Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
J Med Virol ; 86(4): 678-86, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24482297

RESUMEN

Multiple human proteins have been shown to both support and restrict viral replication, and confirmation of virus-associated changes in the expression of these genes is relevant for future therapeutic efforts. In this study a well-characterized panel of 49 individuals either infected with HIV-1 or uninfected was compiled and analyzed for the effect of HIV infection status, viral load, and antiretroviral treatment on specific gene expression. mRNA was extracted and reverse transcribed from purified CD4+ cells, and quantitative real-time PCR was utilized to scrutinize differences in the expression of four host genes that have been demonstrated to either stimulate (HSP90 and LEDGF/p75) or restrict (p21/WAF1 and APOBEC3G) proviral integration. HIV infection status was associated with slight to moderate alterations in the expression of all four genes. After adjusting for age, mRNA expression levels of HSP90, LEDGF/p75 and APOBEC3G were found to all be decreased in infected patients compared to healthy controls by 1.43-, 1.26-, and 4.71-fold, respectively, while p21/WAF1 expression was increased 2.35-fold. Furthermore, individuals receiving raltegravir exhibited a 1.28-fold reduction in LEDGF/p75 compared to those on non-raltegravir antiretroviral treatment. Identification of these and similar HIV-induced changes in gene expression may be valuable for delineating the extent of host cell molecular mechanisms stimulating viral replication.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Citidina Desaminasa/biosíntesis , Infecciones por VIH/inmunología , Proteínas HSP90 de Choque Térmico/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Desaminasa APOBEC-3G , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Adolescente , Adulto , Linfocitos T CD4-Positivos/inmunología , Niño , Estudios Transversales , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Citidina Desaminasa/genética , Femenino , Expresión Génica , Regulación de la Expresión Génica , Infecciones por VIH/tratamiento farmacológico , VIH-1/genética , Proteínas HSP90 de Choque Térmico/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Persona de Mediana Edad , Pirrolidinonas/uso terapéutico , ARN Mensajero/biosíntesis , Raltegravir Potásico , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Adulto Joven
3.
J Vis Exp ; (76)2013 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-23851977

RESUMEN

Lentiviral vectors (LVs) are a powerful means of delivering genetic material to many types of cells. Because of safety concerns associated with these HIV-1 derived vectors, producing large quantities of LVs is challenging. In this paper, we report a method for producing high titers of self-inactivating LVs. We retrovirally transduce the tet-off stable producer cell line GPR to generate a cell line, GPRS, which can express all the viral components, including a dendritic cell-specific glycoprotein, SVGmu. Then, we use concatemeric DNA transfection to transfect the LV transfer plasmid encoding a reporter gene GFP in combination with a selectable marker. Several of the resulting clones can produce LV at a titer 10-fold greater than what we achieve with transient transfection. Plus, these viruses efficiently transduce dendritic cells in vitro and generate a strong T cell immune response to our reporter antigen. This method may be a good option for producing strong LV-based vaccines for clinical studies of cancer or infectious diseases.


Asunto(s)
Células Dendríticas/virología , Lentivirus/genética , Tetraciclina/farmacología , Transfección/métodos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Línea Celular , Células Dendríticas/inmunología , Vectores Genéticos/genética , Células HEK293 , Humanos , Lentivirus/inmunología , Plásmidos/genética
4.
Biomaterials ; 34(12): 3098-109, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23375392

RESUMEN

Liposomes constitute one of the most popular nanocarriers for the delivery of cancer therapeutics. However, since their potency is limited by incomplete drug release and inherent instability in the presence of serum components, their poor delivery occurs in certain circumstances. In this study, we address these shortcomings and demonstrate an alternative liposomal formulation, termed crosslinked multilamellar liposome (CML). With its properties of improved sustainable drug release kinetics and enhanced vesicle stability, CML can achieve controlled delivery of cancer therapeutics. CML stably encapsulated the anticancer drug doxorubicin (Dox) in the vesicle and exhibited a remarkably controlled rate of release compared to that of the unilamellar liposome (UL) with the same lipid composition or Doxil-like liposome (DLL). Our imaging study demonstrated that the CMLs were mainly internalized through a caveolin-dependent pathway and were further trafficked through the endosome-lysosome compartments. Furthermore, in vivo experiments showed that the CML-Dox formulation reduced systemic toxicity and significantly improved therapeutic activity in inhibiting tumor growth compared to that of UL-Dox or DLL-Dox. This drug packaging technology may therefore provide a new treatment option to better manage cancer and other diseases.


Asunto(s)
Antineoplásicos/administración & dosificación , Liposomas , Animales , Portadores de Fármacos , Femenino , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL
5.
Small ; 9(3): 421-9, 2013 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-23038676

RESUMEN

As a consequence of their well-defined nanostructure and intrinsic bioactive functionality, virus-based nanoparticles have shown promise for mediating gene delivery. Adeno-associated virus (AAV) nanoparticles, which possess an excellent safety profile and therapeutic potential, hold potential for use in human gene therapy. However, because of their native tropisms, the applicability of AAV nanoparticles is often limited to restricted ranges of cells or tissues. Thus, retargeting AAV particles to the desired cell populations has continued to be a major research focus in many gene therapy applications. In this study, a general strategy is reported for nanoparticle targeting. This involves the site-specific modification of AAV type 2 (AAV2) by genetically incorporating a short peptide, in this case an aldehyde tag, in the viral capsid. Such a tag can be exploited for site-specific attachment of targeting molecules and allows for further introduction of targeting antibodies or ligands. It is shown that this modification neither affects the level of infectious viral titer nor intracellular trafficking properties. Furthermore, the site-specific conjugation of targeting antibodies could significantly enhance viral transduction to those target cells that have otherwise exhibited very low permissiveness to AAV2 infection. This method also allows the functional incorporation of RGD peptides onto AAV2 for enhanced delivery with implications for cancer gene therapy.


Asunto(s)
Dependovirus/química , Dependovirus/genética , Ingeniería Genética/métodos , Línea Celular , Dependovirus/fisiología , Células HeLa , Células Hep G2 , Humanos , Oligopéptidos/química
6.
Biotechnol Bioeng ; 109(6): 1551-60, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22179950

RESUMEN

Lentiviral vectors (LVs) enveloped with an engineered Sindbis virus glycoprotein can specifically bind to dendritic cells (DCs) through the surface receptor DC-SIGN and induce antigen expression, thus providing an efficient method for delivering DC-directed vaccines. In this study, we constructed a stable producer line (LV-MGFP) for synthesizing DC-SIGN-targeted HIV-1-based LVs (DC-LVs) encoding green fluorescent protein (GFP) by a concatemeric array transfection technique. We demonstrated that the established stable clones could routinely produce vector supernatants with titers above 10(7) transduction units per milliliter (TU/mL) during a continuous 3-month cell passage. The producer cells were also capable of generating similar titers of DC-LVs in serum-free medium. Moreover, the addition of 1-deoxymannojirimycin (DMJ) enabled the producer cells to manufacture DC-LVs with both improved titers and enhanced potency to evoke antigen-specific CD8(+) T cell responses in mice. The stable lines could accommodate the replacement of the internal murine stem cell virus (MSCV) promoter with the human ubiquitin-C (Ubi) promoter in the lentiviral backbone. The resulting DC-LVs bearing Ubi exhibited the enhanced potency to elicit vaccine-specific immunity. Based on accumulated evidence, our studies support the application of this production method in manufacturing DC-LVs for preclinical and clinical testing of novel DC-based immunization.


Asunto(s)
Biotecnología/métodos , Células Dendríticas/inmunología , Vectores Genéticos , VIH-1/genética , Tecnología Farmacéutica/métodos , Transducción Genética , Vacunación/métodos , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular , Medio de Cultivo Libre de Suero , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Virus Sindbis/genética , Coloración y Etiquetado , Técnicas de Cultivo de Tejidos/métodos , Proteínas del Envoltorio Viral/genética , Carga Viral
7.
Virus Res ; 160(1-2): 340-50, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21802459

RESUMEN

Lentiviral vectors (LVs) derived from human immunodeficiency virus type 1 (HIV-1) are promising vehicles for gene delivery because they not only efficiently transduce both dividing and non-dividing cells, but also maintain long-term transgene expression. Development of an LV system capable of transducing cells in a cell type-specific manner can be beneficial for certain applications that rely on targeted gene delivery. Previously it was shown that an inverse fusion strategy that incorporated an HIV-1 receptor (CD4) and its co-receptor (CXCR4 or CCR5) onto vector surfaces could confer to LVs the ability to selectively deliver genes to HIV-1 envelope-expressing cells. To build upon this work, we aim to improve its relatively low transduction efficiency and circumvent its inability to target multiple tropisms of HIV-1 by a single vector. We investigated a method to create LVs co-enveloped with the HIV-1 cellular receptor CD4 and a fusogenic protein derived from the Sindbis virus glycoprotein and tested its efficiency to selectively deliver genes into cells expressing HIV-1 envelope proteins. The engineered LV system yields a higher level of transduction efficiency and a broader tropism towards cells displaying the HIV-1 envelope protein (Env) than the previously developed system. Furthermore, we demonstrated in vitro that this engineered LV can preferentially deliver suicide gene therapy to HIV-1 envelope-expressing cells. We conclude that it is potentially feasible to target LVs towards HIV-1-infected cells by functional co-incorporation of the CD4 and fusogenic proteins, and provide preliminary evidence for further investigation on a potential alternative treatment for eradicating HIV-1-infected cells that produce drug-resistant viruses after highly active antiretroviral therapy (HAART).


Asunto(s)
Antígenos CD4/metabolismo , Expresión Génica , VIH-1/genética , Receptores del VIH/metabolismo , Transducción Genética , Proteínas Virales de Fusión/metabolismo , Productos del Gen env del Virus de la Inmunodeficiencia Humana/metabolismo , Antígenos CD4/genética , Línea Celular , Técnicas de Transferencia de Gen , Ingeniería Genética , Terapia Genética/métodos , Vectores Genéticos , Humanos , Receptores del VIH/genética , Virus Sindbis/genética , Proteínas Virales de Fusión/genética , Acoplamiento Viral , Productos del Gen env del Virus de la Inmunodeficiencia Humana/genética
8.
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
9.
Mol Ther ; 19(5): 942-50, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21326219

RESUMEN

Human embryonic stem (hES) cells are renewable cell sources that have potential applications in regenerative medicine. The development of technologies to produce permanent and site-specific genome modifications is in demand to achieve future medical implementation of hES cells. We report herein that a baculoviral vector (BV) system carrying zinc-finger nucleases (ZFNs) can successfully modify the hES cell genome. BV-mediated transient expression of ZFNs specifically disrupted the CCR5 locus in transduced cells and the modified cells exhibited resistance to HIV-1 transduction. To convert the BV to a gene targeting vector, a DNA donor template and ZFNs were incorporated into the vector. These hybrid vectors yielded permanent site-specific gene addition in both immortalized human cell lines (10%) and hES cells (5%). Modified hES cells were both karyotypically normal and pluripotent. These results suggest that this baculoviral delivery system can be engineered for site-specific genetic manipulation in hES cells.


Asunto(s)
Desoxirribonucleasas/genética , Células Madre Embrionarias/metabolismo , Nucleopoliedrovirus/genética , Receptores CCR5/genética , Línea Celular , Desoxirribonucleasas/metabolismo , Citometría de Flujo , Marcación de Gen , Vectores Genéticos , VIH-1/genética , Humanos , Mutación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción Genética , Transgenes , Dedos de Zinc
10.
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
11.
ACS Nano ; 2(8): 1553-62, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19079775

RESUMEN

This study reports a general method of labeling enveloped viruses with semiconductor quantum dots (QDs) for use in single virus trafficking studies. Retroviruses, including human immunodeficiency virus (HIV), could be successfully tagged with QDs through the membrane incorporation of a short acceptor peptide (AP) that is susceptible to site-specific biotinylation and attachment of streptavidin-conjugated QDs. It was found that this AP tag-based QD labeling had little effect on the viral infectivity and allowed for the study of the kinetics of the internalization of the recombinant lentivirus enveloped with vesicular stomatitis virus glycoprotein (VSVG) into the early endosomes. It also allows for the live cell imaging of the trafficking of labeled virus to the Rab5(+) endosomal compartments. This study further demonstrated by direct visualization of QD-labeled virus that VSVG-pseudotyped lentivirus enters cells independent of clatherin- and caveolin-pathways, while the entry of VSVG-pseudotyped retrovirus occurs via the clathrin pathway. The studies monitoring HIV particles using QD-labeling showed that we could detect single virions on the surface of target cells expressing either CD4/CCR5 or DC-SIGN. Further internalization studies of QD-HIV evidently showed that the clathrin pathway is the major route for DC-SIGN-mediated uptake of viruses. Taken together, our data demonstrate the potential of this QD-labeling for visualizing the dynamic interactions between viruses and target cell structures.


Asunto(s)
Microscopía Fluorescente/métodos , Nanotecnología/métodos , Puntos Cuánticos , Vesiculovirus/fisiología , Vesiculovirus/ultraestructura , Medios de Contraste , Conformación Molecular , Coloración y Etiquetado/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...