Network


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

Hotspot


Dive into the research topics where Nadine Martin is active.

Publication


Featured researches published by Nadine Martin.


Genes & Development | 2012

Ncoa3 functions as an essential Esrrb coactivator to sustain embryonic stem cell self-renewal and reprogramming.

Michelle Percharde; Fabrice Lavial; Jia-Hui Ng; Vibhor Kumar; Rute Alexandra Tomaz; Nadine Martin; Jia-Chi Yeo; Jesús Gil; Shyam Prabhakar; Huck-Hui Ng; Malcolm G. Parker; Véronique Azuara

Embryonic stem cell (ESC) pluripotency depends on a well-characterized gene regulatory network centered on Oct4, Sox2, and Nanog. In contrast, little is known about the identity of the key coregulators and the mechanisms by which they may potentiate transcription in ESCs. Alongside core transcription factors, the orphan nuclear receptor Esrrb (estrogen-related receptor β) is vital for the maintenance of ESC identity and furthermore is uniquely associated with the basal transcription machinery. Here, we show that Ncoa3, an essential coactivator, is required to mediate Esrrb function in ESCs. Ncoa3 interacts with Esrrb via its ligand-binding domain and bridges Esrrb to RNA polymerase II complexes. Functionally, Ncoa3 is critical for both the induction and maintenance of pluripotency. Through chromatin immunoprecipitation (ChIP) sequencing and microarray experiments, we further demonstrate that Ncoa3 shares overlapping gene regulatory functions with Esrrb and cooperates genome-wide with the Oct4-Sox2-Nanog circuitry at active enhancers to up-regulate genes involved in self-renewal and pluripotency. We propose an integrated model of transcriptional and coactivator control, mediated by Ncoa3, for the maintenance of ESC self-renewal and somatic cell reprogramming.


Nature Medicine | 2015

Synthetic lethal targeting of oncogenic transcription factors in acute leukemia by PARP inhibitors.

Maria Esposito; Lu Zhao; Tsz Kan Fung; Jayant K. Rane; Amanda Wilson; Nadine Martin; Jesús Gil; Anskar Y. H. Leung; Alan Ashworth; Chi Wai Eric So

Acute myeloid leukemia (AML) is mostly driven by oncogenic transcription factors, which have been classically viewed as intractable targets using small-molecule inhibitor approaches. Here we demonstrate that AML driven by repressive transcription factors, including AML1-ETO (encoded by the fusion oncogene RUNX1-RUNX1T1) and PML-RARα fusion oncoproteins (encoded by PML-RARA) are extremely sensitive to poly (ADP-ribose) polymerase (PARP) inhibition, in part owing to their suppressed expression of key homologous recombination (HR)-associated genes and their compromised DNA-damage response (DDR). In contrast, leukemia driven by mixed-lineage leukemia (MLL, encoded by KMT2A) fusions with dominant transactivation ability is proficient in DDR and insensitive to PARP inhibition. Intriguingly, genetic or pharmacological inhibition of an MLL downstream target, HOXA9, which activates expression of various HR-associated genes, impairs DDR and sensitizes MLL leukemia to PARP inhibitors (PARPis). Conversely, HOXA9 overexpression confers PARPi resistance to AML1-ETO and PML-RARα transformed cells. Together, these studies describe a potential utility of PARPi-induced synthetic lethality for leukemia treatment and reveal a novel molecular mechanism governing PARPi sensitivity in AML.


The EMBO Journal | 2013

Interplay between Homeobox proteins and Polycomb repressive complexes in p16INK4a regulation

Nadine Martin; Nikolay Popov; Francesca Aguilo; Ana O'Loghlen; Selina Raguz; Ambrosius P. Snijders; Gopuraja Dharmalingam; SiDe Li; Efstathia Thymiakou; Thomas Carroll; Bernd B. Zeisig; Chi Wai Eric So; Gordon Peters; Vasso Episkopou; Martin J. Walsh; Jesús Gil

The INK4/ARF locus regulates senescence and is frequently altered in cancer. In normal cells, the INK4/ARF locus is found silenced by Polycomb repressive complexes (PRCs). Which are the mechanisms responsible for the recruitment of PRCs to INK4/ARF and their other target genes remains unclear. In a genetic screen for transcription factors regulating senescence, we identified the homeodomain‐containing protein HLX1 (H2.0‐like homeobox 1). Expression of HLX1 extends cellular lifespan and blunts oncogene‐induced senescence. Using quantitative proteomics, we identified p16INK4a as the key target mediating the effects of HLX1 in senescence. HLX1 represses p16INK4a transcription by recruiting PRCs and HDAC1. This mechanism has broader implications, as HLX1 also regulates a subset of PRC targets besides p16INK4a. Finally, sampling members of the Homeobox family, we identified multiple genes with ability to repress p16INK4a. Among them, we found HOXA9 (Homeobox A9), a putative oncogene in leukaemia, which also recruits PRCs and HDAC1 to regulate p16INK4a. Our results reveal an unexpected and conserved interplay between homeodomain‐containing proteins and PRCs with implications in senescence, development and cancer.


Trends in Molecular Medicine | 2014

Ageing as developmental decay: insights from p16(INK4a.)

Nadine Martin; David Beach; Jesús Gil

The p16(INK4a) cell cycle regulator is one of the best ageing biomarkers because it is suppressed in early embryogenesis and progressively induced during ageing. p16(INK4a) plays a crucial role in key cell fate decisions which contribute to ageing, such as cellular senescence and stem cell dynamics. Detailed examination of the pathways regulating p16(INK4a) expression has revealed an overlap with those regulating early development. We present the hypothesis that ageing might be primarily driven by gradual functional decay of developmental pathways. To support this, we summarise the role of p16(INK4a) in ageing and our current knowledge on p16(INK4a) regulation. The developmental decay hypothesis implies that the much-evidenced damage associated with all aspects of ageing might be secondary to such decay.


Oncogene | 2015

The nuclear receptor NR2E1/TLX controls senescence.

Ana O'Loghlen; Nadine Martin; Benjamin Krusche; Helen Pemberton; M M Alonso; Hollie Chandler; S Brookes; Simona Parrinello; Gordon Peters; Jesús Gil

The nuclear receptor NR2E1 (also known as TLX or tailless) controls the self-renewal of neural stem cells (NSCs) and has been implied as an oncogene which initiates brain tumors including glioblastomas. Despite NR2E1 regulating targets like p21CIP1 or PTEN we still lack a full explanation for its role in NSC self-renewal and tumorigenesis. We know that polycomb repressive complexes also control stem cell self-renewal and tumorigenesis, but so far, no formal connection has been established between NR2E1 and PRCs. In a screen for transcription factors regulating the expression of the polycomb protein CBX7, we identified NR2E1 as one of its more prominent regulators. NR2E1 binds at the CBX7 promoter, inducing its expression. Notably CBX7 represses NR2E1 as part of a regulatory loop. Ectopic NR2E1 expression inhibits cellular senescence, extending cellular lifespan in fibroblasts via CBX7-mediated regulation of p16INK4a and direct repression of p21CIP1. In addition NR2E1 expression also counteracts oncogene-induced senescence. The importance of NR2E1 to restrain senescence is highlighted through the process of knocking down its expression, which causes premature senescence in human fibroblasts and epithelial cells. We also confirmed that NR2E1 regulates CBX7 and restrains senescence in NSCs. Finally, we observed that the expression of NR2E1 directly correlates with that of CBX7 in human glioblastoma multiforme. Overall we identified control of senescence and regulation of polycomb action as two possible mechanisms that can join those so far invoked to explain the role of NR2E1 in control of NSC self-renewal and cancer.


Cell Cycle | 2013

Co-regulation of senescence-associated genes by oncogenic homeobox proteins and polycomb repressive complexes

Nadine Martin; Selina Raguz; Gopuraja Dharmalingam; Jesús Gil

Cellular senescence is a stable cell cycle arrest that can be induced by stresses such as telomere shortening, oncogene activation or DNA damage. Senescence is a potent anticancer barrier that needs to be circumvented during tumorigenesis. The cell cycle regulator p16INK4a is a key effector upregulated during senescence. Polycomb repressive complexes (PRCs) play a crucial role in silencing the INK4/ARF locus, which encodes for p16INK4a, but the mechanisms by which PRCs are recruited to this locus as well as to other targets remain poorly understood. Recently we discovered the ability of the homeobox proteins HLX1 (H2.0-like homeobox 1) and HOXA9 (Homeobox A9) to bypass senescence. We showed that HLX1 and HOXA9 recruit PRCs to repress INK4a, which constitutes a key mechanism explaining their effects on senescence. Here we provide evidence for the regulation of additional senescence-associated PRC target genes by HLX1 and HOXA9. As both HLX1 and HOXA9 are oncogenes implicated in leukemogenesis, we discuss the implications that the collaboration between Homeobox proteins and PRCs has for senescence and cancer.


Aging Cell | 2015

CBX7 and miR-9 are part of an autoregulatory loop controlling p16(INK) (4a).

Ana O'Loghlen; Sharon Brookes; Nadine Martin; Valentina Rapisarda; Gordon Peters; Jesús Gil

Polycomb repressive complexes (PRC1 and PRC2) are epigenetic regulators that act in coordination to influence multiple cellular processes including pluripotency, differentiation, cancer and senescence. The role of PRCs in senescence can be mostly explained by their ability to repress the INK4/ARF locus. CBX7 is one of five mammalian orthologues of Drosophila Polycomb that forms part of PRC1. Despite the relevance of CBX7 for regulating senescence and pluripotency, we have a limited understanding of how the expression of CBX7 is regulated. Here we report that the miR‐9 family of microRNAs (miRNAS) downregulates the expression of CBX7. In turn, CBX7 represses miR‐9‐1 and miR‐9‐2 as part of a regulatory negative feedback loop. The miR‐9/CBX7 feedback loop is a regulatory module contributing to induction of the cyclin‐dependent kinase inhibitor (CDKI) p16INK4a during senescence. The ability of the miR‐9 family to regulate senescence could have implications for understanding the role of miR‐9 in cancer and aging.


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.


European Journal of Cancer | 2016

Exploiting DNA damage repair defects for effective targeting of acute myeloid leukaemia by PARP inhibitors

Maria Teresa Esposito; Lu Zhao; Tsz Kan Fung; Jayant K. Rane; Amanda Wilson; Nadine Martin; Jesús Gil; Anskar Y. H. Leung; Alan Ashworth; Chi Wai Eric So

Abstract for 24th Biennial Congress of the European Association for Cancer Research, 9–12 July 2016, Manchester, UK. Poster Session: Cancer Genomics, Epigenetics and Genome Instability II: Monday 11 July 2016


Genes & Development | 2016

SWI/SNF regulates a transcriptional program that induces senescence to prevent liver cancer

Luca Tordella; Sadaf Khan; Anja Hohmeyer; Ana Banito; Sabrina Klotz; Selina Raguz; Nadine Martin; Gopuraja Dhamarlingam; Thomas Carroll; José Mario González Meljem; Sumit Deswal; Juan Pedro Martinez-Barbera; Ramón García-Escudero; Johannes Zuber; Lars Zender; Jesús Gil

Collaboration


Dive into the Nadine Martin's collaboration.

Top Co-Authors

Avatar

Jesús Gil

Imperial College London

View shared research outputs
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

Selina Raguz

Imperial College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ana Banito

Imperial College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lu Zhao

King's College London

View shared research outputs
Researchain Logo
Decentralizing Knowledge