Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 148
Filter
1.
Oncogene ; 31(43): 4619-29, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-22249256

ABSTRACT

The epithelial-to-mesenchymal transition (EMT) is a crucial program for the invasion and metastasis of epithelial tumors that involves loss of cell-cell adhesion and increased cell mobility; however, mechanisms underlying this transition are not fully elucidated. Here, we propose a novel mechanism through which the nicotinamide adenine dinucleotide-dependent histone deacetylase SIRT1 regulates EMT in prostate cancer cells through cooperation with the EMT inducing transcription factor ZEB1. We found that forced expression of SIRT1 in non-transformed PZ-HPV-7 prostate epithelial cells disrupts the epithelial morphology concomitant with decreased expression of the epithelial marker, E-cadherin, and increased expression of mesenchymal markers. In contrast, silencing SIRT1 in metastatic prostate tumor cells restores cell-cell adhesion and induces a shift toward an epithelial morphology concomitant with increased expression of E-cadherin and decreased expression of mesenchymal markers. We also found that SIRT1 has a physiologically relevant role in endogenous EMT induced by EGF signaling in prostate cancer cells. We propose that the regulation of EMT by SIRT1 involves modulation of, and cooperation with, the EMT inducing transcription factor ZEB1. Specifically, we show that SIRT1 silencing reduces expression of ZEB1 and that SIRT1 is recruited to the E-cadherin proximal promoter by ZEB1 to deacetylate histone H3 and to reduce binding of RNA polymerase II, ultimately suppressing E-cadherin transcription. We thus identify a necessary role for ZEB1 in SIRT1-mediated EMT. Finally, we show that reduction of SIRT1 decreases prostate cancer cell migration in vitro and metastasis in vivo in immunodeficient mice, which is largely independent of any general effects of SIRT1 on prostate cancer growth and survival. We therefore identify SIRT1 as a positive regulator of EMT and metastatic growth of prostate cancer cells and our findings implicate overexpressed SIRT1 as a potential therapeutic target to reverse EMT and to prevent prostate cancer progression.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplasm Metastasis , Prostatic Neoplasms/pathology , Sirtuin 1/physiology , Transcription Factors/metabolism , Gene Silencing , Humans , Male , Prostatic Neoplasms/metabolism , Sirtuin 1/genetics
2.
J Endocrinol ; 213(1): 37-48, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22159506

ABSTRACT

Sirtuins, which are class III NAD-dependent histone deacetylases that regulate a number of physiological processes, play important roles in the regulation of metabolism, aging, oncogenesis, and cancer progression. Recently, a role for the sirtuins in the regulation of steroid hormone receptor signaling is emerging. In this mini-review, we will summarize current research into the regulation of estrogen, androgen, progesterone, mineralocorticoid, and glucocorticoid signaling by sirtuins in cancer. Sirtuins can regulate steroid hormone signaling through a variety of molecular mechanisms, including acting as co-regulatory transcription factors, deacetylating histones in the promoters of genes with nuclear receptor-binding sites, directly deacetylating steroid hormone nuclear receptors, and regulating pathways that modify steroid hormone receptors through phosphorylation. Furthermore, disruption of sirtuin activity may be an important step in the development of steroid hormone-refractory cancers.


Subject(s)
Neoplasms/genetics , Neoplasms/metabolism , Sirtuin 1/physiology , Humans , Neoplasms/pathology , Signal Transduction/physiology , Sirtuin 1/metabolism
3.
Oncogene ; 28(5): 651-61, 2009 Feb 05.
Article in English | MEDLINE | ID: mdl-19015639

ABSTRACT

HIC1 is a newly discovered tumor suppressor and transcriptional repressor that is frequently silenced in human tumors. HIC1 protein expression has been linked to better outcomes in breast cancers. The molecular mechanism underlying HIC1-mediated transcriptional and growth suppression, and the relevant targets of HIC1-mediated transcriptional modulation, is currently unclear. We have identified an HIC1 DNA-binding site in E2F-responsive gene promoters and demonstrate that HIC1 targets E2F-responsive genes for transcriptional regulation and growth suppression. We and others have recently discovered that Brg1, a central component of the SWI/SNF chromatin-remodeling family, is required for the transcriptional regulation of multiple cell cycle control-related genes, including E2F-responsive promoters. We studied HIC1 interactions with, and dependence upon, Brg1 activity, and found that HIC1 can recruit Brg1 to E2F-responsive promoters and that its transcriptional repression of these genes is dependent upon Brg1. These data indicate that HIC1 is a central molecule in a novel mechanism controlling cell growth and that the disruption of this HIC1-mediated pathway may lead to abnormal cell proliferation and, ultimately, cancer.


Subject(s)
Cell Proliferation , Chromatin Assembly and Disassembly/physiology , Down-Regulation/genetics , Kruppel-Like Transcription Factors/physiology , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/metabolism , DNA Helicases/physiology , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/physiology , Genes, Tumor Suppressor/physiology , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Neoplasms/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Protein Binding , Transcription Factors/metabolism , Transcription Factors/physiology , Transcription, Genetic/genetics , Tumor Cells, Cultured
4.
Oncogene ; 26(50): 7153-7, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17486062

ABSTRACT

The SWI/SNF complex participates as a co-activator in the transcriptional regulation of certain genes. Conversely, we and others have recently established that Brg1 and Brm, the central components of SWI/SNF, act instead as co-repressors for E2F-mediated transcriptional repression, and for the transcription of certain other promoters. We report here that Brg-1 and Brm can switch their mode of function at same promoter between activation and repression by ligand-directed differential coordination with BAF155, BAF170, HDAC1, p300 and prohibitin. This ligand and context-dependent reprogramming of the SWI/SNF complex allows it to differentially serve as either a co-repressor or a co-activator of transcription at the same promoter.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation/physiology , Receptors, Estrogen/metabolism , Repressor Proteins/physiology , Trans-Activators/physiology , Transcription Factors/metabolism , Tumor Suppressor Proteins/physiology , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/physiology , DNA Helicases/metabolism , DNA-Binding Proteins , Estrogens/metabolism , Humans , Ligands , Nuclear Proteins/metabolism , Prohibitins , Receptors, Estrogen/genetics , Transcription Factors/physiology
5.
Curr Pharm Des ; 11(9): 1091-104, 2005.
Article in English | MEDLINE | ID: mdl-15853658

ABSTRACT

The activity of genes encoded by the highly-condensed DNA in cellular nuclei must be precisely regulated. Regulation of the accessibility of gene promoters to transcription complexes is one level of gene regulation and is influenced by histone tail modifications such as acetylation, methylation, and phosphorylation. Acetylation is a reversible modification catalyzed by histone acetyl transferase (HAT) and histone deacetyltransferase (HDAC) enzymes. Histone deacetylation is associated with transcriptional repression of genes, as the removal of acetyl groups from lysine residues allows for tighter electrostatic interactions between DNA and histones, limiting accessibility of the DNA for transcription. Inhibition of HDAC activity permits histones to remain in an acetylated state, and through the resulting alterations in gene regulation, inhibits cell cycle progression, inhibits differentiation, and in some cases induces apoptosis. Inhibition of proliferation by HDAC inhibitors is characterized by arrest at the G1 or G2/M phases of the cell cycle. Many types of tumor cells then undergo programmed cell death. Exposure to HDAC inhibitors may also allow reactivation of tumor suppressor genes which had been silenced by hypoacetylation during tumorigenesis. HDAC inhibitors from a number of chemical classes have shown promise as anti-cancer agents in animal studies and early clinical trials. The development of HDAC inhibitors which specifically target HDAC isozymes, and more detailed understanding of their anti-neoplastic actions, heralds a new epigenetic antitumor therapeutic strategy.


Subject(s)
Antineoplastic Agents/administration & dosage , Cell Cycle/drug effects , Drug Delivery Systems/methods , Histone Deacetylase Inhibitors , Protease Inhibitors/administration & dosage , Animals , Antineoplastic Agents/chemistry , Cell Cycle/physiology , Histone Deacetylases/metabolism , Humans , Protease Inhibitors/chemistry
6.
Curr Opin Oncol ; 13(5): 360-7, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11555713

ABSTRACT

Lymphoproliferative diseases (LPDs) associated with the Epstein-Barr virus (EBV) include non-Hodgkin lymphomas, which occur in the setting of immunosuppression, including that induced by human immunodeficiency virus, and posttransplant lymphoproliferative disorders. These LPDs are characterized by actively proliferating, latently infected EBV-positive B lymphocytes and often follow a rapidly progressive fatal clinical course. Pharmacologic treatment for herpesvirus infections has targeted the virus-specific enzyme, thymidine kinase (TK), with nucleoside analogs. The lack of viral TK expression in EBV-positive tumors, caused by viral latency, however, makes antiviral therapy alone ineffective as an antineoplastic therapy. Arginine butyrate selectively activates the EBV TK gene in latently infected EBV-positive tumor cells. We have developed a strategy for treatment of EBV-associated lymphomas using pharmacologic induction of the latent viral TK gene and enzyme in tumor cells using arginine butyrate, followed by treatment with ganciclovir. A phase I/II trial, using an intrapatient dose escalation of arginine butyrate combined with ganciclovir, is underway. This combination therapy has produced complete clinical responses in 5 of 10 previously refractory patients, with partial responses occurring in 2 additional patients. This virus-targeted antitumor strategy may provide a new therapeutic approach to EBV-associated neoplasms.


Subject(s)
Antineoplastic Agents/pharmacology , Antiviral Agents/therapeutic use , Arginine/analogs & derivatives , Arginine/pharmacology , Butyrates/pharmacology , Epstein-Barr Virus Infections/complications , Ganciclovir/therapeutic use , Gene Expression Regulation, Neoplastic , Herpesvirus 4, Human/genetics , Lymphoma, Non-Hodgkin/therapy , Lymphoma, Non-Hodgkin/virology , Thymidine Kinase/biosynthesis , Antineoplastic Agents/therapeutic use , Arginine/therapeutic use , Butyrates/therapeutic use , Clinical Trials as Topic , Combined Modality Therapy , Enzyme Induction , Herpesvirus 4, Human/pathogenicity , Humans , Prognosis , Thymidine Kinase/metabolism
7.
Transpl Infect Dis ; 3(3): 177-85, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11493400

ABSTRACT

Lymphoproliferative disorders associated with the Epstein-Barr virus (EBV) include non-Hodgkin's lymphoma, Hodgkin's lymphoma, and "post-transplant lymphoproliferative disorders" (PTLD), which occur with immunosuppression after marrow and organ transplantation. PTLD is characterized by actively proliferating, latently infected EBV(+) B-lymphocytes, and often manifests a rapidly progressive fatal clinical course if the immunosuppression cannot be reversed. Lung transplant recipients are a subset of patients at special risk for developing PTLD. The incidence of PTLD development in these patients has been estimated at 5--10%. Whereas immunologic and antiviral therapy have been moderately effective for treating EBV-associated infections in the lytic phase, they have been less useful in the more common latent phase of the disease. One common treatment for herpesvirus infections has targeted the virus-specific enzyme thymidine kinase (TK). The lack of viral TK expression in EBV(+) tumor cells, due to viral latency, makes anti-viral therapy alone ineffective as an anti-neoplastic therapy, however. We have developed a strategy for the treatment of EBV-associated lymphomas/PTLD using pharmacologic induction of the latent viral TK gene and enzyme in the tumor cells, followed by treatment with ganciclovir. Arginine butyrate selectively activates the EBV TK gene in latently EBV-infected human lymphoid cells and tumor cells. A Phase I/II trial has been initiated, employing an intra-patient dose escalation of arginine butyrate combined with ganciclovir. In six patients with EBV-associated lymphomas or PTLD, all of which were resistant to conventional radiation and/or chemotherapy, this combination produced complete clinical responses in four of six patients, with a partial response occurring in a fifth patient. Pathologic examination in two of three patients demonstrated complete necrosis of the EBV lymphoma, with no residual disease, following a single three-week course of the combination therapy. Possible side-effects of the therapy included nausea and reversible lethargy at the highest doses. One patient suffered acute liver failure, thought to be secondary to release of FasL from the necrotic tumor. Analysis of patient-derived tumor cells in culture demonstrated that arginine butyrate produced selective induction of the EBV TK gene, which then conferred sensitivity to ganciclovir, resulting in tumor apoptosis. Additional patient accrual is sought for further evaluation of this therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Arginine/analogs & derivatives , Arginine/pharmacology , Butyrates/pharmacology , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human/genetics , Lung Transplantation/adverse effects , Lymphoproliferative Disorders/etiology , Antineoplastic Agents/therapeutic use , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Arginine/therapeutic use , Butyrates/therapeutic use , Drug Therapy, Combination , Ganciclovir/pharmacology , Ganciclovir/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Viral/drug effects , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/physiology , Humans , Lung Neoplasms/etiology , Lymphoproliferative Disorders/drug therapy , Postoperative Complications/drug therapy , Postoperative Complications/virology , Tumor Cells, Cultured/drug effects , Virus Latency
8.
Front Biosci ; 6: D1008-18, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11487468

ABSTRACT

The BET proteins are a novel class of transcriptional regulators whose members can be found in animals, plants and fungi. Founding members are Human RING3, Drosophila Fsh and yeast Bdf1p. BET proteins are distinguished by an N-terminal bromodomain or bromodomains and an ET domain. As predicted by the presence of the bromodomain(s), these proteins have been found to be associated with chromatin. The poorly characterized ET domain functions as a protein-protein interaction motif and may be part of a serine-kinase activity. Other regions ("modular domains"), which are conserved only in certain BET proteins, are likely to provide sub-family specific functions. Genetic, biochemical and molecular techniques have implicated BET proteins in functions as diverse as meiosis, cell cycle control and homeosis. The data suggest that BET proteins may modulate chromatin structure and affect transcription via a sequence-independent mechanism. This review will attempt to summarize current research on BET proteins and envision where future research is likely to lead.


Subject(s)
Drosophila Proteins , Saccharomyces cerevisiae Proteins , Transcription Factors/physiology , Amino Acid Sequence , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/physiology , Evolution, Molecular , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/physiology , Phylogeny , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/physiology , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Transcription Factors/chemistry
9.
Cell Growth Differ ; 12(6): 297-306, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11432804

ABSTRACT

Oncogenic Ras induces cells to undergo apoptosis after inhibition of protein kinase C (PKC) activity. The integration of differential signaling pathways is required for full execution of apoptosis. In this study, we used Jurkat as well as Fas/FADD-defective cell lines expressing v-ras to determine the upstream elements required for activation of the caspase cascade in PKC/Ras-mediated apoptosis. During this Ras-induced apoptotic process, caspase-8 was activated, possibly through its binding to Fas-associated death domain (FADD), in Jurkat/ras and Jurkat/Fas(m)/ras cells but not in Jurkat/FADD(m)/ras cells. c-Jun NH(2)-terminal kinase (JNK) was activated in all three cell lines expressing ras in response to apoptotic stimulation. Suppression of JNK by dn-JNK1 blocked the interaction of FADD and caspase-8 and partially protected Jurkat/ras and Jurkat/Fas(m)/ras cells from apoptosis. However, dn-JNK1 had no effect on PKC/Ras-induced apoptosis in Jurkat/FADD(m)/ras cells. The results indicate that FADD/caspase-8 signaling is involved in PKC/Ras-mediated apoptosis, and JNK may be an upstream effector of caspase activation.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis , Carrier Proteins/metabolism , Caspases/metabolism , fas Receptor/metabolism , ras Proteins/metabolism , BH3 Interacting Domain Death Agonist Protein , Caspase 8 , Caspase 9 , Cytochrome c Group/metabolism , Enzyme Activation , Fas-Associated Death Domain Protein , Humans , JNK Mitogen-Activated Protein Kinases , Jurkat Cells , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction
10.
Blood ; 97(10): 3259-67, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11342457

ABSTRACT

Current chemotherapeutic and butyrate therapeutics that induce fetal hemoglobin expression generally also suppress erythropoiesis, limiting the production of cells containing fetal hemoglobin (F cells). Recently, selected short-chain fatty acid derivatives (SCFADs) were identified that induce endogenous gamma-globin expression in K562 cells and human burst-forming units-erythroid and that increase proliferation of human erythroid progenitors and a multilineage interleukin-3-dependent hematopoietic cell line. In this report, gamma-globin inducibility by these SCFADs was further demonstrated in mice transgenic for the locus control region and the entire beta-globin gene locus in a yeast artificial chromosome and in 2 globin promoter-reporter assays. Conditioned media experiments strongly suggest that their proliferative activity is a direct effect of the test compounds. Investigation of potential mechanisms of action of these SCFADs demonstrates that these compounds induce prolonged expression of the growth-promoting genes c-myb and c-myc. Both butyrate and specific growth-stimulatory SCFADs induced prolonged signal transducer and activator of transcription (STAT)-5 phosphorylation and activation, and c-cis expression, persisting for more than 120 minutes, whereas with IL-3 alone phosphorylation disappeared within minutes. In contrast to butyrate treatment, the growth-stimulating SCFADs did not result in bulk histone H4 hyperacetylation or induction of p21(Waf/Cip), which mediates the suppression of cellular growth by butyrate. These findings suggest that the absence of bulk histone hyperacetylation and p21 induction, but prolonged induction of cis, myb, myc, and STAT-5 activation, contribute to the cellular proliferation induced by selected SCFADs.


Subject(s)
Cell Division/drug effects , DNA-Binding Proteins/metabolism , Fatty Acids, Volatile/pharmacology , Milk Proteins , Trans-Activators/metabolism , Animals , Butyrates/pharmacology , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins , Globins/biosynthesis , Globins/genetics , Growth Substances/pharmacology , Histones/metabolism , Humans , Immediate-Early Proteins/biosynthesis , Interleukin-3/pharmacology , Kinetics , Mice , Mice, Transgenic , Phosphorylation , Proto-Oncogene Proteins c-myb/biosynthesis , Proto-Oncogene Proteins c-myc/biosynthesis , STAT5 Transcription Factor , Suppressor of Cytokine Signaling Proteins
11.
Endocrinology ; 142(6): 2600-5, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11356710

ABSTRACT

Retinoic acid (RA) slows growth and induces differentiation of tumor cells through activation of RA receptors (RARs). However, melanoma cell lines display highly variable responsiveness to RA, which is a poorly understood phenomenon. By using Northern and Western blot analyses, we show that RA-resistant A375 and RA-responsive S91 melanoma cells express comparable levels of major components of RAR-signaling pathways. However, A375 cells have substantially higher intracellular reactive oxygen species (ROS) levels than S91 cells. Lowering ROS levels in A375 cells through hypoxic culture conditions restores RAR-dependent trans-activity, which could be further enhanced by addition of the antioxidant N-acetyl-cysteine. Hypoxia also enhances RAR activity in the moderately RA-responsive C32 cells, which have intermediate ROS levels. Conversely, increasing oxidative stress in highly RA-responsive S91 and B16 cells, which have low ROS levels, by treatment with H(2)O(2) impairs RAR activity. Consistent with these observations, RA more potently inhibited the proliferation of hypoxic A375 cells than that of normoxic cells. Oxidative states diminish, whereas reducing conditions enhance, DNA binding of retinoid X receptor/RAR heterodimers in vitro, providing a molecular basis for the observed inverse correlation between RAR activity and ROS levels. The redox state of melanoma cells provides a novel, epigenetic control mechanism of RAR activity and RA resistance.


Subject(s)
Drug Resistance, Neoplasm , Melanoma/pathology , Receptors, Retinoic Acid/physiology , Tretinoin/pharmacology , Acetylcysteine/pharmacology , Antioxidants/pharmacology , Blotting, Northern , Blotting, Western , Cell Hypoxia , DNA/metabolism , Dimerization , Gene Expression/drug effects , Hydrogen Peroxide/pharmacology , Luciferases/genetics , Melanoma/drug therapy , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Recombinant Fusion Proteins , Retinoid X Receptors , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured
12.
Am J Physiol Cell Physiol ; 280(4): C897-911, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11245607

ABSTRACT

Hypoxia is known to induce extravasation of lymphocytes and leukocytes during ischemic injury and increase the metastatic potential of malignant lymphoid cells. We have recently identified a new adhesion molecule, hypoxia-activated ligand-1/13 (HAL-1/13), that mediates the hypoxia-induced increases in lymphocyte and neutrophil adhesion to endothelium and hypoxia-mediated invasion of endothelial cell monolayers by tumor cells. In this report, we used expression cloning to identify this molecule as the lupus antigen and DNA-dependent protein kinase-associated nuclear protein, Ku80. The HAL-1/13-Ku80 antigen is present on the surface of leukemic and solid tumor cell lines, including T and B lymphomas, myeloid leukemias, neuroblastoma, rhabdomyosarcoma, and breast carcinoma cells. Transfection and ectopic expression of HAL-1/13-Ku80 on (murine) NIH/3T3 fibroblasts confers the ability of these normally nonadhesive cells to bind to a variety of human lymphoid cell lines. This adhesion can be specifically blocked by HAL-1/13 or Ku80-neutralizing antibodies. Loss of expression variants of these transfectants simultaneously lost their adhesive properties toward human lymphoid cells. Hypoxic exposure of tumor cell lines resulted in upregulation of HAL-1/13-Ku80 expression at the cell surface, mediated by redistribution of the antigen from the nucleus. These studies indicate that the HAL-1/13-Ku80 molecule may mediate, in part, the hypoxia-induced adhesion of lymphocytes, leukocytes, and tumor cells.


Subject(s)
Antigens, Nuclear , DNA Helicases , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Hypoxia/immunology , Hypoxia/metabolism , Lymphocytes/cytology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , 3T3 Cells , Animals , Autoantigens/genetics , Autoantigens/immunology , Autoantigens/metabolism , Cell Adhesion/immunology , Cell Membrane/immunology , Cell Membrane/metabolism , Cloning, Molecular , DNA, Complementary , DNA-Binding Proteins/immunology , Gene Expression/physiology , HeLa Cells , Humans , In Vitro Techniques , Ku Autoantigen , Ligands , Mice , Nuclear Proteins/immunology , Plasmids , Up-Regulation/physiology
13.
J Virol ; 74(20): 9742-8, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11000248

ABSTRACT

We have previously reported that the long terminal repeat (LTR) region of feline leukemia viruses (FeLVs) can enhance expression of certain cellular genes such as the collagenase IV gene and MCP-1 in trans (S. K. Ghosh and D. V. Faller, J. Virol. 73:4931-4940, 1999). Genomic DNA of all healthy feline species also contains LTR-like sequences that are related to exogenous FeLV LTRs. In this study, we evaluated the cellular gene transactivational potential of these endogenous FeLV LTR sequences. Unlike their exogenous FeLV counterparts, neither nearly full-length endogenous FeLV molecular clones (CFE-6 and CFE-16) nor their isolated LTRs were able to activate collagenase IV gene or MCP-1 expression in transient transfection assays. We had also demonstrated previously that production of an RNA transcript from exogenous FeLV LTRs correlates with their transactivational activity. In the present study, we demonstrate that the endogenous FeLV LTRs do not generate LTR-specific RNA transcripts in the feline embryo fibroblast cell line AH927. Furthermore, infection of AH927 cells by an exogenous FeLV subgroup A virus did not induce production of such LTR-specific transcripts from the endogenous proviral genomes, although the LTR-specific transcripts from the exogenous virus were readily detected. Finally, LTR-specific transcripts were not generated in BALB/3T3 cells transiently transfected with isolated CFE-6 LTR, in contrast to transfections with LTRs from exogenous viruses. Our data thus suggest that the inability of endogenous FeLV LTRs in gene transactivation is not due to cell line specificity or presence of any upstream inhibitory cis-acting element. Endogenous, nonleukemogenic FeLV LTRs, therefore, do not transactivate cellular gene expression, and this property appears to be specific to exogenous, leukemogenic FeLVs.


Subject(s)
Leukemia Virus, Feline/genetics , Terminal Repeat Sequences , Transcriptional Activation , 3T3 Cells , Animals , Base Sequence , Cats , Mice , Molecular Sequence Data
14.
J Biol Chem ; 275(50): 39001-11, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-10967125

ABSTRACT

Ras is a well established modulator of apoptosis. Suppression of protein kinase C (PKC) activity can selectively induce apoptosis in cells expressing a constitutively activated Ras protein. We wished to determine whether reactive oxygen species serve as an effector of Ras-mediated apoptosis. Ras-transformed NIH/3T3 cells contained higher basal levels of intracellular H(2)O(2) compared with normal NIH/3T3 cells, and PKC inhibition up-regulated ROS to 5-fold greater levels in Ras-transformed cells than in normal cells. Treatment with N-acetyl-l-cysteine reduced both the basal and inducible levels of intracellular H(2)O(2) in NIH/3T3-Ras cells and antagonized the induction of apoptosis by PKC inhibition. Culturing NIH/3T3-Ras cells in low oxygen conditions, which prevents ROS generation, also inhibited the apoptotic response to PKC inhibition. These results suggest that reactive oxygen species are necessary as downstream effectors of the Ras-mediated apoptotic response to PKC inhibition. However, the generation of ROS alone is not sufficient to induce apoptosis in Ras-transformed cells because inhibition of cell cycle progression prevented the induction of apoptosis in NIH/3T3-Ras cells without inhibiting the generation of intracellular H(2)O(2) observed after PKC inhibition. These findings suggest that continued cell cycle progression of Ras-transformed cells during PKC inhibition is also necessary for the induction of apoptosis.


Subject(s)
Apoptosis , Protein Kinase C/antagonists & inhibitors , Reactive Oxygen Species/metabolism , ras Proteins/physiology , 3T3 Cells , Acetylcysteine/pharmacology , Animals , Antifungal Agents/pharmacology , Butyrates/pharmacology , Cell Cycle/drug effects , Cell Separation , Cell Survival/drug effects , Cyclosporins/pharmacology , DNA Fragmentation/drug effects , Enzyme Inhibitors/pharmacology , Flow Cytometry , Fluoresceins/pharmacology , Free Radical Scavengers/pharmacology , Glyceryl Ethers/pharmacology , Hydrogen Peroxide/metabolism , Hydroxamic Acids/pharmacology , Mice , Mitosis/drug effects , Oxygen/metabolism , Protein Kinase C/metabolism , Time Factors , Transfection , Up-Regulation
15.
Cell Growth Differ ; 11(8): 417-24, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10965846

ABSTRACT

RING3 is a novel, nuclear-localized, serine-threonine kinase that has elevated activity in human leukemias. RING3 transforms NIH/3T3 cells and is activated by mitogenic signals, all of which suggest that it may play a role in cell cycle-responsive transcription. We tested this hypothesis with transient transfection of RING3 into fibroblasts and assayed transactivation of the promoters of cyclin D11 cyclin A, cyclin E, and dihydrofolate reductase (dhfr) genes. RING3 transactivates these promoters in a manner dependent on ras signaling. A kinase-deficient point mutant of RING3 does not transactivate. Mutational analysis of the dhfr promoter reveals that transactivation also depends on the presence of a functional E2F binding site. Furthermore, ectopic expression of Rb protein, a negative regulator of E2F activity, suppresses the RING3-dependent transactivation of this promoter. Consistent with a potential role of E2F in RING3-dependent transcription, anti-RING3 immunoaffinity chromatography or recombinant RING3 protein affinity chromatography of nuclear extracts copurified a protein complex that contains E2F-1 and E2F-2. These data suggest that RING3 is a potentially important regulator of E2F-dependent cell cycle genes.


Subject(s)
Carrier Proteins , Cell Cycle Proteins/genetics , DNA-Binding Proteins , Protein Serine-Threonine Kinases/physiology , Transcription Factors/metabolism , 3T3 Cells , Animals , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone , Cyclins/genetics , Cyclins/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , Macromolecular Substances , Mice , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma-Binding Protein 1 , Time Factors , Transcription Factor DP1 , Transcriptional Activation
16.
J Cell Sci ; 113 ( Pt 17): 3085-91, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10934046

ABSTRACT

RING3 is a novel protein kinase linked to human leukaemia. Its Drosophila homologue female sterile homeotic is a developmental regulator that interacts genetically with trithorax, a human homologue of which is also associated with leukaemia. The RING3 structure contains two mutually related bromodomains that probably assist in the remodelling of chromatin and thereby affect transcription. Consistent with this hypothesis, a RING3-like protein has been identified in the mouse Mediator complex, where it is associated with transcription factors. We show that, whilst RING3 is constitutively localised to the nucleus of exponentially growing HeLa cells, it is delocalised throughout serum-starved fibroblasts. We use immunostaining and confocal microscopy to demonstrate that RING3 translocates to the fibroblast nucleus upon serum stimulation. After translocation, RING3 participates in nuclear protein complexes that include E2F proteins; it transactivates the promoters of several important mammalian cell cycle genes that are dependent on E2F, including dihydrofolate reductase, cyclin D1, cyclin A and cyclin E. We use site-directed mutagenesis of a putative nuclear localisation motif to show that the activation-induced nuclear localisation and consequent transcriptional activity of RING3 depends on a monopartite, classical nuclear localisation sequence. These observations refine and extend the mechanism by which RING3 contributes to E2F-regulated cell cycle progression. Deregulation of this mechanism may be leukaemogenic.


Subject(s)
Cell Nucleus/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Biological Transport/drug effects , Cell Line , Chromosomal Proteins, Non-Histone , Culture Media/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Nuclear Localization Signals , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Recombinant Proteins/metabolism , Transcription Factors , Transcription, Genetic , Transfection
17.
J Biol Chem ; 275(34): 26024-31, 2000 Aug 25.
Article in English | MEDLINE | ID: mdl-10856296

ABSTRACT

During previous studies, we found that mdm2 mRNA levels were elevated in benzo[a]pyrene (BaP, a polycyclic aryl hydrocarbon)-treated cells under conditions of DNA damage-induced cell cycle arrest (Vaziri, C., and Faller, D. V. (1997) J. Biol. Chem. 272, 2762-2769). We have identified potential aryl-hydrocarbon receptor-binding sites in the mdm2 promoter. However, we show that induction of mdm2 mRNA by BaP is entirely dependent upon aryl-hydrocarbon-induced genotoxicity and does not involve direct aryl-hydrocarbon receptor-mediated transcriptional activation of the mdm2 gene. Heterologous mdm2 promoter-reporter constructs containing p53-response elements were not responsive to BaP treatment. Therefore the p53-response elements in the mdm2 promoter are insufficient to confer DNA damage-dependent expression of mdm2. Furthermore, mdm2 transcripts were induced by BaP in p53 null cells from transgenic mice (although both basal and BaP-induced mdm2 expression levels were reduced in these cells relative to p53(+/+) cultures). These data show that p53-mediated mechanisms cannot account for BaP/DNA damage-induced mdm2 expression. Mdm2 promoter-reporter gene assays and nuclear run-off analyses of nascent mdm2 transcripts showed that transcriptional induction was unable to account for the large changes in mdm2 transcript levels following BaP treatment. However, mdm2 mRNA half-life measurements showed stabilization of the mdm2 transcript (from approximately 1 h to >4 h) in response to BaP. To our knowledge, this is the first report of control of mdm2 at the post-transcriptional level and in a p53-independent manner. Transient ectopic expression of mdm2 strongly augmented aryl-hydrocarbon-induced apoptosis, demonstrating that mdm2 levels can have a profound effect on the cellular response to DNA damage. Overall, our results suggest a potentially important link between DNA damage signaling and RNA stability that may be relevant to cell cycle regulation, tumor suppression, and environmental carcinogenesis.


Subject(s)
Benzo(a)pyrene/pharmacology , DNA Damage , Nuclear Proteins , Protein Processing, Post-Translational , Proto-Oncogene Proteins/biosynthesis , Tumor Suppressor Protein p53/metabolism , 3T3 Cells , Animals , Fibroblasts/metabolism , Half-Life , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2 , RNA, Messenger/metabolism , Rats , Transcription, Genetic
18.
Cancer Lett ; 154(2): 163-74, 2000 Jun 30.
Article in English | MEDLINE | ID: mdl-10806304

ABSTRACT

We have recently identified and characterized a new adhesion ligand, HAL1/13 (hypoxia-activated ligand), which mediates the increase in leukocyte adhesion to endothelium under hypoxic conditions (J. Immunol. 155 (1995) 802-810). The HAL1/13 antigen was cloned and found to be identical to p86 subunit of Ku autoantigen. In this study we demonstrate that exposure of neuroblastoma and breast carcinoma cells to hypoxia results in upregulation of HAL1/13 surface expression, coincident with an increased ability of these tumor cells to invade endothelial monolayers, which could be partially attenuated by the anti-HAL1/13 antibody. Hypoxia also potentiated neuroblastoma and breast carcinoma cell transmigration through Matrigel filters. Anti-HAL1/13 antibody inhibited haptotactic locomotion of hypoxic tumor cells on laminin.


Subject(s)
Antigens, Nuclear , Cell Hypoxia , DNA Helicases , DNA-Binding Proteins/physiology , Neoplasm Invasiveness , Nuclear Proteins/physiology , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Cell Adhesion , Cell Movement , Collagen/metabolism , DNA-Binding Proteins/metabolism , Drug Combinations , Endothelium, Vascular/metabolism , Extracellular Matrix/metabolism , Flow Cytometry , Humans , In Vitro Techniques , Ku Autoantigen , Laminin/metabolism , Leukocytes/metabolism , Ligands , Microscopy, Confocal , Models, Theoretical , Neuroblastoma/immunology , Neuroblastoma/metabolism , Nuclear Proteins/metabolism , Oxygen/metabolism , Proteoglycans/metabolism , Time Factors , Tumor Cells, Cultured , Umbilical Veins/metabolism , Up-Regulation
19.
J Biol Chem ; 275(3): 1715-22, 2000 Jan 21.
Article in English | MEDLINE | ID: mdl-10636867

ABSTRACT

The intracellular metabolism of many carcinogenic polycyclic aryl hydrocarbons (PAHs, typified by the ubiquitous pollutant benzo[a]pyrene or B[a]P) generates electrophilic products that react covalently with genomic DNA. Cells that acquire PAH-induced DNA damage undergo growth arrest in a p53-independent manner (Vaziri, C., and Faller, D. V. (1997) J. Biol. Chem. 272, 2762-2769). In this report we have investigated the molecular basis of PAH-induced cell cycle arrest. Mitogenic signaling events involving cyclins D and E, Rb phosphorylation, and transcriptional activation of E2F-responsive genes (including cyclin E and cyclin A) were unaffected in cells containing PAH-damaged DNA. However, PAH-induced growth arrest was associated with post-transcriptional decreases in cyclin A expression. Mitogen-induced expression of cyclin B, an event that is temporally distal to cyclin A expression, was also inhibited in PAH-treated cells. The PAH-induced cell cycle block was transient, and arrested cells resumed DNA synthesis after a prolonged ( approximately 20 h) delay. Resumption of DNA synthesis in PAH-treated cells occurred concomitant with elevated expression of cyclins A and B. PAH-induced cell cycle arrest was overcome by ectopically expressed cyclin A (encoded by a recombinant adenovirus in transiently infected cells). Overall, our results suggest the existence of a DNA damage checkpoint pathway that arrests cell cycle progression via post-transcriptional control of cyclin A expression.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Cyclin A/metabolism , DNA Damage , DNA-Binding Proteins , Gene Expression Regulation , RNA Processing, Post-Transcriptional , 3T3 Cells , Animals , Benzo(a)pyrene/toxicity , Cell Cycle/drug effects , Cyclin A/genetics , Cyclin D1/metabolism , E2F Transcription Factors , Flow Cytometry , Immunoblotting , Mice , Mitogens/metabolism , Mutagens/toxicity , Polycyclic Compounds/toxicity , Retinoblastoma Protein/metabolism , Retinoblastoma-Binding Protein 1 , Signal Transduction/drug effects , Transcription Factor DP1 , Transcription Factors/metabolism
20.
J Biol Chem ; 274(22): 15320-8, 1999 May 28.
Article in English | MEDLINE | ID: mdl-10336417

ABSTRACT

Activation of protein kinase C (PKC) can protect cells from apoptosis induced by various agents, including Fas ligation. To elucidate a possible interaction between Fas-mediated apoptotic signals and activation-related protective signals, we investigated the impact of Fas ligation on PKC activity. We demonstrate that engagement of Fas on human lymphoid Jurkat cells triggered apoptosis, and Fas ligation resulted in partial blockade of cellular PKC activity. The phorbol 12-myristate 13-acetate-mediated translocation of PKCtheta from the cytoplasm to the membrane was inhibited by treatment with anti-Fas antibody, whereas the translocation of PKCalpha or epsilon was not affected. In vitro kinase assay of PKCalpha or epsilon phosphotransferase activity demonstrated that Fas ligation inhibited the ability of PKCalpha to phosphorylate histone H1 as substrate but did not inhibit epsilon isozyme activity. This inhibition of PKCalpha activity mediated by Fas ligation was reversed by okadaic acid, a phosphatase inhibitor, suggesting the involvement of a member of the protein phosphatase 2A subfamily in this component of Fas signaling. Identical patterns of PKC isozyme inhibition were obtained using mouse thymoma cells overexpressing the fas gene (LF(+)). These results suggest that the selective inhibition of a potentially protective, PKC-mediated pathway by Fas activation may, to some extent, contribute to Fas-induced apoptotic signaling.


Subject(s)
Isoenzymes/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Protein Kinase C/antagonists & inhibitors , Animals , Antibodies/pharmacology , Apoptosis , Cell Division , Enzyme Activation , Enzyme Inhibitors/pharmacology , Fas Ligand Protein , Humans , Isoenzymes/metabolism , Jurkat Cells , Kinetics , Membrane Glycoproteins/immunology , Mice , Okadaic Acid/pharmacology , Phosphorylation , Protein Kinase C/metabolism , Protein Kinase C-alpha , Protein Kinase C-epsilon , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...