Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunol Lett ; 194: 29-39, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29273425

RESUMEN

Macromolecules including DNA and proteins serve as important human therapeutics but are limited by their general inability to cross cell membranes. Supercharged proteins have been known as potent tools for delivery of macromolecules into mammalian cells. Thus, the use of these delivery systems is important to reduce the human papillomavirus (HPV)-associated malignancies through improvement of vaccine modalities. In this study, we used a supercharged green fluorescent protein (+36 GFP) for delivery of the full-length HPV16 E7 DNA and protein into mammalian cells and evaluated immune responses, and protective/therapeutic effects of different formulations in C57BL/6 tumor mice model. Our results showed that the complexes of E7 DNA/+36 GFP and also E7 protein/+36 GFP form stable nanoparticles through non-covalent binding with an average size of ∼ 200-300 nm. The efficient delivery of E7 DNA or protein by +36 GFP was detected in HEK-293T cell line for 4 h and 24 h post-transfection. Mice immunization with E7 protein/+36 GFP nanoparticles elicited a higher Th1 cellular immune response with the predominant IgG2a and IFN-γ levels than those induced by E7 protein, E7 DNA, E7 DNA/+36 GFP and control groups (p < .05). Moreover, the E7 DNA/+36 GFP and E7 protein/+36 GFP nanoparticles similarly protected mice against TC-1 tumor challenge (∼67%) as compared to E7 DNA and E7 protein (∼33%), respectively. These data suggest that +36 GFP may provide a promising platform to improve protein and DNA delivery in vitro and in vivo.


Asunto(s)
Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes , Papillomavirus Humano 16 , Proteínas E7 de Papillomavirus , ADN Viral/química , ADN Viral/farmacocinética , ADN Viral/farmacología , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/farmacocinética , Proteínas Fluorescentes Verdes/farmacología , Células HEK293 , Humanos , Proteínas E7 de Papillomavirus/química , Proteínas E7 de Papillomavirus/farmacocinética , Proteínas E7 de Papillomavirus/farmacología
2.
Int J Nanomedicine ; 9: 4609-19, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25356070

RESUMEN

We determined the efficacy and safety of chitosan (CS)-coated poly(lactic-co-glycolic) acid (PLGA) nanoparticles (NPs) as a delivery system for a vaccine to protect chickens against Newcastle disease virus (NDV). The newly constructed vaccine contained DNA (the F gene) of NDV. The Newcastle disease virus (NDV) F gene deoxyribonucleic acid (DNA) plasmid (pFDNA)-CS/PLGA-NPs were spherical (diameter =699.1 ± 5.21 nm [mean ± standard deviation]) and smooth, with an encapsulation efficiency of 98.1% and a Zeta potential of +6.35 mV. An in vitro release assay indicated that CS controlled the burst release of plasmid DNA, such that up to 67.4% of the entire quantity of plasmid DNA was steadily released from the pFDNA-CS/PLGA-NPs. An in vitro expression assay indicated that the expression of nanoparticles (NPs) was maintained in the NPs. In an immunization test with specific pathogen-free chickens, the pFDNA-CS/PLGA-NPs induced stronger cellular, humoral, and mucosal immune responses than the plasmid DNA vaccine alone. The pFDNA-CS/PLGA-NPs did not harm 293T cells in an in vitro assay and did not harm chickens in an in vivo assay. Overall, the results indicated that CS-coated PLGA NPs can serve as an efficient and safe mucosal immune delivery system for NDV DNA vaccine.


Asunto(s)
Quitosano/química , ADN Viral/farmacocinética , Portadores de Fármacos/química , Ácido Láctico/química , Nanopartículas/química , Virus de la Enfermedad de Newcastle/genética , Ácido Poliglicólico/química , Vacunas de ADN/farmacocinética , Animales , Pollos , Quitosano/farmacocinética , ADN Viral/genética , ADN Viral/inmunología , ADN Viral/metabolismo , Portadores de Fármacos/farmacocinética , Inmunoglobulina A/análisis , Ácido Láctico/farmacocinética , Linfocitos/inmunología , Enfermedad de Newcastle/prevención & control , Enfermedad de Newcastle/virología , Ácido Poliglicólico/farmacocinética , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Lágrimas/química , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética , Vacunas de ADN/inmunología
3.
J Control Release ; 158(3): 451-60, 2012 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-22207073

