Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
Cell Death Dis ; 6: e1921, 2015 Oct 15.
Article En | MEDLINE | ID: mdl-26469969

The Notch1 signaling pathway contributes to tumorigenesis by influencing differentiation, proliferation and apoptosis. Here, we demonstrate that inhibition of the Notch1 signaling pathway sensitizes glioblastoma cell lines and glioblastoma initiating cells to apoptosis induced by the death ligand TRAIL. This sensitization occurs through transcriptional upregulation of the death receptor 5 (DR5, TRAIL-R2). The increase in DR5 expression is abrogated by concomitant repression of the transcription factor Sp1, which directly binds to the DR5 promoter in the absence of Notch1 as revealed by chromatin immunoprecipitation. Consistent with these findings, Notch1 inhibition resulted in increased DR5 promoter activity, which was impaired by mutation of one out of two Sp1-binding sites within the proximal DR5 promoter. Moreover, we demonstrate that JNK signaling contributes to the regulation of DR5 expression by Notch1. Taken together, our results identify Notch1 as key driver for TRAIL resistance and suggest Notch1 as a promising target for anti-glioblastoma therapy.


Antineoplastic Agents/pharmacology , Receptor, Notch1/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Sp1 Transcription Factor/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis/drug effects , Caspase 8/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Gene Expression/drug effects , Gene Knockdown Techniques , Humans , Receptor, Notch1/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Signal Transduction , Up-Regulation
2.
Br J Dermatol ; 164(6): 1201-13, 2011 Jun.
Article En | MEDLINE | ID: mdl-21418174

In recent years, the contribution of viruses to cutaneous oncogenesis has steadily gained recognition. The archetype is human herpesvirus 8, which is well established as the causative agent in Kaposi sarcoma. Other viruses believed to play a role in nonmelanoma skin cancer include human papillomavirus and the recently described Merkel cell polyomavirus. We review the mechanisms by which these three viruses interact with the host cell, ultraviolet radiation and immunosuppression to result in carcinogenesis.


Skin Neoplasms/virology , Tumor Virus Infections/complications , Carcinoma, Merkel Cell/virology , Carcinoma, Squamous Cell/virology , Cell Transformation, Neoplastic , Cell Transformation, Viral , Forecasting , Herpesvirus 8, Human , Humans , Immune Tolerance/physiology , Immunosuppression Therapy/adverse effects , Papillomavirus Infections/complications , Polyomavirus Infections/complications , Sarcoma, Kaposi/virology , Ultraviolet Rays/adverse effects
3.
Am J Transplant ; 8(11): 2199-204, 2008 Nov.
Article En | MEDLINE | ID: mdl-18785959

Globally approximately 15% of all malignant tumors are caused by viruses and even a higher percentage is observed in organ transplant recipients (OTR). Here, nonmelanoma skin cancer (NMSC) is the most frequent malignancy, which correlates with cutaneous human papilloma virus (HPV) infection. In the present review, we reflect on some recent general concepts how tumor viruses can either act as direct or indirect carcinogens in the multistep process of carcinogenesis. Immunosuppressive drugs in OTR, which reduce the risk of organ rejection, could be critical in increasing the activation of persisting viral infections, thereby enhancing the probability to develop skin tumors. We discuss virus-induced transformation with special emphasis on the function of HPV as an indirect and HHV-8 as direct carcinogen in the development of NMSC and Kaposi sarcoma (KS), respectively. Moreover, we describe a rodent model system useful to examine future strategies in preventing skin tumor formation in immunosuppressed OTR.


Organ Transplantation/adverse effects , Skin Neoplasms/etiology , Tumor Virus Infections/etiology , Animals , Carcinogens , Disease Models, Animal , Graft Rejection , Herpesvirus 8, Human/metabolism , Humans , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Papillomaviridae/metabolism , Sarcoma, Kaposi/complications , Sarcoma, Kaposi/etiology , Sarcoma, Kaposi/virology , Skin Neoplasms/complications , Skin Neoplasms/virology , Tumor Virus Infections/complications
4.
Br J Dermatol ; 157 Suppl 2: 14-7, 2007 Dec.
Article En | MEDLINE | ID: mdl-18067625

BACKGROUND: Immune response modifiers including imiquimod can be topically applied for the treatment of both genital warts and benign and malignant skin tumours (e.g. actinic keratosis). In an initial pilot study, we examined the response of spontaneously papillomavirus caused skin lesions (e.g. papillomas, keratoacanthomas) vs. chemically induced skin tumours of Mastomys coucha to imiquimod. METHODS: Fourteen spontaneously and 16 chemically [initiation with 7,12-dimethylbenzanthracene (DMBA) followed by repeated 12-O-Tetradecanoylphorbol-13-acetate (TPA) applications] induced skin tumours were treated two to three times per week with 5% imiquimod or placebo. RESULTS: Notably, significant higher regression or growth arrest rates of imiquimod treated animals were observed in chemically vs. spontaneously induced skin tumours [9/14 (64%) vs. 2/11 (18%), P < 0.05]. Regression or growth arrest of both skin tumours from placebo treated animals were similar (1/2 vs. 1/3). Tumour growth of nonresponders was lowest in imiquimod treated animals compared to the placebo or untreated group. CONCLUSIONS: Imiquimod was able to reduce skin tumour growth particularly in chemically induced lesions of Mastomys coucha. The different clearance rates are most likely due to lower differentiation status of the DMBA/TPA-induced tumours, allowing a better uptake of imiquimod than spontaneously induced papillomas or keratoacanthomas, known to be highly keratinized.


Aminoquinolines/therapeutic use , Antineoplastic Agents/therapeutic use , Skin Neoplasms/drug therapy , 9,10-Dimethyl-1,2-benzanthracene , Animals , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Imiquimod , Murinae , Papilloma/drug therapy , Papilloma/pathology , Papilloma/virology , Papillomavirus Infections/complications , Skin Neoplasms/chemically induced , Skin Neoplasms/pathology , Skin Neoplasms/virology , Tetradecanoylphorbol Acetate , Treatment Outcome
5.
J Cancer Res Clin Oncol ; 129(2): 107-13, 2003 Feb.
Article En | MEDLINE | ID: mdl-12669235

PURPOSE: Inhibitors of histone deacetylase, such as sodium butyrate, block proliferation of cervical carcinoma cells by inhibiting the G1 to S transition of the cell cycle. The derivative phenylbutyrate (PB), characterized by its higher pharmacological half-life, and its metabolite phenylacetate (PA) were tested for their growth-inhibitory function on cervical cancer cells differing in their HPV type, copy number, and integration sites. METHODS AND RESULTS: Using flow cytometric and Western blot analyses, we show that a 24-h incubation period with PB, but not with PA, was already sufficient to cause a dose-dependent growth arrest by increasing the G1 fraction with a concomitant drop in the S-phase. Consistent with the cell cycle block, only PB, but not PA, induced the cyclin-dependent kinase inhibitors p21(CIP1) and p27(KIP1). The inhibitory effect was not the result of a non-specific cytotoxic effect of PB, since cessation of cellular growth was already completely reversible 5 h after drug removal. CONCLUSIONS: Due to its broad growth inhibitory properties on different cervical carcinoma cells in vitro, and its low toxic profile demonstrated in preceding clinical studies, PB may serve as an effective drug in handling pre-cancerous lesions and cervical cancer in patients.


Antineoplastic Agents/pharmacology , Carcinoma/drug therapy , Papillomaviridae/classification , Phenylbutyrates/pharmacology , Precancerous Conditions/drug therapy , Uterine Cervical Neoplasms/drug therapy , Blotting, Western , Carcinoma/virology , Female , Flow Cytometry , Humans , Papillomavirus Infections/complications , Precancerous Conditions/virology , Tumor Virus Infections/complications , Uterine Cervical Neoplasms/virology
6.
Int J Cancer ; 93(6): 823-31, 2001 Sep.
Article En | MEDLINE | ID: mdl-11519044

When cervical carcinoma cells were monitored for apoptotic signals, HPV18(+) lines were found to be highly sensitive to agonistic CD95 antibodies or recombinant CD95 ligands after co-exposure with CHX (CD95(S)). In contrast, HPV16(+) cervical carcinoma cells and HPV16-immortalized non-malignant human keratinocytes were CD95-resistant (CD95(R)) under equivalent conditions. Somatic cell hybridization between CD95(S) and CD95(R) cervical carcinoma cell lines revealed that CD95 sensitivity was a dominant trait, which could be correlated with abundant c-Myc and low Bcl-X(L) expression. Although CD95(R) cervical carcinoma cells expressed even higher levels of p53 and CD95 receptor at the surface, resistance could be attributed to the inability to form a functional DISC, necessary for successful transmission of the apoptogenic response. These data indicate that resistance to apoptotic stimuli represents an important immunological escape mechanism during virus-induced carcinogenesis.


Apoptosis , Carcinoma/pathology , Carcinoma/virology , Papillomaviridae/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , fas Receptor/biosynthesis , Blotting, Western , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Separation , Cycloheximide/pharmacology , DNA Fragmentation , Down-Regulation , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Female , Flow Cytometry , HeLa Cells , Humans , Jurkat Cells , Ligands , Membrane Potentials , Mitochondria/metabolism , Phenotype , Protein Synthesis Inhibitors/pharmacology , Recombinant Proteins/metabolism , Sensitivity and Specificity , Time Factors , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , fas Receptor/metabolism
7.
Oncogene ; 20(35): 4768-76, 2001 Aug 09.
Article En | MEDLINE | ID: mdl-11521189

Histone deacetylase (HDAC) inhibitors sodium butyrate and trichostatin A arrest human papillomavirus (HPV)-positive carcinoma cells in G1 to S transition of the cell cycle, which is paralleled by an up-regulation of the cyclin-dependent kinase inhibitors (CKIs) p21CIP1 and p27KIP1 as well as the complete loss of cdk2 activity. Although HPV expression was hitherto thought to be required to maintain a proliferative phenotype of these cells, cdk2 suppression is achieved even in the presence of ongoing viral transcription. While CKIs normally cannot exert their cdk2-inhibitory function in the presence of the viral oncoprotein E7, co-immunoprecipitation experiments revealed that E7 binding is prevented. Increase of p27KIP1 correlates with down-regulation of p45SKP2, a component of the ubiquitin-protein ligase SCF(SKP2) controlling the half-life of regulatory proteins during the cell cycle. HDAC inhibition also triggered an E7-dependent degradation of pRb, while the levels of E2F remained unaffected. The presence of free intracellular E2F and the concomitant up-regulation of CKIs during G1 arrest results in a 'conflicting growth situation', which finally renders the cells to undergo apoptosis. These data provide novel molecular insights into how the transforming potential of HPV can be bypassed and open new therapeutical perspectives for the treatment of cervical cancer.


Apoptosis/drug effects , CDC2-CDC28 Kinases , Cell Transformation, Neoplastic , DNA-Binding Proteins , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Muscle Proteins , Oncogene Proteins, Viral/physiology , Uterine Cervical Neoplasms/drug therapy , Cell Cycle/drug effects , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/analysis , Cyclins/physiology , Female , HeLa Cells , Humans , Microfilament Proteins/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Retinoblastoma Protein/metabolism , Uterine Cervical Neoplasms/virology
8.
Oncogene ; 19(29): 3235-44, 2000 Jul 06.
Article En | MEDLINE | ID: mdl-10918580

The expression of the monocyte-chemoattractant-protein-1 (MCP-1) is closely linked with a non-tumorigenic phenotype in somatic cell hybrids made between the human papillomavirus type 18 (HPV 18) positive cervical carcinoma cell line HeLa and normal human fibroblasts. In contrast, MCP-1 transcription is absent in tumorigenic segregants derived from the same hybrids or in parental HeLa cells. Selectivity of MCP-1 transcription, which is regulated at the level of initiation of transcription, is mainly based on differences in the location and extension of DNAse I-hypersensitive regions (DHSR) at both ends of the gene. While TNF-alpha only moderately increases the sensitivity of pre-existing 5'-DHSRs, a 3'-end DHSR became strongly induced exclusively in non-malignant hybrids. DNA sequencing showed that the 3'-DHSR coincides with an additional AP-1 site located approximately 600 bp downstream of the polyadenylation site. Analyses of AP-1 composition revealed that MCP-1 is only expressed in those cells where jun-family members were mainly heterodimerized with the fos-related protein fra-1. In contrast, in tumorigenic cells the 1: 1 ratio between jun and fra-1 is disturbed and the MCP-1 gene is no longer expressed. Hence, alterations in the heterodimerization pattern of AP-1 and its selective accessibility to opened chromatin may represent a novel regulatory pathway in the regulation of chemokines in malignant and non-malignant HPV-positive cells.


Chemokine CCL2/genetics , Chromatin/physiology , Gene Expression Regulation , Papillomaviridae/genetics , Transcription Factor AP-1/metabolism , Base Sequence , Cell Line , Cell Nucleus/metabolism , Chromosome Mapping , Deoxyribonuclease I/metabolism , Gene Expression Regulation/drug effects , HeLa Cells , Humans , Molecular Sequence Data , RNA, Messenger , Sequence Analysis, DNA , Transcription Factor AP-1/genetics , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/pharmacology
9.
Int J Cancer ; 86(6): 811-7, 2000 Jun 15.
Article En | MEDLINE | ID: mdl-10842195

The transcription factor AP-1 represents a central key element in the expression of human pathogenic papillomaviruses (HPV). We here propose a novel role for AP-1 as an essential component of an intracellular surveillance mechanism negatively controlling the proliferation of HPV-positive cells under in vivo conditions. The dissection of AP-1 composition in cervical-carcinoma cells revealed an inverse relationship between the Fos-related antigen Fra-1 and the tumorigenic phenotype. Cervical-carcinoma cell lines were either negative or expressed only low amounts of Fra-1 (jointly with c-Fos) within their AP-1 complexes. Somatic-cell hybridization technique was used to fuse different HPV-positive malignant cell lines. This resulted either in tumorigenic hybrids or in cells in which the malignant phenotype of the parental fusion partners was completely suppressed. The monitoring of AP-1 composition in electrophoretic mobility super-shift assays showed that the amount of Fra-1 was substantially increased within the AP-1 complex of non-malignant cells. In contrast, Fra-1 was even diminished in malignant hybrids, while c-Fos remained expressed. This correlation suggests that the concentration of Fra-1 within the AP-1 transcription complex might be an important marker for predicting the in vivo growth properties of HPV-positive cells.


Papillomaviridae/isolation & purification , Transcription Factor AP-1/analysis , Uterine Cervical Neoplasms/etiology , Female , Genetic Complementation Test , Humans , Proto-Oncogene Proteins c-fos/analysis , Tumor Cells, Cultured , Uterine Cervical Neoplasms/virology
10.
Oncogene ; 18(21): 3187-98, 1999 May 27.
Article En | MEDLINE | ID: mdl-10359524

AP-1 represents a transcription factor, which plays a pivotal role in initiating and maintaining the expression of human papillomavirus (HPV) oncoproteins E6 and E7 during HPV-linked carcinogenesis of the uterine cervix. AP-1 stands as a synonym for different proteins such as c-Jun, JunB, JunD, c-Fos, FosB as well as the Fos-related antigens Fra-1 and Fra-2, which can either homo- or heterodimerize to build up a functional transcription complex. AP-1 is mainly considered as a positive regulator, which binds to cognate DNA sequences within the viral upstream regulatory region. By using non-tumorigenic HeLa-fibroblast hybrids ('444'), their tumorigenic segregants ('CGL3') as well as HPV 18 positive HeLa cells as a experimental model system, evidence is provided that AP-1 composition differs considerably between these cell lines. In nuclear extracts obtained from non-tumorigenic cells, Jun-family members (in the order c-Jun>JunD>JunB) were mainly heterodimerized with Fra-1, a protein, known to be involved in the abrogation of AP-1 activity under certain experimental conditions. In contrast, Fra-1 concentration is low in extracts from tumorigenic cells. Conversely, c-Fos, the canonical dimerization partner of Jun proteins is expressed in substantial quantity in HeLa- and 'CGL3' cells, but it is completely absent in AP-1 complexes from non-tumorigenic '444' cells. Ectopical expression of c-fos under a heterologous promoter in '444'-cells induces tumorigenicity and a change of the Jun/Fra-1 ratio towards a constellation initially detected in 'CGL3'-and HeLa cells. Furthermore, conversion to tumorigenicity is accompanied with a resistance against TNF-alpha, a cytokine, capable to selectively suppress HPV 18 transcription in formerly non-malignant cells. These data propose a novel role for AP-1 as an essential component of an inter- and intracellular surveillance mechanism negatively controlling HPV transcription in non-tumorigenic cells.


Gene Expression Regulation, Viral/drug effects , Papillomaviridae/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factor AP-1/metabolism , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Blotting, Western , Cell Division , Electrophoresis, Polyacrylamide Gel , Fibroblasts/drug effects , Fibroblasts/metabolism , Genetic Vectors , HeLa Cells , Humans , Hybrid Cells , Neoplasm Invasiveness , Proto-Oncogene Proteins c-fos/genetics , Transcription Factor AP-1/genetics , Transfection
11.
Int J Cancer ; 82(1): 6-11, 1999 Jul 02.
Article En | MEDLINE | ID: mdl-10360812

Chemokines play a central role in the chemotactic activation of immunological effector cells. One of the currently best characterized chemokines is the monocyte-chemo-attractant protein-1 (MCP-1), which is involved in the cross-talk with cells of the monocyte-macrophage lineage. Since macrophages and macrophage-derived cytokines appear to be important in the transcriptional regulation of "high-risk" types of human papillomaviruses (HPV), we monitored MCP-1 expression by in situ hybridization (ISH) in histologically distinct stages of cervical intra-epithelial neoplasms (CIN), cervical cancer and non-HPV-associated cases of erosive endocervicitis. Here, we demonstrate that high-grade dysplasia (CIN III, n = 9) completely lacks both MCP-1 expression and CD68+-macrophage infiltration, while MCP-1-specific signals were occasionally detectable in one out of 5 CIN-II and in one out of 3 CIN-I lesions. Inspection of hyperplastic squamous epithelium adjacent to cervical carcinomas reveals high MCP-1 expression and accumulation of infiltrating macrophages. In contrast, no macrophages could be detected in corresponding hyperplastic tissue areas surrounding CIN-II and CIN-III lesions, although MCP-1 was found to be highly expressed. Finally, in agreement with our earlier in vitro data, invasive carcinomas of the cervix uteri showed MCP-1-specific hybridization signals and macrophage infiltration only in the stroma surrounding the carcinoma cells and in endothelial cells of capillaries, especially at the invasion front of the tumor, while the inner mass of the carcinomas was completely negative. On the other hand, ISH and histochemical evaluation of inflammatory, non-HPV-associated cases of erosive endocervicitis indicate strong MCP-1 expression, which is regularly accompanied by chemotactic appearance of macrophages. These observations indicate that dysregulation of MCP-1-gene expression may represent an important step during HPV-linked carcinogenesis, allowing the escape of virus-positive cells from local immune response.


Chemokine CCL2/genetics , RNA, Messenger/analysis , Uterine Cervical Dysplasia/immunology , Uterine Cervical Neoplasms/immunology , Adult , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Female , Gene Expression Regulation, Neoplastic , Humans , Macrophages/pathology , Middle Aged , Papillomaviridae , Tumor Virus Infections/complications , Tumor Virus Infections/immunology , Uterine Cervical Neoplasms/etiology , Uterine Cervical Neoplasms/pathology , Uterine Cervical Dysplasia/etiology , Uterine Cervical Dysplasia/pathology
12.
Int J Cancer ; 76(5): 639-46, 1998 May 29.
Article En | MEDLINE | ID: mdl-9610719

The glycolytic pathway inhibitor 2-deoxyglucose (2-DG) is capable of suppressing the transcription of the human pathogenic papillomavirus type 18 (HPV 18) in cervical carcinoma cells and derived non-tumorigenic somatic cell hybrids at the level of transcription initiation. HPV down-regulation is selective, since other reference genes are not affected or even up-regulated under the same experimental conditions. Moreover, 2-DG appears to restore the normal half-life of the tumor suppressor gene product p53, because the protein is strongly up-regulated after HPV 18 E6/E7 suppression. The observed 2-DG-effect is not cytotoxic and is reversible after refeeding with fresh medium. HPV 18 suppression by 2-DG can be completely abrogated by simultaneous treatment with the intracellular Ca2+ antagonist TMB-8, indicating that Ca2+, a known intracellular "second messenger", is involved in this process. Elevated c-myc and p53 expression appears to be responsible for the time-dependent accumulation of apoptotic cells after prolonged 2-DG treatment. The finding that 2-DG acts selectively against the expression of a human pathogenic papillomavirus strongly suggests that an appropriate level of glycolysis is not only a peculiarity of growing tumors, but even may be an essential prerequisite for the maintenance of virus-specific E6/E7 gene expression. Our results may have substantial implications for the potential therapeutic application of 2-DG or other glucose derivatives in the treatment of precancerous and malignant HPV-associated lesions.


Antiviral Agents/pharmacology , DNA-Binding Proteins , Deoxyglucose/pharmacology , Oncogene Proteins, Viral/biosynthesis , Papillomaviridae/genetics , Papillomaviridae/metabolism , Transcription, Genetic/drug effects , Adenosine Triphosphate/metabolism , Apoptosis/drug effects , Calcium Channel Blockers/pharmacology , Culture Media , Down-Regulation/drug effects , Gallic Acid/analogs & derivatives , Gallic Acid/pharmacology , Gene Expression Regulation, Viral/drug effects , HeLa Cells , Humans , Oncogene Proteins, Viral/genetics , Papillomaviridae/drug effects , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/biosynthesis
13.
J Virol ; 71(1): 362-70, 1997 Jan.
Article En | MEDLINE | ID: mdl-8985358

Considering the involvement of a redox-regulatory pathway in the expression of human papillomaviruses (HPVs), HPV type 16 (HPV-16)-immortalized human keratinocytes were treated with the antioxidant pyrrolidine-dithiocarbamate (PDTC). PDTC induces elevated binding of the transcription factor AP-1 to its cognate recognition site within the viral regulatory region. Despite of increased AP-1 binding, normally indispensable for efficient HPV-16 transcription, viral gene expression was selectively suppressed at the level of initiation of transcription. Electrophoretic mobility supershift assays showed that the composition of the AP-1 complex, predominantly consisting of Jun homodimers in untreated cells, was altered. Irrespective of enhanced c-fos expression, c-jun was phosphorylated and became primarily heterodimerized with fra-1, which was also induced after PDTC incubation. Additionally, there was also an increased complex formation between c-jun and junB. Because both fra-1 and junB overexpression negatively interferes with c-jun/c-fos trans-activation of AP-1-responsive genes, our results suggest that the observed block in viral transcription is mainly the consequence of an antioxidant-induced reconstitution of the AP-1 transcription complex. Since expression of the c-jun/c-fos gene family is tightly regulated during cellular differentiation, defined reorganization of a central viral transcription factor may represent a novel mechanism controlling the transcription of pathogenic HPVs during keratinocyte differentiation and in the progression to cervical cancer.


Antioxidants/pharmacology , Papillomaviridae/genetics , Pyrrolidines/pharmacology , Thiocarbamates/pharmacology , Transcription Factor AP-1/drug effects , Transcription, Genetic/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Down-Regulation , Genes, p53 , HeLa Cells , Humans , Keratinocytes/cytology , Transcription Factor AP-1/genetics , Tumor Cells, Cultured , Up-Regulation
14.
Virology ; 217(1): 33-41, 1996 Mar 01.
Article En | MEDLINE | ID: mdl-8599218

Human papillomavirus (HPV) type 16 DNA sequences have been found integrated into the host cell genome in a large number of cervical tumors and cell lines derived therefrom. In this study, we have cloned and analyzed the nonoccupied allele corresponding to the integration site of HPV-16 in the cervical cancer cell line SiHa. Our mapping analyses revealed an approximately 7.8-kb deletion of cellular DNA upon viral integration. Computer analysis of 2.3 kb of DNA sequences from the deleted genomic region as well as 1.0 kb of sequences upstream of the viral integration site showed no significant homology to any known human sequences. DNase I mapping experiments on native chromatin demonstrated the existence of two hypersensitive sites in both the HPV-16-containing and nonoccupied alleles located approximately 1.1 and 1.7 kb upstream of the viral integration site. This suggests that viral integration occurred close to putative regulatory sequences and that recombination with host cellular DNA was not followed by a reorganization of the chromatin structure upstream of the integration site. Nuclear run-on and RT-PCR experiments showed HPV-specific transcription spanning the E2, E4, E5, and L1/L2 open reading frames (ORFs) located upstream of the HPV-16 regulatory region (URR). Taken together, our data suggest that the cellular DNA region upstream of the HPV-16 integration site in the SiHa cell line contains regulatory elements affecting transcription of HPV-16 ORFs located upstream of the HPV-16 URR.


Alleles , Papillomaviridae/genetics , Sequence Deletion , Uterine Cervical Neoplasms/virology , Virus Integration/genetics , Base Sequence , Chromatin , Cloning, Molecular , DNA, Viral/genetics , Female , Humans , Molecular Sequence Data , Tumor Cells, Cultured , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
15.
Mol Carcinog ; 14(3): 179-89, 1995 Nov.
Article En | MEDLINE | ID: mdl-7576110

To investigate the effect of tumor-associated macrophages on the in vivo growth properties of cervical carcinoma cells, tumorigenic human papilloma virus (HPV) 18-positive HeLa cells were transfected with an expression vector harboring the cDNA for the macrophage chemoattractant protein-1 JE (MCP-1). Although the endogenous gene is present and not structurally rearranged, its expression seems to be negatively affected by a still unknown mechanism. Inoculation of JE (MCP-1)-negative HeLa cells into nude mice led to rapidly growing tumors, where macrophage infiltration into the inner tumor mass was not detectable immunohistochemically. The activity that attracted mononuclear cells under both in vitro and in vivo condition was reconstituted in HeLa cells after transfection with the JE (MCP-1) expression vector. Heterotransplantation of those cells into immunocompromised animals resulted in significant growth retardation that was accompanied by a strong infiltration of macrophages. On the other hand, in vivo selection of nonmalignant hybrids made between wild-type HeLa cells and normal human fibroblasts in nude mice resulted in tumorigenic segregants 4 mo after inoculation into the animals. Monitoring JE (MCP-1) expression directly within those nodules, we found that transcription was either absent or only weakly detectable. Recultivation of JE (MCP-1)-positive tissue grafts under in vitro conditions revealed that the gene was only marginally inducible by tumor necrosis factor-alpha, a cytokine that normally induces a very strong activation of transcription in nontumorigenic cells. These findings suggest that functional JE (MCP-1) expression and in turn activated macrophages may play a pivotal role in controlling the proliferation rate of HPV-positive cells in vivo.


Chemokine CCL2/genetics , Animals , Cell Division , Chemokine CCL2/physiology , DNA, Complementary/genetics , Gene Expression , Gene Expression Regulation/physiology , HeLa Cells , Humans , Hybrid Cells , Immunohistochemistry , Macrophages/physiology , Mice , Mice, Nude , Papillomaviridae , Promoter Regions, Genetic , RNA, Messenger/genetics , Simian virus 40/genetics , Transfection , Tumor Necrosis Factor-alpha/pharmacology
16.
J Gen Virol ; 75 ( Pt 5): 1157-62, 1994 May.
Article En | MEDLINE | ID: mdl-8176376

Human papillomavirus type 6 (HPV-6) was isolated from a tongue papilloma which subsequently progressed to an invasive carcinoma. Three biopsies were taken from the same patient at different intervals during the tumour development. The HPV-6 genome in all three biopsies contained a GT-rich 94 bp insertion at nucleotide 7350 in the upstream regulatory region (URR). In comparison to previously published HPV-6 DNA isolates, this insertion seems to be the most prevalent and constant modification, not present in the prototype HPV-6b, and allows an improved alignment with the sequence of the HPV-11 genome. The possible biological significance of these GT-rich clusterings at the beginning of the URR, present not only in these HPV-6 isolates but observed in all other 'genital' HPVs also, is discussed.


Carcinoma/etiology , Papillomaviridae/genetics , Papillomavirus Infections/genetics , Tongue Neoplasms/etiology , Tumor Virus Infections/genetics , Aged , Base Sequence , Biopsy , Carcinoma/virology , Gene Rearrangement/genetics , Genome, Viral , Humans , Male , Molecular Sequence Data , Regulatory Sequences, Nucleic Acid/genetics , Sequence Homology, Nucleic Acid , Time Factors , Tongue Neoplasms/virology
17.
J Virol ; 68(4): 2142-50, 1994 Apr.
Article En | MEDLINE | ID: mdl-8138998

Malignant human papillomavirus type 18 (HPV18)-positive cervical carcinoma cells can be reverted to a nonmalignant phenotype by generation of somatic cell hybrids with normal human fibroblasts. Although nontumorigenic hybrids, their tumorigenic segregants, and the parental HeLa cells have similar in vitro properties, inoculation only of nontumorigenic cells into nude mice results in a selective suppression of HPV18 transcription which precedes cessation of cellular growth. Our present study, aimed at understanding the differential regulation in vitro and in vivo, shows that the JE gene, encoding the monocyte chemoattractant protein (MCP-1), is expressed only in nontumorigenic hybrids. Although the gene, including its regulatory region, is intact, no JE (MCP-1) mRNA is detected in the tumorigenic segregants and in other malignant HPV-positive cervical carcinoma cell lines. Tests of several monocyte-derived cytokines showed that only tumor necrosis factor alpha strongly induces the JE (MCP-1) gene in nontumorigenic cells and that this is accompanied by a dose-dependent reduction of HPV transcription. The JE (MCP-1) up-regulation occurs within 2 h and does not require de novo protein synthesis. The response to tumor necrosis factor alpha seems to be mediated by an NF-kappa B-related mechanism, since the induction can be completely abrogated by pretreating the cells with an antioxidant such as pyrrolidine dithiocarbamate. Interestingly, cocultivation of nonmalignant hybrids with monocyte-enriched fractions from human peripheral blood also results in an induction of the JE (MCP-1) gene and a concomitant suppression of HPV18 transcription. Neither effect is observed in malignant cells. These data suggest that JE (MCP-1) may play a pivotal role in the intercellular communication by triggering an intracellular pathway which negatively interferes with viral transcription in HPV-positive nontumorigenic cells.


Carcinoma/genetics , Chemotactic Factors/genetics , Cytokines/genetics , Gene Expression Regulation, Neoplastic , Uterine Cervical Neoplasms/genetics , Base Sequence , Cell Division , Chemokine CCL2 , Culture Techniques/methods , Female , HeLa Cells , Humans , Hybrid Cells , Leukocytes, Mononuclear , Molecular Sequence Data , Papillomaviridae/genetics , RNA, Messenger/analysis , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
20.
J Gen Virol ; 74 ( Pt 5): 791-801, 1993 May.
Article En | MEDLINE | ID: mdl-8388016

Integration of human papillomaviruses (HPVs) into the host genome is considered to be an early and important event in HPV-linked cervical carcinogenesis. Consequently, the viral DNA potentially becomes a target for cellular control mechanisms normally acting on the corresponding integration site. Besides resulting position effects, host-specific DNA methylation may play a functional role in HPV gene regulation. To elucidate the influence of such a kind of epigenetic modification on viral transcription, in vitro methylation studies on HPV-18 upstream regulatory region (URR)-controlled reporter plasmids were carried out. Selective methylation of the viral URR results in a down-regulation of the transcriptional activity, which can be attributed to nonrandom distribution of methyl-acceptor sites clustered within the constitutive enhancer region. In vivo competition experiments show that suppression is not directly mediated by steric hindrance of methyl residues with transcription factors, but rather is due to the association with methyl-CpG DNA-binding proteins. Using a restriction enzyme accessibility assay on both the DNA and chromatin levels, it could be demonstrated that, in vivo, extensively methylated viral DNA is nucleosomally organized, characteristic of transcriptionally inactive chromatin. These data suggest that DNA methylation is an important regulatory pathway in the modulation of HPV expression and as a consequence the proliferation rate of virus-infected cells.


DNA, Viral/metabolism , Gene Expression Regulation, Viral , Papillomaviridae/genetics , Binding, Competitive , Cells, Cultured , Chromatin/ultrastructure , Cloning, Molecular , DNA-Binding Proteins/metabolism , Humans , Methylation , Nucleosomes/metabolism , Transcription, Genetic , Transfection , Tumor Cells, Cultured
...