Network


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

Hotspot


Dive into the research topics where Carolina Vicente-Dueñas is active.

Publication


Featured researches published by Carolina Vicente-Dueñas.


The EMBO Journal | 2009

Cancer induction by restriction of oncogene expression to the stem cell compartment

María Pérez-Caro; César Cobaleda; Inés González-Herrero; Carolina Vicente-Dueñas; Camino Bermejo-Rodríguez; Margarita Sánchez-Beato; Alberto Orfao; Belén Pintado; Teresa Flores; Manuel Sánchez-Martín; Rafael Jiménez; Miguel A. Piris; Isidro Sánchez-García

In human cancers, all cancerous cells carry the oncogenic genetic lesions. However, to elucidate whether cancer is a stem cell‐driven tissue, we have developed a strategy to limit oncogene expression to the stem cell compartment in a transgenic mouse setting. Here, we focus on the effects of the BCR‐ABLp210 oncogene, associated with chronic myeloid leukaemia (CML) in humans. We show that CML phenotype and biology can be established in mice by restricting BCR‐ABLp210 expression to stem cell antigen 1 (Sca1)+ cells. The course of the disease in Sca1‐BCR‐ABLp210 mice was not modified on STI571 treatment. However, BCR‐ABLp210‐induced CML is reversible through the unique elimination of the cancer stem cells (CSCs). Overall, our data show that oncogene expression in Sca1+ cells is all that is required to fully reprogramme it, giving rise to a full‐blown, oncogene‐specified tumour with all its mature cellular diversity, and that elimination of the CSCs is enough to eradicate the whole tumour.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Mutations in early follicular lymphoma progenitors are associated with suppressed antigen presentation

Michael R. Green; Shingo Kihira; Chih Long Liu; Ramesh V. Nair; Raheleh Salari; Andrew J. Gentles; Jonathan M. Irish; Henning Stehr; Carolina Vicente-Dueñas; Isabel Romero-Camarero; Isidro Sánchez-García; Sylvia K. Plevritis; Daniel A. Arber; Serafim Batzoglou; Ronald Levy; Ash A. Alizadeh

Significance Follicular lymphoma (FL) is a disease characterized by multiple relapses that are linked by a common progenitor bearing only a subset of the mutations found within the tumor that presents clinically. Inability to cure this disease may therefore be linked to the failure of current therapies to clear these early tumor-propagating clones. Here we further define the genetic hallmarks of this disease and model the steps in evolution through phylogenetic analysis of serial tumor biopsies. This identified CREBBP mutations as early events in genome evolution that are enriched within tumor cell progenitors and provided evidence that these mutations act by allowing immune evasion. This highlights CREBBP mutations as an attractive therapeutic target in FL and provides insight into their pathogenic mechanism. Follicular lymphoma (FL) is incurable with conventional therapies and has a clinical course typified by multiple relapses after therapy. These tumors are genetically characterized by B-cell leukemia/lymphoma 2 (BCL2) translocation and mutation of genes involved in chromatin modification. By analyzing purified tumor cells, we identified additional novel recurrently mutated genes and confirmed mutations of one or more chromatin modifier genes within 96% of FL tumors and two or more in 76% of tumors. We defined the hierarchy of somatic mutations arising during tumor evolution by analyzing the phylogenetic relationship of somatic mutations across the coding genomes of 59 sequentially acquired biopsies from 22 patients. Among all somatically mutated genes, CREBBP mutations were most significantly enriched within the earliest inferable progenitor. These mutations were associated with a signature of decreased antigen presentation characterized by reduced transcript and protein abundance of MHC class II on tumor B cells, in line with the role of CREBBP in promoting class II transactivator (CIITA)-dependent transcriptional activation of these genes. CREBBP mutant B cells stimulated less proliferation of T cells in vitro compared with wild-type B cells from the same tumor. Transcriptional signatures of tumor-infiltrating T cells were indicative of reduced proliferation, and this corresponded to decreased frequencies of tumor-infiltrating CD4 helper T cells and CD8 memory cytotoxic T cells. These observations therefore implicate CREBBP mutation as an early event in FL evolution that contributes to immune evasion via decreased antigen presentation.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Expression of MALT1 oncogene in hematopoietic stem/progenitor cells recapitulates the pathogenesis of human lymphoma in mice

Carolina Vicente-Dueñas; Lorena Fontan; Inés González-Herrero; Isabel Romero-Camarero; Victor Segura; M. Angela Aznar; Esther Alonso-Escudero; Elena Campos-Sanchez; Lucía Ruiz-Roca; Marcos Barajas-Diego; Ainara Sagardoy; Jose I. Martinez-Ferrandis; Fernando Abollo-Jimenez; Cristina Bertolo; Iván Peñuelas; Francisco Javier García-Criado; María Begoña García-Cenador; Thomas Tousseyn; Xabier Agirre; Felipe Prosper; Federico Garcia-Bragado; Ellen D. McPhail; Izidore S. Lossos; Ming-Qing Du; Teresa Flores; Jesús María Hernández-Rivas; Marcos González; Antonio Salar; Beatriz Bellosillo; Eulogio Conde

Chromosomal translocations involving the MALT1 gene are hallmarks of mucosa-associated lymphoid tissue (MALT) lymphoma. To date, targeting these translocations to mouse B cells has failed to reproduce human disease. Here, we induced MALT1 expression in mouse Sca1+Lin− hematopoietic stem/progenitor cells, which showed NF-κB activation and early lymphoid priming, being selectively skewed toward B-cell differentiation. These cells accumulated in extranodal tissues and gave rise to clonal tumors recapitulating the principal clinical, biological, and molecular genetic features of MALT lymphoma. Deletion of p53 gene accelerated tumor onset and induced transformation of MALT lymphoma to activated B-cell diffuse large-cell lymphoma (ABC-DLBCL). Treatment of MALT1-induced lymphomas with a specific inhibitor of MALT1 proteolytic activity decreased cell viability, indicating that endogenous Malt1 signaling was required for tumor cell survival. Our study shows that human-like lymphomas can be modeled in mice by targeting MALT1 expression to hematopoietic stem/progenitor cells, demonstrating the oncogenic role of MALT1 in lymphomagenesis. Furthermore, this work establishes a molecular link between MALT lymphoma and ABC-DLBCL, and provides mouse models to test MALT1 inhibitors. Finally, our results suggest that hematopoietic stem/progenitor cells may be involved in the pathogenesis of human mature B-cell lymphomas.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Identification of cancer initiating cells in K-Ras driven lung adenocarcinoma

Sara Mainardi; Nieves Mijimolle; Sarah Francoz; Carolina Vicente-Dueñas; Isidro Sánchez-García; Mariano Barbacid

Significance K-RAS oncogene-driven lung adenocarcinomas is one of the most malignant human tumors for which there are no efficacious therapeutic strategies. Here, we have used a mouse tumor model that closely recapitulates this human disease to illustrate that adult lung cells are uniquely sensitive to transformation by this oncogene. Monitoring lung cells at the single-cell level revealed that they respond differently to K-Ras oncogenic signals. Whereas K-Ras–expressing Clara cells required an inflammatory response to yield hyperplasias and adenomas, alveolar type II cells or their committed precursors led to the generation of malignant adenocarcinoma regardless of their surrounding microenvironment. Ubiquitous expression of a resident K-RasG12V oncogene in adult mice revealed that most tissues are resistant to K-Ras oncogenic signals. Indeed, K-RasG12V expression only induced overt tumors in lungs. To identify these transformation-permissive cells, we induced K-RasG12V expression in a very limited number of adult lung cells (0.2%) and monitored their fate by X-Gal staining, a surrogate marker coexpressed with the K-RasG12V oncoprotein. Four weeks later, 30% of these cells had proliferated to form small clusters. However, only SPC+ alveolar type II (ATII) cells were able to form hyperplastic lesions, some of which progressed to adenomas and adenocarcinomas. In contrast, induction of K-RasG12V expression in lung cells by intratracheal infection with adenoviral-Cre particles generated hyperplasias in all regions except the proximal airways. Bronchiolar and bronchioalveolar duct junction hyperplasias were primarily made of CC10+ Clara cells. Some of them progressed to form benign adenomas. However, only alveolar hyperplasias, exclusively made up of SPC+ ATII cells, progressed to yield malignant adenocarcinomas. Adenoviral infection induced inflammatory infiltrates primarily made of T and B cells. This inflammatory response was essential for the development of K-RasG12V–driven bronchiolar hyperplasias and adenomas, but not for the generation of SPC+ ATII lesions. Finally, activation of K-RasG12V during embryonic development under the control of a Sca1 promoter yielded CC10+, but not SPC+, hyperplasias, and adenomas. These results, taken together, illustrate that different types of lung cells can generate benign lesions in response to K-Ras oncogenic signals. However, in adult mice, only SPC+ ATII cells were able to yield malignant adenocarcinomas.


The EMBO Journal | 2013

Function of oncogenes in cancer development: a changing paradigm

Carolina Vicente-Dueñas; Isabel Romero-Camarero; César Cobaleda; Isidro Sánchez-García

Tumour‐associated oncogenes induce unscheduled proliferation as well as genomic and chromosomal instability. According to current models, therapeutic strategies that block oncogene activity are likely to selectively target tumour cells. However, recent evidences have revealed that oncogenes are only essential for the proliferation of some specific tumour cell types, but not all. Indeed, the latest studies of the interactions between the oncogene and its target cell have shown that oncogenes contribute to cancer development not only by inducing proliferation but also by developmental reprogramming of the epigenome. This provides the first evidence that tumorigenesis can be initiated by stem cell reprogramming, and uncovers a new role for oncogenes in the origin of cancer. Here we analyse these evidences and propose an updated model of oncogene function that can explain the full range of genotype–phenotype associations found in human cancer. Finally, we discuss how this vision opens new avenues for developing novel anti‐cancer interventions.


The EMBO Journal | 2012

A novel molecular mechanism involved in multiple myeloma development revealed by targeting MafB to haematopoietic progenitors

Carolina Vicente-Dueñas; Isabel Romero-Camarero; Inés González-Herrero; Esther Alonso-Escudero; Fernando Abollo-Jiménez; Xiaoyu Jiang; Norma C. Gutiérrez; Alberto Orfao; Nieves Marín; Luisa M. Villar; Ma Carmen Fernández Criado; Belén Pintado; Teresa Flores; Diego Alonso-López; Javier De Las Rivas; Rafael Jiménez; Francisco Javier García Criado; María Begoña García Cenador; Izidore S. Lossos; César Cobaleda; Isidro Sánchez-García

Understanding the cellular origin of cancer can help to improve disease prevention and therapeutics. Human plasma cell neoplasias are thought to develop from either differentiated B cells or plasma cells. However, when the expression of Maf oncogenes (associated to human plasma cell neoplasias) is targeted to mouse B cells, the resulting animals fail to reproduce the human disease. Here, to explore early cellular changes that might take place in the development of plasma cell neoplasias, we engineered transgenic mice to express MafB in haematopoietic stem/progenitor cells (HS/PCs). Unexpectedly, we show that plasma cell neoplasias arise in the MafB‐transgenic mice. Beyond their clinical resemblance to human disease, these neoplasias highly express genes that are known to be upregulated in human multiple myeloma. Moreover, gene expression profiling revealed that MafB‐expressing HS/PCs were more similar to B cells and tumour plasma cells than to any other subset, including wild‐type HS/PCs. Consistent with this, genome‐scale DNA methylation profiling revealed that MafB imposes an epigenetic program in HS/PCs, and that this program is preserved in mature B cells of MafB‐transgenic mice, demonstrating a novel molecular mechanism involved in tumour initiation. Our findings suggest that, mechanistically, the haematopoietic progenitor population can be the target for transformation in MafB‐associated plasma cell neoplasias.


Seminars in Cancer Biology | 2010

Cancer as a reprogramming-like disease: implications in tumor development and treatment.

Andrés Castellanos; Carolina Vicente-Dueñas; Elena Campos-Sánchez; Juan J. Cruz; Francisco Javier García-Criado; María Begoña García-Cenador; Pedro A. Lazo; Jesus Perez-Losada; Isidro Sánchez-García

Cancer is a clonal malignant disease originated in a single cell and characterized by the accumulation of partially differentiated cells that are phenotypically reminiscent of normal stages of differentiation. Given the fact that human cancer is diagnosed at later stages and cannot be monitored during its natural evolution, the origin of tumors has been a subject of continuing discussion. Animal models provide a means to determine the identity of the cell-of-origin leading to malignancy and to develop new treatments. Recent findings in mice have shown that cancer stem cells could arise through a reprogramming-like mechanism, suggesting that genetic lesions that initiate the cancer process might be dispensable for tumor progression and maintenance. This review addresses the impact of these results toward a better understanding of carcinogenesis and proposes research avenues for tackling these issues in the future.


Cancer Discovery | 2015

Infection Exposure Is a Causal Factor in B-cell Precursor Acute Lymphoblastic Leukemia as a Result of Pax5-Inherited Susceptibility.

Alberto Martín-Lorenzo; Julia Hauer; Carolina Vicente-Dueñas; Franziska Auer; Inés González-Herrero; Idoia García-Ramírez; Sebastian Ginzel; Ralf Thiele; Stefan N. Constantinescu; Christoph Bartenhagen; Martin Dugas; Michael Gombert; Daniel Schäfer; Oscar Blanco; Andrea Mayado; Alberto Orfao; Diego Alonso-López; Javier De Las Rivas; Cesar Cobaleda; María Begoña García-Cenador; Francisco Javier García-Criado; Isidro Sánchez-García; Arndt Borkhardt

UNLABELLED Earlier in the past century, infections were regarded as the most likely cause of childhood B-cell precursor acute lymphoblastic leukemia (pB-ALL). However, there is a lack of relevant biologic evidence supporting this hypothesis. We present in vivo genetic evidence mechanistically connecting inherited susceptibility to pB-ALL and postnatal infections by showing that pB-ALL was initiated in Pax5 heterozygous mice only when they were exposed to common pathogens. Strikingly, these murine pB-ALLs closely resemble the human disease. Tumor exome sequencing revealed activating somatic, nonsynonymous mutations of Jak3 as a second hit. Transplantation experiments and deep sequencing suggest that inactivating mutations in Pax5 promote leukemogenesis by creating an aberrant progenitor compartment that is susceptible to malignant transformation through accumulation of secondary Jak3 mutations. Thus, treatment of Pax5(+/-) leukemic cells with specific JAK1/3 inhibitors resulted in increased apoptosis. These results uncover the causal role of infection in pB-ALL development. SIGNIFICANCE These results demonstrate that delayed infection exposure is a causal factor in pB-ALL. Therefore, these findings have critical implications for the understanding of the pathogenesis of leukemia and for the development of novel therapies for this disease.


Nature Communications | 2013

Germinal centre protein HGAL promotes lymphoid hyperplasia and amyloidosis via BCR-mediated Syk activation

Isabel Romero-Camarero; Xiaoyu Jiang; Yasodha Natkunam; Xiaoqing Lu; Carolina Vicente-Dueñas; Inés González-Herrero; Teresa Flores; Juan L. García; George McNamara; Christian A. Kunder; Shuchun Zhao; Victor Segura; Lorena Fontan; Jose A. Martinez-Climent; Francisco Javier García-Criado; Jason D. Theis; Ahmet Dogan; Elena Campos-Sanchez; Michael R. Green; Ash A. Alizadeh; César Cobaleda; Isidro Sánchez-García; Izidore S. Lossos

The human germinal centre associated lymphoma (HGAL) gene is specifically expressed in germinal centre B-lymphocytes and germinal centre-derived B-cell lymphomas, but its function is largely unknown. Here we demonstrate that HGAL directly binds Syk in B-cells, increases its kinase activity upon B-cell receptor stimulation and leads to enhanced activation of Syk downstream effectors. To further investigate these findings in vivo, HGAL transgenic mice were generated. Starting from 12 months of age these mice developed polyclonal B-cell lymphoid hyperplasia, hypergammaglobulinemia and systemic reactive AA amyloidosis, leading to shortened survival. The lymphoid hyperplasia in the HGAL transgenic mice are likely attributable to enhanced B-cell receptor signalling as shown by increased Syk phosphorylation, ex vivo B-cell proliferation and increased RhoA activation. Overall, our study shows for the first time that the germinal centre protein HGAL regulates B-cell receptor signalling in B-lymphocytes which, without appropriate control, may lead to B-cell lymphoproliferation.


Cell Cycle | 2012

Loss of p53 exacerbates multiple myeloma phenotype by facilitating the reprogramming of hematopoietic stem/progenitor cells to malignant plasma cells by MafB

Carolina Vicente-Dueñas; Inés González-Herrero; María Begoña García Cenador; Francisco Javier García Criado; Isidro Sánchez-García

Multiple myeloma (MM) is a serious, mostly incurable human cancer of malignant plasma cells. Chromosomal translocations affecting MAFB are present in a significant percentage of multiple myeloma patients. Genetically engineered Sca1-MafB mice, in which MafB expression is limited to hematopoietic stem/progenitor cells (HS/P-Cs), display the phenotypic features of MM. Contrary to many other types of cancer, it is not yet known if the p53 gene plays any essential role in the pathogenesis of this disease. Here, we show, taking advantage of the Sca1-MafB MM mouse model, that loss of p53 does not rescue the multiple myeloma disease, but instead accelerates its development and exacerbates the MM phenotype. Therefore, the efficiency of the MafB-induced MM reprogramming of normal HS/P-Cs to terminally differentiated malignant plasma cells is enhanced by p53 deficiency, in analogy to what happens in reprogramming to pluripotency. These results raise caution about interfering with p53 function when treating multiple myeloma.

Collaboration


Dive into the Carolina Vicente-Dueñas's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

César Cobaleda

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Oscar Blanco

University of Salamanca

View shared research outputs
Top Co-Authors

Avatar

Arndt Borkhardt

University of Düsseldorf

View shared research outputs
Researchain Logo
Decentralizing Knowledge