Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Nature ; 605(7909): 357-365, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35508654

RESUMEN

The entry of mammalian cells into the DNA synthesis phase (S phase) represents a key event in cell division1. According to current models of the cell cycle, the kinase CDC7 constitutes an essential and rate-limiting trigger of DNA replication, acting together with the cyclin-dependent kinase CDK2. Here we show that CDC7 is dispensable for cell division of many different cell types, as determined using chemical genetic systems that enable acute shutdown of CDC7 in cultured cells and in live mice. We demonstrate that another cell cycle kinase, CDK1, is also active during G1/S transition both in cycling cells and in cells exiting quiescence. We show that CDC7 and CDK1 perform functionally redundant roles during G1/S transition, and at least one of these kinases must be present to allow S-phase entry. These observations revise our understanding of cell cycle progression by demonstrating that CDK1 physiologically regulates two distinct transitions during cell division cycle, whereas CDC7 has a redundant function in DNA replication.


Asunto(s)
Proteínas de Ciclo Celular , Fase G1 , Proteínas Serina-Treonina Quinasas , Proteolisis , Fase S , Animales , Proteínas de Ciclo Celular/metabolismo , Replicación del ADN , Ratones , Proteínas Serina-Treonina Quinasas/metabolismo
2.
Mol Cell ; 80(3): 410-422.e6, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33108758

RESUMEN

While effective anti-cancer drugs targeting the CHK1 kinase are advancing in the clinic, drug resistance is rapidly emerging. Here, we demonstrate that CRISPR-mediated knockout of the little-known gene FAM122A/PABIR1 confers cellular resistance to CHK1 inhibitors (CHK1is) and cross-resistance to ATR inhibitors. Knockout of FAM122A results in activation of PP2A-B55α, a phosphatase that dephosphorylates the WEE1 protein and rescues WEE1 from ubiquitin-mediated degradation. The resulting increase in WEE1 protein expression reduces replication stress, activates the G2/M checkpoint, and confers cellular resistance to CHK1is. Interestingly, in tumor cells with oncogene-driven replication stress, CHK1 can directly phosphorylate FAM122A, leading to activation of the PP2A-B55α phosphatase and increased WEE1 expression. A combination of a CHK1i plus a WEE1 inhibitor can overcome CHK1i resistance of these tumor cells, thereby enhancing anti-cancer activity. The FAM122A expression level in a tumor cell can serve as a useful biomarker for predicting CHK1i sensitivity or resistance.


Asunto(s)
Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosfoproteínas/metabolismo , Pirazinas/farmacología , Pirazoles/farmacología , Animales , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Daño del ADN/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Nucleares/metabolismo , Fosfoproteínas/fisiología , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Tirosina Quinasas/genética , Pirazinas/metabolismo , Pirazoles/metabolismo , Transducción de Señal/efectos de los fármacos
3.
Trends Biochem Sci ; 47(12): 1009-1022, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35835684

RESUMEN

Cell cycle-dependent gene transcription is tightly controlled by the retinoblastoma (RB):E2F and DREAM complexes, which repress all cell cycle genes during quiescence. Cyclin-dependent kinase (CDK) phosphorylation of RB and DREAM allows for the expression of two gene sets. The first set of genes, with peak expression in G1/S, is activated by E2F transcription factors (TFs) and is required for DNA synthesis. The second set, with maximum expression during G2/M, is required for mitosis and is coordinated by the MuvB complex, together with B-MYB and Forkhead box M1 (FOXM1). In this review, we summarize the key findings that established the distinct control mechanisms regulating G1/S and G2/M gene expression in mammals and discuss recent advances in the understanding of the temporal control of these genes.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas Represoras , Animales , Proteínas Represoras/genética , Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Mitosis , Quinasas Ciclina-Dependientes/genética , Expresión Génica , Mamíferos
4.
PLoS Pathog ; 13(10): e1006668, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29028833

RESUMEN

Merkel cell carcinoma (MCC) frequently contains integrated copies of Merkel cell polyomavirus DNA that express a truncated form of Large T antigen (LT) and an intact Small T antigen (ST). While LT binds RB and inactivates its tumor suppressor function, it is less clear how ST contributes to MCC tumorigenesis. Here we show that ST binds specifically to the MYC homolog MYCL (L-MYC) and recruits it to the 15-component EP400 histone acetyltransferase and chromatin remodeling complex. We performed a large-scale immunoprecipitation for ST and identified co-precipitating proteins by mass spectrometry. In addition to protein phosphatase 2A (PP2A) subunits, we identified MYCL and its heterodimeric partner MAX plus the EP400 complex. Immunoprecipitation for MAX and EP400 complex components confirmed their association with ST. We determined that the ST-MYCL-EP400 complex binds together to specific gene promoters and activates their expression by integrating chromatin immunoprecipitation with sequencing (ChIP-seq) and RNA-seq. MYCL and EP400 were required for maintenance of cell viability and cooperated with ST to promote gene expression in MCC cell lines. A genome-wide CRISPR-Cas9 screen confirmed the requirement for MYCL and EP400 in MCPyV-positive MCC cell lines. We demonstrate that ST can activate gene expression in a EP400 and MYCL dependent manner and this activity contributes to cellular transformation and generation of induced pluripotent stem cells.


Asunto(s)
Antígenos Virales de Tumores/metabolismo , Carcinoma de Células de Merkel/virología , Transformación Celular Viral/fisiología , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Antígenos Transformadores de Poliomavirus/metabolismo , Carcinoma de Células de Merkel/genética , Carcinoma de Células de Merkel/metabolismo , Línea Celular Tumoral , Humanos , Immunoblotting , Inmunoprecipitación , Poliomavirus de Células de Merkel , Infecciones por Polyomavirus/complicaciones , Infecciones por Polyomavirus/genética , Infecciones por Polyomavirus/metabolismo , Infecciones Tumorales por Virus/complicaciones , Infecciones Tumorales por Virus/genética , Infecciones Tumorales por Virus/metabolismo
5.
bioRxiv ; 2023 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-37873169

RESUMEN

Targeted protein degradation by the ubiquitin-proteasome system is an essential mechanism regulating cellular division. The kinase PLK1 coordinates protein degradation at the G2/M phase of the cell cycle by promoting the binding of substrates to the E3 ubiquitin ligase SCFßTrCP. However, the magnitude to which PLK1 shapes the mitotic proteome has not been characterized. Combining deep, quantitative proteomics with pharmacologic PLK1 inhibition (PLK1i), we identified more than 200 proteins whose abundances were increased by PLK1i at G2/M. We validate many new PLK1-regulated proteins, including several substrates of the cell cycle E3 SCFCyclin F, demonstrating that PLK1 promotes proteolysis through at least two distinct SCF-family E3 ligases. Further, we found that the protein kinase A anchoring protein AKAP2 is cell cycle regulated and that its mitotic degradation is dependent on the PLK1/ßTrCP-signaling axis. Interactome analysis revealed that the strongest interactors of AKAP2 function in signaling networks regulating proliferation, including MAPK, AKT, and Hippo. Altogether, our data demonstrate that PLK1 coordinates a widespread program of protein breakdown at G2/M. We propose that dynamic proteolytic changes mediated by PLK1 integrate proliferative signals with the core cell cycle machinery during cell division. This has potential implications in malignancies where PLK1 is aberrantly regulated.

6.
Cell Rep ; 34(9): 108808, 2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33657372

RESUMEN

To identify genes whose loss confers resistance to CHK1 inhibitors, we perform genome-wide CRISPR-Cas9 screens in non-small-cell lung cancer (NSCLC) cell lines treated with the CHK1 inhibitor prexasertib (CHK1i). Five of the top six hits of the screens, MYBL2 (B-MYB), LIN54, FOXM1, cyclin A2 (CCNA2), and CDC25B, are cell-cycle-regulated genes that contribute to entry into mitosis. Knockout of MMB-FOXM1 complex components LIN54 and FOXM1 reduce CHK1i-induced DNA replication stress markers and premature mitosis during Late S phase. Activation of a feedback loop between the MMB-FOXM1 complex and CDK1 is required for CHK1i-induced premature mitosis in Late S phase and subsequent replication catastrophe, indicating that dysregulation of the S to M transition is necessary for CHK1 inhibitor sensitivity. These findings provide mechanistic insights into small molecule inhibitors currently studied in clinical trials and provide rationale for combination therapies.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Proteínas de Ciclo Celular/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Proteína Forkhead Box M1/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Mitosis/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Pirazinas/farmacología , Pirazoles/farmacología , Transactivadores/metabolismo , Células A549 , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Proteína Forkhead Box M1/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Complejos Multiproteicos , Transducción de Señal , Transactivadores/genética
8.
Cell Rep ; 16(12): 3359-3372, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27653696

RESUMEN

The anaphase promoting complex/cyclosome (APC/C) is an ubiquitin ligase and core component of the cell-cycle oscillator. During G1 phase, APC/C binds to its substrate receptor Cdh1 and APC/C(Cdh1) plays an important role in restricting S-phase entry and maintaining genome integrity. We describe a reciprocal feedback circuit between APC/C and a second ubiquitin ligase, the SCF (Skp1-Cul1-F box). We show that cyclin F, a cell-cycle-regulated substrate receptor (F-box protein) for the SCF, is targeted for degradation by APC/C. Furthermore, we establish that Cdh1 is itself a substrate of SCF(cyclin F). Cyclin F loss impairs Cdh1 degradation and delays S-phase entry, and this delay is reversed by simultaneous removal of Cdh1. These data indicate that the coordinated, temporal ordering of cyclin F and Cdh1 degradation, organized in a double-negative feedback loop, represents a fundamental aspect of cell-cycle control. This mutual antagonism could be a feature of other oscillating systems.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Ciclinas/metabolismo , Retroalimentación Fisiológica/fisiología , Fase S/fisiología , Células HEK293 , Células HeLa , Humanos
9.
Semin Oncol ; 31(3): 374-81, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15190495

RESUMEN

Current conventional surgical and pathological techniques substantially understage colon cancer. This is evidenced by the fact that a significant subset of patients who are stage I and II at the time of colectomy return with distant metastases and ultimately succumb to the disease within the next 5 years. The identification of more nodes within a specimen and the detailed analysis of lymph nodes with advanced pathological techniques such as immunohistochemistry and reverse-transcriptase polymerase chain reaction (RT-PCR) can improve the staging of colon cancer, but are also associated with tremendous financial, time, and labor constraints. Sentinel lymph node (SLN) mapping has provided an avenue of staging colon cancer with high success rates and accuracy rates, while maintaining cost- and time-effectiveness. The ability to reproduce these results is dependent on adherence to the technical details of the procedure, and thereby providing the pathologist with the true SLNs, upon which the advanced pathological studies can be applied. We report our experience of SLN mapping for colon tumors in 209 patients, elaborating on the materials used, technical details, pitfalls, and results of the procedure. Our results show a success rate of 100% (209/209) and an overall accuracy rate for predicting positive or negative metastatic disease of 96.2% (201/209). Nodal metastases were identified in 46.2% (85/184) of patients with invasive disease (stage T1 to T4). The SLN was the exclusive site of metastases in 38.8% (33/85) of these patients, and the nodal disease was detected only as micrometastases in 22.4% (19/85). The skip metastases rate (false negatives) was 9.4% (8/85). SLN mapping is a powerful tool for accurate staging of colon cancer with a high success rate. The upstaging associated with this procedure may reveal disease that might otherwise go undetected by conventional surgical and pathological methods. Those patients who are upstaged can then benefit from adjuvant chemotherapy, which has been shown to improve survival of colon cancer patients with nodal disease by at least 33%.


Asunto(s)
Neoplasias del Colon/patología , Biopsia del Ganglio Linfático Centinela , Neoplasias del Colon/cirugía , Colorantes , Fluoresceína , Humanos , Metástasis Linfática/diagnóstico , Estadificación de Neoplasias/métodos , Radiofármacos , Colorantes de Rosanilina , Biopsia del Ganglio Linfático Centinela/métodos , Azufre Coloidal Tecnecio Tc 99m
10.
Spine (Phila Pa 1976) ; 28(6): E118-20, 2003 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-12642775

RESUMEN

STUDY DESIGN: Descriptive case report. OBJECTIVES: To report the case of a child with medial compartment syndrome of the foot following posterior spinal instrumentation and fusion. SUMMARY OF BACKGROUND DATA: No previous study has reported medial compartment syndrome of the foot following spinal surgery. METHODS: A 15-year-old female with progressive idiopathic scoliosis was taken for posterior instrumentation and fusion. The patient had a history of severe postexertional cramping in the feet following athletics. Surgery progressed uneventfully and the patient was continuously monitored with somatosensory-evoked potentials, which showed no changes. In the recovery room, the patient complained of severe cramping in one foot that was similar to her postexertional cramping. This was lessened with massage and ketorolac. Soreness continued in the foot into postoperative day one and then increased overnight. On the morning of postoperative day 2, pressure in the medial compartment was found to be 97 mm Hg and she was taken for fasciotomy, which found necrosis of the abductor hallucis muscle, and all other compartments of the foot were normal. RESULTS: At the 6-month follow-up, the patient is doing well with no known sequelae. CONCLUSION: This was a very rare case of medial compartment syndrome of the foot following spine surgery. We believe that the patient had a predisposition, whether neurologic or vascular, toward cramping in the foot and that this activity was stimulated by the nerve stimulation during the evoked potential monitoring. Although the patient had thoracic epidural analgesia after surgery, it was not felt to have contributed to the development or result of the compartment syndrome. We strongly advocate for checking patients feet and legs during surgery for overactivity and stress the need for a high index of suspicion for compartment syndrome for unexplained pain after surgery.


Asunto(s)
Síndromes Compartimentales/diagnóstico , Síndromes Compartimentales/etiología , Pie/fisiopatología , Escoliosis/cirugía , Fusión Vertebral/efectos adversos , Adolescente , Síndromes Compartimentales/fisiopatología , Síndromes Compartimentales/cirugía , Femenino , Pie/cirugía , Humanos , Calambre Muscular/etiología , Músculo Esquelético/fisiopatología , Músculo Esquelético/cirugía , Necrosis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA