Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cell Death Discov ; 4: 56, 2018.
Article in English | MEDLINE | ID: mdl-29760954

ABSTRACT

Anaplastic lymphoma kinase (ALK) is an oncogenic receptor tyrosine kinase that is activated by gene amplification and mutation in neuroblastomas. ALK inhibitors can delay the progression of ALK-driven cancers, but are of limited use owing to ALK inhibitor resistance. Here, we show that resistance to ALK inhibitor in ALK-driven neuroblastomas can be attenuated by combination treatment with a p53 activator. Either ALK inhibition or p53 activator treatment induced cell cycle arrest, whereas combination treatment induced apoptosis, and prevented tumour relapse both in vitro and in vivo. This shift toward apoptosis, and away from cell-cycle arrest, in the presence of an ALK inhibitor and a p53 activator, is mediated by inhibition of the ALK-AKT-FOXO3a axis leading to a specific upregulation of SOX4. SOX4 cooperates with p53 to upregulate the pro-apoptotic protein PUMA. These data therefore suggest a novel combination therapy strategy for treating ALK-driven neuroblastomas.

2.
Methods Mol Biol ; 1599: 145-156, 2017.
Article in English | MEDLINE | ID: mdl-28477117

ABSTRACT

Ataxia-telangiectasia mutated (ATM) protein is a member of the phosphatidylinositol 3-phosphate kinase (PI3-K)-related protein kinase (PIKK) family and is implicated in the initiation of signaling pathways following DNA double strand breaks (DSBs) elicited by exposure to ionizing irradiation (IR) or radiomimetic compounds. Loss of function of the ATM gene product results in the human genetic disorder ataxia-telangiectasia (A-T) characterized by neurodegeneration, immunodeficiency, genomic instability, and cancer predisposition. In response to DSBs, ATM is activated and phosphorylates Ser/Thr-Gln (S/T-Q) sequences on numerous proteins participating in DNA-damage responses. Among these proteins, phosphorylation of the tumor suppressor p53 at Ser15 is known as a target for ATM, which leads to the dissociation of MDM2, an E3 ubiquitin ligase, from p53 to prevent MDM2-dependent p53 degradation. Ser46 on p53 is phosphorylated in response to DSBs and contributes to the preferential transactivation of pro-apoptotic genes, such as p53AIP1, Noxa, and PUMA, to prevent tumor formation. Our group have shown that not only ATM preferentially phosphorylates S/T-Q sequences, but also Ser46, which is a noncanonical site with an S-P sequence for ATM. Ser46 on p53 is directly phosphorylated by ATM in a p53 conformation-dependent manner using the ATP analogue-accepting ATM mutant (ATM-AS) system. This protocol summarizes an approach to identify direct numerous targets for ATM kinase and is used to elucidate ATM signaling pathways in the DNA damage responses.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Damage/genetics , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Signal Transduction/genetics , Signal Transduction/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
EMBO Rep ; 18(1): 123-137, 2017 01.
Article in English | MEDLINE | ID: mdl-27856536

ABSTRACT

The ribosomal protein L11 (RPL11) binds and inhibits the MDM2 ubiquitin ligase, thereby promoting p53 stability. Thus, RPL11 acts as a tumor suppressor. Here, we show that GRWD1 (glutamate-rich WD40 repeat containing 1) physically and functionally interacts with RPL11. GRWD1 is localized to nucleoli and is released into the nucleoplasm upon nucleolar stress. Silencing of GRWD1 increases p53 induction by nucleolar stress, whereas overexpression of GRWD1 reduces p53 induction. Furthermore, GRWD1 overexpression competitively inhibits the RPL11-MDM2 interaction and alleviates RPL11-mediated suppression of MDM2 ubiquitin ligase activity toward p53. These effects are mediated by the N-terminal region of GRWD1, including the acidic domain. Finally, we show that GRWD1 overexpression in combination with HPV16 E7 and activated KRAS confers anchorage-independent growth and tumorigenic capacity on normal human fibroblasts. Consistent with this, GRWD1 overexpression is associated with poor prognosis in cancer patients. Taken together, our results suggest that GRWD1 is a novel negative regulator of p53 and a potential oncogene.


Subject(s)
Carrier Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-mdm2/metabolism , Ribosomal Proteins/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Animals , Carrier Proteins/chemistry , Cell Line, Tumor , Cell Transformation, Viral , Disease Models, Animal , Female , Gene Expression , Gene Silencing , Genes, ras , Heterografts , Humans , Mice , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/mortality , Neoplasms/pathology , Prognosis , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Stress, Physiological , Tumor Suppressor Protein p53/metabolism
4.
Sci Rep ; 6: 31758, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27545311

ABSTRACT

Cellular senescence is defined as permanent cell cycle arrest induced by various stresses. Although the p53 transcriptional activity is essential for senescence induction, the downstream genes that are crucial for senescence remain unsolved. Here, by using a developed experimental system in which cellular senescence or apoptosis is induced preferentially by altering concentration of etoposide, a DNA-damaging drug, we compared gene expression profiles of senescent and apoptotic cells by microarray analysis. Subtraction of the expression profile of apoptotic cells identified 20 genes upregulated specifically in senescent cells. Furthermore, 6 out of 20 genes showed p53-dependent upregulation by comparing gene expression between p53-proficient and -deficient cells. These 6 genes were also upregulated during replicative senescence of normal human diploid fibroblasts, suggesting that upregulation of these genes is a general phenomenon in senescence. Among these genes, 2 genes (PRODH and DAO) were found to be directly regulated by p53, and ectopic expression of 4 genes (PRODH, DAO, EPN3, and GPR172B) affected senescence phenotypes induced by etoposide treatment. Collectively, our results identified several proteins as novel downstream effectors of p53-mediated senescence and provided new clues for further research on the complex signalling networks underlying the induction and maintenance of senescence.


Subject(s)
Cellular Senescence/drug effects , Etoposide/pharmacology , Gene Expression Profiling/methods , Transcriptome/drug effects , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Line , Cell Line, Tumor , Cellular Senescence/genetics , Hep G2 Cells , Humans , Immunoblotting , RNA Interference , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
5.
Bioorg Med Chem ; 24(8): 1919-26, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27010502

ABSTRACT

Although several p53-Mdm2-binding disruptors have been identified to date, few studies have been published on p53-Mdmx-interaction inhibitors. In the present study, we demonstrated that o-aminothiophenol derivatives with molecular weights of 200-300 selectively inhibited the p53-Mdmx interaction. S-2-Isobutyramidophenyl 2-methylpropanethioate (K-178) (1c) activated p53, up-regulated the expression of its downstream genes such as p21 and Mdm2, and preferentially inhibited the growth of cancer cells with wild-type p53 over those with mutant p53. Furthermore, we found that the S-isobutyryl-deprotected forms 1b and 3b of 1c and S-2-benzamidophenyl 2-methylpropanethioate (K-181) (3c) preferentially inhibited the p53-Mdmx interaction over the p53-Mdm2 interaction, respectively, by using a Flag-p53 and glutathione S-transferase (GST)-fused protein complex (Mdm2, Mdmx, DAPK1, or PPID). In addition, the interaction of p53 with Mdmx was lost by replacing a sulfur atom with an oxygen atom in 1b and 1c. These results suggest that sulfides such as 1b, 3b, 4b, and 5b interfere with the binding of p53-Mdmx, resulting in the dissociation of the two proteins. Furthermore, the results of oral administration experiments using xenografts in nude mice indicated that 1c reduced the volume of tumor masses to 49.0% and 36.6% that of the control at 100 mg/kg and 150 mg/kg, respectively, in 40 days.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Drug Discovery , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Administration, Oral , Aniline Compounds/administration & dosage , Aniline Compounds/chemistry , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Molecular Weight , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Protein Binding/drug effects , Structure-Activity Relationship , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
6.
Cancer Res ; 76(4): 974-83, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26837769

ABSTRACT

The CD74-Neuregulin1 (NRG1) fusion gene was recently identified as novel driver of invasive mucinous adenocarcinoma, a malignant form of lung cancer. However, the function of the CD74-NRG1 fusion gene in adenocarcinoma pathogenesis and the mechanisms by which it may impart protumorigenic characteristics to cancer stem cells (CSC) is still unclear. In this study, we found that the expression of the CD74-NRG1 fusion gene increased the population of lung cancer cells with CSC-like properties. CD74-NRG1 expression facilitated sphere formation not only of cancer cells, but also of nonmalignant lung epithelial cells. Using a limiting dilution assay in a xenograft model, we further show that the CD74-NRG1 fusion gene enhanced tumor initiation. Mechanistically, we found that CD74-NRG1 expression promoted the phosphorylation of ErbB2/3 and activated the PI3K/Akt/NF-κB signaling pathway. Furthermore, the expression of the secreted insulin-like growth factor 2 (IGF2) and phosphorylation of its receptor, IGF1R, were enhanced in an NF-κB-dependent manner in cells expressing CD74-NRG1. These findings suggest that CD74-NRG1-induced NF-κB activity promotes the IGF2 autocrine/paracrine circuit. Moreover, inhibition of ErbB2, PI3K, NF-κB, or IGF2 suppressed CD74-NRG1-induced tumor sphere formation. Therefore, our study provides a preclinical rationale for developing treatment approaches based on these identified pathways to suppress CSC properties that promote tumor progression and recurrence.


Subject(s)
Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Neuregulin-1/genetics , Neuregulin-1/metabolism , Cell Line, Tumor , Cell Proliferation , Humans , Lung Neoplasms/genetics , Signal Transduction
8.
Mol Endocrinol ; 29(5): 739-55, 2015 May.
Article in English | MEDLINE | ID: mdl-25860340

ABSTRACT

17ß-estradiol (E2)-induced signaling and control of estrogen receptor (ER)α degradation both play a major role in breast cancer cell proliferation. We recently reported the involvement of lysosomal function in both E2-dependent ERα breakdown and E2-induced cell proliferation and thus hypothesized a role for endocytic proteins in ERα signaling. An small interfering RNA screen identified proteins that regulate intracellular endocytic traffic and whose silencing alters E2-induced ERα degradation. One such protein was the clathrin heavy chain (CHC), whose role in E2:ERα signaling to cell proliferation is unknown. Here, we show that CHC physically interacts with ERα in the cytoplasm of breast cancer cells and regulates E2-induced cell proliferation. Surprisingly, the CHC:ERα interaction is required to sustain E2 signaling but is dispensable for ERα degradation. Our data also demonstrate that many membrane trafficking proteins contribute to the regulation of ERα degradation, thus unraveling the contribution of endocytic proteins in E2:ERα signaling.


Subject(s)
Clathrin Heavy Chains/metabolism , Estradiol/physiology , Estrogen Receptor alpha/metabolism , Cell Proliferation , Endocytosis , Humans , MCF-7 Cells , Palmitic Acid/metabolism , Protein Binding , Protein Interaction Mapping , Protein Processing, Post-Translational , Proteolysis , Receptor, IGF Type 1/metabolism , Signal Transduction
9.
Proc Natl Acad Sci U S A ; 111(52): 18691-6, 2014 Dec 30.
Article in English | MEDLINE | ID: mdl-25512506

ABSTRACT

Communication between cancer cells and their microenvironment controls cancer progression. Although the tumor suppressor p53 functions in a cell-autonomous manner, it has also recently been shown to function in a non-cell-autonomous fashion. Although functional defects have been reported in p53 in stromal cells surrounding cancer, including mutations in the p53 gene and decreased p53 expression, the role of p53 in stromal cells during cancer progression remains unclear. We herein show that the expression of α-smooth muscle actin (α-SMA), a marker of cancer-associated fibroblasts (CAFs), was increased by the ablation of p53 in lung fibroblasts. CAFs enhanced the invasion and proliferation of lung cancer cells when cocultured with p53-depleted fibroblasts and required contact between cancer and stromal cells. A comprehensive analysis using a DNA chip revealed that tetraspanin 12 (TSPAN12), which belongs to the tetraspanin protein family, was derepressed by p53 knockdown. TSPAN12 knockdown in p53-depleted fibroblasts inhibited cancer cell proliferation and invasion elicited by coculturing with p53-depleted fibroblasts in vitro, and inhibited tumor growth in vivo. It also decreased CXC chemokine ligand 6 (CXCL6) secretion through the ß-catenin signaling pathway, suggesting that cancer cell contact with TSPAN12 in fibroblasts transduced ß-catenin signaling into fibroblasts, leading to the secretion of CXCL6 to efficiently promote invasion. These results suggest that stroma-derived p53 plays a pivotal role in epithelial cancer progression and that TSPAN12 and CXCL6 are potential targets for lung cancer therapy.


Subject(s)
Fibroblasts/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Signal Transduction , Tetraspanins/metabolism , Animals , Cell Line, Tumor , Chemokine CXCL6/genetics , Chemokine CXCL6/metabolism , Fibroblasts/pathology , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/pathology , Tetraspanins/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , beta Catenin/genetics , beta Catenin/metabolism
10.
Clin Cancer Res ; 20(12): 3087-93, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24727320

ABSTRACT

PURPOSE: To identify druggable oncogenic fusions in invasive mucinous adenocarcinoma (IMA) of the lung, a malignant type of lung adenocarcinoma in which KRAS mutations frequently occur. EXPERIMENTAL DESIGN: From an IMA cohort of 90 cases, consisting of 56 cases (62%) with KRAS mutations and 34 cases without (38%), we conducted whole-transcriptome sequencing of 32 IMAs, including 27 cases without KRAS mutations. We used the sequencing data to identify gene fusions, and then performed functional analyses of the fusion gene products. RESULTS: We identified oncogenic fusions that occurred mutually exclusively with KRAS mutations: CD74-NRG1, SLC3A2-NRG1, EZR-ERBB4, TRIM24-BRAF, and KIAA1468-RET. NRG1 fusions were present in 17.6% (6/34) of KRAS-negative IMAs. The CD74-NRG1 fusion activated HER2:HER3 signaling, whereas the EZR-ERBB4 and TRIM24-BRAF fusions constitutively activated the ERBB4 and BRAF kinases, respectively. Signaling pathway activation and fusion-induced anchorage-independent growth/tumorigenicity of NIH3T3 cells expressing these fusions were suppressed by tyrosine kinase inhibitors approved for clinical use. CONCLUSIONS: Oncogenic fusions act as driver mutations in IMAs without KRAS mutations, and thus represent promising therapeutic targets for the treatment of such IMAs.


Subject(s)
Adenocarcinoma, Mucinous/drug therapy , Lung Neoplasms/drug therapy , Mutation/genetics , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/pathology , Aged , Animals , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Female , Follow-Up Studies , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Middle Aged , NIH 3T3 Cells , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Proto-Oncogene Proteins p21(ras) , Signal Transduction/drug effects
11.
Cell Rep ; 7(2): 527-538, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24726368

ABSTRACT

In lung cancer progression, p53 mutations are more often observed in invasive tumors than in noninvasive tumors, suggesting that p53 is involved in tumor invasion and metastasis. To understand the nature of p53 function as a tumor suppressor, it is crucial to elucidate the detailed mechanism of the alteration in epithelial cells that follow oncogenic KRAS activation and p53 inactivation. Here, we report that KRAS activation induces epithelial-mesenchymal transition and that p53 inactivation is required for cell motility and invasiveness. Furthermore, TSPAN2, a transmembrane protein, is responsible for cell motility and invasiveness elicited by p53 inactivation. TSPAN2 is highly expressed in p53-mutated lung cancer cells, and high expression of TSPAN2 is associated with the poor prognosis of lung adenocarinomas. TSPAN2 knockdown suppresses metastasis to the lungs and liver, enabling prolonged survival. TSPAN2 enhances cell motility and invasiveness by assisting CD44 in scavenging intracellular reactive oxygen species.


Subject(s)
Cell Movement , Lung Neoplasms/metabolism , Nerve Tissue Proteins/metabolism , Tetraspanins/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Lung Neoplasms/pathology , Mice, Nude , Mutation , Neoplasm Invasiveness , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , Tetraspanins/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , ras Proteins/genetics , ras Proteins/metabolism
12.
Mol Cell Biol ; 33(12): 2447-57, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23589328

ABSTRACT

The p53 tumor suppressor protein is a transcription factor controlling various outcomes, such as growth arrest and apoptosis, through the regulation of different sets of target genes. The nuclear mitotic apparatus protein (NuMA) plays important roles in spindle pole organization during mitosis and in chromatin regulation in the nucleus during interphase. Although NuMA has been shown to colocalize with several nuclear proteins, including high-mobility-group proteins I and Y and GAS41, the role of NuMA during interphase remains unclear. Here we report that NuMA binds to p53 to modulate p53-mediated transcription. Acute and partial ablation of NuMA attenuates the induction of the proarrested p21 gene following DNA damage, subsequently causing impaired cell cycle arrest. Interestingly, NuMA knockdown had little effect on the induction of the p53-dependent proapoptotic PUMA gene. Furthermore, NuMA is required for the recruitment of cyclin-dependent kinase 8 (Cdk8), a component of the Mediator complex and a promoter of p53-mediated p21 gene function. These data demonstrate that NuMA is critical for the target selectivity of p53-mediated transcription.


Subject(s)
Antigens, Nuclear/metabolism , Cyclin-Dependent Kinase 8/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Antigens, Nuclear/genetics , Breast Neoplasms/metabolism , Cell Cycle Checkpoints , Cell Cycle Proteins , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , Female , Fibrosarcoma/metabolism , Humans , Lung Neoplasms/metabolism , Nuclear Matrix-Associated Proteins/genetics , Protein Binding , RNA Interference , RNA, Small Interfering , Transcription, Genetic
13.
Nat Med ; 18(3): 375-7, 2012 Feb 12.
Article in English | MEDLINE | ID: mdl-22327624

ABSTRACT

We identified in-frame fusion transcripts of KIF5B (the kinesin family 5B gene) and the RET oncogene, which are present in 1-2% of lung adenocarcinomas (LADCs) from people from Japan and the United States, using whole-transcriptome sequencing. The KIF5B-RET fusion leads to aberrant activation of RET kinase and is considered to be a new driver mutation of LADC because it segregates from mutations or fusions in EGFR, KRAS, HER2 and ALK, and a RET tyrosine kinase inhibitor, vandetanib, suppresses the fusion-induced anchorage-independent growth activity of NIH3T3 cells.


Subject(s)
Adenocarcinoma/genetics , Kinesins/genetics , Lung Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins c-ret/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Anaplastic Lymphoma Kinase , Animals , Cell Transformation, Neoplastic/drug effects , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing , Humans , Japan , Lung Neoplasms/pathology , Mice , NIH 3T3 Cells , Norway , Piperidines/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras) , Quinazolines/pharmacology , Receptor Protein-Tyrosine Kinases/genetics , Receptor, ErbB-2/genetics , United States , ras Proteins/genetics
14.
Cancer Sci ; 103(4): 782-90, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22320325

ABSTRACT

The development of oral squamous cell carcinoma (OSCC) is a multistep process that requires the accumulation of genetic alterations. To identify genes responsible for OSCC development, we performed high-density single nucleotide polymorphism array analysis and genome-wide gene expression profiling on OSCC tumors. These analyses indicated that the absent in melanoma 2 (AIM2) gene and the interferon-inducible gene 16 (IFI16) mapped to the hematopoietic interferon-inducible nuclear proteins. The 200-amino-acid repeat gene cluster in the amplified region of chromosome 1q23 is overexpressed in OSCC. Both AIM2 and IFI16 are cytoplasmic double-stranded DNA sensors for innate immunity and act as tumor suppressors in several human cancers. Knockdown of AIM2 or IFI16 in OSCC cells results in the suppression of cell growth and apoptosis, accompanied by the downregulation of nuclear factor kappa-light-chain-enhancer of activated B cells activation. Because all OSCC cell lines have reduced p53 activity, wild-type p53 was introduced in p53-deficient OSCC cells. The expression of wild-type p53 suppressed cell growth and induced apoptosis via suppression of nuclear factor kappa-light-chain-enhancer of activated B cells activity. Finally, the co-expression of AIM2 and IFI16 significantly enhanced cell growth in p53-deficient cells; in contrast, the expression of AIM2 and/or IFI16 in cells bearing wild-type p53 suppressed cell growth. Moreover, AIM2 and IFI16 synergistically enhanced nuclear factor kappa-light-chain-enhancer of activated B cells signaling in p53-deficient cells. Thus, expression of AIM2 and IFI16 may have oncogenic activities in the OSCC cells that have inactivated the p53 system.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Genes, p53 , Mouth Neoplasms/metabolism , Nuclear Proteins/genetics , Phosphoproteins/genetics , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Chromosomes, Human, Pair 1 , DNA-Binding Proteins , Gene Amplification , Humans , NF-kappa B/metabolism , Proto-Oncogene Proteins c-ets/genetics , Transcription Factor AP-1/genetics
15.
Cancer Sci ; 101(8): 1891-6, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20557307

ABSTRACT

Human papillomavirus (HPV) infection is a causative event for the development of uterine cervical carcinoma. Human papillomavirus (HPV) 16, 18, and 33 DNA has been also detected frequently in lung adenocarcinomas (AdCs) in East Asian countries; however, its prevalence in Japan remains unclear. We therefore screened for HPV 16/18/33 DNA in 297 lung AdCs in a Japanese population by multiplex PCR with type-specific primers. As reported previously, HPV 16 DNA was detected in two cervical cancer cell lines, CaSki and SiHa, while HPV 18 DNA was detected in HeLa cells, and 0.1-1.0 copies of HPV-DNA per cell were detectable by this method. However, with this method, none of the 297 lung AdCs showed positive signals for HPV 16/18/33 DNA, indicating that HPV-DNA is not or is very rarely integrated in lung AdC genomes in the Japanese. Furthermore, none of the lung AdCs showed positive signals by nested PCR with HPV 16/18 type-specific primers. Therefore, we further attempted to detect HPV 16/18/33 DNA in 91 lung cancer cell lines, including 40 AdC cell lines. Among them, 30 have been established in Japan and the remaining 61 in the USA. No HPV signals were obtained in any of the 91 cell lines by either multiplex or nested PCR, while the p53 gene was mutated in 81 of them including 35 of the 40 AdC cell lines. These results indicate that HPV 16/18/33 infection does not play a major role in the development of lung AdC in Japan nor in the USA.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/virology , Human papillomavirus 16/isolation & purification , Human papillomavirus 18/isolation & purification , Lung Neoplasms/genetics , Lung Neoplasms/virology , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , DNA, Viral/analysis , Female , Genes, p53 , Humans , Male , Middle Aged , Mutation , Polymerase Chain Reaction
16.
Mol Cell Biol ; 30(7): 1620-33, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20123963

ABSTRACT

p53 phosphorylation at Ser46 following DNA damage is important for preferential transactivation of proapoptotic genes. Here, we report that ataxia-telangiectasia mutated (ATM) kinase is responsible for Ser46 phosphorylation of p53 during early-phase response to DNA damage. To elucidate the direct phosphorylation of p53 at Ser46 by ATM, an ATM mutant (ATM-AS) sensitive to ATP analogues was engineered. In vitro kinase assays revealed that p53 was phosphorylated at Ser46 by ATM-AS, even when ATP analogues were used as phosphate donors, although this phosphorylation site is not in an SQ motif, a consensus ATM site. Furthermore, Ser46 phosphorylation by ATM was dependent on the N- and C-terminal domains of p53, unlike Ser15 phosphorylation. Immunofluorescence analyses showed that Ser46-phosphorylated p53 was observed as foci in response to DNA damage and colocalized with gamma-H2AX or Ser1981-phosphorylated ATM. These results suggest that ATM phosphorylates a noncanonical serine residue on p53 by mechanisms different from those for the phosphorylation of Ser15.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Glutamine/genetics , Protein Serine-Threonine Kinases/metabolism , Serine , Threonine/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Adenosine Triphosphate/analogs & derivatives , Animals , Ataxia Telangiectasia Mutated Proteins , Base Sequence , Cell Cycle Proteins/genetics , Cell Line , DNA/genetics , DNA/metabolism , DNA/radiation effects , DNA Damage , DNA-Binding Proteins/genetics , Glutamine/metabolism , Humans , Molecular Structure , Phosphorylation , Protein Serine-Threonine Kinases/genetics , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine/genetics , Serine/metabolism , Threonine/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics
17.
J Mol Biol ; 394(3): 460-71, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19766654

ABSTRACT

The p53 pathway is activated in response to various cellular stresses to protect cells from malignant transformation. We have previously shown that clathrin heavy chain (CHC), which is a cytosolic protein regulating endocytosis, is present in nuclei and binds to p53 to promote p53-mediated transcription. However, details of the binding interface between p53 and CHC remain unclear. Here, we report on the binding mode between p53 and CHC using mutation analyses and a structural model of the interaction generated by molecular dynamics. Structural modeling analyses predict that an Asn1288 residue in CHC is crucial for binding to p53. In fact, substitution of this Asn to Ala of CHC diminished its ability to interact with p53, leading to reduced activity to transactivate p53. Surprisingly, this mutation had little effect on receptor-mediated endocytosis. Thus, the function-specific mutation of CHC will clarify physiological roles of CHC in the regulation of the p53 pathway.


Subject(s)
Clathrin Heavy Chains/chemistry , Clathrin Heavy Chains/genetics , Transcriptional Activation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , 3' Untranslated Regions , Amino Acid Sequence , Amino Acid Substitution , Asparagine/chemistry , Base Sequence , Binding Sites/genetics , Cell Line , Clathrin Heavy Chains/metabolism , Conserved Sequence , Endocytosis , HeLa Cells , Humans , In Vitro Techniques , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes , Mutagenesis, Site-Directed , Protein Interaction Domains and Motifs , RNA Interference , RNA, Small Interfering/genetics , Receptors, Transferrin/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Thermodynamics , Tumor Suppressor Protein p53/chemistry
18.
Genes Cells ; 13(4): 375-86, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18363968

ABSTRACT

The p53 gene encodes a multi-functional protein to prevent tumorigenesis. Although there have been many reports of the nuclear functions of p53, little is known about the cytosolic functions of p53. Here, we found that p53 is present in cytosol as well as nuclei under unstressed conditions and binds to clathrin heavy chain (CHC). CHC is known to play a role in receptor-mediated endocytosis. Based on our findings, we examined the effect of p53 on clathrin-mediated endocytosis of epidermal growth factor receptor (EGFR). Surprisingly, p53 co-localized with CHC at the plasma membrane in response to EGF stimulation. In cells with ablated p53 expression by RNAi, EGFR internalization was delayed and intracellular signaling from EGFR was altered. Thus, our findings provide evidence that cytosolic p53 may participate in the regulation of clathrin-mediated endocytosis to control the correct signaling from EGFR.


Subject(s)
Clathrin Heavy Chains/metabolism , Endocytosis/physiology , Tumor Suppressor Protein p53/metabolism , Base Sequence , Cell Line , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Clathrin Heavy Chains/antagonists & inhibitors , Clathrin Heavy Chains/genetics , Cytosol/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Genes, p53 , Humans , Mutation , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction , Tumor Suppressor Protein p53/antagonists & inhibitors
19.
Genes Dev ; 20(9): 1087-99, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16618797

ABSTRACT

The p53 protein is a transcription factor that activates various genes responsible for growth arrest and/or apoptosis in response to DNA damage. Here, we report that clathrin heavy chain (CHC) binds to p53 and contributes to p53-mediated transcription. CHC is known to be a cytosolic protein that functions as a vesicle transporter. We found, however, that CHC exists not only in cytosol but also in nuclei. CHC expression enhances p53-dependent transactivation, whereas the reduction of CHC expression by RNA interference (RNAi) attenuates its transcriptional activity. Moreover, CHC binds to the p53-responsive promoter in vivo and stabilizes p53-p300 interaction to promote p53-mediated transcription. Thus, nuclear CHC is required for the transactivation of p53 target genes and plays a distinct role from clathrin-mediated endocytosis.


Subject(s)
Clathrin Heavy Chains/physiology , Transcriptional Activation , Tumor Suppressor Protein p53/physiology , Cell Line, Tumor , Cell Nucleus/metabolism , Clathrin Heavy Chains/genetics , Cytoplasm/metabolism , Endocytosis , Genes, p53 , Humans , Mutation , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/genetics , Tumor Suppressor Protein p53/genetics , p300-CBP Transcription Factors/metabolism
20.
Acta Neurobiol Exp (Wars) ; 62(3): 153-66, 2002.
Article in English | MEDLINE | ID: mdl-12416393

ABSTRACT

The "protein only" hypothesis holds that the infectious agent causing transmissible spongiform encephalopathies is a conformational isomer of PrP, a host protein predominantly expressed in brain and is strongly supported by many lines of evidence. Prion diseases are so far unique among conformational diseases in that they are transmissible, not only experimentally but also by natural routes, mainly by ingestion. The pathway of prions to the brain has been elucidated in outline. A striking feature of prions is their extraordinary resistance to conventional sterilisation procedures, and their capacity to bind to surfaces of metal and plastic without losing infectivity. This property, first observed in a clinical setting, is now being investigated in experimental settings, both in animals and in cell culture.


Subject(s)
Prion Diseases/genetics , Prions/genetics , Prions/physiology , Animals , Humans , Iatrogenic Disease , Prion Diseases/transmission
SELECTION OF CITATIONS
SEARCH DETAIL