RESUMEN

As an effort to overcome limits of adenovirus (Ad) as a systemic delivery vector for cancer therapy, we developed a novel system using oncolytic Ad plasmid DNA with two bioreducible polymers: arginine-grafted bioreducible poly(disulfide amine)polymer (ABP) and PEG5k-conjugated ABP (ABP5k) in expectation of oncolytic effect caused by progeny viral production followed by replication. The linearized Ad DNAs for active viral replication polyplexed with each polymer were able to replicate only in human cancer cells and produce progeny viruses. The non-immunogenic polymers delivering the DNAs markedly elicited to evade the innate and adaptive immune response. The biodistribution ratio of the polyplexes administered systemically was approximately 99% decreased in liver when compared with naked Ad. Moreover, tumor-to-liver ratio of the Ad DNA delivered by ABP or ABP5k was significantly elevated at 229- or 419-fold greater than that of naked Ad, respectively. The ABP5k improved the chance of the DNA to localize within tumor versus liver with 1.8-fold increased ratio. In conclusion, the innovative and simple system for delivering oncolytic Ad plasmid DNA with the bioreducible polymers, skipping time-consuming steps such as generation and characterization of oncolytic Ad vectors, can be utilized as an alternative approach for cancer therapy.


Asunto(s)
Adenoviridae/genética , Arginina/administración & dosificación , ADN Viral/administración & dosificación , Virus Oncolíticos/genética , Poliaminas/administración & dosificación , Polietilenglicoles/administración & dosificación , Animales , Arginina/farmacocinética , Línea Celular Tumoral , ADN Viral/farmacocinética , Femenino , Vectores Genéticos/administración & dosificación , Células HEK293 , Humanos , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Viroterapia Oncolítica , Plásmidos , Poliaminas/farmacocinética , Polietilenglicoles/farmacocinética , Distribución Tisular
4.
PLoS One ; 6(7): e22524, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21799884

RESUMEN

BACKGROUND: Typically, DNA immunization via the intramuscular route induces specific, Th1-dominant immune responses. However, plasmids expressing viral proteins fused to cytotoxic T lymphocyte antigen 4 (CTLA-4) primed Th2-biased responses and were able to induced effective protection against viral challenge in the woodchuck model. Thus, we addressed the question in the mouse model how the Th1/Th2 bias of primed immune responses by a DNA vaccine influences hepatitis B virus (HBV) clearance. PRINCIPAL FINDINGS: Plasmids expressing HBV core protein (HBcAg) or HBV e antigen and HBcAg fused to the extracellular domain of CTLA-4 (pCTLA-4-HBc), CD27, and full length CD40L were constructed. Immunizations of these DNA plasmids induced HBcAg-specific antibody and cytotoxic T-cell responses in mice, but with different characteristics regarding the titers and subtypes of specific antibodies and intensity of T-cell responses. The plasmid pHBc expressing HBcAg induced an IgG2a-dominant response while immunizations of pCTLA-4-HBc induced a balanced IgG1/IgG2a response. To assess the protective values of the immune responses of different characteristics, mice were pre-immunized with pCTLA-4-HBc and pHBc, and challenged by hydrodynamic injection (HI) of pAAV/HBV1.2. HBV surface antigen (HBsAg) and DNA in peripheral blood and HBcAg in liver tissue were cleared with significantly accelerated kinetics in both groups. The clearance of HBsAg was completed within 16 days in immunized mice while more than 50% of the control mice are still positive for HBsAg on day 22. Stronger HBcAg-specific T-cell responses were primed by pHBc correlating with a more rapid decline of HBcAg expression in liver tissue, while anti-HBs antibody response developed rapidly in the mice immunized with pCTLA-4-HBc, indicating that the Th1/Th2 bias of vaccine-primed immune responses influences the mode of viral clearance. CONCLUSION: Viral clearance could be efficiently achieved by Th1/Th2-balanced immune response, with a small but significant shift in T-cell and B-cell immune responses.


Asunto(s)
Fusión Artificial Génica , Antígeno CTLA-4/genética , Antígenos del Núcleo de la Hepatitis B/genética , Virus de la Hepatitis B/inmunología , Inmunización/métodos , Células Th2/inmunología , Vacunas de ADN/inmunología , Animales , Especificidad de Anticuerpos , Linfocitos B/inmunología , Linfocitos B/virología , Ligando de CD40/genética , Ligando de CD40/inmunología , ADN Viral/administración & dosificación , ADN Viral/farmacocinética , Epítopos/inmunología , Femenino , Antígenos del Núcleo de la Hepatitis B/inmunología , Antígenos e de la Hepatitis B/genética , Antígenos e de la Hepatitis B/inmunología , Virus de la Hepatitis B/metabolismo , Inmunoglobulina G/inmunología , Hígado/inmunología , Hígado/metabolismo , Hígado/virología , Marmota , Tasa de Depuración Metabólica , Ratones , Plásmidos/genética , Células TH1/inmunología , Células TH1/virología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Vacunas de ADN/genética , Vacunas Virales/genética , Vacunas Virales/inmunología
5.
Biomaterials ; 30(29): 5767-74, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19577293

RESUMEN

Recombinant baculoviral vectors efficiently transduce many types of mammalian cells. However, their in vivo applications are hampered by the sensitivity of the virus to complement-mediated inactivation. Based on our observation that the surface charge of baculovirus is negative at neutral pH, we developed a procedure to coat baculoviral vectors with positively charged polyethylenimine 25 kDa, a commonly tested non-viral gene delivery vector, through electrostatic interaction. This coating was effective in protecting baculoviral vectors against human and rat serum-mediated inactivation in vitro, providing transduction efficiencies comparable with that generated by the control virus used under a serum-free condition. Enhanced in vivo gene expression in the liver and spleen was observed after tail vein injection of the coated viruses into mice. When injected directly into human tumor xenografts in nude mice, the coated viruses suppressed tumor development more effectively than uncoated viral vectors. These findings demonstrated the usefulness of using a simple coating method to circumvent a major obstacle to in vivo application of baculoviral vectors. The method may also serve as a flexible platform technology for improved use of the vectors, for example introducing a targeting ligand and minimizing immune responses.


Asunto(s)
Baculoviridae/química , Baculoviridae/genética , ADN Viral/administración & dosificación , ADN Viral/farmacocinética , Portadores de Fármacos/síntesis química , Iminas/química , Polietilenos/química , Suero/química , Transfección/métodos , Animales , Materiales Biocompatibles Revestidos/química , Vectores Genéticos/genética , Humanos , Ensayo de Materiales , Ratones , Ratones Desnudos , Ratas
6.
Anal Sci ; 25(5): 675-80, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19430152

RESUMEN

Real-time quantitative PCR (QPCR) has been proven to be a powerful tool for quantifying specific target DNA sequences. Compared to relative quantification, absolute quantification has the advantage of determining the absolute copy number of a given target, such as pathogen or plasmid DNA in vivo. However, matrix or impurities remaining in a DNA sample after various sample treatment procedures may influence a subsequent DNA analysis. In this work, we have compared methods of sample processing and validated the use of tissue genomic DNA as a universal external standard to facilitate quantification of plasmid DNA in biological matrix, especially addressing the amplification inhibition due to matrix effect and sample complexity. Also, we applied our high-throughput sample preparation and absolute quantification method to determine the distribution of an HIV plasmid DNA vaccine in vivo. Successful application showed the validity and reliability of the method in absolute quantification of a particular gene in vivo.


Asunto(s)
Vacunas contra el SIDA/farmacocinética , ADN Viral/farmacocinética , ADN/análisis , Genoma/genética , Plásmidos/genética , Reacción en Cadena de la Polimerasa/normas , Vacunas de ADN/farmacocinética , Vacunas contra el SIDA/análisis , Animales , ADN/genética , ADN Viral/análisis , Femenino , Dosificación de Gen , VIH/genética , VIH/inmunología , Modelos Lineales , Masculino , Ratones , Plásmidos/análisis , Reacción en Cadena de la Polimerasa/métodos , Estándares de Referencia , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Factores de Tiempo , Distribución Tisular , Vacunas de ADN/análisis
7.
J Biomed Nanotechnol ; 5(1): 106-14, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20055113

RESUMEN

Magnesium phosphate (MgPi) nanoparticles (NPs) encapsulating pSVbetagal and pEGFP have been used as novel non-viral vector for targeted gene delivery. These plasmid DNA loaded magnesium phosphate nanoparticles of diameter 100-130 nm were prepared in water-in-oil microemulsion. In vitro cell viability study carried out on MCF-7, HEK, and COS-7 cells demonstrated that magnesium phosphate nanoparticles have no cytotoxic effect against cell proliferation. In vivo cytotoxicity conducted on Swiss Albino mice indicated no cytotoxic effect 3 months after intraperitoneal administration of 600 mg of void magnesium phosphate nanoparticles per Kg of body weight one-time only. In vitro transfection in COS-7 cells demonstrated that magnesium phosphate nanoparticles showed approximately 100% efficiency when compared to commercial transfecting reagent Polyfect as well as the transfection efficiency of calcium phosphate (CaPi) nanoparticles recently reported. Moreover, to explore the possibility of targeting these nanoparticles to specific tissue, we have surface modified these particles by adsorbing highlyt adhesive polymer, polyacrylic acid (PAA), followed by conjugating the carboxylic groups of the polymer with p-amino-thio-beta-galactopyranoside (PAG) using a cross-linking agent 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide (EDC) and used these particles to target to liver in vivo successfully and more efficiently.


Asunto(s)
ADN Viral/genética , ADN Viral/farmacocinética , Portadores de Fármacos/química , Marcación de Gen/métodos , Vectores Genéticos/genética , Compuestos de Magnesio/química , Nanopartículas/química , Fosfatos/química , Transfección/métodos , Animales , Células COS , Chlorocebus aethiops , Cristalización/métodos , ADN Viral/administración & dosificación , Sustancias Macromoleculares/química , Ensayo de Materiales , Conformación Molecular , Propiedades de Superficie
8.
Cancer Sci ; 99(2): 385-90, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18201270

RESUMEN

The use of replication-selective tumor-specific viruses represents a novel approach for the treatment of neoplastic disease. We constructed an attenuated adenovirus, telomerase-specific replication-selective adenovirus (TRAD), in which the human telomerase reverse transcriptase promoter element drives the expression of the E1A and E1B genes linked with an internal ribosome entry site (IRES). Forty-eight hours after TRAD infection at a multiplicity of infection of 1.0, the cell viability of H1299 human lung cancer cells was consistently less than 50% and therefore this procedure could be used as a potency assay to assess the biological activity of TRAD. We also established a quantitative real-time polymerase chain reaction (PCR) analysis with consensus primers for either the adenovirus E1A or IRES sequence. The linear ranges of quantitation with E1A and IRES primers were 10(3)-10(8) and 10(2)-10(8) plaque-forming units/mL in the plasma, respectively. The PCR analysis demonstrated that the levels of E1A in normal tissues were more than 10(3) lower than in the tumors of A549 human lung tumor xenografts in nu/nmicro mice at 28 days after intratumoral injection. Our results suggest that the cell-killing assay against H1299 cells and real-time PCR can be used to assess the biological activity and biodistribution of TRAD in clinical trials.


Asunto(s)
Adenoviridae/genética , Viroterapia Oncolítica/métodos , Reacción en Cadena de la Polimerasa/métodos , Telomerasa/genética , Animales , Línea Celular , Línea Celular Tumoral , Replicación del ADN , ADN Viral/farmacocinética , Femenino , Genoma Viral , Humanos , Ratones , Ratones Endogámicos BALB C , Modelos Animales
9.
Biotechnol Bioeng ; 98(6): 1230-41, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17615558

RESUMEN

Non-invasive manipulation of multicellular systems is important for medical and biological research. The ability to introduce, remove, or modify molecules in the intracellular environment is pivotal to our understanding of cellular structure and function. Herein, we report on an alternative method for introducing foreign material into developing embryos using the application of femtosecond (fs) laser pulses. When intense fs laser pulses are focused to a sub-micron spot, transient pores are formed, providing a transport pathway for the delivery of exogenous material into embryonic cells. In this study, zebrafish embryos were used as a model system to demonstrate the non-invasiveness of this applied delivery tool. Utilizing optically induced transient pores chorionated and dechorionated zebrafish embryos were successfully loaded with a fluorescent reporter molecule (fluorescein isothiocyanate), Streptavidin-conjugated quantum dots or DNA (Simian-CMV-EGFP). Pore formation was independent of the targeted location, with both blastomere-yolk interface and blastomere pores competent for delivery. Long-term survival of laser manipulated embryos to pec-fin stage was 89% and 100% for dechorionated and chorionated embryos, respectively. To our knowledge, this is the first report of DNA delivery into zebrafish embryos utilizing fs laser pulses.


Asunto(s)
Fluoresceína-5-Isotiocianato/farmacocinética , Rayos Láser , Pez Cebra/embriología , Animales , Antígenos Virales/genética , ADN Viral/química , ADN Viral/genética , ADN Viral/farmacocinética , Femenino , Colorantes Fluorescentes/farmacocinética , Proteínas Inmediatas-Precoces/genética , Espacio Intracelular/metabolismo , Masculino , Nanotecnología/métodos , Permeabilidad , Puntos Cuánticos , Transfección/métodos , Pez Cebra/genética
10.
Hum Gene Ther ; 18(3): 245-56, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17376008

RESUMEN

To translate the potential advantages of recombinant adeno-associated virus type 1 (rAAV1) vectors into a clinical application for muscle-directed gene therapy for alpha1 -antitrypsin (AAT) deficiency, we performed safety studies in 170 C57BL/6 mice and 26 New Zealand White rabbits. A mouse toxicology study included 8 cohorts of 10 mice each (5 per sex). Mice were killed either 21 or 90 days after intramuscular injection of doses ranging up to 1x10(13)vector genomes (VG), equivalent to 4 x 10(14)VG/kg. A mouse biodistribution study was performed in 5 cohorts of 10 mice, receiving intramuscular injections at the same doses; as well as in a lower dose cohort (3 x 10(8) VG; equivalent to 1.2 x 10(10)VG/kg); and in 4 other cohorts (excluding the vehicle control) injected with identical doses intravenously. Finally, biodistribution was examined in rabbits, with serial collection of blood and semen, as well as terminal tissue collection. Two significant findings were present, both of which were dose dependent. First, inflammatory cell infiltrates were detected at the site of injection 21, 60, or 90 days after intramuscular injection of 1 x 10(13)VG. This was not associated with loss of transgene expression. Second, vector DNA sequences were detected in most animals, levels being highest with the highest doses and earliest time points. Vector DNA was also present in liver, spleen, kidneys, and a number of other organs, including the gonads of animals receiving the highest dose. Likewise, vector DNA was present in the semen of male rabbits at higher doses. The copy number of vector DNA in the blood and semen declined over time throughout the study. These two dose-dependent findings have served to guide to the design of a phase 1 human trial of rAAV1-AAT.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/toxicidad , alfa 1-Antitripsina/genética , Animales , ADN Viral/análisis , ADN Viral/farmacocinética , ADN Viral/toxicidad , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/farmacocinética , Genoma Viral , Humanos , Inflamación/inducido químicamente , Inflamación/patología , Inyecciones Intramusculares , Inyecciones Intravenosas , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/química , Músculo Esquelético/patología , Conejos , Distribución Tisular , alfa 1-Antitripsina/análisis , Deficiencia de alfa 1-Antitripsina/terapia
11.
Biol Pharm Bull ; 29(7): 1511-5, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16819202

RESUMEN

An in vitro cell culture model that mimics in vivo extracellular environment would be useful in developing in vivo gene delivery system. In the present study, a parallel flow model was applied to investigate the impact of convective flow on cellular uptake and transfection activity in endothelial cells. LipofectAMINE PLUS and adenovirus were used as model vectors, which bind cells via electrostatic- and ligand-receptor interactions, respectively. Whereas a convective flow increased the total amount of vector passing through the flow chamber by 3 orders of magnitude, uptake was increased by less than 10-fold, suggesting that the flow severely inhibited cellular uptake by reducing the retention time in the chamber and/or by diminishing the affinity between the cell and vector. Moreover, the uptake of both vectors was increased in a shear stress-dependent manner to a comparable extent, suggesting that the effect of flow on the cellular uptake was not significant. In contrast, transfection efficiency (TE), expressed as the transfection activity normalized by the cellular uptake of vectors was dramatically stimulated by shear stress, only when LipofectAMINE PLUS was used. Since the activities of the CMV promoter were unaffected by a shear stress, it is possible that altered intracellular trafficking may responsible for the improvement in lipoplex-mediated TE, presumably related to the cellular uptake pathway.


Asunto(s)
Adenoviridae/fisiología , Encéfalo/virología , ADN Viral/farmacocinética , Células Endoteliales/virología , Animales , Portadores de Fármacos , Lípidos/farmacocinética , Ratones , Transfección , Virología/instrumentación , Virología/métodos
12.
Hum Gene Ther ; 17(7): 705-16, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16839270

RESUMEN

The recombinant adenoviral (Ad) vector is being considered as a cancer vaccine platform because it efficiently induces immune responses to tumor antigens by intradermal immunization. The aims of this study were to evaluate the potential toxicities and biodistribution after a single dose or six weekly intradermal doses of Ad2/gp100v2 and Ad2/MART-1v2, which encode tumor-associated antigens gp100 and MelanA/MART-1, respectively. The only dose-related toxicities associated with intradermal administration of these Ad vectors were inflammatory cell infiltrates in the draining lymph nodes and injection sites that persisted 83 days after administration. The biodistribution of Ad DNA as detected by real-time polymerase chain reaction was largely confined to the injection sites and draining lymph nodes of mice treated with either a single dose or multiple doses of Ad vector and in the spleens of mice treated with multiple doses of Ad vector. Adenoviral DNA was transiently detected in the bone marrow, lung, or blood of only one animal for each site and was below the limit of assay quantification (<10 copies/microg DNA). The vector persisted in the skin and lymph nodes as long as 92 days after the last dose. We conclude that Ad vectors delivered by intradermal administration provide a safe, genetic vaccine delivery platform that induces desirable immune responses at the immunization sites and the lymph nodes that, ultimately, result in immune responses specific to the tumor antigens.


Asunto(s)
Adenoviridae/genética , Vacunas contra el Cáncer/administración & dosificación , Terapia Genética , Glicoproteínas de Membrana/genética , Proteínas de Neoplasias/genética , Animales , Antígenos de Neoplasias , Vacunas contra el Cáncer/farmacocinética , Vacunas contra el Cáncer/toxicidad , ADN Viral/farmacocinética , ADN Viral/toxicidad , Femenino , Vectores Genéticos/administración & dosificación , Inyecciones Intradérmicas , Antígeno MART-1 , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa , Distribución Tisular , Antígeno gp100 del Melanoma
13.
Ann Biomed Eng ; 33(2): 202-8, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15771273

RESUMEN

The efficiency of gene therapy as a pretreatment for saphenous vein coronary artery bypass grafts can be improved by increasing the transport of vector into the tunica media. The purpose of this study was to determine the effect of increasing transmural pressure on vector delivery depth in human saphenous vein segments. Specifically, we introduced adenovirus-sized microspheres luminally to observe changes in transport efficiency into the intimal and medial layers with increasing pressure. Our results indicate that transmural pressures of 100 and 400 mmHg increase the intimal concentration of microspheres as compared to 0 mmHg (p < 0.03), but do not significantly affect medial concentrations. We did not find increasing concentrations with increasing pressure above 100 mmHg. These results suggest that low or intermediate transmural pressures are adequate for intimal vector delivery and that techniques other than increasing pressure are required to deliver gene therapy vectors (> or = 100 nm) to medial smooth muscle cells. Also, our data support previous models designating the internal elastic lamina as the primary barrier to particle transport. Finally, our ex vivo microsphere perfusion experiment represents a novel way to explore functional vein permeabilities to gene therapy vectors and, ultimately, optimize vascular gene therapy protocols.


Asunto(s)
Adenoviridae/genética , ADN Viral/administración & dosificación , ADN Viral/farmacocinética , Terapia Genética/métodos , Vena Safena/metabolismo , Transfección/métodos , Transporte Biológico Activo/fisiología , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Vectores Genéticos/farmacocinética , Humanos , Técnicas In Vitro , Microesferas , Presión
14.
Blood ; 102(7): 2412-9, 2003 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-12791653

RESUMEN

We report the generation and use of pseudotyped adeno-associated viral (AAV) vectors for the liver-specific expression of human blood coagulation factor IX (hFIX). Therefore, an AAV-2 genome encoding the hfIX gene was cross-packaged into capsids of AAV types 1 to 6 using efficient, large-scale technology for particle production and purification. In immunocompetent mice, the resultant vector particles expressed high hFIX levels ranging from 36% (AAV-4) to more than 2000% of normal (AAV-1, -2, and -6), which would exceed curative levels in patients with hemophilia. Expression was dose- and time-dependent, with AAV-6 directing the fastest and strongest onset of hFIX expression at all doses. Interestingly, systemic administration of 2 x 1012 vector particles of AAV-1, -4, or -6 resulted in hFIX levels similar to those achieved by portal vein delivery. For all other serotypes and particle doses, hepatic vector administration yielded up to 84-fold more hFIX protein than tail vein delivery, corroborated by similarly increased vector DNA copy numbers in the liver, and elicited a reduced immune response against the viral capsids. Finally, neutralization assays showed variable immunologic cross-reactions between most of the AAV serotypes. Our technology and findings should facilitate the development of AAV pseudotype-based gene therapies for hemophilia B and other liver-related diseases.


Asunto(s)
Adenoviridae/genética , Factor IX/genética , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Hemofilia B/terapia , Hepatopatías/terapia , Animales , Anticuerpos/sangre , Cápside , Carcinoma Hepatocelular , ADN Viral/farmacocinética , Dosificación de Gen , Regulación Viral de la Expresión Génica , Vectores Genéticos/inmunología , Vectores Genéticos/farmacocinética , Humanos , Riñón/citología , Neoplasias Hepáticas , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Distribución Tisular , Células Tumorales Cultivadas
15.
Asian J Androl ; 4(3): 169-73, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12364970

RESUMEN

AIM: To pretreat sperm at various temperatures before exposure to human papillomavirus (HPV) 16 DNA fragments and to assess the efficiency of HPV carrier sperm to transfect cumulus cells. METHODS: Cumulus cells from follicular aspirates were obtained, pooled and divided into culture dishes containing Sybr Gold-stained HPV DNA carrying sperm that were either pretreated at 4 degree C, 37 degree C or 40 degree C (n = 5). The cells were incubated in 5% CO(2) in air mixture at 37 degree C for 24 hours. The efficiency of sperm to take up fluorescent HPV DNA was determined at hour 0. After incubation, cumulus cell viability was assessed using the eosin method and the percentages of fluorescent cumulus cells determined. RESULTS: Over half of all the cumulus cells became fluorescent with the highest percentage in the 37 degree C group. Sperm pretreated at 4 degree C had the greatest amount of HPV DNA fragments. Total sperm motility was similar for the 3 pretreatment groups. There were no differences in cumulus viability among the groups. CONCLUSION: Sperm pretreated at 37 degree C transferred the greatest amount of fluorescent HPV DNA fragments to the cumulus cells. The HPV DNA was observed in the nuclear and cytoplasmic compartments. The data suggested the possibility of sperm as a vector for the transmission of HPV DNA to the cumulus cells surrounding ovulated oocytes, which might lead to early implantation failures.


Asunto(s)
Papillomaviridae/genética , Espermatozoides/fisiología , Transfección/métodos , ADN Viral/farmacocinética , Humanos , Masculino , Temperatura
16.
Nat Cell Biol ; 3(12): 1092-100, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11781571

RESUMEN

Adenovirus type 2 (Ad2) imports its DNA genome through the nuclear pore complex (NPC) of cells in interphase for viral production. Here we identify the NPC-filament protein CAN/Nup214 as a docking site for incoming Ad2 capsids. Binding to CAN is independent of cytosolic factors. Capsids disassemble at NPCs to free their DNA for import. This process requires binding of nuclear histone H1 to the stably docked capsids and involves H1-import factors, restricting this irreversible process to the proximity of the nucleus. Our results provide a molecular mechanism for disassembly of Ad2 and reveal an unexpected function of histone H1 in virus-mediated DNA import.


Asunto(s)
Adenoviridae/genética , Proteínas de la Cápside , ADN Viral/farmacocinética , Histonas/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Poro Nuclear/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Secuencia de Aminoácidos , Anticuerpos/farmacología , Cápside/genética , Cápside/inmunología , Cápside/metabolismo , Histonas/inmunología , Humanos , Neoplasias Pulmonares , Datos de Secuencia Molecular , Proteínas de Complejo Poro Nuclear/inmunología , Unión Proteica/fisiología , Células Tumorales Cultivadas , alfa Carioferinas/metabolismo , beta Carioferinas/metabolismo
17.
Pharm Res ; 16(4): 503-8, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10227703

RESUMEN

PURPOSE: Plasmid DNA (pDNA) has become an important class of macromolecular agent suitable for non-viral gene therapy as well as DNA vaccination. Our recent study has suggested that pDNA is taken up by mouse peritoneal macrophages via a specific mechanism mediated by a receptor similar to the scavenger receptor (SR). This study was designed to further characterize the pDNA uptake by macrophages in order to elucidate the mechanism. METHODS: The binding and uptake of pDNA labeled with 32P or a fluorescent marker were studied in vitro using cultured Chinese hamster ovary (CHO) cells expressing the class A scavenger receptor (SRA) and peritoneal macrophages from SRA-knockout mice. RESULTS: pDNA binding and uptake by CHO(SRA) cells were minimal and almost identical to that by wild-type CHO cells. Macrophages from the knockout mice showed pronounced pDNA binding and uptake as did the control macrophages. In both types of macrophage, pDNA binding was significantly inhibited by cold pDNA, polyinosinic acid and dextran sulfate but not by polycytidylic acid or Ac-LDL. These results provide direct evidence that SRA is not responsible for the significant binding and subsequent uptake of pDNA by mouse peritoneal macrophages. Further binding experiments revealed that, in addition to polyinosinic acid and dextran sulfate, heparin was a potent inhibitor among a variety of polyanionic compounds such as polynucleotides, anionic polysaccharides and modified proteins including Ox-LDL. CONCLUSIONS: The present study suggest that pDNA binding and uptake by mouse peritoneal macrophages are mediated by a specific mechanism to some defined polyanions not by scavenger receptors. The finding would be an important basis for further studies to elucidate the mechanism(s) of pDNA uptake by macrophages.


Asunto(s)
ADN Viral/farmacocinética , Macrófagos Peritoneales/metabolismo , Plásmidos/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Animales , Sitios de Unión/genética , Células CHO , Células Cultivadas , Cricetinae , Citomegalovirus/genética , Colorantes Fluorescentes , Ratones , Ratones Noqueados , Radioisótopos de Fósforo , Plásmidos/genética , Receptores Inmunológicos/deficiencia , Receptores Depuradores , Receptores Depuradores de Clase A
18.
Mol Gen Genet ; 259(6): 569-76, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9819049

RESUMEN

We have previously shown that, when administered orally to mice, bacteriophage M13 DNA, as a paradigm foreign DNA without homology to the mouse genome, can persist in fragmented form in the gastrointestinal tract, penetrate the intestinal wall, and reach the nuclei of leukocytes, spleen and liver cells. Similar results were obtained when a plasmid containing the gene for the green fluorescent protein (pEGFP-C1) was fed to mice. In spleen, the foreign DNA was detected in covalent linkage to DNA with a high degree of homology to mouse genes, perhaps pseudogenes, or to authentic E. coli DNA. We have now extended these studies to the offspring of mice that were fed regularly during pregnancy with a daily dose of 50 microg of M13 or pEGFP-C1 DNA. Using the polymerase chain reaction (PCR) or the fluorescent in situ hybridization (FISH) method, foreign DNA, orally ingested by pregnant mice, can be discovered in various organs of fetuses and of newborn animals. The M13 DNA fragments have a length of about 830 bp. In various organs of the mouse fetus, clusters of cells contain foreign DNA as revealed by FISH. The foreign DNA is invariably located in the nuclei. We have never found all cells of the fetus to be transgenic for the foreign DNA. This distribution pattern argues for a transplacental pathway rather than for germline transmission which might be expected only after long-time feeding regimens. In rare cells of three different fetuses, whose mothers have been fed with M 13 DNA during gestation, the foreign DNA was detected by FISH in association with both chromatids. Is maternally ingested foreign DNA a potential mutagen for the developing fetus?


Asunto(s)
ADN/farmacocinética , Intercambio Materno-Fetal , Administración Oral , Animales , Animales Recién Nacidos , Bacteriófago M13/genética , ADN/administración & dosificación , ADN Viral/administración & dosificación , ADN Viral/farmacocinética , Femenino , Contenido Digestivo , Proteínas Fluorescentes Verdes , Hibridación Fluorescente in Situ , Riñón/metabolismo , Hígado/metabolismo , Proteínas Luminiscentes/genética , Ratones , Ratones Endogámicos C57BL , Plásmidos/administración & dosificación , Plásmidos/farmacocinética , Embarazo , Bazo/metabolismo , Distribución Tisular
19.
Wien Klin Wochenschr ; 110(2): 40-4, 1998 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-9531678

RESUMEN

Foreign DNA (deoxyribonucleic acid) is part of our environment. Considerable amounts of foreign DNA of very different origin are ingested daily with food. In a series of experiments we fed the DNA of bacteriophage M13 as test DNA to mice and showed that fragments of this DNA survive the passage through the gastrointestinal (GI) tract in small amounts (1-2%). Food-ingested M13 DNA reaches peripheral white blood cells, the spleen and liver via the intestinal epithelia and cells in the Peyer's patches of the intestinal wall. There is evidence to assume that food-ingested foreign DNA can become covalently linked to mouse-like DNA. When M13 DNA is fed to pregnant mice the test DNA can be detected in cells in various organs of the fetuses and of newborn animals, but never in all cells of the mouse fetus. It is likely that the M13 DNA is transferred by the transplacental route and not via the germ line. The consequences of foreign DNA uptake for mutagenesis and oncogenesis have not yet been investigated.


Asunto(s)
ADN/farmacocinética , Fenómenos Fisiológicos del Sistema Digestivo , Exposición a Riesgos Ambientales , Placenta/fisiología , Animales , Bacteriófago M13 , ADN/efectos adversos , ADN Viral/farmacocinética , Femenino , Humanos , Absorción Intestinal , Intercambio Materno-Fetal , Ratones , Embarazo
20.
Folia Biol (Praha) ; 43(3): 123-7, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9338119

RESUMEN

Xenopus laevis spermatozoa and mouse epididymal spermatozoa were compared in their ability to bind plasmid DNA. Spermatozoa of both species are endowed with a very similar binding capacity for plasmid pAPrC DNA carrying the complete Rous sarcoma virus (RSV) DNA. Our experiments failed to detect any substantial differences between both types of sperm cells in the kinetics of DNA binding, maximum DNA binding capacity, effect of various ions, metabolic inhibitors and substrates on DNA binding, etc. Each X. laevis and mouse sperm cell associates, on an average, with about 50 and 45 molecules, respectively, of the plasmid DNA in a DNase resistant form, if spermatozoa were exposed to the DNA at a concentration of 0.5 microgram/5 x 10(6) sperm cells. The uptake of the DNA by both types of sperm cells is strongly inhibited by heparin. The 37-kDa factor IF-1 shown by Zani et al. (1995) to specifically block DNA binding to mouse sperm cells inhibited the interaction between pAPrC DNA and frog spermatozoa with a similar intensity.


Asunto(s)
Virus del Sarcoma Aviar/genética , ADN Viral/farmacocinética , Plásmidos/farmacocinética , Espermatozoides/fisiología , Xenopus laevis/fisiología , Animales , ADN Viral/metabolismo , Filtración , Masculino , Ratones , Ratones Endogámicos BALB C , Plásmidos/metabolismo , Espermatozoides/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...