Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Hum Gene Ther ; 18(2): 171-82, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17328683

ABSTRACT

Self-complementary adeno-associated viral (scAAV) vectors bypass the requirement for viral second-strand DNA synthesis, but the packaging capacity of these vectors ( approximately 2.4 kb) is significantly smaller than that of conventional AAV vectors ( approximately 4.8 kb). We constructed human recombinant green fluorescent protein (hrGFP) expression cassettes ranging from 2.3 to 4.1 kb. Each vector was biologically active, but the transduction efficiency of vectors containing <3.3-kb genomes was significantly higher than those containing 3.5-kb genomes or larger. However, scAAV vectors containing up to approximately 3.3-kb genomes also contained single-stranded genomes, and 3.5-kb and larger genomes were packaged only as single-stranded DNA. These data suggest that the maximum packaging capacity of scAAV vectors is approximately 3.3 kb. The production of single-stranded genomes was not due to repair of the terminal resolution site (trs) in the inverted terminal repeats in the AAV genome, but rather was partly due to the use of AAV helper plasmid, known to lead to higher levels of expression of Rep proteins. The use of a helper plasmid known to lead to reduced levels of Rep proteins led to the generation of scAAV vectors that contained approximately 90% of the viral genomes in double-stranded forms. These studies demonstrate the feasibility of achieving encapsidation of larger genomes into scAAV vectors than was suggested originally, but underscore the need to exercise caution in using the appropriate helper plasmid to generate scAAV stocks capable of high-efficiency transduction that are relatively free of single-stranded DNA-containing vectors.


Subject(s)
DNA, Recombinant/genetics , DNA-Binding Proteins/metabolism , Dependovirus/genetics , Dependovirus/physiology , Genetic Vectors/isolation & purification , Viral Proteins/metabolism , Virus Assembly/physiology , DNA, Complementary/genetics , DNA, Viral/genetics , DNA, Viral/metabolism , Genetic Vectors/genetics , Genome, Viral , HeLa Cells , Humans , Sequence Deletion , Transduction, Genetic , Virus Replication
2.
Hum Gene Ther ; 17(9): 909-20, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16972759

ABSTRACT

Most viral vectors used for gene therapy lack the ability to target a defined cell population. Parvovirus B19 has a restricted tropism for human erythroid progenitor cells and uses activated alpha5beta1 integrins as coreceptors for entry [Weigel-Kelley, K.A., Yoder, M.C., and Srivastava, A. (2003). Blood 102, 3927-3933]. In this study we examined the role of coexpressed integrins in alpha5beta1 integrin coreceptor function. Antibody-mediated cross-linking of beta1, beta2, and beta3 integrins and the integrin-associated protein (IAP) increased parvovirus B19 entry into nontarget K562 cells. Functional silencing of one integrin group, however, reduced the virus uptake- promoting function of a subsequently activated integrin group, indicating that the three integrins did not operate in isolation but through shared signaling pathways. This was further corroborated by direct competition between simultaneously clustered beta2 and beta1 integrins that could be overcome by stabilizing clustered beta1 integrins in a high-affinity conformation. In contrast, parvovirus B19 entry into primary erythroid progenitor cells was characterized by strong clustering-induced beta1 integrin coreceptor activity that was not abolished by subsequent beta2 and beta3 integrin activation and was, in fact, substantially increased in the presence of preclustered beta2 and beta3 integrins. Thus, integrin function is regulated in a cell type-specific manner through coexpressed integrins and preferential parvovirus B19 entry into erythroid progenitor cells is promoted by a robust beta1 integrin response that is enhanced through stable preclustering of coexpressed integrins. These results have implications for other viral vectors that use integrins as receptors/coreceptors and for gene therapy of hematopoietic progenitor cells using parvovirus B19 vectors.


Subject(s)
Integrin alpha5beta1/physiology , Parvovirus B19, Human/physiology , Antibodies, Viral/immunology , Cell Differentiation , Humans , K562 Cells , Parvovirus B19, Human/immunology , Signal Transduction , Transduction, Genetic
3.
Virology ; 353(2): 283-93, 2006 Sep 30.
Article in English | MEDLINE | ID: mdl-16828834

ABSTRACT

We have reported that tyrosine-phosphorylated forms of a cellular protein, FKBP52, inhibit the second-strand DNA synthesis of adeno-associated virus 2 (AAV), leading to inefficient transgene expression from recombinant AAV vectors. To further explore the role of FKBP52 in AAV-mediated transduction, we established murine embryo fibroblasts (MEFs) cultures from FKBP52 wild-type (WT), heterozygous (HE), and knockout (KO) mice. Conventional AAV vectors failed to transduce WT MEFs efficiently, and the transduction efficiency was not significantly increased in HE or KO MEFs. AAV vectors failed to traffic efficiently to the nucleus in these cells. Treatment with hydroxyurea (HU) increased the transduction efficiency of conventional AAV vectors by approximately 25-fold in WT MEFs, but only by approximately 4-fold in KO MEFs. The use of self-complementary AAV (scAAV) vectors, which bypass the requirement of viral second-strand DNA synthesis, revealed that HU treatment increased the transduction efficiency approximately 23-fold in WT MEFs, but only approximately 4-fold in KO MEFs, indicating that the lack of HU treatment-mediated increase in KO MEFs was not due to failure of AAV to undergo viral second-strand DNA synthesis. Following HU treatment, approximately 59% of AAV genomes were present in the nuclear fraction from WT MEFs, but only approximately 28% in KO MEFs, indicating that the pathway by which HU treatment mediates nuclear transport of AAV was impaired in KO MEFs. When KO MEFs were stably transfected with an FKBP52 expression plasmid, HU treatment-mediated increase in the transduction efficiency was restored in these cells, which correlated directly with improved intracellular trafficking. Intact AAV particles were also shown to interact with FKBP52 as well as with dynein, a known cellular protein involved in AAV trafficking. These studies suggest that FKBP52, being a cellular chaperone protein, facilitates intracellular trafficking of AAV, which has implications in the optimal use of recombinant AAV vectors in human gene therapy.


Subject(s)
Dependovirus/metabolism , Genetic Vectors/metabolism , Molecular Chaperones/physiology , Animals , Biological Transport , Cell Nucleus/virology , Cells, Cultured , Dependovirus/genetics , Dyneins/metabolism , Fibroblasts/metabolism , Gene Expression , Genetic Therapy , Genetic Vectors/genetics , Heterozygote , Mice , Mice, Knockout , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Molecular Weight , Tacrolimus/metabolism , Transduction, Genetic
4.
Hum Gene Ther ; 17(3): 321-33, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16544981

ABSTRACT

Conflicting data exist on hematopoietic cell transduction by AAV serotype 2 (AAV2) vectors, and additional AAV serotype vectors have not been evaluated for their efficacy in hematopoietic stem/progenitor cell transduction. We evaluated the efficacy of conventional, single-stranded AAV serotype vectors 1 through 5 in primitive murine hematopoietic stem/progenitor cells in vitro as well as in vivo. In progenitor cell assays using Sca1+ c-kit+ Lin- hematopoietic cells, 9% of the colonies in cultures infected with AAV1 expressed the transgene. Coinfection of AAV1 with self-complementary AAV vectors carrying the gene for T cell protein tyrosine phosphatase (scAAV-TC-PTP) increased the transduction efficiency to 24%, indicating that viral secondstrand DNA synthesis is a rate-limiting step. This was further corroborated by the use of scAAV vectors, which bypass this requirement. In bone marrow transplantation studies involving lethally irradiated syngeneic mice, Sca1+ c-kit+ Lin- cells coinfected with AAV1 +/- scAAV-TC-PTP vectors led to transgene expression in 2 and 7.5% of peripheral blood (PB) cells, respectively, 6 months posttransplantation. In secondary transplantation experiments, 7% of PB cells and 3% of bone marrow (BM) cells expressed the transgene 6 months posttransplantation. Approximately 21% of BM-derived colonies harbored the proviral DNA sequences in integrated forms. These results document that AAV1 is thus far the most efficient vector in transducing primitive murine hematopoietic stem/progenitor cells. Further studies involving scAAV genomes and hematopoietic cell-specific promoters should further augment the transduction efficiency of AAV1 vectors, which should have implications in the optimal use of these vectors in hematopoietic stem cell gene therapy.


Subject(s)
Dependovirus/genetics , Genetic Vectors/administration & dosage , Hematopoietic Stem Cells/metabolism , Protein Tyrosine Phosphatases/genetics , Stem Cells/metabolism , Transduction, Genetic , Animals , Ataxin-1 , Ataxins , Cells, Cultured , DNA, Recombinant/administration & dosage , Dependovirus/classification , Dependovirus/immunology , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/genetics , Hematopoietic Stem Cells/virology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Nucleotidyltransferases/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2 , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins c-kit/genetics , Stem Cells/virology , Transgenes/physiology
5.
Mol Ther ; 10(5): 950-7, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15509512

ABSTRACT

Recombinant vectors based on adeno-associated virus type 2 (AAV) target the liver efficiently, but the transgene expression is limited to approximately 5% of hepatocytes. The lack of efficient transduction is due, in part, to the presence of a cellular protein, FKBP52, phosphorylated forms of which inhibit the viral second-strand DNA synthesis. We have documented that dephosphorylation of FKBP52 at tyrosine residues by the cellular T cell protein tyrosine phosphatase (TC-PTP) enhances AAV-mediated transduction in primary murine hematopoietic cells from TC-PTP-transgenic mice. We have also documented that AAV-mediated transduction is significantly enhanced in hepatocytes in TC-PTP-transgenic as well as in FKBP52-deficient mice because of efficient viral second-strand DNA synthesis. In this study, we evaluated whether co-infection of conventional single-stranded AAV vectors with self-complementary AAV-TC-PTP vectors leads to increased transduction efficiency of conventional AAV vectors in established human cell lines in vitro and in primary murine hepatocytes in vivo. We demonstrate here that scAAV-TC-PTP vectors serve as a helper virus in augmenting the transduction efficiency of conventional AAV vectors in vitro as well as in vivo which correlates directly with the extent of second-strand DNA synthesis of conventional single-stranded AAV vectors. Toxicological studies following tail-vein injections of scAAV-TC-PTP vectors in experimental mice show no evidence of any adverse effect in any of the organs in any of the mice for up to 13 weeks. Thus, this novel co-infection strategy should be useful in circumventing one of the major obstacles in the optimal use of recombinant AAV vectors in human gene therapy.


Subject(s)
Dependovirus/genetics , Genetic Vectors/genetics , Helper Viruses/genetics , Protein Tyrosine Phosphatases/genetics , Transduction, Genetic/methods , Animals , Cell Line, Tumor , DNA, Viral/analysis , Genetic Therapy/methods , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Hepatocytes/chemistry , Hepatocytes/pathology , Humans , Mice , Mice, Inbred C57BL , Protein Tyrosine Phosphatase, Non-Receptor Type 2 , Protein Tyrosine Phosphatases/analysis , Protein Tyrosine Phosphatases/metabolism
6.
Hum Gene Ther ; 15(12): 1207-18, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15684697

ABSTRACT

Controversies abound concerning hematopoietic stem cell transduction by recombinant adeno-associated virus 2 (AAV) vectors. For human hematopoietic cells, we have shown that this problem is related to the extent of expression of the cellular receptor for AAV. At least a small subset of murine hematopoietic cells, on the other hand, does express both the AAV receptor and the coreceptor, yet is transduced poorly. In the present study, we have found that approximately 85% of AAV genomes were present in the cytoplasmic fraction of primary murine c-Kit(+)Lin- hematopoietic cells. However, when mice were injected intraperitoneally with hydroxyurea before isolation of these cells, the extent to which AAV genomes were detected in the cytoplasmic fraction was reduced to approximately 40%, with a corresponding increase to approximately 60% in the nuclear fraction, indicating that hydroxyurea facilitated nuclear transport of AAV. It was apparent, nonetheless, that a significant fraction of the AAV genomes present in the nuclear fraction from cells obtained from hydroxyurea-treated mice was single stranded. We next tested whether the single-stranded AAV genomes were derived from virions that failed to undergo uncoating in the nucleus. A substantial fraction of the signal in the nuclear fraction of hematopoietic cells obtained from hydroxyurea-treated mice was also resistant to DNase I. That AAV particles were intact and biologically active was determined by successful transduction of 293 cells by virions recovered from murine hematopoietic cells 48 hr postinfection. Although hydroxyurea facilitated nuclear transport of AAV, most of the virions failed to undergo uncoating, thereby leading to only a partial improvement in viral second- strand DNA synthesis and transgene expression. A better understanding of the underlying mechanism of viral uncoating has implications in the optimal use of recombinant AAV vectors in hematopoietic stem cell gene therapy.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Dependovirus/genetics , Genetic Vectors/administration & dosage , Hematopoietic Stem Cells/metabolism , Transduction, Genetic/methods , Animals , Cell Line , Cells, Cultured , DNA, Viral/analysis , Female , Gene Expression Regulation , Genetic Therapy/methods , Hematopoietic Stem Cells/virology , Hydroxyurea/pharmacology , Lac Operon/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Genetic , Recombinant Proteins/metabolism , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Virion/physiology
7.
Blood ; 102(12): 3927-33, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-12907437

ABSTRACT

Replication of the pathogenic human parvovirus B19 is restricted to erythroid progenitor cells. Although blood group P antigen has been reported to be the cell surface receptor for parvovirus B19, a number of nonerythroid cells, which express P antigen, are not permissive for parvovirus B19 infection. We have documented that P antigen is necessary for parvovirus B19 binding but not sufficient for virus entry into cells. To test whether parvovirus B19 utilizes a cell surface coreceptor for entry, we used human erythroleukemia cells (K562), which allow parvovirus B19 binding but not entry. We report here that upon treatment with phorbol esters, K562 cells become adherent and permissive for parvovirus B19 entry, which is mediated by alpha 5 beta 1 integrins, but only in their high-affinity conformation. Mature human red blood cells (RBCs), which express high levels of P antigen, but not alpha 5 beta 1 integrins, bind parvovirus B19 but do not allow viral entry. In contrast, primary human erythroid progenitor cells express high levels of both P antigen and alpha 5 beta 1 integrins and allow beta1 integrin-mediated entry of parvovirus B19. Thus, in a natural course of infection, RBCs are likely exploited for a highly efficient systemic dissemination of parvovirus B19.


Subject(s)
Integrin alpha5beta1/physiology , Parvovirus B19, Human/physiology , Receptors, Virus/physiology , Cell Adhesion/drug effects , Erythrocytes/virology , Erythroid Precursor Cells/virology , Humans , Integrin alpha5beta1/chemistry , Integrin beta1/metabolism , Integrin beta1/physiology , K562 Cells , P Blood-Group System/physiology , Phorbol Esters/pharmacology , Protein Conformation , Receptors, Virus/chemistry
8.
J Virol ; 77(4): 2741-6, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12552015

ABSTRACT

The use of adeno-associated virus type 2 (AAV) vectors has gained attention as a potentially useful alternative to the more commonly used retrovirus and adenovirus vectors for human gene therapy. However, the transduction efficiency of AAV vectors varies greatly in different cells and tissues in vitro and in vivo. We have documented that a cellular protein that binds the immunosuppressant drug FK506, termed the FK506-binding protein (FKBP52), interacts with the single-stranded D sequence within the AAV inverted terminal repeats, inhibits viral second-strand DNA synthesis, and consequently limits high-efficiency transgene expression (K. Qing, J. Hansen, K. A. Weigel-Kelley, M. Tan, S. Zhou, and A. Srivastava, J. Virol., 75: 8968-8976, 2001). FKBP52 can be phosphorylated at both tyrosine and serine/threonine residues, but only the phosphorylated forms of FKBP52 interact with the D sequence. Furthermore, the tyrosine-phosphorylated FKBP52 inhibits AAV second-strand DNA synthesis by greater than 90%, and the serine/threonine-phosphorylated FKBP52 causes approximately 40% inhibition, whereas the dephosphorylated FKBP52 has no effect on AAV second-strand DNA synthesis. In the present study, we have identified that the tyrosine-phosphorylated form of FKBP52 is a substrate for the cellular T-cell protein tyrosine phosphatase (TC-PTP). Deliberate overexpression of the murine wild-type (wt) TC-PTP gene, but not that of a cysteine-to-serine (C-S) mutant, caused tyrosine dephosphorylation of FKBP52, leading to efficient viral second-strand DNA synthesis and resulting in a significant increase in AAV-mediated transduction efficiency in HeLa cells in vitro. Both wt and C-S mutant TC-PTP expression cassettes were also used to generate transgenic mice. Primitive hematopoietic stem/progenitor cells from wt TC-PTP-transgenic mice, but not from C-S mutant TC-PTP-transgenic mice, could be successfully transduced by recombinant AAV vectors. These studies corroborate the fact that tyrosine phosphorylation of the cellular FKBP52 protein strongly influences AAV transduction efficiency, which may have important implications in the optimal use of AAV vectors in human gene therapy.


Subject(s)
Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors , Mice, Transgenic , Protein Tyrosine Phosphatases/metabolism , Transgenes , Animals , Genetic Therapy/methods , HeLa Cells , Humans , Mice , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 2 , Protein Tyrosine Phosphatases/genetics , Tacrolimus Binding Proteins/metabolism , Transduction, Genetic , Tyrosine/metabolism
9.
Pathol Biol (Paris) ; 50(5): 295-306, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12116848

ABSTRACT

In an attempt to exploit the remarkable tissue-tropism of the human parvovirus B19 to target human hematopoietic cells of the erythroid lineage, recombinant human adeno-associated virus 2 genomes were encapsidated in parvovirus B19 capsids. Although efficient transduction of primary human hematopoietic cells in the erythroid lineage occurred, a low-level of transgene expression in non-erythroid cells was also detected. These studies suggest that cell surface expression of P antigen, the primary receptor for parvovirus B19, is necessary but not sufficient for parvovirus B19 vector-mediated transduction of human hematopoietic cells. These studies also suggest the existence of a putative cell surface co-receptor, which is required for successful infection of human hematopoietic cells by parvovirus B19.


Subject(s)
DNA, Recombinant , Genetic Vectors , Parvovirus B19, Human/genetics , DNA, Viral/genetics , Dependovirus/genetics , Erythroid Precursor Cells , Gene Expression , Humans , Receptors, Virus/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...