Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Proc Natl Acad Sci U S A ; 116(47): 23625-23635, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31690663

ABSTRACT

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.


Subject(s)
Adenocarcinoma/genetics , Cell Cycle Proteins/metabolism , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/physiology , Transcription Factors/physiology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , CD146 Antigen/metabolism , Cell Movement , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Gene Knockdown Techniques , Genes, Tumor Suppressor , HCT116 Cells , HT29 Cells , Heterografts , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasm Transplantation , RNA Interference , RNA, Small Interfering/genetics , Transcription Factors/deficiency , Transcription Factors/genetics
2.
Proc Natl Acad Sci U S A ; 115(44): E10417-E10426, 2018 10 30.
Article in English | MEDLINE | ID: mdl-30327349

ABSTRACT

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.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , DNA Transposable Elements/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Animals , Apoptosis/genetics , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Diet, High-Fat/adverse effects , Liver/pathology , Mice , Mutagenesis/genetics , Oncogenes/genetics , Signal Transduction/genetics , Up-Regulation/genetics
3.
Proc Natl Acad Sci U S A ; 114(35): E7301-E7310, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28811376

ABSTRACT

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.


Subject(s)
Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , beta Karyopherins/genetics , beta Karyopherins/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Ovarian Epithelial , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor/methods , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Ivermectin/pharmacology , Loss of Function Mutation/genetics , Neoplasms, Glandular and Epithelial/metabolism , Oncogenes , Ovarian Neoplasms/metabolism , Paclitaxel/pharmacology , RNA, Small Interfering/genetics
4.
Biochem Biophys Res Commun ; 513(4): 1027-1034, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31010678

ABSTRACT

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.


Subject(s)
Carcinogenesis/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Liver Neoplasms/pathology , RNA-Binding Proteins/pharmacology , Animals , Cell Line , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Mice , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/pharmacology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
5.
Exp Cell Res ; 369(2): 179-186, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29842878

ABSTRACT

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.


Subject(s)
Mast Cells/cytology , Animals , Cell Culture Techniques/history , Cell Culture Techniques/methods , Cell Differentiation , Cell Separation/history , Cell Separation/methods , History, 19th Century , History, 20th Century , History, 21st Century , Humans , Mast Cells/physiology , Mice , Rats
7.
Nat Cell Biol ; 10(9): 1083-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-19160489

ABSTRACT

The anaphase promoting complex (APC) or cyclosome is a multisubunit E3 ubiquitin ligase. Cdc20 (fizzy (fzy)) or p55CDC, and Cdh1 (Hct1, srw1 or fizzy-related 1 (fzr1)) encode two adaptor proteins that bring substrates to the APC. Both APC-Cdc20 and APC-Cdh1 have been implicated in the control of mitosis through mediating ubiquitination of mitotic regulators, such as cyclin B1 and securin. However, the importance of Cdh1 function in vivo and whether its function is redundant with that of Cdc20 are unclear. Here we have analysed mice lacking Cdh1. We show that Cdh1 is essential for placental development and that its deficiency causes early lethality. Cdhl-deficient mouse embryonic fibroblasts (MEFs) entered replicative senescence prematurely because of stabilization of Ets2 and subsequent activation of p6(Ink4a) expression. These results have uncovered an unexpected role of the APC in maintaining replicative lifespan of MEFs. Further, Cdh1 heterozygous mice show defects in late-phase long-term potentiation (L-LTP) in the hippocampus and are deficient in contextual fear-conditioning, suggesting that Cdh1 has a role in learning and memory.


Subject(s)
Cell Cycle Proteins/metabolism , Cellular Senescence , Memory/physiology , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Cdh1 Proteins , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Embryo Loss/metabolism , Fibroblasts/enzymology , Fibroblasts/pathology , Heterozygote , In Vitro Techniques , Mice , Protein Stability , Proto-Oncogene Protein c-ets-2/metabolism , Substrate Specificity
8.
Res Sq ; 2024 May 15.
Article in English | MEDLINE | ID: mdl-38798564

ABSTRACT

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.

9.
Proc Natl Acad Sci U S A ; 107(32): 14188-93, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20663956

ABSTRACT

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.


Subject(s)
Aneuploidy , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Neoplasms/etiology , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , DNA Damage , Mice , Mice, Transgenic , Reactive Oxygen Species
10.
Cancers (Basel) ; 14(11)2022 May 24.
Article in English | MEDLINE | ID: mdl-35681566

ABSTRACT

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.

11.
Cancers (Basel) ; 14(7)2022 Mar 27.
Article in English | MEDLINE | ID: mdl-35406470

ABSTRACT

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.

12.
Cancer Res ; 81(21): 5413-5424, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34475109

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/genetics , DNA Transposable Elements , Leiomyosarcoma/pathology , Lung Neoplasms/secondary , Mutagenesis, Insertional , Mutation , Uterine Neoplasms/pathology , Animals , Female , Humans , Leiomyosarcoma/etiology , Leiomyosarcoma/metabolism , Lung Neoplasms/etiology , Lung Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , PTEN Phosphohydrolase/physiology , Proto-Oncogene Proteins p21(ras)/physiology , Transposases/genetics , Transposases/metabolism , Uterine Neoplasms/etiology , Uterine Neoplasms/metabolism
15.
Nat Biotechnol ; 34(8): 845-51, 2016 08.
Article in English | MEDLINE | ID: mdl-27398792

ABSTRACT

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.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Colon/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Gene Expression Profiling/methods , Neoplasm Proteins/metabolism , Carcinogenesis/genetics , Cell-Free System/metabolism , Cells, Cultured , Colon/pathology , Genes, Neoplasm/genetics , Humans , Organogenesis , Tissue Engineering/methods
16.
Nat Genet ; 47(2): 142-50, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25559195

ABSTRACT

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.


Subject(s)
Adenocarcinoma/genetics , Adenoma/genetics , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mutagenesis, Insertional/methods , Adenocarcinoma/pathology , Adenoma/pathology , Animals , Biological Evolution , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cohort Studies , Colorectal Neoplasms/pathology , DNA Transposable Elements , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Progression , Female , Gene Knockdown Techniques , Genes, Tumor Suppressor , High-Throughput Nucleotide Sequencing , Humans , Male , Mice , Mice, Inbred C57BL , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Sequence Analysis, DNA , Signal Transduction , Specific Pathogen-Free Organisms , Transcription Factors/genetics , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
17.
Cell Cycle ; 10(4): 671-9, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21301228

ABSTRACT

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.


Subject(s)
Cell Cycle , Cell Nucleolus/metabolism , DNA Damage , Dual-Specificity Phosphatases/metabolism , Protein Kinases/metabolism , Blotting, Western , Cell Line , Cell Nucleolus/enzymology , Cell Nucleolus/genetics , Checkpoint Kinase 1 , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Human/genetics , Chromosomes, Human/physiology , Dual-Specificity Phosphatases/genetics , Genomic Instability , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Phosphorylation , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering , Staurosporine/analogs & derivatives , Staurosporine/pharmacology
18.
Mol Cell Biol ; 31(7): 1470-7, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21262768

ABSTRACT

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.


Subject(s)
Aging, Premature/genetics , Aging, Premature/pathology , DNA Damage , Dual-Specificity Phosphatases/deficiency , Aging, Premature/metabolism , Animals , Cellular Senescence , DNA Repair , Dual-Specificity Phosphatases/metabolism , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Fertility , Fibroblasts/metabolism , Fibroblasts/pathology , G2 Phase , Memory , Mice , Mitosis
19.
Mol Cell Biol ; 30(18): 4463-79, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20624912

ABSTRACT

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.


Subject(s)
Endocytosis/physiology , Focal Adhesions/metabolism , Integrin beta1/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins , Cell Adhesion/physiology , Cell Line , Cell Movement/physiology , Clathrin/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Dynamin II/genetics , Dynamin II/metabolism , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tumor Suppressor Proteins , Zyxin
20.
J Cell Biol ; 185(6): 983-94, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19528295

ABSTRACT

Genomic instability is a hallmark of human cancers. Spindle assembly checkpoint (SAC) is a critical cellular mechanism that prevents chromosome missegregation and therefore aneuploidy by blocking premature separation of sister chromatids. Thus, SAC, much like the DNA damage checkpoint, is essential for genome stability. In this study, we report the generation and analysis of mice carrying a Cdc20 allele in which three residues critical for the interaction with Mad2 were mutated to alanine. The mutant Cdc20 protein (AAA-Cdc20) is no longer inhibited by Mad2 in response to SAC activation, leading to the dysfunction of SAC and aneuploidy. The dysfunction could not be rescued by the additional expression of another Cdc20 inhibitor, BubR1. Furthermore, we found that Cdc20(AAA/AAA) mice died at late gestation, but Cdc20(+/AAA) mice were viable. Importantly, Cdc20(+/AAA) mice developed spontaneous tumors at highly accelerated rates, indicating that the SAC-mediated inhibition of Cdc20 is an important tumor-suppressing mechanism.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Genes, cdc , Neoplasms/genetics , Spindle Apparatus/metabolism , Amino Acid Sequence , Aneuploidy , Animals , Cdc20 Proteins , Cell Cycle Proteins/genetics , Cells, Cultured , Chromosomal Instability , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Karyotyping , Mad2 Proteins , Mice , Molecular Sequence Data , Neoplasms/metabolism , Neoplasms/pathology , Nocodazole/metabolism , Point Mutation , Sequence Alignment , Tubulin Modulators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL