Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Base de datos
Tipo de estudio
Intervalo de año de publicación
1.
PLoS One ; 8(6): e67400, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23840690

RESUMEN

Dendritic cells (DCs) are potent antigen-presenting cells and therefore have enormous potential as vaccine targets. We have previously developed an engineered lentiviral vector (LV) that is pseudotyped with a mutated Sindbis virus glycoprotein (SVGmu), which is capable of targeting DCs through Dendritic Cell-specific ICAM3-grabbing Nonintegrin (DC-SIGN), a receptor that is predominantly expressed by DCs. In this study, we aimed to elucidate the internalization and trafficking mechanisms of this viral vector system through direct visualization of GFP-Vpr-tagged viral particles in target DCs, which was further corroborated by drug inhibition and dominant-negative mutants of cellular proteins that regulate the endocytic traffic. We demonstrated that our engineered LVs enter the cell via receptor-mediated clathrin- and dynamin-dependent endocytosis. Microtubule networks were also involved in a productive infection. Viral vector fusion was low-pH-dependent and occurred in the early endosomal stage of the intracellular transport. Autophagy was also examined for its effect on transduction efficiency, and we observed that enhanced autophage activity reduced vector infectivity, while suppressed autophagy boosted transduction efficiency. This study shed some light on the internalization and trafficking mechanisms of DC-directed LVs and offers some strategies to further improve the efficiency of LV-mediated gene therapy.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Lentivirus/genética , Receptores de Superficie Celular/metabolismo , Internalización del Virus , Autofagia , Clatrina/metabolismo , Células Dendríticas/virología , Endocitosis , Endosomas/metabolismo , Ingeniería Genética , Vectores Genéticos , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Concentración de Iones de Hidrógeno , Lentivirus/fisiología , Lisosomas/fisiología , Microscopía Confocal , Microtúbulos/metabolismo , Análisis de la Célula Individual , Transducción Genética
2.
PLoS One ; 6(6): e21491, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21738680

RESUMEN

Lentiviruses have recently attracted considerable interest for their potential as a genetic modification tool for dendritic cells (DCs). In this study, we explore the ability of lentiviruses enveloped with alphaviral envelope glycoproteins derived from Semliki Forest virus (SFV) to mediate transduction of DCs. We found that SFV glycoprotein (SFV-G)-pseudotyped lentiviruses use C-type lectins (DC-SIGN and L-SIGN) as attachment factors for transduction of DCs. Importantly, SFV-G pseudotypes appear to have enhanced transduction towards C-type lectin-expressing cells when produced under conditions limiting glycosylation to simple high-mannose, N-linked glycans. These results, in addition to the natural DC tropism of SFV-G, offer evidence to support the use of SFV-G-bearing lentiviruses to genetically modify DCs for the study of DC biology and DC-based immunotherapy.


Asunto(s)
Células Dendríticas/virología , Glicoproteínas/metabolismo , Lentivirus/genética , Receptores Virales/metabolismo , Virus de los Bosques Semliki/metabolismo , Transducción Genética/métodos , Proteínas del Envoltorio Viral/metabolismo , Línea Celular , Células Cultivadas , Glicoproteínas/genética , Humanos , Plásmidos , Receptores Virales/genética , Virus de los Bosques Semliki/genética , Proteínas del Envoltorio Viral/genética
3.
Hum Gene Ther ; 22(10): 1281-91, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21452926

RESUMEN

Lentiviral vectors (LVs) pseudotyped with envelope proteins of alphaviruses have recently attracted considerable interest for their potential as gene delivery tools. We report the production of human immunodeficiency virus type 1 (HIV-1)-derived LVs pseudotyped with envelope glycoproteins derived from the Aura virus (AURA). We found that the AURA-glycoprotein-pseudotyped LVs use C-type lectins (DC-SIGN and L-SIGN) as attachment factors. These interactions with DC-SIGN are specific as determined by inhibition assays and appear to facilitate transduction through a pH-dependent pathway. AURA-pseudotyped LVs were used to transduce monocyte-derived dendritic cells (DCs) and the transduction was shown to be DC-SIGN mediated, as illustrated by competitive inhibition with DC-SIGN and L-SIGN antibodies and yeast mannan. Comparisons with LVs enveloped with glycoproteins derived from vesicular stomatitis virus and Sindbis virus suggest that AURA-glycoprotein-bearing LVs might be useful to genetically modify DCs for the study of DC biology and DC-based immunotherapy.


Asunto(s)
Alphavirus/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Dendríticas/metabolismo , Técnicas de Transferencia de Gen , Ingeniería Genética/métodos , Vectores Genéticos/biosíntesis , VIH-1/genética , Lectinas Tipo C/metabolismo , Receptores de Superficie Celular/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Animales , Moléculas de Adhesión Celular/genética , Genes prv/genética , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Lectinas Tipo C/genética , Ratones , Microscopía Confocal , Células 3T3 NIH , Plásmidos/genética , Receptores de Superficie Celular/genética , Transducción Genética/métodos
4.
ACS Nano ; 5(5): 3523-35, 2011 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-21473596

RESUMEN

The unique spectral properties of semiconductor quantum dots (QDs) enable long-term live-cell imaging and ultrasensitive detection of viral particles, which in turn can potentially provide a practical means for detailed analysis of the underlying molecular mechanisms of virus entry. In this study, we report a general method of labeling adeno-associated virus serotype 2 (AAV2) with QDs for enhanced visualization of the intracellular behavior of viruses in living target cells. It was found that the mild conditions required for this QD conjugation reaction allowed for the retention of viral infectivity of AAV2. Furthermore, quantitative analysis of viral motility in living cells suggested that QD-labeling had no significant effect on the intracellular transport properties of AAV2 particles compared to those of conventional organic dye-labeled AAV2. Our imaging study demonstrated that QD-AAV2 was internalized mainly through a clathrin-dependent pathway and then trafficked through various endosomes. It was also observed that QD-AAV2 particles exploit the cytoskeleton network to facilitate their transport within cells, and the labeling study provided evidence that the ubiquitin-proteasome system was likely involved in the intracellular trafficking of AAV2, at least at the level of nuclear transport. Taken together, our findings reveal the potential of this QD-labeling method for monitoring the intracellular dynamics of virus-host cell interactions and interrogating the molecular mechanisms of viral infection in greater detail.


Asunto(s)
Adenoviridae/fisiología , Adenoviridae/ultraestructura , Aumento de la Imagen/métodos , Microscopía Fluorescente/métodos , Puntos Cuánticos , Ensamble de Virus/fisiología , Medios de Contraste
5.
J Biol Eng ; 5(1): 1, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21276219

RESUMEN

BACKGROUND: Dendritic cells (DCs) are antigen-presenting immune cells that interact with T cells and have been widely studied for vaccine applications. To achieve this, DCs can be manipulated by lentiviral vectors (LVs) to express antigens to stimulate the desired antigen-specific T cell response, which gives this approach great potential to fight diseases such as cancers, HIV, and autoimmune diseases. Previously we showed that LVs enveloped with an engineered Sindbis virus glycoprotein (SVGmu) could target DCs through a specific interaction with DC-SIGN, a surface molecule predominantly expressed by DCs. We hypothesized that SVGmu interacts with DC-SIGN in a mannose-dependent manner, and that an increase in high-mannose structures on the glycoprotein surface could result in higher targeting efficiencies of LVs towards DCs. It is known that 1-deoxymannojirimycin (DMJ) can inhibit mannosidase, which is an enzyme that removes high-mannose structures during the glycosylation process. Thus, we investigated the possibility of generating LVs with enhanced capability to modify DCs by supplying DMJ during vector production. RESULTS: Through western blot analysis and binding tests, we were able to infer that binding of SVGmu to DC-SIGN is directly related to amount of high-mannose structures on SVGmu. We also found that the titer for the LV (FUGW/SVGmu) produced with DMJ against 293T.DCSIGN, a human cell line expressing the human DC-SIGN atnibody, was over four times higher than that of vector produced without DMJ. In addition, transduction of a human DC cell line, MUTZ-3, yielded a higher transduction efficiency for the LV produced with DMJ. CONCLUSION: We conclude that LVs produced under conditions with inhibited mannosidase activity can effectively modify cells displaying the DC-specific marker DC-SIGN. This study offers evidence to support the utilization of DMJ in producing LVs that are enhanced carriers for the development of DC-directed vaccines.

6.
Immunol Rev ; 239(1): 45-61, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21198664

RESUMEN

The increasing level of understanding of the lentivirus biology has been instrumental in shaping the design strategy of creating therapeutic lentiviral delivery vectors. As a result, lentiviral vectors have become one of the most powerful gene transfer vehicles. They are widely used for therapeutic purposes as well as in studies of basic biology, due to their unique characteristics. Lentiviral vectors have been successfully employed to mediate durable and efficient antigen expression and presentation in dendritic cells both in vitro and in vivo, leading to the activation of cellular immunity and humoral responses. This capability makes the lentiviral vector an ideal choice for immunizations that target a wide range of cancers and infectious diseases. Further advances into optimizing the vector system and understanding the relationship between the immune system and diseases pathogenesis will only augment the potential benefits and utility of lentiviral vaccines for human health.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Inmunización/métodos , Lentivirus/genética , Neoplasias/inmunología , Linfocitos T/inmunología , Vacunas Virales/inmunología , Virosis/inmunología , Adyuvantes Inmunológicos , Animales , Presentación de Antígeno , Células Presentadoras de Antígenos/inmunología , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/inmunología , Vectores Genéticos , Humanos , Ratones , Neoplasias/terapia , Vacunas Sintéticas , Vacunas Virales/uso terapéutico , Virosis/prevención & control , Virosis/terapia
7.
Microsc Res Tech ; 73(9): 886-900, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20232362

RESUMEN

Membrane fusion plays an essential role in the entry of enveloped viruses into target cells. The merging of viral and target cell membranes is catalyzed by viral fusion proteins, which involves multiple sequential steps in the fusion process. However, the fusion mechanisms mediated by different fusion proteins involve multiple transient intermediates that have not been well characterized. Here, we report a synthetic virus platform that allows us to better understand the different fusion mechanisms driven by the diverse types fusion proteins. The platform consists of lentiviral particles coenveloped with a surface antibody, which serves as the binding protein, along with a fusion protein derived from either influenza virus (HAmu) or Sindbis virus (SINmu). By using a single virus tracking technique, we demonstrated that both HAmu- and SINmu-bearing viruses enter cells through clathrin-dependent endocytosis, but they required different endosomal trafficking routes to initiate viral fusion. Direct observation of single viral fusion events clearly showed that hemifusion mediated by SINmu upon exposure to low pH occurs faster than that mediated by HAmu. Monitoring sequential fusion processes by dual labeling the outer and inner leaflets of viral membranes also revealed that the SINmu-mediated hemifusion intermediate is relatively long-lived as compared with that mediated by HAmu. Taken together, we have demonstrated that the combination of this versatile viral platform with the techniques of single virus tracking can be a powerful tool for revealing molecular details of fusion mediated by various fusion proteins.


Asunto(s)
Membrana Celular/química , Células/química , Proteínas Virales de Fusión/análisis , Internalización del Virus , Virus/química , Línea Celular , Membrana Celular/virología , Células/virología , Humanos , Microscopía Confocal , Proteínas Virales de Fusión/metabolismo , Fenómenos Fisiológicos de los Virus
8.
Immunopharmacol Immunotoxicol ; 32(2): 208-18, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20085508

RESUMEN

Lentiviral vectors (LVs) are efficient gene delivery vehicles suitable for delivering long-term transgene expression in various cell types. Engineering LVs to have the capacity to transduce specific cell types is of great interest to advance the translation of LVs toward the clinic. Here we provide an overview of innovative approaches to target LVs to cells of the immune system. In this overview we distinguish between two types of LV targeting strategies: (i) targeting of the vectors to specific cells by LV surface modifications, and (ii) targeting at the level of transgene transcription by insertion of tissue-specific promoters to drive transgene expression. It is clear that each strategy is of enormous value but ultimately combining these approaches may help reduce the effects of off-target expression and improve the efficiency and safety of LVs for gene therapy.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos , Sistema Inmunológico/metabolismo , Lentivirus/genética , Animales , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Transducción Genética , Transgenes
9.
Hum Gene Ther ; 20(12): 1652-64, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19663564

RESUMEN

It has been demonstrated that nonintegrating lentiviral vectors (NILVs) are efficient in maintaining transgene expression in vitro and in vivo. Gene delivery by NILVs can significantly reduce nonspecific vector integration, which has been shown to cause malignant transformation in patients receiving gene therapy for X-linked severe combined immunodeficiency. Strong and sustained immune responses were observed after a single immunization with NILVs carrying viral antigens. However, there is no report to date that evaluates the efficacy of NILVs in inducing antigen-specific antitumor immunity. Using a well-characterized tumor model, we tested in vivo immunization with a self-inactivating lentiviral vector harboring a defective integrase. A high frequency of ovalbumin peptide (OVAp1)-specific CD8(+) T cells and a substantial antibody response were detected in naive mice immunized with an NILV encoding an OVA transgene. Furthermore, this immunization method completely protected the mice against the growth of E.G7 tumor cells expressing the OVA antigen. Thus, this study provides evidence that immunization using NILVs can be a safe and promising approach for exploring cancer immunotherapy.


Asunto(s)
Antígenos/genética , Vacunas contra el Cáncer/genética , Terapia Genética , Vectores Genéticos/genética , VIH-1/genética , Neoplasias/terapia , Ovalbúmina/genética , Animales , Antígenos/inmunología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/inmunología , Integrasa de VIH/genética , Inmunoterapia Activa , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Neoplasias/inmunología , Ovalbúmina/inmunología , Transducción Genética , Transgenes , Proteínas del Envoltorio Viral/genética , Integración Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA