients' complex and unusual CTD phenotype. This experiment of nature indicates that the integrity of the human JNK1-dependent MAPK signaling pathway is essential for IL-17A- and IL-17F-dependent mucocutaneous immunity to Candida and for the TGF-β-dependent homeostasis of connective tissues.
- Published
- 2019
- Full Text
- View/download PDF
33. Homozygous
- Author
-
Scott B, Drutman, Filomeen, Haerynck, Franklin L, Zhong, David, Hum, Nicholas J, Hernandez, Serkan, Belkaya, Franck, Rapaport, Sarah Jill, de Jong, David, Creytens, Simon J, Tavernier, Katrien, Bonte, Sofie, De Schepper, Jutte, van der Werff Ten Bosch, Lazaro, Lorenzo-Diaz, Andy, Wullaert, Xavier, Bossuyt, Gérard, Orth, Vincent R, Bonagura, Vivien, Béziat, Laurent, Abel, Emmanuelle, Jouanguy, Bruno, Reversade, and Jean-Laurent, Casanova
- Subjects
Keratinocytes ,Male ,Inflammasomes ,Siblings ,Homozygote ,Papillomavirus Infections ,Infant ,NLR Proteins ,Syndrome ,Biological Sciences ,Pedigree ,Child, Preschool ,Gain of Function Mutation ,Cytokines ,Humans ,Female ,Apoptosis Regulatory Proteins ,Respiratory Tract Infections ,Adaptor Proteins, Signal Transducing - Abstract
Juvenile-onset recurrent respiratory papillomatosis (JRRP) is a rare and debilitating childhood disease that presents with recurrent growth of papillomas in the upper airway. Two common human papillomaviruses (HPVs), HPV-6 and -11, are implicated in most cases, but it is still not understood why only a small proportion of children develop JRRP following exposure to these common viruses. We report 2 siblings with a syndromic form of JRRP associated with mild dermatologic abnormalities. Whole-exome sequencing of the patients revealed a private homozygous mutation in NLRP1, encoding Nucleotide-Binding Domain Leucine-Rich Repeat Family Pyrin Domain-Containing 1. We find the NLRP1 mutant allele to be gain of function (GOF) for inflammasome activation, as demonstrated by the induction of inflammasome complex oligomerization and IL-1β secretion in an overexpression system. Moreover, patient-derived keratinocytes secrete elevated levels of IL-1β at baseline. Finally, both patients displayed elevated levels of inflammasome-induced cytokines in the serum. Six NLRP1 GOF mutations have previously been described to underlie 3 allelic Mendelian diseases with differing phenotypes and modes of inheritance. Our results demonstrate that an autosomal recessive, syndromic form of JRRP can be associated with an NLRP1 GOF mutation.
- Published
- 2019
34. Risk of disease progression in low-risk MDS is linked to distinct epigenetic subtypes
- Author
-
Stephen D. Nimer, Maria E. Figueroa, Tingting Qin, Ross L. Levine, Franck Rapaport, Virginia M. Klimek, Raajit K. Rampal, and Jason Sotzen
- Subjects
Risk ,Cancer Research ,Biology ,Article ,Dioxygenases ,Epigenesis, Genetic ,Text mining ,Bone Marrow ,Proto-Oncogene Proteins ,Biomarkers, Tumor ,Humans ,Myelodysplastic syndromes (MDS) ,Epigenetics ,Epigenesis ,Genetics ,DNA methylation ,epigenetics ,Extramural ,business.industry ,Disease progression ,Hematology ,Prognosis ,DNA-Binding Proteins ,Oncology ,CpG site ,Myelodysplastic Syndromes ,low-risk ,Mutation (genetic algorithm) ,Core Binding Factor Alpha 2 Subunit ,Mutation ,Disease Progression ,CpG Islands ,business - Published
- 2019
35. Homozygous NLRP1 gain-of-function mutation in siblings with a syndromic form of recurrent respiratory papillomatosis
- Author
-
Sarah Jill De Jong, Sofie De Schepper, Jean-Laurent Casanova, Jutte van der Werff ten Bosch, David Hum, Emmanuelle Jouanguy, Franklin L. Zhong, Vincent R. Bonagura, Scott Drutman, Nicholas Hernandez, David Creytens, Gérard Orth, Katrien Bonte, Andy Wullaert, Franck Rapaport, Filomeen Haerynck, Laurent Abel, Serkan Belkaya, Xavier Bossuyt, Vivien Béziat, Lazaro Lorenzo-Diaz, Bruno Reversade, Simon Tavernier, Center for Reproductive Medicine, ACS - Diabetes & metabolism, ARD - Amsterdam Reproduction and Development, ACS - Heart failure & arrhythmias, Clinical sciences, Growth and Development, Pediatrics, Reversade, Bruno, Drutman, Scott B., Haerynck, Filomeen, Zhong, Franklin L., Hum, David, Hernandez, Nicholas J., Belkaya, Serkan, Rapaport, Franck, de Jong, Sarah Jill, Creytens, David, Tavernier, Simon J., Bonte, Katrien, De Schepper, Sofie, ten Bosch, Jutte van der Werff, Lorenzo-Diaz, Lazaro, Wullaert, Andy, Bossuyt, Xavier, Orth, Gerard, Bonagura, Vincent R., Beziat, Vivien, Abel, Laurent, Jouanguy, Emmanuelle, Laurent-Casanova, Jean, School of Medicine, and Department of Medical Genetics
- Subjects
Medicine(all) ,Mutation ,Human papillomavirus ,Multidisciplinary ,Medicine ,Medical genetics ,Genetics ,Inflammasome ,NLRP1 ,Recurrent respiratory papillomatosis ,Biology ,medicine.disease_cause ,Phenotype ,Pyrin domain ,symbols.namesake ,inflammasome ,Immunology ,medicine ,Mendelian inheritance ,symbols ,genetics ,Recurrent Respiratory Papillomatosis ,Allele ,Inflammasome complex ,recurrent respiratory papillomatosis ,medicine.drug - Abstract
Juvenile-onset recurrent respiratory papillomatosis (JRRP) is a rare and debilitating childhood disease that presents with recurrent growth of papillomas in the upper airway. Two common human papillomaviruses (HPVs), HPV-6 and -11, are implicated in most cases, but it is still not understood why only a small proportion of children develop JRRP following exposure to these common viruses. We report 2 siblings with a syndromic form of JRRP associated with mild dermatologic abnormalities. Whole-exome sequencing of the patients revealed a private homozygous mutation in NLRP1, encoding Nucleotide-Binding Domain Leucine-Rich Repeat Family Pyrin Domain-Containing 1. We find the NLRP1 mutant allele to be gain of function (GOF) for inflammasome activation, as demonstrated by the induction of inflammasome complex oligomerization and IL-1β secretion in an overexpression system. Moreover, patient-derived keratinocytes secrete elevated levels of IL-1β at baseline. Finally, both patients displayed elevated levels of inflammasome-induced cytokines in the serum. Six NLRP1 GOF mutations have previously been described to underlie 3 allelic Mendelian diseases with differing phenotypes and modes of inheritance. Our results demonstrate that an autosomal recessive, syndromic form of JRRP can be associated with an NLRP1 GOF mutation., United States Department of Health and Human Services; National Institutes of Health (NIH); NIH National Center for Advancing Translational Sciences (NCATS); French National Research Agency (ANR); Integrative Biology of Emerging Infectious Diseases Laboratoire d'Excellence; French Cancer Institute; Strategic Positioning Fund on Genetic Orphan Diseases from A* STAR, Singapore; Jeffrey Modell Foundation; Bijzonder Onderzoeksfonds-Tenure Grant; Cure-AID; European Union ERA-Net for Research Programmes on Rare Diseases; H2020; European Union (European Union); National Research Foundation; St. Giles Foundation; Rockefeller University; Institut National de la Sante et de la Recherche Medicale (Inserm); Paris Descartes University; Shapiro-Silverberg Fund for the Advancement of Translational Research; American Philosophical Society Daland Fellowship in Clinical Investigation; NIH National Center for Research Resources (NCRR)
- Published
- 2019
36. Human IFN-γ immunity to mycobacteria is governed by both IL-12 and IL-23
- Author
-
Cindy S. Ma, Jean-Laurent Casanova, Monika Schmidt, Federico Mele, Gaspard Kerner, James P. Di Santo, Cecilia S. Lindestam Arlehamn, Avneet Heer, Lluis Quintana-Murci, Alessandro Sette, Bernhard Fleckenstein, Tomi Lazarov, Sandra Jovic, Natalie Wong, Stéphanie Boisson-Dupuis, Daniela Latorre, Julia K. Joseph, Bertrand Boisson, Rubén Martínez-Barricarte, Caner Aytekin, Danielle T. Avery, Aydan Ikinciogullari, Figen Dogu, Jean-François Emile, Etienne Patin, Federica Sallusto, Yoann Seeleuthner, Yuval Itan, Laurent Abel, Franck Rapaport, Alejandro Nieto-Patlán, Frederic Geissmann, Satoshi Okada, Fabienne Jabot-Hanin, Stuart G. Tangye, Esther van de Vosse, Geetha Rao, Elissa K. Deenick, Jacinta Bustamante, Mélanie Migaud, Caroline Deswarte, Mohammed Reza Bloursaz, Laura Surace, Benedetta Bigio, Davood Mansouri, Anne Puel, Payam Tabarsi, Xiao-Fei Kong, Gönül Tanır, Vanessa L. Bryant, Noé Ramírez-Alejo, Seyed Alireza Mahdaviani, Janet Markle, St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller University [New York], University of New South Wales [Sydney] (UNSW), Università della Svizzera italiana = University of Italian Switzerland (USI), Shahid Beheshti University of Medical Sciences [Tehran] (SBUMS), Sami Ulus Maternity and Children Training and Research Hospital, The Walter and Eliza Hall Institute of Medical Research (WEHI), University of Melbourne, The Royal Melbourne Hospital, CHU Necker - Enfants Malades [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Imagine - Institut des maladies génétiques (IHU) (Imagine - U1163), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), Immunité Innée - Innate Immunity, Institut Pasteur [Paris] (IP)-Institut National de la Santé et de la Recherche Médicale (INSERM), Icahn School of Medicine at Mount Sinai [New York] (MSSM), Génomique évolutive, modélisation et santé (GEMS), Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Génétique Evolutive Humaine - Human Evolutionary Genetics, Centre de Bioinformatique, Biostatistique et Biologie Intégrative (C3BI), Hiroshima University, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Ankara University School of Medicine [Turkey], Memorial Sloane Kettering Cancer Center [New York], Weill Medical College of Cornell University [New York], King‘s College London, La Jolla Institute for Immunology [La Jolla, CA, États-Unis], University of California [San Diego] (UC San Diego), University of California (UC), Hôpital Ambroise Paré [AP-HP], Leiden University Medical Center (LUMC), Universiteit Leiden, Eidgenössische Technische Hochschule - Swiss Federal Institute of Technology [Zürich] (ETH Zürich), Howard Hughes Medical Institute [New York] (HHMI), Howard Hughes Medical Institute (HHMI)-New York University School of Medicine, NYU System (NYU)-NYU System (NYU)-Rockefeller University [New York]-Columbia University Irving Medical Center (CUIMC), The Laboratory of Human Genetics of Infectious Diseases was supported by grants from the National Institute of Allergy and Infectious Diseases (NIAID) grant numbers 5R37AI095983, R01AI089970, and K99AI127932, the National Center for Research Resources and the National Center for Advancing Sciences (NCATS) of the NIH grant number UL1TR001866, the Rockefeller University, the St. Giles Foundation, the European Research Council (ERC-2010-AdG-268777 and grant no. 323183), Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Descartes University, the Integrative Biology of Emerging Infectious Diseases Laboratory of Excellence (ANR-10-LABX-62-IBEID), and the French National Research Agency (ANR) under the 'Investissement d’avenir' program (grant ANR-10-IAHU-01), ANR-TBPATHGEN (grant ANR-14-CE14-0007-01), ANR-IFNPHOX (grant ANR13-ISV3-0001-01), and ANR-GENMSMD (grant ANR16-CE17-0005-01). This work was supported by NIAID award no. U19AI118626 (to A.S. and F.S.). J.G.M. was funded by the Canadian Institutes of Health Research, the NIH Translational Science Award (CTSA) program (no. UL1 TR000043), the Swiss National Science Foundation (grant no. IZKOZ3_173586), the Charles H. Revson Foundation, and the NIAID (1K99AI127932-01A1). R.M.-B. was supported by the European Molecular Biology Organization (EMBO). N.R.-A. was supported by the National Council of Science and Technology of Mexico (CONACYT, 264011) and the Stony Wold-Herbert Fund Fellowship Grant. Y.I. was supported by the AXA Research Fund. S.G.T., E.K.D., and C.S.M. are supported by research grants and fellowships from the National Health and Medical Research Council of Australia (S.G.T., C.S.M., and E.K.D.) and the Office for Health and Medical Research of the State Government of NSW Australia (C.S.M.). A.S. is supported by NIH research grant HHSN272200900044C. J.B. is supported by SRC2017. The Institute for Research in Biomedicine and F.S. are supported by the Helmut Horten Foundation. S.O. was supported by the Aid for Scientific Research Grant from the Japanese Society for the Promotion of Science (16H05355) and the Practical Research Project for Rare/Intractable Diseases from the Japan Agency for Medical Research and Development., ANR-10-LABX-0062,IBEID,Integrative Biology of Emerging Infectious Diseases(2010), ANR-10-IAHU-0001,Imagine,Institut Hospitalo-Universitaire Imagine(2010), ANR-14-CE14-0007,TBPATHGEN,Dissection de la pathogenèse de la tuberculose par l'identification de défauts monogéniques de l'immunité dans les formes pédiatriques sévères de la maladie(2014), ANR-13-ISV3-0001,IFNGPHOX,L'immunité anti-tuberculeuse dépendante de l'IFN-gamma chez l'homme opère via la NADPH oxydase phagocytaire(2013), ANR-16-CE17-0005,GENMSMD,Dissection génétique de la Susceptibilité Mendélienne aux infections mycobactériennes chez l'homme(2016), European Project: 268777,EC:FP7:ERC,ERC-2010-AdG_20100317,GENTB(2011), European Project: 323183,EC:FP7:ERC,ERC-2012-ADG_20120314,PREDICT(2013), Shahid Beheshti University, Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP), Institut Pasteur [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Génomique évolutive, modélisation et santé (CNRS-UMR2000), Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), University of California, The Rockefeller University, St Giles laboratory of Human Genetics and Infectious Diseases, rockefeller university, Garvan Institute of Medical Research [Darlinghurst, Australia], Dis Training & Res Ctr, Shahid Beheshti University of Medical Sciences, Ecole d'Ingénieurs de Purpan (INPT - EI Purpan), Institut National Polytechnique (Toulouse) (Toulouse INP), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées, Immunité Innée, Imagine - Institut des maladies génétiques (IMAGINE - U1163), Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Centre de Bioinformatique (CBIO), MINES ParisTech - École nationale supérieure des mines de Paris-PSL Research University (PSL), Institut de Génomique d'Evry (IG), Institut de Biologie François JACOB (JACOB), Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay, Sami Ulus Children Hlth & Dis Training & Res Ctr, Ankara, Génétique Humaine des Maladies Infectieuses (Inserm U980), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Descartes - Paris 5 (UPD5), Service d'anatomie pathologique [CHU Necker], CHU Necker - Enfants Malades [AP-HP]-Assistance publique - Hôpitaux de Paris (AP-HP) (APHP), Univ Erlangen Nurnberg, La Jolla Institute for Allergy and Immunology, Laboratoire épidémiologie et oncogénèse des tumeurs digestives, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Garvan Institute for Medical Research, and Institute for Research in Biomedicine
- Subjects
MESH: Interleukin-12 ,0301 basic medicine ,MESH: Interferon-gamma ,MESH: Pedigree ,medicine.medical_treatment ,Immunology ,Population ,Mycobacterium Infections, Nontuberculous ,Biology ,Interleukin-23 ,Article ,Mycobacterium ,Interferon-gamma ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Immunity ,MESH: Mycobacterium ,medicine ,Humans ,Interferon gamma ,education ,Interleukin 12 receptor, beta 1 subunit ,MESH: Interleukin-23 ,education.field_of_study ,MESH: Humans ,General Medicine ,MESH: Mycobacterium Infections, Nontuberculous ,Natural killer T cell ,Interleukin-12 ,Immunity, Innate ,Pedigree ,3. Good health ,030104 developmental biology ,Cytokine ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,Interleukin 12 ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,MESH: Immunity, Innate ,030215 immunology ,medicine.drug - Abstract
International audience; Hundreds of patients with autosomal recessive, complete IL-12p40 or IL-12Rβ1 deficiency have been diagnosed over the last 20 years. They typically suffer from invasive mycobacteriosis and, occasionally, from mucocutaneous candidiasis. Susceptibility to these infections is thought to be due to impairments of IL-12-dependent IFN-γ immunity and IL-23-dependent IL-17A/IL-17F immunity, respectively. We report here patients with autosomal recessive, complete IL-12Rβ2 or IL-23R deficiency, lacking responses to IL-12 or IL-23 only, all of whom, unexpectedly, display mycobacteriosis without candidiasis. We show that αβ T, γδ T, B, NK, ILC1, and ILC2 cells from healthy donors preferentially produce IFN-γ in response to IL-12, whereas NKT cells and MAIT cells preferentially produce IFN-γ in response to IL-23. We also show that the development of IFN-γ-producing CD4 + T cells, including, in particular , mycobacterium-specific TH1* cells (CD45RA− CCR6+), is dependent on both IL-12 and IL-23. Last, we show that IL12RB1, IL12RB2, and IL23R have similar frequencies of deleterious variants in the general population. The comparative rarity of symptomatic patients with IL-12Rβ2 or IL-23R deficiency, relative to IL-12Rβ1 deficiency , is, therefore, due to lower clinical penetrance. There are fewer symptomatic IL-23R-and IL-12Rβ2-deficient than IL-12Rβ1-deficient patients, not because these genetic disorders are rarer, but because the isolated absence of IL-12 or IL-23 is, in part, compensated by the other cytokine for the production of IFN-γ, thereby providing some protection against mycobacteria. These experiments of nature show that human IL-12 and IL-23 are both required for optimal IFN-γ dependent immunity to mycobacteria, both individually and much more so cooperatively.
- Published
- 2018
- Full Text
- View/download PDF
37. Impact of High-Molecular-Risk Mutations on Transplantation Outcomes in Patients with Myelofibrosis
- Author
-
Ross L. Levine, Caroline J McNamara, Damiano Rondelli, Rona Singer Weinberg, Andrew T. Kuykendall, Christopher Famulare, Molly Maloy, Rivka Litvin, Ruben A. Mesa, Rami S. Komrokji, Andrea Arruda, David A. Sallman, Minal Patel, Lonette Sandy, Juan Medina, Amylou C. Dueck, Ronald Hoffman, Vesna Najfeld, Roni Tamari, Franck Rapaport, Hugo Castro-Malaspina, Raajit K. Rampal, Nan Zhang, Vikas Gupta, Sergio Giralt, and John Mascarenhas
- Subjects
Oncology ,Male ,medicine.medical_specialty ,IDH1 ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Article ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Internal medicine ,Medicine ,Humans ,In patient ,Myelofibrosis ,Aged ,Retrospective Studies ,Transplantation ,business.industry ,Hematology ,Middle Aged ,medicine.disease ,Prognosis ,Transplantation outcomes ,Treatment Outcome ,International Prognostic Scoring System ,Primary Myelofibrosis ,030220 oncology & carcinogenesis ,Cohort ,Mutation ,Disease Progression ,Female ,business ,030215 immunology - Abstract
Mutational profiling has demonstrated utility in predicting the likelihood of disease progression in patients with myelofibrosis (MF). However, there is limited data regarding the prognostic utility of genetic profiling in MF patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HCT). We performed high-throughput sequencing of 585 genes on pre-transplant samples from 101 patients with MF who underwent allo-HCT and evaluated the association of mutations and clinical variables with transplantation outcomes. Overall survival (OS) at 5 years post-transplantation was 52%, and relapse-free survival (RFS) was 51.1 % for this cohort. Nonrelapse mortality (NRM) accounted for most deaths. Patient's age, donor's age, donor type, and Dynamic International Prognostic Scoring System score at diagnosis did not predict for outcomes. Mutations known to be associated with increased risk of disease progression, such as ASXL1, SRSF2, IDH1/2, EZH2, and TP53, did not impact OS or RFS. The presence of U2AF1 (P = .007) or DNMT3A (P = .034) mutations was associated with worse OS. A Mutation-Enhanced International Prognostic Scoring System 70 score was available for 80 patients (79%), and there were no differences in outcomes between patients with high risk scores and those with intermediate and low risk scores. Collectively, these data identify mutational predictors of outcome in MF patients undergoing allo-HCT. These genetic biomarkers in conjunction with clinical variables may have important utility in guiding transplantation decision making.
- Published
- 2018
38. CHZ868, a Type II JAK2 Inhibitor, Reverses Type I JAK Inhibitor Persistence and Demonstrates Efficacy in Myeloproliferative Neoplasms
- Author
-
Fabienne Baffert, Nick Socci, Joëlle Rubert, Barbara Spitzer, Michael Zender, Nicolas Ebel, Efthymia Papalexi, Ernesta Dammassa, Anna Sophia McKenney, Ronald Hoffman, Alan H. Shih, William R. Sellers, Ross L. Levine, Emeline Mandon, Rita Andraos, Sophia Chiu, Ke Xu, Raajit K. Rampal, Matthew D. Keller, Dmitry Pankov, Richard J. O'Reilly, Melanie Heinlein, Franck Rapaport, Jihae Ahn, Maria Kleppe, Sara C. Meyer, Francesco Hofmann, Arno Dölemeyer, Priya Koppikar, Masato Murakami, Christoph Gaul, Olga A. Guryanova, Vincent Romanet, Kaitlyn Shank, Thomas Radimerski, Katia Manova, Rona Singer Weinberg, and Agnes Viale
- Subjects
Cancer Research ,Molecular Sequence Data ,Antineoplastic Agents ,medicine.disease_cause ,Article ,Mice ,Myeloproliferative Disorders ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Receptor ,Protein Kinase Inhibitors ,Myeloproliferative neoplasm ,Cell Proliferation ,Mutation ,Janus kinase 2 ,biology ,Sequence Analysis, RNA ,Cell growth ,food and beverages ,Cell Biology ,Janus Kinase 2 ,medicine.disease ,Xenograft Model Antitumor Assays ,3. Good health ,Mice, Inbred C57BL ,Pyrimidines ,Oncology ,Cell culture ,Benzamides ,Immunology ,biology.protein ,Cancer research ,Signal transduction ,Receptors, Thrombopoietin ,Signal Transduction - Abstract
SummaryAlthough clinically tested JAK inhibitors reduce splenomegaly and systemic symptoms, molecular responses are not observed in most myeloproliferative neoplasm (MPN) patients. We previously demonstrated that MPN cells become persistent to type I JAK inhibitors that bind the active conformation of JAK2. We investigated whether CHZ868, a type II JAK inhibitor, would demonstrate activity in JAK inhibitor persistent cells, murine MPN models, and MPN patient samples. JAK2 and MPL mutant cell lines were sensitive to CHZ868, including type I JAK inhibitor persistent cells. CHZ868 showed significant activity in murine MPN models and induced reductions in mutant allele burden not observed with type I JAK inhibitors. These data demonstrate that type II JAK inhibition is a viable therapeutic approach for MPN patients.
- Published
- 2015
- Full Text
- View/download PDF
39. Abstract PD1-4: Somatic leukemogenic mutations associated with infiltrating white blood cells in breast cancer patients
- Author
-
Franck Rapaport, Michael F. Berger, Daoqi You, Agnes Viale, Maria Kleppe, Larry Norton, Robert Benezra, Lennart Bastian, Ross L. Levine, Britta Weigelt, Elizabeth A. Comen, Hannah Wen, Nicolas Socci, Jorge S. Reis-Filho, Brian Blum, Matthew D. Keller, and Edi Brogi
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Myeloid ,business.industry ,Tumor-infiltrating lymphocytes ,Cancer ,medicine.disease_cause ,medicine.disease ,Somatic evolution in cancer ,medicine.anatomical_structure ,Breast cancer ,Oncology ,Cancer cell ,Cancer research ,Medicine ,business ,Carcinogenesis ,Triple-negative breast cancer - Abstract
Background: In the last few decades, theoretical models of cancer growth and progression have long focused on the aberrations of cancer cells alone, such as the abnormal mitotic and invasive characteristics of cancer cells. More recent research across multiple solid tumors suggests a critical interplay between solid tumors and immune regulating cells. Mounting evidence suggests that the immune system can tip the scales of cancer progression, eliciting either an anti-tumor or pro-tumor immune response depending upon varying stimulating and inhibitory factors. Here, we are the first to demonstrate novel mutations including leukemogenic mutations among tumor infiltrating lymphocytes in breast cancer patients. Methods: We obtained 17 primary breast cancer samples from patients who presented for either a lumpectomy or mastectomy as part of an IRB approved biospecimen protocol. Of the 17 patient samples, 13 had triple negative breast cancer, 2 had ER+, HER2+ disease, and 2 had ER+, HER2- disease. In the 17 samples, we used fluorescent activated cell sorting to separate CD45-positive hematopoietic cells from CD45-negative epithelial cells. We then performed exome sequencing of tumor-infiltrating hematopoietic cells to investigate for the presence of pathogenic mutations in tumor-associated leukocytes. In this first step, we identified candidate mutations in known cancer genes, including BCOR, NOTCH2, TET2, NF1, EZH2, and JAK1. As a validation step, we then performed capture-based sequencing of tumor-infiltrating leukocytes in 20 breast cancer samples matched to each patient’s germline DNA sample (buccal swab). In 10 of the 20 patients, we identified and validated somatic mutations. Of note, 6 of these patients harbored mutations known to be associated with leukemia, including DNTM3A, TET2, and BCOR. Most of these mutations were present in at least 5-20% of reads. This suggests that these mutations were present in enriched subclones and were not rare alleles occurring in a minority of hematopoietic stem cells. Lastly, we performed 454 deep sequencing analysis of microdissected tumor DNA samples and confirmed the absence of these mutations in breast cancer cells. Conclusion: Our data demonstrate somatic mutations in tumor infiltrating leukocytes in breast tumors which were not identified in matched germline or tumor DNA samples. Notably, some of these mutations have been implicated in the pathogenesis of lymphoid and myeloid malignancies. This observation suggests a unique relationship between cancer cells and mutant infiltrating leukocytes. We are now investigating the functional interaction between cancer cells and hematopoietic cells. Our findings reframe our understanding of carcinogenesis and offer novel opportunities for cancer detection and treatment. Citation Format: Elizabeth A Comen, Maria Kleppe, Hannah Wen, Britta Weigelt, Lennart Bastian, Brian Blum, Franck T Rapaport, Matt Keller, Nicolas Socci, Agnes Viale, Daoqi You, Robert Benezra, Edi Brogi, Jorge Reis-Filho, Michael Berger, Ross Levine, Larry Norton. Somatic leukemogenic mutations associated with infiltrating white blood cells in breast cancer patients [abstract]. In: Proceedings of the Thirty-Seventh Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2014 Dec 9-13; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2015;75(9 Suppl):Abstract nr PD1-4.
- Published
- 2015
- Full Text
- View/download PDF
40. Mutational Cooperativity Linked to Combinatorial Epigenetic Gain of Function in Acute Myeloid Leukemia
- Author
-
Agnes Viale, Megan A. Hatlen, Magali Cavatore, Cem Meydan, Chen Wei, Martin S. Tallman, Nicholas D. Socci, Todd Hricik, Elisabeth Paietta, Ulrich Steidl, Yanwen Jiang, Brittany Woods, Stephen D. Nimer, Alan H. Shih, Luisa Cimmino, Yongming Sun, Ari Melnick, Ross L. Levine, Alexander Robertson, Suveg Pandey, Iléana Antony-Debré, Iannis Aifantis, Franck Rapaport, Laura Barreyro, Muhamed Baljevic, Elisa de Stanchina, Christopher E. Mason, and Kaitlyn Shank
- Subjects
Cancer Research ,Myeloid ,Population ,Antineoplastic Agents ,Haploinsufficiency ,Biology ,Article ,Dioxygenases ,Epigenesis, Genetic ,fluids and secretions ,hemic and lymphatic diseases ,Proto-Oncogene Proteins ,medicine ,Epigenetics ,Gene Silencing ,Allele ,education ,Regulation of gene expression ,education.field_of_study ,Cytarabine ,Myeloid leukemia ,hemic and immune systems ,Cell Differentiation ,Cell Biology ,DNA Methylation ,medicine.disease ,3. Good health ,DNA-Binding Proteins ,GATA2 Transcription Factor ,Gene Expression Regulation, Neoplastic ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,fms-Like Tyrosine Kinase 3 ,Oncology ,Doxorubicin ,embryonic structures ,DNA methylation ,Mutation ,Cancer research - Abstract
Specific combinations of acute myeloid leukemia (AML) disease alleles, including FLT3 and TET2 mutations, confer distinct biologic features and adverse outcome. We generated mice with mutations in Tet2 and Flt3, which resulted in fully penetrant, lethal AML. Multipotent Tet2(-/-);Flt3(ITD) progenitors (LSK CD48(+)CD150(-)) propagate disease in secondary recipients and were refractory to standard AML chemotherapy and FLT3-targeted therapy. Flt3(ITD) mutations and Tet2 loss cooperatively remodeled DNA methylation and gene expression to an extent not seen with either mutant allele alone, including at the Gata2 locus. Re-expression of Gata2 induced differentiation in AML stem cells and attenuated leukemogenesis. TET2 and FLT3 mutations cooperatively induce AML, with a defined leukemia stem cell population characterized by site-specific changes in DNA methylation and gene expression.
- Published
- 2015
- Full Text
- View/download PDF
41. Varicella-zoster virus CNS vasculitis and RNA polymerase III gene mutation in identical twins
- Author
-
Maibritt Mardahl, Sébastien Fribourg, Madalina E. Carter-Timofte, Carsten Schade Larsen, Trine H. Mogensen, Søren R. Paludan, Jean-Laurent Casanova, Shen-Ying Zhang, Franck Rapaport, Anders F. Hansen, and Mette Christiansen
- Subjects
0301 basic medicine ,viruses ,Gene mutation ,medicine.disease_cause ,Peripheral blood mononuclear cell ,Virus ,Article ,RNA polymerase III ,03 medical and health sciences ,0302 clinical medicine ,Medicine ,Pleocytosis ,Exome sequencing ,030304 developmental biology ,Varicella Zoster Infection ,0303 health sciences ,Mutation ,business.industry ,Varicella zoster virus ,3. Good health ,030104 developmental biology ,Neurology ,Viral replication ,Immunology ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - Abstract
Deficiency in the cytosolic DNA sensor RNA Polymerase III was recently described in children with severe varicella zoster infection in the CNS or lungs. Here we describe a pair of monozygotic female twins, who both experienced severe recurrent CNS vasculitis caused by VZV reactivation. The clinical presentation and findings included recurrent episodes of headache, dizziness, and neurological deficits, cerebrospinal fluid with pleocytosis and intrathecal VZV antibody production, and magnetic resonance scan of the brain showing ischaemic lesions. We performed whole exome sequencing and identified a rare mutation in the Pol III subunit POLR3F. The identified R50W POLR3F mutation is predicted to be damaging by bioinformatics and when tested in functional assays, patient PBMCs exhibited impaired antiviral and inflammatory responses to the PoL III agonist Poly(dA:dT) as well as increased viral replication in patient cells compared to controls. Altogether, these cases add genetic and immunological evidence to the novel association between defects in sensing of AT-rich DNA present in the VZV genome and increased susceptibility to severe manifestations of VZV infection in the CNS in humans.AbbreviationsCADDcombined annotation dependent depletionCSFcerebrospinal fluidDOCKdedicator of cytokinesisIFNGRinterferon gamma receptorMSCmutation significance cut-offNKnatural killerPOLIIIRNA polymerase IIISCIDsevere combined immunodeficiencyTYKtyrosine kinaseVZVvaricella zoster virusWESwhole exome sequencing
- Published
- 2018
- Full Text
- View/download PDF
42. Cooperative Epigenetic Remodeling by TET2 Loss and NRAS Mutation Drives Myeloid Transformation and MEK Inhibitor Sensitivity
- Author
-
Ari Melnick, Sara C. Meyer, Rebecca Maher, Elodie Pronier, Martin S. Tallman, Alan H. Shih, Francine E. Garrett-Bakelman, Hiroyoshi Kunimoto, Franck Rapaport, Abbas Nazir, Justin T. Whitfield, Kaitlyn Shank, Cem Meydan, and Ross L. Levine
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,MAPK/ERK pathway ,Cancer Research ,Myeloid ,Chronic myelomonocytic leukemia ,Mice, Transgenic ,Biology ,Protein Serine-Threonine Kinases ,medicine.disease_cause ,Article ,Dioxygenases ,GTP Phosphohydrolases ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Epigenetics ,Monomeric GTP-Binding Proteins ,Mutation ,Myeloproliferative Disorders ,MEK inhibitor ,Intracellular Signaling Peptides and Proteins ,Membrane Proteins ,Leukemia, Myelomonocytic, Chronic ,medicine.disease ,DNA-Binding Proteins ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Cell Transformation, Neoplastic ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Signal Transduction - Abstract
Summary Mutations in epigenetic modifiers and signaling factors often co-occur in myeloid malignancies, including TET2 and NRAS mutations. Concurrent Tet2 loss and Nras G12D expression in hematopoietic cells induced myeloid transformation, with a fully penetrant, lethal chronic myelomonocytic leukemia (CMML), which was serially transplantable. Tet2 loss and Nras mutation cooperatively led to decrease in negative regulators of mitogen-activated protein kinase (MAPK) activation, including Spry2, thereby causing synergistic activation of MAPK signaling by epigenetic silencing. Tet2/Nras double-mutant leukemia showed preferential sensitivity to MAPK kinase (MEK) inhibition in both mouse model and patient samples. These data provide insights into how epigenetic and signaling mutations cooperate in myeloid transformation and provide a rationale for mechanism-based therapy in CMML patients with these high-risk genetic lesions.
- Published
- 2017
43. Efficacy of ALK5 inhibition in myelofibrosis
- Author
-
Anjali Budhathoki, Cem Murdun, Adam W. Mailloux, Pearlie K. Epling-Burnette, Franck Rapaport, Zhizhuang Joe Zhao, Xiubao Ren, Xuefeng Wang, Lanzhu Yue, Kith Pradhan, Ying Han, Ulrich Steidl, Wanting Tina Ho, Huaquan Wang, Ross L. Levine, Wanke Zhao, Amit Verma, Ling Zhang, Matthias Bartenstein, and Zonghong Shao
- Subjects
0301 basic medicine ,Male ,medicine.medical_treatment ,Cellular differentiation ,Receptor, Transforming Growth Factor-beta Type I ,Mice, Transgenic ,Transforming Growth Factor beta1 ,03 medical and health sciences ,Mice ,Bone Marrow ,Myeloproliferation ,Medicine ,Galunisertib ,Animals ,Humans ,Smad3 Protein ,Myelofibrosis ,Cells, Cultured ,Cell Proliferation ,business.industry ,Mesenchymal stem cell ,Hematopoietic stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,General Medicine ,Janus Kinase 2 ,medicine.disease ,Hematopoietic Stem Cells ,Extramedullary hematopoiesis ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Cytokine ,HEK293 Cells ,Primary Myelofibrosis ,Cancer research ,Quinolines ,Pyrazoles ,Collagen ,business ,Megakaryocytes ,Receptors, Thrombopoietin ,Research Article ,Signal Transduction - Abstract
Myelofibrosis (MF) is a bone marrow disorder characterized by clonal myeloproliferation, aberrant cytokine production, extramedullary hematopoiesis, and bone marrow fibrosis. Although somatic mutations in JAK2, MPL, and CALR have been identified in the pathogenesis of these diseases, inhibitors of the Jak2 pathway have not demonstrated efficacy in ameliorating MF in patients. TGF-β family members are profibrotic cytokines and we observed significant TGF-β1 isoform overexpression in a large cohort of primary MF patient samples. Significant overexpression of TGF-β1 was also observed in murine clonal MPLW515L megakaryocytic cells. TGF-β1 stimulated the deposition of excessive collagen by mesenchymal stromal cells (MSCs) by activating the TGF-β receptor I kinase (ALK5)/Smad3 pathway. MSCs derived from MPLW515L mice demonstrated sustained overproduction of both collagen I and collagen III, effects that were abrogated by ALK5 inhibition in vitro and in vivo. Importantly, use of galunisertib, a clinically active ALK5 inhibitor, significantly improved MF in both MPLW515L and JAK2V617F mouse models. These data demonstrate the role of malignant hematopoietic stem cell (HSC)/TGF-β/MSC axis in the pathogenesis of MF, and provide a preclinical rationale for ALK5 blockade as a therapeutic strategy in MF.
- Published
- 2017
44. Aid is a key regulator of myeloid/erythroid differentiation and DNA methylation in hematopoietic stem/progenitor cells
- Author
-
Abbas Nazir, Hiroyoshi Kunimoto, Franck Rapaport, Kaitlyn Shank, Elodie Pronier, Anna Sophia McKenney, Alan H. Shih, Francine E. Garrett-Bakelman, Ari Melnick, Jayanta Chaudhuri, Ross L. Levine, Benjamin H. Durham, and Cem Meydan
- Subjects
0301 basic medicine ,Regulation of gene expression ,Myeloid ,Myeloid Neoplasia ,Cellular differentiation ,Immunology ,Hematopoietic stem cell ,GATA1 ,hemic and immune systems ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Molecular biology ,Cell biology ,03 medical and health sciences ,Haematopoiesis ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,hemic and lymphatic diseases ,DNA methylation ,CEBPA ,medicine - Abstract
Recent studies have reported that activation-induced cytidine deaminase (AID) and ten-eleven-translocation (TET) family members regulate active DNA demethylation. Genetic alterations of TET2 occur in myeloid malignancies, and hematopoietic-specific loss of Tet2 induces aberrant hematopoietic stem cell (HSC) self-renewal/differentiation, implicating TET2 as a master regulator of normal and malignant hematopoiesis. Despite the functional link between AID and TET in epigenetic gene regulation, the role of AID loss in hematopoiesis and myeloid transformation remains to be investigated. Here, we show that Aid loss in mice leads to expansion of myeloid cells and reduced erythroid progenitors resulting in anemia, with dysregulated expression of Cebpa and Gata1, myeloid/erythroid lineage-specific transcription factors. Consistent with data in the murine context, silencing of AID in human bone marrow cells skews differentiation toward myelomonocytic lineage. However, in contrast to Tet2 loss, Aid loss does not contribute to enhanced HSC self-renewal or cooperate with Flt3-ITD to induce myeloid transformation. Genome-wide transcription and differential methylation analysis uncover the critical role of Aid as a key epigenetic regulator. These results indicate that AID and TET2 share common effects on myeloid and erythroid lineage differentiation, however, their role is nonredundant in regulating HSC self-renewal and in myeloid transformation.
- Published
- 2017
45. Acute myeloid leukemia presenting with panhypopituitarism or diabetes insipidus: a case series with molecular genetic analysis and review of the literature
- Author
-
Peter Kopp, Franck Rapaport, Raajit K. Rampal, Justin Watts, Jessica K. Altman, Elizabeth H. Cull, Ross L. Levine, Martin S. Tallman, and Mark G. Frattini
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Hypopituitarism ,Diagnosis, Differential ,chemistry.chemical_compound ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Genotype ,medicine ,Humans ,neoplasms ,Chromosome 7 (human) ,business.industry ,Hematopoietic Stem Cell Transplantation ,Brain ,Myeloid leukemia ,Hematology ,Middle Aged ,medicine.disease ,Magnetic Resonance Imaging ,Leukemia, Myeloid, Acute ,Leukemia ,Endocrinology ,RUNX1 ,chemistry ,Chromosome 3 ,Mutation ,Diabetes insipidus ,Female ,Chromosomes, Human, Pair 3 ,Chromosome Deletion ,business ,Chromosomes, Human, Pair 7 ,Diabetes Insipidus - Abstract
Central diabetes insipidus (DI) is a rare finding in patients with acute myeloid leukemia (AML), usually occurring in patients with chromosome 3 or 7 abnormalities. We describe four patients with AML and concurrent DI and a fifth patient with AML and panhypopituitarism. Four of five patients had monosomy 7. Three patients had chromosome 3q21q26/EVI-1 gene rearrangements. The molecular genotype of patients with AML and DI is not known. Therefore, we performed gene sequencing of 30 genes commonly mutated in AML in three patients with available leukemia cell DNA. One patient had no identifiable mutations, and two had RUNX1 F158S mutations.
- Published
- 2014
- Full Text
- View/download PDF
46. Abstract 5105: A plug-and-play infrastructure for scalable bioinformatics operations
- Author
-
Juan E. Arango-Ossa, Elli Papaemmanuil, Joseph G. Mccarter, Franck Rapaport, Venkata Yellapantula, Andrew L. Kung, Gunes Gundem, Minal Patel, Juan S. Medina-Martinez, Ross L. Levine, Noushin Farnoud, Gao Teng, Elsa Bernard, Dominik Glodzik, and Max Levine
- Subjects
Cancer Research ,Relational database ,Computer science ,business.industry ,Data management ,Software development ,Linked data ,Bioinformatics ,Metadata ,Oncology ,Scalability ,Information system ,Web application ,business - Abstract
Genome profiling represents a critical pillar for clinical, translational, and basic research studies. Hospitals, core facilities, and research enterprises invest significant resources to generate genomic data sets. Yet, data management and analysis is frequently manual, which demands significant operator time and often results in siloed resources rendering them as single-use assets. Centralization of the genomic capital in a framework that enables automated processing, metadata integration and continuous interrogation maximizes return for investment and serves as the critical catalyst for research innovation, clinical translation and reproducible research. We developed Isabl, a plug-and-play infrastructure for scalable bioinformatics operations. Isabl provides solutions for databasing, assets management, tracking, automated and reproducible data processing. Dynamic reporting and meta-analysis across data assets is enabled. Isabl is built on four main components. First, an individual-centric and extensible relational database with tracking support for samples (temporal, spatial, aliquot), experimental data (assays, platforms, sequencing runs), cohorts (clinical trials, research projects) and versioned bioinformatics applications (assembly aware, tools, results). Second, the database is exposed through a fully featured RESTful API that enables horizontal integration with information systems such as sequencing cores LIMS, variant visualization platforms like cBioPortal, and where applicable, clinical and biospecimen institutional databases. Third, a Software Development Kit (SDK) built for Next Generation Sequencing assets management. The SDK enables automated execution of data import and language-agnostic bioinformatics applications (alignment, variant calling, post-processing) with support for cohort and individual level reporting features. Furthermore, the SDK facilitates dynamic retrieval of results using vertical and horizontal queries (individual and cohort level, respectively). Lastly, Isabl comes with a Single Page Web Application that fosters user interaction with multidisciplinary teams (i.e. researchers, project coordinators, engineers, clinicians) facilitating tracking of analyses, results visualization, and dynamic query processing. Isabl is currently supporting the Memorial Sloan Kettering Genome Pediatrics Precision Medicine Initiative, a prototype platform that delivers integrated, real-time automated reporting of clinical targeted gene re-sequencing, research whole genome and transcriptome profiling data; as well as linked data from pre-clinical models (i.e. PDX) and single cells studies. As an open-source tool, Isabl democratizes access to a purpose built, automated, scalable and fully integrable bioinformatics architecture. Isabl will be available at https://github.com/isabl-io. Citation Format: Juan S. Medina-Martínez, Juan E. Arango-Ossa, Gunes Gundem, Max F. Levine, Minal Patel, Noushin R. Farnoud, Venkata D. Yellapantula, Gao Teng, Joseph G. Mccarter, Elsa Bernard, Franck Rapaport, Dominik Glodzik, Ross L. Levine, Andrew Kung, Elli Papaemmanuil. A plug-and-play infrastructure for scalable bioinformatics operations [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 5105.
- Published
- 2019
- Full Text
- View/download PDF
47. Mutational correlates of response to hypomethylating agent therapy in acute myeloid leukemia
- Author
-
Ross L. Levine, Maria E. Arcila, Martin S. Tallman, Kristina M. Knapp, Shweta Dixit, Virginia M. Klimek, Rami S. Komrokji, Franck Rapaport, Catherine C. Coombs, David A. Sallman, Alan F. List, Abhinita Mohanty, Jeffrey E. Lancet, Raajit K. Rampal, Marcel R.M. van den Brink, Najla Al Ali, Eric Padron, and Sean M. Devlin
- Subjects
Antimetabolites, Antineoplastic ,Myeloid ,Online Only Article ,DNA Methyltransferase 3A ,03 medical and health sciences ,0302 clinical medicine ,Mutation Rate ,Medicine ,Humans ,DNA (Cytosine-5-)-Methyltransferases ,business.industry ,Extramural ,Myeloid leukemia ,Hematology ,DNA Methylation ,medicine.disease ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Hypomethylating agent ,Multicenter study ,030220 oncology & carcinogenesis ,DNA methylation ,Immunology ,Mutation ,Cancer research ,business ,030215 immunology - Published
- 2016
48. Jak1 Integrates Cytokine Sensing to Regulate Hematopoietic Stem Cell Function and Stress Hematopoiesis
- Author
-
Franck Rapaport, Corinne E. Hill, Robert L. Bowman, Maria Kleppe, Matthew D. Keller, Jorge Gandara, Priya Koppikar, Lauren Dong, Christopher E. Mason, Sheng Li, Julie Teruya-Feldstein, Sofie De Groote, Matthew H. Spitzer, Efthymia Papalexi, Garry P. Nolan, and Ross L. Levine
- Subjects
0301 basic medicine ,Myeloid ,Cellular differentiation ,medicine.medical_treatment ,Stem cell factor ,Regenerative Medicine ,Medical and Health Sciences ,Mice ,0302 clinical medicine ,2.1 Biological and endogenous factors ,Myeloid Cells ,Aetiology ,Bone Marrow Transplantation ,Mice, Knockout ,Janus kinase 1 ,Cell Cycle ,Hematopoietic stem cell ,Cell Differentiation ,Hematology ,Biological Sciences ,Cell biology ,Haematopoiesis ,medicine.anatomical_structure ,Cytokine ,Jak1 ,030220 oncology & carcinogenesis ,Interferon Type I ,Molecular Medicine ,Stem Cell Research - Nonembryonic - Non-Human ,cytokine signaling ,Stem cell ,Signal Transduction ,Knockout ,Physiological ,1.1 Normal biological development and functioning ,Biology ,Stress ,Article ,03 medical and health sciences ,Underpinning research ,Stress, Physiological ,Genetics ,medicine ,Animals ,Alleles ,Immunosuppression Therapy ,Inflammatory and immune system ,stress hematopoiesis ,Cell Biology ,Janus Kinase 1 ,Stem Cell Research ,Hematopoietic Stem Cells ,Hematopoiesis ,Enzyme Activation ,030104 developmental biology ,Gene Expression Regulation ,Immunology ,Interleukin-3 ,Developmental Biology - Abstract
JAK1 is a critical effector of pro-inflammatory cytokine signaling and plays important roles in immune function, while abnormal JAK1 activity has been linked to immunological and neoplastic diseases. Specific functions of JAK1 in the context of hematopoiesis, and specifically within hematopoietic stem cells (HSCs), have not clearly been delineated. Here, we show that conditional Jak1 loss in HSCs reduces their self-renewal and markedly alters lymphoid/myeloid differentiation invivo. Jak1-deficient HSCs exhibit decreased competitiveness invivo and are unable to rescue hematopoiesis in the setting of myelosuppression. They exhibit increased quiescence, an inability to enter the cell cycle in response to hematopoietic stress, and a marked reduction in cytokine sensing, including in response to type I interferons and IL-3. Moreover, Jak1 loss is not fully rescued byexpression of a constitutively active Jak2 allele. Together, these data highlight an essential role for Jak1 in HSC homeostasis and stress responses.
- Published
- 2016
49. Increased GVHD-related mortality with broad-spectrum antibiotic use after allogeneic hematopoietic stem cell transplantation in human patients and mice
- Author
-
Silvia Caballero, Franck Rapaport, Hendrik Poeck, Ying Taur, Jennifer Tsai, Marco Calarfiore, Sophia R. Lieberman, Marcel R.M. van den Brink, Camilla Borges Ferreira Gomes, Ke Xu, Katya F. Ahr, Sean M. Devlin, Shannon B. Falconer, Chen Liu, Eric G. Pamer, Robert R. Jenq, George F. Murphy, Kori A. Porosnicu Rodriguez, Lauren F. Young, Jonathan U. Peled, Hillary V. Jay, Alan M. Hanash, Boglarka Gyurkocza, Jyotsna Gupta, Eli L. Moss, Ann E. Slingerland, Jarrod A Dudakov, Suelen M. Perobelli, Odette M. Smith, Ami S. Bhatt, Enrico Velardi, Yusuke Shono, and Melissa D. Docampo
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,medicine.medical_treatment ,Antibiotics ,Cilastatin, Imipenem Drug Combination ,Graft vs Host Disease ,Penicillanic Acid ,Hematopoietic stem cell transplantation ,Aztreonam ,Interleukin-23 ,chemistry.chemical_compound ,Feces ,Mice ,immune system diseases ,polycyclic compounds ,Medicine ,Phylogeny ,biology ,Hematopoietic Stem Cell Transplantation ,General Medicine ,Flow Cytometry ,Anti-Bacterial Agents ,Drug Combinations ,surgical procedures, operative ,Piperacillin, Tazobactam Drug Combination ,Cilastatin ,Female ,Anaerobic bacteria ,Akkermansia muciniphila ,medicine.drug ,medicine.drug_class ,Colon ,Cefepime ,Article ,03 medical and health sciences ,Verrucomicrobia ,Animals ,Humans ,Transplantation, Homologous ,Piperacillin ,business.industry ,biology.organism_classification ,Mucus ,Gastrointestinal Microbiome ,Transplantation ,Mice, Inbred C57BL ,Imipenem ,030104 developmental biology ,chemistry ,Immunology ,business - Abstract
After allogeneic hematopoietic stem cell transplantation (allo-HSCT), intestinal bacteria modulate risks of infection and graft-versus-host disease (GVHD). Neutropenic fever is common and treated with a choice of clinically equivalent antibiotics that target obligately anaerobic bacteria (anaerobes) to varying degrees. We retrospectively examined 857 allo-HSCT recipients and found that treatment of neutropenic fever with imipenem-cilastatin and piperacillin-tazobactam was associated with increased GVHD-related mortality at 5 years (21.5% in imipenem-cilastatin-treated patients vs. 13.1% in untreated patients, p=0.025, and 19.8% in piperacillin-tazobactam-treated patients vs. 11.9% in untreated patients, p=0.007). However, two other antibiotics also used to treat neutropenic fever, aztreonam and cefepime, were not associated with GVHD-related mortality (p=0.78 and p=0.98, respectively). Analysis of stool microbiota composition showed that piperacillin-tazobactam administration was associated with increased compositional perturbation. Studies in mouse models demonstrated similar effects of these antibiotics, as well as aggravated GVHD mortality with imipenem-cilastatin or piperacillin-tazobactm compared to aztreonam (p
- Published
- 2016
50. Distinct evolution and dynamics of epigenetic and genetic heterogeneity in acute myeloid leukemia
- Author
-
Todd Hricik, Richard J D'Andrea, Gail J. Roboz, Luen Bik To, Michael Becker, Selina M. Luger, David Grimwade, Lars Bullinger, Anna L. Brown, Hartmut Döhner, Richard Dillon, Francine E. Garrett-Bakelman, Monica L. Guzman, Ruud Delwel, Konstanze Döhner, Jay P. Patel, Ross L. Levine, Christopher Y. Park, Joy Cannon, Peter J. M. Valk, Franck Rapaport, Bob Löwenberg, Ian D. Lewis, Sheng Li, Mathijs A. Sanders, Martin Carroll, Duane C. Hassane, Stephen S. Chung, Priyanka Vijay, Christopher E. Mason, Donna Neuberg, Alexander E. Perl, Ari Melnick, Hematology, Li, Sheng, Garrett-Bakelman, Francine E, Chung, Stephen S, Sanders, Mathijs A, Brown, Anna L, D'Andrea, Richard J, and Mason, Christopher E
- Subjects
0301 basic medicine ,Adult ,Male ,Tumour heterogeneity ,Biology ,Somatic evolution in cancer ,General Biochemistry, Genetics and Molecular Biology ,Epigenesis, Genetic ,Evolution, Molecular ,03 medical and health sciences ,Cytosine ,Genetic Heterogeneity ,Germline mutation ,Humans ,acute myeloid leukaemia ,Epigenetics ,Cancer epigenetics ,Allele ,Promoter Regions, Genetic ,Alleles ,Proportional Hazards Models ,Genetics ,Genetic heterogeneity ,Gene Expression Regulation, Leukemic ,Sequence Analysis, RNA ,High-Throughput Nucleotide Sequencing ,General Medicine ,Sequence Analysis, DNA ,DNA Methylation ,Middle Aged ,Prognosis ,cancer epigenetics ,3. Good health ,Survival Rate ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Tumor progression ,Multivariate Analysis ,Cancer research ,Disease Progression ,CpG Islands ,Female ,tumour heterogeneity - Abstract
Genetic heterogeneity contributes to clinical outcome and progression of most tumors, but little is known about allelic diversity for epigenetic compartments, and almost no data exist for acute myeloid leukemia (AML). We examined epigenetic heterogeneity as assessed by cytosine methylation within defined genomic loci with four CpGs (epialleles), somatic mutations, and transcriptomes of AML patient samples at serial time points. We observed that epigenetic allele burden is linked to inferior outcome and varies considerably during disease progression. Epigenetic and genetic allelic burden and patterning followed different patterns and kinetics during disease progression. We observed a subset of AMLs with high epiallele and low somatic mutation burden at diagnosis, a subset with high somatic mutation and lower epiallele burdens at diagnosis, and a subset with a mixed profile, suggesting distinct modes of tumor heterogeneity. Genes linked to promoter-associated epiallele shifts during tumor progression showed increased single-cell transcriptional variance and differential expression, suggesting functional impact on gene regulation. Thus, genetic and epigenetic heterogeneity can occur with distinct kinetics likely to affect the biological and clinical features of tumors. Refereed/Peer-reviewed
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.