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.
Hum Gene Ther Clin Dev ; 26(2): 103-12, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26086763

RESUMO

Preclinical studies to assess biodistribution, safety, and initial efficacy of ART-I02, an adeno-associated type 5 (rAAV5) vector expressing human interferon ß (hIFN-ß), were performed in a total of 24 rhesus monkeys with collagen-induced arthritis. All monkeys were naïve or showed limited neutralizing antibody (Nab) titers to AAV5 at the start of the study. Animals were injected with a single intra-articular dose of ART-I02 or placebo, consisting of 3.2×10(13) vg (Dose A=maximum feasible dose), 4.58×10(12) vg (Dose B), or placebo in the first affected finger joint, the ipsilateral knee, and ankle joint at the same time point. Animals were monitored for clinical parameters and well-being with a maximum of 4 weeks, with the option that the severity of arthritis could necessitate an earlier time point of sacrifice. No adverse events were noted after injection of ART-I02. No abnormalities were observed after histological evaluation of all organs. At both dose levels, immunohistochemical staining indicated expression of hIFN-ß. In animals injected with Dose A, we observed stabilization or a reduction in swelling in the finger joint in which vector was administered. The highest copy numbers of vector DNA were detected in synovial tissue of the injected joint and the draining lymph node of the injected knee. High titers of Nab to rAAV5 were observed at the end of the study. Five monkeys developed an rAAV5-specific T-cell response. Two monkeys developed Nab to hIFN-ß. In conclusion, intra-articular injection of ART-I02 was well-tolerated and did not induce adverse events. After administration of Dose A of ART-I02, we observed a beneficial effect on joint swelling, substantiated by decreased histological inflammation and bone erosion scores. A GMP vector for clinical application has been manufactured and is currently being tested in GLP rodent studies, with the aim to move forward to a clinical trial.


Assuntos
Artrite Experimental/terapia , Dependovirus/genética , Interferon beta/genética , Animais , Articulação do Tornozelo/patologia , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Feminino , Articulações dos Dedos/patologia , Terapia Genética , Vetores Genéticos , Humanos , Injeções Intra-Articulares , Interferon beta/metabolismo , Articulação do Joelho/patologia , Macaca mulatta , Masculino , Distribuição Tecidual , Resultado do Tratamento
2.
PLoS One ; 9(2): e86453, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24498275

RESUMO

Recombinant adeno-associated vectors (rAAV) are commonly purified by either chromatography or equilibrium CsCl gradient. Nevertheless, even after purification various cellular proteins often associate with rAAV vector capsids. Such co-purifying cellular proteins may raise concern about safety of gene therapy. Here we report identification and characterization of the co-purifying cellular protein in the vector preparations by using a combination of two proteomics approaches, GeLC-MS (gel electrophoresis liquid chromatography-mass spectrometry) and 2DE (two-dimensional gel electrophoresis). Most prominent bands revealed by Coomassie Blue staining were mostly similar to the AAV capsid proteins. Posttranslational modifications of capsid proteins were detected by the proteomics analysis. A total of 13 cellular proteins were identified in the rAAV vectors purified by two rounds of cesium chloride gradient centrifugation, including 9 by the GeLC-MS analysis and 4 by the 2DE analysis. Selected cellular proteins were verified by western blot. Furthermore, the cellular proteins could be consistently found associated with different AAV serotypes and carrying different transgenes. Yet, the proteins were not integral components of the viral capsis since a stringent washing procedure by column purification could remove them. These co-purified proteins in AAV vector preparations may have a role in various stages of the AAV life cycle.


Assuntos
Proteínas do Capsídeo/análise , Dependovirus/metabolismo , Proteoma/análise , Proteômica/métodos , Western Blotting , Proteínas do Capsídeo/isolamento & purificação , Cromatografia Líquida , Dependovirus/genética , Eletroforese em Gel Bidimensional , Terapia Genética/métodos , Terapia Genética/normas , Vetores Genéticos/genética , Humanos , Espectrometria de Massas , Proteoma/isolamento & purificação , Reprodutibilidade dos Testes , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
3.
N Engl J Med ; 368(16): 1509-1518, 2013 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-23527958

RESUMO

Chimeric antigen receptor-modified T cells with specificity for CD19 have shown promise in the treatment of chronic lymphocytic leukemia (CLL). It remains to be established whether chimeric antigen receptor T cells have clinical activity in acute lymphoblastic leukemia (ALL). Two children with relapsed and refractory pre-B-cell ALL received infusions of T cells transduced with anti-CD19 antibody and a T-cell signaling molecule (CTL019 chimeric antigen receptor T cells), at a dose of 1.4×10(6) to 1.2×10(7) CTL019 cells per kilogram of body weight. In both patients, CTL019 T cells expanded to a level that was more than 1000 times as high as the initial engraftment level, and the cells were identified in bone marrow. In addition, the chimeric antigen receptor T cells were observed in the cerebrospinal fluid (CSF), where they persisted at high levels for at least 6 months. Eight grade 3 or 4 adverse events were noted. The cytokine-release syndrome and B-cell aplasia developed in both patients. In one child, the cytokine-release syndrome was severe; cytokine blockade with etanercept and tocilizumab was effective in reversing the syndrome and did not prevent expansion of chimeric antigen receptor T cells or reduce antileukemic efficacy. Complete remission was observed in both patients and is ongoing in one patient at 11 months after treatment. The other patient had a relapse, with blast cells that no longer expressed CD19, approximately 2 months after treatment. Chimeric antigen receptor-modified T cells are capable of killing even aggressive, treatment-refractory acute leukemia cells in vivo. The emergence of tumor cells that no longer express the target indicates a need to target other molecules in addition to CD19 in some patients with ALL.


Assuntos
Antígenos CD19 , Imunoterapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores de Antígenos de Linfócitos T , Linfócitos T/imunologia , Criança , Quimera , Feminino , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Indução de Remissão
4.
Mol Ther ; 20(9): 1737-49, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22735378

RESUMO

The hepatitis C virus (HCV) chronically infects 2% of the world population and effective treatment is limited by long duration and significant side-effects. Here, we describe a novel drug, intended as a "single-shot " therapy, which expresses three short hairpin RNAs (shRNAs) that simultaneously target multiple conserved regions of the HCV genome as confirmed in vitro by knockdown of an HCV replicon system. Using a recombinant adeno-associated virus (AAV) serotype 8 vector for delivery, comprehensive transduction of hepatocytes was achieved in vivo in a nonhuman primate (NHP) model following a single intravenous injection. However, dose ranging studies performed in 13 NHP resulted in high-expression levels of shRNA from wild-type (wt) Pol III promoters and dose-dependent hepatocellular toxicity, the first demonstration of shRNA-related toxicity in primates, establishing that the hepatotoxicity arises from highly conserved features of the RNA interference (RNAi) pathway. In the second generation drug, each promoter was re-engineered to reduce shRNA transcription to levels that circumvent toxicity but still inhibit replicon activity. In vivo testing of this modified construct in 18 NHPs showed conservation of hepatocyte transduction but complete elimination of hepatotoxicity, even with sustained shRNA expression for 50 days. These data support progression to a clinical study for treatment of HCV infection.


Assuntos
Genoma Viral , Hepacivirus/genética , Hepatite C Crônica/terapia , Hepatócitos/virologia , Fígado/virologia , RNA Interferente Pequeno/genética , RNA Viral/antagonistas & inibidores , Animais , DNA Polimerase III/genética , Dependovirus/genética , Engenharia Genética , Terapia Genética , Vetores Genéticos , Hepatite C Crônica/virologia , Hepatócitos/patologia , Injeções Intravenosas , Fígado/patologia , Macaca fascicularis , Camundongos , Regiões Promotoras Genéticas , RNA Viral/genética , Replicon , Transdução Genética , Replicação Viral
5.
Lancet ; 374(9701): 1597-605, 2009 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-19854499

RESUMO

BACKGROUND: Gene therapy has the potential to reverse disease or prevent further deterioration of vision in patients with incurable inherited retinal degeneration. We therefore did a phase 1 trial to assess the effect of gene therapy on retinal and visual function in children and adults with Leber's congenital amaurosis. METHODS: We assessed the retinal and visual function in 12 patients (aged 8-44 years) with RPE65-associated Leber's congenital amaurosis given one subretinal injection of adeno-associated virus (AAV) containing a gene encoding a protein needed for the isomerohydrolase activity of the retinal pigment epithelium (AAV2-hRPE65v2) in the worst eye at low (1.5 x 10(10) vector genomes), medium (4.8 x 10(10) vector genomes), or high dose (1.5 x 10(11) vector genomes) for up to 2 years. FINDINGS: AAV2-hRPE65v2 was well tolerated and all patients showed sustained improvement in subjective and objective measurements of vision (ie, dark adaptometry, pupillometry, electroretinography, nystagmus, and ambulatory behaviour). Patients had at least a 2 log unit increase in pupillary light responses, and an 8-year-old child had nearly the same level of light sensitivity as that in age-matched normal-sighted individuals. The greatest improvement was noted in children, all of whom gained ambulatory vision. The study is registered with ClinicalTrials.gov, number NCT00516477. INTERPRETATION: The safety, extent, and stability of improvement in vision in all patients support the use of AAV-mediated gene therapy for treatment of inherited retinal diseases, with early intervention resulting in the best potential gain. FUNDING: Center for Cellular and Molecular Therapeutics at the Children's Hospital of Philadelphia, Foundation Fighting Blindness, Telethon, Research to Prevent Blindness, F M Kirby Foundation, Mackall Foundation Trust, Regione Campania Convenzione, European Union, Associazione Italiana Amaurosi Congenita di Leber, Fund for Scientific Research, Fund for Research in Ophthalmology, and National Center for Research Resources.


Assuntos
Proteínas de Transporte/genética , Proteínas do Olho/genética , Terapia Genética/métodos , Atrofia Óptica Hereditária de Leber/terapia , Adolescente , Adulto , Fatores Etários , Cegueira/congênito , Cegueira/genética , Criança , Adaptação à Escuridão , Dependovirus/genética , Progressão da Doença , Relação Dose-Resposta a Droga , Eletrorretinografia , Feminino , Vetores Genéticos/genética , Vetores Genéticos/uso terapêutico , Humanos , Injeções , Masculino , Mutação/genética , Nistagmo Fisiológico , Atrofia Óptica Hereditária de Leber/diagnóstico , Atrofia Óptica Hereditária de Leber/genética , Segurança , Resultado do Tratamento , Acuidade Visual , Adulto Jovem , cis-trans-Isomerases
6.
Mol Ther ; 17(6): 1022-30, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19293773

RESUMO

The assessment of the risk of germline transmission of vector-coded sequences is critical for clinical translation of gene transfer strategies. We used rabbit models to analyze the risk of germline transmission of adeno-associated viral (AAV) vectors. Intravenous injection of AAV-2 or AAV-8 resulted in liver-mediated, long-term expression of therapeutic levels of human factor IX (hFIX) in a dose-dependent manner. In high-dose cohorts, AAV-8 resulted in twofold higher levels of circulating hFIX and of vector DNA in liver compared to AAV-2. Vector sequences were found in the semen of all rabbits. The kinetics of vector clearance from semen was dose- and time-dependent but serotype-independent. No late recurrence of AAV-8 sequences was found in the semen over several consecutive cycles of spermatogenesis. In a novel rabbit model, AAV-2 or AAV-8 sequences were detected in the semen of vasectomized animals that lack germ cells. Therefore, structures of the genitourinary (GU) tract, as well as the testis, contribute significantly to vector shedding in the semen. Collectively, data from these two models suggest that the risk of inadvertent germline transmission in males by AAV-8 vectors is low, similar to that of AAV-2, and that AAV dissemination to the semen is in part modulated by host-dependent factors.


Assuntos
Adenoviridae/genética , Adenoviridae/metabolismo , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Células Germinativas/metabolismo , Animais , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Fator IX/genética , Vetores Genéticos/administração & dosagem , Humanos , Fígado/metabolismo , Masculino , Coelhos , Sêmen/metabolismo , Testículo/anatomia & histologia , Sistema Urogenital/anatomia & histologia , Vasectomia
7.
Mol Ther ; 17(1): 144-52, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18941440

RESUMO

In a gene therapy clinical trial for hemophilia B, adeno-associated virus 2 (AAV2) capsid-specific CD8(+) T cells were previously implicated in the elimination of vector-transduced hepatocytes, resulting in loss of human factor IX (hFIX) transgene expression. To test the hypothesis that expression of AAV2 cap DNA impurities in the AAV2-hFIX vector was the source of epitopes presented on transduced cells, transcription of cap was assessed by quantitative reverse transcription-PCR (Q-RT-PCR) following transduction of target cells with the vector used in the clinical trial. Transcriptional profiling was also performed for residual Amp(R), and adenovirus E2A and E4. Although trace amounts of DNA impurities were present in the clinical vector, transcription of these sequences was not detected after transduction of human hepatocytes, nor in mice administered a dose 26-fold above the highest dose administered in the clinical study. Two methods used to minimize encapsidated DNA impurities in the clinical vector were: (i) a vector (cis) production plasmid with a backbone exceeding the packaging limit of AAV; and (ii) a vector purification step that achieved separation of the vector from vector-related impurities (e.g., empty capsids). In conclusion, residual cap expression was undetectable following transduction with AAV2-hFIX clinical vectors. Preformed capsid protein is implicated as the source of epitopes recognized by CD8(+) T cells that eliminated vector-transduced cells in the clinical study.


Assuntos
Dependovirus/genética , Dependovirus/imunologia , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Transcrição Gênica/genética , Transcrição Gênica/imunologia , Animais , Capsídeo/imunologia , Linhagem Celular , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase
8.
N Engl J Med ; 358(21): 2240-8, 2008 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-18441370

RESUMO

Leber's congenital amaurosis (LCA) is a group of inherited blinding diseases with onset during childhood. One form of the disease, LCA2, is caused by mutations in the retinal pigment epithelium-specific 65-kDa protein gene (RPE65). We investigated the safety of subretinal delivery of a recombinant adeno-associated virus (AAV) carrying RPE65 complementary DNA (cDNA) (ClinicalTrials.gov number, NCT00516477 [ClinicalTrials.gov]). Three patients with LCA2 had an acceptable local and systemic adverse-event profile after delivery of AAV2.hRPE65v2. Each patient had a modest improvement in measures of retinal function on subjective tests of visual acuity. In one patient, an asymptomatic macular hole developed, and although the occurrence was considered to be an adverse event, the patient had some return of retinal function. Although the follow-up was very short and normal vision was not achieved, this study provides the basis for further gene therapy studies in patients with LCA.


Assuntos
Cegueira/terapia , Proteínas de Transporte/genética , Proteínas do Olho/genética , Terapia Genética , Vetores Genéticos , Degeneração Retiniana/terapia , Adulto , Cegueira/congênito , Cegueira/genética , Cegueira/patologia , DNA Complementar , Dependovirus/genética , Técnicas de Transferência de Genes , Humanos , Injeções , Mutação , Regiões Promotoras Genéticas , Reflexo Pupilar , Retina/patologia , Degeneração Retiniana/congênito , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Acuidade Visual , cis-trans-Isomerases
9.
Mol Ther ; 16(3): 458-65, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18209734

RESUMO

We evaluated the safety and efficacy of an optimized adeno-associated virus (AAV; AAV2.RPE65) in animal models of the RPE65 form of Leber congenital amaurosis (LCA). Protein expression was optimized by addition of a modified Kozak sequence at the translational start site of hRPE65. Modifications in AAV production and delivery included use of a long stuffer sequence to prevent reverse packaging from the AAV inverted-terminal repeats, and co-injection with a surfactant. The latter allows consistent and predictable delivery of a given dose of vector. We observed improved electroretinograms (ERGs) and visual acuity in Rpe65 mutant mice. This has not been reported previously using AAV2 vectors. Subretinal delivery of 8.25 x 10(10) vector genomes in affected dogs was well tolerated both locally and systemically, and treated animals showed improved visual behavior and pupillary responses, and reduced nystagmus within 2 weeks of injection. ERG responses confirmed the reversal of visual deficit. Immunohistochemistry confirmed transduction of retinal pigment epithelium cells and there was minimal toxicity to the retina as judged by histopathologic analysis. The data demonstrate that AAV2.RPE65 delivers the RPE65 transgene efficiently and quickly to the appropriate target cells in vivo in animal models. This vector holds great promise for treatment of LCA due to RPE65 mutations.


Assuntos
Cegueira/terapia , Dependovirus/genética , Animais , Cegueira/genética , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Cães , Eletrorretinografia , Proteínas do Olho/genética , Proteínas do Olho/fisiologia , Vetores Genéticos/genética , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Atrofia Óptica Hereditária de Leber/genética , Atrofia Óptica Hereditária de Leber/terapia , cis-trans-Isomerases
10.
Proc Natl Acad Sci U S A ; 104(32): 13104-9, 2007 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-17664425

RESUMO

Previous studies have documented that 0.1 approximately 1% of input recombinant adeno-associated virus (rAAV) vectors could be stabilized and lead to transgene expression. To characterize the steps involving massive AAV DNA loss, we designed an"AAV footprinting" strategy that can track newly formed AAV dsDNA genomes. This strategy is based on an ROSA26R mouse model or cell line that carries a lacZ gene flanked by two loxP sites. When it is transduced by a rAAV vector carrying the Cre recombinase, the lacZ gene can be activated and remain active even when rAAV genomes are later lost. By using this sensitive AAV footprinting technique, we confirmed the existence of transient AAV dsDNA that went undetected by conventional DNA methods. Although these dsDNA intermediates could be efficiently formed in almost every cell and were competent for mRNA transcription and protein synthesis in vivo, they got lost continuously. Only a small fraction was eventually stabilized for sustained gene expression. Although both rAAV2 and rAAV8 can potentially have similar levels of dsDNA formation, AAV8 dsDNA was formed much faster than that of AAV2, which explains why rAAV8 is more efficient than rAAV2 in transducing the liver. Collectively, our studies suggested that rather than receptor binding, viral entry, and ssDNA to dsDNA conversion, the instability of newly formed AAV dsDNA was the primary contributing factor for the low rAAV transduction efficacy. The uncoating step significantly influenced the stability of AAV transient dsDNA. The identification of transient AAV dsDNA provided a new pathway for improving rAAV transduction.


Assuntos
DNA Viral/genética , Dependovirus/genética , Genoma Viral , Recombinação Genética , Transdução Genética , Fosfatase Alcalina/genética , Terapia Genética , Vetores Genéticos , Instabilidade Genômica , Integrases/genética , Fígado/metabolismo , Plasmídeos
11.
J Virol Methods ; 140(1-2): 183-92, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17196264

RESUMO

Adeno-associated virus (AAV) empty capsids typically co-purify with genome containing AAV2 vectors purified by column chromatography. This study describes a method to remove empty capsids from genome containing vector particles by anion exchange chromatography. The separation is based on the slightly less anionic character of empty particles compared to vectors. Detailed methods to achieve AAV2 vector purification and particle separation using cation exchange resin POROS 50HS followed by anion exchange resin Q-Sepharose(xl) are described. Chromatographic separation of AAV2 particles was achieved using gradients based on sodium acetate and ammonium acetate, and was optimal at pH 8.5. Efficient removal of particle surface nucleic acid impurities was found to be important to achieve good particle separation. In a large scale experiment performed using partially purified vector containing a mixture of 1.56 x 10(14)vg and 2.52 x 10(15) empty capsids as a starting material, the optimized anion exchange chromatography method resulted in a vector peak of 1.15 x 10(14)vg containing 0.25 x 10(14) empty capsids, corresponding to 74% vector yield and 86-fold reduction in empty capsids in the vector product.


Assuntos
Cromatografia por Troca Iônica/métodos , Dependovirus/isolamento & purificação , Vetores Genéticos/isolamento & purificação , Vírion , Capsídeo/metabolismo , Capsídeo/ultraestrutura , Dependovirus/classificação , Dependovirus/genética , Vetores Genéticos/ultraestrutura , Concentração de Íons de Hidrogênio , Vírion/ultraestrutura
12.
Hum Gene Ther ; 17(1): 46-54, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16409124

RESUMO

Adeno-associated virus (AAV) serotype 1 (AAV1) has been shown to be more effective than the well-studied AAV serotype 2 (AAV2) in muscle gene transfer. Replacement of amino acids 350 to 430 of AAV2 VP1 with the corresponding amino acids from VP1 of AAV1 resulted in a hybrid vector, termed AAV-221-IV, which behaved similarly to AAV1 in vitro and in vivo in muscle. Intramuscular injection of 1x10(11) vector particles per mouse of hybrid vector carrying a human FIX transgene in CD4 knockout mice resulted in an average level of human FIX in the plasma of 450 ng/ml, 4- to 10-fold higher than in mice injected with an AAV2 vector carrying the same transgene, and 80% of the transgene levels in animals treated with the same dose of AAV1. DNA analysis of injected muscle showed a 10-fold higher copy number after gene delivery by the hybrid vector compared with AAV2. A comparison of total DNA versus DNA from intact virus particles suggests a higher stability of hybrid virus particles. These results suggest that changes in the AAV capsid have an effect on virus-cell receptor interaction, and also influence trafficking and processing of the virus particle in the cell. This "hybrid vector" retains the heparin-binding sites of AAV2 and, therefore, can be purified by passage through a heparin-Sepharose column with the same efficiency as AAV2. When tested in vivo, either in CD4 knockout mice or in a hemophilic mouse model, the heparin-purified hybrid vector showed >10-fold higher activity than similarly purified AAV2. This demonstrates the utility of this hybrid vector in the performance of large-scale heparin column purification to generate a vector with a high expression profile for muscle-directed gene delivery. Initiation of clinical studies with this hybrid vector may be facilitated because it differs from AAV2 by only nine amino acids.


Assuntos
Dependovirus/fisiologia , Terapia Genética , Vetores Genéticos/normas , Hemofilia B/terapia , Animais , Células COS , Proteínas do Capsídeo/genética , Células Cultivadas , Chlorocebus aethiops , Dependovirus/genética , Dependovirus/imunologia , Dependovirus/isolamento & purificação , Fator IX/análise , Fator IX/biossíntese , Expressão Gênica/genética , Células HeLa , Humanos , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Fenótipo , Homologia de Sequência de Aminoácidos , Transdução Genética
13.
J Virol ; 78(24): 13678-86, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15564477

RESUMO

Adeno-associated virus (AAV) is a unique gene transfer vector which takes approximately 4 to 6 weeks to reach its expression plateau. The mechanism for this slow-rise expression profile was proposed to be inefficient second-strand DNA synthesis from the input single-stranded (ss) DNA viral genome. In order to clarify the status of ss AAV genomes, we generated AAV vectors labeled with bromodeoxyuridine (BrdU), a nucleotide analog that can be incorporated into the AAV genome and packaged into infectious virions. Since BrdU-DNA can be detected only by an anti-BrdU antibody when DNA is in an ss form, not in a double-stranded (ds) form, ss AAV genomes with BrdU can be readily tracked in situ. Although ss AAV DNA was abundant by Southern blot analysis, free ss AAV genomes were not detectable after AAV transduction by this new detection method. Further Southern blot analysis of viral DNA and virions revealed that ss AAV DNA was protected within virions. Extracted cellular fractions demonstrated that viral particles in host cells remained infectious. In addition, a significant amount of AAV genomes was degraded after AAV transduction. Therefore, we conclude that the amount of free ss DNA is not abundant during AAV transduction. AAV transduction is limited by the steps that affect AAV ss DNA release (i.e., uncoating) before second-strand DNA synthesis can occur. AAV ss DNA released from viral uncoating is either converted into ds DNA efficiently or degraded by cellular DNA repair mechanisms as damaged DNA. This study elucidates a mechanism that can be exploited to develop new strategies to improve AAV vector transduction efficiency.


Assuntos
DNA de Cadeia Simples/metabolismo , Dependovirus/metabolismo , Genoma Viral , Recombinação Genética , Transdução Genética , Bromodesoxiuridina/metabolismo , DNA/metabolismo , DNA Viral/metabolismo , Dependovirus/genética , Dependovirus/patogenicidade , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos
14.
Mech Dev ; 120(10): 1113-26, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14568101

RESUMO

The Drosophila Pax-6 homologs eyeless (ey) and twin of eyeless (toy) are expressed in the eyes and in the central nervous system (CNS). In addition to the pivotal functions in eye development, previous studies revealed that ey also plays important roles in axonal development of the mushroom bodies, centers for associative learning and memory. It has been reported that a second intron enhancer that contains several Pax-6 binding sites mainly controls the eye-specific expression, but the DNA sequences that control CNS expression are unknown. In this work, we have dissected transcriptional enhancer elements of the ey gene that are required for the CNS expression in various developmental stages. We first show that CNS expression is independent of the eye-specific enhancer of the second intron. By systematic reporter studies, we have identified several discrete DNA elements in the 5' upstream region and in the second intron that cooperatively interact to generate most of the ey expression pattern in the CNS. DNA sequence comparison between the ey genes of distant Drosophila species has identified conserved modules that might be bound by the upstream regulatory factors of the ey gene in CNS development. Furthermore, by RNA interference and mutant studies, we show that ey expression in the brain is independent of the activity of toy and ey itself whereas in the eye primordia it requires both, supporting the notion that ey and toy are regulated by parallel and independent regulatory cascades in brain development.


Assuntos
Encéfalo/embriologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Animais , Encéfalo/metabolismo , Elementos de DNA Transponíveis , Proteínas de Ligação a DNA/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Elementos Facilitadores Genéticos , Proteínas do Olho , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Íntrons , Camundongos , Dados de Sequência Molecular , Especificidade de Órgãos , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados , Proteínas Repressoras , Transativadores/genética , Transativadores/metabolismo
15.
Mol Ther ; 7(3): 419-25, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12668138

RESUMO

Although most animal experiments with recombinant adeno-associated virus (AAV) vectors have been based on AAV serotype 2, recent studies showed that AAV vectors based on AAV serotype 1 performed more efficiently in muscle and other tissues. On the other hand, AAV2-based vectors can be readily purified by heparin column. To combine the advantages of both types of vectors, we developed a strategy to generate chimeric vectors by using a mixture of AAV helper plasmids encoding both serotypes in the transfection process. Because the AAV packaging machinery cannot distinguish between closely related AAV1 and AAV2 capsid proteins, each packaged virion contains capsid proteins from both serotypes. As expected, the resulting chimeric vectors could be purified by heparin column. Neutralizing antibody assays showed that the chimeric vectors can be inhibited by either AAV1 or AAV2 antiserum. In vivo, the chimeric vectors direct levels of expression similar to those of AAV1 in muscle or AAV2 in liver; that is, they combine the best transduction characteristics of both parent vectors. In summary, this study provides a straightforward method for combining various properties of different AAV serotypes into one vector. Potential limitations of the chimeric vectors are also discussed.


Assuntos
Dependovirus/genética , Terapia Genética , Vetores Genéticos , Proteínas Recombinantes de Fusão/genética , Animais , Primers do DNA/genética , DNA Recombinante/genética , DNA Viral/química , DNA Viral/genética , Ensaio de Imunoadsorção Enzimática , Fator IX/metabolismo , Técnicas de Transferência de Genes , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Sorotipagem , Transdução Genética , Transfecção , Transgenes , alfa 1-Antitripsina/metabolismo
16.
J Virol ; 77(4): 2768-74, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12552020

RESUMO

Muscle is an attractive target for gene delivery because of its mass and because vectors can be delivered in a noninvasive fashion. Adeno-associated virus (AAV) has been shown to be effective for muscle-targeted gene transfer. Recent progress in characterization of AAV serotype 1 (AAV1) and AAV6 demonstrated that these two AAV serotypes are far more efficient in transducing muscle than is the traditionally used AAV2. Since all cis elements are identical in these vectors, the potential determinants for their differences in transducing muscle appear to be located within the AAV capsid proteins. In the present study, a series of AAV capsid mutants were generated to identify the major regions affecting AAV transduction efficiency in muscle. Replacement of amino acids 350 to 736 of AAV2 VP1 with the corresponding amino acids from VP1 of AAV1 resulted in a hybrid vector that behaved very similarly to AAV1 in vitro and in vivo in muscle. Characterization of additional mutants carrying smaller regions of the AAV1 VP1 amino acid sequence in the AAV2 capsid protein suggested that amino acids 350 to 430 of VP1 function as a major tissue tropism determinant. Further analysis showed that the heparin binding domain and the major antigenic determinants in the AAV capsid region were not necessary for the efficiency of AAV1 transduction of muscle.


Assuntos
Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Dependovirus/patogenicidade , Músculos/virologia , Transdução Genética , Sequência de Aminoácidos , Proteínas do Capsídeo/química , Dependovirus/genética , Vetores Genéticos , Humanos , Dados de Sequência Molecular , Mutação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...