Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Virol ; 82(6): 2727-40, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18199646

RESUMEN

Adeno-associated virus (AAV) vectors are associated with relatively mild host immune responses in vivo. Although AAV induces very weak innate immune responses, neutralizing antibodies against the vector capsid and transgene still occur. To understand further the basis of the antiviral immune response to AAV vectors, studies were performed to characterize AAV interactions with macrophages. Primary mouse macrophages and human THP-1 cells transduced in vitro using an AAV serotype 2 (AAV2) vector encoding green fluorescent protein did not result in measurable transgene expression. An assessment of internalized vector genomes showed that AAV2 vector uptake was enhanced in the presence of normal but not heat-inactivated or C3-depleted mouse/human serum. Enhanced uptake in the presence of serum coincided with increased macrophage activation as determined by the expression of NF-kappaB-dependent genes such as macrophage inflammatory protein 2 (MIP-2), interleukin-1beta (IL-1beta), IL-8, and MIP-1beta. AAV vector serotypes 1 and 8 also activated human and mouse macrophages in a serum-dependent manner. Immunoprecipitation studies demonstrated the binding of iC3b complement protein to the AAV2 capsid in human serum. AAV2 did not activate the alternative pathway of the complement cascade and lacked cofactor activity for factor I-mediated degradation of C3b to iC3b. Instead, our results suggest that the AAV capsid also binds complement regulatory protein factor H. In vivo, complement receptor 1/2- and C3-deficient mice displayed impaired humoral immunity against AAV2 vectors, with a delay in antibody development and significantly lower neutralizing antibody titers. These results show that the complement system is an essential component of the host immune response to AAV.


Asunto(s)
Proteínas del Sistema Complemento/fisiología , Dependovirus/inmunología , Vectores Genéticos/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Línea Celular , Dependovirus/genética , Expresión Génica , Humanos , Inmunoprecipitación , Activación de Macrófagos , Macrófagos/inmunología , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL
2.
Mol Ther ; 16(8): 1467-73, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18560418

RESUMEN

We have developed a highly specific and robust new method for labeling adeno-associated virus (AAV) vector particles with either biophysical probes or targeting ligands. Our approach uses the Escherichia coli enzyme biotin ligase (BirA), which ligates biotin to a 15-amino-acid biotin acceptor peptide (BAP) in a sequence-specific manner. In this study we demonstrate that by using a ketone isotere of biotin as a cofactor we can ligate this probe to BAP-modified AAV capsids. Because ketones are absent from AAV, BAP-modified AAV particles can be tagged with the ketone probe and then specifically conjugated to hydrazide- or hydroxylamine-functionalized molecules. We demonstrate this two-stage modification methodology in the context of a mammalian cell lysate for the labeling of AAV vector particles with various fluorophores, and for the attachment of a synthetic cyclic arginine-glycine-aspartate (RGD) peptide (c(RGDfC)) to target integrin receptors that are present on neovasculature. Fluorophore labeling allowed the straightforward determination of intracellular particle distribution. Ligand conjugation mediated a significant increase in the transduction of endothelial cells in vitro, and permitted the intravascular targeting of AAV vectors to tumor-associated vasculature in vivo. These results suggest that this approach holds significant promise for future studies aimed at understanding and modifying AAV vector-cellular interactions.


Asunto(s)
Ligasas de Carbono-Nitrógeno/química , Dependovirus/química , Proteínas de Escherichia coli/química , Proteínas Represoras/química , Animales , Biotina/química , Biotina/metabolismo , Ligasas de Carbono-Nitrógeno/genética , Ligasas de Carbono-Nitrógeno/metabolismo , Línea Celular , Dependovirus/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Femenino , Colorantes Fluorescentes/química , Técnicas de Transferencia de Gen , Vectores Genéticos/química , Vectores Genéticos/genética , Células HeLa , Humanos , Ligandos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Desnudos , Técnicas de Sonda Molecular , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Péptidos Cíclicos/química , Péptidos Cíclicos/genética , Péptidos Cíclicos/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
3.
Acta Pharmacol Sin ; 29(12): 1440-50, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19026163

RESUMEN

AIM: The aim of our study was to develop an effective gene delivery system for ovarian cancer gene therapy. METHODS: The expression of heparin sulfate proteoglycan (HSPG) and integrins alpha(upsilon)beta(3) and alpha(upsilon)beta(5) were analyzed with flow cytometry on 2 human ovarian cancer cell lines (OVCAR-3 and SKOV-3ip). The gene transduction efficiencies were evaluated with recombinant adeno-associated viral vector (rAAV)2-green fluorescent protein or rAAV2-lactase Z followed by flow cytometry or cytohistochemistry staining. The effect of 17beta-estradiol on ovarian cancer cell proliferation, HSPG, the expressions of integrins alpha(upsilon)beta(3) and alpha(upsilon)beta(5), and adeno-associated viral vector (AAV)2-mediated gene transduction were determined. RESULTS: In the present study, we found: (1) a variation in HSPG and the expressions of integrins alpha(upsilon)beta(3) and alpha(upsilon)beta(5) between OVCAR-3 and SKOV-3ip; (2) that 17beta-estradiol was shown to significantly stimulate cell proliferation and integrin beta(5) expression in certain ovarian cancer cell lines; and (3) integrintargeted A520/N584RGD-rAAV2, which has alternative interactivity with integrins and abrogates the binding capacity HSPG, showed much higher gene transduction efficiency in ovarian cancer cells than rAAV2 in the presence/absence of 17beta-estradiol. Moreover, this RGD-modified rAAV2 exerted more efficient transduction in ovarian cancer cells in response to 17beta-estradiol. CONCLUSION: Our findings implied that A520/N584RGD-rAAV2 may offer great potential for ovarian cancer treatment in vivo.


Asunto(s)
Dependovirus , Estradiol/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Animales , Antivirales/farmacología , Muerte Celular/fisiología , Línea Celular Tumoral/efectos de los fármacos , Dependovirus/genética , Dependovirus/metabolismo , Femenino , Ganciclovir/farmacología , Terapia Genética , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Heparina/análogos & derivados , Heparina/metabolismo , Humanos , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/metabolismo , Neoplasias Ováricas/terapia , Proteoglicanos/metabolismo , Receptores de Vitronectina/genética , Receptores de Vitronectina/metabolismo , Transducción Genética
4.
Hum Gene Ther ; 17(3): 353-61, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16544984

RESUMEN

Recombinant adeno-associated virus (AAV) vectors are promising in the context of gene therapy because of their ability to mediate efficient gene transfer and stable gene expression. AAV2 uses heparin sulfate as its primary receptor, which is widely expressed on the various tissues and organs. This limits the application of AAV2 in targeting specific tissues. To make an AAV2 vector with modified tropism, we constructed various AAV2 capsid mutants by inserting RGD-4C peptide at position 520 and/or at position 584. Eight mutants were generated, identified, and characterized. Heparin-binding ability was completely abrogated in five mutants, and partially reduced in three mutants. Solid-phase ELISA and gene transduction assays confirmed that the novel tropism is determined by the introduced RGD epitope, which binds to cellular integrin receptor. Our observations suggest that simultaneous modification at both sites, tentatively involved in heparin binding, results in altered tropism and improved transduction efficiency in vitro.


Asunto(s)
Proteínas de la Cápside/genética , Dependovirus/genética , Vectores Genéticos , Heparina/metabolismo , Mutagénesis Insercional , Animales , Sitios de Unión , Proteínas de la Cápside/metabolismo , Células Cultivadas/metabolismo , Células Cultivadas/virología , Ensayo de Inmunoadsorción Enzimática , Técnicas de Transferencia de Gen , Terapia Genética , Células HeLa/metabolismo , Células HeLa/virología , Humanos , Riñón/metabolismo , Riñón/virología , Ratones , Mutación/genética , Oligopéptidos/genética , Oligopéptidos/metabolismo , Transducción Genética , Tropismo
5.
Hum Gene Ther ; 17(7): 741-50, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16839273

RESUMEN

Gene-eluting stents are being evaluated in animals as an alternative approach to inhibiting in-stent restenosis. Adeno-associated virus type 2 (AAV2) and adenovirus are commonly used for gene transfer applications. We tested the hypothesis that these vectors can achieve prolonged and localized gene delivery to the vessel wall, using stents as delivery platforms. AdbetaGal (5 x 10(9) plaque-forming units) and AAV2betaGal (5.3 x 10(9) DNase-resistant particles) were used to coat BiodivYsio stents with matrix HI coating (Abbott Vascular Devices, Galway, Ireland). After balloon injury, external iliac arteries of New Zealand White rabbits were stented. The reverse transcription-polymerase chain reaction was used to assess viral spread. Expression of LacZ was demonstrated with both vectors at five time points (3, 7, 14, 21, and 28 days). In the adenovirus group the median percentage of cells expressing the transgene on day 3 was 2.73%, which increased to a median expression of 7.31% at 28 days (p > 0.05). Expression was localized to medial cells on day 3, but was observed predominantly in neointimal cells on day 28. In the AAV group, day 3 expression was 5.78%, which decreased to 2.12% on day 28 (p = 0.05). No systemic dissemination of virus was seen in any group. Adenovirus- and AAV2-coated stents can be used to deliver genes to the blood vessel wall for up to 28 days.


Asunto(s)
Adenoviridae/genética , Vasos Sanguíneos/metabolismo , Dependovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Stents , Animales , Vasos Sanguíneos/citología , Vasos Sanguíneos/enzimología , Expresión Génica , Vectores Genéticos/genética , Conejos , Transgenes , beta-Galactosidasa/análisis , beta-Galactosidasa/genética
6.
Am J Vet Res ; 67(7): 1145-55, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16817735

RESUMEN

OBJECTIVE: To evaluate host cell permissiveness and cytotoxic effects of recombinant and modified adenoviral vectors in equine chondrocytes, synovial cells, and bone marrow-derived mesenchymal stem cells (BMD-MSCs). SAMPLE POPULATION: Articular cartilage, synovium, and bone marrow from 15 adult horses. PROCEDURES: Equine chondrocytes, synovial cells, and BMD-MSCs and human carcinoma (HeLa) cells were cultured and infected with an E-1-deficient adenovirus vector encoding the beta-galactosidase gene or the green fluorescent protein gene (Ad-GFP) and with a modified E-1-deficient vector with the arg-gly-asp capsid peptide insertion and containing the GFP gene (Ad-RGD-GFP). Percentages of transduced cells, total and transduced cell counts, and cell viability were assessed 2 and 7 days after infection. RESULTS: -Permissiveness to adenoviral vector infection was significantly different among cell types and was ranked in decreasing order as follows: HeLa cells > BMD-MSCs > chondrocytes > synovial cells. Morphologic signs of cytotoxicity were evident in HeLa cells but not in equine cells. Numbers of transduced cells decreased by day 7 in all cell types except equine BMD-MSCs. Transduction efficiency was not significantly different between the Ad-GFP and Ad-RGD-GFP vectors. CONCLUSION AND CLINICAL RELEVANCE: Sufficient gene transfer may be achieved by use of an adenovirus vector in equine cells. High vector doses can be used in equine cells because of relative resistance to cytotoxic effects in those cells. Greater permissiveness and sustained expression of transgenes in BMD-MSCs make them a preferential cell target for gene therapy in horses.


Asunto(s)
Adenoviridae/fisiología , Adenoviridae/patogenicidad , Condrocitos/citología , Terapia Genética/veterinaria , Caballos , Células Madre/citología , Membrana Sinovial/citología , Adenoviridae/genética , Animales , Células de la Médula Ósea , Condrocitos/virología , Expresión Génica , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Células HeLa , Caballos/virología , Humanos , Células Madre/virología , Membrana Sinovial/virología
7.
AIDS Patient Care STDS ; 30(12): 545-550, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27905841

RESUMEN

HIV-1 infection requires the presence of the CD4 receptor on the target cell surface and a coreceptor, predominantly CC-chemokine receptor 5 (CCR5). It has been shown that individuals who are homozygous for a defective CCR5 gene are protected from HIV-1 infection. A novel self-inactivating lentiviral vector LVsh5/C46 (Cal-1) has been engineered to block HIV-1 infection with two viral entry inhibitors, conferring resistance to HIV-1 infection from both CCR5 and CXCR4 tropic strains. Cal-1 encodes a short hairpin RNA (sh5) to downregulate CCR5 and C46, an HIV-1 fusion inhibitor. Gene therapy by Cal-1 is aimed at transducing CD4+ T cells and CD34+ hematopoietic stem/progenitor cells in an autologous transplant setting. Pre-clinical safety and efficacy studies in vitro and in vivo (humanized mouse model and nonhuman primates) have shown that Cal-1 is safe with no indication of any toxicity risk and acts to decrease viral load and increase CD4 counts. Two clinical trials are underway using Cal-1: a phase I/II study to assess safety and feasibility in an adult HIV-1-positive population not on antiretroviral therapy (ART); and a second Fred Hutchinson Investigator Initiated phase I study to assess safety and feasibility in adults with HIV-1-associated non-Hodgkin or Hodgkin lymphoma.


Asunto(s)
Terapia Genética/métodos , Infecciones por VIH/terapia , VIH-1/fisiología , Fusión de Membrana , Receptores CCR5 , Receptores del VIH/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/biosíntesis , Adulto , Animales , Terapia Biológica/métodos , Antagonistas de los Receptores CCR5 , Recuento de Linfocito CD4 , Ensayos Clínicos como Asunto , Regulación hacia Abajo , Infecciones por VIH/genética , Infecciones por VIH/virología , VIH-1/genética , Células Madre Hematopoyéticas , Humanos , Receptores CCR5/biosíntesis , Receptores CXCR4 , Proteínas Recombinantes de Fusión/genética , Carga Viral
8.
Mol Ther Methods Clin Dev ; 3: 16007, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26958575

RESUMEN

We have focused on gene therapy approaches to induce functional cure/remission of HIV-1 infection. Here, we evaluated the safety and efficacy of the clinical grade anti-HIV lentiviral vector, Cal-1, in pigtailed macaques (Macaca nemestrina). Cal-1 animals exhibit robust levels of gene marking in myeloid and lymphoid lineages without measurable adverse events, suggesting that Cal-1 transduction and autologous transplantation of hematopoietic stem cells are safe, and lead to long-term, multilineage engraftment following myeloablative conditioning. Ex vivo, CD4+ cells from transplanted animals undergo positive selection in the presence of simian/human immunodeficiency virus (SHIV). In vivo, Cal-1 gene-marked cells are evident in the peripheral blood and in HIV-relevant tissue sites such as the gastrointestinal tract. Positive selection for gene-marked cells is observed in blood and tissues following SHIV challenge, leading to maintenance of peripheral blood CD4+ T-cell counts in a normal range. Analysis of Cal-1 lentivirus integration sites confirms polyclonal engraftment of gene-marked cells. Following infection, a polyclonal, SHIV-resistant clonal repertoire is established. These findings offer strong preclinical evidence for safety and efficacy of Cal-1, present a new method for tracking protected cells over the course of virus-mediated selective pressure in vivo, and reveal previously unobserved dynamics of virus-dependent T-cell selection.

9.
Neurosurgery ; 76(2): 216-25; discussion 225, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25549186

RESUMEN

BACKGROUND: After injection into muscle and peripheral nerves, a variety of viral vectors undergo retrograde transport to lower motor neurons. However, because of its attractive safety profile and durable gene expression, adeno-associated virus (AAV) remains the only vector to have been applied to the human nervous system for the treatment of neurodegenerative disease. Nonetheless, only a very small fraction of intramuscularly injected AAV vector arrives at the spinal cord. OBJECTIVE: To engineer a novel AAV vector by inserting a neuronal targeting peptide (Tet1), with binding properties similar to those of tetanus toxin, into the AAV1 capsid. METHODS: Integral to this approach was the use of structure-based design to increase the effectiveness of functional capsid engineering. This approach allowed the optimization of scaffolding regions for effective display of the foreign epitope while minimizing disruption of the native capsid structure. We also validated an approach by which low-titer tropism-modified AAV vectors can be rescued by particle mosaicism with unmodified capsid proteins. RESULTS: Importantly, our rationally engineered AAV1-based vectors exhibited markedly enhanced transduction of cultured motor neurons, diminished transduction of nontarget cells, and markedly superior retrograde delivery compared with unmodified AAV1 vector. CONCLUSION: This approach promises a significant advancement in the rational engineering of AAV vectors for diseases of the nervous system and other organs.


Asunto(s)
Proteínas de Unión al ADN/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/síntesis química , Proteínas Proto-Oncogénicas/genética , Transducción Genética/métodos , Animales , Proteínas de la Cápside/química , Dependovirus/genética , Terapia Genética/métodos , Células HEK293 , Células HeLa , Humanos , Oxigenasas de Función Mixta , Estructura Cuaternaria de Proteína , Ratas , Ratas Sprague-Dawley
10.
Mol Ther Nucleic Acids ; 4: e236, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25872029

RESUMEN

We described earlier a dual-combination anti-HIV type 1 (HIV-1) lentiviral vector (LVsh5/C46) that downregulates CCR5 expression of transduced cells via RNAi and inhibits HIV-1 fusion via cell surface expression of cell membrane-anchored C46 antiviral peptide. This combinatorial approach has two points of inhibition for R5-tropic HIV-1 and is also active against X4-tropic HIV-1. Here, we utilize the humanized bone marrow, liver, thymus (BLT) mouse model to characterize the in vivo efficacy of LVsh5/C46 (Cal-1) vector to engineer cellular resistance to HIV-1 pathogenesis. Human CD34+ hematopoietic stem/progenitor cells (HSPC) either nonmodified or transduced with LVsh5/C46 vector were transplanted to generate control and treatment groups, respectively. Control and experimental groups displayed similar engraftment and multilineage hematopoietic differentiation that included robust CD4+ T-cell development. Splenocytes isolated from the treatment group were resistant to both R5- and X4-tropic HIV-1 during ex vivo challenge experiments. Treatment group animals challenged with R5-tropic HIV-1 displayed significant protection of CD4+ T-cells and reduced viral load within peripheral blood and lymphoid tissues up to 14 weeks postinfection. Gene-marking and transgene expression were confirmed stable at 26 weeks post-transplantation. These data strongly support the use of LVsh5/C46 lentiviral vector in gene and cell therapeutic applications for inhibition of HIV-1 infection.

11.
Viruses ; 6(1): 54-68, 2013 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-24381033

RESUMEN

Human immunodeficiency virus type 1 (HIV-1) infection of target cells requires CD4 and a co-receptor, predominantly the chemokine receptor CCR5. CCR5-delta32 homozygosity results in a truncated protein providing natural protection against HIV infection-this without detrimental effects to the host-and transplantation of CCR5-delta32 stem cells in a patient with HIV ("Berlin patient") achieved viral eradication. As a more feasible approach gene-modification strategies are being developed to engineer cellular resistance to HIV using autologous cells. We have developed a dual therapeutic anti-HIV lentiviral vector (LVsh5/C46) that down-regulates CCR5 and inhibits HIV-1 fusion via cell surface expression of the gp41-derived peptide, C46. This construct, effective against multiple strains of both R5- and X4-tropic HIV-1, is being tested in Phase I/II trials by engineering HIV-resistant hematopoietic cells.


Asunto(s)
Antagonistas de los Receptores CCR5 , Terapia Genética/métodos , Infecciones por VIH/terapia , VIH-1/fisiología , Receptores del VIH/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/biosíntesis , Terapia Biológica/métodos , Ensayos Clínicos como Asunto , Infecciones por VIH/virología , VIH-1/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/fisiología , Células Madre Hematopoyéticas/virología , Humanos , Receptores CCR5/biosíntesis , Receptores del VIH/biosíntesis , Proteínas Recombinantes de Fusión/genética
12.
Arthritis ; 2012: 735472, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22288012

RESUMEN

Intra-articular gene therapy has potential for the treatment of osteoarthritis and rheumatoid arthritis. To quantify in vitro relative gene transduction, equine chondrocytes and synovial cells were treated with adenovirus vectors (Ad), serotype 2 adeno-associated virus vectors (rAAV2), or self-complementary (sc) AAV2 vectors carrying green fluorescent protein (GFP). Using 6 horses, bilateral metacarpophalangeal joints were injected with Ad, rAAV2, or scAAV2 vectors carrying GFP genes to assess the in vivo joint inflammation and neutralizing antibody (NAb) titer in serum and joint fluid. In vitro, the greater transduction efficiency and sustained gene expression were achieved by scAAV2 compared to rAAV2 in equine chondrocytes and synovial cells. In vivo, AAV2 demonstrated less joint inflammation than Ad, but similar NAb titer. The scAAV2 vectors can induce superior gene transduction than rAAV2 in articular cells, and both rAAV2 and scAAV2 vectors were showed to be safer for intra-articular use than Ad vectors.

13.
Methods Mol Biol ; 807: 273-300, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22034035

RESUMEN

Adeno-associated virus (AAV) has become a versatile vector platform. In recent years, powerful -techniques for the generation of tropism-modified vectors (rAAV-targeting vectors) and for investigation of virus-cell interaction were developed. The following chapter describes strategies for insertion of peptide ligands into the viral capsid and the subsequent characterization of capsid mutants, for producing mosaic capsids and for labeling the viral capsid chemically or genetically.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/genética , Cápside/química , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Reacción en Cadena de la Polimerasa , Virión/química
14.
J Orthop Res ; 28(2): 149-55, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19658157

RESUMEN

This study quantified and compared the transduction efficiencies of adenoviral (Ad), Arg-Gly-Asp (RGD)-modified Ad, adeno-associated viral serotype 2 (AAV2), and self-complementary AAV2 (scAAV2) vectors within full-thickness osteoarthritic (OA) and unaffected canine cartilage explants in vitro. Intraarticular administration of Ad and scAAV2 vectors was performed to determine the ability of these vectors to transduce unaffected guinea pig cartilage in vivo. Following explant exposure to vector treatment or control, the onset and surface distribution of reporter gene expression was monitored daily with fluorescent microscopy. At termination, explants were divided: one half was digested for analysis using flow cytometry; the remaining portion was used for histology and immunohistochemistry (IHC). Intact articular joints were collected for real-time RT-PCR and IHC to detect reporter gene expression following injection of selected vectors. Ad vector transduced focal areas along the perimeters of explants; the remaining vectors transduced chondrocytes across 100% of the surface. Greater mean transduction efficiencies were found with both AAV2 vectors as compared to the Ad vector (p < or = 0.026). Ad and Ad-RGD vectors transduced only superficial chondrocytes of OA and unaffected cartilage. Uniform reporter gene expression from AAV2 and scAAV2 was detected in the tangential and transitional zones of OA cartilage, but not deeper zones. AAV2 and scAAV2 vectors achieved partial and full-thickness transduction of unaffected cartilage. In vivo work revealed that scAAV2 vector, but not Ad vector, transduced deeper zones of cartilage and menisci. This study demonstrates that AAV2 and scAAV2 are reliable vectors for use in cartilage in vitro and in vivo.


Asunto(s)
Adenoviridae/genética , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Osteoartritis/terapia , Transducción Genética , Animales , Cartílago Articular/patología , Modelos Animales de Enfermedad , Perros , Expresión Génica , Genes Reporteros/genética , Vectores Genéticos/uso terapéutico , Cobayas , Osteoartritis/patología , Resultado del Tratamiento
15.
J Orthop Res ; 26(6): 764-71, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18241059

RESUMEN

This study evaluated healing of equine metatarsal osteotomies and ostectomies in response to percutaneous injection of adenoviral (Ad) bone morphogenetic protein (BMP)-2, Ad-BMP-6, or beta-galactosidase protein vector control (Ad-LacZ) administered 14 days after surgery. Radiographic and quantitative computed tomographic assessment of bone formation indicated greater and earlier mineralized callus in both the osteotomies and ostectomies of the metatarsi injected with Ad-BMP-2 or Ad-BMP-6. Peak torque to failure and torsional stiffness were greater in osteotomies treated with Ad-BMP-2 than Ad-BMP-6, and both Ad-BMP-2- and Ad-BMP-6-treated osteotomies were greater than Ad-LacZ or untreated osteotomies. Gene expression of ostectomy mineralized callus 8 weeks after surgery indicated upregulation of genes related to osteogenesis compared to intact metatarsal bone. Expression of transforming growth factor beta-1, cathepsin H, and gelsolin-like capping protein were greater in Ad-BMP-2- and Ad-BMP-6-treated callus compared to Ad-LacZ-treated or untreated callus. Evidence of tissue biodistribution of adenovirus in distant organs was not identified by quantitative PCR, despite increased serum antiadenoviral vector antibody. This study demonstrated a greater relative potency of Ad-BMP-2 over Ad-BMP-6 in accelerating osteotomy healing when administered in this regimen, although both genes were effective at increasing bone at both osteotomy and ostectomy sites.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Curación de Fractura/genética , Fracturas Óseas/terapia , Terapia Genética/métodos , Osteogénesis/genética , Osteotomía , Factor de Crecimiento Transformador beta/genética , Adenoviridae/genética , Animales , Fenómenos Biomecánicos , Proteína Morfogenética Ósea 2 , Proteína Morfogenética Ósea 6 , Callo Óseo/diagnóstico por imagen , Callo Óseo/fisiopatología , Modelos Animales de Enfermedad , Fracturas Óseas/diagnóstico por imagen , Fracturas Óseas/fisiopatología , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Caballos , Humanos , Operón Lac , Huesos Metatarsianos/diagnóstico por imagen , Huesos Metatarsianos/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Tomografía Computarizada por Rayos X , Torque , Torsión Mecánica
16.
Gynecol Oncol ; 103(3): 1054-62, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16870238

RESUMEN

OBJECTIVES: Capsid-modified AAV vectors can mediate enhanced gene transfer to neoplasms characterized by low AAV receptor expression. Here we sought to determine the therapeutic potential of a capsid-modified AAV vector for gene therapy of ovarian carcinoma (OvCa). METHODS: We tested a panel of OvCa cell lines for AAV2-mediated gene transduction and for sensitivity to ganciclovir (GCV) following AAVHSVtk administration. Levels of AAV internalization and attachment receptor were assessed by flow cytometry and immunohistochemistry. The role of receptors in AAV-mediated gene transfer was assessed by competition assays. Finally, we examined the ability of a modified vector with an integrin-binding RGD motif inserted into the AAV capsid to improve gene delivery to OvCa and enhance AAVHSVtk/GCV-mediated killing by cytotoxicity assay. RESULTS: All OvCa cell lines were poorly transduced with AAV2 vectors and showed variably sensitive to AAVHSVtk/GCV. While OvCa cell lines expressed AAV2 internalization receptors (alphav integrins), expression of the AAV2 attachment receptor, HSPG, was variable and not detected on many lines. Analysis of archived clinical specimens showed no detectable HSPG expression on approximately 45% of primary human tumors. Gene transfer to OvCa was increased several fold using the RGD-modified vector. Gene transfer was independent of HSPG and specific to the targeted receptor. Importantly, the RGD-modified capsid markedly increased the ability of the AAVHSVtk to kill OvCa cells in the presence of GCV. CONCLUSIONS: The development of AAV vectors targeted to cell surface receptors other than HSPG will be critical to the advancement of AAV-mediated gene therapy for treating OvCa.


Asunto(s)
Vectores Genéticos , Proteoglicanos de Heparán Sulfato/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Adenoviridae/metabolismo , Cápside/química , Línea Celular Tumoral , Femenino , Citometría de Flujo , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Humanos , Inmunohistoquímica , Neoplasias Ováricas/patología , Plásmidos/metabolismo
17.
Mol Ther ; 14(1): 97-106, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16624620

RESUMEN

The development of rationally designed targeted gene delivery vectors is an important focus for gene therapy. While genetic modification of AAV can produce vectors with modified tropism, incorporation of targeting peptides into the structural context of the AAV virion often results in loss of function or loss of virion integrity. To address this issue, we have developed a targeting system using metabolically biotinylated AAV. We generated serotype 1, 2, 3, 4, and 5 AAV capsids with small peptide insertions that are metabolically biotinylated in packaging cells during vector production by coexpression of the Escherichia coli BirA, biotin ligase, gene. Biotin moieties are exposed on the surface of assembled AAV particles and can interact with avidin. Metabolically biotinylated AAV vectors produced in this manner maintained endogenous titer and tissue tropism, could be purified on monomeric avidin resin, and could be retargeted to cells engineered to express an artificial avidin-biotin receptor. This technology provides not only a single platform for the purification of multiple AAV vector serotypes, but also a means for the development of multiple targeted AAV vectors utilizing a single capsid modification via straightforward avidin-biotin ligand coupling.


Asunto(s)
Biotinilación/métodos , Dependovirus/genética , Vectores Genéticos/aislamiento & purificación , Secuencia de Aminoácidos , Avidina/química , Biotina/química , Western Blotting , Proteínas de la Cápside/genética , Línea Celular , Dependovirus/clasificación , Terapia Genética/métodos , Vectores Genéticos/química , Vectores Genéticos/genética , Células HeLa , Humanos , Péptidos/genética , Péptidos/metabolismo , Plásmidos/genética , Serotipificación
18.
J Gene Med ; 8(5): 589-602, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16506246

RESUMEN

BACKGROUND: Glaucoma is a chronic eye disease which leads to irreversible blindness. The trabecular meshwork tissue controls intraocular pressure (IOP), which is the major risk factor for glaucoma. Gene therapy treatment of chronic diseases requires the use of long-term expression, low toxicity and lack of immune response vectors. Adeno-associated viruses (AAV) possess these characteristics but have been unable to transduce the trabecular meshwork. Because of the importance of regulating elevated IOP by long-term gene therapy, we investigated mechanisms of AAV transduction to the human trabecular meshwork (TM). METHODS: Primary human trabecular meshwork cells (HTM) and perfused organ cultures were infected with rAAV2-GFP, RGD-pseudotyped rAAV2-GFP alone, or combined with recombinant DeltaE1/E3 adenoviruses. Intracellular rAAV2 DNA and RNA were measured by relative quantitative and real-time TaqMan polymerase chain reaction (PCR). Host transcriptome was analyzed using high-density oligonucleotide microarrays. One transduction mechanism was tested using self-complementary AAV (scAAV). RESULTS: The dramatic transduction enhancement obtained upon co-infection of rAAV2 with DeltaE1/E3 adenoviruses provides insights into transduction mechanisms in the HTM. Even if not transduced, rAAV2 enters TM cells. GeneChip analysis showed significant changes in host genes involved in cell cycle and DNA replication. Consequently, scAAV-GFP transduction was highly efficient. Other transduction-enhancement genes included coxsackie adenovirus receptor (CAR) and genes relevant to trabecular meshwork function. CONCLUSIONS: The rate-limiting step of AAV transduction was not viral entry failure but, at least in part, host downregulation of DNA replication. Additional specific host genes might be involved. Our study revealed genes and mechanisms which led for the first time to efficient AAV transduction of the HTM.


Asunto(s)
Dependovirus/genética , Glaucoma/terapia , Malla Trabecular/patología , Transducción Genética , Cámara Anterior/metabolismo , Cámara Anterior/patología , Línea Celular , Perfilación de la Expresión Génica , Terapia Genética/métodos , Vectores Genéticos , Glaucoma/genética , Glaucoma/patología , Proteínas Fluorescentes Verdes/genética , Humanos , Presión Intraocular/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Técnicas de Cultivo de Órganos , Perfusión , Malla Trabecular/metabolismo
19.
J Virol ; 79(23): 14781-92, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16282478

RESUMEN

Although adeno-associated virus (AAV) infection is common in humans, the biology of natural infection is poorly understood. Since it is likely that many primary AAV infections occur during childhood, we set out to characterize the frequency and complexity of circulating AAV isolates in fresh and archived frozen human pediatric tissues. Total cellular DNA was isolated from 175 tissue samples including freshly collected tonsils (n = 101) and archived frozen samples representing spleen (n = 21), lung (n = 16), muscle (n = 15), liver (n = 19), and heart (n = 3). Samples were screened for the presence of AAV and adenovirus sequences by PCR using degenerate primers. AAV DNA was detected in 7 of 101 (7%) tonsil samples and two of 74 other tissues (one spleen and one lung). Adenovirus sequences were identified in 19 of 101 tonsils (19%), but not in any other tissues. Complete capsid gene sequences were recovered from all nine AAV-positive tissues. Sequence analyses showed that eight of the capsid sequences were AAV2-like (approximately 98% amino acid identity), while the single spleen isolate was intermediate between serotypes 2 and 3. Comparison to the available AAV2 crystal structure revealed that the majority of the amino acid substitutions mapped to surface-exposed hypervariable domains. To further characterize the AAV capsid structure in these samples, we used a novel linear rolling-circle amplification method to amplify episomal AAV DNA and isolate infectious molecular clones from several human tissues. Serotype 2-like viruses were generated from these DNA clones and interestingly, failed to bind to a heparin sulfate column. Inspection of the capsid sequence from these two clones (and the other six AAV2-like isolates) revealed that they lacked arginine residues at positions 585 and 588 of the capsid protein, which are thought to be essential for interaction with the heparin sulfate proteoglycan coreceptor. These data provide a framework with which to explore wild-type AAV persistence in vivo and provide additional tools to further define the biodistribution and form of AAV in human tissues.


Asunto(s)
Proteínas de la Cápside/metabolismo , Dependovirus/genética , Infecciones por Parvoviridae/genética , Secuencia de Aminoácidos , Proteínas de la Cápside/genética , Niño , ADN Viral/química , Dependovirus/aislamiento & purificación , Heparina/metabolismo , Humanos , Datos de Secuencia Molecular , Filogenia , Reacción en Cadena de la Polimerasa
20.
Mol Ther ; 7(4): 515-25, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12727115

RESUMEN

Recombinant adeno-associated virus (AAV) has become an attractive vector system for a number of gene therapy paradigms. However, the utility of AAV vectors is often limited by the absence of heparan sulfate proteoglycan (HSPG), the virus's primary attachment receptor, on the desired target cell population. In order to achieve HSPG-independent gene delivery, several groups have shown that the endogenous tropism of AAV can be expand by genetically altering the viral capsid. However, the parameters of this developing technology have yet to be defined and it has not yet been determined if these modified vectors actually infect cells via these engineered interactions. Previously we constructed a series of insertion mutants spanning the AAV capsid protein gene and identified specific sites that can tolerate the insertion of small exogenous peptides. Here we describe a number of sites within the AAV capsid gene that can be used for the insertion of integrin-targeting peptide epitopes. Incorporation of an Arg-Gly-Asp (RGD)-containing peptide at these sites enables AAV to infect integrin-expressing cells independent of HSPG. Mutant AAV vectors displaying these peptide ligands can be produced to wild-type titer and have been shown to specifically interact with the targeted integrin receptors and mediate infection via this interaction. We report significant increases in gene transfer to Raji, K562, and SKOV-3 cell lines that express integrin, but little HSPG, suggesting that rAAV vectors displaying RGD peptides may be of great utility for treatment of neoplasms characterized by the deficiency of HSPG expression. We have also demonstrated that due to their expanded tropism, these novel vectors are capable of efficient transduction of AAV2-resistant tumors in vivo suggesting that they may offer significant therapeutic advantages.


Asunto(s)
Proteínas de la Cápside/genética , Dependovirus/genética , Vectores Genéticos , Heparina/análogos & derivados , Oligopéptidos/genética , Animales , Transporte Biológico , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Técnicas de Cultivo de Célula , Dependovirus/metabolismo , Epítopos/metabolismo , Femenino , Vectores Genéticos/genética , Células HeLa , Heparina/metabolismo , Humanos , Integrinas/metabolismo , Células K562 , Ratones , Ratones SCID , Oligopéptidos/química , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Proteoglicanos/metabolismo , Células Tumorales Cultivadas , Virión/genética , Virión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA