Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Gen Virol ; 94(Pt 2): 284-292, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23100360

RESUMEN

European bat lyssaviruses type 1 (EBLV-1) and type 2 (EBLV-2) circulate within bat populations throughout Europe and are capable of causing disease indistinguishable from that caused by classical rabies virus (RABV). However, the determinants of viral fitness and pathogenicity are poorly understood. Full-length genome clones based on the highly attenuated, non-neuroinvasive, RABV vaccine strain (SAD-B19) were constructed with the glycoprotein (G) of either SAD-B19 (SN), of EBLV-1 (SN-1) or EBLV-2 (SN-2). In vitro characterization of SN-1 and SN-2 in comparison to wild-type EBLVs demonstrated that the substitution of G affected the final virus titre and antigenicity. In vivo, following peripheral infection with a high viral dose (10(4) f.f.u.), animals infected with SN-1 had reduced survivorship relative to infection with SN, resulting in survivorship similar to animals infected with EBLV-1. The histopathological changes and antigen distribution observed for SN-1 were more representative of those observed with SN than with EBLV-1. EBLV-2 was unable to achieve a titre equivalent to that of the other viruses. Therefore, a reduced-dose experiment (10(3) f.f.u.) was undertaken in vivo to compare EBLV-2 and SN-2, which resulted in 100 % survivorship for all recombinant viruses (SN, SN-1 and SN-2) while clinical disease developed in mice infected with the EBLVs. These data indicate that interspecies replacement of G has an effect on virus titre in vitro, probably as a result of suboptimal G-matrix protein interactions, and influences the survival outcome following a peripheral challenge with a high virus titre in mice.


Asunto(s)
Glicoproteínas/metabolismo , Lyssavirus/genética , Lyssavirus/patogenicidad , Proteínas Virales/metabolismo , Factores de Virulencia/metabolismo , Animales , Antígenos Virales/genética , Antígenos Virales/inmunología , Antígenos Virales/metabolismo , Encéfalo/patología , Encéfalo/virología , Modelos Animales de Enfermedad , Glicoproteínas/genética , Glicoproteínas/inmunología , Histocitoquímica , Inmunohistoquímica , Lyssavirus/inmunología , Ratones , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Recombinación Genética , Infecciones por Rhabdoviridae/patología , Infecciones por Rhabdoviridae/virología , Análisis de Supervivencia , Carga Viral , Proteínas Virales/genética , Proteínas Virales/inmunología , Factores de Virulencia/genética , Factores de Virulencia/inmunología
2.
RNA ; 16(12): 2553-63, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20940339

RESUMEN

Effective small interfering RNA (siRNA)-mediated therapeutics require the siRNA to be delivered into the cellular RNA-induced silencing complex (RISC). Quantitative information of this essential delivery step is currently inferred from the efficacy of gene silencing and siRNA uptake in the tissue. Here we report an approach to directly quantify siRNA in the RISC in rodents and monkey. This is achieved by specific immunoprecipitation of the RISC from tissue lysates and quantification of small RNAs in the immunoprecipitates by stem-loop PCR. The method, expected to be independent of delivery vehicle and target, is label-free, and the throughput is acceptable for preclinical animal studies. We characterized a lipid-formulated siRNA by integrating these approaches and obtained a quantitative perspective on siRNA tissue accumulation, RISC loading, and gene silencing. The described methodologies have utility for the study of silencing mechanism, the development of siRNA therapeutics, and clinical trial design.


Asunto(s)
Técnicas de Transferencia de Gen , ARN Interferente Pequeño/genética , Animales , Animales Modificados Genéticamente , Anticuerpos/aislamiento & purificación , Anticuerpos/metabolismo , Anticuerpos/farmacología , Especificidad de Anticuerpos , Proteínas Argonautas , Células Cultivadas , Factor 2 Eucariótico de Iniciación/inmunología , Factor 2 Eucariótico de Iniciación/metabolismo , Estudios de Evaluación como Asunto , Femenino , Silenciador del Gen/fisiología , Marcación de Gen/métodos , Técnicas de Transferencia de Gen/normas , Humanos , Inmunoprecipitación/métodos , Inmunoprecipitación/normas , Macaca mulatta , Ratones , Ratones Endogámicos ICR , Unión Proteica , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Roedores
3.
Mol Ther ; 18(1): 171-80, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19738601

RESUMEN

Despite recent progress, systemic delivery remains the major hurdle for development of safe and effective small inhibitory RNA (siRNA)-based therapeutics. Encapsulation of siRNA into liposomes is a promising option to overcome obstacles such as low stability in serum and inefficient internalization by target cells. However, a major liability of liposomes is the potential to induce an acute inflammatory response, thereby increasing the risk of numerous adverse effects. In this study, we characterized a liposomal siRNA delivery vehicle, LNP201, which is capable of silencing an mRNA target in mouse liver by over 80%. The biodistribution profile, efficacy after single and multiple doses, mechanism of action, and inflammatory toxicity are characterized for LNP201. Furthermore, we demonstrate that the glucocorticoid receptor (GR) agonist dexamethasone (Dex) inhibits LNP201-induced cytokine release, inflammatory gene induction, and mitogen-activated protein kinase (MAPK) phosphorylation in multiple tissues. These data present a possible clinical strategy for increasing the safety profile of siRNA-based drugs while maintaining the potency of gene silencing.


Asunto(s)
Dexametasona/uso terapéutico , Nanopartículas/efectos adversos , ARN Interferente Pequeño/inmunología , ARN Interferente Pequeño/metabolismo , Animales , Femenino , Silenciador del Gen , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Ratones , Nanopartículas/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Receptores de Glucocorticoides/agonistas
4.
Mol Cancer Ther ; 17(2): 544-553, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29282298

RESUMEN

Colorectal carcinomas harbor well-defined genetic abnormalities, including aberrant activation of Wnt/ß-catenin and MAPK pathways, often simultaneously. Although the MAPK pathway can be targeted using potent small-molecule drugs, including BRAF and MEK inhibitors, ß-catenin inhibition has been historically challenging. RNAi approaches have advanced to the stage of clinical viability and are especially well suited for transcriptional modulators, such as ß-catenin. In this study, we report therapeutic effects of combined targeting of these pathways with pharmacologic agents. Using a recently described tumor-selective nanoparticle containing a ß-catenin-targeting RNAi trigger, in combination with the FDA-approved MEK inhibitor (MEKi) trametinib, we demonstrate synergistic tumor growth inhibition in in vivo models of colorectal cancer, melanoma, and hepatocellular carcinoma. At dose levels that were insufficient to significantly impact tumor growth as monotherapies, combination regimens resulted in synergistic efficacy and complete tumor growth inhibition. Importantly, dual MEKi/RNAi therapy dramatically improved survival of mice bearing colorectal cancer liver metastases. In addition, pharmacologic silencing of ß-catenin mRNA was effective against tumors that are inherently resistant or that acquire drug-induced resistance to trametinib. These results provide a strong rationale for clinical evaluation of this dual-targeting approach for cancers harboring Wnt/ß-catenin and MAPK pathway mutations. Mol Cancer Ther; 17(2); 544-53. ©2017 AACR.


Asunto(s)
Neoplasias Colorrectales/terapia , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Piridonas/farmacología , Pirimidinonas/farmacología , ARN Mensajero/genética , ARN Interferente Pequeño/administración & dosificación , beta Catenina/genética , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Sinergismo Farmacológico , Silenciador del Gen , Xenoinjertos , Humanos , Neoplasias Hepáticas Experimentales/secundario , Quinasas Quinasa Quinasa PAM/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Desnudos , Nanopartículas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo
5.
Mol Cancer Ther ; 15(9): 2143-54, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27390343

RESUMEN

The Wnt/ß-catenin pathway is among the most frequently altered signaling networks in human cancers. Despite decades of preclinical and clinical research, efficient therapeutic targeting of Wnt/ß-catenin has been elusive. RNA interference (RNAi) technology silences genes at the mRNA level and therefore can be applied to previously undruggable targets. Lipid nanoparticles (LNP) represent an elegant solution for the delivery of RNAi-triggering oligonucleotides to disease-relevant tissues, but have been mostly restricted to applications in the liver. In this study, we systematically tuned the composition of a prototype LNP to enable tumor-selective delivery of a Dicer-substrate siRNA (DsiRNA) targeting CTNNB1, the gene encoding ß-catenin. This formulation, termed EnCore-R, demonstrated pharmacodynamic activity in subcutaneous human tumor xenografts, orthotopic patient-derived xenograft (PDX) tumors, disseminated hematopoietic tumors, genetically induced primary liver tumors, metastatic colorectal tumors, and murine metastatic melanoma. DsiRNA delivery was homogeneous in tumor sections, selective over normal liver and independent of apolipoprotein-E binding. Significant tumor growth inhibition was achieved in Wnt-dependent colorectal and hepatocellular carcinoma models, but not in Wnt-independent tumors. Finally, no evidence of accelerated blood clearance or sustained liver transaminase elevation was observed after repeated dosing in nonhuman primates. These data support further investigation to gain mechanistic insight, optimize dose regimens, and identify efficacious combinations with standard-of-care therapeutics. Mol Cancer Ther; 15(9); 2143-54. ©2016 AACR.


Asunto(s)
Neoplasias/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , beta Catenina/genética , Animales , Apolipoproteínas E/química , Apolipoproteínas E/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Lípidos/química , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Melanoma Experimental , Ratones , Nanopartículas/química , Metástasis de la Neoplasia , Neoplasias/metabolismo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/química , Relación Estructura-Actividad , Vía de Señalización Wnt , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
6.
J Infect Dis ; 195(7): 980-8, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17330788

RESUMEN

We analyzed the safety and immunogenicity of attenuated rabies virus vectors expressing simian-human immunodeficiency virus (SHIV)-1(89.6P) Env or simian immunodeficiency virus (SIV)(mac239) Gag in rhesus macaques. Four test macaques were immunized with both vaccine constructs, and 2 control macaques received an empty rabies vector. Seroconversion against rabies virus glycoprotein (G) and SHIV(89.6P) Env was detected after the initial immunization, but no cellular responses against SHIV antigens were observed. HIV/SIV-specific immune responses were not enhanced by boosts with the same vectors. Therefore, we constructed vectors expressing SHIV(89.6P) Env and SIV(mac239) Gag in which the rabies G was replaced with the G protein of vesicular stomatitis virus (VSV). Two years after initial immunization, a boost with the rabies-VSV G vectors resulted in SIV/HIV-specific immune responses. Upon challenge with SHIV(89.6P) test macaques controlled the infection, whereas control macaques had high levels of viremia and a profound loss of CD4(+) T cells, with 1 control macaque dying of an AIDS-like disease.


Asunto(s)
Productos del Gen env/inmunología , Productos del Gen gag/inmunología , Vacunas contra el SIDAS/uso terapéutico , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Vacunas Sintéticas/uso terapéutico , Animales , Cartilla de ADN , Productos del Gen env/genética , Productos del Gen gag/genética , Vectores Genéticos , Macaca mulatta , Masculino , ARN Viral/análisis , Virus de la Rabia/genética , Virus de la Inmunodeficiencia de los Simios/genética
7.
Virology ; 344(2): 363-77, 2006 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-16226782

RESUMEN

Recombinant rabies virus (RV) vaccine strain-based vectors expressing HIV-1 antigens have been shown to induce strong and long-lasting cellular but modest humoral responses against the expressed antigens in mice. However, an effective vaccine against HIV-1 may require stronger responses, and the development of such an immune response may depend on the presence of certain cytokines at the time of the inoculation. Here, we describe several new RV-based vaccine vehicles expressing HIV-1 Gag or envelope (Env) and murine IL-2 or IL-4. Cells infected with recombinant RVs expressed high levels of functional IL-2 or IL-4 in culture supernatants in addition to HIV-1 proteins. The recombinant RV expressing IL-4 was highly attenuated in a cytokine-independent manner, indicating that the insertion of two foreign genes into the RV genome is mainly responsible for the attenuation observed. The expression of IL-4 resulted in a decrease in the cellular immune response against HIV-1 Gag and Env when compared with the parental virus not expressing IL-4 and only 2 of 20 mice seroconverted to HIV-1 Env after two inoculations. The IL-2-expressing RV was completely apathogenic after direct intracranial inoculation of mice. In addition, mice immunized with IL-2 maintained strong anti-HIV-1 Gag and Env cellular responses and consistently induced seroconversion against HIV-1 Env after two inoculations. This suggests the potential use of IL-2 in RV-based HIV-1 vaccine strategies, which may require the induction of both arms of the immune response.


Asunto(s)
Vacunas contra el SIDA/inmunología , Antígenos VIH/inmunología , VIH-1/inmunología , Interleucina-2/inmunología , Rhabdoviridae/genética , Vacunas contra el SIDA/genética , Animales , Línea Celular , Proliferación Celular , Femenino , Productos del Gen env/genética , Productos del Gen env/inmunología , Productos del Gen gag/genética , Productos del Gen gag/inmunología , Vectores Genéticos/genética , Antígenos VIH/genética , VIH-1/genética , Interleucina-2/genética , Interleucina-4/inmunología , Ratones , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
8.
Virology ; 331(1): 82-93, 2005 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-15582655

RESUMEN

Recombinant rhabdovirus vectors expressing human immunodeficiency virus (HIV) and/or simian immunodeficiency virus (SIV) proteins have been shown to induce strong immune responses in mice and rhesus macaques. However, the finding that such responses protect rhesus macaques from AIDS-like disease but not from infection indicates that further improvements for these vectors are needed. Here, we designed a prime-boost schedule consisting of a rabies virus (RV) vaccine strain and a recombinant vesicular stomatitis virus (VSV) both expressing HIV Envelope (Env). Mice were primed and boosted with the two vaccine vehicles by different routes and in different combinations. Mucosal and systemic humoral responses were assessed using enzyme linked immunosorbent assay (ELISA) while the cellular immune response was determined by an IFN-gamma ELISPOT assay. We found that an immunization combination of RV and VSV elicited the highest titers of anti-Env antibodies and the greatest amount of Env-specific IFN-gamma secreting cells pre- and post-challenge with a recombinant vaccinia virus expressing HIV(89.6) Env. Furthermore, intramuscular immunization did not induce antigen-specific mucosal antibodies while intranasal inoculation stimulated vector-specific IgA antibodies in vaginal washings and serum. Our results show that it is feasible to elicit robust cellular and humoral anti-HIV responses using two different live attenuated Rhabdovirus vectors to sequentially prime and boost.


Asunto(s)
Productos del Gen env/inmunología , VIH-1/inmunología , Rhabdoviridae/fisiología , Vacunas contra el SIDA/inmunología , Animales , Ensayo de Inmunoadsorción Enzimática , Femenino , Vectores Genéticos/fisiología , Anticuerpos Anti-VIH/biosíntesis , Inmunidad Celular , Inmunidad Mucosa , Esquemas de Inmunización , Interferón gamma/sangre , Ratones , Ratones Endogámicos BALB C , Virus de la Rabia/inmunología , Vacunas Sintéticas/inmunología , Virus de la Estomatitis Vesicular Indiana/fisiología
9.
Proc Natl Acad Sci U S A ; 101(25): 9405-10, 2004 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-15197258

RESUMEN

Rabies virus (RV) nucleoprotein (N) tightly encapsidates the genomic and antigenomic RNA of RV to form the viral ribonucleoprotein (RNP) complex. Antigens, such as N, presented in a highly organized structure are sufficient and even desirable to activate B cells to proliferate and produce antibodies. In addition to activating B cells to proliferate, it has been shown that RV N in the RNP complex induces potent T helper cell responses resulting in long-lasting and strong humoral immune responses against RV. The possibility to systematically incorporate foreign genes into the genome of RV and produce a recombinant virus allows us to examine whether the immunogenicity of foreign antigens can be enhanced by incorporation into the RV RNP structure. To test this hypothesis we constructed a recombinant RV expressing a RV N-GFP fusion protein. The chimeric N-GFP fusion protein was efficiently expressed and incorporated into RV RNP and virions. Moreover, the recombinant RNP induces a strong humoral immune response against GFP in mice. In contrast, mice inoculated with GFP alone or a combination of wild-type RV RNPs and GFP did not trigger any GFP-specific humoral responses using the same immunization schedule. These data indicate the usefulness of RV-based vectors as killed vaccines against other infectious diseases.


Asunto(s)
Antígenos Virales/fisiología , Virus de la Rabia/fisiología , Ribonucleoproteínas/fisiología , Proteínas Virales/fisiología , Animales , Secuencia de Bases , Linfocitos T CD4-Positivos/inmunología , Cartilla de ADN , Femenino , Genes Reporteros , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa , Virus de la Rabia/genética , Virus de la Rabia/inmunología , Proteínas Recombinantes de Fusión/inmunología , Recombinación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA