Network


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

Hotspot


Dive into the research topics where Ana Banito is active.

Publication


Featured researches published by Ana Banito.


Cell | 2008

Chemokine Signaling via the CXCR2 Receptor Reinforces Senescence

Juan Carlos Acosta; Ana O'Loghlen; Ana Banito; Maria V. Guijarro; Arnaud Augert; Selina Raguz; Marzia Fumagalli; Marco Da Costa; Celia Brown; Nikolay Popov; Yoshihiro Takatsu; Jonathan Melamed; Fabrizio d'Adda di Fagagna; David Bernard; Eva Hernando; Jesús Gil

Cells enter senescence, a state of stable proliferative arrest, in response to a variety of cellular stresses, including telomere erosion, DNA damage, and oncogenic signaling, which acts as a barrier against malignant transformation in vivo. To identify genes controlling senescence, we conducted an unbiased screen for small hairpin RNAs that extend the life span of primary human fibroblasts. Here, we report that knocking down the chemokine receptor CXCR2 (IL8RB) alleviates both replicative and oncogene-induced senescence (OIS) and diminishes the DNA-damage response. Conversely, ectopic expression of CXCR2 results in premature senescence via a p53-dependent mechanism. Cells undergoing OIS secrete multiple CXCR2-binding chemokines in a program that is regulated by the NF-kappaB and C/EBPbeta transcription factors and coordinately induce CXCR2 expression. CXCR2 upregulation is also observed in preneoplastic lesions in vivo. These results suggest that senescent cells activate a self-amplifying secretory network in which CXCR2-binding chemokines reinforce growth arrest.


Genes & Development | 2009

Senescence impairs successful reprogramming to pluripotent stem cells

Ana Banito; Sheikh Tamir Rashid; Juan Carlos Acosta; Si De Li; Carlos Filipe Pereira; Imbisaat Geti; Sandra Pinho; José C.R. Silva; Véronique Azuara; Martin J. Walsh; Ludovic Vallier; Jesús Gil

Somatic cells can be reprogrammed into induced pluripotent stem (iPS) cells by overexpressing combinations of factors such as Oct4, Sox2, Klf4, and c-Myc. Reprogramming is slow and stochastic, suggesting the existence of barriers limiting its efficiency. Here we identify senescence as one such barrier. Expression of the four reprogramming factors triggers senescence by up-regulating p53, p16(INK4a), and p21(CIP1). Induction of DNA damage response and chromatin remodeling of the INK4a/ARF locus are two of the mechanisms behind senescence induction. Crucially, ablation of different senescence effectors improves the efficiency of reprogramming, suggesting novel strategies for maximizing the generation of iPS cells.


Nature Cell Biology | 2013

A complex secretory program orchestrated by the inflammasome controls paracrine senescence

Juan Carlos Acosta; Ana Banito; Torsten Wuestefeld; Athena Georgilis; Peggy Janich; Jennifer P. Morton; Dimitris Athineos; Tae-Won Kang; Felix Lasitschka; Mindaugas Andrulis; Gloria Pascual; Kelly J. Morris; Sadaf Khan; Hong Jin; Gopuraja Dharmalingam; Ambrosius P. Snijders; Thomas J. Carroll; David Capper; Catrin Pritchard; Gareth J. Inman; Thomas Longerich; Owen J. Sansom; Lars Zender; Jesús Gil

Oncogene-induced senescence (OIS) is crucial for tumour suppression. Senescent cells implement a complex pro-inflammatory response termed the senescence-associated secretory phenotype (SASP). The SASP reinforces senescence, activates immune surveillance and paradoxically also has pro-tumorigenic properties. Here, we present evidence that the SASP can also induce paracrine senescence in normal cells both in culture and in human and mouse models of OIS in vivo. Coupling quantitative proteomics with small-molecule screens, we identified multiple SASP components mediating paracrine senescence, including TGF-β family ligands, VEGF, CCL2 and CCL20. Amongst them, TGF-β ligands play a major role by regulating p15INK4b and p21CIP1. Expression of the SASP is controlled by inflammasome-mediated IL-1 signalling. The inflammasome and IL-1 signalling are activated in senescent cells and IL-1α expression can reproduce SASP activation, resulting in senescence. Our results demonstrate that the SASP can cause paracrine senescence and impact on tumour suppression and senescence in vivo.


Genes & Development | 2009

Histone demethylase JMJD3 contributes to epigenetic control of INK4a/ARF by oncogenic RAS

Marta Barradas; Emma Anderton; Juan Carlos Acosta; SiDe Li; Ana Banito; Marc Rodriguez-Niedenführ; Goedele N. Maertens; Michaela S. Banck; Ming-Ming Zhou; Martin J. Walsh; Gordon Peters; Jesús Gil

The INK4a/ARF tumor suppressor locus, a key executor of cellular senescence, is regulated by members of the Polycomb group (PcG) of transcriptional repressors. Here we show that signaling from oncogenic RAS overrides PcG-mediated repression of INK4a by activating the H3K27 demethylase JMJD3 and down-regulating the methyltransferase EZH2. In human fibroblasts, JMJD3 activates INK4a, but not ARF, and causes p16(INK4a)-dependent arrest. In mouse embryo fibroblasts, Jmjd3 activates both Ink4a and Arf and elicits a p53-dependent arrest, echoing the effects of RAS in this system. Our findings directly implicate JMJD3 in the regulation of INK4a/ARF during oncogene-induced senescence and suggest that JMJD3 has the capacity to act as a tumor suppressor.


Cell Stem Cell | 2012

MicroRNA Regulation of Cbx7 Mediates a Switch of Polycomb Orthologs during ESC Differentiation

Ana O'Loghlen; Ana M. Muñoz-Cabello; Alexandre Gaspar-Maia; Hsan-Au Wu; Ana Banito; Natalia Kunowska; Tomas Racek; Helen Pemberton; Patrizia Beolchi; Fabrice Lavial; Osamu Masui; Michiel Vermeulen; Thomas Carroll; Johannes Graumann; Edith Heard; Niall Dillon; Véronique Azuara; Ambrosius P. Snijders; Gordon Peters; Emily Bernstein; Jesús Gil

Summary The Polycomb Group (PcG) of chromatin modifiers regulates pluripotency and differentiation. Mammalian genomes encode multiple homologs of the Polycomb repressive complex 1 (PRC1) components, including five orthologs of the Drosophila Polycomb protein (Cbx2, Cbx4, Cbx6, Cbx7, and Cbx8). We have identified Cbx7 as the primary Polycomb ortholog of PRC1 complexes in embryonic stem cells (ESCs). The expression of Cbx7 is downregulated during ESC differentiation, preceding the upregulation of Cbx2, Cbx4, and Cbx8, which are directly repressed by Cbx7. Ectopic expression of Cbx7 inhibits differentiation and X chromosome inactivation and enhances ESC self-renewal. Conversely, Cbx7 knockdown induces differentiation and derepresses lineage-specific markers. In a functional screen, we identified the miR-125 and miR-181 families as regulators of Cbx7 that are induced during ESC differentiation. Ectopic expression of these miRNAs accelerates ESC differentiation via regulation of Cbx7. These observations establish a critical role for Cbx7 and its regulatory miRNAs in determining pluripotency.


Cell Stem Cell | 2010

ESCs Require PRC2 to Direct the Successful Reprogramming of Differentiated Cells toward Pluripotency

Carlos Filipe Pereira; Francesco M. Piccolo; Tomomi Tsubouchi; Stephan Sauer; Natalie K. Ryan; Ludovica Bruno; David Landeira; Joana Santos; Ana Banito; Jesús Gil; Haruhiko Koseki; Matthias Merkenschlager; Amanda G. Fisher

Embryonic stem cells (ESCs) are pluripotent, self-renewing, and have the ability to reprogram differentiated cell types to pluripotency upon cellular fusion. Polycomb-group (PcG) proteins are important for restraining the inappropriate expression of lineage-specifying factors in ESCs. To investigate whether PcG proteins are required for establishing, rather than maintaining, the pluripotent state, we compared the ability of wild-type, PRC1-, and PRC2-depleted ESCs to reprogram human lymphocytes. We show that ESCs lacking either PRC1 or PRC2 are unable to successfully reprogram B cells toward pluripotency. This defect is a direct consequence of the lack of PcG activity because it could be efficiently rescued by reconstituting PRC2 activity in PRC2-deficient ESCs. Surprisingly, the failure of PRC2-deficient ESCs to reprogram somatic cells is functionally dominant, demonstrating a critical requirement for PcG proteins in the chromatin-remodeling events required for the direct conversion of differentiated cells toward pluripotency.


Nature Cell Biology | 2015

mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype

Nicolás Herranz; Suchira Gallage; Massimiliano Mellone; Torsten Wuestefeld; Sabrina Klotz; Christopher J. Hanley; Selina Raguz; Juan Carlos Acosta; Andrew J. Innes; Ana Banito; Athena Georgilis; Alex Montoya; Katharina Wolter; Gopuraja Dharmalingam; Peter Faull; Thomas Carroll; Juan Pedro Martinez-Barbera; Pedro R. Cutillas; Florian Reisinger; Mathias Heikenwalder; Richard A. Miller; Dominic J. Withers; Lars Zender; Gareth J. Thomas; Jesús Gil

Senescent cells secrete a combination of factors collectively known as the senescence-associated secretory phenotype (SASP). The SASP reinforces senescence and activates an immune surveillance response, but it can also show pro-tumorigenic properties and contribute to age-related pathologies. In a drug screen to find new SASP regulators, we uncovered the mTOR inhibitor rapamycin as a potent SASP suppressor. Here we report a mechanism by which mTOR controls the SASP by differentially regulating the translation of the MK2 (also known as MAPKAPK2) kinase through 4EBP1. In turn, MAPKAPK2 phosphorylates the RNA-binding protein ZFP36L1 during senescence, inhibiting its ability to degrade the transcripts of numerous SASP components. Consequently, mTOR inhibition or constitutive activation of ZFP36L1 impairs the non-cell-autonomous effects of senescent cells in both tumour-suppressive and tumour-promoting contexts. Altogether, our results place regulation of the SASP as a key mechanism by which mTOR could influence cancer, age-related diseases and immune responses.


EMBO Reports | 2010

Induced pluripotent stem cells and senescence: learning the biology to improve the technology.

Ana Banito; Jesús Gil

The discovery that adult somatic cells can be reprogrammed into pluripotent cells by expressing a combination of factors associated with pluripotency holds immense promise for a wide range of biotechnological and therapeutic applications. However, some hurdles—such as improving the low reprogramming efficiencies and ensuring the pluripotent potential, genomic integrity and safety of the resulting cells—must be overcome before induced pluripotent stem cells (iPSCs) can be used for clinical purposes. Several groups have recently shown that key tumour suppressors—such as members of the p53 and p16INK4a/retinoblastoma networks—control the efficiency of iPSC generation by activating cell‐intrinsic programmes such as senescence. Here, we discuss the implications of these discoveries for improving the safety and efficiency of iPSC generation, and for increasing our understanding of different aspects of basic biology—such as the control of pluripotency or the mechanisms involved in the generation of cancer stem cells.


Cell Cycle | 2008

Control of senescence by CXCR2 and its ligands.

Juan Carlos Acosta; Ana O'Loghlen; Ana Banito; Selina Raguz; Jesús Gil

Senescence is an irreversible growth arrest with important physiological implications as it contributes to tumour suppression and may have a role in aging. During senescence, cells suffer profound phenotypic changes affecting amongst others cell morphology and chromatin structure. Senescent cells also undergo significant transcriptional changes, such as the increased production of a plethora of different secreted factors, which are the basis of the so-called senescence-associated secretory phenotype. While some of these factors have been previously shown to possess different pro-tumorigenic activities, we recently demonstrated that the secretion of CXCR2-binding chemokines (such as IL-8 or GROα) by senescent cells contribute to reinforce senescence via activation of the p53 pathway. Importantly, our data adds to that presented by several groups suggesting that also other factors secreted during senescence (such as PAI-1, IGFBP-7 or IL-6) contribute to the senescent response. Here, we discuss our findings in the context of the emerging role for secreted factors in regulating senescence through paracrine and/or autocrine mechanisms


Genes & Development | 2017

Coupling shRNA screens with single-cell RNA-seq identifies a dual role for mTOR in reprogramming-induced senescence

Marieke Aarts; Athena Georgilis; Meryam Beniazza; Patrizia Beolchi; Ana Banito; Thomas Carroll; Marizela Kulisic; Daniel F. Kaemena; Gopuraja Dharmalingam; Nadine Martin; Wolf Reik; Johannes Zuber; Keisuke Kaji; Tamir Chandra; Jesús Gil

Expression of the transcription factors OCT4, SOX2, KLF4, and cMYC (OSKM) reprograms somatic cells into induced pluripotent stem cells (iPSCs). Reprogramming is a slow and inefficient process, suggesting the presence of safeguarding mechanisms that counteract cell fate conversion. One such mechanism is senescence. To identify modulators of reprogramming-induced senescence, we performed a genome-wide shRNA screen in primary human fibroblasts expressing OSKM. In the screen, we identified novel mediators of OSKM-induced senescence and validated previously implicated genes such as CDKN1A We developed an innovative approach that integrates single-cell RNA sequencing (scRNA-seq) with the shRNA screen to investigate the mechanism of action of the identified candidates. Our data unveiled regulation of senescence as a novel way by which mechanistic target of rapamycin (mTOR) influences reprogramming. On one hand, mTOR inhibition blunts the induction of cyclin-dependent kinase (CDK) inhibitors (CDKIs), including p16INK4a, p21CIP1, and p15INK4b, preventing OSKM-induced senescence. On the other hand, inhibition of mTOR blunts the senescence-associated secretory phenotype (SASP), which itself favors reprogramming. These contrasting actions contribute to explain the complex effect that mTOR has on reprogramming. Overall, our study highlights the advantage of combining functional screens with scRNA-seq to accelerate the discovery of pathways controlling complex phenotypes.

Collaboration


Dive into the Ana Banito's collaboration.

Top Co-Authors

Avatar

Jesús Gil

Imperial College London

View shared research outputs
Top Co-Authors

Avatar

Selina Raguz

Imperial College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ana O'Loghlen

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alex Montoya

Imperial College London

View shared research outputs
Researchain Logo
Decentralizing Knowledge