Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 20(21)2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31683862

ABSTRACT

Mucosotropic human papillomaviruses (HPVs) cause prevalent anogenital infections, some of which can progress to cancers. It is imperative to identify efficacious drug candidates, as there are few therapeutic options. We have recapitulated a robust productive program of HPV-18 in organotypic raft cultures of primary human keratinocytes. The HPV E7 protein induces S phase reentry, along with DNA damage response (DDR) in differentiated cells to support viral DNA amplification. A number of small molecule inhibitors of DDR regulators are in clinical use or clinical trials to treat cancers. Here, we used our raft culture system to examine effects of inhibitors of ATR/Chk1 and ATM/Chk2 on HPV infection. The inhibitors impaired S-phase reentry and progression as well as HPV DNA amplification. The Chk1 inhibitor MK-8776 was most effective, reducing viral DNA amplification by 90-99% and caused DNA damage and apoptosis, preferentially in HPV infected cells. We found that this sensitivity was imparted by the E7 protein and report that MK-8776 also caused extensive cell death of cervical cancer cell lines. Furthermore, it sensitized the cells to cisplatin, commonly used to treat advanced cervical cancer. Based on these observations, the Chk1 inhibitors could be potential effective agents to be re-purposed to treat the spectrum of HPV infections in single or combination therapy.


Subject(s)
DNA, Viral/antagonists & inhibitors , Human papillomavirus 18/metabolism , Keratinocytes/drug effects , Papillomavirus E7 Proteins/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Virus Replication/drug effects , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cells, Cultured , Cisplatin/pharmacology , DNA, Viral/genetics , DNA, Viral/metabolism , Female , HeLa Cells , Human papillomavirus 18/genetics , Human papillomavirus 18/physiology , Humans , Keratinocytes/metabolism , Keratinocytes/virology , Male , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/genetics , Papillomavirus Infections/prevention & control , Papillomavirus Infections/virology , Protein Kinase Inhibitors/pharmacology , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , Virus Replication/genetics
3.
Proc Natl Acad Sci U S A ; 115(47): E11138-E11147, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30385631

ABSTRACT

Human papillomaviruses (HPVs) cause epithelial proliferative diseases. Persistent infection of the mucosal epithelia by the high-risk genotypes can progress to high-grade dysplasia and cancers. Viral transcription and protein activities are intimately linked to regulation by histone acetyltransferases and histone deacetylases (HDACs) that remodel chromatin and regulate gene expression. HDACs are also essential to remodel and repair replicating chromatin to enable the progression of replication forks. As such, Vorinostat (suberoylanilide hydroximic acid), and other pan-HDAC inhibitors, are used to treat lymphomas. Here, we investigated the effects of Vorinostat on productive infection of the high-risk HPV-18 in organotypic cultures of primary human keratinocytes. HPV DNA amplifies in the postmitotic, differentiated cells of squamous epithelia, in which the viral oncoproteins E7 and E6 establish a permissive milieu by destabilizing major tumor suppressors, the pRB family proteins and p53, respectively. We showed that Vorinostat significantly reduced these E6 and E7 activities, abrogated viral DNA amplification, and inhibited host DNA replication. The E7-induced DNA damage response, which is critical for both events, was also compromised. Consequently, Vorinostat exposure led to DNA damage and triggered apoptosis in HPV-infected, differentiated cells, whereas uninfected tissues were spared. Apoptosis was attributed to highly elevated proapoptotic Bim isoforms that are known to be repressed by EZH2 in a repressor complex containing HDACs. Two other HDAC inhibitors, Belinostat and Panobinostat, also inhibited viral DNA amplification and cause apoptosis. We suggest that HDAC inhibitors are promising therapeutic agents to treat benign HPV infections, abrogate progeny virus production, and hence interrupt transmission.


Subject(s)
DNA Replication/drug effects , DNA-Binding Proteins/metabolism , Histone Deacetylase Inhibitors/pharmacology , Human papillomavirus 18/genetics , Oncogene Proteins, Viral/metabolism , Papillomavirus Infections/drug therapy , Vorinostat/pharmacology , Apoptosis/drug effects , Bcl-2-Like Protein 11/metabolism , Cells, Cultured , DNA Repair/drug effects , DNA, Viral/genetics , Histones/metabolism , Human papillomavirus 18/drug effects , Humans , Hydroxamic Acids/pharmacology , Keratinocytes/virology , Mucous Membrane/virology , Panobinostat/pharmacology , Papillomavirus Infections/prevention & control , Papillomavirus Infections/transmission , Sulfonamides/pharmacology , Tumor Suppressor Protein p53/metabolism
5.
Ann Plast Surg ; 76(4): 446-52, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26954733

ABSTRACT

Wound healing involves a number of factors that results in the production of a "closed" wound. Studies have shown, in animal models, acceleration of wound healing with the addition of adipose-derived stromal cells (ADSC). The cause for the positive effect which these cells have on wound healing has not been elucidated. We have previously shown that addition of ADSC to the dermal equivalent in 3-dimensional skin cultures accelerates reepithelialization. We now demonstrate that conditioned media (CM) from cultured ADSC produced a similar rate of healing. This result suggests that a feedback from the 3-dimensional epithelial cultures to ADSC was not necessary to effect the accelerated reepithelialization. Mass spectrometry of CM from ADSC and primary human fibroblasts revealed differences in secretomes, some of which might have roles in the accelerating wound healing. Thus, the use of CM has provided some preliminary information on a possible mode of action.


Subject(s)
Mesenchymal Stem Cells/physiology , Re-Epithelialization/physiology , Subcutaneous Fat, Abdominal/cytology , Tissue Culture Techniques/methods , Culture Media, Conditioned , Fibroblasts/physiology , Humans , Skin , Wound Healing/physiology
6.
Proc Natl Acad Sci U S A ; 110(19): 7542-9, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23572574

ABSTRACT

Human papillomaviruses (HPVs) amplify in differentiated strata of a squamous epithelium. The HPV E7 protein destabilizes the p130/retinoblastoma susceptibility protein family of tumor suppressors and reactivates S-phase reentry, thereby facilitating viral DNA amplification. The high-risk HPV E6 protein destabilizes the p53 tumor suppressor and many other host proteins. However, the critical E6 targets relevant to viral DNA amplification have not been identified, because functionally significant E6 mutants are not stably maintained in transfected cells. Using Cre-loxP recombination, which efficiently generates HPV genomic plasmids in transfected primary human keratinocytes, we have recapitulated a highly productive infection of HPV-18 in organotypic epithelial cultures. By using this system, we now report the characterization of four HPV-18 E6 mutations. An E6 null mutant accumulated high levels of p53 and amplified very poorly. p53 siRNA or ectopic WT E6 partially restored amplification, whereas three missense E6 mutations that did not effectively destabilize p53 complemented the null mutant poorly. Unexpectedly, in cis, two of the missense mutants amplified, albeit to a lower extent than the WT and only in cells with undetectable p53. These observations and others implicate p53 and additional host proteins in regulating viral DNA amplification and also suggest an inhibitory effect of E6 overexpression. We show that high levels of viral DNA amplification are critical for late protein expression and report several previously undescribed viral RNAs, including bicistronic transcripts predicted to encode E5 and L2 or an alternative form of E1^E4 and L1.


Subject(s)
DNA, Viral/metabolism , DNA-Binding Proteins/genetics , Genes, p53 , Human papillomavirus 18/genetics , Mutation , Oncogene Proteins, Viral/genetics , Cells, Cultured , Genes, Tumor Suppressor , Genetic Complementation Test , Genome, Viral , Humans , Integrases/metabolism , Keratinocytes/cytology , Mutation, Missense , Phenotype , Plasmids/metabolism , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/metabolism
7.
Ann Plast Surg ; 68(5): 501-4, 2012 May.
Article in English | MEDLINE | ID: mdl-22510896

ABSTRACT

Adipose tissue is a known reservoir of multipotent mesenchymal stem cells, which can be manipulated in culture to produce cells with different phenotypes. The goal of this study was to determine whether the addition of these multipotential cells to organotypic, human skin equivalent cultures would accelerate wound healing after laser injury. For our initial studies, we were able to obtain 3-dimensional raft cultures from adult skin explanted directly onto the dermal equivalent containing human fibroblasts with or without adipose-derived stromal cells (ADSCs). Two days after laser injury, the raft cultures of skin explants that contained ADSCs had a completely healed multilayered epidermis, whereas the control raft culture without the adipose-derived cells still had areas of injury. With this encouraging outcome, these experiments were then repeated in a raft culture system initiated from dissociated primary adult human keratinocytes on the humanized dermal equivalent. Again, the cultures containing ADSCs healed faster than the control cultures. In conclusion, these data provide support to our hypothesis that ADSCs are an excellent and readily available source of factors necessary for accelerated wound healing and tissue regeneration.


Subject(s)
Adipose Tissue/cytology , Burns/therapy , Radiation Injuries/therapy , Skin Physiological Phenomena , Skin/radiation effects , Stromal Cells/transplantation , Wound Healing , Adipocytes , Adult , Animals , Cells, Cultured , Fibroblasts , Humans , Keratinocytes , Lasers, Gas/adverse effects , Rats , Tissue Culture Techniques
8.
Mol Cancer ; 10: 71, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21663621

ABSTRACT

BACKGROUND: High-risk human papillomavirus (hrHPV) infections are causally related to cervical cancer development. The additional (epi)genetic alterations driving malignant transformation of hrHPV-infected cells however, are not yet fully elucidated. In this study we experimentally assessed the role of the PI3-kinase pathway and its regulator PIK3CA, which is frequently altered in cervical cancer, in HPV-induced transformation. METHODS: Cervical carcinomas and ectocervical controls were assessed for PIK3CA mRNA and protein expression by quantitative RT-PCR and immunohistochemical staining, respectively. A longitudinal in vitro model system of hrHPV-transfected keratinocytes, representing the immortal and anchorage independent phenotype, was assayed for PI3-kinase activation and function using chemical pathway inhibition i.e. LY294002 treatment, and PIK3CA RNA interference. Phenotypes examined included cellular viability, migration, anchorage independent growth and differentiation. mRNA expression of hTERT and HPV16 E6E7 were studied using quantitative RT-PCR and Northern blotting. RESULTS: Cervical carcinomas showed significant overexpression of PIK3CA compared to controls. During HPV-induced transformation in vitro, expression of the catalytic subunit PIK3CA as well as activation of downstream effector PKB/AKT progressively increased in parallel. Inhibition of PI3-kinase signalling in HPV16-transfected keratinocytes by chemical interference or siRNA-mediated silencing of PIK3CA resulted in a decreased phosphorylation of PKB/AKT. Moreover, blockage of PI3-kinase resulted in reduced cellular viability, migration, and anchorage independent growth. These properties were accompanied with a downregulation of HPV16E7 and hTERT mRNA expression. In organotypic raft cultures of HPV16- and HPV18-immortalized cells, phosphorylated PKB/AKT was primarily seen in differentiated cells staining positive for cytokeratin 10 (CK10). Upon PI3-kinase signalling inhibition, there was a severe impairment in epithelial tissue development as well as a dramatic reduction in p-PKB/AKT and CK10. CONCLUSION: The present data indicate that activation of the PI3-kinase/PKB/AKT pathway through PIK3CA regulates various transformed phenotypes as well as growth and differentiation of HPV-immortalized cells and may therefore play a pivotal role in HPV-induced carcinogenesis.


Subject(s)
Alphapapillomavirus/physiology , Cell Transformation, Neoplastic/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Carcinoma/physiopathology , Cell Differentiation/drug effects , Cell Line, Transformed , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Chromones/pharmacology , Class I Phosphatidylinositol 3-Kinases , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Humans , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Uterine Cervical Neoplasms/physiopathology
9.
J Biol Chem ; 286(17): 15473-82, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21321122

ABSTRACT

The productive program of human papillomaviruses occurs in differentiated squamous keratinocytes. We have previously shown that HPV-18 DNA amplification initiates in spinous cells in organotypic cultures of primary human keratinocytes during prolonged G(2) phase, as signified by abundant cytoplasmic cyclin B1 (Wang, H. K., Duffy, A. A., Broker, T. R., and Chow, L. T. (2009) Genes Dev. 23, 181-194). In this study, we demonstrated that the E7 protein, which induces S phase reentry in suprabasal cells by destabilizing the p130 pocket protein (Genovese, N. J., Banerjee, N. S., Broker, T. R., and Chow, L. T. (2008) J. Virol. 82, 4862-4873), also elicited extensive G(2) responses. Western blots and indirect immunofluorescence assays were used to probe for host proteins known to control G(2)/M progression. E7 expression induced cytoplasmic accumulation of cyclin B1 and cdc2 in the suprabasal cells. The elevated cdc2 had inactivating phosphorylation on Thr(14) or Tyr(15), and possibly both, due to an increase in the responsible Wee1 and Myt1 kinases. In cells that harbored cytoplasmic cyclin B1 or cdc2, there was also an accumulation of the phosphatase-inactive cdc25C phosphorylated on Ser(216), unable to activate cdc2. Moreover, E7 expression induced elevated expression of phosphorylated ATM (Ser(1981)) and the downstream phosphorylated Chk1, Chk2, and JNKs, kinases known to inactivate cdc25C. Similar results were observed in primary human keratinocyte raft cultures in which the productive program of HPV-18 took place. Collectively, this study has revealed the mechanisms by which E7 induces prolonged G(2) phase in the differentiated cells following S phase induction.


Subject(s)
G2 Phase , Keratinocytes/pathology , Keratinocytes/virology , Papillomavirus E7 Proteins/physiology , S Phase , Cell Cycle Proteins/metabolism , Cell Differentiation , DNA-Binding Proteins/physiology , Humans , Oncogene Proteins, Viral/physiology , Phosphorylation
10.
J Virol ; 82(10): 4862-73, 2008 May.
Article in English | MEDLINE | ID: mdl-18321970

ABSTRACT

The E7 proteins of human papillomaviruses (HPVs) promote S-phase reentry in differentiated keratinocytes of the squamous epithelia to support viral DNA amplification. In this study, we showed that nuclear p130 was present in the differentiated strata of several native squamous epithelia susceptible to HPV infection. In contrast, p130 was below the level of detection in HPV-infected patient specimens. In submerged and organotypic cultures of primary human keratinocytes, the E7 proteins of the high-risk mucosotrophic HPV-18, the benign cutaneous HPV-1, and, to a lesser extent, the low-risk mucosotropic HPV-11 destabilized p130. This E7 activity depends on an intact pocket protein binding domain and a casein kinase II (CKII) phosphorylation motif. Coimmunoprecipitation experiments showed that both E7 domains were important for binding to p130 in extracts of organotypic cultures. Metabolic labeling in vivo demonstrated that E7 proteins were indeed phosphorylated in a CKII motif-dependent manner. Moreover, the efficiencies of the E7 proteins of various HPV types or mutations to induce S-phase reentry in spinous cells correlated with their relative abilities to bind and to destabilize p130. Collectively, these data support the notion that p130 controls the homeostasis of the differentiated keratinocytes and is therefore targeted by E7 for degradation to establish conditions permissive for viral DNA amplification.


Subject(s)
Cell Cycle , Keratinocytes/chemistry , Keratinocytes/virology , Papillomaviridae/physiology , Papillomavirus E7 Proteins/metabolism , Retinoblastoma-Like Protein p130/metabolism , Amino Acid Sequence , Cells, Cultured , Epithelium/chemistry , Epithelium/virology , Human papillomavirus 11/physiology , Human papillomavirus 18/physiology , Humans , Molecular Sequence Data , Papillomavirus E7 Proteins/chemistry , Phosphorylation , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , Retinoblastoma-Like Protein p130/chemistry
11.
Breast Cancer Res Treat ; 105(2): 157-67, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17221158

ABSTRACT

PURPOSE: Alternative and complementary therapeutic strategies need to be developed for metastatic breast cancer. Virotherapy is a novel therapeutic approach for the treatment of cancer in which the replicating virus itself is the anticancer agent. However, the success of virotherapy has been limited due to inefficient virus delivery to the tumor site. The present study addresses the utility of human mesenchymal stem cells (hMSCs) as intermediate carriers for conditionally replicating adenoviruses (CRAds) to target metastatic breast cancer in vivo. EXPERIMENTAL DESIGN: HMSC were transduced with CRAds. We used a SCID mouse xenograft model to examine the effects of systemically injected CRAd loaded hMSC or CRAd alone on the growth of MDA-MB-231 derived pulmonary metastases (experimental metastases model) in vivo and on overall survival. RESULTS: Intravenous injection of CRAd loaded hMSCs into mice with established MDA-MB-231 pulmonary metastatic disease homed to the tumor site and led to extended mouse survival compared to mice treated with CRAd alone. CONCLUSION: Injected hMSCs transduced with CRAds suppressed the growth of pulmonary metastases, presumably through viral amplification in the hMSCs. Thus, hMSCs may be an effective platform for the targeted delivery of CRAds to distant cancer sites such as metastatic breast cancer.


Subject(s)
Adenoviridae/genetics , Breast Neoplasms/therapy , Genetic Therapy , Lung Neoplasms/therapy , Mesenchymal Stem Cells , Receptors, CXCR4/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Genetic Vectors , Humans , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Mice , Mice, SCID , Promoter Regions, Genetic , Receptors, CXCR4/metabolism , Survival Rate , Transplantation, Heterologous , Tumor Cells, Cultured
12.
J Virol ; 80(13): 6517-24, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16775338

ABSTRACT

The productive program of human papillomaviruses (HPVs) in epithelia is tightly linked to squamous differentiation. The E7 proteins of high-risk HPV genotypes efficiently inactivate the pRB family of proteins that control the cell cycle, triggering S phase in suprabasal keratinocytes. This ability has until now not been demonstrated for the low-risk HPV-6 or HPV-11 E7 proteins. An inducible system in which HPV-16 E7 is fused to the ligand binding domain of the human estrogen receptor (ER) was described by Smith-McCune et al. (K. Smith-McCune, D. Kalman, C. Robbins, S. Shivakumar, L. Yuschenkoff, and J. M. Bishop, Proc. Natl. Acad. Sci. USA 96:6999-7004, 1999). In the absence of hormone, E7ER is cytoplasmic, and upon addition of 17beta-estradiol, it translocates to the nucleus. Using organotypic epithelial raft cultures developed from primary human keratinocytes, we show that 16E7ER promotes either S-phase reentry or p21cip1 accumulation in differentiated keratinocytes in a stochastic manner as early as 6 h postinduction with 17beta-estradiol. A vector expressing the ER moiety alone had no effect. These observations prove unequivocally that the E7 protein drives S-phase reentry in postmitotic, differentiated keratinocytes rather than preventing S-phase exit while the cells ascend through the epithelium. HPV-11 E7ER and, much less efficiently, HPV-6 E7ER also promoted S-phase reentry by differentiated cells upon exposure to 17beta-estradiol. S-phase induction required the consensus pRB binding motif. We propose that the elevated nuclear levels of the low-risk HPV E7 protein afforded by the inducible system account for the positive results. These observations are entirely consistent with the fact that low-risk HPV genotypes replicate in the differentiated strata in patient specimens, as do the high-risk HPVs.


Subject(s)
Cell Transformation, Viral , Human papillomavirus 16/metabolism , Keratinocytes/metabolism , Oncogene Proteins, Viral/metabolism , Receptors, Estrogen/metabolism , S Phase , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Amino Acid Motifs/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Movement/drug effects , Cell Movement/genetics , Cell Nucleus/metabolism , Cell Transformation, Viral/drug effects , Cell Transformation, Viral/genetics , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytoplasm/metabolism , Epithelium/metabolism , Epithelium/pathology , Epithelium/virology , Estradiol/pharmacology , Genotype , Human papillomavirus 16/genetics , Humans , Keratinocytes/pathology , Keratinocytes/virology , Male , Oncogene Proteins, Viral/genetics , Papillomavirus Infections/genetics , Papillomavirus Infections/metabolism , Papillomavirus Infections/pathology , Protein Binding/drug effects , Protein Binding/genetics , Receptors, Estrogen/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Risk Factors , S Phase/genetics , Time Factors , Virus Replication/drug effects , Virus Replication/genetics
13.
Methods Mol Med ; 119: 187-202, 2005.
Article in English | MEDLINE | ID: mdl-16353336

ABSTRACT

The productive phase of human papillomavirus (HPV) infection is dependent on squamous differentiation of epithelial keratinocytes. Organotypic culture systems of primary human keratinocytes (PHKs) or immortalized keratinocytes that contain HPV genomes were developed to recapitulate this permissive environment. A complementary approach to determine the functions of individual HPV genes and to examine the virus-host interactions is to introduce the gene, alone or in combination, into keratinocytes that are then grown in organotypic cultures. The success of the latter approach depends on the methodology of retrovirus-mediated gene transfer, which can transduce the viral gene or genes into an entire population of PHKs. In this chapter, we describe the strategies and methods of retrovirus-mediated gene transfer into keratinocytes grown into organotypic cultures.


Subject(s)
Gene Expression Regulation, Viral , Gene Transfer Techniques , Keratinocytes/cytology , Keratinocytes/virology , Papillomaviridae/genetics , Retroviridae/genetics , Cell Culture Techniques/methods , Cell Line , Cloning, Molecular , Genetic Vectors , Genome, Viral , Humans , Plasmids , Viral Proteins/genetics
14.
J Virol ; 78(20): 11172-86, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15452237

ABSTRACT

Primary keratinocytes immortalized by human papillomaviruses (HPVs), along with HPV-induced cervical carcinoma cell lines, are excellent models for investigating neoplastic progression to cancer. By simultaneously visualizing viral DNA and nascent viral transcripts in interphase nuclei, we demonstrated for the first time a selection for a single dominant papillomavirus transcription center or domain (PVTD) independent of integrated viral DNA copy numbers or loci. The PVTD did not associate with several known subnuclear addresses but was almost always perinucleolar. Silent copies of the viral genome were activated by growth in the DNA methylation inhibitor 5-azacytidine. HPV-immortalized keratinocytes supertransduced with HPV oncogenes and selected for marker gene coexpression underwent crisis, and the surviving cells transcribed only the newly introduced genes. Thus, transcriptional selection in response to environmental changes is a dynamic process to achieve optimal gene expression for cell survival. This phenomenon may be critical in clonal selection during carcinogenesis. Examination of HPV-associated cancers supports this hypothesis.


Subject(s)
Cell Transformation, Neoplastic , Gene Expression Regulation, Viral , Oncogene Proteins, Viral/metabolism , Papillomaviridae/pathogenicity , Selection, Genetic , Transcription, Genetic , Cell Line, Transformed , Cell Nucleolus/virology , DNA, Viral/analysis , Humans , In Situ Hybridization, Fluorescence , Keratinocytes/virology , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , Tyramine , Virus Integration
15.
J Virol ; 78(17): 9041-50, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15308700

ABSTRACT

The effects of human papillomavirus type 18 (HPV-18) E6 and E7 proteins on global patterns of host gene expression in primary human keratinocytes grown in organotypic raft culture system were assessed. Primary human keratinocytes were infected with retroviruses that express the wild-type HPV-18 E6 and E7 genes from the native differentiation-dependent HPV enhancer-promoter. Total RNA was isolated from raft cultures and used to generate probes for querying Affymetrix U95A microarrays, which contain >12,500 human gene sequences. Quadruplicate arrays of each E6/E7-transduced and empty vector-transduced samples were analyzed by 16 pairwise comparisons. Transcripts altered in > or =12 comparisons were selected for further analysis. With this approach, HPV-18 E6/E7 expression significantly altered the expression of 1,381 genes. A large increase in transcripts associated with DNA and RNA metabolism was observed, with major increases noted for transcription factors, splicing factors, and DNA replication elements, among others. Multiple genes associated with protein translation were downregulated. In addition, major alterations were found in transcripts associated with the cell cycle and cell differentiation. Our study provides a systematic description of transcript changes brought about by HPV-18 E6/E7 in a physiologically relevant model and should furnish a solid source of information to guide future studies.


Subject(s)
Cell Culture Techniques/methods , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation , Keratinocytes/metabolism , Keratinocytes/virology , Oncogene Proteins, Viral/metabolism , Cell Differentiation , Cells, Cultured , DNA/metabolism , DNA Replication , DNA-Binding Proteins/genetics , G2 Phase , Humans , Keratinocytes/cytology , Mitosis , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Viral/genetics , Papillomaviridae , Protein Biosynthesis , RNA, Messenger/analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spindle Apparatus/physiology , Transcription Factors/genetics , Transduction, Genetic
16.
Mol Cancer Ther ; 3(4): 437-49, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15078987

ABSTRACT

We have generated novel conditionally replicative adenoviruses (CRAds) targeted to melanoma cells. In these adenoviruses, the E4 region (AdDelta24TyrE4) or both E1 and E4 regions (Ad2xTyr) were controlled by a synthetic tyrosinase enhancer/promoter (Tyr2E/P) specific for melanocytes. The properties of these CRAds were compared with wild-type adenovirus (Adwt) and our previous CRAd with a targeted E1A CRII mutation (AdTyrDelta24) in submerged cultures of melanoma cells and nonmelanoma control cells. We showed that AdDelta24TyrE4 had a cell type selectivity similar to AdTyrDelta24 but had a distinct block in viral reproduction in nonmelanoma cells and that Ad2xTyr had an augmented selectivity for melanoma cells. These viruses were additionally tested in organotypic cultures of melanoma cell lines, primary human keratinocytes (PHKs), or mixed cell populations. Unexpectedly, the CRAds exhibited somewhat different cell type selectivity profiles in these cultures relative to those observed in submerged cultures, demonstrating the importance of multiple assay systems. Specifically, AdTyrDelta24 and Ad2xTyr were selective for melanoma cells, whereas AdDelta24TyrE4 exhibited no selectivity, similar to Adwt. AdTyrDelta24 and Ad2xTyr were strongly attenuated in their ability to lyse PHKs in organotypic cultures. Furthermore, Ad2xTyr had a superior melanoma selectivity in organotypic cultures of cocultivated melanoma cells and PHKs. The enhanced selectivity for melanoma cells exhibited by Ad2xTyr provides a window of opportunity for therapeutic application. These studies also demonstrate that organotypic cultures derived from mixtures of tumor and normal cells represent a promising new model for analysis of CRAd specificity and toxicity.


Subject(s)
Adenovirus E1A Proteins/genetics , Adenovirus E4 Proteins/genetics , Adenoviruses, Human/physiology , Enhancer Elements, Genetic/genetics , Melanoma/therapy , Melanoma/virology , Monophenol Monooxygenase/genetics , Promoter Regions, Genetic/genetics , Adenovirus E1A Proteins/metabolism , Adenovirus E4 Proteins/metabolism , Adenoviruses, Human/genetics , Adenoviruses, Human/growth & development , Adenoviruses, Human/metabolism , Cell Line, Tumor , Cell Survival , Cells, Cultured , Coculture Techniques , DNA Replication , Genetic Vectors/genetics , Genetic Vectors/isolation & purification , Genetic Vectors/metabolism , Genetic Vectors/toxicity , Humans , Keratinocytes/virology , Membrane Microdomains , Microscopy, Fluorescence , Organ Specificity , Virus Replication
17.
J Virol ; 77(11): 6533-40, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12743310

ABSTRACT

Human oncolytic adenoviruses have been used in clinical trials targeting cancers of epithelial origin. To gain a better understanding of the infectious cycle of adenovirus in normal human squamous tissues, we examined the viral infection process in organotypic cultures of primary human keratinocytes. We show that for the infection to occur, wounding of the epithelium is required. In addition, infection appears to initiate at the basal or parabasal cells that express the high-affinity coxsackievirus-adenovirus receptor, CAR, whereas the productive phase takes place in differentiated cells. This is due, at least in part, to the differentiation-dependent activation of the E1A and E2A early promoters and E4 promoters. We also show that adenovirus infection triggers a response mediated by the abnormal accumulation of cyclin E and p21cip1 proteins similar to the one previously observed in human papillomavirus-infected tissues. However, the virus seems to be able to overcome it, at least partially.


Subject(s)
Adenoviridae/pathogenicity , Adenovirus Early Proteins/metabolism , Gene Expression Regulation, Viral , Keratinocytes/virology , Virus Replication , Adenoviridae/physiology , Adenovirus Early Proteins/genetics , Cell Differentiation , Cells, Cultured , Cyclin E/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Epithelium/injuries , Humans , Keratinocytes/cytology , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...