Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Arch Virol ; 165(3): 671-681, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31942645

RESUMEN

Dengue virus (DENV) is the most common mosquito-borne viral disease. The World Health Organization estimates that 400 million new cases of dengue fever occur every year. Approximately 500,000 individuals develop severe and life-threatening complications from dengue fever, such as dengue shock syndrome (DSS) and dengue hemorrhagic fever (DHF), which cause 22,000 deaths yearly. Currently, there are no specific licensed therapeutics to treat DENV illness. We have previously shown that the MEK/ERK inhibitor U0126 inhibits the replication of the flavivirus yellow fever virus. In this study, we demonstrate that the MEK/ERK inhibitor AZD6244 has potent antiviral efficacy in vitro against DENV-2, DENV-3, and Saint Louis encephalitis virus (SLEV). We also show that it is able to protect AG129 mice from a lethal challenge with DENV-2 (D2S20). The molecule is currently undergoing phase III clinical trials for the treatment of non-small-cell lung cancer. The effect of AZD6244 on the DENV life cycle was attributed to a blockade of morphogenesis. Treatment of AG129 mice twice daily with oral doses of AZD6244 (100 mg/kg/day) prevented the animals from contracting dengue hemorrhagic fever (DHF)-like lethal disease upon intravenous infection with 1 × 105 PFU of D2S20. The effectiveness of AZD6244 was observed even when the treatment of infected animals was initiated 1-2 days postinfection. This was also followed by a reduction in viral copy number in both the serum and the spleen. There was also an increase in IL-1ß and TNF-α levels in mice that were infected with D2S20 and treated with AZD6244 in comparison to infected mice that were treated with the vehicle only. These data demonstrate the potential of AZD6244 as a new therapeutic agent to treat DENV infection and possibly other flavivirus diseases.


Asunto(s)
Antivirales/uso terapéutico , Bencimidazoles/uso terapéutico , Virus del Dengue/crecimiento & desarrollo , Dengue Grave/prevención & control , Animales , Línea Celular , Cricetinae , Virus del Dengue/efectos de los fármacos , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Interleucina-1beta/sangre , Ratones , Dengue Grave/virología , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/sangre
2.
PLoS Pathog ; 11(9): e1005148, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26325270

RESUMEN

Vaccinia virus A33 is an extracellular enveloped virus (EEV)-specific type II membrane glycoprotein that is essential for efficient EEV formation and long-range viral spread within the host. A33 is a target for neutralizing antibody responses against EEV. In this study, we produced seven murine anti-A33 monoclonal antibodies (MAbs) by immunizing mice with live VACV, followed by boosting with the soluble A33 homodimeric ectodomain. Five A33 specific MAbs were capable of neutralizing EEV in the presence of complement. All MAbs bind to conformational epitopes on A33 but not to linear peptides. To identify the epitopes, we have adetermined the crystal structures of three representative neutralizing MAbs in complex with A33. We have further determined the binding kinetics for each of the three antibodies to wild-type A33, as well as to engineered A33 that contained single alanine substitutions within the epitopes of the three crystallized antibodies. While the Fab of both MAbs A2C7 and A20G2 binds to a single A33 subunit, the Fab from MAb A27D7 binds to both A33 subunits simultaneously. A27D7 binding is resistant to single alanine substitutions within the A33 epitope. A27D7 also demonstrated high-affinity binding with recombinant A33 protein that mimics other orthopoxvirus strains in the A27D7 epitope, such as ectromelia, monkeypox, and cowpox virus, suggesting that A27D7 is a potent cross-neutralizer. Finally, we confirmed that A27D7 protects mice against a lethal challenge with ectromelia virus.


Asunto(s)
Anticuerpos Neutralizantes/metabolismo , Glicoproteínas de Membrana/antagonistas & inhibidores , Modelos Moleculares , Orthopoxvirus/fisiología , Infecciones por Poxviridae/virología , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Tropismo Viral , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/genética , Anticuerpos Neutralizantes/uso terapéutico , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Complejo Antígeno-Anticuerpo/química , Complejo Antígeno-Anticuerpo/genética , Complejo Antígeno-Anticuerpo/metabolismo , Chlorocebus aethiops , Femenino , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos BALB C , Mutación , Orthopoxvirus/inmunología , Infecciones por Poxviridae/inmunología , Infecciones por Poxviridae/prevención & control , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico , Vacunas Sintéticas/química , Vacunas Sintéticas/genética , Vacunas Sintéticas/metabolismo , Vacunas Sintéticas/uso terapéutico , Células Vero , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Vacunas Virales/química , Vacunas Virales/genética , Vacunas Virales/metabolismo , Vacunas Virales/uso terapéutico
3.
J Virol ; 87(17): 9610-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23804638

RESUMEN

Adenoviruses (Ads) are promising vectors for therapeutic interventions in humans. When injected into the bloodstream, Ad vectors can bind several vitamin K-dependent blood coagulation factors, which contributes to virus sequestration in the liver by facilitating transduction of hepatocytes. Although both coagulation factors FVII and FX bind the hexon protein of human Ad serotype 5 (HAdv5) with a very high affinity, only FX appears to play a role in mediating Ad-hepatocyte transduction in vivo. To understand the discrepancy between efficacy of FVII binding to hexon and its apparently poor capacity for supporting virus cell entry, we analyzed the HAdv5-FVII complex by using high-resolution cryo-electron microscopy (cryo-EM) followed by molecular dynamic flexible fitting (MDFF) simulations. The results indicate that although hexon amino acids T423, E424, and T425, identified earlier as critical for FX binding, are also involved in mediating binding of FVII, the FVII GLA domain sits within the surface-exposed hexon trimer depression in a different orientation from that found for FX. Furthermore, we found that when bound to hexon, two proximal FVII molecules interact via their serine protease (SP) domains and bury potential heparan sulfate proteoglycan (HSPG) receptor binding residues within the dimer interface. In contrast, earlier cryo-EM studies of the Ad-FX interaction showed no evidence of dimer formation. Dimerization of FVII bound to Ad may be a contributing mechanistic factor for the differential infectivity of Ad-FX and Ad-FVII complexes, despite high-affinity binding of both these coagulation factors to the virus.


Asunto(s)
Adenovirus Humanos/genética , Adenovirus Humanos/fisiología , Factor VII/química , Factor VII/metabolismo , Factor X/química , Factor X/metabolismo , Vectores Genéticos , Animales , Células CHO , Cricetinae , Cricetulus , Células HEK293 , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Internalización del Virus
4.
Mol Ther ; 17(12): 2121-30, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19755961

RESUMEN

One of the significant hurdles toward safe and efficacious systemic treatment of cancer with oncolytic adenoviruses (Ads) is dose-limiting hepatotoxicity that prevents the increase of a therapeutic dose. In this study, we expanded the therapeutic window of oncolytic serotype 5 Ad (Ad5) by a genetic modification of hypervariable loop 5 (HVR5) in the capsid protein hexon that prevented infection of hepatocytes due to ablation of binding to blood factors. This oncolytic virus, Ad-GL-HB, had significantly reduced levels of hepatocyte transduction in immunocompetent and immunodeficient mice as compared to parental virus Ad-GL. The hepatocyte detargeting decreased liver damage and increased the maximum tolerated dose of Ad-GL-HB tenfold relative to that of Ad-GL. Intravenous (i.v.) injection of Ad-GL or Ad-GL-HB into tumor-bearing mice produced equally increased survival rates demonstrating that while Ad-GL-HB detargeted hepatocytes, it sustained tumor cell infection after systemic administration. The significantly improved safety of the virus allowed it to be used at increased doses for improved systemic antitumor efficacy. Our results suggest that hexon modifications provide valuable strategies for systemic oncolytic Ad therapy.


Asunto(s)
Adenoviridae/genética , Proteínas de la Cápside/genética , Vectores Genéticos/uso terapéutico , Neoplasias Experimentales/terapia , Viroterapia Oncolítica , Virus Oncolíticos/genética , Animales , Femenino , Hepatocitos/metabolismo , Humanos , Luminiscencia , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Desnudos , Neoplasias Experimentales/genética , Neoplasias Experimentales/virología , Transducción Genética
5.
J Virol ; 81(23): 12918-26, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17881437

RESUMEN

A short open reading frame named the "U exon," located on the adenovirus (Ad) l-strand (for leftward transcription) between the early E3 region and the fiber gene, is conserved in mastadenoviruses. We have observed that Ad5 mutants with large deletions in E3 that infringe on the U exon display a mild growth defect, as well as an aberrant Ad E2 DNA-binding protein (DBP) intranuclear localization pattern and an apparent failure to organize replication centers during late infection. Mutants in which the U exon DNA is reconstructed have a reversed phenotype. Chow et al. (L. T. Chow et al., J. Mol. Biol. 134:265-303, 1979) described mRNAs initiating in the region of the U exon and spliced to downstream sequences in the late DBP mRNA leader and the DBP-coding region. We have cloned this mRNA (as cDNA) from Ad5 late mRNA; the predicted protein is 217 amino acids, initiating in the U exon and continuing in frame in the DBP leader and in the DBP-coding region but in a different reading frame from DBP. Polyclonal and monoclonal antibodies generated against the predicted U exon protein (UXP) showed that UXP is approximately 24K in size by immunoblot and is a late protein. At 18 to 24 h postinfection, UXP is strongly associated with nucleoli and is found throughout the nucleus; later, UXP is associated with the periphery of replication centers, suggesting a function relevant to Ad DNA replication or RNA transcription. UXP is expressed by all four species C Ads. When expressed in transient transfections, UXP complements the aberrant DBP localization pattern of UXP-negative Ad5 mutants. Our data indicate that UXP is a previously unrecognized protein derived from a novel late l-strand transcription unit.


Asunto(s)
Adenovirus Humanos/genética , Adenovirus Humanos/fisiología , Proteínas Virales/genética , Proteínas Virales/fisiología , Proteínas E2 de Adenovirus/análisis , Nucléolo Celular/química , Núcleo Celular/química , Clonación Molecular , Humanos , Immunoblotting , Datos de Secuencia Molecular , Peso Molecular , Sistemas de Lectura Abierta , ARN Mensajero/genética , ARN Viral/genética , Análisis de Secuencia de ADN , Eliminación de Secuencia , Proteínas Virales/biosíntesis , Proteínas Virales/química , Replicación Viral/fisiología
6.
Mol Ther ; 15(3): 598-607, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17191072

RESUMEN

Novel approaches are needed to improve the antitumor potency and to increase the cancer specificity of oncolytic adenoviruses (Ad). We hypothesized that the combination of interferon-alpha (IFN-alpha) expression with a specific mutation in the e1a gene of Ad could target vector replication to genetic defects in the IFN-alpha pathway resulting in both improved antitumor efficacy and reduced toxicity. The conditionally replicative Ad vector KD3-IFN carries the dl1101/1107 mutation in the e1a gene that eliminates binding of E1A proteins to p300/CBP and pRb. KD3-IFN expresses human IFN-alpha in concurrence with vector replication and overexpresses the adenovirus death protein (ADP; E3-11.6K). The antitumor activity of KD3-IFN was significantly higher than that of a control vector in established human hepatocellular carcinoma tumors in immunodeficient mice and in hamster kidney cancer tumors in immunocompetent Syrian hamsters. The dl1101/1107 mutation rendered Ad replication sensitive to the antiviral effect of IFN-alpha in normal as opposed to cancer cells. These results translated to reduced vector toxicity upon systemic administration to C57BL/6 mice. The combination of Ad oncolysis, ADP overexpression, and IFN-alpha-mediated immunotherapy represents a three-pronged approach for increasing the anticancer efficacy of replicative Ads. Exploiting the dl1101/1107 mutation provides a mechanism for additional selectivity of IFN-alpha-expressing replication-competent Ads.


Asunto(s)
Adenoviridae/genética , Proteínas E1A de Adenovirus/metabolismo , Interferón-alfa/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Virus Oncolíticos/genética , Proteínas E1A de Adenovirus/genética , Animales , Línea Celular , Supervivencia Celular , Cricetinae , Femenino , Expresión Génica , Terapia Genética , Vectores Genéticos/genética , Genoma Viral/genética , Interferón-alfa/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/terapia , Ratones , Ratones Desnudos , Mutación/genética , Tasa de Supervivencia , Factores de Tiempo , Transgenes/genética , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Ther ; 15(3): 598-607, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28182898

RESUMEN

Novel approaches are needed to improve the antitumor potency and to increase the cancer specificity of oncolytic adenoviruses (Ad). We hypothesized that the combination of interferon-alpha (IFN-α) expression with a specific mutation in the e1a gene of Ad could target vector replication to genetic defects in the IFN-α pathway resulting in both improved antitumor efficacy and reduced toxicity. The conditionally replicative Ad vector KD3-IFN carries the dl1101/1107 mutation in the e1a gene that eliminates binding of E1A proteins to p300/CBP and pRb. KD3-IFN expresses human IFN-α in concurrence with vector replication and overexpresses the adenovirus death protein (ADP; E3-11.6K). The antitumor activity of KD3-IFN was significantly higher than that of a control vector in established human hepatocellular carcinoma tumors in immunodeficient mice and in hamster kidney cancer tumors in immunocompetent Syrian hamsters. The dl1101/1107 mutation rendered Ad replication sensitive to the antiviral effect of IFN-α in normal as opposed to cancer cells. These results translated to reduced vector toxicity upon systemic administration to C57BL/6 mice. The combination of Ad oncolysis, ADP overexpression, and IFN-α-mediated immunotherapy represents a three-pronged approach for increasing the anticancer efficacy of replicative Ads. Exploiting the dl1101/1107 mutation provides a mechanism for additional selectivity of IFN-α-expressing replication-competent Ads.

8.
Methods Mol Med ; 130: 223-35, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17401177

RESUMEN

Adenovirus research often requires purified high-titer virus stocks and accurate virus titers for use in experiments. Accurate titers are important for quantitative, interpretable, and reproducible results. This is especially true when there are comparisons of different mutant viruses following infection. This chapter details the large-scale preparation of adenovirus (either replication-competent or replication-defective) in spinner cultures (e.g., KB, HeLa, or 293 cells). Protocols for harvesting cells and isolation of adenovirus by CsCl banding are presented. Methods for titering adenovirus by plaque assay are presented along with a discussion of how plaque assays can be used to determine the kinetics of cell killing and cytolysis by adenoviruses.


Asunto(s)
Adenoviridae/clasificación , Adenoviridae/aislamiento & purificación , Cesio , Cloruros , Adenoviridae/genética , Línea Celular , Centrifugación por Gradiente de Densidad/métodos , Células HeLa , Humanos , Indicadores y Reactivos , Células KB , Riñón
9.
Cancer Res ; 64(10): 3638-44, 2004 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15150123

RESUMEN

We have constructed a novel oncolytic adenovirus (Ad) vector named VRX-009 that combines enhanced cell spread with tumor-specific replication. Enhanced spread, which could significantly increase antitumor efficacy, is mediated by overexpression of the Ad cytolytic protein named ADP (also known as E3-11.6K). Replication of VRX-009 is restricted to cells with a deregulated wnt signal transduction pathway by replacement of the wild-type Ad E4 promoter with a synthetic promoter consisting of five consensus binding sites for the T-cell factor transcription factor. Tumor-selective replication is indicated by several lines of evidence. VRX-009 expresses E4ORF3, a representative Ad E4 protein, only in colon cancer cell lines. Furthermore, VRX-009 replicates preferentially in colon cancer cell lines as evidenced by virus productivity 2 orders of magnitude higher in SW480 colon cancer cells than in A549 lung cancer cells. Replication in primary human bronchial epithelial cells and human umbilical vein endothelial cells was also significantly lower than in SW480 cells. When tested in human tumor xenografts in nude mice, VRX-009 effectively suppressed the growth of SW480 colon tumors but not of A549 lung tumors. VRX-009 may provide greater level of antitumor efficacy than standard oncolytic Ad vectors in tumors in which a defect in wnt signaling increases the level of nuclear beta-catenin.


Asunto(s)
Adenoviridae/fisiología , Proteínas E3 de Adenovirus/fisiología , Neoplasias/terapia , Neoplasias/virología , Proteínas Proto-Oncogénicas/genética , Adenoviridae/genética , Proteínas E3 de Adenovirus/biosíntesis , Proteínas E3 de Adenovirus/genética , Proteínas E4 de Adenovirus/biosíntesis , Proteínas E4 de Adenovirus/genética , Animales , División Celular/fisiología , Línea Celular Tumoral , Efecto Citopatogénico Viral , Endotelio Vascular/metabolismo , Endotelio Vascular/virología , Vectores Genéticos/genética , Humanos , Ratones , Neoplasias/genética , Plásmidos/genética , Proteínas Proto-Oncogénicas/fisiología , Transducción de Señal , Replicación Viral , Proteínas Wnt , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Hum Gene Ther ; 16(1): 139-46, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15703497

RESUMEN

Oncolytic human adenovirus (Ad) vectors exert their antitumor effect by replicating in and lysing tumor cells. These vectors are commonly evaluated in immunodeficient mice bearing human tumor xenografts. However, this model suffers because the mice are immunodeficient and are not permissive for human Ads. We have developed a cotton rat model to test the selectivity, immunogenicity, and efficacy of oncolytic Ad vectors. The cotton rat is a rodent species that is semipermissive for human Ads. We show that the cotton cancer rat cell line LCRT supports the replication of human Ad in tissue culture and that the cells are destroyed on virus replication. When injected subcutaneously, LCRT cells formed tumors in immunocompetent cotton rats, and the growth of these tumors was delayed by the injection of an oncolytic Ad vector. Replication of the Ad vector in the tumor was demonstrated by sampling tumor tissue and isolating infectious virus particles at various times after intratumoral injection of the virus. We propose that the cotton rat can be used as an animal model to evaluate oncolytic Ad vectors.


Asunto(s)
Adenovirus Humanos/fisiología , Modelos Animales de Enfermedad , Vectores Genéticos , Neoplasias/terapia , Infecciones por Adenovirus Humanos/terapia , Infecciones por Adenovirus Humanos/virología , Animales , Femenino , Terapia Genética , Humanos , Neoplasias/virología , Ratas , Sigmodontinae , Replicación Viral/fisiología
11.
Cancer Gene Ther ; 12(7): 617-26, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15761475

RESUMEN

Avian adenovirus CELO is a novel adenovirus vector system with the advantages of efficient production, high virion stability, and the absence of crossreactivity with Ad5-neutralizing antibodies. In this study, we evaluated the anticancer efficacy of a CELO vector encoding the herpes simplex virus type 1 thymidine kinase, a prodrug-activating therapeutic gene. Vectors carrying the gene for HSV-tk or EGFP under the control of the HCMV promoter in place of the "nonessential" region of the CELO genome were constructed. Anticancer activity of the CELO-TK vector was studied in vitro, in human and murine tumor cells in cell culture, and in vivo, in established subcutaneous murine B16 melanoma tumors in C57BL/6 mice. The CELO-TK vector mediated delivery of functional HSV-tk to tumor cell lines in cell culture. Comparison of the CELO-TK vector to a first-generation human adenovirus type 5 vector Ad5-TK in cultured H1299 cells showed equal levels of functional activity at increasing multiplicities of infection with CELO-based vector. CELO vectors allowed for transduction and expression of EGFP and HSV-tk genes in subcutaneous melanoma tumors in C57BL/6 mice. Intratumoral injections of CELO-TK followed by ganciclovir administration resulted in suppression of tumor growth and significantly increased the median of survival. The results of the study demonstrated the efficacy of CELO vector as a vehicle for the delivery of prodrug-activating genes such as HSV-tk to tumor cells in vitro and in vivo.


Asunto(s)
Adenovirus A Aviar/genética , Terapia Genética , Vectores Genéticos , Melanoma Experimental/terapia , Simplexvirus/enzimología , Timidina Quinasa/genética , Animales , Línea Celular Tumoral , Femenino , Proteínas Fluorescentes Verdes/genética , Humanos , Melanoma Experimental/genética , Melanoma Experimental/virología , Ratones , Ratones Endogámicos C57BL , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/terapia , Neoplasias Cutáneas/virología , Transducción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Virus Res ; 108(1-2): 149-59, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15681065

RESUMEN

The majority of proteins encoded in the early 3 (E3) region of human subgroup C adenoviruses function to modulate the host immune response. For example, gp19K, one of these E3 proteins, prevents the major histocompatibility complex type I (MHC-I) from presenting viral antigens on the surface of the infected cell. Other E3 proteins, such as the RID and 14.7K proteins, counteract the effector phase of the cellular immune response. In order to study further the effects of these proteins, we constructed an E1-/E3- adenovirus vector, Ad/E3, that contains all the E3 genes with the exception of the cytolytic adp gene, inserted into the deleted E1 region. The transcription of the E3 genes in this vector is driven by a CMV promoter in place of the native E3 promoter. Ad/E3 expressed close to wild-type adenovirus levels of all E3 proteins, and these proteins appear to function normally in cell culture. For example, in Ad/E3-infected cells, surface expression of MHC-I was down-regulated, as was cell surface display of death receptors Fas and TRAIL Receptor 1. A human cell line of lung origin (A549), which was rapidly rejected after transplantation into C57BL/6 mice, was protected for an extended time from the host immune response after infection with an Ad/E3, and went through a number of divisions in immunocompetent mice. These latter results indicate that the E3 proteins protect cells from destruction by the immune system.


Asunto(s)
Proteínas E3 de Adenovirus/fisiología , Rechazo de Injerto , Terapia de Inmunosupresión , Trasplantes , Proteínas E3 de Adenovirus/genética , Adenovirus Humanos/genética , Adenovirus Humanos/fisiología , Animales , Línea Celular Tumoral , Transformación Celular Viral , Femenino , Vectores Genéticos , Humanos , Inmunocompetencia , Ratones , Ratones Endogámicos C57BL , Trasplante Heterólogo
13.
PLoS Negl Trop Dis ; 9(6): e0003850, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26086739

RESUMEN

Monkeypox virus (MPXV) is the etiological agent of human (MPX). It is an emerging orthopoxvirus zoonosis in the tropical rain forest of Africa and is endemic in the Congo-basin and sporadic in West Africa; it remains a tropical neglected disease of persons in impoverished rural areas. Interaction of the human population with wildlife increases human infection with MPX virus (MPXV), and infection from human to human is possible. Smallpox vaccination provides good cross-protection against MPX; however, the vaccination campaign ended in Africa in 1980, meaning that a large proportion of the population is currently unprotected against MPXV infection. Disease control hinges on deterring zoonotic exposure to the virus and, barring that, interrupting person-to-person spread. However, there are no FDA-approved therapies against MPX, and current vaccines are limited due to safety concerns. For this reason, new studies on pathogenesis, prophylaxis and therapeutics are still of great interest, not only for the scientific community but also for the governments concerned that MPXV could be used as a bioterror agent. In the present study, a new vaccination strategy approach based on three recombinant bovine herpesvirus 4 (BoHV-4) vectors, each expressing different MPXV glycoproteins, A29L, M1R and B6R were investigated in terms of protection from a lethal MPXV challenge in STAT1 knockout mice. BoHV-4-A-CMV-A29LgD106ΔTK, BoHV-4-A-EF1α-M1RgD106ΔTK and BoHV-4-A-EF1α-B6RgD106ΔTK were successfully constructed by recombineering, and their capacity to express their transgene was demonstrated. A small challenge study was performed, and all three recombinant BoHV-4 appeared safe (no weight-loss or obvious adverse events) following intraperitoneal administration. Further, BoHV-4-A-EF1α-M1RgD106ΔTK alone or in combination with BoHV-4-A-CMV-A29LgD106ΔTK and BoHV-4-A-EF1α-B6RgD106ΔTK, was shown to be able to protect, 100% alone and 80% in combination, STAT1(-/-) mice against mortality and morbidity. This work demonstrated the efficacy of BoHV-4 based vectors and the use of BoHV-4 as a vaccine-vector platform.


Asunto(s)
Antígenos Virales/inmunología , Herpesvirus Bovino 4/fisiología , Monkeypox virus/inmunología , Mpox/prevención & control , Factor de Transcripción STAT1/metabolismo , Vacunas Virales/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Bovinos , Línea Celular , Regulación de la Expresión Génica , Vectores Genéticos , Herpesvirus Bovino 4/inmunología , Humanos , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Factor de Transcripción STAT1/genética , Transfección , Vacunas Virales/genética
14.
Cancer Gene Ther ; 10(3): 193-200, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12637940

RESUMEN

We have described three potential adenovirus type 5 (Ad5)-based replication-competent cancer gene therapy vectors named KD1, KD3, and VRX-007. All three vectors overexpress an Ad5 protein named Adenovirus Death Protein (ADP, also named E3-11.6 K protein). ADP is required for efficient lysis of Ad5-infected cells and spread of virus from cell to cell, and thus its overexpression increases the oncolytic activity of the vectors. KD1 and KD3 contain mutations in the Ad5 E1A gene that knock out binding of the E1A proteins to cellular p300/CBP and pRB; these mutations allow KD1 and KD3 to grow well in cancer cells but not in normal cells. VRX-007 has wild-type E1A. Here we report that radiation increases the oncolytic activity of KD1, KD3, and VRX-007. This increased activity was observed in cultured cells, and it was not because of radiation-induced replication of the vectors. The combination of radiation plus KD3 suppressed the growth of A549 lung adenocarcinoma xenografts in nude mice more efficiently than radiation alone or KD3 alone. The combination of ADP-overexpressing vectors and radiation may have potential in treating cancer.


Asunto(s)
Adenoviridae/genética , Proteínas E3 de Adenovirus/genética , Terapia Genética , Neoplasias Experimentales/radioterapia , Neoplasias Experimentales/terapia , Proteínas E3 de Adenovirus/metabolismo , Animales , Línea Celular Tumoral , Terapia Combinada , Relación Dosis-Respuesta en la Radiación , Femenino , Vectores Genéticos , Humanos , Neoplasias Pulmonares/terapia , Ratones , Ratones Desnudos , Neoplasias Experimentales/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Cancer Gene Ther ; 9(8): 651-4, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12136425

RESUMEN

The successful clinical application of adenovirus (Ad) in cancer control has been of limited success because of the current inability to infect the majority of cancer cells with a large amount of vector. In this study, we show that when human lung tumors growing in immunodeficient nude mice were coinfected with a replication-defective (RD) Ad vector expressing green fluorescent protein and a replication-competent (RC) Ad vector named KD3, KD3 enhanced the expression of green fluorescent protein throughout the tumor. Also, KD3 and another RC vector named KD1 complemented the expression of luciferase from a RD vector in a human liver tumor xenotransplant in nude mice. Altogether, these results suggest that the combination of a RD vector with a RC vector might be a more effective treatment for cancer than either vector alone due to more widespread dissemination of the virus.


Asunto(s)
Adenoviridae/genética , Supervivencia Celular/genética , Virus Defectuosos/genética , Vectores Genéticos , Transgenes , Replicación Viral , Adenoviridae/fisiología , Animales , Proteínas Fluorescentes Verdes , Humanos , Luciferasas/genética , Proteínas Luminiscentes/genética , Ratones , Ratones Desnudos , Células Tumorales Cultivadas
16.
Int Rev Immunol ; 23(1-2): 75-111, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-14690856

RESUMEN

In the evolutionary battle between viruses and their hosts, viruses have armed themselves with weapons to defeat the host's attacks on infected cells. Various proteins encoded in the adenovirus (Ad) E3 transcription unit protect cells from killing mediated by cytotoxic T cells and death-inducing cytokines such as tumor necrosis factor (TNF), Fas ligand, and TNF-related apoptosis-inducing ligand (TRAIL). The viral protein E3-gp19 K blocks MHC class-I-restricted antigen presentation, which diminishes killing by cytotoxic T cells. The receptor internalization and degradation (RID) complex (formerly E3-10.4 K/14.5 K) stimulates the clearance from the cell surface and subsequent degradation of the receptors for Fas ligand and TRAIL, thereby preventing the action of these important immune mediators. RID also downmodulates the epidermal growth factor receptor (EGFR), although what role, if any, this function has in immune regulation is uncertain. In addition, RID antagonizes TNF-mediated apoptosis and inflammation through a mechanism that does not primarily involve receptor downregulation. E3-6.7 K functions together with RID in downregulating some TRAIL receptors and may block apoptosis independently of other E3 proteins. Furthermore, E3-14.7 K functions as a general inhibitor of TNF-mediated apoptosis and blocks TRAIL-induced apoptosis. Finally, after expending great effort to maintain cell viability during the early part of the virus replication cycle, Ads lyse the cell to allow efficient virus release and dissemination. To perform this task subgroup C Ads synthesize a protein late in infection named ADP (formerly E3-11.6 K) that is required for efficient virus release. This review focuses on recent experiments aimed at discovering the mechanism of action of these critically important viral proteins.


Asunto(s)
Adenoviridae/patogenicidad , Proteínas E3 de Adenovirus/fisiología , Adenoviridae/genética , Adenoviridae/fisiología , Infecciones por Adenoviridae/patología , Infecciones por Adenoviridae/fisiopatología , Infecciones por Adenoviridae/virología , Proteínas E3 de Adenovirus/genética , Animales , Apoptosis/fisiología , Modelos Animales de Enfermedad , Vectores Genéticos , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Ratones , Ratones Transgénicos , Modelos Biológicos , Transcripción Genética
17.
Cell Rep ; 3(6): 1840-6, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23770239

RESUMEN

Although molecular components that execute noninflammatory apoptotic cell death are well defined, molecular pathways that trigger necrotic cell death remain poorly characterized. Here, we show that in response to infection with adenovirus or Listeria monocytogenes, macrophages in vivo undergo rapid proinflammatory necrotic death that is controlled by interferon-regulatory factor 3 (IRF3). The transcriptional activity of IRF3 is, surprisingly, not required for the induction of necrosis, and it proceeds normally in mice deficient in all known regulators of necrotic death or IRF3 activation, including RIPK3, caspases 1, 8, or 11, STING, and IPS1/MAVS. Although L. monocytogenes triggers necrosis to promote the infection, IRF3-dependent necrosis is required for reducing pathogen burden in the models of disseminated infection with adenovirus. Therefore, our studies implicate IRF3 as a principal and nonredundant component of a physiologically regulated necrotic cell-death pathway that operates as an effective innate immune mechanism of host protection against disseminated virus infection.


Asunto(s)
Infecciones por Adenovirus Humanos/inmunología , Adenovirus Humanos/inmunología , Factor 3 Regulador del Interferón/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Macrófagos/microbiología , Macrófagos/patología , Infecciones por Adenovirus Humanos/patología , Animales , Caspasas/metabolismo , Inmunidad Innata/inmunología , Factor 3 Regulador del Interferón/deficiencia , Factor 3 Regulador del Interferón/genética , Listeriosis/patología , Macrófagos/inmunología , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Necrosis/inmunología , Necrosis/patología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal
18.
Methods Mol Biol ; 797: 35-52, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21948467

RESUMEN

Oncolytic (replication-competent) adenoviruses (Ads) represent the most advanced platform for cancer gene therapy. These viral vectors ablate tumors by killing tumor cells in the process of virus replication. As progeny virions are released, they infect remaining cancer cells, generating a bystander effect. Ads engineered for increased cancer specificity produce less damage to normal tissues. First-generation oncolytic Ads have demonstrated acceptable levels of safety while the efficacy was observed only in combination with chemotherapy and/or radiation. Second-generation oncolytic Ads are armed with therapeutic transgenes to increase release, spread, and bystander effect for enhancing the efficacy. Third-generation oncolytic Ads are armed vectors with capsid modifications for transductional detargeting from normal tissues and targeting to cancer cells. Chemical modification of the capsid additionally improves therapeutic window. Here, we describe methods for generation and characterization of advanced-generation oncolytic Ads.


Asunto(s)
Adenoviridae/química , Adenoviridae/genética , Terapia Genética , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/química , Virus Oncolíticos/genética , Adenoviridae/aislamiento & purificación , Animales , Cápside/química , Vectores Genéticos , Humanos , Ratones , Ratones Desnudos , Virus Oncolíticos/aislamiento & purificación
19.
Science ; 338(6108): 795-8, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-23019612

RESUMEN

Although coagulation factors play a role in host defense for "living fossils" such as horseshoe crabs, the role of the coagulation system in immunity in higher organisms remains unclear. We modeled the interface of human species C adenovirus (HAdv) interaction with coagulation factor X (FX) and introduced a mutation that abrogated formation of the HAdv-FX complex. In vivo genome-wide transcriptional profiling revealed that FX-binding-ablated virus failed to activate a distinct network of nuclear factor κB-dependent early-response genes that are activated by HAdv-FX complex downstream of TLR4/MyD88/TRIF/TRAF6 signaling. Our study implicates host factor "decoration" of the virus as a mechanism to trigger an innate immune sensor that responds to a misplacement of coagulation FX from the blood into intracellular macrophage compartments upon virus entry into the cell.


Asunto(s)
Infecciones por Adenoviridae/inmunología , Adenovirus Humanos/inmunología , Adenovirus Humanos/metabolismo , Factor X/metabolismo , Inmunidad Innata , Infecciones por Adenoviridae/metabolismo , Infecciones por Adenoviridae/virología , Adenovirus Humanos/genética , Animales , Células CHO , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Línea Celular Tumoral , Cricetinae , Cricetulus , Microscopía por Crioelectrón , Citocinas/metabolismo , Factor X/química , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Hepatocitos/virología , Humanos , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Simulación de Dinámica Molecular , Mutación , FN-kappa B/metabolismo , Transducción de Señal , Internalización del Virus
20.
Hum Gene Ther ; 21(2): 179-90, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19788385

RESUMEN

Oncolytic virotherapy makes use of the natural ability of viruses to infect and kill cancer cells. Adenovirus serotype 5 (Ad5) has been approved for use in humans as a therapy for solid cancers. In this study, we have tested whether Ad5 and low-seroprevalence adenoviruses can be used as oncolytics for multiple myeloma (MM). We show that Ad5 productively infects most myeloma cell lines, replicates to various degrees, and mediates oncolytic cell killing in vitro and in vivo. Comparison of Ad5 with low-seroprevalence Ads on primary marrow samples from MM patients revealed striking differences in the abilities of different adenoviral serotypes to kill normal CD138(-) cells and CD138(+) MM cells. Ad5 and Ad6 from species C and Ad26 and Ad48 from species D all mediated killing of CD138(+) cells with low-level killing of CD138(-) cells. In contrast, Ad11, Ad35, Ad40, and Ad41 mediated weak oncolytic effects in all of the cells. Comparison of cell binding, cell entry, and replication revealed that Ad11 and Ad35 bound MM cells 10 to 100 times better than other serotypes. However, after this efficient interaction, Ad11 and Ad35 viral DNA was not replicated and cell killing did not occur. In contrast, Ad5, Ad6, Ad26, and Ad48 all replicated 10- to 100-fold in MM cells and this correlated with cell killing. These data suggest that Ad5 and other low-seroprevalence adenoviruses may have utility as oncolytic agents against MM and other hematologic malignancies.


Asunto(s)
Adenoviridae/patogenicidad , Mieloma Múltiple/terapia , Mieloma Múltiple/virología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Adenoviridae/clasificación , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Línea Celular Tumoral , Efecto Citopatogénico Viral , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Virus Oncolíticos/genética , Virus Oncolíticos/metabolismo , Resultado del Tratamiento , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA