scispace - formally typeset
Open AccessJournal ArticleDOI

A genome-wide microRNA screen identifies regulators of tetraploid cell proliferation.

Reads0
Chats0
TLDR
A comprehensive gain-of-function genome-wide screen to identify microRNAs (miRNAs) that are sufficient to promote the proliferation of tetraploid cells reveals several avenues through which tetraPloid cells may regain the proliferative capacity necessary to drive tumorigenesis.
Abstract
Tetraploid cells, which are most commonly generated by errors in cell division, are genomically unstable and have been shown to promote tumorigenesis. Recent genomic studies have estimated that ∼40% of all solid tumors have undergone a genome-doubling event during their evolution, suggesting a significant role for tetraploidy in driving the development of human cancers. To safeguard against the deleterious effects of tetraploidy, nontransformed cells that fail mitosis and become tetraploid activate both the Hippo and p53 tumor suppressor pathways to restrain further proliferation. Tetraploid cells must therefore overcome these antiproliferative barriers to ultimately drive tumor development. However, the genetic routes through which spontaneously arising tetraploid cells adapt to regain proliferative capacity remain poorly characterized. Here, we conducted a comprehensive gain-of-function genome-wide screen to identify microRNAs (miRNAs) that are sufficient to promote the proliferation of tetraploid cells. Our screen identified 23 miRNAs whose overexpression significantly promotes tetraploid proliferation. The vast majority of these miRNAs facilitate tetraploid growth by enhancing mitogenic signaling pathways (e.g., miR-191-3p); however, we also identified several miRNAs that impair the p53/p21 pathway (e.g., miR-523-3p), and a single miRNA (miR-24-3p) that potently inactivates the Hippo pathway via down-regulation of the tumor suppressor gene NF2. Collectively, our data reveal several avenues through which tetraploid cells may regain the proliferative capacity necessary to drive tumorigenesis.

read more

Citations
More filters

MicroRNAs: Target Recognition and Regulatory Functions

TL;DR: In this article, a review outlines the current understanding of miRNA target recognition in animals and discusses the widespread impact of miRNAs on both the expression and evolution of protein-coding genes.
Journal ArticleDOI

Reciprocal Crosstalk Between YAP1/Hippo Pathway and the p53 Family Proteins: Mechanisms and Outcomes in Cancer

TL;DR: This review dissects the critical points of crosstalk between the YAP1/Hippo pathway components, with a focus on YAP 1, and the p53 tumor suppressor protein family.
Dissertation

Whole genome doubling propagates chromosomal instability and accelerates cancer genome evolution

TL;DR: In this paper, the effect of spontaneous tetraploidization on colorectal cancer genomes has been investigated using a siRNA screen of genes commonly mutated in genome-doubled tumours of multiple cancer types.
Book ChapterDOI

Giants and monsters: Unexpected characters in the story of cancer recurrence.

TL;DR: The clinical significance of PGCC, the need for standardized nomenclature and molecular markers, as well as possible avenues to develop therapies aimed at PGCC and the process of neosis are discussed.
Journal ArticleDOI

Modelling chromosome structural and copy number changes to understand cancer genomes.

TL;DR: The obtained findings reveal that genomic alterations strongly affect the cellular physiology and, importantly, instigate further genomic instability.
References
More filters
Journal ArticleDOI

MicroRNAs: Target Recognition and Regulatory Functions

TL;DR: The current understanding of miRNA target recognition in animals is outlined and the widespread impact of miRNAs on both the expression and evolution of protein-coding genes is discussed.
Journal ArticleDOI

Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control

TL;DR: It is demonstrated that in mammalian cells, the transcription coactivator YAP (Yes-associated protein), is inhibited by cell density via the Hippo pathway, and YAP overexpression regulates gene expression in a manner opposite to cell density, and is able to overcome cell contact inhibition.
Journal ArticleDOI

Visualizing Spatiotemporal Dynamics of Multicellular Cell-Cycle Progression

TL;DR: Time-lapse imaging is performed to explore the spatiotemporal patterns of cell-cycle dynamics during the epithelial-mesenchymal transition of cultured cells, the migration and differentiation of neural progenitors in brain slices, and the development of tumors across blood vessels in live mice.
Related Papers (5)