Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 158(4): 833-848, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25126788

RESUMEN

Genetically unstable tetraploid cells can promote tumorigenesis. Recent estimates suggest that ∼37% of human tumors have undergone a genome-doubling event during their development. This potentially oncogenic effect of tetraploidy is countered by a p53-dependent barrier to proliferation. However, the cellular defects and corresponding signaling pathways that trigger growth suppression in tetraploid cells are not known. Here, we combine RNAi screening and in vitro evolution approaches to demonstrate that cytokinesis failure activates the Hippo tumor suppressor pathway in cultured cells, as well as in naturally occurring tetraploid cells in vivo. Induction of the Hippo pathway is triggered in part by extra centrosomes, which alter small G protein signaling and activate LATS2 kinase. LATS2 in turn stabilizes p53 and inhibits the transcriptional regulators YAP and TAZ. These findings define an important tumor suppression mechanism and uncover adaptive mechanisms potentially available to nascent tumor cells that bypass this inhibitory regulation.


Asunto(s)
Citocinesis , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Línea Celular Tumoral , Centrosoma/metabolismo , Células Epiteliales/metabolismo , Hepatocitos/metabolismo , Vía de Señalización Hippo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Tetraploidía , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
2.
Nature ; 619(7968): 184-192, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37286600

RESUMEN

Transcriptional heterogeneity due to plasticity of the epigenetic state of chromatin contributes to tumour evolution, metastasis and drug resistance1-3. However, the mechanisms that cause this epigenetic variation are incompletely understood. Here we identify micronuclei and chromosome bridges, aberrations in the nucleus common in cancer4,5, as sources of heritable transcriptional suppression. Using a combination of approaches, including long-term live-cell imaging and same-cell single-cell RNA sequencing (Look-Seq2), we identified reductions in gene expression in chromosomes from micronuclei. With heterogeneous penetrance, these changes in gene expression can be heritable even after the chromosome from the micronucleus has been re-incorporated into a normal daughter cell nucleus. Concomitantly, micronuclear chromosomes acquire aberrant epigenetic chromatin marks. These defects may persist as variably reduced chromatin accessibility and reduced gene expression after clonal expansion from single cells. Persistent transcriptional repression is strongly associated with, and may be explained by, markedly long-lived DNA damage. Epigenetic alterations in transcription may therefore be inherently coupled to chromosomal instability and aberrations in nuclear architecture.


Asunto(s)
Inestabilidad Cromosómica , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Micronúcleos con Defecto Cromosómico , Neoplasias , Transcripción Genética , Humanos , Cromatina/genética , Cromatina/metabolismo , Cromosomas/genética , Células Clonales/metabolismo , Daño del ADN/genética , Neoplasias/genética , Neoplasias/patología , Análisis de Expresión Génica de una Sola Célula
3.
Nature ; 618(7967): 1024-1032, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37198482

RESUMEN

Focal copy-number amplification is an oncogenic event. Although recent studies have revealed the complex structure1-3 and the evolutionary trajectories4 of oncogene amplicons, their origin remains poorly understood. Here we show that focal amplifications in breast cancer frequently derive from a mechanism-which we term translocation-bridge amplification-involving inter-chromosomal translocations that lead to dicentric chromosome bridge formation and breakage. In 780 breast cancer genomes, we observe that focal amplifications are frequently connected to each other by inter-chromosomal translocations at their boundaries. Subsequent analysis indicates the following model: the oncogene neighbourhood is translocated in G1 creating a dicentric chromosome, the dicentric chromosome is replicated, and as dicentric sister chromosomes segregate during mitosis, a chromosome bridge is formed and then broken, with fragments often being circularized in extrachromosomal DNAs. This model explains the amplifications of key oncogenes, including ERBB2 and CCND1. Recurrent amplification boundaries and rearrangement hotspots correlate with oestrogen receptor binding in breast cancer cells. Experimentally, oestrogen treatment induces DNA double-strand breaks in the oestrogen receptor target regions that are repaired by translocations, suggesting a role of oestrogen in generating the initial translocations. A pan-cancer analysis reveals tissue-specific biases in mechanisms initiating focal amplifications, with the breakage-fusion-bridge cycle prevalent in some and the translocation-bridge amplification in others, probably owing to the different timing of DNA break repair. Our results identify a common mode of oncogene amplification and propose oestrogen as its mechanistic origin in breast cancer.


Asunto(s)
Neoplasias de la Mama , Receptor alfa de Estrógeno , Amplificación de Genes , Oncogenes , Translocación Genética , Femenino , Humanos , Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Oncogenes/genética , Translocación Genética/genética , Genoma Humano/genética , Roturas del ADN de Doble Cadena , Especificidad de Órganos
4.
Nature ; 606(7916): 930-936, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35477155

RESUMEN

Chromothripsis is a catastrophic mutational process that promotes tumorigenesis and causes congenital disease1-4. Chromothripsis originates from aberrations of nuclei called micronuclei or chromosome bridges5-8. These structures are associated with fragile nuclear envelopes that spontaneously rupture9,10, leading to DNA damage when chromatin is exposed to the interphase cytoplasm. Here we identify a mechanism explaining a major fraction of this DNA damage. Micronuclei accumulate large amounts of RNA-DNA hybrids, which are edited by adenine deaminases acting on RNA (ADAR enzymes) to generate deoxyinosine. Deoxyinosine is then converted into abasic sites by a DNA base excision repair (BER) glycosylase, N-methyl-purine DNA glycosylase11,12 (MPG). These abasic sites are cleaved by the BER endonuclease, apurinic/apyrimidinic endonuclease12 (APE1), creating single-stranded DNA nicks that can be converted to DNA double strand breaks by DNA replication or when closely spaced nicks occur on opposite strands13,14. This model predicts that MPG should be able to remove the deoxyinosine base from the DNA strand of RNA-DNA hybrids, which we demonstrate using purified proteins and oligonucleotide substrates. These findings identify a mechanism for fragmentation of micronuclear chromosomes, an important step in generating chromothripsis. Rather than breaking any normal chromosome, we propose that the eukaryotic cytoplasm only damages chromosomes with pre-existing defects such as the DNA base abnormality described here.


Asunto(s)
Cromosomas , Cromotripsis , Citoplasma , Daño del ADN , Reparación del ADN , Cromatina/metabolismo , Cromatina/patología , Cromosomas/metabolismo , Citoplasma/metabolismo , ADN/metabolismo , ADN Glicosilasas/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , ARN/metabolismo
5.
Cell ; 150(1): 151-64, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22727045

RESUMEN

Cellular wound healing, enabling the repair of membrane damage, is ubiquitous in eukaryotes. One aspect of the wound healing response is the redirection of a polarized cytoskeleton and the secretory machinery to the damage site. Although there has been recent progress in identifying conserved proteins involved in wound healing, the mechanisms linking these components into a coherent response are not defined. Using laser damage in budding yeast, we demonstrate that local cell wall/membrane damage triggers the dispersal of proteins from the site of polarized growth, enabling their accumulation at the wound. We define a protein-kinase-C-dependent mechanism that mediates the destruction of the formin Bni1 and the exocyst component Sec3. This degradation is essential to prevent competition between the site of polarized growth and the wound. Mechanisms to overcome competition from a pre-existing polarized cytoskeleton may be a general feature of effective wound healing in polarized cells.


