Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Trends Genet ; 39(12): 954-967, 2023 12.
Article in English | MEDLINE | ID: mdl-37714734

ABSTRACT

The overwhelming majority of proliferating somatic human cells are diploid, and this genomic state is typically maintained across successive cell divisions. However, failures in cell division can induce a whole-genome doubling (WGD) event, in which diploid cells transition to a tetraploid state. While some WGDs are developmentally programmed to produce nonproliferative tetraploid cells with specific cellular functions, unscheduled WGDs can be catastrophic: erroneously arising tetraploid cells are ill-equipped to cope with their doubled cellular and chromosomal content and quickly become genomically unstable and tumorigenic. Deciphering the genetics that underlie the genesis, physiology, and evolution of whole-genome doubled (WGD+) cells may therefore reveal therapeutic avenues to selectively eliminate pathological WGD+ cells.


Subject(s)
Neoplasms , Tetraploidy , Humans , Neoplasms/genetics , Cell Division , Genome/genetics , Cell Physiological Phenomena
2.
Mol Cell ; 83(12): 1964-1966, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37327773

ABSTRACT

Lin et al.1 demonstrate that acentric chromosome fragments generated within micronuclei are tethered by the CIP2A-TOPBP1 complex during mitosis, thus promoting clustered segregation of the fragments to a single daughter cell nucleus and facilitating re-ligation with limited chromosomal scattering and loss.


Subject(s)
Cell Nucleus , Chromosomes , Mitosis , Chromosome Segregation
3.
Cells ; 12(7)2023 03 23.
Article in English | MEDLINE | ID: mdl-37048053

ABSTRACT

Hyperactive sphingosine 1-phosphate (S1P) signaling is associated with a poor prognosis of triple-negative breast cancer (TNBC). Despite recent evidence that links the S1P receptor 1 (S1P1) to TNBC cell survival, its role in TNBC invasion and the underlying mechanisms remain elusive. Combining analyses of human TNBC cells with zebrafish xenografts, we found that phosphorylation of S1P receptor 1 (S1P1) at threonine 236 (T236) is critical for TNBC dissemination. Compared to luminal breast cancer cells, TNBC cells exhibit a significant increase of phospho-S1P1 T236 but not the total S1P1 levels. Misexpression of phosphorylation-defective S1P1 T236A (alanine) decreases TNBC cell migration in vitro and disease invasion in zebrafish xenografts. Pharmacologic disruption of S1P1 T236 phosphorylation, using either a pan-AKT inhibitor (MK2206) or an S1P1 functional antagonist (FTY720, an FDA-approved drug for treating multiple sclerosis), suppresses TNBC cell migration in vitro and tumor invasion in vivo. Finally, we show that human TNBC cells with AKT activation and elevated phospho-S1P1 T236 are sensitive to FTY720-induced cytotoxic effects. These findings indicate that the AKT-enhanced phosphorylation of S1P1 T236 mediates much of the TNBC invasiveness, providing a potential biomarker to select TNBC patients for the clinical application of FTY720.


Subject(s)
Fingolimod Hydrochloride , Sphingosine-1-Phosphate Receptors , Triple Negative Breast Neoplasms , Animals , Humans , Fingolimod Hydrochloride/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Threonine , Triple Negative Breast Neoplasms/drug therapy , Zebrafish/metabolism
4.
Nat Commun ; 13(1): 4109, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35840569

ABSTRACT

Melanomas and other solid tumors commonly have increased ploidy, with near-tetraploid karyotypes being most frequently observed. Such karyotypes have been shown to arise through whole-genome doubling events that occur during early stages of tumor progression. The generation of tetraploid cells via whole-genome doubling is proposed to allow nascent tumor cells the ability to sample various pro-tumorigenic genomic configurations while avoiding the negative consequences that chromosomal gains or losses have in diploid cells. Whereas a high prevalence of whole-genome doubling events has been established, the means by which whole-genome doubling arises is unclear. Here, we find that BRAFV600E, the most common mutation in melanomas, can induce whole-genome doubling via cytokinesis failure in vitro and in a zebrafish melanoma model. Mechanistically, BRAFV600E causes decreased activation and localization of RhoA, a critical cytokinesis regulator. BRAFV600E activity during G1/S phases of the cell cycle is required to suppress cytokinesis. During G1/S, BRAFV600E activity causes inappropriate centriole amplification, which is linked in part to inhibition of RhoA and suppression of cytokinesis. Together these data suggest that common abnormalities of melanomas linked to tumorigenesis - amplified centrosomes and whole-genome doubling events - can be induced by oncogenic BRAF and other mutations that increase RAS/MAPK pathway activity.


Subject(s)
Melanoma , Proto-Oncogene Proteins B-raf , Animals , Cell Line, Tumor , Cytokinesis/genetics , Melanoma/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Tetraploidy , Zebrafish/genetics , Zebrafish/metabolism
5.
Nat Commun ; 13(1): 3732, 2022 06 29.
Article in English | MEDLINE | ID: mdl-35768444

ABSTRACT

Melanoma is commonly driven by activating mutations in the MAP kinase BRAF; however, oncogenic BRAF alone is insufficient to promote melanomagenesis. Instead, its expression induces a transient proliferative burst that ultimately ceases with the development of benign nevi comprised of growth-arrested melanocytes. The tumor suppressive mechanisms that restrain nevus melanocyte proliferation remain poorly understood. Here we utilize cell and murine models to demonstrate that oncogenic BRAF leads to activation of the Hippo tumor suppressor pathway, both in melanocytes in vitro and nevus melanocytes in vivo. Mechanistically, we show that oncogenic BRAF promotes both ERK-dependent alterations in the actin cytoskeleton and whole-genome doubling events, which independently reduce RhoA activity to promote Hippo activation. We also demonstrate that functional impairment of the Hippo pathway enables oncogenic BRAF-expressing melanocytes to bypass nevus formation and rapidly form melanomas. Our data reveal that the Hippo pathway enforces the stable arrest of nevus melanocytes and represents a critical barrier to melanoma development.


Subject(s)
Melanoma , Nevus , Skin Neoplasms , Animals , Melanocytes/metabolism , Melanoma/pathology , Mice , Mutation , Nevus/genetics , Nevus/metabolism , Nevus/pathology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/pathology
6.
Nucleic Acids Res ; 49(16): 9424-9443, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34365507

ABSTRACT

RNA provides the framework for the assembly of some of the most intricate macromolecular complexes within the cell, including the spliceosome and the mature ribosome. The assembly of these complexes relies on the coordinated association of RNA with hundreds of trans-acting protein factors. While some of these trans-acting factors are RNA-binding proteins (RBPs), others are adaptor proteins, and others still, function as both. Defects in the assembly of these complexes results in a number of human pathologies including neurodegeneration and cancer. Here, we demonstrate that Silencing Defective 2 (SDE2) is both an RNA binding protein and also a trans-acting adaptor protein that functions to regulate RNA splicing and ribosome biogenesis. SDE2 depletion leads to widespread changes in alternative splicing, defects in ribosome biogenesis and ultimately complete loss of cell viability. Our data highlight SDE2 as a previously uncharacterized essential gene required for the assembly and maturation of the complexes that carry out two of the most fundamental processes in mammalian cells.


Subject(s)
Alternative Splicing/genetics , DNA-Binding Proteins/genetics , RNA Splicing/genetics , Ribosomes/genetics , Genes, Essential/genetics , Humans , RNA-Binding Proteins/genetics , Spliceosomes/genetics
8.
Nature ; 590(7846): 492-497, 2021 02.
Article in English | MEDLINE | ID: mdl-33505027

ABSTRACT

Whole-genome doubling (WGD) is common in human cancers, occurring early in tumorigenesis and generating genetically unstable tetraploid cells that fuel tumour development1,2. Cells that undergo WGD (WGD+ cells) must adapt to accommodate their abnormal tetraploid state; however, the nature of these adaptations, and whether they confer vulnerabilities that can be exploited therapeutically, is unclear. Here, using sequencing data from roughly 10,000 primary human cancer samples and essentiality data from approximately 600 cancer cell lines, we show that WGD gives rise to common genetic traits that are accompanied by unique vulnerabilities. We reveal that WGD+ cells are more dependent than WGD- cells on signalling from the spindle-assembly checkpoint, DNA-replication factors and proteasome function. We also identify KIF18A, which encodes a mitotic kinesin protein, as being specifically required for the viability of WGD+ cells. Although KIF18A is largely dispensable for accurate chromosome segregation during mitosis in WGD- cells, its loss induces notable mitotic errors in WGD+ cells, ultimately impairing cell viability. Collectively, our results suggest new strategies for specifically targeting WGD+ cancer cells while sparing the normal, non-transformed WGD- cells that comprise human tissue.


Subject(s)
Genome, Human/genetics , Mitosis/drug effects , Neoplasms/genetics , Neoplasms/pathology , Tetraploidy , Abnormal Karyotype/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Genes, Lethal/genetics , Humans , Kinesins/deficiency , Kinesins/genetics , Kinesins/metabolism , M Phase Cell Cycle Checkpoints/drug effects , Male , Mitosis/genetics , Proteasome Endopeptidase Complex/metabolism , Reproducibility of Results , Spindle Apparatus/drug effects
9.
Nat Cell Biol ; 22(2): 246-256, 2020 02.
Article in English | MEDLINE | ID: mdl-32015438

ABSTRACT

The Hippo and mammalian target of rapamycin complex 1 (mTORC1) pathways are the two predominant growth-control pathways that dictate proper organ development. We therefore explored potential crosstalk between these two functionally relevant pathways to coordinate their growth-control functions. We found that the LATS1 and LATS2 kinases, the core components of the Hippo pathway, phosphorylate S606 of Raptor, an essential component of mTORC1, to attenuate mTORC1 activation by impairing the interaction of Raptor with Rheb. The phosphomimetic Raptor-S606D knock-in mutant led to a reduction in cell size and proliferation. Compared with Raptor+/+ mice, RaptorD/D knock-in mice exhibited smaller livers and hearts, and a significant inhibition of elevation in mTORC1 signalling induced by Nf2 or Lats1 and Lats2 loss. Thus, our study reveals a direct link between the Hippo and mTORC1 pathways to fine-tune organ growth.


Subject(s)
Gene Expression Regulation, Developmental , Mechanistic Target of Rapamycin Complex 1/genetics , Protein Serine-Threonine Kinases/genetics , Ras Homolog Enriched in Brain Protein/genetics , Regulatory-Associated Protein of mTOR/genetics , Tumor Suppressor Proteins/genetics , Animals , CRISPR-Cas Systems , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Gene Editing , HCT116 Cells , HEK293 Cells , HeLa Cells , Heterografts , Hippo Signaling Pathway , Humans , Liver/abnormalities , Liver/metabolism , MCF-7 Cells , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Nude , Mice, Transgenic , Myocardium/metabolism , Myocardium/pathology , Neurofibromin 2/deficiency , Neurofibromin 2/genetics , Organ Size , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , Regulatory-Associated Protein of mTOR/metabolism , Signal Transduction , Tumor Suppressor Proteins/deficiency
10.
Nat Commun ; 10(1): 1547, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30948712

ABSTRACT

The Hippo pathway maintains tissue homeostasis by negatively regulating the oncogenic transcriptional co-activators YAP and TAZ. Though functional inactivation of the Hippo pathway is common in tumors, mutations in core pathway components are rare. Thus, understanding how tumor cells inactivate Hippo signaling remains a key unresolved question. Here, we identify the kinase STK25 as an activator of Hippo signaling. We demonstrate that loss of STK25 promotes YAP/TAZ activation and enhanced cellular proliferation, even under normally growth-suppressive conditions both in vitro and in vivo. Notably, STK25 activates LATS by promoting LATS activation loop phosphorylation independent of a preceding phosphorylation event at the hydrophobic motif, which represents a form of Hippo activation distinct from other kinase activators of LATS. STK25 is significantly focally deleted across a wide spectrum of human cancers, suggesting STK25 loss may represent a common mechanism by which tumor cells functionally impair the Hippo tumor suppressor pathway.


Subject(s)
Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Cell Line , Cell Proliferation , Genes, Tumor Suppressor , Hippo Signaling Pathway , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
11.
Methods Mol Biol ; 1893: 203-214, 2019.
Article in English | MEDLINE | ID: mdl-30565136

ABSTRACT

The advent of CRISPR has revolutionized genomic engineering, and harnessing its power to regulate levels of the transcriptional co-activators YAP and TAZ represents an exciting new opportunity in the field of Hippo signaling. Initially repurposed from the microbial immune system to perform highly specific gene knockouts, CRISPR technology has now been expanded to modulate the transcriptional activity of any gene of interest in mammalian systems. Here, we describe strategies to employ CRISPR to genetically knock out the genes encoding for YAP (YAP1) or TAZ (WWTR1) in mammalian cell lines, as well as briefly outline an approach for utilizing CRISPR to transcriptionally modulate YAP/TAZ levels.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Nuclear Proteins/genetics , Transcription Factors/genetics , Acyltransferases , Cell Cycle Proteins , Gene Editing , Gene Expression Regulation , Gene Knockout Techniques , Genetic Vectors , Hippo Signaling Pathway , Humans , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA, Guide, Kinetoplastida , Signal Transduction , Transcription Factors/metabolism
12.
Mol Biol Cell ; 29(13): 1682-1692, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29791254

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.


Subject(s)
Genetic Testing , MicroRNAs/genetics , Tetraploidy , Adaptor Proteins, Signal Transducing/metabolism , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation/genetics , Humans , MicroRNAs/metabolism , Mitogens/metabolism , Neurofibromin 2/metabolism , Phosphoproteins/metabolism , Signal Transduction , Transcription Factors , Tumor Suppressor Protein p53/metabolism , YAP-Signaling Proteins
13.
J Vis Exp ; (135)2018 05 14.
Article in English | MEDLINE | ID: mdl-29806834

ABSTRACT

Live-cell imaging is a powerful technique that can be used to directly visualize biological phenomena in single cells over extended periods of time. Over the past decade, new and innovative technologies have greatly enhanced the practicality of live-cell imaging. Cells can now be kept in focus and continuously imaged over several days while maintained under 37 °C and 5% CO2 cell culture conditions. Moreover, multiple fields of view representing different experimental conditions can be acquired simultaneously, thus providing high-throughput experimental data. Live-cell imaging provides a significant advantage over fixed-cell imaging by allowing for the direct visualization and temporal quantitation of dynamic cellular events. Live-cell imaging can also identify variation in the behavior of single cells that would otherwise have been missed using population-based assays. Here, we describe live-cell imaging protocols to assess cell fate decisions following treatment with the anti-mitotic drug paclitaxel. We demonstrate methods to visualize whether mitotically arrested cells die directly from mitosis or slip back into interphase. We also describe how the fluorescent ubiquitination-based cell cycle indicator (FUCCI) system can be used to assess the fraction of interphase cells born from mitotic slippage that are capable of re-entering the cell cycle. Finally, we describe a live-cell imaging method to identify nuclear envelope rupture events.


Subject(s)
Cell Line, Tumor/drug effects , Culture Techniques/methods , Paclitaxel/therapeutic use , Cell Differentiation , Humans , Paclitaxel/pharmacology
14.
Oncotarget ; 8(45): 78556-78572, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-29108249

ABSTRACT

Pancreatic ductal adenocarcinomas (PDACs) are highly aggressive malignancies, associated with poor clinical prognosis and limited therapeutic options. Oncogenic KRAS mutations are found in over 90% of PDACs, playing a central role in tumor progression. Global gene expression profiling of PDAC reveals 3-4 major molecular subtypes with distinct phenotypic traits and pharmacological vulnerabilities, including variations in oncogenic KRAS pathway dependencies. PDAC cell lines of the aberrantly differentiated endocrine exocrine (ADEX) subtype are robustly KRAS-dependent for survival. The KRAS gene is located on chromosome 12p11-12p12, a region amplified in 5-10% of primary PDACs. Within this amplicon, we identified co-amplification of KRAS with the STK38L gene in a subset of primary human PDACs and PDAC cell lines. Therefore, we determined whether PDAC cell lines are dependent on STK38L expression for proliferation and viability. STK38L encodes a serine/threonine kinase, which shares homology with Hippo pathway kinases LATS1/2. We show that STK38L expression is elevated in a subset of primary PDACs and PDAC cell lines displaying ADEX subtype characteristics, including overexpression of mutant KRAS. RNAi-mediated depletion of STK38L in a subset of ADEX subtype cell lines inhibits cellular proliferation and induces apoptosis. Concomitant with these effects, STK38L depletion causes increased expression of the LATS2 kinase and the cell cycle regulator p21. LATS2 depletion partially rescues the cytostatic and cytotoxic effects of STK38L depletion. Lastly, high STK38L mRNA expression is associated with decreased overall patient survival in PDACs. Collectively, our findings implicate STK38L as a candidate targetable vulnerability in a subset of molecularly-defined PDACs.

15.
Sci Transl Med ; 9(414)2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29093183

ABSTRACT

Despite intense efforts, the cure rates of childhood and adult solid tumors are not satisfactory. Resistance to intensive chemotherapy is common, and targets for molecular therapies are largely undefined. We have found that the majority of childhood solid tumors, including rhabdoid tumors, neuroblastoma, medulloblastoma, and Ewing sarcoma, express an active DNA transposase, PGBD5, that can promote site-specific genomic rearrangements in human cells. Using functional genetic approaches, we discovered that mouse and human cells deficient in nonhomologous end joining (NHEJ) DNA repair cannot tolerate the expression of PGBD5. In a chemical screen of DNA damage signaling inhibitors, we identified AZD6738 as a specific sensitizer of PGBD5-dependent DNA damage and apoptosis. We found that expression of PGBD5, but not its nuclease activity-deficient mutant, was sufficient to induce sensitivity to AZD6738. Depletion of endogenous PGBD5 conferred resistance to AZD6738 in human tumor cells. PGBD5-expressing tumor cells accumulated unrepaired DNA damage in response to AZD6738 treatment and underwent apoptosis in both dividing and G1-phase cells in the absence of immediate DNA replication stress. Accordingly, AZD6738 exhibited nanomolar potency against most neuroblastoma, medulloblastoma, Ewing sarcoma, and rhabdoid tumor cells tested while sparing nontransformed human and mouse embryonic fibroblasts in vitro. Finally, treatment with AZD6738 induced apoptosis and regression of human neuroblastoma and medulloblastoma tumors engrafted in immunodeficient mice in vivo. This effect was potentiated by combined treatment with cisplatin, including substantial antitumor activity against patient-derived primary neuroblastoma xenografts. These findings delineate a therapeutically actionable synthetic dependency induced in PGBD5-expressing solid tumors.


Subject(s)
DNA Repair/drug effects , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/pathology , Pyrimidines/therapeutic use , Sulfoxides/therapeutic use , Transposases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Child , DNA Damage , DNA End-Joining Repair/drug effects , Drug Synergism , Humans , Indoles , Mice , Mice, Nude , Models, Biological , Morpholines , Pyrimidines/pharmacology , Signal Transduction , Sulfonamides , Sulfoxides/pharmacology , Transposases/metabolism , Xenograft Model Antitumor Assays
16.
Mol Biol Cell ; 27(21): 3210-3213, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27799497

ABSTRACT

The nuclear envelope, composed of two lipid bilayers and numerous accessory proteins, has evolved to house the genetic material of all eukaryotic cells. In so doing, the nuclear envelope provides a physical barrier between chromosomes and the cytoplasm. Once believed to be highly stable, recent studies demonstrate that the nuclear envelope is prone to rupture. These rupture events expose chromosomal DNA to the cytoplasmic environment and have the capacity to promote DNA damage. Thus nuclear rupture may be an unappreciated mechanism of mutagenesis.


Subject(s)
Nuclear Envelope/metabolism , Nuclear Envelope/physiology , Cell Nucleus/metabolism , Cell Nucleus/physiology , Chromosomes/metabolism , Cytoplasm/metabolism , DNA Damage/physiology , Eukaryotic Cells/metabolism , Genomic Instability/genetics , Neoplasms/genetics , Nuclear Proteins/metabolism
17.
Methods Mol Biol ; 1413: 393-401, 2016.
Article in English | MEDLINE | ID: mdl-27193862

ABSTRACT

Tetraploid cells are genetically unstable and have the capacity to promote the development and/or progression of human malignancies. It is now estimated that ~40 % of all solid tumors have passed through a tetraploid intermediate stage at some point during their development. Understanding the biological characteristics of tetraploid cells that impart oncogenic properties is therefore a highly relevant and fundamentally important aspect of cancer biology. Here, we describe strategies to efficiently generate and purify tetraploid cells for use in cell biological studies.


Subject(s)
Cell Separation , Mitosis , Tetraploidy , Cell Cycle , Cell Line , Cytokinesis , Flow Cytometry , Humans
18.
Chromosome Res ; 24(1): 93-104, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26582635

ABSTRACT

Centrosome amplification is a common feature of both solid and hematological human malignancies. Extra centrosomes are not merely innocent bystanders in cancer cells, but rather promote tumor progression by disrupting normal cellular architecture and generating chromosome instability. Consequently, centrosome amplification correlates with advanced tumor grade and overall poor clinical prognosis. By contrast, extra centrosomes are adversely tolerated in non-transformed cells and hinder cell proliferation. This suggests that in addition to acquiring extra centrosomes, cancer cells must also adapt to overcome the deleterious consequences associated with them. Here, we review evidence that implicates core components of the Hippo tumor suppressor pathway as having key roles in both the direct and indirect regulation of centrosome number. Intriguingly, functional inactivation of the Hippo pathway, which is common across broad spectrum of human cancers, likely represents one key adaptation that enables cancer cells to tolerate extra centrosomes.


Subject(s)
Cell Proliferation , Centrosome/metabolism , Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Hippo Signaling Pathway , Humans , Neoplasms/genetics , Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Tumor Suppressor Proteins/genetics
19.
Environ Mol Mutagen ; 56(7): 563-80, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25784636

ABSTRACT

Genomic instability leads to a wide spectrum of genetic changes, including single nucleotide mutations, structural chromosome alterations, and numerical chromosome changes. The accepted view on how these events are generated predicts that separate cellular mechanisms and genetic events explain the occurrence of these types of genetic variation. Recently, new findings have shed light on the complexity of the mechanisms leading to structural and numerical chromosome aberrations, their intertwining pathways, and their dynamic evolution, in somatic as well as in germ cells. In this review, we present a critical analysis of these recent discoveries in this area, with the aim to contribute to a deeper knowledge of the molecular networks leading to adverse outcomes in humans following exposure to environmental factors. The review illustrates how several technological advances, including DNA sequencing methods, bioinformatics, and live-cell imaging approaches, have contributed to produce a renewed concept of the mechanisms causing genomic instability. Special attention is also given to the specific pathways causing genomic instability in mammalian germ cells. Remarkably, the same scenario emerged from some pioneering studies published in the 1980s to 1990s, when the evolution of polyploidy, the chromosomal effects of spindle poisons, the fate of micronuclei, were intuitively proposed to share mechanisms and pathways. Thus, an old working hypothesis has eventually found proper validation.


Subject(s)
Chromosomes, Human/genetics , Genomic Instability , Animals , Chromosome Aberrations , DNA Damage , DNA Replication , Germ Cells/physiology , Humans , Mitosis , Neoplasms/genetics , Neoplasms/pathology
20.
Science ; 347(6219): 273-7, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25593184

ABSTRACT

Cancer cells rely on telomerase or the alternative lengthening of telomeres (ALT) pathway to overcome replicative mortality. ALT is mediated by recombination and is prevalent in a subset of human cancers, yet whether it can be exploited therapeutically remains unknown. Loss of the chromatin-remodeling protein ATRX associates with ALT in cancers. Here, we show that ATRX loss compromises cell-cycle regulation of the telomeric noncoding RNA TERRA and leads to persistent association of replication protein A (RPA) with telomeres after DNA replication, creating a recombinogenic nucleoprotein structure. Inhibition of the protein kinase ATR, a critical regulator of recombination recruited by RPA, disrupts ALT and triggers chromosome fragmentation and apoptosis in ALT cells. The cell death induced by ATR inhibitors is highly selective for cancer cells that rely on ALT, suggesting that such inhibitors may be useful for treatment of ALT-positive cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Pyrazines/pharmacology , Sulfones/pharmacology , Telomere Homeostasis , Telomere/drug effects , Telomere/metabolism , Apoptosis , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle , Cell Line, Tumor , DNA Helicases/genetics , DNA Helicases/metabolism , Gene Knockdown Techniques , Glioma/drug therapy , Glioma/genetics , HeLa Cells , Homologous Recombination , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Promyelocytic Leukemia Protein , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Replication Protein A/metabolism , Telomerase/metabolism , Telomere/genetics , Telomeric Repeat Binding Protein 2/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , X-linked Nuclear Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...