Network


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

Hotspot


Dive into the research topics where Laura Ciudad is active.

Publication


Featured researches published by Laura Ciudad.


Blood | 2010

Immunomodulatory effect of 5-azacytidine (5-azaC): potential role in the transplantation setting

Luis Ignacio Sánchez-Abarca; Silvia Gutierrez-Cosio; Carlos Santamaría; Teresa Caballero-Velázquez; B. Blanco; Carmen Herrero-Sánchez; Juan L. Garcia; Soraya Carrancio; Pilar Hernandez-Campo; Francisco Javier Gonzalez; Teresa Flores; Laura Ciudad; Esteban Ballestar; Consuelo del Cañizo; Jesús F. San Miguel; José A. Pérez-Simón

Cytokine genes are targets of multiple epigenetic mechanisms in T lymphocytes. 5-azacytidine (5-azaC) is a nucleoside-based DNA methyltransferase inhibitor that induces demethylation and gene reactivation. In the current study, we analyzed the effect of 5-azaC in T-cell function and observed that 5-azaC inhibits T-cell proliferation and activation, blocking cell cycle in the G(0) to G(1) phase and decreasing the production of proinflammatory cytokines such as tumor necrosis factor-alpha and interferon-gamma. This effect was not attributable to a proapoptotic effect of the drug but to the down-regulation of genes involved in T-cell cycle progression and activation such as CCNG2, MTCP1, CD58, and ADK and up-regulation of genes that induce cell-growth arrest, such as DCUN1D2, U2AF2, GADD45B, or p53. A longer exposure to the drug leads to demethylation of FOXP3 promoter, overexpression of FOXP3, and expansion of regulatory T cells. Finally, the administration of 5-azaC after transplantation prevented the development of graft-versus-host disease, leading to a significant increase in survival in a fully mismatched bone marrow transplantation mouse model. In conclusion, the current study shows the effect of 5-azaC in T lymphocytes and illustrates its role in the allogeneic transplantation setting as an immunomodulatory drug, describing new pathways that must be explored to prevent graft-versus-host disease.


Nucleic Acids Research | 2012

Pre-B cell to macrophage transdifferentiation without significant promoter DNA methylation changes

Javier Rodríguez-Ubreva; Laura Ciudad; David Gomez-Cabrero; Maribel Parra; Lars H. Bussmann; Alessandro di Tullio; Eric M. Kallin; Jesper Tegnér; Thomas Graf; Esteban Ballestar

Transcription factor-induced lineage reprogramming or transdifferentiation experiments are essential for understanding the plasticity of differentiated cells. These experiments helped to define the specific role of transcription factors in conferring cell identity and played a key role in the development of the regenerative medicine field. We here investigated the acquisition of DNA methylation changes during C/EBPα-induced pre-B cell to macrophage transdifferentiation. Unexpectedly, cell lineage conversion occurred without significant changes in DNA methylation not only in key B cell- and macrophage-specific genes but also throughout the entire set of genes differentially methylated between the two parental cell types. In contrast, active and repressive histone modification marks changed according to the expression levels of these genes. We also demonstrated that C/EBPα and RNA Pol II are associated with the methylated promoters of macrophage-specific genes in reprogrammed macrophages without inducing methylation changes. Our findings not only provide insights about the extent and hierarchy of epigenetic events in pre-B cell to macrophage transdifferentiation but also show an important difference to reprogramming towards pluripotency where promoter DNA demethylation plays a pivotal role.


Molecular Cancer Research | 2011

Long-Range Epigenetic Silencing Associates with Deregulation of Ikaros Targets in Colorectal Cancer Cells

Biola M. Javierre; Javier Rodríguez-Ubreva; Fatima Al-Shahrour; Marina Corominas; Osvaldo Graña; Laura Ciudad; Xabier Agirre; David G. Pisano; Alfonso Valencia; Jose Roman-Gomez; María José Calasanz; Felipe Prosper; Manel Esteller; Rogelio González-Sarmiento; Esteban Ballestar

Transcription factors are common targets of epigenetic inactivation in human cancer. Promoter hypermethylation and subsequent silencing of transcription factors can lead to further deregulation of their targets. In this study, we explored the potential epigenetic deregulation in cancer of Ikaros family genes, which code for essential transcription factors in cell differentiation and exhibit genetic defects in hematologic neoplasias. Unexpectedly, our analysis revealed that Ikaros undergoes very specific promoter hypermethylation in colorectal cancer, including in all the cell lines studied and around 64% of primary colorectal adenocarcinomas, with increasing proportions in advanced Dukes stages. Ikaros hypermethylation occurred in the context of a novel long-range epigenetic silencing (LRES) region. Reintroduction of Ikaros in colorectal cancer cells, ChIP-chip analysis, and validation in primary samples led us to identify a number of direct targets that are possibly related with colorectal cancer progression. Our results not only provide the first evidence that LRES can have functional specific effects in cancer but also identify several deregulated Ikaros targets that may contribute to progression in colorectal adenocarcinoma. Mol Cancer Res; 9(8); 1139–51. ©2011 AACR.


Nature Communications | 2015

Monozygotic twins discordant for common variable immunodeficiency reveal impaired DNA demethylation during naïve-to-memory B-cell transition

Virginia C. Rodríguez-Cortez; Lucia del Pino-Molina; Javier Rodríguez-Ubreva; Laura Ciudad; David Gomez-Cabrero; José M. Urquiza; Jesper Tegnér; Carlos Rodríguez-Gallego; Eduardo López-Granados; Esteban Ballestar

Common variable immunodeficiency (CVID), the most frequent primary immunodeficiency characterized by loss of B-cell function, depends partly on genetic defects, and epigenetic changes are thought to contribute to its aetiology. Here we perform a high-throughput DNA methylation analysis of this disorder using a pair of CVID-discordant MZ twins and show predominant gain of DNA methylation in CVID B cells with respect to those from the healthy sibling in critical B lymphocyte genes, such as PIK3CD, BCL2L1, RPS6KB2, TCF3 and KCNN4. Individual analysis confirms hypermethylation of these genes. Analysis in naive, unswitched and switched memory B cells in a CVID patient cohort shows impaired ability to demethylate and upregulate these genes in transitioning from naive to memory cells in CVID. Our results not only indicate a role for epigenetic alterations in CVID but also identify relevant DNA methylation changes in B cells that could explain the clinical manifestations of CVID individuals.


Genome Biology | 2015

NF-κB-direct activation of microRNAs with repressive effects on monocyte-specific genes is critical for osteoclast differentiation

Lorenzo de la Rica; Antonio Garcia-Gomez; Natalia R. Comet; Javier Rodríguez-Ubreva; Laura Ciudad; Roser Vento-Tormo; Damiana Álvarez-Errico; Mireia García; Carmen Gómez-Vaquero; Esteban Ballestar

BackgroundMonocyte-to-osteoclast conversion is a unique terminal differentiation process that is exacerbated in rheumatoid arthritis and bone metastasis. The mechanisms implicated in upregulating osteoclast-specific genes involve transcription factors, epigenetic regulators and microRNAs (miRNAs). It is less well known how downregulation of osteoclast-inappropriate genes is achieved.ResultsIn this study, analysis of miRNA expression changes in osteoclast differentiation from human primary monocytes revealed the rapid upregulation of two miRNA clusters, miR-212/132 and miR-99b/let-7e/125a. We demonstrate that they negatively target monocyte-specific and immunomodulatory genes like TNFAIP3, IGF1R and IL15. Depletion of these miRNAs inhibits osteoclast differentiation and upregulates their targets. These miRNAs are also upregulated in other inflammatory monocytic differentiation processes. Most importantly, we demonstrate for the first time the direct involvement of Nuclear Factor kappa B (NF-κB) in the regulation of these miRNAs, as well as with their targets, whereby NF-κB p65 binds the promoters of these two miRNA clusters and NF-κB inhibition or depletion results in impaired upregulation of their expression.ConclusionsOur results reveal the direct involvement of NF-κB in shutting down certain monocyte-specific genes, including some anti-inflammatory activities, through a miRNA-dependent mechanism for proper osteoclast differentiation.


Molecular and Cellular Biology | 2014

C/EBPa-Mediated Activation of MicroRNAs 34a and 223 Inhibits Lef1 Expression To Achieve Efficient Reprogramming into Macrophages

Javier Rodríguez-Ubreva; Laura Ciudad; Chris van Oevelen; Maribel Parra; Thomas Graf; Esteban Ballestar

ABSTRACT MicroRNAs (miRNAs) exert negative effects on gene expression and influence cell lineage choice during hematopoiesis. C/EBPa-induced pre-B cell-to-macrophage transdifferentiation provides an excellent model to investigate the contribution of miRNAs to hematopoietic cell identity, especially because the two cell types involved fall into separate lymphoid and myeloid branches. In this process, efficient repression of the B cell-specific program is essential to ensure transdifferentation and macrophage function. miRNA profiling revealed that upregulation of miRNAs is highly predominant compared with downregulation and that C/EBPa directly regulates several upregulated miRNAs. We also determined that miRNA 34a (miR-34a) and miR-223 sharply accelerate C/EBPa-mediated transdifferentiation, whereas their depletion delays this process. These two miRNAs affect the transdifferentiation efficiency and activity of macrophages, including their lipopolysaccharide (LPS)-dependent inflammatory response. miR-34a and miR-223 directly target and downregulate the lymphoid transcription factor Lef1, whose ectopic expression delays transdifferentiation to an extent similar to that seen with miR-34a and miR-223 depletion. In addition, ectopic introduction of Lef1 in macrophages causes upregulation of B cell markers, including CD19, Pax5, and Ikzf3. Our report demonstrates the importance of these miRNAs in ensuring the erasure of key B cell transcription factors, such as Lef1, and reinforces the notion of their essential role in fine-tuning the control required for establishing cell identity.


Scientific Reports | 2017

Activation-induced cytidine deaminase targets SUV4-20-mediated histone H4K20 trimethylation to class-switch recombination sites

Virginia C. Rodríguez-Cortez; Paloma Martínez-Redondo; Francesc Català-Moll; Javier Rodríguez-Ubreva; Antonio Garcia-Gomez; Ganesh Poorani-Subramani; Laura Ciudad; Henar Hernando; Arantxa Pérez-García; José M. Urquiza; Almudena R. Ramiro; Javier M. Di Noia; Alejandro Vaquero; Esteban Ballestar

Activation-induced cytidine deaminase (AID) triggers antibody diversification in B cells by catalysing deamination and subsequently mutating immunoglobulin (Ig) genes. Association of AID with RNA Pol II and occurrence of epigenetic changes during Ig gene diversification suggest participation of AID in epigenetic regulation. AID is mutated in hyper-IgM type 2 (HIGM2) syndrome. Here, we investigated the potential role of AID in the acquisition of epigenetic changes. We discovered that AID binding to the IgH locus promotes an increase in H4K20me3. In 293F cells, we demonstrate interaction between co-transfected AID and the three SUV4-20 histone H4K20 methyltransferases, and that SUV4-20H1.2, bound to the IgH switch (S) mu site, is replaced by SUV4-20H2 upon AID binding. Analysis of HIGM2 mutants shows that the AID truncated form W68X is impaired to interact with SUV4-20H1.2 and SUV4-20H2 and is unable to bind and target H4K20me3 to the Smu site. We finally show in mouse primary B cells undergoing class-switch recombination (CSR) that AID deficiency associates with decreased H4K20me3 levels at the Smu site. Our results provide a novel link between SUV4-20 enzymes and CSR and offer a new aspect of the interplay between AID and histone modifications in setting the epigenetic status of CSR sites.


Leukemia Research | 2012

Epigenetic regulation of PRAME in acute myeloid leukemia is different compared to CD34+ cells from healthy donors: effect of 5-AZA treatment.

Silvia Gutierrez-Cosio; L. de la Rica; Esteban Ballestar; Carlos Santamaría; Luis Ignacio Sánchez-Abarca; Teresa Caballero-Velázquez; B. Blanco; Cristina Calderón; Carmen Herrero-Sánchez; Soraya Carrancio; Laura Ciudad; Consuelo del Cañizo; J. F. San Miguel; José A. Pérez-Simón

PRAME is a tumor associated antigen (TAA) of particular interest since it is widely expressed by lymphoid and myeloid malignancies. Several studies have associated high PRAME RNA levels with good prognosis in acute myeloid leukemia (AML). PRAME expression is regulated at the epigenetic level. For this reason inhibitors of DNA methylation, such as 5-azacytidine, can modulate the expression of this TAAs. In the current study we analyzed the effect of 5-azaC on the expression of PRAME in blasts versus CD34+ cells from healthy donors in an attempt to increase its expression, thus inducing a potential target for therapeutic strategies.


Journal of Translational Medicine | 2011

Epigenetic alterations in autoimmune disease

Esteban Ballestar; Biola M. Javierre; Lorenzo de la Rica; David Gomez-Cabrero; Jesper Tegnér; Carmen Gómez-Vaquero; Javier Narváez; Henar Hernando; Virginia C Rodriguez; Roser Vento; Laura Ciudad; Javier Rodríguez-Ubreva

Background In the past years, we have witnessed unprecedented attention for the study of epigenetic alterations in the context of a variety of complex disorders, including autoimmune diseases [1]. This is in part due to the observation that genetics is insufficient to entirely explain the predisposition to their pathogenesis. The environmental influence is well illustrated by the existence of partial concordance for susceptibility to disease in monozygotic twins. In connection with this epigenetic mechanisms regulate gene expression and are sensitive to external stimuli, bridging the gap between environmental and genetic factors. There is now considerable evidence of the existence of epigenetic alterations, particularly DNA methylation alterations, in diseases like systemic lupus erythematosus or rheumatoid arthritis [1]. Most of the studies were initially performed by using candidate-gene approaches, although the increasing availability of high-throughput methods is providing better methods for the screening of epigenetic alterations in these diseases [2].


Archive | 2014

MacrophagesEfficient Reprogramming into 34a and 223 Inhibits Lef1 Expression To C/EBPa-Mediated Activation of MicroRNAs

Maribel Parra; Thomas Graf; Esteban Ballestar; Javier Rodríguez-Ubreva; Laura Ciudad; Chris van Oevelen

Collaboration


Dive into the Laura Ciudad's collaboration.

Top Co-Authors

Avatar

Esteban Ballestar

Instituto de Salud Carlos III

View shared research outputs
Top Co-Authors

Avatar

Javier Rodríguez-Ubreva

Barcelona Biomedical Research Park

View shared research outputs
Top Co-Authors

Avatar

Carlos Santamaría

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thomas Graf

Pompeu Fabra University

View shared research outputs
Top Co-Authors

Avatar

Maribel Parra

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

B. Blanco

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Belén Blanco

Autonomous University of Madrid

View shared research outputs
Researchain Logo
Decentralizing Knowledge