Asunto(s)
Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/fisiología , Polaridad Celular , Citoesqueleto/metabolismo , Eucariontes/citología , Eucariontes/fisiología , Proteínas de Microfilamentos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
6.
Mol Cell ; 73(5): 915-929.e6, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30849395

RESUMEN

DNA replication errors generate complex chromosomal rearrangements and thereby contribute to tumorigenesis and other human diseases. One mechanism that triggers these errors is mitotic entry before the completion of DNA replication. To address how mitosis might affect DNA replication, we used Xenopus egg extracts. When mitotic CDK (Cyclin B1-CDK1) is used to drive interphase egg extracts into a mitotic state, the replicative CMG (CDC45/MCM2-7/GINS) helicase undergoes ubiquitylation on its MCM7 subunit, dependent on the E3 ubiquitin ligase TRAIP. Whether replisomes have stalled or undergone termination, CMG ubiquitylation is followed by its extraction from chromatin by the CDC48/p97 ATPase. TRAIP-dependent CMG unloading during mitosis is also seen in C. elegans early embryos. At stalled forks, CMG removal results in fork breakage and end joining events involving deletions and templated insertions. Our results identify a mitotic pathway of global replisome disassembly that can trigger replication fork collapse and DNA rearrangements.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ciclina B1/metabolismo , Daño del ADN , Replicación del ADN , ADN/biosíntesis , Reordenamiento Génico , Mitosis , Proteínas Quinasas/metabolismo , Proteínas de Xenopus/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ciclo Celular/genética , Ciclina B1/genética , ADN/genética , Reparación del ADN , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Proteínas de Mantenimiento de Minicromosoma/genética , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Proteínas de Xenopus/genética , Xenopus laevis/genética , Xenopus laevis/metabolismo , ADN Polimerasa theta
7.
Cell ; 144(1): 9-10, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21215363

RESUMEN

A report in this issue describes "chromothripsis," a new mechanism for genetic instability in cancer cells. Chromothripsis appears to be a cataclysmic event in which a single chromosome is fragmented and then reassembled. The phenomenon raises important questions of how chromosome rearrangements can be confined to defined genome segments.

8.
Cell ; 140(1): 74-87, 2010 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-20074521

RESUMEN

We report that eight heterozygous missense mutations in TUBB3, encoding the neuron-specific beta-tubulin isotype III, result in a spectrum of human nervous system disorders that we now call the TUBB3 syndromes. Each mutation causes the ocular motility disorder CFEOM3, whereas some also result in intellectual and behavioral impairments, facial paralysis, and/or later-onset axonal sensorimotor polyneuropathy. Neuroimaging reveals a spectrum of abnormalities including hypoplasia of oculomotor nerves and dysgenesis of the corpus callosum, anterior commissure, and corticospinal tracts. A knock-in disease mouse model reveals axon guidance defects without evidence of cortical cell migration abnormalities. We show that the disease-associated mutations can impair tubulin heterodimer formation in vitro, although folded mutant heterodimers can still polymerize into microtubules. Modeling each mutation in yeast tubulin demonstrates that all alter dynamic instability whereas a subset disrupts the interaction of microtubules with kinesin motors. These findings demonstrate that normal TUBB3 is required for axon guidance and maintenance in mammals.


Asunto(s)
Tubulina (Proteína)/metabolismo , Secuencia de Aminoácidos , Animales , Axones/metabolismo , Encéfalo/embriología , Encéfalo/metabolismo , Supervivencia Celular , Niño , Discapacidades del Desarrollo , Femenino , Humanos , Cinesinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microtúbulos/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Mutación Missense , Transporte de Proteínas , Tubulina (Proteína)/química , Tubulina (Proteína)/genética
9.
Nature ; 561(7724): 551-555, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30232450

RESUMEN

Defects in the architecture or integrity of the nuclear envelope are associated with a variety of human diseases1. Micronuclei, one common nuclear aberration, are an origin for chromothripsis2, a catastrophic mutational process that is commonly observed in cancer3-5. Chromothripsis occurs after micronuclei spontaneously lose nuclear envelope integrity, which generates chromosome fragmentation6. Disruption of the nuclear envelope exposes DNA to the cytoplasm and initiates innate immune proinflammatory signalling7. Despite its importance, the basis of the fragility of the micronucleus nuclear envelope  is not known. Here we show that micronuclei undergo defective nuclear envelope assembly. Only 'core' nuclear envelope proteins8,9 assemble efficiently on lagging chromosomes, whereas 'non-core' nuclear envelope proteins8,9, including nuclear pore complexes (NPCs), do not. Consequently, micronuclei fail to properly import key proteins that are necessary for the integrity of the nuclear envelope and genome. We show that spindle microtubules block assembly of NPCs and other non-core nuclear envelope proteins on lagging chromosomes, causing an irreversible defect in nuclear envelope assembly. Accordingly, experimental manipulations that position missegregated chromosomes away from the spindle correct defective nuclear envelope assembly, prevent spontaneous nuclear envelope disruption, and suppress DNA damage in micronuclei. Thus, during mitotic exit in metazoan cells, chromosome segregation and nuclear envelope assembly are only loosely coordinated by the timing of mitotic spindle disassembly. The absence of precise checkpoint controls may explain why errors during mitotic exit are frequent and often trigger catastrophic genome rearrangements4,5.


Asunto(s)
Cromotripsis , Micronúcleos con Defecto Cromosómico , Mitosis , Membrana Nuclear/metabolismo , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Segregación Cromosómica , Cromosomas Humanos/metabolismo , ADN/metabolismo , Daño del ADN , Inestabilidad Genómica , Humanos , Microtúbulos/metabolismo , Poro Nuclear/metabolismo , Huso Acromático/metabolismo
10.
Annu Rev Genet ; 49: 183-211, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26442848

RESUMEN

Chromosomal rearrangements are generally thought to accumulate gradually over many generations. However, DNA sequencing of cancer and congenital disorders uncovered a new pattern in which multiple rearrangements arise all at once. The most striking example, chromothripsis, is characterized by tens or hundreds of rearrangements confined to a single chromosome or to local regions over a few chromosomes. Genomic analysis of chromothripsis and the search for its biological mechanism have led to new insights on how chromosome segregation errors can generate mutagenesis and changes to the karyotype. Here, we review the genomic features of chromothripsis and summarize recent progress on understanding its mechanism. This includes reviewing new work indicating that one mechanism to generate chromothripsis is through the physical isolation of chromosomes in abnormal nuclear structures (micronuclei). We also discuss connections revealed by recent genomic analysis of cancers between chromothripsis, chromosome bridges, and ring chromosomes.


Asunto(s)
Evolución Biológica , Cromosomas/genética , Cariotipo , Mutagénesis/fisiología , Cromosomas Humanos/genética , Reordenamiento Génico , Humanos , Micronúcleos con Defecto Cromosómico , Membrana Nuclear/genética , Cromosomas en Anillo
11.
Mol Cell ; 54(3): 512-25, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24703952

RESUMEN

Excluding 53BP1 from chromatin is required to attenuate the DNA damage response during mitosis, yet the functional relevance and regulation of this exclusion are unclear. Here we show that 53BP1 is phosphorylated during mitosis on two residues, T1609 and S1618, located in its well-conserved ubiquitination-dependent recruitment (UDR) motif. Phosphorylating these sites blocks the interaction of the UDR motif with mononuclesomes containing ubiquitinated histone H2A and impedes binding of 53BP1 to mitotic chromatin. Ectopic recruitment of 53BP1-T1609A/S1618A to mitotic DNA lesions was associated with significant mitotic defects that could be reversed by inhibiting nonhomologous end-joining. We also reveal that protein phosphatase complex PP4C/R3ß dephosphorylates T1609 and S1618 to allow the recruitment of 53BP1 to chromatin in G1 phase. Our results identify key sites of 53BP1 phosphorylation during mitosis, identify the counteracting phosphatase complex that restores the potential for DDR during interphase, and establish the physiological importance of this regulation.


Asunto(s)
Roturas del ADN de Doble Cadena , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Procesamiento Proteico-Postraduccional , Secuencia de Aminoácidos , Fase G1 , Células HeLa , Humanos , Mitosis , Datos de Secuencia Molecular , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación , Unión Proteica , Transporte de Proteínas , Proteína 1 de Unión al Supresor Tumoral P53
12.
RNA ; 25(3): 352-363, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30538148

RESUMEN

The accurate inheritance of genetic material is a basic necessity in all domains of life and an unexpectedly large number of RNA processing factors are required for mitotic progression and genome stability. NRDE2 (nuclear RNAi defective-2) is an evolutionarily conserved protein originally discovered for its role in nuclear RNA interference (RNAi) and heritable gene silencing in Caenorhabditis elegans (C. elegans). The function of the human NRDE2 gene remains poorly understood. Here we show that human NRDE2 is an essential protein required for suppressing intron retention in a subset of pre-mRNAs containing short, GC-rich introns with relatively weak 5' and 3' splice sites. NRDE2 preferentially interacts with components of the U5 small nuclear ribonucleoprotein (snRNP), the exon junction complex, and the RNA exosome. Interestingly, NRDE2-depleted cells exhibit greatly increased levels of genomic instability and DNA damage, as well as defects in centrosome maturation and mitotic progression. We identify the essential centriolar satellite protein, CEP131, as a direct NRDE2-regulated target. NRDE2 specifically binds to and promotes the efficient splicing of CEP131 pre-mRNA, and depleting NRDE2 dramatically reduces CEP131 protein expression, contributing to impaired recruitment of critical centrosomal proteins (e.g., γ-tubulin and Aurora Kinase A) to the spindle poles during mitosis. Our work establishes a conserved role for human NRDE2 in RNA splicing, characterizes the severe genomic instability phenotypes observed upon loss of NRDE2, and highlights the direct regulation of CEP131 splicing as one of multiple mechanisms through which such phenotypes might be explained.


Asunto(s)
Factores de Empalme de ARN/metabolismo , Empalme del ARN , Proteínas de Unión al ARN/metabolismo , Línea Celular , Regulación de la Expresión Génica , Humanos , Intrones , Interferencia de ARN , Precursores del ARN/genética , Precursores del ARN/metabolismo , ARN Interferente Pequeño/genética
13.
Nature ; 522(7555): 179-84, 2015 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-26017310

RESUMEN

Genome sequencing has uncovered a new mutational phenomenon in cancer and congenital disorders called chromothripsis. Chromothripsis is characterized by extensive genomic rearrangements and an oscillating pattern of DNA copy number levels, all curiously restricted to one or a few chromosomes. The mechanism for chromothripsis is unknown, but we previously proposed that it could occur through the physical isolation of chromosomes in aberrant nuclear structures called micronuclei. Here, using a combination of live cell imaging and single-cell genome sequencing, we demonstrate that micronucleus formation can indeed generate a spectrum of genomic rearrangements, some of which recapitulate all known features of chromothripsis. These events are restricted to the mis-segregated chromosome and occur within one cell division. We demonstrate that the mechanism for chromothripsis can involve the fragmentation and subsequent reassembly of a single chromatid from a micronucleus. Collectively, these experiments establish a new mutational process of which chromothripsis is one extreme outcome.


Asunto(s)
Rotura Cromosómica , Daño del ADN , Micronúcleos con Defecto Cromosómico , Línea Celular , Supervivencia Celular , Segregación Cromosómica/genética , Variaciones en el Número de Copia de ADN/genética , Reordenamiento Génico/genética , Inestabilidad Genómica/genética , Humanos , Mutación/genética , Neoplasias/genética , Fase S/genética , Análisis de la Célula Individual
14.
Nature ; 519(7543): 349-52, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25731168

RESUMEN

Polyploidy is observed across the tree of life, yet its influence on evolution remains incompletely understood. Polyploidy, usually whole-genome duplication, is proposed to alter the rate of evolutionary adaptation. This could occur through complex effects on the frequency or fitness of beneficial mutations. For example, in diverse cell types and organisms, immediately after a whole-genome duplication, newly formed polyploids missegregate chromosomes and undergo genetic instability. The instability following whole-genome duplications is thought to provide adaptive mutations in microorganisms and can promote tumorigenesis in mammalian cells. Polyploidy may also affect adaptation independently of beneficial mutations through ploidy-specific changes in cell physiology. Here we perform in vitro evolution experiments to test directly whether polyploidy can accelerate evolutionary adaptation. Compared with haploids and diploids, tetraploids undergo significantly faster adaptation. Mathematical modelling suggests that rapid adaptation of tetraploids is driven by higher rates of beneficial mutations with stronger fitness effects, which is supported by whole-genome sequencing and phenotypic analyses of evolved clones. Chromosome aneuploidy, concerted chromosome loss, and point mutations all provide large fitness gains. We identify several mutations whose beneficial effects are manifest specifically in the tetraploid strains. Together, these results provide direct quantitative evidence that in some environments polyploidy can accelerate evolutionary adaptation.


Asunto(s)
Adaptación Fisiológica/genética , Evolución Biológica , Poliploidía , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/fisiología , Aneuploidia , Cromosomas Fúngicos/genética , Células Clonales/citología , Células Clonales/metabolismo , Diploidia , Aptitud Genética/genética , Haploidia , Tasa de Mutación , Mutación Puntual/genética , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/metabolismo , Factores de Tiempo
15.
Genes Dev ; 27(23): 2513-30, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24298051

RESUMEN

Recent genome sequencing studies have identified several classes of complex genomic rearrangements that appear to be derived from a single catastrophic event. These discoveries identify ways that genomes can be altered in single large jumps rather than by many incremental steps. Here we compare and contrast these phenomena and examine the evidence that they arise "all at once." We consider the impact of massive chromosomal change for the development of diseases such as cancer and for evolution more generally. Finally, we summarize current models for underlying mechanisms and discuss strategies for testing these models.


Asunto(s)
Evolución Molecular , Reordenamiento Génico/genética , Genoma Humano/genética , Aberraciones Cromosómicas , Inestabilidad Genómica , Humanos
16.
Nature ; 510(7503): 167-71, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24739973

RESUMEN

Centrosome amplification has long been recognized as a feature of human tumours; however, its role in tumorigenesis remains unclear. Centrosome amplification is poorly tolerated by non-transformed cells and, in the absence of selection, extra centrosomes are spontaneously lost. Thus, the high frequency of centrosome amplification, particularly in more aggressive tumours, raises the possibility that extra centrosomes could, in some contexts, confer advantageous characteristics that promote tumour progression. Using a three-dimensional model system and other approaches to culture human mammary epithelial cells, we find that centrosome amplification triggers cell invasion. This invasive behaviour is similar to that induced by overexpression of the breast cancer oncogene ERBB2 (ref. 4) and indeed enhances invasiveness triggered by ERBB2. Our data indicate that, through increased centrosomal microtubule nucleation, centrosome amplification increases Rac1 activity, which disrupts normal cell-cell adhesion and promotes invasion. These findings demonstrate that centrosome amplification, a structural alteration of the cytoskeleton, can promote features of malignant transformation.


Asunto(s)
Neoplasias de la Mama/patología , Transformación Celular Neoplásica/patología , Centrosoma/patología , Genes erbB-2 , Aneuploidia , Mama/citología , Mama/patología , Neoplasias de la Mama/genética , Adhesión Celular , Línea Celular , Transformación Celular Neoplásica/genética , Progresión de la Enfermedad , Activación Enzimática , Células Epiteliales/citología , Células Epiteliales/patología , Humanos , Microtúbulos/química , Microtúbulos/metabolismo , Microtúbulos/patología , Invasividad Neoplásica/patología , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Proteína de Unión al GTP rac1/metabolismo
17.
Blood ; 129(4): 497-508, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-27756750

RESUMEN

Acute myeloid leukemia (AML) is a heterogeneous disease with complex molecular pathophysiology. To systematically characterize AML's genetic dependencies, we conducted genome-scale short hairpin RNA screens in 17 AML cell lines and analyzed dependencies relative to parallel screens in 199 cell lines of other cancer types. We identified 353 genes specifically required for AML cell proliferation. To validate the in vivo relevance of genetic dependencies observed in human cell lines, we performed a secondary screen in a syngeneic murine AML model driven by the MLL-AF9 oncogenic fusion protein. Integrating the results of these interference RNA screens and additional gene expression data, we identified the transcription factor ZEB2 as a novel AML dependency. ZEB2 depletion impaired the proliferation of both human and mouse AML cells and resulted in aberrant differentiation of human AML cells. Mechanistically, we showed that ZEB2 transcriptionally represses genes that regulate myeloid differentiation, including genes involved in cell adhesion and migration. In addition, we found that epigenetic silencing of the miR-200 family microRNAs affects ZEB2 expression. Our results extend the role of ZEB2 beyond regulating epithelial-mesenchymal transition (EMT) and establish ZEB2 as a novel regulator of AML proliferation and differentiation.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Proteínas de Homeodominio/genética , Leucemia Mieloide Aguda/genética , MicroARNs/genética , Proteínas Represoras/genética , Animales , Adhesión Celular , Diferenciación Celular , Línea Celular Tumoral , Movimiento Celular , Epigénesis Genética , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/metabolismo , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , MicroARNs/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Transducción de Señal , Transcripción Genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc
18.
Mol Cell ; 43(5): 751-63, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21884976

RESUMEN

The kinesin-8 family of microtubule motors plays a critical role in microtubule length control in cells. These motors have complex effects on microtubule dynamics: they destabilize growing microtubules yet stabilize shrinking microtubules. The budding yeast kinesin-8, Kip3, accumulates on plus ends of growing but not shrinking microtubules. Here we identify an essential role of the tail domain of Kip3 in mediating both its destabilizing and its stabilizing activities. The Kip3 tail promotes Kip3's accumulation at the plus ends and facilitates the destabilizing effect of Kip3. However, the Kip3 tail also inhibits microtubule shrinkage and is required for promoting microtubule rescue by Kip3. These effects of the tail domain are likely to be mediated by the tubulin- and microtubule-binding activities that we describe. We propose a concentration-dependent model for the coordination of the destabilizing and stabilizing activities of Kip3 and discuss its relevance to cellular microtubule organization.


Asunto(s)
Cinesinas/metabolismo , Microtúbulos/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Humanos , Cinesinas/química , Cinesinas/genética , Modelos Biológicos , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo
19.
Nat Rev Genet ; 13(3): 189-203, 2012 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-22269907

RESUMEN

Genetic instability, which includes both numerical and structural chromosomal abnormalities, is a hallmark of cancer. Whereas the structural chromosome rearrangements have received substantial attention, the role of whole-chromosome aneuploidy in cancer is much less well-understood. Here we review recent progress in understanding the roles of whole-chromosome aneuploidy in cancer, including the mechanistic causes of aneuploidy, the cellular responses to chromosome gains or losses and how cells might adapt to tolerate these usually detrimental alterations. We also explore the role of aneuploidy in cellular transformation and discuss the possibility of developing aneuploidy-specific therapies.


Asunto(s)
Aneuploidia , Transformación Celular Neoplásica/patología , Inestabilidad Cromosómica , Aberraciones Cromosómicas , Neoplasias/genética , Humanos , Neoplasias/terapia
20.
Nature ; 550(7676): 340-341, 2017 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-28976957

Asunto(s)
Neoplasias , Biología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA