Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Res Sq ; 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38798564

RESUMEN

Studying lung adenocarcinoma (LUAD) early carcinogenesis is challenging, primarily due to the lack of LUAD precursors specimens. We amassed multi-omics data from 213 LUAD and LUAD precursors to identify molecular features underlying LUAD precancer evolution. We observed progressively increasing mutations, chromosomal aberrations, whole genome doubling and genomic instability from precancer to invasive LUAD, indicating aggravating chromosomal instability (CIN). Telomere shortening, a crucial genomic alteration linked to CIN, emerged at precancer stage. Moreover, later-stage lesions demonstrated increasing cancer stemness and decreasing alveolar identity, suggesting epithelial de-differentiation during early LUAD carcinogenesis. The innate immune cells progressively diminished from precancer to invasive LUAD, concomitant with a gradual recruitment of adaptive immune cells (except CD8+ and gamma-delta T cells that decreased in later stages) and upregulation of numerous immune checkpoints, suggesting LUAD precancer evolution is associated with a shift from innate to adaptive immune response and immune evasion mediated by various mechanisms.

3.
Cancers (Basel) ; 14(11)2022 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-35681566

RESUMEN

Hepatocellular carcinoma (HCC) is one of the deadliest cancers worldwide and the only cancer with an increasing incidence in the United States. Recent advances in sequencing technology have enabled detailed profiling of liver cancer genomes and revealed extensive inter- and intra-tumor heterogeneity, making it difficult to identify driver genes for HCC. To identify HCC driver genes, we performed transposon mutagenesis screens in a mouse HBV model of HCC and discovered many candidate cancer genes (SB/HBV-CCGs). Here, we show that one of these genes, RNF125 is a potent anti-proliferative tumor suppressor gene in HCC. RNF125 is one of nine CCGs whose expression was >3-fold downregulated in human HCC. Depletion of RNF125 in immortalized mouse liver cells led to tumor formation in transplanted mice and accelerated growth of human liver cancer cell lines, while its overexpression inhibited their growth, demonstrating the tumor-suppressive function of RNF125 in mouse and human liver. Whole-transcriptome analysis revealed that RNF125 transcriptionally suppresses multiple genes involved in cell proliferation and/or liver regeneration, including Egfr, Met, and Il6r. Blocking Egfr or Met pathway expression inhibited the increased cell proliferation observed in RNF125 knockdown cells. In HCC patients, low expression levels of RNF125 were correlated with poor prognosis demonstrating an important role for RNF125 in HCC. Collectively, our results identify RNF125 as a novel anti-proliferative tumor suppressor in HCC.

4.
Cancers (Basel) ; 14(7)2022 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-35406470

RESUMEN

Exosomes are crucial extracellular vesicles (EVs) with a diameter of approximately 30-200 nm. They are released by most cell types in their extracellular milieu and carry various biomolecules, including proteins and nucleic acids. Exosomes are increasingly studied in various diseases, including cancer, due to their role in local and distant cell-cell communication in which they can promote tumor growth, cancer progression, and metastasis. Interestingly, a tremendous number of exosomes is released by malignant cancer cells, and these are then taken up by autologous and heterologous recipient stromal cells such as immune cells, cancer stem cells, and endothelial cells. All these events demand an enormous amount of energy and require that exosomes remain stable while having the capacity to reach distant sites and cross physical barriers. Nevertheless, there is a dearth of research pertaining to the energy sources of exosomes, and questions remain about how they maintain their motility in the tumor microenvironment (TME) and beyond. Moreover, exosomes can produce adenosine triphosphate (ATP), an important energy molecule required by all cells, and mitochondria have been identified as one of the exosomal cargoes. These findings strengthen the prospect of exosomal communication via transfer of mitochondria and the bioenergetics of target recipient cells. In the TME, the accumulation of ATP and lactate may facilitate the entry of exosomes into cancer cells to promote metastasis, as well as help to target cancer cells at the tumor site. This review highlights how exosomes obtain sufficient energy to thrive in the TME and communicate with distant physiological destinations.

5.
Cancer Res ; 81(21): 5413-5424, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34475109

RESUMEN

Uterine leiomyosarcoma (ULMS) is a malignancy, which arises from the uterine smooth muscle. Because of its rarity, aggressive nature, and extremely poor prognosis, the molecular mechanisms driving ULMS remain elusive. To identify candidate cancer genes (CCG) driving ULMS, we conducted an in vivo Sleeping Beauty (SB) transposon mutagenesis screen in uterine myometrium-specific, PTEN knockout, KRAS mutant (PTEN KO/KRAS) mice. ULMS quickly developed in SB PTEN KO/KRAS mice, but not in PTEN KO/KRAS mice, demonstrating the critical importance of SB mutagenesis for driving ULMS in this model. Subsequent sequencing of SB insertion sites in these tumors identified 19 ULMS CCGs that were significantly enriched in known cancer genes. Among them, Zfp217 and Sfmbt2 functioned at early stages of tumor initiation and appeared to be oncogenes. Expression of ZNF217, the human homolog of ZFP217, was shown to be elevated in human ULMS compared with paired normal uterine smooth muscle, where it negatively correlated with patient prognosis. Inhibition of ZNF217 suppressed, whereas overexpression induced, proliferation, survival, migration, and stemness of human ULMS. In a second ex vivo ULMS SB metastasis screen, three CCGs were identified that may drive ULMS metastasis to the lung. One of these CCGs, Nrd1 (NRDC in humans), showed stronger expression in human metastatic tumors compared with primary ULMS and negatively associated with patient survival. NRDC knockdown impaired migration and adhesion without affecting cell proliferation, whereas overexpression had the opposite effect. Together, these results reveal novel mechanism driving ULMS tumorigenesis and metastasis and identify ZNF217 and NRDC as potential targets for ULMS therapy. SIGNIFICANCE: An in vivo Sleeping Beauty transposon mutagenesis screen identifies candidate cancer genes that drive initiation and progression of uterine leiomyosarcoma and may serve as therapeutic targets.


Asunto(s)
Biomarcadores de Tumor/genética , Elementos Transponibles de ADN , Leiomiosarcoma/patología , Neoplasias Pulmonares/secundario , Mutagénesis Insercional , Mutación , Neoplasias Uterinas/patología , Animales , Femenino , Humanos , Leiomiosarcoma/etiología , Leiomiosarcoma/metabolismo , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfohidrolasa PTEN/fisiología , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Transposasas/genética , Transposasas/metabolismo , Neoplasias Uterinas/etiología , Neoplasias Uterinas/metabolismo
7.
Proc Natl Acad Sci U S A ; 116(47): 23625-23635, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31690663

RESUMEN

Myocardin-related transcription factor B (MRTFB) is a candidate tumor-suppressor gene identified in transposon mutagenesis screens of the intestine, liver, and pancreas. Using a combination of cell-based assays, in vivo tumor xenograft assays, and Mrtfb knockout mice, we demonstrate here that MRTFB is a human and mouse colorectal cancer (CRC) tumor suppressor that functions in part by inhibiting cell invasion and migration. To identify possible MRTFB transcriptional targets, we performed whole transcriptome RNA sequencing in MRTFB siRNA knockdown primary human colon cells and identified 15 differentially expressed genes. Among the top candidate tumor-suppressor targets were melanoma cell adhesion molecule (MCAM), a known tumor suppressor, and spindle apparatus coiled-coil protein 1 (SPDL1), which has no confirmed role in cancer. To determine whether these genes play a role in CRC, we knocked down the expression of MCAM and SPDL1 in human CRC cells and showed significantly increased invasion and migration of tumor cells. We also showed that Spdl1 expression is significantly down-regulated in Mrtfb knockout mouse intestine, while lower SPDL1 expression levels are significantly associated with reduced survival in CRC patients. Finally, we show that depletion of MCAM and SPDL1 in human CRC cells significantly increases tumor development in xenograft assays, further confirming their tumor-suppressive roles in CRC. Collectively, our findings demonstrate the tumor-suppressive role of MRTFB in CRC and identify several genes, including 2 tumor suppressors, that act downstream of MRTFB to regulate tumor growth and survival in CRC patients.


Asunto(s)
Adenocarcinoma/genética , Proteínas de Ciclo Celular/metabolismo , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/fisiología , Factores de Transcripción/fisiología , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Antígeno CD146/metabolismo , Movimiento Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Femenino , Técnicas de Silenciamiento del Gen , Genes Supresores de Tumor , Células HCT116 , Células HT29 , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Interferencia de ARN , ARN Interferente Pequeño/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
9.
Biochem Biophys Res Commun ; 513(4): 1027-1034, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31010678

RESUMEN

Colorectal cancer (CRC) and liver cancer are the second and fourth leading causes of cancer-related deaths in the whole world, respectively, and each year over 1.6 million people die from these diseases. To identify driver genes in CRC and liver cancer, we have performed Sleeping Beauty transposon mutagenesis screens in mouse models. Zinc finger RNA binding protein, ZFR, was one of the novel candidate cancer genes identified in these forward genetic screens. Consistent with this discovery, a pan-cancer analysis of sequencing results of thousands of human cancer genomes demonstrated that ZFR is a potential potent oncogene. In this study, we aimed to investigate ZFR's roles in both types of cancer and found that overexpression of ZFR in CRC and liver cancer cells led to accelerated tumor development. Consistently, knockdown of ZFR resulted in significantly decelerated tumor development. ZFR overexpression also promoted tumor development of immortalized mouse liver cells. ZFR overexpression and shRNA knockdown led to accelerated and decelerated cell proliferation, respectively, indicating that ZFR promotes tumor development mainly by regulating cell proliferation. To identify ZFR's targets in transcription, we performed whole transcriptome sequencing using ZFR small interfering RNAs in a primary human colon cell line. All potential target genes were validated by real time PCR. FAM49B was a tumor suppressor candidate for ZFR targets. When we knocked down the expression of FAM49B in CRC and liver cancer cells, we observed significantly accelerated cell proliferation, consistent with the results with ZFR overexpression. The results presented here demonstrate the oncogenic role of ZFR in both CRC and liver cancer, providing a potential drug target for both cancers' treatment. We also identified ZFR's potential transcriptional targets, and further investigations on those targets, especially FAM49B, will help us understand more about the important role of ZFR in digestive system cancers.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Neoplasias Hepáticas/patología , Proteínas de Unión al ARN/farmacología , Animales , Línea Celular , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/farmacología , Ratones , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/farmacología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
10.
Proc Natl Acad Sci U S A ; 115(44): E10417-E10426, 2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30327349

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is the fastest rising cause of hepatocellular carcinoma (HCC) in Western countries; however, the molecular mechanisms that cause NAFLD-HCC remain elusive. To identify molecular drivers of NAFLD-HCC, we performed Sleeping Beauty (SB) transposon mutagenesis screens in liver-specific Pten knockout and in high-fat diet-fed mice, which are murine models of NAFLD-HCC. SB mutagenesis accelerated liver tumor formation in both models and identified 588 and 376 candidate cancer genes (CCGs), respectively; 257 CCGs were common to both screens and were enriched in signaling pathways known to be important for human HCC. Comparison of these CCGs with those identified in a previous SB screen of hepatitis B virus-induced HCC identified a core set of 141 CCGs that were mutated in all screens. Forty-one CCGs appeared specific for NAFLD-HCC, including Sav1, a component of the Hippo signaling pathway and the most frequently mutated gene identified in both NAFLD-HCC screens. Liver-specific deletion of Sav1 was found to promote hepatic lipid accumulation, apoptosis, and fibrogenesis, leading to the acceleration of hepatocarcinogenesis in liver-specific Pten mutant mice. Sav1/Pten double-mutant livers also showed a striking up-regulation of markers of liver progenitor cells (LPCs), along with synergistic activation of Yap, which is a major downstream effector of Hippo signaling. Lastly, Yap activation, in combination with Pten inactivation, was found to accelerate cell growth and sphere formation of LPCs in vitro and induce their malignant transformation in allografts. Our forward genetic screens in mice have thus identified pathways and genes driving the development of NAFLD-HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Elementos Transponibles de ADN/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/patología , Animales , Apoptosis/genética , Carcinogénesis/genética , Carcinogénesis/patología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Dieta Alta en Grasa/efectos adversos , Hígado/patología , Ratones , Mutagénesis/genética , Oncogenes/genética , Transducción de Señal/genética , Regulación hacia Arriba/genética
11.
Exp Cell Res ; 369(2): 179-186, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29842878

RESUMEN

Mast cells (MCs) are tissue-based stationary effector cells that form the immune system's first-line defense against various challenges. They are developed from the bone marrow-derived progenitors to complete their differentiation and maturation in the tissues where they eventually establish residence. MCs have been implicated in many diseases, such as allergy, parasitic infection, and neoplastic disorders. Immortalized MC lines, such as RBL-2H3, HMC-1, and LAD-2, are useful for investigating the biological functions of MC only to some extents due to the restriction of degranulation evaluation, in vivo injection and other factors. Over the past few decades, technologies for acquiring primarily MCs have been continually optimized, and novel protocols have been proposed. However, no relevant publications have analyzed and summarized these techniques. In this review, the classical approaches for extracting MCs are generalized, and new methods with potential values are introduced. We also evaluate the advantages and applicability of diverse MC models. Since MCs exhibit substantial plasticity and functional diversity due to different origins, it is both necessary and urgent to select a reliable and suitable source of MCs for a particular study.


Asunto(s)
Mastocitos/citología , Animales , Técnicas de Cultivo de Célula/historia , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Separación Celular/historia , Separación Celular/métodos , Historia del Siglo XIX , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Mastocitos/fisiología , Ratones , Ratas
12.
Proc Natl Acad Sci U S A ; 114(35): E7301-E7310, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28811376

RESUMEN

Epithelial ovarian cancer (EOC) is a deadly cancer, and its prognosis has not been changed significantly during several decades. To seek new therapeutic targets for EOC, we performed an in vivo dropout screen in human tumor xenografts using a pooled shRNA library targeting thousands of druggable genes. Then, in follow-up studies, we performed a second screen using a genome-wide CRISPR/Cas9 library. These screens identified 10 high-confidence drug targets that included well-known oncogenes such as ERBB2 and RAF1, and novel oncogenes, notably KPNB1, which we investigated further. Genetic and pharmacological inhibition showed that KPNB1 exerts its antitumor effects through multiphase cell cycle arrest and apoptosis induction. Mechanistically, proteomic studies revealed that KPNB1 acts as a master regulator of cell cycle-related proteins, including p21, p27, and APC/C. Clinically, EOC patients with higher expression levels of KPNB1 showed earlier recurrence and worse prognosis than those with lower expression levels of KPNB1. Interestingly, ivermectin, a Food and Drug Administration-approved antiparasitic drug, showed KPNB1-dependent antitumor effects on EOC, serving as an alternative therapeutic toward EOC patients through drug repositioning. Last, we found that the combination of ivermectin and paclitaxel produces a stronger antitumor effect on EOC both in vitro and in vivo than either drug alone. Our studies have thus identified a combinatorial therapy for EOC, in addition to a plethora of potential drug targets.


Asunto(s)
Neoplasias Glandulares y Epiteliales/genética , Neoplasias Ováricas/genética , beta Carioferinas/genética , beta Carioferinas/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Carcinoma Epitelial de Ovario , Puntos de Control del Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Ivermectina/farmacología , Mutación con Pérdida de Función/genética , Neoplasias Glandulares y Epiteliales/metabolismo , Oncogenes , Neoplasias Ováricas/metabolismo , Paclitaxel/farmacología , ARN Interferente Pequeño/genética
13.
Nat Biotechnol ; 34(8): 845-51, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27398792

RESUMEN

Refined cancer models are needed to bridge the gaps between cell line, animal and clinical research. Here we describe the engineering of an organotypic colon cancer model by recellularization of a native human matrix that contains cell-populated mucosa and an intact muscularis mucosa layer. This ex vivo system recapitulates the pathophysiological progression from APC-mutant neoplasia to submucosal invasive tumor. We used it to perform a Sleeping Beauty transposon mutagenesis screen to identify genes that cooperate with mutant APC in driving invasive neoplasia. We identified 38 candidate invasion-driver genes, 17 of which, including TCF7L2, TWIST2, MSH2, DCC, EPHB1 and EPHB2 have been previously implicated in colorectal cancer progression. Six invasion-driver genes that have not, to our knowledge, been previously described were validated in vitro using cell proliferation, migration and invasion assays and ex vivo using recellularized human colon. These results demonstrate the utility of our organoid model for studying cancer biology.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Colon/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Perfilación de la Expresión Génica/métodos , Proteínas de Neoplasias/metabolismo , Carcinogénesis/genética , Sistema Libre de Células/metabolismo , Células Cultivadas , Colon/patología , Genes Relacionados con las Neoplasias/genética , Humanos , Organogénesis , Ingeniería de Tejidos/métodos
14.
Nat Genet ; 47(2): 142-50, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25559195

RESUMEN

To provide a more comprehensive understanding of the genes and evolutionary forces driving colorectal cancer (CRC) progression, we performed Sleeping Beauty (SB) transposon mutagenesis screens in mice carrying sensitizing mutations in genes that act at different stages of tumor progression. This approach allowed us to identify a set of genes that appear to be highly relevant for CRC and to provide a better understanding of the evolutionary forces and systems properties of CRC. We also identified six genes driving malignant tumor progression and a new human CRC tumor-suppressor gene, ZNF292, that might also function in other types of cancer. Our comprehensive CRC data set provides a resource with which to develop new therapies for treating CRC.


Asunto(s)
Adenocarcinoma/genética , Adenoma/genética , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Mutagénesis Insercional/métodos , Adenocarcinoma/patología , Adenoma/patología , Animales , Evolución Biológica , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Estudios de Cohortes , Neoplasias Colorrectales/patología , Elementos Transponibles de ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Genes Supresores de Tumor , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Análisis de Secuencia de ADN , Transducción de Señal , Organismos Libres de Patógenos Específicos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Cell Cycle ; 10(15): 2414, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21734460
16.
Aging (Albany NY) ; 3(5): 456, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21666283
17.
Cell Cycle ; 10(4): 671-9, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21301228

RESUMEN

Chk1 is a critical effector of DNA damage checkpoints necessary for the maintenance of chromosome integrity during cell cycle progression. Here we report, that Chk1 co-localized with the nucleolar marker, fibrillarin in response to radiation-induced DNA damage in human cells. Interestingly, in vitro studies using GST pull down assays identified the dual-specificity serine/threonine nucleolar phosphatase Cdc14B as a Chk1 substrate. Furthermore, Chk1, but not a kinase-dead Chk1 control, was shown to phosphorylate Cdc14B using an in vitro kinase assay. Co-immunoprecipitation experiments using exogenous Cdc14B transfected into human cells confirmed the interaction of Cdc14B and Chk1 during cell cycle. In addition, reduction of Chk1 levels via siRNA or UCN-01 treatment demonstrated that Chk1 activation following DNA damage was required for Cdc14B export from the nucleolus. These studies have revealed a novel interplay between Chk1 kinase and Cdc14B phosphatase involving radiation-induced nucleolar shuttling to facilitate error-free cell cycle progression and prevent genomic instability.


Asunto(s)
Ciclo Celular , Nucléolo Celular/metabolismo , Daño del ADN , Fosfatasas de Especificidad Dual/metabolismo , Proteínas Quinasas/metabolismo , Western Blotting , Línea Celular , Nucléolo Celular/enzimología , Nucléolo Celular/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas Humanos/genética , Cromosomas Humanos/fisiología , Fosfatasas de Especificidad Dual/genética , Inestabilidad Genómica , Células HeLa , Humanos , Immunoblotting , Inmunoprecipitación , Fosforilación , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Estaurosporina/análogos & derivados , Estaurosporina/farmacología
18.
Mol Cell Biol ; 31(7): 1470-7, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21262768

RESUMEN

The Cdc14 dual-specificity phosphatase plays a key role in the mitotic exit of budding yeast cells. Mammals have two homologues, Cdc14a and Cdc14b. Unlike the yeast counterpart, neither Cdc14a nor Cdc14b seems to be essential for mitotic exit. To determine the physiological function of Cdc14b, we generated mice deficient in the phosphatase. The mutant mice were viable and did not display overt abnormalities. However, these mice developed signs of aging at much younger ages than the wild-type mice. At the cellular level, the Cdc14b-deficient mouse embryonic fibroblasts (MEFs) grew more slowly than the controls at later passages as a result of increased rates of senescence. Consistent with these premature-aging phenotypes, Cdc14b-deficient cells accumulated more endogenous DNA damage than the wild-type cells, and more Cdc14b-deficient MEFs entered senescence than control MEFs in response to exogenous DNA damage. However, no deficiencies in DNA damage checkpoint response were detected in Cdc14b mutant cells, suggesting that the function of Cdc14b is required for efficient DNA damage repair.


Asunto(s)
Envejecimiento Prematuro/genética , Envejecimiento Prematuro/patología , Daño del ADN , Fosfatasas de Especificidad Dual/deficiencia , Envejecimiento Prematuro/metabolismo , Animales , Senescencia Celular , Reparación del ADN , Fosfatasas de Especificidad Dual/metabolismo , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Fertilidad , Fibroblastos/metabolismo , Fibroblastos/patología , Fase G2 , Memoria , Ratones , Mitosis
19.
Mol Cell Biol ; 30(18): 4463-79, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20624912

RESUMEN

Cell migration requires the regulated disassembly of focal adhesions, but the underlying mechanisms remain poorly defined. We have previously shown that focal adhesion disassembly requires the dynamin 2- and clathrin-dependent endocytosis of ligand-activated beta1 integrins. Here, we identify type I phosphatidylinositol phosphate kinase beta (PIPKIbeta), an enzyme that generates phosphatidylinositol-4,5-bisphosphate (PI4,5P(2)), as a key regulator of this process. We found that knockdown of PIPKIbeta by RNA interference blocks the internalization of active beta1 integrins and impairs focal adhesion turnover and cell migration. These defects are caused by the failure to target the endocytic machinery, including clathrin adaptors and dynamin 2, to focal adhesion sites. As a consequence, depletion of PIPKIbeta blocks clathrin assembly at adhesion plaques and prevents complex formation between dynamin 2 and focal adhesion kinase (FAK), a critical step in focal adhesion turnover. Together, our findings identify PIPKIbeta as a novel regulator of focal adhesion disassembly and suggest that PIPKIbeta spatially regulates integrin endocytosis at adhesion sites to control cell migration.


Asunto(s)
Endocitosis/fisiología , Adhesiones Focales/metabolismo , Integrina beta1/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Adhesión Celular/fisiología , Línea Celular , Movimiento Celular/fisiología , Clatrina/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Dinamina II/genética , Dinamina II/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Interferencia de ARN , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Supresoras de Tumor , Zixina
20.
Proc Natl Acad Sci U S A ; 107(32): 14188-93, 2010 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-20663956

RESUMEN

The spindle assembly checkpoint (SAC) is essential for proper sister chromatid segregation. Defects in this checkpoint can lead to chromosome missegregation and aneuploidy. An increasing body of evidence suggests that aneuploidy can play a causal role in tumorigenesis. However, mutant mice that are prone to aneuploidy have only mild tumor phenotypes, suggesting that there are limiting factors in the aneuploidy-induced tumorigenesis. Here we provide evidence that p53 is such a limiting factor. We show that aneuploidy activates p53 and that loss of p53 drastically accelerates tumor development in two independent aneuploidy models. The p53 activation depends on the ataxia-telangiectasia mutated (ATM) gene product and increased levels of reactive oxygen species. Thus, the ATM-p53 pathway safeguards not only DNA damage but also aneuploidy.


Asunto(s)
Aneuploidia , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Neoplasias/etiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Daño del ADN , Ratones , Ratones Transgénicos , Especies Reactivas de Oxígeno
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA