Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Virus Res ; 274: 197771, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31577935

RESUMO

We compared the phenotypes of three mutant AAV2 viruses containing mutations in arginine amino acids (R585, R588 and R484) previously shown to be involved in AAV2 heparan sulfate binding. The transduction efficiencies of wild type and mutant viruses were determined in the eye, the brain and peripheral organs following subretinal, striatal and intravenous injection, respectively, in mice and rats. We found that each of the three mutants (the single mutant R585A; the double mutant R585, 588A; and the triple mutant R585, 588, 484A) had a unique phenotype compared to wt and each other. R585A was completely defective for transducing peripheral organs via intravenous injection, suggesting that R585A may be useful for targeting peripheral organs by substitution of peptide ligands in the capsid surface. In the brain, all three mutants displayed widespread transduction, with the double mutant R585, 588A displaying the greatest spread and the greatest number of transduced neurons. The double mutant was also extremely efficient for retrograde transport, while the triple mutant was almost completely defective for retrograde transport. This suggested that R484 may be directly involved in interaction with the transport machinery. Finally, the double mutant also displayed improved transduction of the eye compared to wild type and the other mutants.


Assuntos
Proteínas do Capsídeo/genética , Capsídeo/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Parvovirinae/fisiologia , Animais , Transporte Axonal/genética , Proteínas do Capsídeo/metabolismo , Dependovirus , Feminino , Masculino , Camundongos , Mutação , Parvovirinae/genética , Parvovirinae/metabolismo , Fenótipo , Ligação Proteica , Ratos , Tropismo Viral/genética
2.
Differentiation ; 78(1): 35-44, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19427096

RESUMO

The Hematopoietic- and neurologic-expressed sequence 1 (Hn1) gene encodes a small protein that is highly conserved among species. Hn1 expression is upregulated in regenerating neural tissues, including the axotomized adult rodent facial motor nerve and dedifferentiating retinal pigment epithelial cells of the Japanese newt. It is also expressed in numerous tissues during embryonic development as well as in regions of the adult brain that exhibit high plasticity. Hn1 has also been reported as a marker for human ovarian carcinoma and it is expressed in high-grade human gliomas. This study was directed toward understanding the function of Hn1 in a murine melanoma cell line. Hn1 mRNA and protein were identified in B16.F10 cells and in tumors formed from these cells. Inhibition of Hn1 protein expression with siRNA increased melanogenesis. Hn1-depleted cells expressed higher levels of the melanogenic proteins tyrosinase and Trp2 and an increased interaction between actin and Rab27a. The in vitro cell growth rate of Hn1-depleted cells was significantly reduced due to G1/S cell cycle arrest. This was consistent with a reduction in the phosphorylation of retinoblastoma protein as well as lower levels of p27 and increased expression of p21. Decreased expression of c-Met, the receptor for hepatocyte growth factor, was also detected in the Hn1-depleted cells, however HGF-dependent stimulation of phosphorylated-ERK was unaffected. Hn1 depletion also led to increased basal levels of phosphorylated p38 MAPK, while basal ERK phosphorylation was reduced. Moreover, Hn1-depleted cells had reduced expression of transcription factors MITF and USF-1, and increased expression of TFE3. These data, coupled with reports on Hn1 expression in regeneration and development, suggest that Hn1 functions as a suppressor of differentiation in cells undergoing repair or proliferation.


Assuntos
Ciclo Celular/genética , Diferenciação Celular/genética , Melaninas/genética , Melanoma Experimental/genética , Proteínas do Tecido Nervoso/genética , Adenoviridae/genética , Animais , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Escherichia coli/genética , Técnica Direta de Fluorescência para Anticorpo , Corantes Fluorescentes/metabolismo , Vetores Genéticos , Imuno-Histoquímica , Hibridização In Situ , Indóis/metabolismo , Melaninas/fisiologia , Melanoma Experimental/fisiopatologia , Camundongos , Proteínas Associadas aos Microtúbulos , Proteínas do Tecido Nervoso/fisiologia , Fenótipo , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes de Fusão/fisiologia , Transdução Genética
3.
Pathol Oncol Res ; 15(3): 437-44, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19145478

RESUMO

The hematopoietic- and neurologic-expressed sequence 1 (Hn1) gene encodes a highly conserved protein that is expressed in developing and regenerating tissues. In this study, Hn1 expression was evaluated in human and murine malignant gliomas. Hn1 mRNA and protein were detected in the murine GL261 glioma cell line and in GL261 brain tumors in vivo. HN1 is also expressed in human U118MG and U87MG cell lines. Evaluation of human brain tumors using an anti-Hn1 polyclonal antibody detected strong immunoreactivity in high-grade (WHO III and IV) malignant gliomas. The rate of GL261 cell proliferation in vitro was unaltered by Hn1 depletion using an anti-Hn1 siRNA. However, tumors established from Hn1-depleted GL261 cells formed significantly smaller volumes than those established from control-treated cells. These data suggest a role for Hn1 in the biology of malignant brain tumors.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Animais , Northern Blotting , Western Blotting , Neoplasias Encefálicas/patologia , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Glioma/patologia , Humanos , Hibridização In Situ , Camundongos , Proteínas Associadas aos Microtúbulos , RNA Mensageiro/análise , RNA Interferente Pequeno , Análise Serial de Tecidos
4.
Proc Natl Acad Sci U S A ; 105(22): 7827-32, 2008 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-18511559

RESUMO

Recombinant adeno-associated virus 2 (AAV2) vectors are in use in several Phase I/II clinical trials, but relatively large vector doses are needed to achieve therapeutic benefits. Large vector doses also trigger an immune response as a significant fraction of the vectors fails to traffic efficiently to the nucleus and is targeted for degradation by the host cell proteasome machinery. We have reported that epidermal growth factor receptor protein tyrosine kinase (EGFR-PTK) signaling negatively affects transduction by AAV2 vectors by impairing nuclear transport of the vectors. We have also observed that EGFR-PTK can phosphorylate AAV2 capsids at tyrosine residues. Tyrosine-phosphorylated AAV2 vectors enter cells efficiently but fail to transduce effectively, in part because of ubiquitination of AAV capsids followed by proteasome-mediated degradation. We reasoned that mutations of the surface-exposed tyrosine residues might allow the vectors to evade phosphorylation and subsequent ubiquitination and, thus, prevent proteasome-mediated degradation. Here, we document that site-directed mutagenesis of surface-exposed tyrosine residues leads to production of vectors that transduce HeLa cells approximately 10-fold more efficiently in vitro and murine hepatocytes nearly 30-fold more efficiently in vivo at a log lower vector dose. Therapeutic levels of human Factor IX (F.IX) are also produced at an approximately 10-fold reduced vector dose. The increased transduction efficiency of tyrosine-mutant vectors is due to lack of capsid ubiquitination and improved intracellular trafficking to the nucleus. These studies have led to the development of AAV vectors that are capable of high-efficiency transduction at lower doses, which has important implications in their use in human gene therapy.


Assuntos
Dependovirus/genética , Vetores Genéticos , Mutação Puntual , Transdução Genética , Tirosina/genética , Animais , Capsídeo/metabolismo , Núcleo Celular/metabolismo , Terapia Genética , Células HeLa , Hepatócitos/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Ubiquitinação
5.
Hum Gene Ther ; 18(2): 171-82, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17328683

RESUMO

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.


Assuntos
DNA Recombinante/genética , Proteínas de Ligação a DNA/metabolismo , Dependovirus/genética , Dependovirus/fisiologia , Vetores Genéticos/isolamento & purificação , Proteínas Virais/metabolismo , Montagem de Vírus/fisiologia , DNA Complementar/genética , DNA Viral/genética , DNA Viral/metabolismo , Vetores Genéticos/genética , Genoma Viral , Células HeLa , Humanos , Deleção de Sequência , Transdução Genética , Replicação Viral
6.
Virology ; 353(2): 283-93, 2006 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-16828834

RESUMO

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.


Assuntos
Dependovirus/metabolismo , Vetores Genéticos/metabolismo , Chaperonas Moleculares/fisiologia , Animais , Transporte Biológico , Núcleo Celular/virologia , Células Cultivadas , Dependovirus/genética , Dineínas/metabolismo , Fibroblastos/metabolismo , Expressão Gênica , Terapia Genética , Vetores Genéticos/genética , Heterozigoto , Camundongos , Camundongos Knockout , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Peso Molecular , Tacrolimo/metabolismo , Transdução Genética
7.
Hum Genet ; 119(6): 571-603, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16612615

RESUMO

During the past decade, in vivo administration of viral gene transfer vectors for treatment of numerous human diseases has been brought from bench to bedside in the form of clinical trials, mostly aimed at establishing the safety of the protocol. In preclinical studies in animal models of human disease, adeno-associated viral (AAV) vectors have emerged as a favored gene transfer system for this approach. These vectors are derived from a replication-deficient, non-pathogenic parvovirus with a single-stranded DNA genome. Efficient gene transfer to numerous target cells and tissues has been described. AAV is particularly efficient in transduction of non-dividing cells, and the vector genome persists predominantly in episomal forms. Substantial correction, and in some instances complete cure, of genetic disease has been obtained in animal models of hemophilia, lysosomal storage disorders, retinal diseases, disorders of the central nervous system, and other diseases. Therapeutic expression often lasted for months to years. Treatments of genetic disorders, cancer, and other acquired diseases are summarized in this review. Vector development, results in animals, early clinical experience, as well as potential hurdles and challenges are discussed.


Assuntos
Dependovirus , Técnicas de Transferência de Genes , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/terapia , Vetores Genéticos , Terapia Genética , Humanos
8.
Hum Gene Ther ; 17(3): 321-33, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16544981

RESUMO

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.


Assuntos
Dependovirus/genética , Vetores Genéticos/administração & dosagem , Células-Tronco Hematopoéticas/metabolismo , Proteínas Tirosina Fosfatases/genética , Células-Tronco/metabolismo , Transdução Genética , Animais , Ataxina-1 , Ataxinas , Células Cultivadas , DNA Recombinante/administração & dosagem , Dependovirus/classificação , Dependovirus/imunologia , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/virologia , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Nucleotidiltransferases/genética , Proteína Tirosina Fosfatase não Receptora Tipo 2 , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Células-Tronco/virologia , Transgenes/fisiologia
9.
Anticancer Res ; 26(1A): 311-7, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16475712

RESUMO

BACKGROUND: Recombinant adeno-associated viral vectors serotype 2 (rAAV2) can transduce several tissues with high efficiency. MATERIALS AND METHODS: The transduction efficiency of rAAV2 in pancreatic and colon cancer was compared to that of recombinant adenovirus (rAd) in vitro and in vivo using green fluorescent protein (GFP). RESULTS: With the exception of SU.86.86, the percentage of GFP-positive cells was below 10% at a multiplicity of infection (MOI) of 100 for rAAV2. At the same MOI, almost 100% of cells expressed GFP when rAd was used. However, the transduction efficiency for rAA V2 was comparable to that of rAd when coinfected with wt adenovirus, leading to a dramatic increase in the amount of double-stranded rAAV2 DNA. Similar results were obtained in vivo. While widespread GFP expression was readily detected in all xenografts injected with rAd, only one section of all tumors injected with rAAV2 contained GFP-positive cells. CONCLUSION: The results of this study indicate that rAAV2 might be useful for an ex vivo approach in cancer gene therapy, but it does not seem to be feasible for the in vivo treatment of malignant tumors.


Assuntos
Adenoviridae/genética , Neoplasias do Colo/genética , Neoplasias do Colo/virologia , Dependovirus/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/virologia , Transdução Genética/métodos , Animais , Neoplasias do Colo/terapia , Citomegalovirus/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Células HT29 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Neoplasias Pancreáticas/terapia , Fator 1 de Elongação de Peptídeos/genética , Regiões Promotoras Genéticas , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Mol Ther ; 12(6): 1217-25, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16213797

RESUMO

Scalable production of rAAV vectors remains a major obstacle to the clinical application of this prototypical gene therapy vector. A recently developed baculovirus-based production protocol (M. Urabe et al., 2002, Hum. Gene Ther. 13, 1935-1943) found limited applications due to the system's design. Here we report a detailed analysis of the stability of the original baculovirus system components BacRep, BacVP, and transgene cassette-containing BacGFP. All of the baculovirus helpers analyzed were prone to passage-dependent loss-of-function deletions resulting in considerable decreases in rAAV titers. To alleviate the instability and to extend the baculovirus platform to other rAAV serotypes, we have modified both Rep- and Cap-encoding components of the original system. The modifications include a parvoviral phospholipase A2 domain swap allowing production of infectious rAAV8 vectors in vivo. Alternatively, an infectious rAAV8 (or rAAV5) vector incorporating the AAV2 VP1 capsid protein in a mosaic vector particle with AAV8 capsid proteins was produced using a novel baculovirus vector. In this vector, the level of AAV2 VP1 expression is controlled with a "riboswitch," a self-cleaving ribozyme controlled by toyocamycin in the "ON" mode. The redesigned baculovirus system improves our capacity for rAAV manufacturing by making this production platform more applicable to other existing serotypes.


Assuntos
Baculoviridae/genética , Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Animais , Western Blotting , Capsídeo/química , Terapia Genética/métodos , Proteínas de Fluorescência Verde/metabolismo , Insetos , Camundongos , Modelos Genéticos , Fosfolipases A/química , Fosfolipases A2 , Plasmídeos/metabolismo , Estrutura Terciária de Proteína , Transdução Genética , Transgenes
11.
Mol Ther ; 11(6): 856-65, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15922956

RESUMO

Over the past decade, AAV-based vectors have emerged as promising candidates for gene therapeutic applications. Despite the broad tropism of the first eight serotypes identified, certain cell types are refractory to transduction with AAV-based vectors. Furthermore, for certain applications the targeting of specific cell types is desirable. To improve on present methods to alter AAV2 tropism, we take advantage of AAV2 mosaics. Here, we show that AAV2 mosaics have improved infectivity compared with all-mutant virions. Using an AAV2 mutant that contains the immunoglobulin-binding Z34C fragment of protein A, we demonstrate the utility of AAV2 mosaics to alter AAV2 tropism. This system allows us to transduce selectively and efficiently MO7e and Jurkat cells. The use of AAV2 mosaics with a protein A fragment inserted into their capsid, together with targeting antibodies, is a versatile method that allows the specific transduction of a wide array of cell types.


Assuntos
Proteínas do Capsídeo/genética , Dependovirus/genética , Transdução Genética/métodos , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Dependovirus/fisiologia , Vetores Genéticos/genética , Células HeLa , Humanos , Células Jurkat , Mutação , Proteína Estafilocócica A/metabolismo
12.
Hum Gene Ther ; 16(4): 408-16, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15871672

RESUMO

Recombinant gene delivery vehicles based on adeno-associated virus (rAAV) have emerged as promising vectors for the correction of genetic and acquired human disease states. These vectors possess many characteristics, including low pathogenicity and immunogenicity, and long-term gene expression after a single administered dose, that make them leading candidates for clinical gene therapy applications. Yet, the broad tissue tropism of the available AAV serotypes remains a disadvantage for the safest, most effective in vivo delivery of transgenes to target tissues. In addition, clinically relevant cell types exist that are poorly transduced by current rAAV vectors. As a result, increased efforts are now being made to tailor the tropism of rAAV to improve their transduction and selectivity profiles. Flexible, diverse methodologies have emerged that allow more control over the cell surface receptors rAAV employs for cell entry. These novel rAAV production strategies have resulted in unique vectors characterized by unique capsid protein sequences that employ alternative receptors, and have provided a better understanding of many basic aspects of the rAAV life cycle. This review aims to summarize the genetic methods currently being employed to customize rAAV capsids.


Assuntos
Capsídeo , Dependovirus/genética , Vetores Genéticos/genética , Tropismo , Animais , Terapia Genética/métodos , Vetores Genéticos/farmacologia , Humanos , Mosaicismo , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução Genética , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/metabolismo , Cultura de Vírus
13.
J Virol ; 78(12): 6595-609, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15163751

RESUMO

Direct insertion of amino acid sequences into the adeno-associated virus type 2 (AAV) capsid open reading frame (cap ORF) is one strategy currently being developed for retargeting this prototypical gene therapy vector. While this approach has successfully resulted in the formation of AAV particles that have expanded or retargeted viral tropism, the inserted sequences have been relatively short, linear receptor binding ligands. Since many receptor-ligand interactions involve nonlinear, conformation-dependent binding domains, we investigated the insertion of full-length peptides into the AAV cap ORF. To minimize disruption of critical VP3 structural domains, we confined the insertions to residue 138 within the VP1-VP2 overlap, which has been shown to be on the surface of the particle following insertion of smaller epitopes. The insertion of coding sequences for the 8-kDa chemokine binding domain of rat fractalkine (CX3CL1), the 18-kDa human hormone leptin, and the 30-kDa green fluorescent protein (GFP) after residue 138 failed to lead to formation of particles due to the loss of VP3 expression. To test the ability to complement these insertions with the missing capsid proteins in trans, we designed a system for producing AAV vectors in which expression of one capsid protein is isolated and combined with the remaining two capsid proteins expressed separately. Such an approach allows for genetic modification of a specific capsid protein across its entire coding sequence leaving the remaining capsid proteins unaffected. An examination of particle formation from the individual components of the system revealed that genome-containing particles formed as long as the VP3 capsid protein was present and demonstrated that the VP2 capsid protein is nonessential for viral infectivity. Viable particles composed of all three capsid proteins were obtained from the capsid complementation groups regardless of which capsid proteins were supplied separately in trans. Significant overexpression of VP2 resulted in the formation of particles with altered capsid protein stoichiometry. The key finding was that by using this system we successfully obtained nearly wild-type levels of recombinant AAV-like particles with large ligands inserted after residue 138 in VP1 and VP2 or in VP2 exclusively. While insertions at residue 138 in VP1 significantly decreased infectivity, insertions at residue 138 that were exclusively in VP2 had a minimal effect on viral assembly or infectivity. Finally, insertion of GFP into VP1 and VP2 resulted in a particle whose trafficking could be temporally monitored by using confocal microscopy. Thus, we have demonstrated a method that can be used to insert large (up to 30-kDa) peptide ligands into the AAV particle. This system allows greater flexibility than current approaches in genetically manipulating the composition of the AAV particle and, in particular, may allow vector retargeting to alternative receptors requiring interaction with full-length conformation-dependent peptide ligands.


Assuntos
Proteínas do Capsídeo/metabolismo , Dependovirus/genética , Dependovirus/patogenicidade , Mutação , Peptídeos/genética , Sequência de Bases , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Dependovirus/metabolismo , Células HeLa , Humanos , Microscopia Confocal , Dados de Sequência Molecular , Plasmídeos , Vírion/metabolismo , Montagem de Vírus
14.
J Virol ; 77(12): 6995-7006, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12768018

RESUMO

The adeno-associated virus type 2 (AAV2) uses heparan sulfate proteoglycan (HSPG) as its primary cellular receptor. In order to identify amino acids within the capsid of AAV2 that contribute to HSPG association, we used biochemical information about heparin and heparin sulfate, AAV serotype protein sequence alignments, and data from previous capsid studies to select residues for mutagenesis. Charged-to-alanine substitution mutagenesis was performed on individual residues and combinations of basic residues for the production and purification of recombinant viruses that contained a green fluorescent protein (GFP) reporter gene cassette. Intact capsids were assayed for their ability to bind to heparin-agarose in vitro, and virions that packaged DNA were assayed for their ability to transduce normally permissive cell lines. We found that mutation of arginine residues at position 585 or 588 eliminated binding to heparin-agarose. Mutation of residues R484, R487, and K532 showed partial binding to heparin-agarose. We observed a general correlation between heparin-agarose binding and infectivity as measured by GFP transduction; however, a subset of mutants that partially bound heparin-agarose (R484A and K532A) were completely noninfectious, suggesting that they had additional blocks to infectivity that were unrelated to heparin binding. Conservative mutation of positions R585 and R588 to lysine slightly reduced heparin-agarose binding and had comparable effects on infectivity. Substitution of AAV2 residues 585 through 590 into a location predicted to be structurally equivalent in AAV5 generated a hybrid virus that bound to heparin-agarose efficiently and was able to package DNA but was noninfectious. Taken together, our results suggest that residues R585 and R588 are primarily responsible for heparin sulfate binding and that mutation of these residues has little effect on other aspects of the viral life cycle. Interactive computer graphics examination of the AAV2 VP3 atomic coordinates revealed that residues which contribute to heparin binding formed a cluster of five basic amino acids that presented toward the icosahedral threefold axis from the surrounding spike protrusion. Three other kinds of mutants were identified. Mutants R459A, H509A, and H526A/K527A bound heparin at levels comparable to that of wild-type virus but were defective for transduction. Another mutant, H358A, was defective for capsid assembly. Finally, an R459A mutant produced significantly lower levels of full capsids, suggesting a packaging defect.


Assuntos
Sequência de Aminoácidos , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Dependovirus/patogenicidade , Proteoglicanas de Heparan Sulfato/metabolismo , Sequência de Bases , Proteínas do Capsídeo/genética , Dependovirus/genética , Regulação Viral da Expressão Gênica , Células HeLa , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Receptores Virais/metabolismo , Transdução Genética
15.
J Virol ; 76(22): 11505-17, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12388712

RESUMO

We examined cytoplasmic trafficking and nuclear translocation of adeno-associated virus type 2 (AAV) by using Alexa Fluor 488-conjugated wild-type AAV, A20 monoclonal antibody immunocytochemistry, and subcellular fractionation techniques followed by DNA hybridization. Our results indicated that in the absence of adenovirus (Ad), AAV enters the cell rapidly and escapes from early endosomes with a t(1/2) of about 10 min postinfection. Cytoplasmically distributed AAV accumulated around the nucleus and persisted perinuclearly for 16 to 24 h. Viral uncoating occurred before or during nuclear entry beginning about 12 h postinfection, when viral protein and DNA were readily detected in the nucleus. Few, if any, intact AAV capsids were found in the nucleus. In the presence of Ad, however, cytoplasmic AAV quickly translocated into the nucleus as intact particles as early as 40 min after coinfection, and this facilitated nuclear translocation of AAV was not blocked by the nuclear pore complex inhibitor thapsigargan. The rapid nuclear translocation of intact AAV capsids in the presence of Ad suggested that one or more Ad capsid proteins might be altering trafficking. Indeed, coinfection with empty Ad capsids also resulted in the appearance of AAV DNA in nuclei within 40 min. Escape from early endosomes did not seem to be affected by Ad coinfection.


Assuntos
Adenovírus Humanos/patogenicidade , Núcleo Celular/virologia , Dependovirus/patogenicidade , Vírus Auxiliares/patogenicidade , Adenovírus Humanos/fisiologia , Citoplasma/virologia , DNA Viral/análise , Dependovirus/fisiologia , Corantes Fluorescentes/metabolismo , Células HeLa , Vírus Auxiliares/fisiologia , Humanos , Poro Nuclear/efeitos dos fármacos , Tapsigargina/farmacologia
16.
Anticancer Res ; 22(6A): 3325-30, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12530082

RESUMO

BACKGROUND: Silencing of the viral CMV immediate early enhancer promoter can be a problem in certain cell types when engineering stable cell lines. MATERIALS AND METHODS: We compared the efficacy of the CMV promoter to the promoter of the elongation factor-1 alpha (EF-1 alpha) for the generation of stable colon carcinoma cell lines (HT-29). Green fluorescent protein (GFP) expression cassettes were delivered by recombinant adeno-associated virus (AAV) which is known for its ability to stably transduce cells. Stable cell lines were characterized in vitro by FACS and in vivo after HT-29 clones were grown as xenografts in nude mice. RESULTS: Stable HT-29 clones with > 97% of all cells homogeneously expressing GFP were generated with the EF-1 alpha promoter. In contrast in clones carrying the CMV promoter, only up to 60% of the cells were GFP-positive with expression levels varying widely between cells. Superinfection with wild-type adenovirus induced GFP expression in more than 90% of the cells indicating that the CMV promoter was silenced. In vivo the tumors carrying the EF-1 alpha promoter were homogeneously GFP-positive, whereas the CMV promoter gave rise to a scattered pattern of GFP expression. CONCLUSION: This study underlines the importance of the promoter for the generation of stable cell lines. In addition it demonstrates that recombinant AAV can effectively be used as a gene delivery system for this purpose.


Assuntos
Citomegalovirus/genética , Dependovirus/genética , Células HT29/fisiologia , Fator 1 de Elongação de Peptídeos/genética , Regiões Promotoras Genéticas/genética , Transfecção/métodos , Animais , Genes Reporter , Proteínas de Fluorescência Verde , Células HT29/patologia , Humanos , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Camundongos , Camundongos Nus , Transplante de Neoplasias , Transgenes , Transplante Heterólogo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA