Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Cell Biol ; 222(2)2023 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-36459065

RESUMEN

α/ß-Tubulin posttranslational modifications (PTMs) generate microtubule diversity, but whether they account for cancer cell resistance to microtubule-targeting drugs remains unknown. Here, we performed a pilot dissection of the "cancer tubulin code" using the NCI-60 cancer cell panel. We found that acetylated, detyrosinated, and ∆2-α-tubulin that typically accumulate on stable microtubules were uncoupled in many cancer cells. Acetylated α-tubulin did not affect microtubule dynamics, whereas its levels correlated with, but were not required for, taxol-induced cytotoxicity. In contrast, experimental increase of α-tubulin detyrosination, and/or depletion of the detyrosination-sensitive microtubule-depolymerizing enzyme MCAK, enhanced taxol-induced cytotoxicity by promoting cell death in mitosis and the subsequent interphase, without causing a cumulative effect. Interestingly, only increased detyrosinated α-tubulin aggravated taxol-induced spindle multipolarity. Overall, we identified high α-tubulin acetylation as a potential biomarker for cancer cell response to taxol and uncovered a mechanistic link between α-tubulin detyrosination and the suppression of MCAK activity in taxol-induced cytotoxicity, likely by promoting chromosome missegregation, regardless of spindle defects.


Asunto(s)
Cinesinas , Microtúbulos , Paclitaxel , Tubulina (Proteína) , Mitosis , Paclitaxel/farmacología , Procesamiento Proteico-Postraduccional , Tubulina (Proteína)/metabolismo , Humanos , Línea Celular Tumoral , Cinesinas/metabolismo
2.
Curr Biol ; 32(19): 4240-4254.e5, 2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-36057259

RESUMEN

Chromosome alignment to the spindle equator is a hallmark of mitosis thought to promote chromosome segregation fidelity in metazoans. Yet chromosome alignment is only indirectly supervised by the spindle assembly checkpoint (SAC) as a byproduct of chromosome bi-orientation, and the consequences of defective chromosome alignment remain unclear. Here, we investigated how human cells respond to chromosome alignment defects of distinct molecular nature by following the fate of live HeLa cells after RNAi-mediated depletion of 125 proteins previously implicated in chromosome alignment. We confirmed chromosome alignment defects upon depletion of 108/125 proteins. Surprisingly, in all confirmed cases, depleted cells frequently entered anaphase after a delay with misaligned chromosomes. Using depletion of prototype proteins resulting in defective chromosome alignment, we show that misaligned chromosomes often satisfy the SAC and directly missegregate without lagging behind in anaphase. In-depth analysis of specific molecular perturbations that prevent proper kinetochore-microtubule attachments revealed that misaligned chromosomes that missegregate frequently result in micronuclei. Higher-resolution live-cell imaging indicated that, contrary to most anaphase lagging chromosomes that correct and reintegrate the main nuclei, misaligned chromosomes are a strong predictor of micronuclei formation in a cancer cell model of chromosomal instability, but not in non-transformed near-diploid cells. We provide evidence supporting that intrinsic differences in kinetochore-microtubule attachment stability on misaligned chromosomes account for this distinct outcome. Thus, misaligned chromosomes that satisfy the SAC may represent a previously overlooked mechanism driving chromosomal/genomic instability during cancer cell division, and we unveil genetic conditions predisposing for these events.


Asunto(s)
Cinetocoros , Neoplasias , Segregación Cromosómica , Cromosomas , Células HeLa , Humanos , Puntos de Control de la Fase M del Ciclo Celular , Mitosis , Neoplasias/metabolismo , Huso Acromático/metabolismo
3.
Cell Rep ; 37(6): 109783, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34758324

RESUMEN

Micronuclei are a hallmark of cancer and several other human disorders. Recently, micronuclei were implicated in chromothripsis, a series of massive genomic rearrangements that may drive tumor evolution and progression. Here, we show that Aurora B kinase mediates a surveillance mechanism that integrates error correction during anaphase with spatial control of nuclear envelope reassembly to prevent micronuclei formation. Using high-resolution live-cell imaging of human cancer and non-cancer cells, we uncover that anaphase lagging chromosomes are more frequent than previously anticipated, yet they rarely form micronuclei. Micronuclei formation from anaphase lagging chromosomes is prevented by a midzone-based Aurora B phosphorylation gradient that stabilizes kinetochore-microtubule attachments and assists spindle forces required for anaphase error correction while delaying nuclear envelope reassembly on lagging chromosomes, independently of microtubule density. We propose that a midzone-based Aurora B phosphorylation gradient actively monitors and corrects frequent chromosome segregation errors to prevent micronuclei formation during human cell division.


Asunto(s)
Anafase , Aurora Quinasa B/metabolismo , Segregación Cromosómica , Cinetocoros/enzimología , Micronúcleos con Defecto Cromosómico , Membrana Nuclear/enzimología , Huso Acromático/enzimología , Células HeLa , Humanos , Mecanotransducción Celular , Membrana Nuclear/genética , Fosforilación , Huso Acromático/genética , Factores de Tiempo
4.
Methods Cell Biol ; 158: 91-116, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32423652

RESUMEN

During mitosis, spindle microtubules dynamically attach to and detach from kinetochores in a precise and regulated fashion. To ensure mitotic fidelity, kinetochore-microtubule (k-MT) attachments must be stable enough to satisfy the spindle assembly checkpoint (SAC), but sufficiently unstable to facilitate the correction of maloriented attachments. Different methods are available to assess k-MT stability in both live and fixed cells, but a comparative survey of these methods has not yet been reported. Here, we evaluate several quantitative and semiquantitative methods for determining k-MT stability and apply each technique to illustrate changes in spindle microtubule dynamics upon perturbation with physiologically relevant concentrations of microtubule stabilizing (Taxol) and destabilizing (UMK57 and nocodazole) compounds. We discuss the utility of each technique for defining specific features of spindle microtubule dynamics and k-MT attachment stability.


Asunto(s)
Técnicas Citológicas/métodos , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Calcio/farmacología , Línea Celular , Respuesta al Choque por Frío/efectos de los fármacos , Humanos , Imagenología Tridimensional , Cinetocoros/efectos de los fármacos , Luz , Microtúbulos/efectos de los fármacos , Nocodazol/farmacología , Huso Acromático/efectos de los fármacos , Huso Acromático/metabolismo
5.
J Cell Biol ; 219(4)2020 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-32328631

RESUMEN

Incorrect kinetochore-microtubule attachments during mitosis can lead to chromosomal instability, a hallmark of human cancers. Mitotic error correction relies on the kinesin-13 MCAK, a microtubule depolymerase whose activity in vitro is suppressed by α-tubulin detyrosination-a posttranslational modification enriched on long-lived microtubules. However, whether and how MCAK activity required for mitotic error correction is regulated by α-tubulin detyrosination remains unknown. Here we found that detyrosinated α-tubulin accumulates on correct, more stable, kinetochore-microtubule attachments. Experimental manipulation of tubulin tyrosine ligase (TTL) or carboxypeptidase (Vasohibins-SVBP) activities to constitutively increase α-tubulin detyrosination near kinetochores compromised efficient error correction, without affecting overall kinetochore microtubule stability. Rescue experiments indicate that MCAK centromeric activity was required and sufficient to correct the mitotic errors caused by excessive α-tubulin detyrosination independently of its global impact on microtubule dynamics. Thus, microtubules are not just passive elements during mitotic error correction, and the extent of α-tubulin detyrosination allows centromeric MCAK to discriminate correct vs. incorrect kinetochore-microtubule attachments, thereby promoting mitotic fidelity.


Asunto(s)
Centrómero/metabolismo , Cinesinas/metabolismo , Mitosis , Tubulina (Proteína)/metabolismo , Línea Celular Tumoral , Humanos , Microtúbulos/metabolismo
6.
Elife ; 82019 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-31424385

RESUMEN

According to the prevailing 'clock' model, chromosome decondensation and nuclear envelope reformation when cells exit mitosis are byproducts of Cdk1 inactivation at the metaphase-anaphase transition, controlled by the spindle assembly checkpoint. However, mitotic exit was recently shown to be a function of chromosome separation during anaphase, assisted by a midzone Aurora B phosphorylation gradient - the 'ruler' model. Here we found that Cdk1 remains active during anaphase due to ongoing APC/CCdc20- and APC/CCdh1-mediated degradation of B-type Cyclins in Drosophila and human cells. Failure to degrade B-type Cyclins during anaphase prevented mitotic exit in a Cdk1-dependent manner. Cyclin B1-Cdk1 localized at the spindle midzone in an Aurora B-dependent manner, with incompletely separated chromosomes showing the highest Cdk1 activity. Slowing down anaphase chromosome motion delayed Cyclin B1 degradation and mitotic exit in an Aurora B-dependent manner. Thus, a crosstalk between molecular 'rulers' and 'clocks' licenses mitotic exit only after proper chromosome separation.


Asunto(s)
Anafase , Aurora Quinasa B/metabolismo , Proteína Quinasa CDC2/metabolismo , Ciclina B1/metabolismo , Proteínas de Drosophila/metabolismo , Animales , Línea Celular , Drosophila , Humanos , Proteolisis , Análisis Espacio-Temporal
7.
Cell Cycle ; 18(15): 1702-1713, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31179849

RESUMEN

Chromosomal instability (CIN) is defined as a high rate of whole chromosome loss or gain and is a hallmark of many aneuploid solid tumors. CIN positively correlates with poor patient prognosis and chemotherapeutic resistance. Despite this clinical importance, the role of CIN in tumor initiation, growth and/or progression remains poorly understood. To date, the only strategies developed to determine how CIN contributes to tumorigenesis have relied on transgenic mouse models that deliberately increase the rate of chromosomal mis-segregation. Here we develop a strain of transgenic mice that is designed to strategically decrease the rate of chromosome mis-segregation and suppress CIN. These animals modestly overexpress the kinesin-13 microtubule depolymerase Kif2b, a strategy proven successful in restoring faithful chromosome segregation to human cancer cells in culture. Using the LA2 K-Ras G12D-induced model for lung cancer, we show that Kif2b expression reduces the number of chromosome segregation defects but does not change the incidence of lung tumor lesions. However, pulmonary tumors were significantly larger in animals expressing Kif2b and those tumors exhibited elevated rates of Ki-67 positive cells relative to controls. Thus, in lung cancers driven by mutations in K-Ras, CIN has little impact on tumor initiation but suppresses tumor growth. These data support a model in which CIN imposes a burden on tumor cells, and that enhancement of mitotic fidelity results in accelerated tumor growth.


Asunto(s)
Adenoma/genética , Transformación Celular Neoplásica/genética , Inestabilidad Cromosómica/genética , Cinesinas/metabolismo , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Adenoma/metabolismo , Adenoma/patología , Aneuploidia , Animales , Línea Celular , Transformación Celular Neoplásica/metabolismo , Segregación Cromosómica/genética , Fibroblastos , Cinesinas/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas p21(ras)/genética , Regulación hacia Arriba
8.
J Cell Biol ; 218(4): 1086-1088, 2019 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-30858193

RESUMEN

Chromosome alignment is a hallmark of mitosis in metazoans, but the physiological relevance of this orderly behavior has remained unclear. In this issue, Fonseca et al. (2019. J. Cell Biol. https://doi.org/10.1083/jcb.201807228) show that chromosome alignment ensures mitotic fidelity by promoting interchromosomal compaction during anaphase.


Asunto(s)
Anafase , Cromosomas , Metafase , Mitosis
9.
Methods Cell Biol ; 144: 33-74, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29804676

RESUMEN

Mitosis is an essential process that takes place in all eukaryotes and involves the equal division of genetic material from a parental cell into two identical daughter cells. During mitosis, chromosome movement and segregation are orchestrated by a specialized structure known as the mitotic spindle, composed of a bipolar array of microtubules. The fundamental structure of microtubules comprises of α/ß-tubulin heterodimers that associate head-to-tail and laterally to form hollow filaments. In vivo, microtubules are modified by abundant and evolutionarily conserved tubulin posttranslational modifications (PTMs), giving these filaments the potential for a wide chemical diversity. In recent years, the concept of a "tubulin code" has emerged as an extralayer of regulation governing microtubule function. A range of tubulin isoforms, each with a diverse set of PTMs, provides a readable code for microtubule motors and other microtubule-associated proteins. This chapter focuses on the complexity of tubulin PTMs with an emphasis on detyrosination and summarizes the methods currently used in our laboratory to experimentally manipulate these modifications and study their impact in mitosis.


Asunto(s)
Técnicas Citológicas/métodos , Mitosis , Tubulina (Proteína)/metabolismo , Línea Celular Tumoral , Humanos , Lentivirus/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteínas Motoras Moleculares/metabolismo , Procesamiento Proteico-Postraduccional , ARN Interferente Pequeño/metabolismo , Transducción Genética , Tirosina/metabolismo
10.
Cell Rep ; 17(7): 1755-1763, 2016 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-27829147

RESUMEN

Karyotype diversity is a hallmark of solid tumors that contributes to intratumor heterogeneity. This diversity is generated by persistent chromosome mis-segregation associated with chromosomal instability (CIN). CIN correlates with tumor relapse and is thought to promote drug resistance by creating a vast genomic landscape through which karyotypically unique clones survive lethal drug selection. We explore this proposition using a small molecule (UMK57) that suppresses chromosome mis-segregation in CIN cancer cells by potentiating the activity of the kinesin-13 protein MCAK. Sublethal doses of UMK57 destabilize kinetochore-microtubule (k-MT) attachments during mitosis to increase chromosome segregation fidelity. Surprisingly, chromosome mis-segregation rebounds in UMK57-treated cancer cells within a few days. This rapid relapse is driven by alterations in the Aurora B signaling pathway that hyper-stabilize k-MT attachments and is reversible following UMK57 removal. Thus, cancer cells display adaptive resistance to therapies targeting CIN through rapid and reversible changes to mitotic signaling networks.


Asunto(s)
Antineoplásicos/farmacología , Inestabilidad Cromosómica/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias/patología , Aurora Quinasa B/metabolismo , Línea Celular Tumoral , Cromosomas Humanos/metabolismo , Humanos , Cinesinas/metabolismo
11.
Mol Cancer Ther ; 15(11): 2758-2766, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27550941

RESUMEN

Despite advances in targeted therapy, lung cancer remains the most common cause of cancer-related mortality in the United States. Chromosomal instability is a prominent feature in lung cancer and, because it rarely occurs in normal cells, it represents a potential therapeutic target. Our prior work discovered that lung cancer cells undergo anaphase catastrophe in response to inhibition of cyclin-dependent kinase 2 (CDK2), followed by apoptosis and reduced growth. In this study, the effects and mechanisms of the multi-CDK inhibitor dinaciclib on lung cancer cells were investigated. We sought to determine the specificity of CDK-dependent induction of anaphase catastrophe. Live cell imaging provided direct evidence that dinaciclib caused multipolar cell divisions resulting in extensive chromosome missegregation. Genetic knockdown of dinaciclib CDK targets revealed that repression of CDK2 and CDK1, but not CDK5 or CDK9, triggered anaphase catastrophe in lung cancer cells. Overexpression of CP110, which is a mediator of CDK2 inhibitor-induced anaphase catastrophe (and a CDK1 and 2 phosphorylation substrate), antagonized anaphase catastrophe and apoptosis following dinaciclib treatment. Consistent with our previous findings, acquisition of activated KRAS sensitized lung cancer cells to dinaciclib-mediated anaphase catastrophe and cell death. Combining dinaciclib with the mitotic inhibitor taxol augmented anaphase catastrophe induction and reduced cell viability of lung cancer cells. Thus, the multi-CDK inhibitor dinaciclib causes anaphase catastrophe in lung cancer cells and should be investigated as a potential therapeutic for wild-type and KRAS-mutant lung cancer, individually or in combination with taxanes. Mol Cancer Ther; 15(11); 2758-66. ©2016 AACR.


Asunto(s)
Anafase/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Proteína Quinasa CDC2/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Compuestos de Piridinio/farmacología , Animales , Proteínas de Ciclo Celular/metabolismo , División Celular/efectos de los fármacos , División Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Óxidos N-Cíclicos , Resistencia a Antineoplásicos/genética , Humanos , Indolizinas , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Taxoides/farmacología
12.
PLoS Genet ; 12(2): e1005865, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26871722

RESUMEN

Somatic mutations of the cohesin complex subunit STAG2 are present in diverse tumor types. We and others have shown that STAG2 inactivation can lead to loss of sister chromatid cohesion and alterations in chromosome copy number in experimental systems. However, studies of naturally occurring human tumors have demonstrated little, if any, correlation between STAG2 mutational status and aneuploidy, and have further shown that STAG2-deficient tumors are often euploid. In an effort to provide insight into these discrepancies, here we analyze the effect of tumor-derived STAG2 mutations on the protein composition of cohesin and the expected mitotic phenotypes of STAG2 mutation. We find that many mutant STAG2 proteins retain their ability to interact with cohesin; however, the presence of mutant STAG2 resulted in a reduction in the ability of regulatory subunits WAPL, PDS5A, and PDS5B to interact with the core cohesin ring. Using AAV-mediated gene targeting, we then introduced nine tumor-derived mutations into the endogenous allele of STAG2 in cultured human cells. While all nonsense mutations led to defects in sister chromatid cohesion and a subset induced anaphase defects, missense mutations behaved like wild-type in these assays. Furthermore, only one of nine tumor-derived mutations tested induced overt alterations in chromosome counts. These data indicate that not all tumor-derived STAG2 mutations confer defects in cohesion, chromosome segregation, and ploidy, suggesting that there are likely to be other functional effects of STAG2 inactivation in human cancer cells that are relevant to cancer pathogenesis.


Asunto(s)
Anafase , Antígenos Nucleares/genética , Segregación Cromosómica , Mutación/genética , Neoplasias/genética , Secuencia de Bases , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Cromátides/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas Humanos/genética , Codón sin Sentido/genética , Dependovirus/metabolismo , Marcación de Gen , Células HCT116 , Humanos , Datos de Secuencia Molecular , Mutación Missense/genética , Unión Proteica , Subunidades de Proteína/metabolismo , Cohesinas
13.
Mol Cancer Ther ; 14(11): 2576-85, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26304236

RESUMEN

Chromosomal instability (CIN) is a hallmark of solid tumor biology and is implicated in carcinogenesis. Preferentially eliminating malignant cells by targeting CIN and aneuploidy is an attractive antineoplastic strategy. We previously reported that CDK2 antagonism causes lung cancer cells to undergo anaphase catastrophe and apoptosis through inhibition of phosphorylation of the centrosomal protein CP110. Cells with activating KRAS mutations were particularly sensitive to CDK2 inhibition due to downregulation of CP110 protein levels. This study investigated mechanisms of CDK2 antagonism that mediate anaphase catastrophe via changes in CP110 protein expression and how activated KRAS affects CP110 levels in lung cancers. Site-directed mutagenesis revealed candidate CDK phosphorylation sites of CP110 (residues Ser 170 and Thr 194) critical for conferring anaphase catastrophe by altering centrosome clustering in mitosis. Intriguingly, KRAS mutation can promote CP110 protein degradation by upregulating the ubiquitin ligase SCF(cyclinF), which targets CP110 protein for destabilization. Finally, CDK2 inhibitor response was enhanced when combined with knockdown of the deubiquitinase USP33 that in turn accelerates CP110 protein degradation. Thus, this study provides molecular pharmacologic insights into how CP110 expression regulates response to CDK2 inhibition. An improved understanding of in vitro antineoplastic mechanisms of combining CDK2 antagonism with induced CP110 repression provides a rationale for exploring clinical consequences of this strategy. Taken together, preclinical findings obtained from combining CDK2 inhibition with USP33 repression have implications for treating patients with non-small cell lung cancers.


Asunto(s)
Anafase/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/metabolismo , Fosfoproteínas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Ubiquitina Tiolesterasa/metabolismo , Anafase/genética , Animales , Sitios de Unión/genética , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Centrosoma/efectos de los fármacos , Centrosoma/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Ciclinas/metabolismo , Humanos , Immunoblotting , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Proteínas Asociadas a Microtúbulos/genética , Mutación , Fosfoproteínas/genética , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Purinas/farmacología , Interferencia de ARN , Roscovitina , Serina/genética , Serina/metabolismo , Treonina/genética , Treonina/metabolismo , Ubiquitina Tiolesterasa/genética
14.
Curr Biol ; 25(13): R538-42, 2015 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-26126276

RESUMEN

The terms 'haploid' and 'diploid' that describe single (n) and double (2n) chromosome sets in cells were coined by the Polish-German botanist Eduard Strasburger and originate from the Greek terms haplóos meaning 'single' and diplóos meaning 'double'. The term 'ploidy' was subsequently derived to describe the total chromosome content of cells. Consequently, the term 'euploid' refers to a chromosome complement that is an exact multiple of the haploid number. Therefore, haploids and diploids are both cases of normal euploidy. Euploid types that have more than two sets of chromosomes are 'polyploid' such as 'triploid' (3n), 'tetraploid' (4n), 'pentaploid' (5n), and so forth. There are various natural euploid states with some organisms existing as haploids (fungi), diploids (most mammals), and polyploids (plants).


Asunto(s)
Aneuploidia , Segregación Cromosómica/fisiología , Modelos Genéticos , Fenotipo , Huso Acromático/fisiología , Proliferación Celular/fisiología , Dosificación de Gen , Humanos , Cinetocoros/fisiología , Telómero/fisiología
15.
Cancer Res ; 75(10): 2029-38, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25808870

RESUMEN

Aneuploidy is frequently detected in human cancers and is implicated in carcinogenesis. Pharmacologic targeting of aneuploidy is an attractive therapeutic strategy, as this would preferentially eliminate malignant over normal cells. We previously discovered that CDK2 inhibition causes lung cancer cells with more than two centrosomes to undergo multipolar cell division leading to apoptosis, defined as anaphase catastrophe. Cells with activating KRAS mutations were especially sensitive to CDK2 inhibition. Mechanisms of CDK2-mediated anaphase catastrophe and how activated KRAS enhances this effect were investigated. Live-cell imaging provided direct evidence that following CDK2 inhibition, lung cancer cells develop multipolar anaphase and undergo multipolar cell division with the resulting progeny apoptotic. The siRNA-mediated repression of the CDK2 target and centrosome protein CP110 induced anaphase catastrophe of lung cancer cells. In contrast, CP110 overexpression antagonized CDK2 inhibitor-mediated anaphase catastrophe. Furthermore, activated KRAS mutations sensitized lung cancer cells to CDK2 inhibition by deregulating CP110 expression. Thus, CP110 is a critical mediator of CDK2 inhibition-driven anaphase catastrophe. Independent examination of murine and human paired normal-malignant lung tissues revealed marked upregulation of CP110 in malignant versus normal lung. Human lung cancers with KRAS mutations had significantly lower CP110 expression as compared with KRAS wild-type cancers. Thus, a direct link was found between CP110 and CDK2 inhibitor antineoplastic response. CP110 plays a mechanistic role in response of lung cancer cells to CDK2 inhibition, especially in the presence of activated KRAS mutations.


Asunto(s)
Anafase/efectos de los fármacos , Antineoplásicos/farmacología , Proteínas de Ciclo Celular/metabolismo , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Neoplasias Pulmonares/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Fosfoproteínas/metabolismo , Purinas/farmacología , Animales , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Quinasa 2 Dependiente de la Ciclina/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Proteínas Asociadas a Microtúbulos/genética , Mutación , Fosfoproteínas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Roscovitina , Proteínas ras/genética
16.
Front Oncol ; 3: 164, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23825799

RESUMEN

Most solid tumors are characterized by abnormal chromosome numbers (aneuploidy) and karyotypic profiling has shown that the majority of these tumors are heterogeneous and chromosomally unstable. Chromosomal instability (CIN) is defined as persistent mis-segregation of whole chromosomes and is caused by defects during mitosis. Large-scale genome sequencing has failed to reveal frequent mutations of genes encoding proteins involved in mitosis. On the contrary, sequencing has revealed that most mutated genes in cancer fall into a limited number of core oncogenic signaling pathways that regulate the cell cycle, cell growth, and apoptosis. This led to the notion that the induction of oncogenic signaling is a separate event from the loss of mitotic fidelity, but a growing body of evidence suggests that oncogenic signaling can deregulate cell cycle progression, growth, and differentiation as well as cause CIN. These new results indicate that the induction of CIN can no longer be considered separately from the cancer-associated driver mutations. Here we review the primary causes of CIN in mitosis and discuss how the oncogenic activation of key signal transduction pathways contributes to the induction of CIN.

17.
Chromosoma ; 120(1): 83-96, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20862486

RESUMEN

Centromeres are specialized chromosomal domains that direct mitotic kinetochore assembly and are defined by the presence of CENP-A (CID in Drosophila) and CENP-C. While the role of CENP-A appears to be highly conserved, functional studies in different organisms suggest that the precise role of CENP-C in kinetochore assembly is still under debate. Previous studies in vertebrate cells have shown that CENP-C inactivation causes mitotic delay, chromosome missegregation, and apoptosis; however, in Drosophila, the role of CENP-C is not well-defined. We have used RNA interference depletion in S2 cells to address this question and we find that depletion of CENP-C causes a kinetochore null phenotype, and consequently, the spindle checkpoint, kinetochore-microtubule interactions, and spindle size are severely misregulated. Importantly, we show that CENP-C is required for centromere identity as CID, MEI-S332, and chromosomal passenger proteins fail to localize in CENP-C depleted cells, suggesting a tight communication between the inner kinetochore proteins and centromeres. We suggest that CENP-C might fulfill the structural roles of the human centromere-associated proteins not identified in Drosophila.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Cromosomas de Insectos/metabolismo , Proteínas de Drosophila/metabolismo , Cinetocoros/metabolismo , Mitosis/fisiología , Animales , Línea Celular , Proteínas Cromosómicas no Histona/genética , Cromosomas de Insectos/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Humanos , Microtúbulos/genética , Microtúbulos/metabolismo , Interferencia de ARN
18.
Biochem Soc Trans ; 38(6): 1667-75, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21118145

RESUMEN

The kinetochore is a complex molecular machine that serves as the interface between sister chromatids and the mitotic spindle. The kinetochore assembles at a particular chromosomal locus, the centromere, which is essential to maintain genomic stability during cell division. The kinetochore is a macromolecular puzzle of subcomplexes assembled in a hierarchical manner and fulfils three main functions: microtubule attachment, chromosome and sister chromatid movement, and regulation of mitotic progression though the spindle assembly checkpoint. In the present paper we compare recent results on the assembly, organization and function of the kinetochore in human and Drosophila cells and conclude that, although essential functions are highly conserved, there are important differences that might help define what is a minimal chromosome segregation machinery.


Asunto(s)
Centrómero/metabolismo , Segregación Cromosómica , Drosophila melanogaster/genética , Cinetocoros/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Células HeLa , Humanos , Microtúbulos/metabolismo , Mapeo de Interacción de Proteínas , Huso Acromático/metabolismo
19.
Mol Biol Cell ; 18(3): 850-63, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17182852

RESUMEN

The spindle assembly checkpoint is essential to maintain genomic stability during cell division. We analyzed the role of the putative Drosophila Mad2 homologue in the spindle assembly checkpoint and mitotic progression. Depletion of Mad2 by RNAi from S2 cells shows that it is essential to prevent mitotic exit after spindle damage, demonstrating its conserved role. Mad2-depleted cells also show accelerated transit through prometaphase and premature sister chromatid separation, fail to form metaphases, and exit mitosis soon after nuclear envelope breakdown with extensive chromatin bridges that result in severe aneuploidy. Interestingly, preventing Mad2-depleted cells from exiting mitosis by a checkpoint-independent arrest allows congression of normally condensed chromosomes. More importantly, a transient mitotic arrest is sufficient for Mad2-depleted cells to exit mitosis with normal patterns of chromosome segregation, suggesting that all the associated phenotypes result from a highly accelerated exit from mitosis. Surprisingly, if Mad2-depleted cells are blocked transiently in mitosis and then released into a media containing a microtubule poison, they arrest with high levels of kinetochore-associated BubR1, properly localized cohesin complex and fail to exit mitosis revealing normal spindle assembly checkpoint activity. This behavior is specific for Mad2 because BubR1-depleted cells fail to arrest in mitosis under these experimental conditions. Taken together our results strongly suggest that Mad2 is exclusively required to delay progression through early stages of prometaphase so that cells have time to fully engage the spindle assembly checkpoint, allowing a controlled metaphase-anaphase transition and normal patterns of chromosome segregation.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Metafase , Huso Acromático/metabolismo , Anafase , Animales , Cinetocoros/metabolismo , Proteínas Mad2 , Fenotipo , Interferencia de ARN , Intercambio de Cromátides Hermanas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA