Network


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

Hotspot


Dive into the research topics where Guillem Clot is active.

Publication


Featured researches published by Guillem Clot.


Nature Genetics | 2012

Epigenomic analysis detects widespread gene-body DNA hypomethylation in chronic lymphocytic leukemia.

Marta Kulis; Simon Heath; Marina Bibikova; Ana C. Queirós; Alba Navarro; Guillem Clot; Alejandra Martínez-Trillos; Giancarlo Castellano; Isabelle Brun-Heath; Magda Pinyol; Sergio Barberán-Soler; Panagiotis Papasaikas; Pedro Jares; Sílvia Beà; Daniel Rico; Simone Ecker; Miriam Rubio; Romina Royo; Vincent T. Ho; Brandy Klotzle; Lluis Hernández; Laura Conde; Mónica López-Guerra; Dolors Colomer; Neus Villamor; Marta Aymerich; María Rozman; Mònica Bayés; Marta Gut; Josep Lluís Gelpí

We have extensively characterized the DNA methylomes of 139 patients with chronic lymphocytic leukemia (CLL) with mutated or unmutated IGHV and of several mature B-cell subpopulations through the use of whole-genome bisulfite sequencing and high-density microarrays. The two molecular subtypes of CLL have differing DNA methylomes that seem to represent epigenetic imprints from distinct normal B-cell subpopulations. DNA hypomethylation in the gene body, targeting mostly enhancer sites, was the most frequent difference between naive and memory B cells and between the two molecular subtypes of CLL and normal B cells. Although DNA methylation and gene expression were poorly correlated, we identified gene-body CpG dinucleotides whose methylation was positively or negatively associated with expression. We have also recognized a DNA methylation signature that distinguishes new clinico-biological subtypes of CLL. We propose an epigenomic scenario in which differential methylation in the gene body may have functional and clinical implications in leukemogenesis.


Nature | 2015

Non-coding recurrent mutations in chronic lymphocytic leukaemia.

Xose S. Puente; Sílvia Beà; Rafael Valdés-Mas; Neus Villamor; Jesús Gutiérrez-Abril; José I. Martín-Subero; Marta Munar; Carlota Rubio-Perez; Pedro Jares; Marta Aymerich; Tycho Baumann; Renée Beekman; Laura Belver; Anna Carrió; Giancarlo Castellano; Guillem Clot; Enrique Colado; Dolors Colomer; Dolors Costa; Julio Delgado; Anna Enjuanes; Xavier Estivill; Adolfo A. Ferrando; Josep Lluís Gelpí; Blanca González; S. Gonzalez; Marcos González; Marta Gut; Jesús María Hernández-Rivas; Mónica López-Guerra

Chronic lymphocytic leukaemia (CLL) is a frequent disease in which the genetic alterations determining the clinicobiological behaviour are not fully understood. Here we describe a comprehensive evaluation of the genomic landscape of 452 CLL cases and 54 patients with monoclonal B-lymphocytosis, a precursor disorder. We extend the number of CLL driver alterations, including changes in ZNF292, ZMYM3, ARID1A and PTPN11. We also identify novel recurrent mutations in non-coding regions, including the 3′ region of NOTCH1, which cause aberrant splicing events, increase NOTCH1 activity and result in a more aggressive disease. In addition, mutations in an enhancer located on chromosome 9p13 result in reduced expression of the B-cell-specific transcription factor PAX5. The accumulative number of driver alterations (0 to ≥4) discriminated between patients with differences in clinical behaviour. This study provides an integrated portrait of the CLL genomic landscape, identifies new recurrent driver mutations of the disease, and suggests clinical interventions that may improve the management of this neoplasia.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Landscape of somatic mutations and clonal evolution in mantle cell lymphoma

Sílvia Beà; Rafael Valdés-Mas; Alba Navarro; Itziar Salaverria; David Martín-García; Pedro Jares; Eva Giné; Magda Pinyol; Cristina Royo; Ferran Nadeu; Laura Conde; Manel Juan; Guillem Clot; Pedro Vizán; Luciano Di Croce; Diana A. Puente; Mónica López-Guerra; Alexandra Moros; Gaël Roué; Marta Aymerich; Neus Villamor; Lluis Colomo; Antonio Martínez; Alexandra Valera; José I. Martín-Subero; Virginia Amador; Luis Hernández; María Rozman; Anna Enjuanes; Pilar Forcada

Significance This is a comprehensive whole-genome/whole-exome analysis of mantle cell lymphoma (MCL). We sequenced 29 MCL cases and validated the findings by target sequencing of 172 additional tumors. We identified recurrent mutations in genes regulating chromatin modification and genes such as NOTCH2 that have a major impact on clinical outcome. Additionally, we demonstrated the subclonal heterogeneity of the tumors already at diagnosis and the modulation of the mutational architecture in the progression of the disease. The identification of new molecular mechanisms may open perspectives for the management of MCL patients. Mantle cell lymphoma (MCL) is an aggressive tumor, but a subset of patients may follow an indolent clinical course. To understand the mechanisms underlying this biological heterogeneity, we performed whole-genome and/or whole-exome sequencing on 29 MCL cases and their respective matched normal DNA, as well as 6 MCL cell lines. Recurrently mutated genes were investigated by targeted sequencing in an independent cohort of 172 MCL patients. We identified 25 significantly mutated genes, including known drivers such as ataxia-telangectasia mutated (ATM), cyclin D1 (CCND1), and the tumor suppressor TP53; mutated genes encoding the anti-apoptotic protein BIRC3 and Toll-like receptor 2 (TLR2); and the chromatin modifiers WHSC1, MLL2, and MEF2B. We also found NOTCH2 mutations as an alternative phenomenon to NOTCH1 mutations in aggressive tumors with a dismal prognosis. Analysis of two simultaneous or subsequent MCL samples by whole-genome/whole-exome (n = 8) or targeted (n = 19) sequencing revealed subclonal heterogeneity at diagnosis in samples from different topographic sites and modulation of the initial mutational profile at the progression of the disease. Some mutations were predominantly clonal or subclonal, indicating an early or late event in tumor evolution, respectively. Our study identifies molecular mechanisms contributing to MCL pathogenesis and offers potential targets for therapeutic intervention.


Cancer Research | 2012

Molecular subsets of mantle cell lymphoma defined by the IGHV mutational status and SOX11 expression have distinct biologic and clinical features

Alba Navarro; Guillem Clot; Cristina Royo; Pedro Jares; Anastasia Hadzidimitriou; Andreas Agathangelidis; Vasilis Bikos; Nikos Darzentas; Theodora Papadaki; Itziar Salaverria; Magda Pinyol; Xavier Puig; Jara Palomero; Maria Carmela Vegliante; Virgina Amador; Alejandra Martínez-Trillos; Lenka Stefancikova; Adrian Wiestner; Wyndham H. Wilson; Christiane Pott; María José Calasanz; Nicola Trim; Wendy N. Erber; Birgitta Sander; German Ott; Andreas Rosenwald; Dolors Colomer; Eva Giné; Reiner Siebert; Armando López-Guillermo

Mantle cell lymphoma (MCL) is a heterogeneous disease with most patients following an aggressive clinical course, whereas others having an indolent behavior. We conducted an integrative and multidisciplinary analysis of 177 MCL to determine whether the immunogenetic features of the clonotypic B-cell receptors (BcR) may identify different subsets of tumors. Truly unmutated (100% identity) IGHV genes were found in 24% cases, 40% were minimally/borderline mutated (99.9%-97%), 19% significantly mutated (96.9%-95%), and 17% hypermutated (<95%). Tumors with high or low mutational load used different IGHV genes, and their gene expression profiles were also different for several gene pathways. A gene set enrichment analysis showed that MCL with high and low IGHV mutations were enriched in memory and naive B-cell signatures, respectively. Furthermore, the highly mutated tumors had less genomic complexity, were preferentially SOX11-negative, and showed more frequent nonnodal disease. The best cut-off of germline identity of IGHV genes to predict survival was 97%. Patients with high and low mutational load had significant different outcome with 5-year overall survival (OS) of 59% and 40%, respectively (P = 0.004). Nodal presentation and SOX11 expression also predicted for poor OS. In a multivariate analysis, IGHV gene status and SOX11 expression were independent risk factors. In conclusion, these observations suggest the idea that MCL with mutated IGHV, SOX11-negativity, and nonnodal presentation correspond to a subtype of the disease with more indolent behavior.


Genome Research | 2014

Transcriptome characterization by RNA sequencing identifies a major molecular and clinical subdivision in chronic lymphocytic leukemia

Pedro G. Ferreira; Pedro Jares; Daniel Rico; Gonzalo Gómez-López; Alejandra Martínez-Trillos; Neus Villamor; Simone Ecker; Abel Gonzalez-Perez; David G. Knowles; Jean Monlong; Rory Johnson; Víctor Quesada; Sarah Djebali; Panagiotis Papasaikas; Mónica López-Guerra; Dolors Colomer; Cristina Royo; Maite Cazorla; Magda Pinyol; Guillem Clot; Marta Aymerich; María Rozman; Marta Kulis; David Tamborero; Anaı̈s Gouin; Julie Blanc; Marta Gut; Ivo Gut; Xose S. Puente; David G. Pisano

Chronic lymphocytic leukemia (CLL) has heterogeneous clinical and biological behavior. Whole-genome and -exome sequencing has contributed to the characterization of the mutational spectrum of the disease, but the underlying transcriptional profile is still poorly understood. We have performed deep RNA sequencing in different subpopulations of normal B-lymphocytes and CLL cells from a cohort of 98 patients, and characterized the CLL transcriptional landscape with unprecedented resolution. We detected thousands of transcriptional elements differentially expressed between the CLL and normal B cells, including protein-coding genes, noncoding RNAs, and pseudogenes. Transposable elements are globally derepressed in CLL cells. In addition, two thousand genes-most of which are not differentially expressed-exhibit CLL-specific splicing patterns. Genes involved in metabolic pathways showed higher expression in CLL, while genes related to spliceosome, proteasome, and ribosome were among the most down-regulated in CLL. Clustering of the CLL samples according to RNA-seq derived gene expression levels unveiled two robust molecular subgroups, C1 and C2. C1/C2 subgroups and the mutational status of the immunoglobulin heavy variable (IGHV) region were the only independent variables in predicting time to treatment in a multivariate analysis with main clinico-biological features. This subdivision was validated in an independent cohort of patients monitored through DNA microarrays. Further analysis shows that B-cell receptor (BCR) activation in the microenvironment of the lymph node may be at the origin of the C1/C2 differences.


Nature Genetics | 2015

Whole-genome fingerprint of the DNA methylome during human B cell differentiation.

Marta Kulis; Angelika Merkel; Simon Heath; Ana C. Queirós; Ronald Schuyler; Giancarlo Castellano; Renée Beekman; Emanuele Raineri; Anna Esteve; Guillem Clot; Néria Verdaguer-Dot; Martí Duran-Ferrer; Nuria Russiñol; Roser Vilarrasa-Blasi; Simone Ecker; Vera Pancaldi; Daniel Rico; Lidia Agueda; Julie Blanc; David C. Richardson; Laura Clarke; Avik Datta; Marien Pascual; Xabier Agirre; Felipe Prosper; Diego Alignani; Bruno Paiva; Gersende Caron; Thierry Fest; Marcus O. Muench

We analyzed the DNA methylome of ten subpopulations spanning the entire B cell differentiation program by whole-genome bisulfite sequencing and high-density microarrays. We observed that non-CpG methylation disappeared upon B cell commitment, whereas CpG methylation changed extensively during B cell maturation, showing an accumulative pattern and affecting around 30% of all measured CpG sites. Early differentiation stages mainly displayed enhancer demethylation, which was associated with upregulation of key B cell transcription factors and affected multiple genes involved in B cell biology. Late differentiation stages, in contrast, showed extensive demethylation of heterochromatin and methylation gain at Polycomb-repressed areas, and genes with apparent functional impact in B cells were not affected. This signature, which has previously been linked to aging and cancer, was particularly widespread in mature cells with an extended lifespan. Comparing B cell neoplasms with their normal counterparts, we determined that they frequently acquire methylation changes in regions already undergoing dynamic methylation during normal B cell differentiation.


Blood | 2013

CCND2 rearrangements are the most frequent genetic events in cyclin D1(-) mantle cell lymphoma.

Itziar Salaverria; Cristina Royo; Alejandra Carvajal-Cuenca; Guillem Clot; Alba Navarro; Alejandra Valera; Joo Y. Song; Renata Woroniecka; Grzegorz Rymkiewicz; Wolfram Klapper; Elena Hartmann; Pierre Sujobert; Iwona Wlodarska; Judith A. Ferry; Philippe Gaulard; German Ott; Andreas Rosenwald; Armando López-Guillermo; Leticia Quintanilla-Martinez; Nancy Lee Harris; Elaine S. Jaffe; Reiner Siebert; Elias Campo; Sílvia Beà

Cyclin D1(-) mantle cell lymphomas (MCLs) are not well characterized, in part because of the difficulties in their recognition. SOX11 has been identified recently as a reliable biomarker of MCL that is also expressed in the cyclin D1(-) variant. We investigated 40 lymphomas with MCL morphology and immunophenotype that were negative for cyclin D1 expression/t(11;14)(q13;q32) but positive for SOX11. These tumors presented clinically with generalized lymphadenopathy, advanced stage, and poor outcome (5-year overall survival, 48%). Chromosomal rearrangements of the CCND2 locus were detected in 55% of the cases, with an IG gene as partner in 18 of 22, in particular with light chains (10 IGK@ and 5 IGL@). No mutations in the phosphorylation motifs of CCND1, CCND2, or CCND3 were detected. The global genomic profile and the high complexity of the 32 cyclin D1(-) SOX11(+) MCL patients analyzed by copy number arrays were similar to the conventional cyclin D1/SOX11 MCL. 17p deletions and high Ki67 expression conferred a significantly worse outcome for the patients. This comprehensive characterization of a large series of cyclin D1(-) MCL patients indicates that these tumors are clinically and biologically similar to the conventional cyclin D1(+) MCL and provides a basis for the proper identification and clinical management of these patients.


Blood | 2013

SOX11 regulates PAX5 expression and blocks terminal B-cell differentiation in aggressive mantle cell lymphoma

Maria Carmela Vegliante; Jara Palomero; Patricia Pérez-Galán; Gaël Roué; Giancarlo Castellano; Alba Navarro; Guillem Clot; Alexandra Moros; Helena Suárez-Cisneros; Sílvia Beà; Luis Hernández; Anna Enjuanes; Pedro Jares; Neus Villamor; Dolors Colomer; José I. Martín-Subero; Elias Campo; Virginia Amador

Mantle cell lymphoma (MCL) is one of the most aggressive lymphoid neoplasms whose pathogenesis is not fully understood. The neural transcription factor SOX11 is overexpressed in most MCL but is not detected in other mature B-cell lymphomas or normal lymphoid cells. The specific expression of SOX11 in MCL suggests that it may be an important element in the development of this tumor, but its potential function is not known. Here, we show that SOX11 promotes tumor growth in a MCL-xenotransplant mouse model. Using chromatin immunoprecipitation microarray analysis combined with gene expression profiling upon SOX11 knockdown, we identify target genes and transcriptional programs regulated by SOX11 including the block of mature B-cell differentiation, modulation of cell cycle, apoptosis, and stem cell development. PAX5 emerges as one of the major SOX11 direct targets. SOX11 silencing downregulates PAX5, induces BLIMP1 expression, and promotes the shift from a mature B cell into the initial plasmacytic differentiation phenotype in both primary tumor cells and an in vitro model. Our results suggest that SOX11 contributes to tumor development by altering the terminal B-cell differentiation program of MCL and provide perspectives that may have clinical implications in the diagnosis and design of new therapeutic strategies.


web science | 2012

Non-nodal type of mantle cell lymphoma is a specific biological and clinical subgroup of the disease

Cristina Royo; Alba Navarro; Guillem Clot; Itziar Salaverria; Eva Giné; Pedro Jares; Dolors Colomer; Adrian Wiestner; Wyndham H. Wilson; Maria Carmela Vegliante; Verònica Fernàndez; Elena Hartmann; Nicola Trim; Wendy N. Erber; Steven H. Swerdlow; Wolfram Klapper; Martin J. S. Dyer; Manuel Vargas-Pabón; German Ott; Andreas Rosenwald; Reiner Siebert; Armando López-Guillermo; Elias Campo; Sílvia Beà

Non-nodal type of mantle cell lymphoma is a specific biological and clinical subgroup of the disease


Leukemia | 2015

A B-cell epigenetic signature defines three biologic subgroups of chronic lymphocytic leukemia with clinical impact.

Ana C. Queirós; Neus Villamor; Guillem Clot; Alejandra Martínez-Trillos; Marta Kulis; Arcadi Navarro; Eva Maria Murga Penas; Sandrine Jayne; Aneela Majid; Julia Richter; Anke K. Bergmann; J Kolarova; Cristina Royo; Nuria Russiñol; Giancarlo Castellano; M Pinyol; Sílvia Beà; Itziar Salaverria; Mónica López-Guerra; Dolors Colomer; M. Aymerich; María Rozman; Julio Delgado; Eva Giné; Marcos González-Díaz; Xose S. Puente; Reiner Siebert; Martin J. S. Dyer; Carlos López-Otín; Rozman C

Prospective identification of patients with chronic lymphocytic leukemia (CLL) destined to progress would greatly facilitate their clinical management. Recently, whole-genome DNA methylation analyses identified three clinicobiologic CLL subgroups with an epigenetic signature related to different normal B-cell counterparts. Here, we developed a clinically applicable method to identify these subgroups and to study their clinical relevance. Using a support vector machine approach, we built a prediction model using five epigenetic biomarkers that was able to classify CLL patients accurately into the three subgroups, namely naive B-cell-like, intermediate and memory B-cell-like CLL. DNA methylation was quantified by highly reproducible bisulfite pyrosequencing assays in two independent CLL series. In the initial series (n=211), the three subgroups showed differential levels of IGHV (immunoglobulin heavy-chain locus) mutation (P<0.001) and VH usage (P<0.03), as well as different clinical features and outcome in terms of time to first treatment (TTT) and overall survival (P<0.001). A multivariate Cox model showed that epigenetic classification was the strongest predictor of TTT (P<0.001) along with Binet stage (P<0.001). These findings were corroborated in a validation series (n=97). In this study, we developed a simple and robust method using epigenetic biomarkers to categorize CLLs into three subgroups with different clinicobiologic features and outcome.

Collaboration


Dive into the Guillem Clot's collaboration.

Top Co-Authors

Avatar

Sílvia Beà

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Elias Campo

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Alba Navarro

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Pedro Jares

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Magda Pinyol

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge