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










Publication year range
2.
Cancer Biol Ther ; 19(12): 1174-1184, 2018.
Article in English | MEDLINE | ID: mdl-30067431

ABSTRACT

Oncolytic adenoviruses (Ads) are cancer selective tumoricidal agents; however their mechanism of Ad-mediated cancer cell lysis, or oncolysis, remains undefined. This report focuses upon the autophagy mediator c-JUN n-terminal kinase (JNK) and its effects upon Ad oncolysis and replication. Previously, E1b-deleted Ads have been used to treat several hundred cancer patients with limited clinical efficacy. We hypothesize that by studying the potential interactions between E1b and JNK, mechanisms to improve oncolytic Ad design and cancer therapeutic efficacy may be elucidated. To test this hypothesis, E1b was selectively deleted from the Ad genome. These studies indicated that Ads encoding E1b induced JNK phosphorylation predominately occurred via E1b-19K. The expression of another crucial Ad gene E1a was then overexpressed by the CMV promoter via the replication competent Ad vector Adhz69; these data indicated that E1A also induced JNK phosphorylation. To assess the effects of host cell JNK expression upon Ad oncolysis and replication, siRNA targeting JNK1 and JNK2 (JNK1/2) were utilized. The oncolysis and replication of the E1b-19K wild-type Ads Ad5 and Adhz63 were significantly attenuated following JNK1/2 siRNA transfection. However the oncolytic effects and replication of the E1b-19K deleted Ad Adhz60 were not altered by JNK1/2 siRNA transfection, further implicating the crucial role of E1b-19K for Ad oncolysis and replication via JNK phosphorylation. This study has demonstrated for the first time that JNK is an intriguing molecular marker associated with enhanced Ad virotherapy efficacy, influencing future Ad vector design.


Subject(s)
Adenoviridae , Genetic Vectors , JNK Mitogen-Activated Protein Kinases/metabolism , Neoplasms/metabolism , Oncolytic Virotherapy , Oncolytic Viruses , Virus Replication , Adenoviridae/genetics , Adenovirus E1B Proteins/genetics , Adenovirus E1B Proteins/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Expression , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Host-Pathogen Interactions , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Mice , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Phosphorylation , Xenograft Model Antitumor Assays
3.
Viruses ; 8(12)2016 12 20.
Article in English | MEDLINE | ID: mdl-27999391

ABSTRACT

Adenoviruses (Ads) have been extensively manipulated for the development of cancer selective replication, leading to cancer cell death or oncolysis. Clinical studies using E1-modified oncolytic Ads have shown that this therapeutic platform was safe, but with limited efficacy, indicating the necessity of targeting other viral genes for manipulation. To improve the therapeutic efficacy of oncolytic Ads, we treated the entire Ad genome repeatedly with UV-light and have isolated AdUV which efficiently lyses cancer cells as reported previously (Wechman, S. L. et al. Development of an Oncolytic Adenovirus with Enhanced Spread Ability through Repeated UV Irradiation and Cancer Selection. Viruses2016, 8, 6). In this report, we show that no mutations were observed in the early genes (E1 or E4) of AdUV while several mutations were observed within the Ad late genes which have structural or viral DNA packaging functions. This study also reported the increased release of AdUV from cancer cells. In this study, we found that AdUV inhibits tumor growth following intratumoral injection. These results indicate the potentially significant role of the viral late genes, in particular the DNA packaging genes, to enhance Ad oncolysis.


Subject(s)
Adenoviridae/genetics , Adenoviridae/physiology , DNA Packaging , Mutation , Oncolytic Viruses/genetics , Oncolytic Viruses/physiology , Virus Release , Adenoviridae/radiation effects , DNA Mutational Analysis , Oncolytic Viruses/radiation effects , Ultraviolet Rays , Viral Proteins/genetics
4.
Viruses ; 7(11): 5767-79, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26561828

ABSTRACT

Various viruses have been studied and developed for oncolytic virotherapies. In virotherapy, a relatively small amount of viruses used in an intratumoral injection preferentially replicate in and lyse cancer cells, leading to the release of amplified viral particles that spread the infection to the surrounding tumor cells and reduce the tumor mass. Adenoviruses (Ads) are most commonly used for oncolytic virotherapy due to their infection efficacy, high titer production, safety, easy genetic modification, and well-studied replication characteristics. Ads with deletion of E1b55K preferentially replicate in and destroy cancer cells and have been used in multiple clinical trials. H101, one of the E1b55K-deleted Ads, has been used for the treatment of late-stage cancers as the first approved virotherapy agent. However, the mechanism of selective replication of E1b-deleted Ads in cancer cells is still not well characterized. This review will focus on three potential molecular mechanisms of oncolytic replication of E1b55K-deleted Ads. These mechanisms are based upon the functions of the viral E1B55K protein that are associated with p53 inhibition, late viralmRNAexport, and cell cycle disruption.


Subject(s)
Adenoviridae/physiology , Adenovirus E1B Proteins/deficiency , Oncolytic Viruses/physiology , Virus Replication , Adenoviridae/genetics , Adenovirus E1B Proteins/genetics , Gene Deletion , Humans , Oncolytic Viruses/genetics
5.
BMC Cancer ; 15: 716, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26475304

ABSTRACT

BACKGROUND: Clinical trials have indicated that preclinical results obtained with human tumor xenografts in mouse models may overstate the potential of adenovirus (Ad)-mediated oncolytic therapies. We have previously demonstrated that the replication of human Ads depends on cyclin E dysregulation or overexpression in cancer cells. ED-1 cell derived from mouse lung adenocarcinomas triggered by transgenic overexpression of human cyclin E may be applied to investigate the antitumor efficacy of oncolytic Ads. METHODS: Ad-cycE was used to target cyclin E overexpression in ED-1 cells and repress tumor growth in a syngeneic mouse model for investigation of oncolytic virotherapies. RESULTS: Murine ED-1 cells were permissive for human Ad replication and Ad-cycE repressed ED-1 tumor growth in immunocompetent FVB mice. ED-1 cells destroyed by oncolytic Ads in tumors were encircled in capsule-like structures, while cells outside the capsules were not infected and survived the treatment. CONCLUSION: Ad-cycE can target cyclin E overexpression in cancer cells and repress tumor growth in syngeneic mouse models. The capsule structures formed after Ad intratumoral injection may prevent viral particles from spreading to the entire tumor.


Subject(s)
Adenocarcinoma/therapy , Cyclin E/biosynthesis , Lung Neoplasms/therapy , Oncolytic Virotherapy , Adenocarcinoma/genetics , Adenoviridae/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin E/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Isografts , Lung Neoplasms/genetics , Mice , Oncolytic Viruses , Xenograft Model Antitumor Assays
6.
Cancer Biol Ther ; 15(9): 1256-67, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24972095

ABSTRACT

Epidemiological studies suggest that high intake of cruciferous vegetables is associated with a lower risk of cancer. Experiments have shown that indole-3-carbinol (I3C), a naturally occurring compound derived from cruciferous vegetables, exhibits potent anticarcinogenic properties in a wide range of cancers. In this study, we showed that higher doses of I3C (≥400 µM) induced apoptotic cancer cell death and lower doses of I3C (≤200 µM) repressed cancer cell growth concurrently with suppressed expression of cyclin E and its partner CDK2. Notably, we found that pretreatment with low doses of I3C enhanced Ad-mediated oncolysis and cytotoxicity of human carcinoma cells by synergistic upregulation of apoptosis. Thus, the vegetable compound I3C as a dietary supplement may benefit cancer prevention and improve Ad oncolytic therapies.


Subject(s)
Adenoviridae , Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Indoles/pharmacology , Oncolytic Viruses , Vegetables/chemistry , Cell Cycle Checkpoints , Cell Line, Tumor , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Drug Synergism , Humans
7.
Virol J ; 10: 293, 2013 Sep 23.
Article in English | MEDLINE | ID: mdl-24059864

ABSTRACT

BACKGROUND: Combination of oncolytic adenoviruses (Ads) and chemotherapy drugs has shown promising therapeutic results and is considered as a potential approach for cancer therapy. We previously have shown that autophagy may generate decomposed cellular molecules that can be used as nutrition to support virus replication in cancer cells. In this study, we evaluated a unique combination of the novel oncolytic Ad-cycE with rapamycin, an autophagy inducer and first-line chemotherapeutic drug. METHODS: The combination of oncolytic Ad-cycE and the autophagy inducer rapamycin was assessed for enhanced antitumor effect. We also evaluated the combined effects of rapamycin and Ad-cycE on cancer cell viability. The interaction between Ad-cycE and rapamycin was analyzed with Calcusyn (Biosoft, Ferguson, MO). RESULTS: We show that rapamycin induces autophagy, enhances Ad E1A expression and increases Ad oncolytic replication. Combination of rapamycin and Ad-cycE elicits stronger cytotoxicity than single treatment alone. The analyzed data indicates that the Ad-cycE and rapamycin combination has a significantly synergistic antitumor effect. CONCLUSIONS: Our study provides a new insight into vector development and demonstrates the novel roles of autophagy in adenovirus replication. The combination of autophagy-induced chemotherapy and oncolytic virotherapy may be a new approach to improve future cancer treatment.


Subject(s)
Adenoviridae/growth & development , Autophagy , Oncolytic Viruses/growth & development , Sirolimus/metabolism , Adenoviridae/physiology , Cell Line , Cell Survival , Humans , Viral Load , Virus Replication
8.
PLoS One ; 8(2): e57340, 2013.
Article in English | MEDLINE | ID: mdl-23437375

ABSTRACT

Adenoviruses (Ads) with deletion of E1b55K preferentially replicate in cancer cells and have been used in cancer therapies. We have previously shown that Ad E1B55K protein is involved in induction of cyclin E for Ad replication, but this E1B55K function is not required in cancer cells in which deregulation of cyclin E is frequently observed. In this study, we investigated the interaction of cyclin E and CDK2 in Ad-infected cells. Ad infection significantly increased the large form of cyclin E (cyclin EL), promoted cyclin E/CDK2 complex formation and increased CDK2 phosphorylation at the T160 site. Activated CDK2 caused pRb phosphorylation at the S612 site. Repression of CDK2 activity with the chemical inhibitor roscovitine or with specific small interfering RNAs significantly decreased pRb phosphorylation, with concomitant repression of viral replication. Our results suggest that Ad-induced cyclin E activates CDK2 that targets the transcriptional repressor pRb to generate a cellular environment for viral productive replication. This study reveals a new molecular basis for oncolytic replication of E1b-deleted Ads and will aid in the development of new strategies for Ad oncolytic virotherapies.


Subject(s)
Adenoviridae/genetics , Cyclin E/genetics , Cyclin-Dependent Kinase 2/genetics , Gene Expression Regulation, Neoplastic/drug effects , Oncogene Proteins/genetics , Retinoblastoma Protein/genetics , Viral Proteins/genetics , Adenoviridae/metabolism , Cell Line, Tumor , Cyclin E/agonists , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/virology , Genes, Reporter , Green Fluorescent Proteins , HEK293 Cells , Host-Pathogen Interactions , Humans , Oncogene Proteins/agonists , Oncogene Proteins/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , RNA, Small Interfering/genetics , Retinoblastoma Protein/antagonists & inhibitors , Retinoblastoma Protein/metabolism , Roscovitine , Viral Proteins/metabolism , Virus Replication/drug effects
9.
Virology ; 433(2): 538-47, 2012 Nov 25.
Article in English | MEDLINE | ID: mdl-23021422

ABSTRACT

Adenovirus-mediated gene transfer into a tumor mass can be improved by combining it with conditionally-replicating adenovirus (CRAd) when both vectors co-infect the same cancer cell. We investigated the efficiency of enhancing transgene expression and effectiveness of cancer killing of two advenoviruses (Ads), one expressing E2F-1 (AdE2F-1) and another expressing a truncated form of E2F-1 that lacks the transactivation domain (AdE2Ftr), when combined with oncolytic Adhz60. We found that AdE2F-1 with Adhz60 actually decreased E2F-1 expression and viral replication through a mechanism apparently involving repression of the cyclin-E promoter and decreased expression of early and late structural proteins necessary for viral replication. In contrast, AdE2Ftr with Adhz60 resulted in increased E2Ftr expression, AdE2Ftr replication, and cancer cell death both in vitro and in vivo. These results indicate that AdE2Ftr coupled with a CRAd enhances AdE2Ftr-mediated cancer cell death.


Subject(s)
Adenoviridae/genetics , E2F1 Transcription Factor/genetics , Neoplasms/therapy , Oncolytic Viruses/genetics , Adenoviridae/physiology , Adenovirus E1A Proteins/metabolism , Animals , Cell Death , Cell Line, Tumor , Cyclin E/genetics , Cyclin E/metabolism , Down-Regulation , Gene Expression , Humans , Mice , Mice, Nude , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Promoter Regions, Genetic , Transduction, Genetic , Virus Replication , Xenograft Model Antitumor Assays
10.
Cancer Biol Ther ; 13(12): 1195-204, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22892845

ABSTRACT

Melanoma is generally resistant to chemotherapy, which may be related to defects in death receptor signaling and to defects in induction of apoptosis. Forkhead family transcription factors induce the expression of death receptor ligands such as Fas ligand (Fas-L) resulting in apoptosis. We therefore investigated whether a triple mutant form of forkhead transcription factor FKHRL1 (FKHRL1/TM) can enhance Fas-L mediated-apoptosis in melanoma cells. Two melanoma cells A2058 or DM6 were tested for their sensitivity to agonistic anti-Fas antibody (CH-11); adenovirus expressing FKHRL1/TM (Ad-FKHRL1/TM) was assessed for its capability to induce activation of the caspase pathway; the role of Fas-L in the Ad-FKHRL1/TM mediated-cell death was also assessed in vitro. Ad-FKHRL1/TM antitumor activity in vivo was also evaluated in a mouse melanoma xenograft model. We found that DM6 melanoma cells were more resistant to Fas/Fas-L-mediated apoptosis induced by agonistic anti-Fas antibody than A2058 melanoma cells. Ectopic expression of FKHRL1/TM in melanoma cells upregulated Fas-L expression, decreased procaspase-8 levels, and significantly increased Fas/FasL-mediated cell death in both cells lines; this induced cell death was partially blocked by a Fas/Fas-L antagonist. Importantly, Ad-FKHRL1/TM treatment of subcutaneous melanoma xenografts in mice resulted in approximately 70% decrease in tumor size compared with controls. These data indicate that overexpression of FKHRL1/TM can induce the Fas-L pathway in melanoma cells. Ad-FKHRL1/TM therefore might represent a promising vector for melanoma treatment.


Subject(s)
Apoptosis/genetics , Fas Ligand Protein/metabolism , Forkhead Transcription Factors/genetics , Melanoma , Adenoviridae , Animals , Cell Survival , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Melanoma/genetics , Melanoma/metabolism , Mice , Muscle, Smooth, Vascular/cytology , Mutation , Signal Transduction , Xenograft Model Antitumor Assays
11.
Virology ; 416(1-2): 9-15, 2011 Jul 20.
Article in English | MEDLINE | ID: mdl-21575980

ABSTRACT

Adenoviruses with deletion of E1b have been used in clinical trials to treat cancers that are resistant to conventional therapies. The efficacy of viral replication within cancer cells determines the results of oncolytic therapy, which remains poorly understood and requires further improvement. In this report, we show that adenoviruses induce autophagy by increasing the conversion of LC3-I to LC3-II and the formation of the Atg12-Atg5 complex. Inhibition of autophagy with 3-methyladenine (3MA) resulted in a decreased synthesis of adenovirus structural proteins, and thereby a poor viral replication; promotion of autophagy with rapamycin increased adenovirus yield. This study indicates that adenovirus-induced autophagy correlates positively with virus replication and oncolytic cell death, and that autophagy may generate nutrients that can be used for building viral progeny particles. These results further suggest that chemotherapeutic agents that increase cancer cell autophagy may improve the efficacy of oncolytic virotherapy.


Subject(s)
Adenoviridae/physiology , Autophagy/physiology , Virus Replication/physiology , Adenine/analogs & derivatives , Adenine/pharmacology , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Adenovirus E1B Proteins/genetics , Adenovirus E1B Proteins/metabolism , Autophagy-Related Protein 12 , Autophagy-Related Protein 5 , Cell Line, Tumor , Gene Expression Regulation, Viral , Humans , Microtubule-Associated Proteins/metabolism , Mutation , Small Ubiquitin-Related Modifier Proteins/metabolism
12.
Tumour Biol ; 28(2): 111-22, 2007.
Article in English | MEDLINE | ID: mdl-17287612

ABSTRACT

OBJECTIVES: In the present study, we investigate the role of apoptosis signal-regulating kinase 1 (ASK1) mitogen-activated protein (MAP) kinase signal pathways in E2F-1-mediated apoptosis. METHODS: A gene expression profile in response to E2F-1 overexpression was performed by cDNA microarray analysis and confirmed by real-time reverse-transcription polymerase chain reaction. Kinase activities were assayed by Western blot analysis or kinase assay. Apoptosis was assessed by morphologic inspection and flow-cytometric analysis. Cytotoxicity was monitored by MTT assay. RESULTS: E2F-1 upregulated the expression of ASK1 8-fold compared to the Ad-LacZ-infected control in SK-MEL-2 melanoma cells, which was confirmed by reverse-transcription polymerase chain reaction. Sequence analysis showed that there are 2 putative E2F-1 DNA binding sites in the ASK1 promoter region. Truncated E2F-1 protein, which lacks the transactivation domain, failed to upregulate ASK1, suggesting that ASK1 was regulated at the transcriptional level by E2F-1. E2F-1 overexpression resulted in the transient activation of c-Jun N-terminal kinase (JNK); however, dominant negative mutant ASK1 had no effect on E2F-1 cytotoxicity and JNK activation. p38 was not activated by E2F-1, and inhibition of p38 had no effect on E2F-1-mediated cell death. The ASK1 kinase assay showed that ASK1 activity was not upregulated in response to E2F1 overexpression. The inhibition of ASK1 upstream kinase-AKT can enhance E2F-1-mediated cell death. Moreover, an adenovirus expressing truncated E2F-1 keeps the ability of inducing apoptosis in melanoma cells. CONCLUSIONS: ASK1 expression is upregulated by E2F-1 at the transcription level, but the upregulation of ASK1 expression by E2F-1 was not coordinated with an increased ASK1 activity. The ASK1-JNK/p38 pathway does not appear to play a crucial role in E2F-1-induced apoptosis.


Subject(s)
Apoptosis , E2F1 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , MAP Kinase Kinase Kinase 5/metabolism , Melanoma/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , E2F1 Transcription Factor/genetics , Gene Expression Profiling , Humans , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinase 5/genetics , Melanoma/genetics , Melanoma/pathology , Oligonucleotide Array Sequence Analysis , Signal Transduction , Transfection , Tumor Cells, Cultured , Up-Regulation , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Virology ; 350(2): 418-28, 2006 Jul 05.
Article in English | MEDLINE | ID: mdl-16542696

ABSTRACT

Adenoviruses with deletion of E1b gene can selectively replicate in cancer cells. The underlying mechanisms in tumor-selective replication of E1b-deleted adenoviruses are insufficiently understood. Identifying genes with altered expression patterns caused by the E1B proteins in virus-infected cells will further increase our understanding of E1B functions and provide insight into the tumor-selective replication of E1b-mutated adenoviruses on the molecular level. An approach based on large-scale gene array was applied to analyze molecular changes affected by viral E1B. We identified a total of 345 genes with expression changes of two-fold or greater affected by wild-type adenovirus compared with its E1b-deleted counterpart. The gene array data were confirmed by quantitative real-time PCR and Western blot. E1B proteins affect the expression of a diverse range of genes involved in cell cycle regulation, apoptosis, stress responses and angiogenesis. This is the first study of the global profile of gene expression altered by the viral E1B proteins in human lung cells, and the majority of the genes were previously not known to be affected by the viral proteins. The data presented in this study will lead to more detailed analysis of E1B functions and may also lead to development of new agents and approaches for oncolytic therapy.


Subject(s)
Adenovirus E1B Proteins/genetics , Gene Expression Profiling , Lung/virology , Enzymes/genetics , Gene Expression Regulation , Humans , Nucleic Acid Hybridization , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics
14.
Gene ; 344: 67-77, 2005 Jan 03.
Article in English | MEDLINE | ID: mdl-15656974

ABSTRACT

It has been shown that adenovirus-mediated overexpression of E2F-1 can efficiently induce apoptosis in cancer cells with little effect on normal cells. However, the mechanisms by which E2F-1 induces apoptosis remains poorly understood. The goal of this study was to evaluate changes in gene expression in response to E2F-1 in order to help elucidate the mechanisms by which E2F-1 causes apoptosis. Therefore, we used a quantitative microarray assay to identify the genes regulated by E2F-1 in melanoma cells. By gene expression profiling, we first screened a proprietary list of about 12,000 genes. Overexpression of E2F-1 in melanoma cells resulted in two-fold or greater alteration in the level of expression of 452 genes compared to vehicle-treated control cells. Most of the affected genes were not known to be responsive to E2F-1 prior to this study. E2F-1 adenoviral infection of these cells was found to affect the expression of a diverse range of genes, including oncogenes, transcription factors and genes involved in signal transduction, cell cycle regulation, cell proliferation and apoptosis, as well as other genes with unknown function. Changes in expression of 17 of these genes were confirmed by quantitative real-time polymerase chain reaction (PCR). This is first application of the microarray technique in the study of the global profile of genes regulated by E2F-1 in melanoma cells. This study leads to an increased understanding of the biochemical pathways involved in E2F-1-induced apoptosis and possibly to the identification of new therapeutic targets.


Subject(s)
Apoptosis/genetics , Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , Gene Expression Profiling , Transcription Factors/physiology , Adenoviridae/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cluster Analysis , DNA-Binding Proteins/genetics , E2F Transcription Factors , E2F1 Transcription Factor , Genetic Vectors/genetics , Humans , Melanoma/genetics , Melanoma/pathology , Oligonucleotide Array Sequence Analysis/methods , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription Factors/genetics , Transfection
15.
Cancer Gene Ther ; 11(9): 585-93, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15338010

ABSTRACT

Apoptotic pathways are initiated as a cellular defense mechanism to eliminate adenovirus-infected cells. We have investigated how E1A-induced apoptosis interferes with viral replication in cancer cells. We found that E1B19K alone can efficiently suppress E1A-induced apoptosis in cancer cells. Viruses deleted for both E1B19K and E1B55K resulted in cellular DNA degradation. However, less than 20% of human lung cancer cells infected with a virus deleted for both E1B19K and E1B55 K had evidence of chromatin condensation and multiple-micronuclei formation (apoptotic hallmarks); these cells could not produce infectious viral particles. The majority of cancer cells infected with viruses deleted for the entire E1b gene did not undergo extended apoptosis and produced abundant viral progeny. Thus, only a fraction of cancer cells underwent apoptosis and did not allow E1b-deleted viruses to replicate, while the majority of cancer cells were resistant to E1A-induced apoptosis and could support virus-selective replication. The results of this study imply that, in addition to inhibiting E1A-induced apoptosis, E1B proteins may contribute other important roles in the viral life cycle. Our results also suggest that combining virus-induced apoptosis and selective viral replication into one vector will be a novel approach to destroy cancer cells.


Subject(s)
Adenoviridae Infections/virology , Adenovirus E1A Proteins/physiology , Adenovirus E1B Proteins/genetics , Adenoviruses, Human/physiology , Apoptosis , Gene Deletion , Lung Neoplasms/virology , Virus Replication , Adenoviridae Infections/pathology , Blotting, Western , Chromatin/metabolism , DNA, Viral/genetics , DNA, Viral/metabolism , Genetic Vectors , Humans , Lung Neoplasms/pathology , Micronuclei, Chromosome-Defective , Tumor Cells, Cultured
16.
Cancer Res ; 63(12): 3073-8, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12810631

ABSTRACT

The adenovirus E1A proteins are involved in the transcriptional activation of viral and cellular genes needed for controlling cell cycle and virus replication. Undifferentiated embryonic carcinoma cells have the ability to produce an E1A-like activity that can induce the expression of E1A-targeted adenoviral and cellular genes in the absence of the E1A products. Differentiated embryonic carcinoma cells lose the ability to produce the E1A-like activity. In this study, we investigated the E1A-like activity in cancer cells with an adenovirus having a mutated E1a gene. The mutation is generated by the insertion of a large DNA fragment in the E1a gene and interrupts the COOH-terminal region of both the E1A 12S and 13S proteins. The E1a-mutated virus can efficiently replicate in HepG2 and Hep3B liver cancer cells and produce high titers of virus. Replication of the E1a-mutated virus inhibits tumor formation and destroys tumors in vivo. The results obtained in this study imply that cancer cells may produce an E1A-like activity to support the selective replication of mutated virus in cancer cells. In addition, we found that although the E1a-mutated virus could not replicate in Huh1.cl2 liver cells, the viral DNA could amplify in the cells. This result suggests that replication of adenoviral DNA is necessary, but not sufficient, for generating infectious viral progeny and destroying tumor cells.


Subject(s)
Adenovirus E1A Proteins/genetics , Biological Therapy , Carcinoma, Hepatocellular/pathology , Defective Viruses/physiology , Liver Neoplasms/pathology , Mastadenovirus/physiology , Adenovirus E1A Proteins/deficiency , Adenovirus E1A Proteins/physiology , Animals , Carcinoma, Hepatocellular/therapy , Cytopathogenic Effect, Viral , DNA, Viral/genetics , Defective Viruses/genetics , Humans , Liver Neoplasms/therapy , Liver Neoplasms/virology , Mastadenovirus/genetics , Mice , Mice, Nude , Mutagenesis, Insertional , Tumor Cells, Cultured/virology , Virus Integration , Virus Replication , Xenograft Model Antitumor Assays
17.
J Gene Med ; 4(5): 498-509, 2002.
Article in English | MEDLINE | ID: mdl-12221643

ABSTRACT

BACKGROUND: The helper-dependent (HD) adenoviral (Ad) vector relies on a helper virus to provide viral proteins for vector amplification. HD-Ad vectors can significantly increase therapeutic gene expression and improve safety. However, the yield of an HD-Ad vector is generally lower than that of an E1-deleted first-generation vector, likely due to the alterations in viral E3 or packaging regions of a helper virus that attenuate its replication and complementing for an HD-Ad vector. METHODS: To study this question and improve HD-Ad vector production, we have generated four different helper viruses with a wild-type or deleted E3 region, and with a relocated loxP. We have also constructed a first-generation vector with a wild-type E3 region and without the loxP site. We compared the replication of these viruses in Cre-positive and -negative cells and studied their complementing for HD-Ad vector production. RESULTS: Viruses with deleted E3 formed smaller plaques and produced lower titer compared with viruses containing the E3 region. The site where a loxP is inserted can also affect virus replication. Higher yield of HD-Ad vector was obtained when a helper virus with wild-type E3 was used. We also showed that deletion of the packaging signal in a helper virus through loxP/Cre interaction decreased the viral DNA complementing ability. CONCLUSIONS: Although the E3 region is not essential for adenovirus replication in vivo, deletion of this region attenuates virus replication. Production of HD-Ad vector can be further improved by modifications in helper virus structure.


Subject(s)
Adenoviridae/genetics , Genetic Vectors , Helper Viruses/genetics , Adenoviridae/growth & development , Adenovirus E3 Proteins/genetics , Helper Viruses/growth & development , Integrases/metabolism , Plasmids/genetics , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...