Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Cell Biol ; 222(12)2023 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-37955972

RESUMEN

DNA Topoisomerase IIA (Topo IIA) is an enzyme that alters the topological state of DNA and is essential for the separation of replicated sister chromatids and the integrity of cell division. Topo IIA dysfunction activates cell cycle checkpoints, resulting in arrest in either the G2-phase or metaphase of mitosis, ultimately triggering the abscission checkpoint if non-disjunction persists. These events, which directly or indirectly monitor the activity of Topo IIA, have become of major interest as many cancers have deficiencies in Topoisomerase checkpoints, leading to genome instability. Recent studies into how cells sense Topo IIA dysfunction and respond by regulating cell cycle progression demonstrate that the Topo IIA G2 checkpoint is distinct from the G2-DNA damage checkpoint. Likewise, in mitosis, the metaphase Topo IIA checkpoint is separate from the spindle assembly checkpoint. Here, we integrate mechanistic knowledge of Topo IIA checkpoints with the current understanding of how cells regulate progression through the cell cycle to accomplish faithful genome transmission and discuss the opportunities this offers for therapy.


Asunto(s)
ADN-Topoisomerasas de Tipo II , Inhibidores de Topoisomerasa II , Proteínas de Ciclo Celular/metabolismo , ADN-Topoisomerasas de Tipo II/genética , ADN-Topoisomerasas de Tipo II/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular , Mitosis , Inhibidores de Topoisomerasa II/farmacología
2.
Cancer Res ; 82(9): 1762-1773, 2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35247890

RESUMEN

Topoisomerase 2a (Topo2a)-dependent G2 arrest engenders faithful segregation of sister chromatids, yet in certain tumor cell lines where this arrest is dysfunctional, a PKCε-dependent failsafe pathway can be triggered. Here we elaborate on recent advances in understanding the underlying mechanisms associated with this G2 arrest by determining that p53-p21 signaling is essential for efficient arrest in cell lines, in patient-derived cells, and in colorectal cancer organoids. Regulation of this p53 axis required the SMC5/6 complex, which is distinct from the p53 pathways observed in the DNA damage response. Topo2a inhibition specifically during S phase did not trigger G2 arrest despite affecting completion of DNA replication. Moreover, in cancer cells reliant upon the alternative lengthening of telomeres (ALT) mechanism, a distinct form of Topo2a-dependent, p53-independent G2 arrest was found to be mediated by BLM and Chk1. Importantly, the previously described PKCε-dependent mitotic failsafe was engaged in hTERT-positive cells when Topo2a-dependent G2 arrest was dysfunctional and where p53 was absent, but not in cells dependent on the ALT mechanism. In PKCε knockout mice, p53 deletion elicited tumors were less aggressive than in PKCε-replete animals and exhibited a distinct pattern of chromosomal rearrangements. This evidence suggests the potential of exploiting synthetic lethality in arrest-defective hTERT-positive tumors through PKCε-directed therapeutic intervention. SIGNIFICANCE: The identification of a requirement for p53 in stringent Topo2a-dependent G2 arrest and engagement of PKCε failsafe pathways in arrest-defective hTERT-positive cells provides a therapeutic opportunity to induce selective synthetic lethality.


Asunto(s)
ADN-Topoisomerasas de Tipo II/metabolismo , Neoplasias , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteína p53 Supresora de Tumor , Animales , Línea Celular Tumoral , Daño del ADN , Humanos , Ratones , Neoplasias/genética , Fase S , Transducción de Señal/genética , Proteína p53 Supresora de Tumor/metabolismo
3.
Biochem J ; 478(12): 2247-2263, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-34143863

RESUMEN

A requirement for PKCε in exiting from the Aurora B dependent abscission checkpoint is associated with events at the midbody, however, the recruitment, retention and action of PKCε in this compartment are poorly understood. Here, the prerequisite for 14-3-3 complex assembly in this pathway is directly linked to the phosphorylation of Aurora B S227 at the midbody. However, while essential for PKCε control of Aurora B, 14-3-3 association is shown to be unnecessary for the activity-dependent enrichment of PKCε at the midbody. This localisation is demonstrated to be an autonomous property of the inactive PKCε D532N mutant, consistent with activity-dependent dissociation. The C1A and C1B domains are necessary for this localisation, while the C2 domain and inter-C1 domain (IC1D) are necessary for retention at the midbody. Furthermore, it is shown that while the IC1D mutant retains 14-3-3 complex proficiency, it does not support Aurora B phosphorylation, nor rescues division failure observed with knockdown of endogenous PKCε. It is concluded that the concerted action of multiple independent events facilitates PKCε phosphorylation of Aurora B at the midbody to control exit from the abscission checkpoint.


Asunto(s)
Proteínas 14-3-3/metabolismo , Aurora Quinasa B/metabolismo , Citocinesis , Proteína Quinasa C-epsilon/metabolismo , Proteínas 14-3-3/genética , Aurora Quinasa B/genética , Células HEK293 , Humanos , Fosforilación , Proteína Quinasa C-epsilon/genética , Transducción de Señal , Huso Acromático
4.
Nat Rev Cancer ; 21(1): 51-63, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33177705

RESUMEN

The maturing mutational landscape of cancer genomes, the development and application of clinical interventions and evolving insights into tumour-associated functions reveal unexpected features of the protein kinase C (PKC) family of serine/threonine protein kinases. These advances include recent work showing gain or loss-of-function mutations relating to driver or bystander roles, how conformational constraints and plasticity impact this class of proteins and how emergent cancer-associated properties may offer opportunities for intervention. The profound impact of the tumour microenvironment, reflected in the efficacy of immune checkpoint interventions, further prompts to incorporate PKC family actions and interventions in this ecosystem, informed by insights into the control of stromal and immune cell functions. Drugging PKC isoforms has offered much promise, but when and how is not obvious.


Asunto(s)
Neoplasias/enzimología , Proteína Quinasa C/fisiología , Animales , Humanos , Isoenzimas/fisiología , Mutación , Fosforilación , Regiones Promotoras Genéticas , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Microambiente Tumoral
5.
Cancer Res ; 80(22): 4946-4959, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-32998996

RESUMEN

Chromosomal instability (CIN) comprises continual gain and loss of chromosomes or parts of chromosomes and occurs in the majority of cancers, often conferring poor prognosis. Because of a scarcity of functional studies and poor understanding of how genetic or gene expression landscapes connect to specific CIN mechanisms, causes of CIN in most cancer types remain unknown. High-grade serous ovarian carcinoma (HGSC), the most common subtype of ovarian cancer, is the major cause of death due to gynecologic malignancy in the Western world, with chemotherapy resistance developing in almost all patients. HGSC exhibits high rates of chromosomal aberrations and knowledge of causative mechanisms would represent an important step toward combating this disease. Here we perform the first in-depth functional characterization of mechanisms driving CIN in HGSC in seven cell lines that accurately recapitulate HGSC genetics. Multiple mechanisms coexisted to drive CIN in HGSC, including elevated microtubule dynamics and DNA replication stress that can be partially rescued to reduce CIN by low doses of paclitaxel and nucleoside supplementation, respectively. Distinct CIN mechanisms indicated relationships with HGSC-relevant therapy including PARP inhibition and microtubule-targeting agents. Comprehensive genomic and transcriptomic profiling revealed deregulation of various genes involved in genome stability but were not directly predictive of specific CIN mechanisms, underscoring the importance of functional characterization to identify causes of CIN. Overall, we show that HGSC CIN is complex and suggest that specific CIN mechanisms could be used as functional biomarkers to indicate appropriate therapy. SIGNIFICANCE: These findings characterize multiple deregulated mechanisms of genome stability that lead to CIN in ovarian cancer and demonstrate the benefit of integrating analysis of said mechanisms into predictions of therapy response.


Asunto(s)
Inestabilidad Cromosómica , Cistadenocarcinoma Seroso/genética , Neoplasias Ováricas/genética , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Inestabilidad Cromosómica/fisiología , Segregación Cromosómica , Cistadenocarcinoma Seroso/tratamiento farmacológico , Cistadenocarcinoma Seroso/patología , Variaciones en el Número de Copia de ADN , Daño del ADN , Replicación del ADN/fisiología , Femenino , Inestabilidad Genómica , Humanos , Microtúbulos/fisiología , Clasificación del Tumor , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Ftalazinas/uso terapéutico , Piperazinas/uso terapéutico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico
6.
Adv Biol Regul ; 78: 100759, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33039823

RESUMEN

Associated with their roles as targets for tumour promoters, there has been a long-standing interest in how members of the protein kinase C (PKC) family act to modulate cell growth and division. This has generated a great deal of observational data, but has for the most part not afforded clear mechanistic insights into the control mechanisms at play. Here, we review the roles of PKCε in protecting transformed cells from non-disjunction. In this particular cell cycle context, there is a growing understanding of the pathways involved, affording biomarker and interventional insights and opportunities.


Asunto(s)
Genómica , Neoplasias/genética , Proteína Quinasa C-epsilon/metabolismo , Proliferación Celular , Humanos , Neoplasias/enzimología , Neoplasias/patología
8.
Nat Commun ; 11(1): 1396, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-32170202

RESUMEN

The Aurora B abscission checkpoint delays cytokinesis until resolution of DNA trapped in the cleavage furrow. This process involves PKCε phosphorylation of Aurora B S227. Assessing if this PKCε-Aurora B module provides a more widely exploited genome-protective control for the cell cycle, we show Aurora B phosphorylation at S227 by PKCε also occurs during mitosis. Expression of Aurora B S227A phenocopies inhibition of PKCε in by-passing the delay and resolution at anaphase entry that is associated with non-disjunction and catenation of sister chromatids. Implementation of this anaphase delay is reflected in PKCε activation following cell cycle dependent cleavage by caspase 7; knock-down of caspase 7 phenocopies PKCε loss, in a manner rescued by ectopically expressing/generating a free PKCε catalytic domain. Molecular dynamics indicates that Aurora B S227 phosphorylation induces conformational changes and this manifests in a profound switch in specificity towards S29 TopoIIα phosphorylation, a response necessary for catenation resolution during mitosis.


Asunto(s)
Anafase/fisiología , Aurora Quinasa B/metabolismo , Metafase/fisiología , Mitosis/fisiología , Anafase/genética , Aurora Quinasa B/química , Aurora Quinasa B/genética , Caspasa 7/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cromátides/metabolismo , Cromatina , Cromosomas/metabolismo , Células HEK293 , Humanos , Metafase/genética , Mitosis/genética , Simulación de Dinámica Molecular , Fosforilación , Proteína Quinasa C-epsilon/metabolismo , Proteínas Recombinantes , Transducción de Señal/fisiología
9.
Nucleic Acids Res ; 47(6): 2906-2921, 2019 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-30590722

RESUMEN

The Topo2a-dependent arrest is associated with faithful segregation of sister chromatids and has been identified as dysfunctional in numerous tumour cell lines. This genome-protecting pathway is poorly understood and its characterization is of significant interest, potentially offering interventional opportunities in relation to synthetic lethal behaviours in arrest-defective tumours. Using the catalytic Topo2a inhibitor ICRF193, we have performed a genome-wide siRNA screen in arrest-competent, non-transformed cells, to identify genes essential for this arrest mechanism. In addition, we have counter-screened several DNA-damaging agents and demonstrate that the Topo2a-dependent arrest is genetically distinct from DNA damage checkpoints. We identify the components of the SMC5/6 complex, including the activity of the E3 SUMO ligase NSE2, as non-redundant players that control the timing of the Topo2a-dependent arrest in G2. We have independently verified the NSE2 requirement in fibroblasts from patients with germline mutations that cause severely reduced levels of NSE2. Through imaging Topo2a-dependent G2 arrested cells, an increased interaction between Topo2a and NSE2 is observed at PML bodies, which are known SUMOylation hotspots. We demonstrate that Topo2a is SUMOylated in an ICRF193-dependent manner by NSE2 at a novel non-canonical site (K1520) and that K1520 sumoylation is required for chromosome segregation but not the G2 arrest.


Asunto(s)
ADN-Topoisomerasas de Tipo II/genética , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Ligasas/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Sumoilación/genética , Proteínas de Ciclo Celular/genética , Línea Celular , Proteínas Cromosómicas no Histona/genética , Daño del ADN/efectos de los fármacos , Dicetopiperazinas , Fibroblastos/efectos de los fármacos , Genoma Humano/genética , Mutación de Línea Germinal/genética , Humanos , Complejos Multiproteicos/genética , Piperazinas/farmacología , Proteínas de Unión a Poli-ADP-Ribosa/antagonistas & inhibidores , Interferencia de ARN , Ubiquitina-Proteína Ligasas/genética
10.
Elife ; 72018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29712619

RESUMEN

While targeted therapy against HER2 is an effective first-line treatment in HER2+ breast cancer, acquired resistance remains a clinical challenge. The pseudokinase HER3, heterodimerisation partner of HER2, is widely implicated in the resistance to HER2-mediated therapy. Here, we show that lapatinib, an ATP-competitive inhibitor of HER2, is able to induce proliferation cooperatively with the HER3 ligand neuregulin. This counterintuitive synergy between inhibitor and growth factor depends on their ability to promote atypical HER2-HER3 heterodimerisation. By stabilising a particular HER2 conformer, lapatinib drives HER2-HER3 kinase domain heterocomplex formation. This dimer exists in a head-to-head orientation distinct from the canonical asymmetric active dimer. The associated clustering observed for these dimers predisposes to neuregulin responses, affording a proliferative outcome. Our findings provide mechanistic insights into the liabilities involved in targeting kinases with ATP-competitive inhibitors and highlight the complex role of protein conformation in acquired resistance.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proliferación Celular , Lapatinib/farmacología , Neurregulina-1/metabolismo , Multimerización de Proteína , Receptor ErbB-2/química , Receptor ErbB-3/química , Adenosina Trifosfato/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Femenino , Humanos , Fosforilación , Conformación Proteica , Inhibidores de Proteínas Quinasas/farmacología , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Transducción de Señal , Células Tumorales Cultivadas
11.
Mol Cancer Res ; 16(1): 3-15, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29021232

RESUMEN

To form a proper mitotic spindle, centrosomes must be duplicated and driven poleward in a timely and controlled fashion. Improper timing of centrosome separation and errors in mitotic spindle assembly may lead to chromosome instability, a hallmark of cancer. Protein kinase C epsilon (PKCε) has recently emerged as a regulator of several cell-cycle processes associated with the resolution of mitotic catenation during the metaphase-anaphase transition and in regulating the abscission checkpoint. However, an engagement of PKCε in earlier (pre)mitotic events has not been addressed. Here, we now establish that PKCε controls prophase-to-metaphase progression by coordinating centrosome migration and mitotic spindle assembly in transformed cells. This control is exerted through cytoplasmic dynein function. Importantly, it is also demonstrated that the PKCε dependency of mitotic spindle organization is correlated with the nonfunctionality of the TOPO2A-dependent G2 checkpoint, a characteristic of many transformed cells. Thus, PKCε appears to become specifically engaged in a programme of controls that are required to support cell-cycle progression in transformed cells, advocating for PKCε as a potential cancer therapeutic target.Implications: The close relationship between PKCε dependency for mitotic spindle organization and the nonfunctionality of the TOPO2A-dependent G2 checkpoint, a hallmark of transformed cells, strongly suggests PKCε as a therapeutic target in cancer. Mol Cancer Res; 16(1); 3-15. ©2017 AACR.


Asunto(s)
Centrosoma/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Huso Acromático/metabolismo , Humanos , Puntos de Control de la Fase M del Ciclo Celular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...