Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 492
Filter
1.
FASEB J ; 32(10): 5647-5660, 2018 10.
Article in English | MEDLINE | ID: mdl-29750576

ABSTRACT

Melanoma is among the most aggressive and treatment-resistant human cancers. Aberrant histone H3 methylation at Lys 9 (H3K9) correlates with carcinogenic gene silencing, but the significance of suppressor of variegation 3-9 homolog 1 (SUV39H1), an H3K9-specific methyltransferase, in melanoma initiation and progression remains unclear. Here, we show that SUV39H1-mediated H3K9 trimethylation facilitates retinoblastoma ( RB) 1 promoter CpG island methylation by interacting with DNA methyltransferase 3A and decreasing RB mRNA and protein in melanoma cells. Reduced RB abundance, in turn, impairs E2F1 transcriptional inhibition, leading to increased peptidyl-prolyl cis-trans isomerase never-in-mitosis A (NIMA)-interacting 1 (PIN1) levels, human keratinocyte neoplastic cell transformation, and melanoma tumorigenesis via enhanced rapidly accelerated fibrosarcoma 1(RAF1)-MEK-ERK signaling pathway activation. In a synergistic model with B16-F1 murine melanoma cells, SUV39H1 and PIN1 overexpression increased melanoma growth, which was abrogated by their inhibition in SUV39H1-overexpressing B16-F1 mice. SUV39H1 also positively correlated with PIN1 expression in human melanoma. Our studies establish SUV39H1 as an oncogene in melanoma and underscore the role of chromatin factors in regulating tumorigenesis.-Kim, G., Kim, J.-Y., Lim, S.-C., Lee, K. Y., Kim, O., Choi, H. S. SUV39H1/DNMT3A-dependent methylation of the RB1 promoter stimulates PIN1 expression and melanoma development.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA, Neoplasm/metabolism , Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Methyltransferases/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/biosynthesis , Repressor Proteins/metabolism , Retinoblastoma Protein/biosynthesis , Animals , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , DNA, Neoplasm/genetics , HEK293 Cells , Humans , Melanoma/genetics , Melanoma/pathology , Methylation , Methyltransferases/genetics , Mice , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Repressor Proteins/genetics , Retinoblastoma Protein/genetics
2.
J Cutan Pathol ; 46(9): 659-664, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31012122

ABSTRACT

BACKGROUND: Porocarcinomas are rare aggressive carcinomas that harbor tumor suppressor mutations and must be distinguished from benign entities such as poromas. METHODS: To determine whether altered expression of these genes was diagnostically informative, we examined p53, Rb, and p16 staining patterns in 15 poromas and 16 porocarcinomas. RESULTS: Poromas consistently displayed diffuse strong expression of Rb in all but one case that displayed focal loss (1/15, 7%), and no evidence of aberrancy in p53 or p16. Porocarcinomas displayed diffuse or focal loss of Rb expression in 9/16 (56%) cases, diffuse loss or overexpression of p53 in 8/15 (53%), and diffuse loss or overexpression of p16 in 6/14 (43%). Diffuse aberrancy in p53 and Rb expression correlated with tumor mutations in TP53 and RB1, respectively, whereas focal Rb loss was associated with wild type RB1. Diffuse p16 overexpression correlated with Rb loss rather than CDKN2A mutation. For porocarcinomas with all three stains evaluable, 10/13 (77%) displayed aberrancy in at least one marker. CONCLUSIONS: Our findings suggest that altered p53, p16, and/or Rb expression is relatively specific to porocarcinoma in comparison with poroma. Technical limitations of this panel, including possible focal Rb loss, must be kept in mind, especially in limited samples.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Eccrine Porocarcinoma , Gene Expression Regulation, Neoplastic , Poroma , Retinoblastoma Protein/biosynthesis , Sweat Gland Neoplasms , Tumor Suppressor Protein p53/biosynthesis , Adult , Aged , Aged, 80 and over , Eccrine Porocarcinoma/metabolism , Eccrine Porocarcinoma/pathology , Female , Humans , Male , Middle Aged , Poroma/metabolism , Poroma/pathology , Sweat Gland Neoplasms/metabolism , Sweat Gland Neoplasms/pathology
3.
PLoS Genet ; 11(7): e1005355, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26176933

ABSTRACT

The origin of most ovarian tumors is undefined. Here, we report development of a novel mouse model in which conditional inactivation of the tumor suppressor gene Rb1 in oocytes leads to the formation of ovarian teratomas (OTs). While parthenogenetically activated ooctyes are a known source of OT in some mutant mouse models, enhanced parthenogenetic propensity in vitro was not observed for Rb1-deficient oocytes. Further analyses revealed that follicle recruitment and growth is disrupted in ovaries of mice with conditional inactivation of Rb1, leading to abnormal accumulation of secondary/preantral follicles. These findings underpin the concept that miscues between the germ cell and somatic compartments cause premature oocyte activation and the formation of OTs. Furthermore, these results suggest that defects in folliculogenesis and a permissive genetic background are sufficient to drive OT development, even in the absence of enhanced parthenogenetic activation. Thus, we have discovered a novel role of Rb1 in regulating the entry of primordial oocytes into the pool of growing follicles and signaling between the oocyte and granulosa cells during the protracted process of oocyte growth. Our findings, coupled with data from studies of other OT models, suggest that defects in the coordinated regulation between growth of the oocyte and somatic components in follicles are an underlying cause of OT formation.


Subject(s)
Oocytes/growth & development , Oogenesis , Ovarian Follicle/growth & development , Retinoblastoma Protein/genetics , Animals , Disease Models, Animal , Female , Humans , Meiosis/genetics , Mice , Oocytes/pathology , Ovarian Follicle/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Retinoblastoma Protein/biosynthesis , Signal Transduction , Teratoma/genetics , Teratoma/pathology
4.
J Virol ; 89(1): 799-810, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25355877

ABSTRACT

UNLABELLED: Although monocytes and macrophages are targets of HIV-1-mediated immunopathology, the impact of high viremia on activation-induced monocyte apoptosis relative to monocyte and macrophage activation changes remains undetermined. In this study, we determined constitutive and oxidative stress-induced monocyte apoptosis in uninfected and HIV(+) individuals across a spectrum of viral loads (n = 35; range, 2,243 to 1,355,998 HIV-1 RNA copies/ml) and CD4 counts (range, 26 to 801 cells/mm(3)). Both constitutive apoptosis and oxidative stress-induced apoptosis were positively associated with viral load and negatively associated with CD4, with an elevation in apoptosis occurring in patients with more than 40,000 (4.6 log) copies/ml. As expected, expression of Rb1 and interferon-stimulated genes (ISGs), plasma soluble CD163 (sCD163) concentration, and the proportion of CD14(++) CD16(+) intermediate monocytes were elevated in viremic patients compared to those in uninfected controls. Although CD14(++) CD16(+) frequencies, sCD14, sCD163, and most ISG expression were not directly associated with a change in apoptosis, sCD14 and ISG expression showed an association with increasing viral load. Multivariable analysis of clinical values and monocyte gene expression identified changes in IFI27, IFITM2, Rb1, and Bcl2 expression as determinants of constitutive apoptosis (P = 3.77 × 10(-5); adjusted R(2) = 0.5983), while changes in viral load, IFITM2, Rb1, and Bax expression were determinants of oxidative stress-induced apoptosis (P = 5.59 × 10(-5); adjusted R(2) = 0.5996). Our data demonstrate differential activation states in monocytes between levels of viremia in association with differences in apoptosis that may contribute to greater monocyte turnover with high viremia. IMPORTANCE: This study characterized differential monocyte activation, apoptosis, and apoptosis-related gene expression in low- versus high-level viremic HIV-1 patients, suggesting a shift in apoptosis regulation that may be associated with disease state. Using single and multivariable analysis of monocyte activation parameters and gene expression, we supported the hypothesis that monocyte apoptosis in HIV disease is a reflection of viremia and activation state with contributions from gene expression changes within the ISG and Bcl2 gene families. Understanding monocyte apoptosis response may inform HIV immunopathogenesis, retention of infected macrophages, and monocyte turnover in low- or high-viral-load states.


Subject(s)
Apoptosis , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , Monocytes/immunology , Viral Load , Adult , Aged, 80 and over , Chronic Disease , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Monocytes/physiology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Retinoblastoma Protein/biosynthesis , Young Adult
5.
Tumour Biol ; 37(4): 4831-40, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26520443

ABSTRACT

miR-449a, a novel tumor suppressor, is deregulated in various malignancies, including prostate cancer. Overexpression of miR-449a induces cell cycle arrest, apoptosis, and senescence, but its role in response to ionizing radiation and underlying molecular mechanism are still unknown. Here, we report that miR-449a enhances radiation-induced G2/M phase arrest and apoptosis through modulating pRb/E2F1 and sensitizes prostate cancer cells to X-ray radiation. In wild-type Rb PC-3 cells, overexpression of miR-449a enhances radiation-induced G2/M arrest and apoptosis and promotes the sensitivity to X-ray radiation. While mutant Rb DU-145 cells are resistant to the X-ray radiation despite in the presence of miR-449a. The cell cycle distribution of DU-145 cells is not significantly altered by miR-449a in the response to ionizing radiation. Furthermore, elevated miR-449a downregulates cell cycle regulator CDC25A and oncogene HDAC1. By targeting genes involved in controlling pRb/E2F1 activity, miR-449a regulates cell cycle progression and apoptosis and consequently enhances the radiosensitivity of PC-3 cells. Thus, miR-449a, as a miRNA component of the Rb pathway, promotes the radiosensitivity of PC-3 cells through regulating pRb/E2F1.


Subject(s)
E2F1 Transcription Factor/genetics , MicroRNAs/genetics , Prostatic Neoplasms/radiotherapy , Radiation Tolerance/genetics , Retinoblastoma Protein/genetics , Apoptosis/radiation effects , Cell Cycle Checkpoints/radiation effects , Cell Line, Tumor , Cell Proliferation/radiation effects , E2F1 Transcription Factor/biosynthesis , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Male , MicroRNAs/biosynthesis , Prostate/radiation effects , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Retinoblastoma Protein/biosynthesis , Signal Transduction/radiation effects , X-Rays
6.
Nucleic Acids Res ; 42(18): 11447-61, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25245946

ABSTRACT

ZNF509 is unique among POK family proteins in that four isoforms are generated by alternative splicing. Short ZNF509 (ZNF509S1, -S2 and -S3) isoforms contain one or two out of the seven zinc-fingers contained in long ZNF509 (ZNF509L). Here, we investigated the functions of ZNF509 isoforms in response to DNA damage, showing isoforms to be induced by p53. Intriguingly, to inhibit proliferation of HCT116 and HEK293 cells, we found that ZNF509L activates p21/CDKN1A transcription, while ZNF509S1 induces RB. ZNF509L binds to the p21/CDKN1A promoter either alone or by interacting with MIZ-1 to recruit the co-activator p300 to activate p21/CDKN1A transcription. In contrast, ZNF509S1 binds to the distal RB promoter to interact and interfere with the MIZF repressor, resulting in derepression and transcription of RB. Immunohistochemical analysis revealed that ZNF509 is highly expressed in normal epithelial cells, but was completely repressed in tumor tissues of the colon, lung and skin, indicating a possible role as a tumor suppressor.


Subject(s)
Cell Cycle Checkpoints , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA-Binding Proteins/metabolism , Retinoblastoma Protein/genetics , Transcription Factors/metabolism , Transcriptional Activation , Cell Line , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , DNA Damage , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , Kruppel-Like Transcription Factors/metabolism , Neoplasms/metabolism , Promoter Regions, Genetic , Protein Isoforms/biosynthesis , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Retinoblastoma Protein/biosynthesis , Stress, Physiological/genetics , Transcription Factors/biosynthesis , Transcription Factors/chemistry , Transcription Factors/genetics , Tumor Suppressor Protein p53/metabolism , Zinc Fingers , p300-CBP Transcription Factors/metabolism
7.
Carcinogenesis ; 36(6): 666-75, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25896445

ABSTRACT

Chronic inflammation is an important risk factor for lung cancer. Therefore, identification of chemopreventive agents that suppress inflammation-driven lung cancer is indispensable. We studied the efficacy of combinations of indole-3-carbinol (I3C) and silibinin (Sil), 20 µmol/g diet each, against mouse lung tumors induced by 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) and driven by lipopolysaccharide (LPS), a potent inflammatory agent and constituent of tobacco smoke. Mice treated with NNK + LPS developed 14.7±4.1 lung tumors/mouse, whereas mice treated with NNK + LPS and given combinations of I3C and Sil had 7.1±4.5 lung tumors/mouse, corresponding to a significant reduction of 52%. Moreover, the number of largest tumors (>1.0mm) was significantly reduced from 6.3±2.9 lung tumors/mouse in the control group to 1.0±1.3 and 1.6±1.8 lung tumors/mouse in mice given I3C + Sil and I3C alone, respectively. These results were paralleled by significant reductions in the level of proinflammatory and procarcinogenic proteins (pSTAT3, pIκBα and COX-2) and proteins that regulate cell proliferation (pAkt, cyclin D1, CDKs 2, 4, 6 and pRB). Further studies in premalignant bronchial cells showed that the antiproliferative effects of I3C + Sil were higher than the individual compounds and these effects were mediated by targeting cyclin D1, CDKs 2, 4 and 6 and pRB. I3C + Sil suppressed cyclin D1 by reducing its messenger RNA level and by enhancing its proteasomal degradation. Our results showed the potential lung cancer chemopreventive effects of I3C + Sil in smokers/former smokers with chronic pulmonary inflammatory conditions.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Indoles/pharmacology , Inflammation/drug therapy , Lung Neoplasms/drug therapy , Silymarin/pharmacology , Animals , Anticarcinogenic Agents/pharmacology , Cell Cycle Proteins/biosynthesis , Cell Line, Tumor , Cell Proliferation , Chemoprevention , Cyclin D1/biosynthesis , Cyclin D1/genetics , Cyclin-Dependent Kinases/biosynthesis , Cyclooxygenase 2/biosynthesis , Drug Combinations , Female , Humans , I-kappa B Proteins/biosynthesis , Interleukin-6/biosynthesis , Lipopolysaccharides , Lung/pathology , Mice , Mice, Inbred A , NF-KappaB Inhibitor alpha , Nitrosamines/adverse effects , Proto-Oncogene Proteins c-akt/biosynthesis , Random Allocation , Retinoblastoma Protein/biosynthesis , STAT3 Transcription Factor/biosynthesis , Silybin , Smoke/adverse effects , Tumor Necrosis Factor-alpha/biosynthesis
8.
J Biol Chem ; 289(7): 4116-25, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24371144

ABSTRACT

Ras mutations are frequent in cancer cells where they drive proliferation and resistance to apoptosis. However in primary cells, mutant Ras instead can cause oncogene-induced senescence, a tumor suppressor function linked to repression of the polycomb factor Bmi1, which normally regulates cell cycle inhibitory cyclin-dependent kinase inhibitors (cdki). It is unclear how Ras causes repression of Bmi1 in primary cells to suppress tumor formation while inducing the gene in cancer cells to drive tumor progression. Ras also induces the EMT transcription factor ZEB1 to trigger tumor invasion and metastasis. Beyond its well-documented role in EMT, ZEB1 is important for maintaining repression of cdki. Indeed, heterozygous mutation of ZEB1 is sufficient for elevated cdki expression, leading to premature senescence of primary cells. A similar phenotype is evident with Bmi1 mutation. We show that activation of Rb1 in response to mutant Ras causes dominant repression of ZEB1 in primary cells, but loss of the Rb1 pathway is a hallmark of cancer cells and in the absence of such Rb1 repression Ras induces ZEB1 in cancer cells. ZEB1 represses miR-200 in the context of a mutual repression loop. Because miR-200 represses Bmi1, induction of ZEB1 leads to induction of Bmi1. Rb1 pathway status then dictates the opposing effects of mutant Ras on the ZEB1-miR-200 loop in primary versus cancer cells. This loop not only triggers EMT, surprisingly we show it acts downstream of Ras to regulate Bmi1 expression and thus the critical decision between oncogene-induced senescence and tumor initiation.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , MicroRNAs/metabolism , Neoplasms/metabolism , Oncogene Protein p21(ras)/biosynthesis , Polycomb Repressive Complex 1/biosynthesis , Proto-Oncogene Proteins/biosynthesis , RNA, Neoplasm/metabolism , Retinoblastoma Protein/biosynthesis , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cellular Senescence/genetics , Homeodomain Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , MicroRNAs/genetics , Mutation , Neoplasms/genetics , Neoplasms/pathology , Oncogene Protein p21(ras)/genetics , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins/genetics , RNA, Neoplasm/genetics , Retinoblastoma Protein/genetics , Transcription Factors/genetics , Zinc Finger E-box-Binding Homeobox 1
9.
Int J Cancer ; 135(6): 1286-96, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24510775

ABSTRACT

Multidrug resistance (MDR) remains a major obstacle to effective chemotherapy treatment in ovarian cancer. In our study, paclitaxel-resistant ovarian cancer patients and cell lines had decreased miR-145 levels and expressed high levels of Sp1 and Cdk6. Introducing miR-145 into SKOV3/PTX and A2780/PTX cells led to a reduction in Cdk6 and Sp1 along with downregulation of P-gp and pRb. These changes resulted in increased accumulation of antineoplastic drugs and G1 cell cycle arrest, which rendered the cells more sensitive to paclitaxel in vitro and in vivo. These effects could be reversed by reintroducing Sp1 or Cdk6 into cells expressing high levels of miR-145, resulting in restoration of P-gp and pRb levels. Furthermore, we confirmed that both Cdk6 and Sp1 are targets of miR-145. Intriguingly, demethylation with 5-aza-dC led to reactivation of miR-145 expression in drug-resistant ovarian cancer cell lines, which also resulted in increased sensitivity to paclitaxel. Collectively, these findings begin to elucidate the role of miR-145 as an important regulator of chemoresistance in ovarian cancer by controlling both Cdk6 and Sp1.


Subject(s)
Cyclin-Dependent Kinase 6/metabolism , MicroRNAs/administration & dosage , MicroRNAs/metabolism , Ovarian Neoplasms/therapy , Paclitaxel/pharmacology , Sp1 Transcription Factor/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase 6/biosynthesis , Cyclin-Dependent Kinase 6/genetics , Drug Resistance, Neoplasm , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Paclitaxel/pharmacokinetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Sp1 Transcription Factor/biosynthesis , Sp1 Transcription Factor/genetics , Transfection , Xenograft Model Antitumor Assays
10.
Cancer Sci ; 105(5): 506-11, 2014 May.
Article in English | MEDLINE | ID: mdl-24628854

ABSTRACT

Our previous study implied a correlation between inhibitors of differentiation-1 (Id-1) and cervical cancer development. However, how Id-1 contributes to cervical carcinogenesis is unknown. In the present study, we used an in vitro transformation model to investigate the role of Id-1 in the transformation of cervical cells. Human papillomavirus (HPV)-immortalized cervical epithelial cells (H8) were successfully transformed by exposure to the carcinogen N-nitrosopyrrolidine (NPYR). The expression of both Id-1 RNA and protein was significantly increased in transformed H8 cells, suggesting a possible role of Id-1 in cervical cell transformation. Ectopic expression of Id-1 in H8 cells potentiated NPYR-induced cell transformation. In contrast, silencing of Id-1 suppressed NPYR-induced H8 cell transformation. In addition, the expression of HPV E6 and E7 oncoproteins was upregulated while that of the tumor suppressors p53 and pRb was suppressed after H8 cell transformation. Our results suggest that Id-1 plays an oncogenic role in HPV-related cervical carcinogenesis, which sheds light on cervical cancer development mechanisms and implies that Id-1 is a potential target for cervical cancer prevention and therapy.


Subject(s)
Cell Transformation, Neoplastic/pathology , Human papillomavirus 16 , Inhibitor of Differentiation Protein 1/metabolism , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Repressor Proteins/metabolism , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/virology , Animals , Carcinogens/toxicity , Cell Differentiation , Cell Line , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Epithelial Cells/drug effects , Epithelial Cells/virology , Female , Humans , Inhibitor of Differentiation Protein 1/genetics , Mice , Mice, Nude , N-Nitrosopyrrolidine/toxicity , Neoplasm Transplantation , Oncogene Proteins, Viral/biosynthesis , Papillomavirus E7 Proteins/biosynthesis , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering , Repressor Proteins/biosynthesis , Retinoblastoma Protein/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , Up-Regulation , Uterine Cervical Neoplasms/chemically induced
11.
Mod Pathol ; 27(2): 204-13, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23887295

ABSTRACT

Triple-negative breast carcinomas represent a tumor group of pivotal clinical importance given the lack of target therapies. The prognostic significance of triple-negative breast carcinomas remains unclear because of their histological and molecular heterogeneity. Currently, neither prognostic nor predictive factors are available for these tumors. Retinoblastoma (Rb) pathway loss has been linked to clinical outcome in various cancer types, including breast cancer. We investigated the association between Rb and p16 protein expression and clinical outcome in no-special-type triple-negative breast carcinomas. Immunohistochemical staining for Rb, p16, p53 and CK5 was carried out on a section from archival specimens of 117 no-special-type triple-negative breast carcinomas. Immunopositive p16 (p16+) and immunonegative Rb (Rb-) staining were seen in 49.5% and in 24.8% of tumors, respectively. There was an inverse correlation between p16+ and Rb- (P<0.001). P16+ was correlated with G3 grade (P<0.001), high Ki-67 (P=0.03), p53 overexpression (P<0.001) and CK5 immunopositivity (P=0.01). Rb- was not associated with any clinicopathologic variable. Follow-up and therapy data were available in 95 patients. In 20 patients treated with surgery only, neither p16+ nor Rb- immunostaining were associated with disease-free survival and overall survival. In 75 patients treated with adjuvant chemotherapy, p16+ was associated with good response to therapy with significant increased disease-free survival (P=0.001) and showed a trend towards a statistical significance for increased overall survival (P=0.056); Rb- were not associated with disease-free survival and overall survival. In multivariate analysis, p16+ was independently associated with disease-free and overall survival, with a hazard ratio of 0.18 (95% CI: 0.06-0.51; P=0.001) and 0.21 (95% CI: 0.06-0.74; P=0.015), respectively. In patients with no-special-type triple-negative breast carcinomas, p16+ is related to good response to adjuvant chemotherapy and can be considered the best surrogate marker for Rb pathway loss.


Subject(s)
Biomarkers, Tumor/analysis , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Retinoblastoma Protein/biosynthesis , Triple Negative Breast Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Female , Humans , Immunohistochemistry , Middle Aged , Prognosis , Triple Negative Breast Neoplasms/mortality
12.
J Biol Chem ; 287(11): 7896-906, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22262832

ABSTRACT

We previously showed that depletion of the retinoblastoma protein (RB) induces down-regulation of the adhesion molecule E-cadherin and thereby triggers the epithelial-mesenchymal transition. To further characterize the effect of RB inactivation on the phenotype of cancer cells, we have now examined RB expression in human breast cancer cell lines and clinical specimens. We found that RB-inactive cells exhibit a mesenchymal-like morphology and are highly invasive. We also found that ZEB proteins, transcriptional repressors of the E-cadherin gene, are markedly up-regulated in these cells in a manner sensitive to the miR-200 family of microRNAs. Moreover, depletion of ZEB in RB-inactive cells suppressed cell invasiveness and proliferation and induced epithelial marker expression. These results implicate ZEB in induction of the epithelial-mesenchymal transition, as well as in maintenance of the mesenchymal phenotype in RB-inactive cells. We also developed a screening program for inhibitors of ZEB1 expression and thereby identified several cyclin-dependent kinase inhibitors that blocked both ZEB1 expression and RB phosphorylation. Together, our findings suggest that RB inactivation contributes to tumor progression not only through loss of cell cycle control but also through up-regulation of ZEB expression and induction of an invasive phenotype.


Subject(s)
Biomarkers, Tumor/biosynthesis , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/biosynthesis , Retinoblastoma Protein/biosynthesis , Transcription Factors/biosynthesis , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cadherins/biosynthesis , Cadherins/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Drug Screening Assays, Antitumor , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Homeodomain Proteins/genetics , Humans , MicroRNAs/biosynthesis , MicroRNAs/genetics , Neoplasm Invasiveness , Protein Kinase Inhibitors/pharmacology , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Retinoblastoma Protein/genetics , Transcription Factors/genetics , Up-Regulation/drug effects , Up-Regulation/genetics , Zinc Finger E-box-Binding Homeobox 1
13.
Int J Cancer ; 132(1): 63-71, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22514107

ABSTRACT

Judging the carcinogenicity of human papillomavirus (HPV) types rarely found in cervical cancer (CxCa) is hindered by lack of studies of their biological activity in cancer tissues. To asses transcriptional activity of HPV types, we have developed ultra-short amplimer, splice-site specific, E6*I mRNA RT-PCR assays for 12 high-risk (HR)-HPV (IARC Group 1) and eight probable/possible high-risk (pHR)-HPV types (IARC Group 2A/B carcinogens). Previously unreported E6*I splice sites of the six pHR-HPV types 26, 53, 67, 70, 73 and 82 were identified by cloning and sequencing. We analyzed 97 formalin-fixed paraffin-embedded (FFPE) Mongolian CxCa biopsies for presence of HPV DNA by two sensitive genotyping assays, for E6*I transcripts of all HR-/pHR-HPV types identified and for expression of HPV surrogate markers p16(INK4a), pRb and p53. E6*I of at least one HR-/pHR-HPV was expressed in 94 (98%) of cancer tissues including seven with pHR-HPV types 26, 66, 70 or 82 as single transcribed types. Fifty-eight of E6*I mRNA transcribing cases were analyzable by immunohistochemistry and displayed p16(INK4a) overexpression in 57 (98%), pRb downregulation in 56 (97%) and p53 downregulation in 36 (62%) tissues. The newly developed E6*I mRNA RT-PCR assays appeared to be highly sensitive method to analyze HPV transcription in FFPE materials. Our finding of viral oncogene transcription of pHR-HPV types 26, 66, 70 and 82 in cervical tumors, in the absence of any other transcriptionally active HR-type and with p16(INK4a) overexpression and pRb downregulation, may support a reassessment of the carcinogenicity classification of these pHR-HPV types.


Subject(s)
Papillomaviridae/genetics , Papillomavirus Infections/virology , Uterine Cervical Neoplasms/virology , Adult , Aged , Cyclin-Dependent Kinase Inhibitor p16 , DNA, Viral/genetics , Female , Genotype , Humans , Immunohistochemistry/methods , Middle Aged , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Papillomavirus Infections/genetics , Papillomavirus Infections/metabolism , RNA Splice Sites/genetics , RNA, Messenger/genetics , RNA, Viral/genetics , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/genetics , Risk Factors , Transcription, Genetic/genetics , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism
14.
Cell Biol Toxicol ; 29(4): 283-91, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23955090

ABSTRACT

Metolachlor, a commonly used herbicide in the Midwestern USA, functions by inhibiting chlorophyll and protein synthesis in target plants. Herbicide exposure has led to detrimental effects in several organisms, affecting their growth and behavior; however, its mechanism of action in nontarget organisms is not yet clear. The EPA does not currently have enforceable regulations for maximal limits allowed in drinking water. Previous growth studies from our lab have demonstrated that increasing metolachlor concentrations and increasing time of exposure results in decreased growth of liver cells. The objective of this study was to elucidate a mechanism for this decrease of HepG2 cell growth after herbicide exposure. Results show that metolachlor at environmentally relevant levels (50-100 ppb) that previously led to decreased cell number does not lead to cell death by either necrosis or apoptosis. However, it was demonstrated that the levels of the retinoblastoma protein including two of its hyperphosphorylated forms are decreased in metolachlor exposed cells possibly leading to cell cycle arrest. The levels of another protein involved in cell cycle progression, p53, a mediator in the DNA damage response of cells, was not significantly altered except at the highest level of metolachlor (1,000 ppb) and after a 72-h exposure. These results suggest that the decrease in cell number after low-level metolachlor exposure is most likely due to an alteration in the cell cycle and not due to cell death in human liver cells.


Subject(s)
Acetamides/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Herbicides/pharmacology , Acetamides/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Herbicides/metabolism , Humans , Liver/cytology , Liver/drug effects , Phosphorylation/drug effects , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/metabolism
15.
Int J Cancer ; 128(7): 1532-45, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-20503270

ABSTRACT

There is increasing use of multiple molecular markers to predict prognosis in human cancer. Our aim was to examine the prognostic significance of cyclin D1 and retinoblastoma (pRb) expression in association with human papillomavirus (HPV) status in oropharyngeal squamous cell carcinoma. Clinical records and specimens of 226 patients with follow-up from 1 to 235 months postdiagnosis were retrieved. Tumor HPV status was determined by HPV E6-targeted multiplex real-time PCR/p16 semiquantitative immunohistochemistry and cyclin D1 and pRb expression by semiquantitative immunohistochemistry. Determinants of recurrence and mortality hazards were modeled using Cox regression with censoring at dates of last follow-up. The HPV-positivity rate was 37% (91% type 16). HPV was a predictor of recurrence, an event (recurrence or death) and death after adjustment for clinicopathological variables. There were inverse relationships between HPV status and cyclin D1 and pRb. On univariate analysis, cyclin D1 predicted locoregional recurrence, event and death and pRb predicted event and death. Within the HPV-positive group, after adjusting for clinicopathological factors, patients with cyclin D1-positive cancers had up to a eightfold increased risk of poor outcome relative to those with cyclin D1-negative tumors. However, within the HPV-negative group, there was only a very small adjusted increased risk. A combination of pRb and HPV did not provide additional prognostic information. Our data provide the first evidence that a combination of HPV and cyclin D1 provides more prognostic information in oropharyngeal cancer than HPV alone. If findings are confirmed, treatment based on HPV and cyclin D1 may improve outcomes.


Subject(s)
Carcinoma, Squamous Cell/virology , Cyclin D1/metabolism , Oropharyngeal Neoplasms/virology , Papillomavirus Infections/metabolism , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/metabolism , Female , Humans , Male , Middle Aged , Oropharyngeal Neoplasms/metabolism , Prognosis , Recurrence , Retinoblastoma Protein/biosynthesis , Treatment Outcome
16.
J Reprod Dev ; 57(4): 492-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21519154

ABSTRACT

Mammalian preimplantation embryos enter the S phase immediately after the end of the M phase; their cell cycle lacks a substantial G1 phase. Previously, we suggested that the absence of the G1 phase was attributable to a loss of retinoblastoma protein (RB) function, which is required for suppression of S phase entrance and that this loss of RB function in turn was attributable to the low RB expression level during preimplantation development in mouse embryos. The present study aimed to examine whether or not RB inhibition by CDK4/6-cyclin D-dependent phosphorylation is involved in the loss of RB function in preimplantation mouse embryos by the expression of p16(INK4a), a potent endogenous inhibitor of CDK4/6-cyclin D. First, the decrease in RB expression between the four-cell and morula stages was confirmed in in vivo-derived mouse embryos. We then examined the efficiency of the p16(INK4a) expression vector in inhibiting RB phosphorylation and cell cycle progression using NIH-3T3 cells and obtained gradual RB dephosphorylation and a significantly lower proliferation rate in p16(INK4a)-transfected cells than in control cells. This indicated the successful p16(INK4a) effects on cell-cycle progression by the vector used. On the other hand, the development rate of mouse embryos injected with the p16(INK4a) expression vector was the same as that of the control embryos, although p16(INK4a) expression was detected at mRNA and protein levels in the former group but not in the control group. These results suggest that RB phosphorylation is not involved in RB dysfunction or in the lack of a G1 phase in mouse embryos and that the decrease in RB expression is important for preimplantation-embryo-specific cell-cycle regulation. Moreover, the present study indicates the similarity between preimplantation embryos and cancer cells, which p16(INK4a) expression does not arrest at the G1 phase.


Subject(s)
Blastocyst/cytology , Retinoblastoma Protein/biosynthesis , Animals , Cell Cycle , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Primers/genetics , Female , G1 Phase , Gene Expression Regulation, Developmental , Mice , Mice, Inbred ICR , NIH 3T3 Cells , Phosphorylation , RNA, Messenger/metabolism
17.
Med Oral Patol Oral Cir Bucal ; 16(3): e323-9, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21196892

ABSTRACT

OBJECTIVE: This study aimed to characterize alteration in the immunohistochemical expression of pRb in normal tissue of the salivary gland surrounding pleomorphic adenoma, the tumor cells of pleomorphic adenomas, and carcinoma arising in pleomorphic adenoma. STUDY DESIGN: A selected series of 29 cases of pleomorphic adenomas, and 27 cases of carcinoma ex-pleomorphic adenoma (undifferentiated and adenocarcinoma types) were examined. RESULTS: The results showed that pRb expression was negative in the components of normal tissue of the salivary gland surrounding pleomorphic adenoma. PRb expression in pleomorphic adenomas shows that 2 cases out of 29 (6.9%) strongly expressed in the duct cells, 7/29 (24.1%) cases showed moderate staining. pRb nuclear staining in myxochondroid was identified in 10/29 (34.5%) cases of pleomorphic adenomas with weak staining, 6/29 (20.7%) with moderate staining. pRb was strongly expressed in carcinoma cells in 19 out of 27 cases (70.4%). CONCLUSION: This study suggests the alteration of pRb expression would increase from pleomorphic adenoma to carcinoma arising in pleomorphic adenomas (6.9% versus 70.4%).


Subject(s)
Adenocarcinoma/metabolism , Adenoma, Pleomorphic/metabolism , Carcinoma/metabolism , Retinoblastoma Protein/biosynthesis , Salivary Gland Neoplasms/metabolism , Adenocarcinoma/chemistry , Adenoma, Pleomorphic/chemistry , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma/chemistry , Female , Humans , Immunohistochemistry , Male , Middle Aged , Retinoblastoma Protein/analysis , Salivary Gland Neoplasms/chemistry , Young Adult
18.
J Biol Chem ; 284(52): 36234-36239, 2009 Dec 25.
Article in English | MEDLINE | ID: mdl-19880526

ABSTRACT

Williams-Beuren syndrome (WBS), an autosomal dominant genetic disorder, is characterized by a unique cognitive profile and craniofacial defects. WBS results from a microdeletion at the chromosomal location 7q11.23 that encompasses the genes encoding the members of TFII-I family of transcription factors. Given that the haploinsufficiency for TFII-I is causative to the craniofacial phenotype in humans, we set out to analyze the effect of post-transcriptional silencing of TFII-I during BMP-2-driven osteoblast differentiation in the C2C12 cell line. Our results show that TFII-I plays an inhibitory role in regulating genes that are essential in osteogenesis and intersects with the bone-specific transcription factor Runx2 and the retinoblastoma protein, pRb. Identification of pathways regulated by TFII-I family transcription factors may begin to shed light on the molecular determinants of WBS.


Subject(s)
Antigens, Differentiation/biosynthesis , Osteoblasts/metabolism , Osteogenesis , RNA Interference , Transcription Factors, TFII/metabolism , Williams Syndrome/metabolism , Animals , Antigens, Differentiation/genetics , Bone Morphogenetic Protein 2/pharmacology , COS Cells , Cell Differentiation/drug effects , Cell Differentiation/genetics , Chlorocebus aethiops , Chromosome Deletion , Chromosomes, Human, Pair 7/genetics , Chromosomes, Human, Pair 7/metabolism , Core Binding Factor Alpha 1 Subunit/biosynthesis , Core Binding Factor Alpha 1 Subunit/genetics , Humans , Mice , NIH 3T3 Cells , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/genetics , Transcription Factors, TFII/genetics , Williams Syndrome/genetics
19.
J Virol ; 83(23): 12590-600, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19793824

ABSTRACT

Hepatitis C virus (HCV) infection is associated with the development of hepatocellular carcinoma and probably also non-Hodgkin's B-cell lymphoma. The molecular mechanisms of HCV-associated carcinogenesis are unknown. Here we demonstrated that peripheral blood mononuclear cells obtained from hepatitis C patients and hepatocytes infected with HCV in vitro showed frequent chromosomal polyploidy. HCV infection or the expression of viral core protein alone in hepatocyte culture or transgenic mice inhibited mitotic spindle checkpoint function because of reduced Rb transcription and enhanced E2F-1 and Mad2 expression. The silencing of E2F-1 by RNA interference technology restored the function of mitotic checkpoint in core-expressing cells. Taken together, these data suggest that HCV infection may inhibit the mitotic checkpoint to induce polyploidy, which likely contributes to neoplastic transformation.


Subject(s)
Hepacivirus/physiology , Hepatocytes/virology , Host-Pathogen Interactions , Leukocytes, Mononuclear/virology , Polyploidy , Virus Replication , Animals , Calcium-Binding Proteins/biosynthesis , Cell Cycle Proteins/biosynthesis , E2F1 Transcription Factor/biosynthesis , Gene Expression Regulation , Gene Silencing , Humans , Mad2 Proteins , Mice , Mice, Transgenic , Repressor Proteins/biosynthesis , Retinoblastoma Protein/biosynthesis
20.
Am J Pathol ; 174(6): 2107-15, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19389926

ABSTRACT

Uterine carcinosarcomas (UCSs) are considered to represent true examples of the epithelial-mesenchymal transition. Akt plays a key role in the induction of epithelial-mesenchymal transition, but little is known about its involvement in tumorigenesis. Here we examined the functional roles of the Akt/beta-catenin pathway in UCSs. In clinical samples, phospho-Akt (pAkt) expression was found to be significantly increased in mesenchymal compared with epithelial components, exhibiting both positive and negative correlations with nuclear beta-catenin and E-cadherin, respectively. Expression levels of the transcription factor Slug were also significantly up-regulated in the mesenchymal components and strongly correlated with both pAkt and nuclear beta-catenin. In endometrial cancer cell lines, active Akt induced the stabilization of nuclear beta-catenin through the phosphorylation of GSK-3beta, and this, in turn, led to the transactivation of Slug, which was mediated by nuclear beta-catenin. Moreover, Slug overexpression itself caused repression of E-cadherin, with subtle changes in cell morphology. In addition, knockdown of the retinoblastoma gene product (Rb) up-regulated pAkt and repressed E-cadherin, consistent with the in vivo finding of significantly decreased Rb expression in mesenchymal components. These findings suggest that changes in the Akt/beta-catenin pathway, as well as alterations in Rb expression, may be essential for both the establishment and maintenance of phenotypic characteristics of UCSs, playing key roles in the regulation of E-cadherin through the transactivation of the Slug gene.


Subject(s)
Cadherins/biosynthesis , Carcinosarcoma/metabolism , Proto-Oncogene Proteins c-akt/biosynthesis , Transcription Factors/biosynthesis , Uterine Neoplasms/metabolism , beta Catenin/biosynthesis , Blotting, Western , Cadherins/genetics , Carcinosarcoma/genetics , Female , Fluorescent Antibody Technique , Gene Expression , Humans , Immunohistochemistry , Polymerase Chain Reaction , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Snail Family Transcription Factors , Transcription Factors/genetics , Transcriptional Activation , Transfection , Uterine Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL