Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
J Gene Med ; 13(9): 487-96, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21850667

ABSTRACT

BACKGROUND: Epstein-Barr virus (EBV) causes a range of life-threatening B-lymphocyte malignancies but, despite the use of various strategies, treatment remains problematic. METHODS: In the present study, we developed a non-integrating lentiviral vector (NILV) that mediates specific killing of EBV nuclear antigen 1 (EBNA1)-expressing cells with minimal toxicity to EBNA1-negative cells. The EBV family of repeats (FR) was cloned intok the NILV genome upstream of various transgenes. RESULTS: The presence of the FR in the NILV genome induced transcriptional up-regulation and prolonged the expression of a transgene specifically in EBNA1-positive B cells. Transgene expression from an FR-containing NILV was also prolonged in EBV-transformed cells compared to an FR-negative NILV. We found that the delivery of an FR-containing NILV encoding herpes simplex virus 1 thymidine kinase (TK) lead to the killing of more than 99% of EBNA1-positive B cells with minimal toxicity to EBNA1-negative cells in the presence of gancyclovir. EBNA1-positive cells were not killed by an FR-negative vector containing the TK gene. An FR-TK-containing NILV also specifically killed EBNA1-containing cells in a mixed population of EBNA1-positive and EBNA1-negative cells, thus confirming that NILV-FR-TK-mediated killing is specific for EBNA1-expressing cells. CONCLUSIONS: Transgene expression from our NILVs is both EBNA1-specific and dependent upon the presence of the FR. The results obtained in the present study indicate that NILVs have potential use in the treatment of EBV-associated B cell malignancies.


Subject(s)
Epstein-Barr Virus Nuclear Antigens/metabolism , Genes, Transgenic, Suicide , Genetic Vectors/genetics , HIV-1/genetics , Lymphoma, B-Cell/therapy , Cell Line, Transformed , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Ganciclovir/pharmacology , Gene Expression Regulation , Gene Order , Gene Transfer Techniques , Genetic Therapy , HEK293 Cells , Humans , Lymphoma, B-Cell/virology , Thymidine Kinase/genetics , Transcription, Genetic , Transduction, Genetic , Up-Regulation/genetics
3.
J Gene Med ; 13(6): 342-52, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21674737

ABSTRACT

BACKGROUND: Adeno-associated virus type 2 (AAV) has the ability to target integration of its DNA into a specific locus of the human genome. Site-specific AAV integration is mediated by viral Rep proteins, although the role of cellular factors involved in this process is largely unknown. Recent studies provide evidence showing that cellular DNA repair proteins are involved in targeted integration of AAV, although their specific roles are not well defined. METHODS: In the present study, we investigated the interaction between Rep and proteins of the back-up nonhomologous end-joining pathway (B-NHEJ). We then analyzed the effect of one of these proteins, poly(ADP-ribose) polymerase 1 (PARP1) on AAV integration. RESULTS: We show that AAV Rep interacts with B-NHEJ members DNA ligase III and PARP1 but does not associate with the scaffolding factor XRCC1. Moreover, PARP1 and Rep bind directly and not via DNA-protein interactions. We also found that Rep increases the enzymatic activity of PARP1 potentially through the endonuclease activity of Rep. Finally, we demonstrate that both chemical inhibition of PARP1 and PARP1 depletion using small hairpin RNA enhance integration of the AAV genome in HeLa cells. CONCLUSIONS: The findings of the present study indicate that manipulation of PARP1 activity could be used as a tool for developing new, effective AAV-based therapies for the treatment of genetic diseases and cancer.


Subject(s)
DNA-Binding Proteins/physiology , Dependovirus/genetics , Genome, Viral/genetics , Poly(ADP-ribose) Polymerases/physiology , Viral Proteins/physiology , Virus Integration/physiology , Blotting, Southern , Blotting, Western , DNA Primers/genetics , DNA-Binding Proteins/metabolism , Genetic Vectors/genetics , HeLa Cells , Humans , Immunoprecipitation , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Polymerase Chain Reaction , Viral Proteins/metabolism
4.
Hum Gene Ther ; 22(11): 1445-51, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21506732

ABSTRACT

Adeno-associated viral (AAV) vectors preferentially integrate into the genome of cells that are defective in DNA repair, such as occurs with DNA-PKcs deficiency or poly(ADP-ribose) polymerase-1 down-regulation. As the tumor suppressor protein p53 regulates the transcription of many genes involved in DNA repair, we sought to determine whether functional p53 affects the efficiency of AAV integration. p53 is mutated in more than 50% of cancers, and site-specific integration of AAV into the AAVS1 site of human chromosome 19 has frequently been observed in transformed cancer cell lines, but rarely seen in primary cells or in vivo. We therefore hypothesized that p53-negative cells would be more permissive to AAV integration than p53-positive cells. The integration of a rep- and green fluorescent protein-encoding recombinant AAV vector was quantified in p53-expressing and p53-deficient HCT116 colon cancer cells. Our results show that there is a higher efficiency of AAV integration in p53-negative cells compared with p53-positive cells, indicating that p53 does indeed inhibit AAV integration. Further experiments suggest that this p53-mediated block to AAV integration is likely to be through binding of p53 to the AAV Rep protein and the consequent inhibition of Rep activity during AAV integration.


Subject(s)
Dependovirus/physiology , Tumor Suppressor Protein p53/genetics , Dependovirus/genetics , Genetic Vectors , HCT116 Cells , HeLa Cells , Humans , Tumor Suppressor Protein p53/metabolism , Virus Integration
5.
Mol Ther ; 15(11): 1947-54, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17700544

ABSTRACT

Human immunodeficiency virus (HIV)-based lentiviral vectors (LVs) hold immense promise for gene delivery applications because of their relatively large packaging capacity and their ability to infect a range of cell types. The genome of HIV non-specifically integrates into the host genome, and this promotes efficient, stable transgene expression in dividing cells. However, integration can also be problematic because of variations in gene expression among cells, possible gene silencing and, most importantly, insertional mutagenesis which can lead to undesirable effects such as malignant transformation. In order to alleviate these problems, we have developed a range of non-integrating LVs (NILVs) by introducing point mutations into the catalytic site, chromosome binding site, and viral DNA binding site of the viral integrase (IN). In addition, we have mutated the IN attachment (att) sites within the HIV long terminal repeats (LTRs). All of the vectors produced show efficient reverse transcription and transgene expression in dividing cells and prolonged expression in non-dividing myotubes. Finally, we show that NILV can be used for achieving highly effective gene transfer and expression in muscle in vivo.


Subject(s)
Genetic Vectors/genetics , Lentivirus/genetics , Muscles/metabolism , Transduction, Genetic/methods , Animals , Cell Line , DNA, Viral/genetics , Gene Expression , Genome, Viral/genetics , Humans , Integrases/chemistry , Integrases/genetics , Integrases/metabolism , Mice , Mutation/genetics , Transgenes/genetics
6.
Hum Gene Ther ; 18(6): 483-9, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17523890

ABSTRACT

Vectors based on lentiviruses have become potent tools for efficient gene transfer to multiple cell types both in vitro and in vivo. In part this is attributable to the stability of transduction afforded by integration into the target cell genome. However, evidence indicates that episomal forms of the vector can also be harnessed for effective gene expression. Nonintegrating vectors retain the high transduction efficiency and broad tropism of conventional lentiviruses but avoid the potential problems associated with the nonspecific integration of a transgene. In this respect they are particularly useful in postmitotic tissue because the vector genome is not diluted out through cell division. Here we discuss the various mutations that may be introduced into human immunodeficiency virus-based lentiviral vectors to achieve efficient transduction, and the mechanisms by which these vectors are effective. We also discuss their potential application to gene therapy and the treatment of genetic disease.


Subject(s)
Genetic Therapy/methods , Genetic Vectors , Lentivirus/genetics , Virus Integration/genetics , Animals , Humans
7.
Proc Natl Acad Sci U S A ; 101(16): 6200-5, 2004 Apr 20.
Article in English | MEDLINE | ID: mdl-15071185

ABSTRACT

Systemic administration of adenovirus and adenovirus vectors induces a robust innate and adaptive immune response in a variety of animal models. In tumor necrosis factor (TNF)(-/-) mice, a diminished immune response to adenovirus (Ad) infection has been attributed to compromised dendritic cell (DC) maturation. In this report, we investigated the mechanisms responsible for Ad-mediated activation and maturation of DC. Ad infection induced high levels of TNF-alpha expression by murine bone marrow-derived DC, comparable to levels observed with lipopolysaccharide exposure. Ad-induced TNF-alpha production was necessary for DC maturation and acts in an autocrine manner. Unlike TNF-alpha production associated with exposure to lipopolysaccharide, Ad induction of TNF-alpha was not dependent on the MyD88 signaling pathway. In contrast, Ad-induced TNF-alpha production and DC maturation were dependent on signaling by phosphoinositide-3-OH kinase (PI3K), as determined by wortmannin and LY294002 blocking experiments. The adenovirus capsid protein penton contains a well characterized arginine-glycine-aspartic acid integrin-binding domain that stimulates PI3K in fibroblast cell lines. When this region of the penton was mutated, TNF-alpha expression and bone marrow-derived DC maturation were attenuated. We propose that integrin-mediated PI3K induction of NF-kappaB activates an autocrine TNF-alpha pathway required for DC maturation in response to Ad.


Subject(s)
Adenoviridae/physiology , Dendritic Cells/cytology , Phosphatidylinositol 3-Kinases/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Adaptor Proteins, Signal Transducing , Animals , Antigens, Differentiation/metabolism , Base Sequence , Cell Differentiation/drug effects , DNA Primers , Dendritic Cells/enzymology , Dendritic Cells/metabolism , Enzyme Inhibitors/pharmacology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88 , Oligopeptides/metabolism , Phosphoinositide-3 Kinase Inhibitors , Polymerase Chain Reaction , Receptors, Immunologic/metabolism
8.
Hum Gene Ther ; 15(1): 47-61, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14965377

ABSTRACT

We have used a plasmid-based transfection model of the adeno-associated virus (AAV) Rep-mediated site-specific integration (RMSSI) pathway to characterize the stability and expression of a site-specifically integrated transgene (either green fluorescent protein [GFP] or chloramphenicol acetyltransferase [CAT]). Three plasmids containing the AAV p5 integration efficiency element (p5IEE) have been used to study integration and transgene expression in HeLa cells: (1) pRepGFP(itr+) contains both AAV ITRs, rep, and p5IEE and can be used as either a plasmid or rAAV vehicle for integration; (2) pRepGFP(itr-) contains the AAV rep gene and the p5IEE; (3) pAd-p5CAT contains only the 138-bp p5IEE of AAV. The data presented demonstrate that in the absence of drug selection, all three constructs undergo site-specific integration (efficiencies of between 10 and 40% of transduced cell lines). At 6 weeks posttransfection most cell lines that underwent RMSSI also expressed the appropriate transgene product. By 18 weeks posttransfection cell lines that were established with rep in cis to the transgene showed a decline in transgene expression as well as a loss of transgene DNA. In many cell lines, there appears to be transgene-containing DNA that does not contribute to gene expression. Data support a model of gene expression and transgene instability through a Rep-mediated pathway. In contrast to rep-containing cell lines, clonal cell lines containing p5IEECAT (with Rep provided in trans) maintained both the integrated transgene and transgene expression throughout the entire experimental time course (18 weeks).


Subject(s)
Chromosomes, Human, Pair 19 , Genetic Therapy , Transgenes , Virus Integration , DNA-Binding Proteins/metabolism , Dependovirus/genetics , Genetic Vectors , HeLa Cells , Humans , Plasmids , Repetitive Sequences, Nucleic Acid , Viral Proteins/metabolism
9.
J Virol ; 77(1): 57-67, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12477810

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV/human herpesvirus 8 [HHV-8]) is a gamma-2-herpesvirus responsible for Kaposi's sarcoma as well as primary effusion lymphoma (PEL). KSHV is a lymphotropic virus that has pirated many mammalian genes involved in inflammation, cell cycle control, and angiogenesis. Among these is the early lytic viral G protein-coupled receptor (vGPCR), a homologue of the human interleukin-8 (IL-8) receptor. When expressed, vGPCR is constitutively active and can signal via mitogen- and stress-activated kinases. In certain models it activates the transcriptional potential of NF-kappaB and activator protein 1 (AP-1) and induces vascular endothelial growth factor (VEGF) production. Despite its importance to the pathogenesis of all KSHV-mediated disease, little is known about vGPCR activity in hematopoietic cells. To study the signaling potential and downstream effects of vGPCR in such cells, we have developed PEL cell lines that express vGPCR under the control of an inducible promoter. The sequences required for tetracycline-mediated induction were cloned into a plasmid containing adeno-associated virus type 2 elements to enhance integration efficiency. This novel plasmid permitted studies of vGPCR activity in naturally infected KSHV-positive lymphocytes. We show that vGPCR activates ERK-2 and p38 in PEL cells. In addition, it increases the transcription of reporter genes under the control of AP-1, NF-kappaB, CREB, and NFAT, a Ca(2+)-dependent transcription factor important to KSHV lytic gene expression. vGPCR also increases the transcription of KSHV open reading frames 50 and 57, thereby displaying broad potential to affect viral transcription patterns. Finally, vGPCR signaling results in increased PEL cell elaboration of KSHV vIL-6 and VEGF, two growth factors involved in KSHV-mediated disease pathogenesis.


Subject(s)
Herpesvirus 8, Human/pathogenicity , Lymphoma, Non-Hodgkin/virology , Nuclear Proteins , Receptors, Chemokine/physiology , Viral Proteins/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , DNA-Binding Proteins/metabolism , Endothelial Growth Factors/biosynthesis , Enzyme Activation , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Interleukin-6/biosynthesis , Lymphokines/biosynthesis , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , NFATC Transcription Factors , Promoter Regions, Genetic , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
10.
Proc Natl Acad Sci U S A ; 99(19): 12381-5, 2002 Sep 17.
Article in English | MEDLINE | ID: mdl-12221283

ABSTRACT

Adeno-associated virus (AAV) undergoes site-specific integration into human chromosome 19 through a deletion-substitution mechanism at the well characterized AAVS1 site. We have shown previously that a cis element within the left end of the AAV genome enhances the efficiency of Rep-mediated site-specific integration into chromosome 19 when present in inverted terminal repeat-containing recombinant AAV (rAAV) plasmids. We now demonstrate that a 138-bp cis element, the p5 integration efficiency element (p5IEE), mediates efficient integration. The p5IEE is not only required for efficient site-specific integration, it is also sufficient. Integration mediated by the p5IEE occurs in the absence of the AAV inverted terminal-repeat elements. The data presented in this study demonstrate that the p5IEE is a multifunctional element, serving as the highly regulatable Rep promoter and the primary substrate for targeted integration.


Subject(s)
Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 19/virology , DNA-Binding Proteins/metabolism , Dependovirus/genetics , Viral Proteins/metabolism , Virus Integration/genetics , Base Sequence , Binding Sites/genetics , DNA, Viral/genetics , DNA, Viral/metabolism , Dependovirus/physiology , HeLa Cells , Humans , Molecular Sequence Data , Plasmids/genetics , Promoter Regions, Genetic
11.
J Virol ; 76(11): 5411-21, 2002 Jun.
Article in English | MEDLINE | ID: mdl-11991970

ABSTRACT

The initial aim of this study was to combine attributes of adeno-associated virus (AAV) and adenovirus (Ad) gene therapy vectors to generate an Ad-AAV hybrid vector allowing efficient site-specific integration with Ad vectors. In executing our experimental strategy, we found that, in addition to the known incompatibility of Rep expression and Ad growth, an equally large obstacle was presented by the inefficiency of the integration event when using traditional recombinant AAV (rAAV) vectors. This study has addressed both of these problems. We have shown that a first-generation Ad can be generated that expresses Rep proteins at levels consistent with those found in wild-type AAV (wtAAV) infections and that Rep-mediated AAV persistence can occur in the presence of first-generation Ad vectors. Our finding that traditional rAAV plasmid vectors lack integration potency compared to wtAAV plasmid constructs (10- to 100-fold differences) was unexpected but led to the discovery of a previously unidentified AAV integration enhancer sequence element which functions in cis to an AAV inverted terminal repeat-flanked target gene. rAAV constructs containing left-end AAV sequence, including the p5-rep promoter sequence, integrate efficiently in a site-specific manner. The identification of this novel AAV integration enhancer element is consistent with previous studies, which have indicated that a high frequency of wtAAV recombinant junction formation occurs in the vicinity of the p5 promoter, and recent studies have demonstrated a role for this region in AAV DNA replication. Understanding the contribution of this element to the mechanism of AAV integration will be critical to the use of AAV vectors for targeted gene transfer applications.


Subject(s)
Adenoviruses, Human/genetics , DNA-Binding Proteins/genetics , Dependovirus/genetics , Genetic Vectors/genetics , Promoter Regions, Genetic , Viral Proteins/genetics , DNA, Viral , Gene Expression , Gene Transfer Techniques , Humans , Plasmids , Recombination, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...