Genomic and epigenomic insights into the origin, pathogenesis, and clinical behavior of mantle cell lymphoma subtypes

التفاصيل البيبلوغرافية
العنوان: Genomic and epigenomic insights into the origin, pathogenesis, and clinical behavior of mantle cell lymphoma subtypes
المؤلفون: Xose S. Puente, Ana Muntañola, Ander Diaz-Navarro, José I. Martín-Subero, Blanca Espinet, Roser Vilarrasa-Blasi, Guillem Clot, Cristina López, Marta Kulis, Ferran Nadeu, Rafael Valdés-Mas, Vicente Chapaprieta, Marta Aymerich, Dolors Costa, Elias Campo, David Torrents, Sílvia Beà, Jesús Gutiérrez-Abril, Armando López-Guillermo, David Martín-García, Pedro Jares, Ralf Küppers, Eva Giné, Inmaculada Ribera-Cortada, Montserrat Puiggròs, Giancarlo Castellano, Dolors Colomer, Romina Royo, Martí Duran-Ferrer, Alba Navarro, Reiner Siebert
المصدر: Blood. 136:1419-1432
بيانات النشر: American Society of Hematology, 2020.
سنة النشر: 2020
مصطلحات موضوعية: Adult, Male, Immunology, Medizin, Immunoglobulins, Lymphoma, Mantle-Cell, Biology, Biochemistry, Epigenesis, Genetic, Transcriptome, medicine, Humans, Cyclin D1, Epigenetics, Aged, Cell Proliferation, Epigenomics, Aged, 80 and over, Gene Rearrangement, Regulation of gene expression, Genomics, Cell Biology, Hematology, Gene rearrangement, DNA Methylation, Middle Aged, medicine.disease, Chromatin, Gene Expression Regulation, Neoplastic, Mutation, DNA methylation, Cancer research, Female, Mantle cell lymphoma
الوصف: Mantle cell lymphoma (MCL) is a mature B-cell neoplasm initially driven by CCND1 rearrangement with 2 molecular subtypes, conventional MCL (cMCL) and leukemic non-nodal MCL (nnMCL), that differ in their clinicobiological behavior. To identify the genetic and epigenetic alterations determining this diversity, we used whole-genome (n = 61) and exome (n = 21) sequencing (74% cMCL, 26% nnMCL) combined with transcriptome and DNA methylation profiles in the context of 5 MCL reference epigenomes. We identified that open and active chromatin at the major translocation cluster locus might facilitate the t(11;14)(q13;32), which modifies the 3-dimensional structure of the involved regions. This translocation is mainly acquired in precursor B cells mediated by recombination-activating genes in both MCL subtypes, whereas in 8% of cases the translocation occurs in mature B cells mediated by activation-induced cytidine deaminase. We identified novel recurrent MCL drivers, including CDKN1B, SAMHD1, BCOR, SYNE1, HNRNPH1, SMARCB1, and DAZAP1. Complex structural alterations emerge as a relevant early oncogenic mechanism in MCL, targeting key driver genes. Breakage-fusion-bridge cycles and translocations activated oncogenes (BMI1, MIR17HG, TERT, MYC, and MYCN), generating gene amplifications and remodeling regulatory regions. cMCL carried significant higher numbers of structural variants, copy number alterations, and driver changes than nnMCL, with exclusive alterations of ATM in cMCL, whereas TP53 and TERT alterations were slightly enriched in nnMCL. Several drivers had prognostic impact, but only TP53 and MYC aberrations added value independently of genomic complexity. An increasing genomic complexity, together with the presence of breakage-fusion-bridge cycles and high DNA methylation changes related to the proliferative cell history, defines patients with different clinical evolution.
تدمد: 1528-0020
0006-4971
URL الوصول: https://explore.openaire.eu/search/publication?articleId=doi_dedup___::bbc500617eb285de60b0c605c689d1b3
https://doi.org/10.1182/blood.2020005289
حقوق: OPEN
رقم الأكسشن: edsair.doi.dedup.....bbc500617eb285de60b0c605c689d1b3
قاعدة البيانات: OpenAIRE