Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Cells ; 10(4)2021 04 19.
Article in English | MEDLINE | ID: mdl-33921698

ABSTRACT

MRCKα is a ubiquitously expressed serine/threonine kinase involved in cell contraction and F-actin turnover, which is highly amplified in human breast cancer and part of a gene expression signature for bad prognosis. Nothing is known about the in vivo function of MRCKα. To explore MRCKα function in development and in breast cancer, we generated mice lacking a functional MRCKα gene. Mice were born close to the Mendelian ratio and showed no obvious phenotype including a normal mammary gland formation. Assessing breast cancer development using the transgenic MMTV-PyMT mouse model, loss of MRCKα did not affect tumor onset, tumor growth and metastasis formation. Deleting MRCKα and its related family member MRCKß in two triple-negative breast cancer cell lines resulted in reduced invasion of MDA-MB-231 cells, but did not affect migration of 4T1 cells. Further genomic analysis of human breast cancers revealed that MRCKα is frequently co-amplified with the oncogenes ARID4B and AKT3 which might contribute to the prognostic value of MRCKα expression. Collectively, these data suggest that MRCKα might be a prognostic marker for breast cancer, but probably of limited functional importance.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Carcinogenesis/pathology , Mammary Neoplasms, Animal/metabolism , Mammary Tumor Virus, Mouse/physiology , Myotonin-Protein Kinase/metabolism , Protein Serine-Threonine Kinases/metabolism , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Animals , Antigens, Neoplasm/metabolism , Base Sequence , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Collagen/pharmacology , Disease Models, Animal , Female , Gels/pharmacology , Humans , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/genetics , Mammary Tumor Virus, Mouse/drug effects , Mice , Mice, Knockout , Mutation/genetics , Myosins/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/metabolism , Phenotype , Phosphorylation/drug effects , Polymerization/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Triple Negative Breast Neoplasms/pathology
2.
Antiviral Res ; 134: 192-206, 2016 10.
Article in English | MEDLINE | ID: mdl-27633452

ABSTRACT

Replication of the human immunodeficiency virus type 1 (HIV-1) is dependent on eIF5A hypusination. Hypusine is formed post-translationally on the eIF5A precursor by two consecutive enzymatic steps; a reversible reaction involving the enzyme deoxyhypusine synthase (DHS) and an irreversible step involving the enzyme deoxyhypusine hydroxylase (DOHH). In this study we explored the effect of inhibiting DOHH activity and therefore eIF5A hypusination, on HIV-1 gene expression. Results show that the expression of proteins from an HIV-1 molecular clone is reduced when DOHH activity is inhibited by Deferiprone (DFP) or Ciclopirox (CPX). Next we evaluated the requirement of DOHH activity for internal ribosome entry site (IRES)-mediated translation initiation driven by the 5'untranslated region (5'UTR) of the full length HIV-1 mRNA. Results show that HIV-1 IRES activity relies on DOHH protein concentration and enzymatic activity. Similar results were obtained for IRES-dependent translation initiation mediated by 5'UTR of the human T-cell lymphotropic virus type 1 (HTLV-1) and the mouse mammary tumor virus (MMTV) mRNAs. Interestingly, activity of the poliovirus IRES, was less sensitive to the targeting of DOHH suggesting that not all viral IRESs are equally dependent on the cellular concentration or the activity of DOHH. In summary we present evidence indicating that the cellular concentration of DOHH and its enzymatic activity play a role in HIV-1, HTLV-1 and MMTV IRES-mediated translation initiation.


Subject(s)
5' Untranslated Regions , HIV-1/genetics , HIV-1/physiology , Human T-lymphotropic virus 1/genetics , Mammary Tumor Virus, Mouse/genetics , Mixed Function Oxygenases/antagonists & inhibitors , Animals , Ciclopirox , Deferiprone , Gene Expression , HEK293 Cells , HIV-1/drug effects , HeLa Cells , Humans , Mammary Tumor Virus, Mouse/drug effects , Mice , Mixed Function Oxygenases/drug effects , Peptide Initiation Factors/drug effects , Protein Biosynthesis/drug effects , Pyridones/pharmacology , RNA, Messenger/drug effects , RNA-Binding Proteins/drug effects , Eukaryotic Translation Initiation Factor 5A
3.
Liver Int ; 35(4): 1442-50, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25302564

ABSTRACT

BACKGROUND & AIMS: The NOD.c3c4 mouse model develops autoimmune biliary disease characterized by spontaneous granulomatous cholangitis, antimitochondrial antibodies and liver failure. This model for primary biliary cirrhosis (PBC) has evidence of biliary infection with mouse mammary tumour virus (MMTV), suggesting that the virus may have a role in cholangitis development and progression of liver disease in this mouse model. We tested the hypothesis that MMTV infection is associated with cholangitis in the NOD.c3c4 mouse model by investigating whether antiretroviral therapy impacts on viral levels and liver disease. METHODS: NOD.c3c4 mice were treated with combination antiretroviral therapy. Response to treatment was studied by measuring MMTV RNA in the liver, liver enzyme levels in serum and liver histology using a modified Ishak score. RESULTS: Combination therapy with the reverse transcriptase inhibitors, tenofovir and emtricitabine, resulted in a significant reduction in serum liver enzyme levels, attenuation of cholangitis and decreased MMTV levels in the livers of NOD.c3c4 mice. Furthermore, treatment with the retroviral protease inhibitors, lopinavir and ritonavir, in addition to the reverse transcriptase inhibitors, resulted in further decrease in MMTV levels and attenuation of liver disease in this model. CONCLUSIONS: The attenuation of cholangitis with regimens containing the reverse transcriptase inhibitors, tenofovir and emtricitabine, and the protease inhibitors, lopinavir and ritonavir, suggests that retroviral infection may play a role in the development of cholangitis in this model.


Subject(s)
Anti-Retroviral Agents/pharmacology , Cholangitis/drug therapy , Liver Cirrhosis, Biliary/drug therapy , Mammary Tumor Virus, Mouse/drug effects , Retroviridae Infections/drug therapy , Tumor Virus Infections/drug therapy , Amino Acid Sequence , Animals , Biomarkers/blood , Cholangitis/blood , Cholangitis/immunology , Cholangitis/virology , Disease Models, Animal , Drug Combinations , Drug Therapy, Combination , Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination/pharmacology , Female , Lamivudine/pharmacology , Liver Cirrhosis, Biliary/blood , Liver Cirrhosis, Biliary/immunology , Liver Cirrhosis, Biliary/virology , Lopinavir/pharmacology , Mammary Tumor Virus, Mouse/enzymology , Mammary Tumor Virus, Mouse/genetics , Mammary Tumor Virus, Mouse/pathogenicity , Mice, Inbred NOD , Molecular Sequence Data , Protease Inhibitors/pharmacology , RNA, Viral/blood , Retroviridae Infections/blood , Retroviridae Infections/immunology , Retroviridae Infections/virology , Reverse Transcriptase Inhibitors/pharmacology , Ritonavir/pharmacology , Time Factors , Tumor Virus Infections/blood , Tumor Virus Infections/immunology , Tumor Virus Infections/virology , Viral Load , Zidovudine/pharmacology
4.
J Int Med Res ; 40(6): 2073-82, 2012.
Article in English | MEDLINE | ID: mdl-23321163

ABSTRACT

OBJECTIVE: Soy isoflavone is associated with modification of breast cancer risk. Effects of dietary isoflavone on breast tissue carcinogenesis under varying endogenous oestrogen contexts were investigated. METHODS: Five-week-old mouse mammary tumour virus (MMTV)-erbB2 female transgenic mice (n = 180) were divided into three equal groups: low-, normal- and high-oestrogen groups. Each group was then subdivided into an experimental group (given soybean feed) and a control group (given control feed). RESULTS: In the high-oestrogen environment, breast cancer incidence was significantly lower in the experimental versus the control group, whereas in the low-oestrogen environment, breast cancer incidence was significantly higher in the experimental versus the control group. There were no between-group differences in mean breast tumour latency, mean largest tumour diameter and breast tumour tissue vascular endothelial growth factor levels. CONCLUSIONS: Dietary soy isoflavones promote breast cancer at low oestrogen levels but inhibit breast cancer at high oestrogen levels. This effect may only occur during the initiation stage of breast cancer.


Subject(s)
Estrogens/pharmacology , Genes, erbB-2/genetics , Isoflavones/pharmacology , Mammary Neoplasms, Animal/diet therapy , Mammary Tumor Virus, Mouse/genetics , Animals , Diet , Estrogens/metabolism , Female , Isoflavones/administration & dosage , Mammary Neoplasms, Animal/virology , Mammary Tumor Virus, Mouse/drug effects , Mice , Mice, Transgenic , Ovariectomy , Soybean Proteins/pharmacology , Glycine max , Vascular Endothelial Growth Factor A/analysis
5.
J Biomed Biotechnol ; 2011: 416905, 2011.
Article in English | MEDLINE | ID: mdl-21253530

ABSTRACT

Histone deacetylases (HDACs) have been shown to be required for basal or inducible transcription at a variety of genes by poorly understood mechanisms. We demonstrated previously that HDAC inhibition rapidly repressed transcription from the mouse mammary tumor virus (MMTV) promoter by a mechanism that does not require the binding of upstream transcription factors. In the current study, we find that HDACs work through the core promoter sequences of MMTV as well as those of several cellular genes to facilitate transcriptional initiation through deacetylation of nonhistone proteins.


Subject(s)
Histone Deacetylases/metabolism , Mammary Tumor Virus, Mouse/genetics , Promoter Regions, Genetic , Animals , Cell Line, Tumor , HeLa Cells , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Mammary Tumor Virus, Mouse/drug effects , Mice , Models, Genetic , RNA Polymerase II/metabolism , TATA Box/genetics , Transcription, Genetic/drug effects
6.
Liver Int ; 30(6): 871-7, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20492501

ABSTRACT

BACKGROUND/AIM: Up to one-third of patients with primary biliary cirrhosis (PBC) experience recurrent disease following liver transplantation, which is associated with earlier and more severe recurrence in patients treated with tacrolimus as compared with cyclosporine A (CsA). As the latter has known antiviral activity, we hypothesized that CsA has the ability to inhibit the betaretrovirus characterized from patients with PBC. METHODS: We investigated whether CsA, the cyclosporine analogue NIM811, tacrolimus and other compounds can modulate the mouse mammary tumour virus production from Mm5MT cells. Viral load was evaluated in the cell supernatants by quantifying reverse transcriptase (RT) levels and betaretrovirus RNA. RESULTS: A significant correlation was observed with increasing concentrations of CsA and NIM811, and decreasing of RT levels (rho-0.59, P=0.04 and rho-0.74, P=0.006 respectively), whereas tacrolimus had no significant effect (rho-0.27, P=0.4). At a dose of 3 microg/ml, CsA, NIM811 and the human immunodeficiency virus aspartyl protease inhibitor, lopinavir, were all associated with greater than three-fold reduction in the betaretrovirus RNA production from Mm5MT cells as compared with tacrolimus (P<0.005). CONCLUSIONS: These studies demonstrate that the cyclophilin inhibitors CsA and NIM811 have antiviral activity against betaretrovirus production in vitro.


Subject(s)
Antiviral Agents/pharmacology , Cyclosporine/pharmacology , Immunosuppressive Agents/pharmacology , Liver Cirrhosis, Biliary/virology , Mammary Tumor Virus, Mouse/drug effects , Virus Replication/drug effects , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Liver Cirrhosis, Biliary/surgery , Liver Transplantation , Lopinavir , Mammary Tumor Virus, Mouse/genetics , Mammary Tumor Virus, Mouse/growth & development , Mice , Pyrimidinones/pharmacology , RNA, Viral/biosynthesis , RNA-Directed DNA Polymerase/metabolism , Recurrence , Tacrolimus/pharmacology , Viral Load
7.
Nucl Recept Signal ; 7: e008, 2009 Oct 02.
Article in English | MEDLINE | ID: mdl-20087429

ABSTRACT

Transcription from the mouse mammary tumor virus (MMTV) promoter can be induced by progestins. The progesterone receptor (PR) binds to a cluster of five hormone responsive elements (HREs) and activates the promoter by synergistic interactions with the ubiquitous transcription factor, nuclear factor 1 (NF1). Progesterone treatment of cells in culture leads to activation of the Src/Ras/Erk/Msk1 cascade. Selective inhibition of Erk, or its target kinase Msk1, interferes with chromatin remodeling and blocks MMTV activation. A complex of activated PR, Erk and Msk1 is recruited to promoter after 5 min of hormone treatment and phosphorylates histone H3 at serine 10. This modification promotes the displacement of HP1gamma and subsequent chromatin remodeling. Progestin treatment leads to the recruitment of the BAF complex, which selectively displaces histones H2A and H2B from the nucleosome containing the HREs. The acetyltransferase PCAF is also required for induction of progesterone target genes and acetylates histone H3 at K14, an epigenetic mark, which interacts with Brg1 and Brm, anchoring the BAF complex to chromatin. In nucleosomes assembled on either MMTV or mouse rDNA promoter sequences, SWI/SNF displaces histones H2A and H2B from MMTV, but not from the rDNA nucleosome. Thus, the outcome of nucleosome remodeling by purified SWI/SNF depends on DNA sequence. The resultant H3/H4 tetramer particle is then the substrate for subsequent events in induction. Thus, initial activation of the MMTV promoter requires activation of several kinases and PCAF leading to phosphoacetylation of H3, and recruitment of BAF with subsequent removal of H2A/H2B.


Subject(s)
Chromatin/metabolism , Extracellular Signal-Regulated MAP Kinases/physiology , Gene Expression Regulation, Viral/drug effects , Genes, Viral/drug effects , Genes, Viral/genetics , Mammary Tumor Virus, Mouse/drug effects , Mammary Tumor Virus, Mouse/genetics , Progestins/pharmacology , Animals , Histones/metabolism , Humans
8.
J Nutr ; 138(12): 2316-22, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19022951

ABSTRACT

Transposable elements such as long terminal repeats (LTR) constitute approximately 45% of the human genome; transposition events impair genome stability. Fifty-four promoter-active retrotransposons have been identified in humans. Epigenetic mechanisms are important for transcriptional repression of retrotransposons, preventing transposition events, and abnormal regulation of genes. Here, we demonstrate that the covalent binding of the vitamin biotin to lysine-12 in histone H4 (H4K12bio) and lysine-9 in histone H2A (H2AK9bio), mediated by holocarboxylase synthetase (HCS), is an epigenetic mechanism to repress retrotransposon transcription in human and mouse cell lines and in primary cells from a human supplementation study. Abundance of H4K12bio and H2AK9bio at intact retrotransposons and a solitary LTR depended on biotin supply and HCS activity and was inversely linked with the abundance of LTR transcripts. Knockdown of HCS in Drosophila melanogaster enhances retrotransposition in the germline. Importantly, we demonstrated that depletion of H4K12bio and H2AK9bio in biotin-deficient cells correlates with increased production of viral particles and transposition events and ultimately decreases chromosomal stability. Collectively, this study reveals a novel diet-dependent epigenetic mechanism that could affect cancer risk.


Subject(s)
DNA Transposable Elements , Histones/chemistry , Histones/metabolism , Adult , Animals , Biotin/administration & dosage , Biotinylation , Carbon-Nitrogen Ligases/antagonists & inhibitors , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Cell Line , Chromosome Aberrations , Cytosine/metabolism , DNA Methylation , Dietary Supplements , Drosophila melanogaster , Epigenesis, Genetic , Female , Humans , Jurkat Cells , Male , Mammary Tumor Virus, Mouse/drug effects , Mammary Tumor Virus, Mouse/physiology , Mice , Middle Aged , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Terminal Repeat Sequences , Transcription, Genetic/drug effects , Virus Assembly/drug effects , Young Adult
9.
Retrovirology ; 4: 73, 2007 Oct 11.
Article in English | MEDLINE | ID: mdl-17931409

ABSTRACT

BACKGROUND: The role of mouse mammary tumor virus (MMTV) as a causative agent in human breast carcinogenesis has recently been the subject of renewed interest. The proposed model is based on the detection of MMTV sequences in human breast cancer but not in healthy breast tissue. One of the main drawbacks to this model, however, was that until now human cells had not been demonstrated to sustain productive MMTV infection. RESULTS: Here, we show for the first time the rapid spread of mouse mammary tumor virus, MMTV(GR), in cultured human mammary cells (Hs578T), ultimately leading to the infection of every cell in culture. The replication of the virus was monitored by quantitative PCR, quantitative RT-PCR and immunofluorescence imaging. The infected human cells expressed, upon cultivation with dexamethasone, MMTV structural proteins and released spiked B-type virions, the infectivity of which could be neutralized by anti-MMTV antibody. Replication of the virus was efficiently blocked by an inhibitor of reverse transcription, 3'-azido-3'-deoxythymidine. The human origin of the infected cells was confirmed by determining a number of integration sites hosting the provirus, which were unequivocally identified as human sequences. CONCLUSION: Taken together, our results show that human cells can support replication of mouse mammary tumor virus.


Subject(s)
Breast Neoplasms/virology , Mammary Tumor Virus, Mouse/drug effects , Animals , Cell Line , DNA, Viral/analysis , DNA, Viral/genetics , Humans , Mammary Tumor Virus, Mouse/genetics , Mammary Tumor Virus, Mouse/physiology , Polymerase Chain Reaction/methods , Tumor Virus Infections/immunology , Virus Replication/drug effects , Zidovudine/pharmacology
10.
Toxicol Appl Pharmacol ; 211(1): 11-9, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-15972221

ABSTRACT

In reporter assays for detection of dioxins, the dioxin-responsive element (DRE) is generally used as a sensor sequence. In several systems, the CYP1A1 promoter containing DREs (DRE(cyp)) is inserted into a part of the long terminal repeat of mouse mammary tumor virus (LTR(MMTV)) to improve sensitivity of assays. We found that DRE(cyp)-LTR(MMTV) responds not only to dioxins and dioxin-like compounds but also to forskolin, a cAMP-elevating agent. This effect was dose-dependent and reproduced by other cAMP-elevating agents including 8-bromo-cAMP and 3-isobutyl-methylxanthine. The cAMP response element (CRE) and CRE-like sequences were absent in DRE(cyp)-LTR(MMTV) and not involved in this process. In contrast to the effect of dioxin, the activation of DRE(cyp)-LTR(MMTV) by cAMP was independent of the aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor for DRE. Furthermore, neither DRE(cyp), LTR(MMTV) nor the consensus sequence of DRE alone was activated in response to cAMP. These data elucidated for the first time that the combination of DRE(cyp) with LTR(MMTV) causes a peculiar response to cAMP and suggested that use of AhR antagonists is essential to exclude false-positive responses of DRE(cyp)-LTR(MMTV)-based bioassays for detection and quantification of dioxins and dioxin-like compounds.


Subject(s)
Biological Assay/methods , Cyclic AMP/pharmacology , Gene Expression Regulation/drug effects , Genes, Reporter/drug effects , Promoter Regions, Genetic/drug effects , Animals , Base Sequence , Cell Line, Tumor , Colforsin/pharmacology , Consensus Sequence/genetics , Cytochrome P-450 CYP1A1/genetics , Dioxins/analysis , Dose-Response Relationship, Drug , Environmental Pollutants/analysis , Enzyme Activators/pharmacology , False Positive Reactions , Hepatocytes , Mammary Tumor Virus, Mouse/drug effects , Mammary Tumor Virus, Mouse/genetics , Mice , Molecular Sequence Data , Receptors, Aryl Hydrocarbon/drug effects , Response Elements/drug effects , Response Elements/genetics , Terminal Repeat Sequences/drug effects , Terminal Repeat Sequences/genetics , Transfection
11.
Ann N Y Acad Sci ; 1089: 59-72, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17261755

ABSTRACT

Transcription from the mouse mammary tumor virus (MMTV) promoter can be induced by glucocorticoids or progestins. Progesterone treatment of cultured cells carrying an integrated single copy of an MMTV transgene leads to recruitment of progesterone receptor (PR), SWI/SNF, and SNF2h-related complexes to MMTV promoter. Recruitment is accompanied by selective displacement of histones H2A and H2B from the nucleosome B. In nucleosomes assembled on promoter sequences, SWI/SNF displaces histones H2A and H2B from MMTV nucleosome B, but not from other MMTV nucleosomes or from an rDNA promoter nucleosome. Thus, the outcome of nucleosome remodeling by purified SWI/SNF depends on the DNA sequence. On the other hand, 5 min after hormone treatment, the cytoplasmic signaling cascade Src/Ras/Erk is activated via an interaction of PR with the estrogen receptor, which activates Src. As a consequence of Erk activation PR is phosphorylated, Msk1 is activated, and a ternary complex PR-Erk-Msk1 is recruited to MMTV nucleosome B. Msk1 phosphorylates H3 at serine 10, which is followed by acetylation at lysine 14, displacement of HP1gamma, and recruitment of Brg1, PCAF, and RNA polymerase II. Blocking Erk activation or Msk1 activity prevents induction of the MMTV transgene. Thus, the rapid nongenomic effects of progestins are essential for their transcriptional effects on certain progestin target genes. In rat endometrial stromal cells, picomolar concentrations of progestins trigger the cross talk of PR with ERbeta that activates the Erk and Akt kinase pathways leading to cell proliferation in the absence of direct transcriptional effects of the ligand-activated PR. Thus, depending on the cellular context rapid kinase activation and transcriptional effect play different roles in the physiological response to progestins.


Subject(s)
Cell Proliferation/drug effects , Chromatin Assembly and Disassembly , Gene Expression Regulation , Mammary Tumor Virus, Mouse/drug effects , Progestins/pharmacology , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Animals , Mammary Tumor Virus, Mouse/genetics , Mice , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Protein Kinases/metabolism , Rats , Signal Transduction
12.
Biochem J ; 367(Pt 2): 381-91, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12097136

ABSTRACT

Phe-119 in the reverse transcriptase (RT) of mouse mammary tumour virus (MMTV) is homologous with Tyr-115 in HIV type 1 (HIV-1) RT and to Phe-155 in murine leukaemia virus (MLV) RT. By mutating these residues in HIV-1 and MLV RTs (which are strict DNA polymerases) the enzymes were shown to function also as RNA polymerases. Owing to the uniqueness of MMTV as a type B retrovirus, we have generated a Phe-119-Val mutant of MMTV RT to study the involvement of this residue in affecting the catalytic features of this RT. The data presented here show that the mutant MMTV RT can incorporate both deoxyribonucleosides and ribonucleosides while copying either RNA or DNA. In addition, this mutant RT shows resistance to nucleoside analogues and an enhanced fidelity of DNA synthesis; all relative to the wild-type enzyme. The Phe-119-Val mutant is also different from the wild-type enzyme in its preference for most template primers tested and in its ability to synthesize DNA under non-processive and processive conditions. Overall, it is likely that the aromatic side chain of Phe-119 is located at the dNTP-binding site of MMTV RT and thus might be part of a putative "steric gate" that prevents the incorporation of nucleoside triphosphates. Since the only three-dimensional structures of RTs published so far are those of HIV-1 and MLV, it is likely that MMTV RT folds quite similarly to these RTs.


Subject(s)
DNA, Viral/biosynthesis , Mammary Tumor Virus, Mouse/metabolism , Phenylalanine , RNA-Directed DNA Polymerase/genetics , RNA-Directed DNA Polymerase/metabolism , Ribose/metabolism , Binding Sites , Drug Resistance, Viral , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , Kinetics , Mammary Tumor Virus, Mouse/drug effects , Mammary Tumor Virus, Mouse/genetics , Nucleosides/pharmacology , Point Mutation , RNA, Viral/biosynthesis , RNA-Directed DNA Polymerase/drug effects , Ribonuclease H/metabolism , Ribonucleosides/metabolism , Valine
13.
Virology ; 275(1): 98-106, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-11017791

ABSTRACT

Expression of mouse mammary tumor virus (MMTV)-encoded superantigens in B lymphocytes are required for viral transmission and pathogenesis. Due to problems with detection and quantification of the superantigen protein, most reports about the mechanism of superantigen expression from the viral sag gene rely on the quantitative analysis of putative sag mRNAs. The description of multiple promoters as a source of putative sag mRNA has complicated the situation even further. All conclusions about the level of superantigen protein expression based on these data remain circumstantial. To test the effect of the glucocorticoid hormone dexamethasone on the total superantigen expression from an infectious MMTV provirus we used a quantitative assay that is based on a superantigen-luciferase fusion protein. MMTV gene expression from the major promoter in the 5' long terminal repeat (LTR) is strongly induced in the presence of glucocorticoid hormones. We now demonstrate that, in the presence of dexamethasone, sag gene expression is reduced despite increased transcription from the MMTV 5' LTR and increased amounts of putative sag mRNA initiated at the LTR promoter. These data show that the expression of the MMTV sag gene does not correlate with the activity of the major LTR promoter and thus differs from all other MMTV genes.


Subject(s)
Gene Expression Regulation, Viral/drug effects , Genes, Viral/genetics , Glucocorticoids/pharmacology , Mammary Tumor Virus, Mouse/drug effects , Mammary Tumor Virus, Mouse/immunology , Superantigens/genetics , Animals , Cell Line , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Genes, Reporter/genetics , Glucocorticoids/therapeutic use , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/virology , Mammary Tumor Virus, Mouse/genetics , Mice , Promoter Regions, Genetic/genetics , Proviruses/drug effects , Proviruses/genetics , Proviruses/immunology , RNA, Messenger/analysis , RNA, Messenger/genetics , RNA, Viral/analysis , RNA, Viral/genetics , Recombinant Fusion Proteins/genetics , Terminal Repeat Sequences/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Transfection
14.
Exp Cell Res ; 260(1): 160-5, 2000 Oct 10.
Article in English | MEDLINE | ID: mdl-11010820

ABSTRACT

Inhibition of the ligand-activated androgen receptor (AR) by antiandrogens plays an important role in the treatment of various hyperandrogenic disorders including prostate cancer. However, the molecular mechanisms of antiandrogen activity in vivo remain unclear. In this study we analyzed the effects of cyproterone acetate (CPA), flutamide (F), and hydroxyflutamide (OHF) on transcriptional activation and chromatin remodeling of the genomically integrated mouse mammary tumor virus (MMTV) promoter. This promoter has provided an excellent model system to study the impact of steroid hormones on transcriptional activation in the context of a defined chromatin structure. The MMTV hormone response element is positioned on a phased nucleosome, which becomes remodeled in response to steroids. We utilized this model system in mouse L-cell fibroblasts that contain a stably integrated MMTV promoter. In these cells, dihydrotestosterone (DHT) induced a large increase of AR protein levels that correlated with transcriptional activation and chromatin remodeling of the MMTV promoter. Coadministration of DHT and CPA or DHT and OHF in these cells inhibited the increase of AR levels, which resulted in a strong blockage of transcriptional activation and chromatin remodeling of the MMTV promoter. In contrast, F had no significant influence on these activities. We conclude that a major portion of the antiandrogenic effects of CPA and OHF in vivo are mediated by the reduction of AR levels.


Subject(s)
Androgen Antagonists/pharmacology , Chromatin/drug effects , Mammary Tumor Virus, Mouse/drug effects , Mammary Tumor Virus, Mouse/genetics , Promoter Regions, Genetic/drug effects , Animals , Base Sequence , Cyproterone Acetate/pharmacology , DNA Probes/genetics , Female , Flutamide/analogs & derivatives , Flutamide/pharmacology , L Cells , Male , Mice , Receptors, Androgen/drug effects , Receptors, Androgen/metabolism , Transcriptional Activation/drug effects
15.
J Virol ; 72(12): 9428-35, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9811675

ABSTRACT

Integration of mouse mammary tumor virus (MMTV) near the int genes results in the inappropriate expression of these proto-oncogenes and initiates events that lead to the formation of mammary adenocarcinomas. In most cases, the MMTV provirus integrates in a transcriptional orientation opposite that of the int genes. We have used a novel, vector-based system designed to recapitulate the integration of MMTV upstream of the int-2 promoter. Compared to a cellular promoter or another retroviral promoter, the MMTV long terminal repeat (LTR) in this configuration is particularly efficacious at activating the int-2 promoter. The sequences responsible for enhancing the activity of the int-2 promoter map to two domains in the 5' end of the MMTV LTR. One domain is a previously defined element; the second is an element delineated by these studies that acts synergistically with the first. Both of these elements display mammary cell-specific activity. Thus, even though the MMTV promoter itself is weak without hormonal stimulation, viral integration can position the 5' LTR elements to efficiently activate transcription from cellular proto-oncogenes. Other functional elements in the LTR have little effect on the activation of the int-2 promoter. Even stimulation of the MMTV promoter with steroid hormones only modestly activates transcription from the int-2 promoter, suggesting that the 5' elements of the LTR are the predominant determinants of the tissue- and orientation-specific activation of cellular promoters by MMTV.


Subject(s)
Mammary Tumor Virus, Mouse/genetics , Mammary Tumor Virus, Mouse/pathogenicity , Proto-Oncogenes , Animals , Base Sequence , Cell Line , DNA/genetics , Enhancer Elements, Genetic , Female , Fibroblast Growth Factor 3 , Fibroblast Growth Factors/genetics , Gene Expression Regulation , Genetic Vectors , HeLa Cells , Humans , Mammary Neoplasms, Experimental/etiology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/virology , Mammary Tumor Virus, Mouse/drug effects , Mice , Promoter Regions, Genetic/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogenes/drug effects , Proviruses/genetics , Retroviridae Infections/etiology , Retroviridae Infections/genetics , Retroviridae Infections/virology , Steroids/pharmacology , Terminal Repeat Sequences , Transfection , Tumor Virus Infections/etiology , Tumor Virus Infections/genetics , Tumor Virus Infections/virology , Virus Integration/genetics
16.
J Biol Chem ; 272(35): 21803-10, 1997 Aug 29.
Article in English | MEDLINE | ID: mdl-9268310

ABSTRACT

We have studied the transcriptional behavior of the mouse mammary tumor virus long repeat (MMTV-LTR) promoter during a prolonged exposure to glucocorticoids. When integrated into XC-derived cells, MMTV-LTR expression reached its maximum during the first day of dexamethasone treatment, but longer exposure to the hormone resulted in the deactivation of the promoter. In contrast, glucocorticoid-responsive resident genes or MMTV-based transiently transfected plasmids maintained or even increased their mRNA levels during the same period of hormone treatment. An integrated chimeric construct containing the hormone-responsive elements from MMTV-LTR but in different sequence context became also deactivated after a prolonged hormone treatment but with a deactivation kinetics significantly slower than constructs containing the entire, chromatin-positioning MMTV-LTR sequence. The decrease on MMTV-LTR-driven transcription was concomitant with a parallel closure of the MMTV-LTR chromatin and with a decrease in glucocorticoid receptor (GR) concentration in the cell. We concluded that the chromatin-organized MMTV-LTR promoter is particularly sensitive to any decrease on GR levels. We propose that chromatin structure may contribute decisively to the differential expression of MMTV-LTR by two mechanisms: limiting MMTV-LTR accessibility to activating transcription factors and accelerating its shutting down upon a decrease on GR levels.


Subject(s)
Glucocorticoids/pharmacology , Mammary Tumor Virus, Mouse/genetics , Promoter Regions, Genetic , Repetitive Sequences, Nucleic Acid/genetics , Animals , Blotting, Western , Cells, Cultured , Chromatin/metabolism , Cricetinae , Deoxyribonuclease I/metabolism , Genes, Reporter , Kinetics , Mammary Tumor Virus, Mouse/drug effects , Mice , Promoter Regions, Genetic/drug effects , RNA, Messenger/metabolism , Receptors, Glucocorticoid/metabolism , Repetitive Sequences, Nucleic Acid/drug effects , Transfection
17.
Cancer Immunol Immunother ; 44(5): 249-56, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9247559

ABSTRACT

We have previously developed an experimental model for the xenogenization of malignant lymphoma. From highly tumorigenic S49 mouse lymphoma cells that proliferate in suspension culture (designated T-25), we selected variant clones that grew as an adherent monolayer (designated T-25-Adh) and were non-tumorigenic in syngeneic mice. Furthermore, priming of syngeneic hosts with T-25-Adh cells protected them against subsequent challenges with the tumorigenic T-25 cells. Several lines of evidence have indicated that antigens of an endogenous mouse mammary tumor virus (MMTV) are involved in the immunogenicity of T-25-Adh cells. Since interferon (IFN) is known to affect retroviral assembly and maturation on the cell membrane, we have studied the effects of IFN on endogenous MMTV-related structures, as well as on the immunogenicity of T-25-Adh cells. We observed that mouse alpha and beta interferons affect the morphogenesis of intracellular MMTV-related precursors in the immunogenic T-25-Adh cells, but not in tumorigenic T-25 cells. From T-25-Adh cells we selected variants that were either high responders or low responders to the above-mentioned interferon effect. The high-response variants were significantly more protective against tumorigenic T-25 cells than the low-response variants. Involvement of MMTV-related antigens in the immune response of the host to T-25-Adh cells was further suggested by immunoelectron-microscopical analysis, demonstrating that antisera from mice, immunized with T-25-Adh cells, interacted specifically with cell-surface MMTV budding particles. These findings indicate a novel method for xenogenization of lymphoma cells by IFN. Since endogenous retroviruses are present in all tissues of the mouse, this approach might be applicable to a wide variety of tumors.


Subject(s)
Cancer Vaccines/immunology , Lymphoma/prevention & control , Animals , Antigenic Variation , Cancer Vaccines/genetics , Interferons/pharmacology , Lymphoma/genetics , Lymphoma/immunology , Male , Mammary Tumor Virus, Mouse/drug effects , Mice , Mice, Inbred BALB C , Transplantation, Isogeneic , Vaccines, Attenuated
18.
Biochem Biophys Res Commun ; 210(2): 329-37, 1995 May 16.
Article in English | MEDLINE | ID: mdl-7755607

ABSTRACT

Oenothein B, a macrocircular dimeric ellagitannin, was found to be a potent and specific inhibitor of poly(ADP-ribose) glycohydrolase. Oenothein B suppressed glucocorticoid-sensitive mouse mammary tumor virus (MMTV) transcription in 34I cells. This suppression was accompanied by inhibition of glucocorticoid-induced endogeneous de-poly(ADP-ribosyl)ation of high mobility group (HMG) 14 and 17 proteins. These results suggest that de-poly(ADP-ribosyl)ation of these proteins may be closely connected with the events initiating glucocorticoid-sensitive MMTV gene transcription.


Subject(s)
Gene Expression Regulation, Viral/drug effects , Glycoside Hydrolases/antagonists & inhibitors , Hydrolyzable Tannins , Mammary Tumor Virus, Mouse/drug effects , Tannins/pharmacology , Animals , High Mobility Group Proteins/metabolism , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Inbred C3H , RNA, Messenger/genetics , Tumor Cells, Cultured
19.
Proc Natl Acad Sci U S A ; 92(6): 2076-80, 1995 Mar 14.
Article in English | MEDLINE | ID: mdl-7892227

ABSTRACT

The anticancer drug cis-diamminedichloro-platinum(II) (cisplatin) covalently modifies DNA, and these lesions are thought to lead to cell death by inhibiting DNA and RNA synthesis. By using in vivo analysis techniques, we have investigated the influence of cisplatin on hormone-induced transcription from the mouse mammary tumor virus (MMTV) promoter. Cisplatin substantially reduced glucocorticoid-induced expression from the MMTV promoter stably incorporated into mouse tumor cells. The glucocorticoid-receptor-dependent chromatin remodeling and loading of transcription factors that is a signature response of this promoter in the context of chromatin were significantly reduced by cisplatin but not by the clinically ineffective trans-isomer trans-diamminedichloroplatinum(II) (transplatin). Additional in vivo studies on transiently introduced nonchromatin MMTV templates demonstrated that cisplatin modification of DNA blocked binding of the transcription factor NF1. These results provide strong evidence that cisplatin influences transcription by interfering with the opening of repressive chromatin structures and by blocking transcription factor binding directly, each of which could contribute substantially to its toxicity.


Subject(s)
Chromatin/drug effects , Cisplatin/pharmacology , Mammary Tumor Virus, Mouse/genetics , Promoter Regions, Genetic/drug effects , Repetitive Sequences, Nucleic Acid , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Animals , Base Sequence , Cell Line , Chloramphenicol O-Acetyltransferase/biosynthesis , Chromatin/ultrastructure , DNA, Viral/isolation & purification , DNA, Viral/metabolism , Dexamethasone/pharmacology , Luciferases/biosynthesis , Mammary Tumor Virus, Mouse/drug effects , Mice , Molecular Sequence Data , Neurofibromin 1 , Proteins/isolation & purification , Proteins/metabolism , Templates, Genetic , Transcription Factors/drug effects , Transfection
20.
J Exp Med ; 180(6): 2347-51, 1994 Dec 01.
Article in English | MEDLINE | ID: mdl-7525852

ABSTRACT

Mouse mammary tumor virus (MMTV) encodes a superantigen (SAg) that promotes stable infection and virus transmission. Upon subcutaneous MMTV injection, infected B cells present SAg to SAg-reactive T cells leading to a strong local immune response in the draining lymph node (LN) that peaks after 6 d. We have used the reverse transcriptase inhibitor 3'-azido-3'-deoxythymidine (AZT) to dissect in more detail the mechanism of SAg-dependent enhancement of MMTV infection in this system. Our data show that no detectable B or T cell response to SAg occurs in AZT pretreated mice. However, if AZT treatment is delayed 1-2 d after MMTV injection, a normal SAg-dependent local immune response is observed on day 6. Quantitation of viral DNA in draining LN of these infected mice indicates that a 4,000-fold increase in the absolute numbers of infected cells occurs between days 2 and 6 despite the presence of AZT. Furthermore MMTV DNA was found preferentially in surface IgG+ B cells of infected mice and was not detectable in SAg-reactive T cells. Collectively our data suggest that MMTV infection occurs preferentially in B cells without SAg involvement and is completed 1-2 d after virus challenge. Subsequent amplification of MMTV infection between days 2 and 6 requires SAg expression and occurs in the absence of any further requirement for reverse transcription. We therefore conclude that clonal expansion of infected B cells via cognate interaction with SAg-reactive T cells is the predominant mechanism for increasing the level of MMTV infection. Since infected B cells display a memory (surface IgG+) phenotype, both clonal expansion and possibly longevity of the virus carrier cells may contribute to stable MMTV infection.


Subject(s)
B-Lymphocytes/immunology , Mammary Tumor Virus, Mouse/physiology , RNA-Directed DNA Polymerase/metabolism , Superantigens/biosynthesis , T-Lymphocytes/immunology , Virus Replication , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/virology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , DNA, Viral/analysis , DNA, Viral/biosynthesis , Kinetics , Lymph Nodes/immunology , Lymph Nodes/virology , Mammary Tumor Virus, Mouse/drug effects , Mammary Tumor Virus, Mouse/pathogenicity , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Phenotype , Polymerase Chain Reaction , T-Lymphocytes/drug effects , T-Lymphocytes/virology , Virus Replication/drug effects , Zidovudine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...