Network


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

Hotspot


Dive into the research topics where Maite Huarte is active.

Publication


Featured researches published by Maite Huarte.


Molecular Cell | 2012

LincRNA-p21 Suppresses Target mRNA Translation

Je-Hyun Yoon; Kotb Abdelmohsen; Subramanya Srikantan; Xiaoling Yang; Jennifer L. Martindale; Supriyo De; Maite Huarte; Ming Zhan; Kevin G. Becker; Myriam Gorospe

Mammalian long intergenic noncoding RNAs (lincRNAs) are best known for modulating transcription. Here we report a posttranscriptional function for lincRNA-p21 as a modulator of translation. Association of the RNA-binding protein HuR with lincRNA-p21 favored the recruitment of let-7/Ago2 to lincRNA-p21, leading to lower lincRNA-p21 stability. Under reduced HuR levels, lincRNA-p21 accumulated in human cervical carcinoma HeLa cells, increasing its association with JUNB and CTNNB1 mRNAs and selectively lowering their translation. With elevated HuR, lincRNA-p21 levels declined, which in turn derepressed JunB and β-catenin translation and increased the levels of these proteins. We propose that HuR controls translation of a subset of target mRNAs by influencing lincRNA-p21 levels. Our findings uncover a role for lincRNA as a posttranscriptional inhibitor of translation.


Genome Biology | 2013

Pint lincRNA connects the p53 pathway with epigenetic silencing by the Polycomb repressive complex 2

Oskar Marín-Béjar; Francesco P. Marchese; Alejandro Athie; Yolanda Sánchez; Jovanna González; Victor Segura; Lulu Huang; Isabel Moreno; Alfons Navarro; Mariano Monzo; Jesús García-Foncillas; John L. Rinn; Shuling Guo; Maite Huarte

BackgroundThe p53 transcription factor is located at the core of a complex wiring of signaling pathways that are critical for the preservation of cellular homeostasis. Only recently it has become clear that p53 regulates the expression of several long intergenic noncoding RNAs (lincRNAs). However, relatively little is known about the role that lincRNAs play in this pathway.ResultsHere we characterize a lincRNA named Pint (p53 induced noncoding transcript). We show that Pint is a ubiquitously expressed lincRNA that is finely regulated by p53. In mouse cells, Pint promotes cell proliferation and survival by regulating the expression of genes of the TGF-β, MAPK and p53 pathways. Pint is a nuclear lincRNA that directly interacts with the Polycomb repressive complex 2 (PRC2), and is required for PRC2 targeting of specific genes for H3K27 tri-methylation and repression. Furthermore, Pint functional activity is highly dependent on PRC2 expression. We have also identified Pint human ortholog (PINT), which presents suggestive analogies with the murine lincRNA. PINT is similarly regulated by p53, and its expression significantly correlates with the same cellular pathways as the mouse ortholog, including the p53 pathway. Interestingly, PINT is downregulated in colon primary tumors, while its overexpression inhibits the proliferation of tumor cells, suggesting a possible role as tumor suppressor.ConclusionsOur results reveal a p53 autoregulatory negative mechanism where a lincRNA connects p53 activation with epigenetic silencing by PRC2. Additionally, we show analogies and differences between the murine and human orthologs, identifying a novel tumor suppressor candidate lincRNA.


Nucleic Acid Therapeutics | 2013

Long Non-Coding RNAs: Challenges for Diagnosis and Therapies

Yolanda Sánchez; Maite Huarte

Long non-coding RNAs (lncRNAs) have emerged as one of the largest and more diverse classes of cellular transcripts. The growing evidence suggests that lncRNAs are key regulatory molecules present at virtually every level of cellular physiology, and their alterations are associated with multiple human diseases. Here we provide a general overview of the known roles of lncRNAs in different diseases, as well as their imminent application as biomarkers and therapeutic targets. We also discuss the challenges and possible strategies for these clinical applications. It is unquestionable that our knowledge of lncRNAs not only adds a new dimension to the molecular architecture of human disease, but also opens up a whole new range of opportunities for treatment.


Epigenetics | 2014

Long non-coding RNAs and chromatin modifiers: Their place in the epigenetic code

Francesco P. Marchese; Maite Huarte

The emergence of long non-coding RNAs (lncRNAs) has shaken up our conception of gene expression regulation, as lncRNAs take prominent positions as components of cellular networks. Several cellular processes involve lncRNAs, and a significant number of them have been shown to function in cooperation with chromatin modifying enzymes to promote epigenetic activation or silencing of gene expression. Different model mechanisms have been proposed to explain how lncRNAs achieve regulation of gene expression by interacting with the epigenetic machinery. Here we describe these models in light of the current knowledge of lncRNAs, such as Xist and HOTAIR, and discuss recent literature on the role of the three-dimensional structure of the genome in the mechanism of action of lncRNAs and chromatin modifiers.


Genome Biology | 2017

The multidimensional mechanisms of long noncoding RNA function

Francesco P. Marchese; Ivan Raimondi; Maite Huarte

A major shift in our understanding of genome regulation has emerged recently. It is now apparent that the majority of cellular transcripts do not code for proteins, and many of them are long noncoding RNAs (lncRNAs). Increasingly, studies suggest that lncRNAs regulate gene expression through diverse mechanisms. We review emerging mechanistic views of lncRNAs in gene regulation in the cell nucleus. We discuss the functional interactions that lncRNAs establish with other molecules as well as the relationship between lncRNA transcription and function. While some of these mechanisms are specific to lncRNAs, others might be shared with other types of genes.


Trends in Cell Biology | 2011

To repress or not to repress: This is the guardian's question

John L. Rinn; Maite Huarte

p53 is possibly the most central tumor suppressor gene of our cells, integrating stress signals to activate a transcriptional program responsible for maintaining cellular homeostasis. Many of the downstream effects of p53 are a consequence of its activity as a transcription factor, resulting in the induction of multiple target genes. In addition to gene activation, however, gene repression is an essential part of the p53 cellular response. Despite extensive research efforts towards the elucidation of p53 functions, the molecular mechanisms and biological consequences of gene repression by p53 have not been studied extensively. We review our current knowledge of the mechanisms and biological consequences of p53 repression, with special attention to recently discovered mechanisms of repression that involve non-coding RNA molecules, an emerging aspect of regulation in the p53 cellular network.


Biochimica et Biophysica Acta | 2016

Expanding the p53 regulatory network: LncRNAs take up the challenge

Elena Grossi; Yolanda Sánchez; Maite Huarte

Long noncoding RNAs (lncRNAs) are rapidly emerging as important regulators of gene expression in a wide variety of physiological and pathological cellular processes. In particular, a number of studies revealed that some lncRNAs participate in the p53 pathway, the unquestioned protagonist of tumor suppressor response. Indeed, several lncRNAs are not only part of the large pool of genes coordinated by p53 transcription factor, but are also required by p53 to fine-tune its response and to fully accomplish its tumor suppressor program. In this review we will discuss the current and fast growing knowledge about the contribution of lncRNAs to the complexity of the p53 network, the different mechanisms by which they affect gene regulation in this context, and their involvement in cancer. The incipient impact of lncRNAs in the p53 biological response may encourage the development of therapies and diagnostic methods focused on these noncoding molecules. This article is part of a Special Issue entitled: Clues to long noncoding RNA taxonomy1, edited by Dr. Tetsuro Hirose and Dr. Shinichi Nakagawa.


Molecular Cell | 2016

A Long Noncoding RNA Regulates Sister Chromatid Cohesion

Francesco P. Marchese; Elena Grossi; Oskar Marín-Béjar; Sanjay Kumar Bharti; Ivan Raimondi; Jovanna González; Dannys Jorge Martínez-Herrera; Alejandro Athie; Alicia Amadoz; Robert M. Brosh; Maite Huarte

Long noncoding RNAs (lncRNAs) are involved in diverse cellular processes through multiple mechanisms. Here, we describe a previously uncharacterized human lncRNA, CONCR (cohesion regulator noncoding RNA), that is transcriptionally activated by MYC and is upregulated in multiple cancer types. The expression of CONCR is cell cycle regulated, and it is required for cell-cycle progression and DNA replication. Moreover, cells depleted of CONCR show severe defects in sister chromatid cohesion, suggesting an essential role for CONCR in cohesion establishment during cell division. CONCR interacts with and regulates the activity of DDX11, a DNA-dependent ATPase and helicase involved in DNA replication and sister chromatid cohesion. These findings unveil a direct role for an lncRNA in the establishment of sister chromatid cohesion by modulating DDX11 enzymatic activity.


Methods of Molecular Biology | 2015

RNA Pulldown Protocol for In Vitro Detection and Identification of RNA-Associated Proteins

Oskar Marín-Béjar; Maite Huarte

Recent advances in genomics have revealed that cells encode thousands of noncoding RNA molecules that do not require translation to proteins to perform their biological roles. Although very little is known about the mechanisms of function of noncoding RNAs, these undoubtedly involve the interaction with multiple cellular proteins. Here we describe a detailed RNA pulldown protocol for the in vitro detection of proteins specifically interacting with a long RNA molecule.


Cell Reports | 2017

Long Noncoding RNA PURPL Suppresses Basal p53 Levels and Promotes Tumorigenicity in Colorectal Cancer.

Xiao Ling Li; Murugan Subramanian; Matthew F. Jones; Ritu Chaudhary; Deepak K. Singh; Xinying Zong; Berkley Gryder; Sivasish Sindri; Min Mo; Aaron J. Schetter; Xinyu Wen; Swetha Parvathaneni; Dickran Kazandjian; Lisa M. Miller Jenkins; Wei Tang; Fathi Elloumi; Jennifer L. Martindale; Maite Huarte; Yuelin Zhu; Ana I. Robles; Susan M. Frier; Frank Rigo; Maggie Cam; Stefan Ambs; Sudha Sharma; Curtis C. Harris; Mary Dasso; Kannanganattu V. Prasanth; Ashish Lal

Basal p53 levels are tightly suppressed under normal conditions. Disrupting this regulation results in elevated p53 levels to induce cell cycle arrest, apoptosis, and tumor suppression. Here, we report the suppression of basal p53 levels by a nuclear, p53-regulated long noncoding RNA that we termed PURPL (p53 upregulated regulator of p53 levels). Targeted depletion of PURPL in colorectal cancer cells results in elevated basal p53 levels and induces growth defects in cell culture and in mouse xenografts. PURPL associates with MYBBP1A, a protein that binds to and stabilizes p53, and inhibits the formation of the p53-MYBBP1A complex. In the absence of PURPL, MYBBP1A interacts with and stabilizes p53. Silencing MYBBP1A significantly rescues basal p53 levels and proliferation in PURPL-deficient cells, suggesting that MYBBP1A mediates the effect of PURPL in regulating p53. These results reveal a p53-PURPL auto-regulatory feedback loop and demonstrate a role for PURPL in maintaining basal p53 levels.

Collaboration


Dive into the Maite Huarte's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge