Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cancer Res ; 22(5): 423-439, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38324016

RESUMEN

NDC80 complex (NDC80C) is composed of four subunits (SPC24, SPC25, NDC80, and NUF2) and is vital for kinetochore-microtubule (KT-MT) attachment during mitosis. Paradoxically, NDC80C also functions in the activation of the spindle-assembly checkpoint (SAC). This raises an interesting question regarding how mitosis is regulated when NDC80C levels are compromised. Using a degron-mediated depletion system, we found that acute silencing of SPC24 triggered a transient mitotic arrest followed by mitotic slippage. SPC24-deficient cells were unable to sustain SAC activation despite the loss of KT-MT interaction. Intriguingly, our results revealed that other subunits of the NDC80C were co-downregulated with SPC24 at a posttranslational level. Silencing any individual subunit of NDC80C likewise reduced the expression of the entire complex. We found that the SPC24-SPC25 and NDC80-NUF2 subcomplexes could be individually stabilized using ectopically expressed subunits. The synergism of SPC24 downregulation with drugs that promote either mitotic arrest or mitotic slippage further underscored the dual roles of NDC80C in KT-MT interaction and SAC maintenance. The tight coordinated regulation of NDC80C subunits suggests that targeting individual subunits could disrupt mitotic progression and provide new avenues for therapeutic intervention. IMPLICATIONS: These results highlight the tight coordinated regulation of NDC80C subunits and their potential as targets for antimitotic therapies.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas del Citoesqueleto , Mitosis , Proteínas Nucleares , Humanos , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Células HeLa , Proteínas del Citoesqueleto/metabolismo , Proteínas del Citoesqueleto/genética , Cinetocoros/metabolismo , Puntos de Control de la Fase M del Ciclo Celular/genética , Huso Acromático/metabolismo , Subunidades de Proteína/metabolismo , Subunidades de Proteína/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/genética
2.
Cell Death Dis ; 15(1): 2, 2024 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-38172496

RESUMEN

Mitotic catastrophe induced by prolonged mitotic arrest is a major anticancer strategy. Although antiapoptotic BCL2-like proteins, including BCL-XL, are known to regulate apoptosis during mitotic arrest, adaptive changes in their expression can complicate loss-of-function studies. Our studies revealed compensatory alterations in the expression of BCL2 and MCL1 when BCL-XL is either downregulated or overexpressed. To circumvent their reciprocal regulation, we utilized a degron-mediated system to acutely silence BCL-XL just before mitosis. Our results show that in epithelial cell lines including HeLa and RPE1, BCL-XL and BCL2 acted collaboratively to suppress apoptosis during both unperturbed cell cycle and mitotic arrest. By tagging BCL-XL and BCL2 with a common epitope, we estimated that BCL-XL was less abundant than BCL2 in the cell. Nonetheless, BCL-XL played a more prominent antiapoptotic function than BCL2 during interphase and mitotic arrest. Loss of BCL-XL led to mitotic cell death primarily through a BAX-dependent process. Furthermore, silencing of BCL-XL led to the stabilization of MCL1, which played a significant role in buffering apoptosis during mitotic arrest. Nevertheless, even in a MCL1-deficient background, depletion of BCL-XL accelerated mitotic apoptosis. These findings underscore the pivotal involvement of BCL-XL in controlling timely apoptosis during mitotic arrest, despite adaptive changes in the expression of other BCL2-like proteins.


Asunto(s)
Apoptosis , Proteínas Proto-Oncogénicas c-bcl-2 , Humanos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Proteína bcl-X/genética , Proteína bcl-X/metabolismo , Línea Celular Tumoral , Apoptosis/genética
3.
J Cell Sci ; 136(8)2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36995025

RESUMEN

Switching genes on and off on cue is a cornerstone for understanding gene functions. One contemporary approach for loss-of-function studies of essential genes involves CRISPR-mediated knockout of the endogenous locus in conjunction with the expression of a rescue construct, which can subsequently be turned off to produce a gene inactivation effect in mammalian cell lines. A broadening of this approach would involve simultaneously switching on a second construct to interrogate the functions of a gene in the pathway. In this study, we developed a pair of switches that were independently controlled by both inducible promoters and degrons, enabling the toggling between two constructs with comparable kinetics and tightness. The gene-OFF switch was based on TRE transcriptional control coupled with auxin-induced degron-mediated proteolysis. A second independently controlled gene-ON switch was based on a modified ecdysone promoter and mutated FKBP12-derived destabilization domain degron, allowing acute and tuneable gene activation. This platform facilitates efficient generation of knockout cell lines containing a two-gene switch that is regulated tightly and can be flipped within a fraction of the time of a cell cycle.


Asunto(s)
Regulación de la Expresión Génica , Ácidos Indolacéticos , Animales , Línea Celular , Ácidos Indolacéticos/farmacología , Proteolisis , Regiones Promotoras Genéticas/genética , Mamíferos/metabolismo
4.
Int J Mol Sci ; 24(4)2023 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-36835147

RESUMEN

Whole-genome duplication (WGD) is one of the most common genomic abnormalities in cancers. WGD can provide a source of redundant genes to buffer the deleterious effect of somatic alterations and facilitate clonal evolution in cancer cells. The extra DNA and centrosome burden after WGD is associated with an elevation of genome instability. Causes of genome instability are multifaceted and occur throughout the cell cycle. Among these are DNA damage caused by the abortive mitosis that initially triggers tetraploidization, replication stress and DNA damage associated with an enlarged genome, and chromosomal instability during the subsequent mitosis in the presence of extra centrosomes and altered spindle morphology. Here, we chronicle the events after WGD, from tetraploidization instigated by abortive mitosis including mitotic slippage and cytokinesis failure to the replication of the tetraploid genome, and finally, to the mitosis in the presence of supernumerary centrosomes. A recurring theme is the ability of some cancer cells to overcome the obstacles in place for preventing WGD. The underlying mechanisms range from the attenuation of the p53-dependent G1 checkpoint to enabling pseudobipolar spindle formation via the clustering of supernumerary centrosomes. These survival tactics and the resulting genome instability confer a subset of polyploid cancer cells proliferative advantage over their diploid counterparts and the development of therapeutic resistance.


Asunto(s)
Carcinogénesis , Duplicación de Gen , Inestabilidad Genómica , Neoplasias , Humanos , Ciclo Celular , Centrosoma/metabolismo , Inestabilidad Cromosómica , Mitosis , Poliploidía , Huso Acromático , Carcinogénesis/genética , Neoplasias/genética
5.
J Biol Chem ; 299(3): 102957, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36717077

RESUMEN

Cyclin A and CDC25A are both activators of cyclin-dependent kinases (CDKs): cyclin A acts as an activating subunit of CDKs and CDC25A a phosphatase of the inhibitory phosphorylation sites of the CDKs. In this study, we uncovered an inverse relationship between the two CDK activators. As cyclin A is an essential gene, we generated a conditional silencing cell line using a combination of CRISPR-Cas9 and degron-tagged cyclin A. Destruction of cyclin A promoted an acute accumulation of CDC25A. The increase of CDC25A after cyclin A depletion occurred throughout the cell cycle and was independent on cell cycle delay caused by cyclin A deficiency. Moreover, we determined that the inverse relationship with cyclin A was specific for CDC25A and not for other CDC25 family members or kinases that regulate the same sites in CDKs. Unexpectedly, the upregulation of CDC25A was mainly caused by an increase in transcriptional activity instead of a change in the stability of the protein. Reversing the accumulation of CDC25A severely delayed G2-M in cyclin A-depleted cells. Taken together, these data provide evidence of a compensatory mechanism involving CDC25A that ensures timely mitotic entry at different levels of cyclin A.


Asunto(s)
Ciclina A , Quinasas Ciclina-Dependientes , Fosfatasas cdc25 , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo , Ciclo Celular , División Celular , Ciclina A/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Fosforilación
6.
Cell Death Differ ; 30(3): 753-765, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36329234

RESUMEN

The anti-apoptotic MCL1 is critical for delaying apoptosis during mitotic arrest. MCL1 is degraded progressively during mitotic arrest, removing its anti-apoptotic function. We found that knockout of components of ubiquitin ligases including APC/C, SCF complexes, and the mitochondrial ubiquitin ligase MARCH5 did not prevent mitotic degradation of MCL1. Nevertheless, MARCH5 determined the initial level of MCL1-NOXA network upon mitotic entry and hence the window of time during MCL1 was present during mitotic arrest. Paradoxically, although knockout of MARCH5 elevated mitotic MCL1, mitotic apoptosis was in fact enhanced in a BAK-dependent manner. Mitotic apoptosis was accelerated after MARCH5 was ablated in both the presence and absence of MCL1. Cell death was not altered after disrupting other MARCH5-regulated BCL2 family members including NOXA, BIM, and BID. Disruption of the mitochondrial fission factor DRP1, however, reduced mitotic apoptosis in MARCH5-disrupted cells. These data suggest that MARCH5 regulates mitotic apoptosis through MCL1-independent mechanisms including mitochondrial maintenance that can overcome the stabilization of MCL1.


Asunto(s)
Proteínas de la Membrana , Ubiquitina-Proteína Ligasas , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de la Membrana/metabolismo , Apoptosis , Ubiquitinas
7.
Methods Mol Biol ; 2329: 1-18, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34085211

RESUMEN

The cell cycle is the sequence of events through which a cell duplicates its genome, grows, and divides. Key cell cycle transitions are driven by oscillators comprising of protein kinases and their regulators. Different cell cycle oscillators are inextricably linked to ensure orderly activation of oscillators. A recurring theme in their regulation is the abundance of autoamplifying loops that ensure switch-like and unidirectional cell cycle transitions. The periodicity of many cell cycle oscillators is choreographed by inherent mechanisms that promote automatic inactivation, often involving dephosphorylation and ubiquitin-mediated protein degradation. These inhibitory signals are subsequently suppressed to enable the next cell cycle to occur. Although the activation and inactivation of cell cycle oscillators are in essence autonomous during the unperturbed cell cycle, a number of checkpoint mechanisms are able to halt the cell cycle until preconditions or defects are addressed. Together, these mechanisms orchestrate orderly progression of the cell cycle to produce more cells and to safeguard genome stability.


Asunto(s)
Ciclo Celular , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Inestabilidad Genómica , Animales , ADN/metabolismo , Regulación de la Expresión Génica , Humanos , Fosforilación , Proteolisis , Transducción de Señal
8.
Mol Biol Cell ; 32(14): 1320-1330, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33979199

RESUMEN

Loss-of-function analysis is one of the major arsenals we have for understanding gene functions in mammalian cells. For analysis of essential genes, the major challenge is to develop simple methodologies for tight and rapid inducible gene inactivation. One approach involves CRISPR-Cas9-mediated disruption of the endogenous locus in conjunction with the expression of a rescue construct, which can subsequently be turned off to produce a gene inactivation effect. Here we describe the development of a set of Sleeping Beauty transposon-based vectors for expressing auxin-inducible degron (AID)-tagged genes under the regulation of a tetracycline-controlled promoter. The dual transcriptional and degron-mediated post-translational regulation allows rapid and tight silencing of protein expression in mammalian cells. We demonstrated that both non-essential and essential genes could be targeted in human cell lines using a one-step transfection method. Moreover, multiple genes could be simultaneously or sequentially targeted, allowing inducible inactivation of multiple genes. These resources enable highly efficient generation of conditional gene silencing cell lines to facilitate functional studies of essential genes.


Asunto(s)
Regulación de la Expresión Génica/genética , Silenciador del Gen/fisiología , Ingeniería Genética/métodos , Animales , Sistemas CRISPR-Cas , Línea Celular , Expresión Génica/genética , Regulación de la Expresión Génica/fisiología , Genes Esenciales/genética , Genes Reporteros/genética , Vectores Genéticos/genética , Humanos , Ácidos Indolacéticos/metabolismo , Mutación con Pérdida de Función/genética , Regiones Promotoras Genéticas/genética , Transfección , Transposasas/genética , Transposasas/metabolismo
9.
Mutat Res ; 821: 111716, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32738522

RESUMEN

It is well established that Aurora kinases perform critical functions during mitosis. It has become increasingly clear that the Aurora kinases also perform a myriad of non-mitotic functions including DNA damage response. The available evidence indicates that inhibition Aurora kinase A (AURKA) may contribute to the G2 DNA damage checkpoint through AURKA's functions in PLK1 and CDC25B activation. Both AURKA and Aurora kinase B (AURKB) are also essential in mitotic DNA damage response that guard against DNA damage-induced chromosome segregation errors, including the control of abscission checkpoint and prevention of micronuclei formation. Dysregulation of Aurora kinases can trigger DNA damage in mitosis that is sensed in the subsequent G1 by a p53-dependent postmitotic checkpoint. Aurora kinases are themselves linked to the G1 DNA damage checkpoint through p53 and p73 pathways. Finally, several lines of evidence provide a connection between Aurora kinases and DNA repair and apoptotic pathways. Although more studies are required to provide a comprehensive picture of how cells respond to DNA damage, these findings indicate that both AURKA and AURKB are inextricably linked to pathways guarding against DNA damage. They also provide a rationale to support more detailed studies on the synergism between small-molecule inhibitors against Aurora kinases and DNA-damaging agents in cancer therapies.


Asunto(s)
Antineoplásicos/uso terapéutico , Aurora Quinasas/antagonistas & inhibidores , Daño del ADN , Reparación del ADN , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Animales , Humanos , Neoplasias/genética , Neoplasias/patología
10.
Oncogene ; 39(13): 2819-2834, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32029899

RESUMEN

Mitotic slippage involves cells exiting mitosis without proper chromosome segregation. Although degradation of cyclin B1 during prolonged mitotic arrest is believed to trigger mitotic slippage, its upstream regulation remains obscure. Whether mitotic slippage is caused by APC/CCDC20 activity that is able to escape spindle-assembly checkpoint (SAC)-mediated inhibition, or is actively promoted by a change in SAC activity remains an outstanding issue. We found that a major culprit for mitotic slippage involves reduction of MAD2 at the kinetochores, resulting in a progressive weakening of SAC during mitotic arrest. A further level of control of the timing of mitotic slippage is through p31comet-mediated suppression of MAD2 activation. The loss of kinetochore MAD2 was dependent on APC/CCDC20, indicating a feedback control of APC/C to SAC during prolonged mitotic arrest. The gradual weakening of SAC during mitotic arrest enables APC/CCDC20 to degrade cyclin B1, cumulating in the cell exiting mitosis by mitotic slippage.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Puntos de Control del Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Mad2/metabolismo , Mitosis/genética , Proteínas Nucleares/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Proteínas Cdc20/genética , Proteínas Cdc20/metabolismo , Proteínas de Ciclo Celular/genética , Ciclina B1/metabolismo , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Cinetocoros/metabolismo , Proteínas Nucleares/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , ARN Interferente Pequeño/metabolismo , Huso Acromático/metabolismo
11.
Mol Cancer Ther ; 19(1): 123-134, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31597711

RESUMEN

PARP inhibitors have emerged as effective chemotherapeutic agents for BRCA1/BRCA2-deficient cancers. Another DNA damage response protein, ATM, is also increasingly being recognized as a target for synthetic lethality with PARP inhibitors. As ATM functions in both cell cycle arrest and DNA repair after DNA damage, how cells respond to inhibition of ATM and PARP1 is yet to be defined precisely. We found that loss of ATM function, either in an ATM-deficient background or after treatment with ATM inhibitors (KU-60019 or AZD0156), results in spontaneous DNA damage and an increase in PARylation. When PARP1 is also deleted or inhibited with inhibitors (olaparib or veliparib), the massive increase in DNA damage activates the G2 DNA damage checkpoint kinase cascade involving ATR, CHK1/2, and WEE1. Our data indicated that the role of ATM in DNA repair is critical for the synergism with PARP inhibitors. Bypass of the G2 DNA damage checkpoint in the absence of ATM functions occurs only after a delay. The relative insensitivity of PARP1-deficient cells to PARP inhibitors suggested that other PARP isoforms played a relatively minor role in comparison with PARP1 in synergism with ATMi. As deletion of PARP1 also increased sensitivity to ATM inhibitors, trapping of PARP1 on DNA may not be the only mechanism involved in the synergism between PARP1 and ATM inhibition. Collectively, these studies provide a mechanistic foundation for therapies targeting ATM and PARP1.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Daño del ADN/genética , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Sinergismo Farmacológico , Humanos
12.
Cell Death Dis ; 10(4): 314, 2019 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-30952840

RESUMEN

Disrupting microtubule dynamics with spindle poisons activates the spindle-assembly checkpoint (SAC) and induces mitotic cell death. However, mitotic exit can occur prematurely without proper chromosomal segregation or cytokinesis by a process termed mitotic slippage. It remains controversial whether mitotic slippage increases the cytotoxicity of spindle poisons or the converse. Altering the SAC induces either mitotic cell death or mitotic slippage. While knockout of MAD2-binding protein p31comet strengthened the SAC and promoted mitotic cell death, knockout of TRIP13 had the opposite effect of triggering mitotic slippage. We demonstrated that mitotic slippage prevented mitotic cell death caused by spindle poisons, but reduced subsequent long-term survival. Weakening of the SAC also reduced cell survival in response to spindle perturbation insufficient for triggering mitotic slippage, of which mitotic exit was characterized by displaced chromosomes during metaphase. In either mitotic slippage or mitotic exit with missegregated chromosomes, cell death occurred only after one cell cycle following mitotic exit and increased progressively during subsequent cell cycles. Consistent with these results, transient inhibition of the SAC using an MPS1 inhibitor acted synergistically with spindle perturbation in inducing chromosome missegregation and cytotoxicity. The specific temporal patterns of cell death after mitotic exit with weakened SAC may reconcile the contradictory results from many previous studies.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Muerte Celular , Segregación Cromosómica , Puntos de Control de la Fase M del Ciclo Celular , Mitosis , Huso Acromático/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/genética , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Muerte Celular/genética , Segregación Cromosómica/efectos de los fármacos , Células HCT116 , Células HeLa , Humanos , Cinética , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase M del Ciclo Celular/genética , Puntos de Control de la Fase M del Ciclo Celular/fisiología , Micronúcleos con Defecto Cromosómico/efectos de los fármacos , Mitosis/efectos de los fármacos , Mitosis/genética , Mitosis/fisiología , Venenos/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Huso Acromático/genética
13.
Cell Cycle ; 18(2): 238-248, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30582405

RESUMEN

Characterizing the functions of essential cell cycle control genes requires tight and rapid inducible gene inactivation. Drawbacks of current conditional depletion approaches include slow responses and incomplete depletion. We demonstrated that by integrating the tetracycline-controlled promoter system and the auxin-inducible degron (AID) system together, AID-tagged proteins can be downregulated more efficiently than the individual technology alone. When used in conjunction with CRISPR-Cas9-mediated disruption of the endogenous locus, this system facilitates the analysis of essential genes by allowing rapid and tight conditional depletion, as we have demonstrated using several cell cycle-regulatory genes including cyclin A, CDK2, and TRIP13. The vectors constructed in this study allow expression of AID-fusion proteins under the control of tetracycline-controlled promoters and should be useful in studies requiring rapid and tight suppression of gene expression in mammalian cells.


Asunto(s)
Sistemas CRISPR-Cas/genética , Ácidos Indolacéticos/metabolismo , Proteolisis , Tetraciclinas/metabolismo , Activación Transcripcional/genética , ATPasas Asociadas con Actividades Celulares Diversas/genética , Proteínas de Ciclo Celular/genética , Células Clonales , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Ciclina A/genética , Quinasa 2 Dependiente de la Ciclina/genética , Expresión Génica , Técnicas de Inactivación de Genes , Células HeLa , Humanos , Elementos de Respuesta/genética , Retroviridae/genética , Transfección
14.
Cell Rep ; 22(6): 1439-1450, 2018 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-29425500

RESUMEN

The spindle assembly checkpoint (SAC) prevents premature segregation of chromosomes during mitosis. This process requires structural remodeling of MAD2 from O-MAD2 to C-MAD2 conformation. After the checkpoint is satisfied, C-MAD2 is reverted to O-MAD2 to allow anaphase-promoting complex/cyclosome (APC/C) to trigger anaphase. Recently, the AAA+-ATPase TRIP13 was shown to act in concert with p31comet to catalyze C- to O-MAD2. Paradoxically, although C-MAD2 is present in TRIP13-deficient cells, the SAC cannot be activated. Using a degron-mediated system to uncouple TRIP13 from O- and C-MAD2 equilibrium, we demonstrated that the loss of TRIP13 did not immediately abolish the SAC, but the resulting C-MAD2-only environment was insufficient to enable the SAC. These results favor a model in which MAD2-CDC20 interaction is coupled directly to the conversion of O- to C-MAD2 instead of one that involves unliganded C-MAD2. TRIP13 replenishes the O-MAD2 pool for activation by unattached kinetochores.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Segregación Cromosómica/fisiología , Puntos de Control de la Fase M del Ciclo Celular/fisiología , Proteínas Mad2/metabolismo , Proteínas Cdc20/metabolismo , Células HeLa , Humanos , Cinetocoros/metabolismo
16.
Neuron ; 96(5): 1041-1054.e5, 2017 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-29103808

RESUMEN

Mutations of DISC1 (disrupted-in-schizophrenia 1) have been associated with major psychiatric disorders. Despite the hundreds of DISC1-binding proteins reported, almost nothing is known about how DISC1 interacts with other proteins structurally to impact human brain development. Here we solved the high-resolution structure of DISC1 C-terminal tail in complex with its binding domain of Ndel1. Mechanistically, DISC1 regulates Ndel1's kinetochore attachment, but not its centrosome localization, during mitosis. Functionally, disrupting DISC1/Ndel1 complex formation prolongs mitotic length and interferes with cell-cycle progression in human cells, and it causes cell-cycle deficits of radial glial cells in the embryonic mouse cortex and human forebrain organoids. We also observed similar deficits in organoids derived from schizophrenia patient induced pluripotent stem cells (iPSCs) with a DISC1 mutation that disrupts its interaction with Ndel1. Our study uncovers a new mechanism of action for DISC1 based on its structure, and it has implications for how genetic insults may contribute to psychiatric disorders.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Mitosis/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Neurogénesis/genética , Neuronas/fisiología , Animales , Proteínas Portadoras/química , Ciclo Celular , Femenino , Células HeLa , Humanos , Inmunohistoquímica , Masculino , Ratones , Modelos Moleculares , Proteínas del Tejido Nervioso/química , Células-Madre Neurales , Neuronas/patología , Células Madre Pluripotentes , Embarazo , Unión Proteica , Esquizofrenia/patología
17.
Oncotarget ; 8(24): 38811-38824, 2017 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-28415588

RESUMEN

Understanding cancer stem cell (CSC) maintenance pathways is critical for the development of CSC-targeting therapy. Here, we investigated the functional role of the cyclin D1-dependent activation of Smad2/3 and Smad4 in hepatocellular carcinoma (HCC) CSCs and in HCC primary tumors. Cyclin D1 sphere-derived xenograft tumor models were employed to evaluate the therapeutic effects of a Smad inhibitor in combination with chemotherapy. Cyclin D1 overexpression confers stemness properties by enhancing single sphere formation, enhancing the CD90+ and EpCAM+ population, increasing stemness gene expression, and increasing chemoresistance. Cyclin D1 interacts with and activates Smad2/3 and Smad4 to result in cyclin D1-Smad2/3-Smad4 signaling-regulated liver CSC self-renewal. The cyclin D1-dependent activation of Smad2/3 and Smad4 is also found in HCC patients and predicts disease progression. A Smad inhibitor impaired cyclin D1-Smad-mediated self-renewal, resulting in the chemosensitization. Thus, pretreatment with a Smad inhibitor followed by chemotherapy not only successfully suppressed tumor growth but also eliminated 57% of the tumors in a cyclin D1 sphere-derived xenograft model. Together, The cyclin D1-mediated activation of Smad2/3 and Smad4 is an important regulatory mechanism in liver CSC self-renewal and stemness. Accordingly, a Smad inhibitor induced CSC differentiation and consequently significant chemosensitization, which could be an effective strategy to target CSCs.


Asunto(s)
Carcinoma Hepatocelular/patología , Ciclina D1/metabolismo , Resistencia a Antineoplásicos , Células Madre Neoplásicas/patología , Proteína Smad2/metabolismo , Proteína Smad4/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones SCID , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Pronóstico , Transducción de Señal/efectos de los fármacos , Proteína Smad2/antagonistas & inhibidores , Proteína Smad4/antagonistas & inhibidores , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Methods Mol Biol ; 1524: 189-201, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27815904

RESUMEN

HeLa is one of the oldest and most commonly used cell lines in biomedical research. Owing to the ease of which they can be effectively synchronized by various methods, HeLa cells have been used extensively for studying the cell cycle. Here, we describe several protocols for synchronizing HeLa cells from different phases of the cell cycle, including G1 phase using the HMG-CoA reductase inhibitor lovastatin, S phase with a double thymidine block procedure, and G2 phase with the CDK1 inhibitor RO-3306. Cells can also be enriched in mitosis using nocodazole and mechanical shake-off. Releasing the cells from these blocks enables researchers to follow gene expression and other events through the cell cycle. We also describe several protocols, including flow cytometry, BrdU labeling, immunoblotting, and time-lapse microscopy, for validating the synchrony of the cells and monitoring the progression of the cell cycle.


Asunto(s)
Ciclo Celular/genética , Ciclo Celular/efectos de los fármacos , Ciclinas/metabolismo , Citometría de Flujo , Fase G1/efectos de los fármacos , Fase G1/genética , Fase G2/efectos de los fármacos , Fase G2/genética , Células HeLa , Humanos , Immunoblotting , Lovastatina/farmacología , Nocodazol/farmacología , Quinolinas/farmacología , Fase S/efectos de los fármacos , Fase S/genética , Tiazoles/farmacología
19.
Oncotarget ; 7(25): 38718-38730, 2016 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-27231850

RESUMEN

Microtubule inhibitors including taxanes and vinca alkaloids are among the most widely used anticancer agents. Disrupting the microtubules activates the spindle-assembly checkpoint and traps cells in mitosis. Whether cells subsequently undergo mitotic cell death is an important factor for the effectiveness of the anticancer agents. Given that apoptosis accounts for the majority of mitotic cell death induced by microtubule inhibitors, we performed a systematic study to determine which members of the anti-apoptotic BCL-2 family are involved in determining the duration of mitotic block before cell death or slippage. Depletion of several anti-apoptotic BCL-2-like proteins significantly shortened the time before apoptosis. Among these proteins, BCL-W has not been previously characterized to play a role in mitotic cell death. Although the expression of BCL-W remained constant during mitotic block, it varied significantly between different cell lines. Knockdown of BCL-W with siRNA or disruption of the BCL-W gene with CRISPR-Cas9 speeded up mitotic cell death. Conversely, overexpression of BCL-W delayed mitotic cell death, extending the mitotic block to allow mitotic slippage. Taken together, these results showed that BCL-W contributes to the threshold of anti-apoptotic activity during mitosis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Microtúbulos/metabolismo , Mitosis , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Moduladores de Tubulina/farmacología , Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Muerte Celular/efectos de los fármacos , Linaje de la Célula , Relación Dosis-Respuesta a Droga , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Histonas/metabolismo , Humanos , Paclitaxel/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
20.
Sci Rep ; 6: 22230, 2016 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-26923777

RESUMEN

The G2 DNA damage checkpoint is one of the most important mechanisms controlling G2-mitosis transition. The kinase Greatwall (MASTL in human) promotes normal G2-mitosis transition by inhibiting PP2A via ARPP19 and ENSA. In this study, we demonstrate that MASTL is critical for maintaining genome integrity after DNA damage. Although MASTL did not affect the activation of DNA damage responses and subsequent repair, it determined the timing of entry into mitosis and the subsequent fate of the recovering cells. Constitutively active MASTL promoted dephosphorylation of CDK1(Tyr15) and accelerated mitotic entry after DNA damage. Conversely, downregulation of MASTL or ARPP19/ENSA delayed mitotic entry. Remarkably, APC/C was activated precociously, resulting in the damaged cells progressing from G2 directly to G1 and skipping mitosis all together. Collectively, these results established that precise control of MASTL is essential to couple DNA damage to mitosis through the rate of mitotic entry and APC/C activation.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Puntos de Control del Ciclo Celular , Daño del ADN , Proteínas Asociadas a Microtúbulos/metabolismo , Mitosis , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Quinasa CDC2/metabolismo , Proteínas Cdh1/metabolismo , Línea Celular , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...