Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Antiviral Res ; 195: 105179, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34530009

RESUMEN

Orthopoxviruses such as variola and monkeypox viruses continue to threaten the human population. Monkeypox virus is endemic in central and western Africa and outbreaks have reached as far as the U.S. Although variola virus, the etiologic agent of smallpox, has been eradicated by a successful vaccination program, official and likely clandestine stocks of the virus exist. Moreover, studies with ectromelia virus (the etiological agent of mousepox) have revealed that IL-4 recombinant viruses are significantly more virulent than wild-type viruses even in mice treated with vaccines and/or antivirals. For these reasons, it is critical that antiviral modalities are developed to treat these viruses should outbreaks, or deliberate dissemination, occur. Currently, 2 antivirals (brincidofovir and tecovirimat) are in the U.S. stockpile allowing for emergency use of the drugs to treat smallpox. Both antivirals have advantages and disadvantages in a clinical and emergency setting. Here we report on the efficacy of a recombinant immunoglobulin (rVIG) that demonstrated efficacy against several orthopoxviruses in vitro and in vivo in both a prophylactic and therapeutic fashion. A single intraperitoneal injection of rVIG significantly protected mice when given up to 14 days before or as late as 6 days post challenge. Moreover, rVIG reduced morbidity, as measured by weight-change, as well as several previously established biomarkers of disease. In rVIG treated mice, we found that vDNA levels in blood were significantly reduced, as was ALT (a marker of liver damage) and infectious virus levels in the liver. No apparent adverse events were observed in rVIG treated mice, suggesting the immunoglobulin is well tolerated. These findings suggest that recombinant immunoglobulins could be candidates for further evaluation and possible licensure under the FDA Animal Rule.


Asunto(s)
Antivirales/uso terapéutico , Inmunoglobulinas/uso terapéutico , Orthopoxvirus/efectos de los fármacos , Viruela/tratamiento farmacológico , Vaccinia/tratamiento farmacológico , Animales , Antivirales/administración & dosificación , Benzamidas , Línea Celular , Chlorocebus aethiops , Citosina/análogos & derivados , Femenino , Humanos , Isoindoles , Ratones , Ratones Endogámicos BALB C , Organofosfonatos , Viruela/prevención & control , Viruela/virología , Vacuna contra Viruela/administración & dosificación , Vacunas de ADN/administración & dosificación , Vaccinia/prevención & control , Vaccinia/virología
2.
Vaccine ; 37(36): 5371-5381, 2019 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-31331771

RESUMEN

Influenza world-wide causes significant morbidity and mortality annually, and more severe pandemics when novel strains evolve to which humans are immunologically naïve. Because of the high viral mutation rate, new vaccines must be generated based on the prevalence of circulating strains every year. New approaches to induce more broadly protective immunity are urgently needed. Previous research has demonstrated that influenza-specific T cells can provide broadly heterotypic protective immunity in both mice and humans, supporting the rationale for developing a T cell-targeted universal influenza vaccine. We used state-of-the art immunoinformatic tools to identify putative pan-HLA-DR and HLA-A2 supertype-restricted T cell epitopes highly conserved among > 50 widely diverse influenza A strains (representing hemagglutinin types 1, 2, 3, 5, 7 and 9). We found influenza peptides that are highly conserved across influenza subtypes that were also predicted to be class I epitopes restricted by HLA-A2. These peptides were found to be immunoreactive in HLA-A2 positive but not HLA-A2 negative individuals. Class II-restricted T cell epitopes that were highly conserved across influenza subtypes were identified. Human CD4+ T cells were reactive with these conserved CD4 epitopes, and epitope expanded T cells were responsive to both H1N1 and H3N2 viruses. Dendritic cell vaccines pulsed with conserved epitopes and DNA vaccines encoding these epitopes were developed and tested in HLA transgenic mice. These vaccines were highly immunogenic, and more importantly, vaccine-induced immunity was protective against both H1N1 and H3N2 influenza challenges. These results demonstrate proof-of-principle that conserved T cell epitopes expressed by widely diverse influenza strains can induce broadly protective, heterotypic influenza immunity, providing strong support for further development of universally relevant multi-epitope T cell-targeting influenza vaccines.


Asunto(s)
Vacunas contra la Influenza/uso terapéutico , Gripe Humana/inmunología , Gripe Humana/prevención & control , Animales , Biología Computacional , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/metabolismo , Femenino , Humanos , Inmunidad Celular/inmunología , Inmunidad Celular/fisiología , Virus de la Influenza A/inmunología , Virus de la Influenza A/patogenicidad , Masculino , Ratones , Linfocitos T/inmunología , Linfocitos T/metabolismo
3.
Am J Physiol Regul Integr Comp Physiol ; 316(5): R525-R534, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30811246

RESUMEN

Double-stranded (ds) RNA, both synthetic and produced during virus replication, rapidly stimulates MAPK and NF-κB signaling that results in expression of the inflammatory genes inducible nitric oxide synthase, cyclooxygenase 2, and IL-1ß by macrophages. Using biochemical and genetic approaches, we have identified the chemokine ligand-binding C-C chemokine receptor type 5 (CCR5) as a cell surface signaling receptor required for macrophage expression of inflammatory genes in response to dsRNA. Activation of macrophages by synthetic dsRNA does not require known dsRNA receptors, as poly(inosinic:cytidylic) acid [poly(I:C)] activates signaling pathways leading to expression of inflammatory genes to similar levels in wild-type and Toll-like receptor 3- or melanoma differentiation antigen 5-deficient macrophages. In contrast, macrophage activation in response to poly(I:C) is attenuated in macrophages isolated from mice lacking CCR5. These findings support a role for CCR5 as a cell surface signaling receptor that participates in activation of inflammatory genes in macrophages in response to the viral dsRNA mimetic poly(inosinic:cytidylic) acid by pathways that are distinct from classical dsRNA receptor-mediated responses.


Asunto(s)
Inflamación/metabolismo , Activación de Macrófagos/efectos de los fármacos , Macrófagos Peritoneales/efectos de los fármacos , Poli I-C/farmacología , Receptores CCR5/agonistas , Transducción de Señal/efectos de los fármacos , Animales , Citocinas/genética , Citocinas/metabolismo , Regulación de la Expresión Génica , Inflamación/genética , Inflamación/inmunología , Helicasa Inducida por Interferón IFIH1/deficiencia , Helicasa Inducida por Interferón IFIH1/genética , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células RAW 264.7 , Receptores CCR5/genética , Receptores CCR5/metabolismo , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo
4.
BMC Pharmacol Toxicol ; 19(1): 80, 2018 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-30514402

RESUMEN

BACKGROUND: Several tyrosine kinase inhibitors (TKIs) developed as anti-cancer drugs, also have anti-viral activity due to their ability to disrupt productive replication and dissemination in infected cells. Consequently, such drugs are attractive candidates for "repurposing" as anti-viral agents. However, clinical evaluation of therapeutics against infectious agents associated with high mortality, but low or infrequent incidence, is often unfeasible. The United States Food and Drug Administration formulated the "Animal Rule" to facilitate use of validated animal models for conducting anti-viral efficacy studies. METHODS: To enable such efficacy studies of two clinically approved TKIs, nilotinib, and imatinib, we first conducted comprehensive pharmacokinetic (PK) studies in relevant rodent and non-rodent animal models. PK of these agents following intravenous and oral dosing were evaluated in C57BL/6 mice, prairie dogs, guinea pigs and Cynomolgus monkeys. Plasma samples were analyzed using an LC-MS/MS method. Secondarily, we evaluated the utility of allometry-based inter-species scaling derived from previously published data to predict the PK parameters, systemic clearance (CL) and the steady state volume of distribution (Vss) of these two drugs in prairie dogs, an animal model not tested thus far. RESULTS: Marked inter-species variability in PK parameters and resulting oral bioavailability was observed. In general, elimination half-lives of these agents in mice and guinea pigs were much shorter (1-3 h) relative to those in larger species such as prairie dogs and monkeys. The longer nilotinib elimination half-life in prairie dogs (i.v., 6.5 h and oral, 7.5 h), facilitated multiple dosing PK and safety assessment. The allometry-based predicted values of the Vss and CL were within 2.0 and 2.5-fold, respectively, of the observed values. CONCLUSIONS: Our results suggest that prairie dogs and monkeys may be suitable rodent and non-rodent species to perform further efficacy testing of these TKIs against orthopoxvirus infections. The use of rodent models such as C57BL/6 mice and guinea pigs for assessing pre-clinical anti-viral efficacy of these two TKIs may be limited due to short elimination and/or low oral bioavailability. Allometry-based correlations, derived from existing literature data, may provide initial estimates, which may serve as a useful guide for pre-clinical PK studies in untested animal models.


Asunto(s)
Antineoplásicos/farmacocinética , Antivirales/farmacocinética , Mesilato de Imatinib/farmacocinética , Proteínas Tirosina Quinasas/farmacocinética , Pirimidinas/farmacocinética , Administración Intravenosa , Administración Oral , Animales , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos , Femenino , Cobayas , Macaca fascicularis , Masculino , Ratones Endogámicos C57BL , Sciuridae
5.
Cell Rep ; 25(9): 2369-2378.e4, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30485806

RESUMEN

Tracking antigen-specific T cell responses over time within individuals is difficult because of lack of knowledge of antigen-specific TCR sequences, limitations in sample size, and assay sensitivities. We hypothesized that analyses of high-throughput sequencing of TCR clonotypes could provide functional readouts of individuals' immunological histories. Using high-throughput TCR sequencing, we develop a database of TCRß sequences from large cohorts of mice before (naive) and after smallpox vaccination. We computationally identify 315 vaccine-associated TCR sequences (VATS) that are used to train a diagnostic classifier that distinguishes naive from vaccinated samples in mice up to 9 months post-vaccination with >99% accuracy. We determine that the VATS library contains virus-responsive TCRs by in vitro expansion assays and virus-specific tetramer sorting. These data outline a platform for advancing our capabilities to identify pathogen-specific TCR sequences, which can be used to identify and quantitate low-frequency pathogen-specific TCR sequences in circulation over time with exceptional sensitivity.


Asunto(s)
Rastreo Celular , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Receptores de Antígenos de Linfocitos T/metabolismo , Virus/metabolismo , Secuencia de Aminoácidos , Animales , Células Clonales , Femenino , Biblioteca de Genes , Masculino , Ratones Endogámicos C57BL , Orthopoxvirus , Péptidos/química , Infecciones por Poxviridae/virología , Receptores de Antígenos de Linfocitos T/química , Vacunación
6.
Antiviral Res ; 149: 41-47, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29129708

RESUMEN

Chronic hepatitis B virus infection cannot be cured by current therapies, so new treatments are urgently needed. We recently identified novel inhibitors of the hepatitis B virus ribonuclease H that suppress viral replication in cell culture. Here, we employed immunodeficient FRG KO mice whose livers had been engrafted with primary human hepatocytes to ask whether ribonuclease H inhibitors can suppress hepatitis B virus replication in vivo. Humanized FRG KO mice infected with hepatitis B virus were treated for two weeks with the ribonuclease H inhibitors #110, an α-hydroxytropolone, and #208, an N-hydroxypyridinedione. Hepatitis B virus viral titers and S and e antigen plasma levels were measured. Treatment with #110 and #208 caused significant reductions in plasma viremia without affecting hepatitis B virus S or e antigen levels, and viral titers rebounded following treatment cessation. This is the expected pattern for inhibitors of viral DNA synthesis. Compound #208 suppressed viral titers of both hepatitis B virus genotype A and C isolates. These data indicate that Hepatitis B virus replication can be suppressed during infection in an animal by inhibiting the viral ribonuclease H, validating the ribonuclease H as a novel target for antiviral drug development.


Asunto(s)
Antivirales/farmacología , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/fisiología , Ribonucleasa H/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Animales , Antivirales/administración & dosificación , Antivirales/farmacocinética , Replicación del ADN/efectos de los fármacos , Genotipo , Hepatitis B/tratamiento farmacológico , Hepatitis B/virología , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Proyectos Piloto , Resultado del Tratamiento
7.
Viruses ; 9(8)2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28763036

RESUMEN

Taterapox virus (TATV), which was isolated from an African gerbil (Tatera kempi) in 1975, is the most closely related virus to variola; however, only the original report has examined its virology. We have evaluated the tropism of TATV in vivo in small animals. We found that TATV does not infect Graphiurus kelleni, a species of African dormouse, but does induce seroconversion in the Mongolian gerbil (Meriones unguiculatus) and in mice; however, in wild-type mice and gerbils, the virus produces an unapparent infection. Following intranasal and footpad inoculations with 1 × 106 plaque forming units (PFU) of TATV, immunocompromised stat1-/- mice showed signs of disease but did not die; however, SCID mice were susceptible to intranasal and footpad infections with 100% mortality observed by Day 35 and Day 54, respectively. We show that death is unlikely to be a result of the virus mutating to have increased virulence and that SCID mice are capable of transmitting TATV to C57BL/6 and C57BL/6 stat1-/- animals; however, transmission did not occur from TATV inoculated wild-type or stat1-/- mice. Comparisons with ectromelia (the etiological agent of mousepox) suggest that TATV behaves differently both at the site of inoculation and in the immune response that it triggers.


Asunto(s)
Orthopoxvirus/fisiología , Infecciones por Poxviridae/virología , Tropismo Viral , Animales , Antivirales/uso terapéutico , Modelos Animales de Enfermedad , Virus de la Ectromelia/genética , Virus de la Ectromelia/fisiología , Ectromelia Infecciosa/virología , Especificidad del Huésped , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Orthopoxvirus/genética , Orthopoxvirus/inmunología , Orthopoxvirus/aislamiento & purificación , Infecciones por Poxviridae/tratamiento farmacológico , Infecciones por Poxviridae/inmunología , Infecciones por Poxviridae/transmisión , Factor de Transcripción STAT1/deficiencia , Factor de Transcripción STAT1/genética
8.
Antiviral Res ; 139: 112-116, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28039021

RESUMEN

Orthopoxviruses continue to pose a significant threat to the population as potential agents of bioterrorism. An intentional release of natural or engineered variola virus (VARV) or monkeypox viruses would cause mortality and morbidity in the target population. To address this, antivirals have been developed and evaluated in animal models of smallpox and monkeypox. One such antiviral, brincidofovir (BCV, previously CMX001), has demonstrated high levels of efficacy against orthopoxviruses in animal models and is currently under clinical evaluation for prevention and treatment of diseases caused by cytomegaloviruses and adenoviruses. In this study we use the mousepox model of smallpox to evaluate the relationship between the magnitude of the infectious virus dose and an efficacious BCV therapy outcome when treatment is initiated concomitant with detection of ectromelia virus viral DNA (vDNA) in mouse buccal swabs. We found that vDNA could be detected in buccal swabs of some, but not all infected mice over a range of challenge doses by day 3 or 4 postexposure, when initiation of BCV treatment was efficacious, suggesting that detection of vDNA in buccal swabs could be used as a trigger to initiate BCV treatment of an entire potentially exposed population. However, buccal swabs of some mice did not become positive until 5 days postexposure, when initiation of BCV therapy failed to protect mice that received high doses of virus. And finally, the data suggest that the therapeutic window for efficacious BCV treatment decreases as the virus infectious dose increases. Extrapolating these findings to VARV, the data suggest that treatment should be initiated as soon as possible after exposure and not rely on a diagnostic tool such as the measurement of vDNA in buccal cavity swabs; however, consideration should be given to the fact that the behavior/disease-course of VARV in humans is different from that of ectromelia virus in the mouse.


Asunto(s)
Antivirales/uso terapéutico , Citosina/análogos & derivados , ADN Viral/efectos de los fármacos , Virus de la Ectromelia/efectos de los fármacos , Ectromelia Infecciosa/tratamiento farmacológico , Mucosa Bucal/virología , Organofosfonatos/uso terapéutico , Animales , Antivirales/administración & dosificación , Citosina/administración & dosificación , Citosina/uso terapéutico , ADN Viral/aislamiento & purificación , Modelos Animales de Enfermedad , Ectromelia Infecciosa/virología , Ratones , Organofosfonatos/administración & dosificación , Orthopoxvirus/efectos de los fármacos , Viruela/tratamiento farmacológico , Viruela/virología
9.
PLoS One ; 11(4): e0153705, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27100298

RESUMEN

Results from studies comparing the diversity and specificity of the TCR repertoires expressed by conventional (Tconv) and regulatory (Treg) CD4+ T cell have varied depending on the experimental system employed. We developed a new model in which T cells express a single fixed TCRα chain, randomly rearranged endogenous TCRß chains, and a Foxp3-GFP reporter. We purified CD4+Foxp3- and CD4+Foxp3+ cells, then performed biased controlled multiplex PCR and high throughput sequencing of endogenous TCRß chains. We identified >7,000 different TCRß sequences in the periphery of 5 individual mice. On average, ~12% of TCR sequences were expressed by both conventional and regulatory populations within individual mice. The CD4+ T cells that expressed shared TCR sequences were present at higher frequencies compared to T cells expressing non-shared TCRs. Furthermore, nearly all (>90%) of the TCR sequences that were shared within mice were identical at the DNA sequence level, indicating that conventional and regulatory T cells that express shared TCRs are derived from common clones. Analysis of TCR repertoire overlap in the thymus reveals that a large proportion of Tconv and Treg sharing observed in the periphery is due to clonal expansion in the thymus. Together these data show that there are a limited number of TCR sequences shared between Tconv and Tregs. Also, Tconv and Tregs sharing identical TCRs are found at relatively high frequencies and are derived from common progenitors, of which a large portion are generated in the thymus.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Factores de Transcripción Forkhead/fisiología , Receptores de Antígenos de Linfocitos T/fisiología , Linfocitos T Reguladores/inmunología , Timo/inmunología , Secuencia de Aminoácidos , Animales , Diferenciación Celular , Linaje de la Célula , Células Clonales , Femenino , Citometría de Flujo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Homología de Secuencia de Aminoácido , Timo/citología , Timo/metabolismo
10.
J Biol Chem ; 291(1): 447-61, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26546677

RESUMEN

Exogenous cytokine therapy can induce systemic toxicity, which might be prevented by activating endogenously produced cytokines in local cell niches. Here we developed antibody-based activators of cytokine signaling (AcCS), which recognize cytokines only when they are bound to their cell surface receptors. AcCS were developed for type I interferons (IFNs), which induce cellular activities by binding to cell surface receptors IFNAR1 and IFNAR2. As a potential alternative to exogenous IFN therapy, AcCS were shown to potentiate the biological activities of natural IFNs by ∼100-fold. Biochemical and structural characterization demonstrates that the AcCS stabilize the IFN-IFNAR2 binary complex by recognizing an IFN-induced conformational change in IFNAR2. Using IFN mutants that disrupt IFNAR1 binding, AcCS were able to enhance IFN antiviral potency without activating antiproliferative responses. This suggests AcCS can be used to manipulate cytokine signaling for basic science and possibly for therapeutic applications.


Asunto(s)
Citocinas/inmunología , Fragmentos de Inmunoglobulinas/inmunología , Receptores de Citocinas/inmunología , Transducción de Señal , Antivirales/química , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Fragmentos de Inmunoglobulinas/farmacología , Interferón-alfa/farmacología , Cinética , Mutación/genética , Fosforilación , Conformación Proteica , Receptor de Interferón alfa y beta/química , Receptor de Interferón alfa y beta/metabolismo , Reproducibilidad de los Resultados , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/efectos de los fármacos
11.
Immunity ; 43(6): 1112-24, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26680205

RESUMEN

CD8(+) T cells and NK cells protect from viral infections by killing virally infected cells and secreting interferon-γ. Several inhibitory receptors limit the magnitude and duration of these anti-viral responses. NKG2A, which is encoded by Klrc1, is a lectin-like inhibitory receptor that is expressed as a heterodimer with CD94 on NK cells and activated CD8(+) T cells. Previous studies on the impact of CD94/NKG2A heterodimers on anti-viral responses have yielded contrasting results and the in vivo function of NKG2A remains unclear. Here, we generated Klrc1(-/-) mice and found that NKG2A is selectively required for resistance to ectromelia virus (ECTV). NKG2A functions intrinsically within ECTV-specific CD8(+) T cells to limit excessive activation, prevent apoptosis, and preserve the specific CD8(+) T cell response. Thus, although inhibitory receptors often cause T cell exhaustion and viral spreading during chronic viral infections, NKG2A optimizes CD8(+) T cell responses during an acute poxvirus infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Asesinas Naturales/inmunología , Subfamília C de Receptores Similares a Lectina de Células NK/inmunología , Infecciones por Poxviridae/inmunología , Animales , Citotoxicidad Inmunológica/inmunología , Modelos Animales de Enfermedad , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
PLoS Pathog ; 11(6): e1004941, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26107264

RESUMEN

Naïve anti-viral CD8+ T cells (TCD8+) are activated by the presence of peptide-MHC Class I complexes (pMHC-I) on the surface of professional antigen presenting cells (pAPC). Increasing the number of pMHC-I in vivo can increase the number of responding TCD8+. Antigen can be presented directly or indirectly (cross presentation) from virus-infected and uninfected cells, respectively. Here we determined the relative importance of these two antigen presenting pathways in mousepox, a natural disease of the mouse caused by the poxvirus, ectromelia (ECTV). We demonstrated that ECTV infected several pAPC types (macrophages, B cells, and dendritic cells (DC), including DC subsets), which directly presented pMHC-I to naïve TCD8+ with similar efficiencies in vitro. We also provided evidence that these same cell-types presented antigen in vivo, as they form contacts with antigen-specific TCD8+. Importantly, the number of pMHC-I on infected pAPC (direct presentation) vastly outnumbered those on uninfected cells (cross presentation), where presentation only occurred in a specialized subset of DC. In addition, prior maturation of DC failed to enhance antigen presentation, but markedly inhibited ECTV infection of DC. These results suggest that direct antigen presentation is the dominant pathway in mice during mousepox. In a broader context, these findings indicate that if a virus infects a pAPC then the presentation by that cell is likely to dominate over cross presentation as the most effective mode of generating large quantities of pMHC-I is on the surface of pAPC that endogenously express antigens. Recent trends in vaccine design have focused upon the introduction of exogenous antigens into the MHC Class I processing pathway (cross presentation) in specific pAPC populations. However, use of a pantropic viral vector that targets pAPC to express antigen endogenously likely represents a more effective vaccine strategy than the targeting of exogenous antigen to a limiting pAPC subpopulation.


Asunto(s)
Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Activación de Linfocitos/inmunología , Animales , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Macrófagos/inmunología , Ratones Endogámicos C57BL , Ratones Transgénicos , Péptidos/inmunología , Péptidos/metabolismo , Fenotipo
13.
PLoS Pathog ; 11(3): e1004770, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25822986

RESUMEN

Imatinib mesylate (Gleevec) inhibits Abl1, c-Kit, and related protein tyrosine kinases (PTKs) and serves as a therapeutic for chronic myelogenous leukemia and gastrointestinal stromal tumors. Imatinib also has efficacy against various pathogens, including pathogenic mycobacteria, where it decreases bacterial load in mice, albeit at doses below those used for treating cancer. We report that imatinib at such low doses unexpectedly induces differentiation of hematopoietic stem cells and progenitors in the bone marrow, augments myelopoiesis but not lymphopoiesis, and increases numbers of myeloid cells in blood and spleen. Whereas progenitor differentiation relies on partial inhibition of c-Kit by imatinib, lineage commitment depends upon inhibition of other PTKs. Thus, imatinib mimics "emergency hematopoiesis," a physiological innate immune response to infection. Increasing neutrophil numbers by adoptive transfer sufficed to reduce mycobacterial load, and imatinib reduced bacterial load of Franciscella spp., which do not utilize imatinib-sensitive PTKs for pathogenesis. Thus, potentiation of the immune response by imatinib at low doses may facilitate clearance of diverse microbial pathogens.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Francisella/inmunología , Infecciones por Bacterias Gramnegativas/inmunología , Mesilato de Imatinib/farmacología , Mielopoyesis/efectos de los fármacos , Neutrófilos/inmunología , Animales , Diferenciación Celular/inmunología , Recuento de Leucocitos , Ratones , Mielopoyesis/inmunología
14.
PLoS One ; 10(3): e0118685, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25751266

RESUMEN

Ectromelia virus (ECTV) causes mousepox in mice, a disease very similar to smallpox in humans. ECTV and variola virus (VARV), the agent of smallpox, are closely related orthopoxviruses. Mousepox is an excellent small animal model to study the genetic and immunologic basis for resistance and susceptibility of humans to smallpox. Resistance to mousepox is dependent on a strong polarized type 1 immune response, associated with robust natural killer (NK) cell, cytotoxic T lymphocyte (CTL) and gamma interferon (IFN-γ) responses. In contrast, ECTV-susceptible mice generate a type 2 response, associated with weak NK cell, CTL and IFN-γ responses but robust IL-4 responses. Nonetheless, susceptible strains infected with mutant ECTV lacking virus-encoded IFN-γ binding protein (vIFN-γbp) (ECTV-IFN-γbpΔ) control virus replication through generation of type 1 response. Since the IL-4/IL-13/STAT-6 signaling pathways polarize type 2/T helper 2 (Th2) responses with a corresponding suppression of IFN-γ production, we investigated whether the combined absence of vIFN-γbp, and one or more host genes involved in Th2 response development, influence generation of protective immunity. Most mutant mouse strains infected with wild-type (WT) virus succumbed to disease more rapidly than WT animals. Conversely, the disease outcome was significantly improved in WT mice infected with ECTV-IFN-γbpΔ but absence of IL-4/IL-13/STAT-6 signaling pathways did not provide any added advantage. Deficiency in IL-13 or STAT-6 resulted in defective CTL responses, higher mortality rates and accelerated deaths. Deficiencies in IL-4/IL-13/STAT-6 signaling pathways significantly reduced the numbers of IFN-γ producing CD4 and CD8 T cells, indicating an absence of a switch to a Th1-like response. Factors contributing to susceptibility or resistance to mousepox are far more complex than a balance between Th1 and Th2 responses.


Asunto(s)
Citocinas/genética , Virus de la Ectromelia/patogenicidad , Ectromelia Infecciosa/inmunología , Células Th2/metabolismo , Proteínas Virales/genética , Animales , Línea Celular , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Virus de la Ectromelia/genética , Virus de la Ectromelia/inmunología , Ectromelia Infecciosa/mortalidad , Ectromelia Infecciosa/virología , Técnicas de Silenciamiento del Gen , Interferón gamma/genética , Interferón gamma/metabolismo , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos BALB C , Transducción de Señal , Proteínas Virales/inmunología
15.
Antiviral Res ; 112: 38-46, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25453340

RESUMEN

There are no drugs approved specifically to treat disseminated adenovirus (Ad) infections in humans. Cidofovir is active against Ad in cell culture, and it is used frequently in the clinic with disseminated infection in pediatric transplant patients; however, controlled clinical studies have not been conducted to prove the anti-Ad efficacy of cidofovir. Brincidofovir, a lipid-linked derivative of cidofovir, which has strong activity against Ad in cell culture and in animal models, is a promising new drug currently in clinical trials. Ribavirin, which has modest activity against some Ad types in cell culture, has been used in the clinic against disseminated Ad, but the efficacy of ribavirin is unknown. In the current study, we have examined the activity of cidofovir, brincidofovir, and ribavirin against disseminated Ad5 infection in the immunosuppressed Syrian hamster model. Hamsters are immunosuppressed by treatment with cyclophosphamide, then infected intravenously with Ad5, leading to disseminated Ad5 infection, especially in the liver. We found that cidofovir and brincidofovir have excellent activity against Ad5 pathology and replication in the liver, even when administered therapeutically starting at 3 days post-challenge with Ad5. Ribavirin did not have anti-Ad5 activity in our model. Our data support the use of cidofovir and brincidofovir in humans for the treatment of disseminated Ad infections in humans.


Asunto(s)
Infecciones por Adenoviridae/tratamiento farmacológico , Infecciones por Adenoviridae/patología , Adenovirus Humanos/crecimiento & desarrollo , Citosina/análogos & derivados , Organofosfonatos/uso terapéutico , Ribavirina/uso terapéutico , Infecciones por Adenoviridae/virología , Alanina Transaminasa/sangre , Animales , Peso Corporal , Línea Celular , Cidofovir , Citosina/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Humanos , Huésped Inmunocomprometido , Hígado/patología , Hígado/virología , Mesocricetus , Análisis de Supervivencia , Resultado del Tratamiento , Carga Viral
16.
Antimicrob Agents Chemother ; 58(12): 7171-81, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25224011

RESUMEN

Adenovirus infections of immunocompromised patients can develop into deadly multiorgan or systemic disease. The virus is especially threatening for pediatric allogeneic hematopoietic stem cell transplant recipients; according to some studies, 10% or more of these patients succumb to disease resulting from adenovirus infection. At present, there is no drug approved for the treatment or prevention of adenovirus infections. Compounds that are approved to treat other virus infections are used off-label to combat adenovirus, but only anecdotal evidence of the efficacy of these drugs exists. Ganciclovir, a drug approved for the treatment of herpesvirus infection, was previously reported to be effective against human adenoviruses in vitro. To model adenovirus infections in immunocompromised humans, we examined ganciclovir's efficacy in immunosuppressed Syrian hamsters intravenously infected with type 5 human adenovirus (Ad5). This animal model is permissive for Ad5 replication, and the animals develop symptoms similar to those seen in humans. We demonstrate that ganciclovir suppresses Ad5 replication in the liver of infected hamsters and that it mitigates the consequences of Ad5 infections in these animals when administered prophylactically or therapeutically. We show that ganciclovir inhibits Ad5 DNA synthesis and late gene expression. The mechanism of action for the drug is not clear; preliminary data suggest that it exerts its antiadenoviral effect by directly inhibiting the adenoviral DNA polymerase. While more extensive studies are required, we believe that ganciclovir is a promising drug candidate to treat adenovirus infections. Brincidofovir, a drug with proven activity against Ad5, was used as a positive control in the prophylactic experiment.


Asunto(s)
Infecciones por Adenoviridae/tratamiento farmacológico , Adenovirus Humanos/efectos de los fármacos , Antivirales/farmacología , Ganciclovir/farmacología , Huésped Inmunocomprometido , Proteínas Virales/antagonistas & inhibidores , Infecciones por Adenoviridae/inmunología , Infecciones por Adenoviridae/mortalidad , Infecciones por Adenoviridae/virología , Adenovirus Humanos/genética , Adenovirus Humanos/crecimiento & desarrollo , Adenovirus Humanos/patogenicidad , Animales , Peso Corporal/efectos de los fármacos , Línea Celular Tumoral , Citosina/análogos & derivados , Citosina/farmacología , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Femenino , Expresión Génica , Humanos , Masculino , Mesocricetus , Organofosfonatos/farmacología , Análisis de Supervivencia , Transaminasas/sangre , Carga Viral/efectos de los fármacos , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral/efectos de los fármacos
17.
PLoS Pathog ; 10(8): e1004326, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25122471

RESUMEN

Poxviruses contain large dsDNA genomes encoding numerous open reading frames that manipulate cellular signalling pathways and interfere with the host immune response. The NF-κB signalling cascade is an important mediator of innate immunity and inflammation, and is tightly regulated by ubiquitination at several key points. A critical step in NF-κB activation is the ubiquitination and degradation of the inhibitor of kappaB (IκBα), by the cellular SCFß-TRCP ubiquitin ligase complex. We show here that upon stimulation with TNFα or IL-1ß, Orthopoxvirus-infected cells displayed an accumulation of phosphorylated IκBα, indicating that NF-κB activation was inhibited during poxvirus infection. Ectromelia virus is the causative agent of lethal mousepox, a natural disease that is fatal in mice. Previously, we identified a family of four ectromelia virus genes (EVM002, EVM005, EVM154 and EVM165) that contain N-terminal ankyrin repeats and C-terminal F-box domains that interact with the cellular SCF ubiquitin ligase complex. Since degradation of IκBα is catalyzed by the SCFß-TRCP ubiquitin ligase, we investigated the role of the ectromelia virus ankyrin/F-box protein, EVM005, in the regulation of NF-κB. Expression of Flag-EVM005 inhibited both TNFα- and IL-1ß-stimulated IκBα degradation and p65 nuclear translocation. Inhibition of the NF-κB pathway by EVM005 was dependent on the F-box domain, and interaction with the SCF complex. Additionally, ectromelia virus devoid of EVM005 was shown to inhibit NF-κB activation, despite lacking the EVM005 open reading frame. Finally, ectromelia virus devoid of EVM005 was attenuated in both A/NCR and C57BL/6 mouse models, indicating that EVM005 is required for virulence and immune regulation in vivo.


Asunto(s)
Virus de la Ectromelia/patogenicidad , Ectromelia Infecciosa/metabolismo , FN-kappa B/metabolismo , Proteínas Virales/metabolismo , Animales , Virus de la Ectromelia/inmunología , Virus de la Ectromelia/metabolismo , Ectromelia Infecciosa/inmunología , Citometría de Flujo , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , FN-kappa B/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Virales/inmunología , Virulencia/fisiología
18.
Antiviral Res ; 111: 42-52, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25128688

RESUMEN

Natural orthopoxvirus outbreaks such as vaccinia, cowpox, cattlepox and buffalopox continue to cause morbidity in the human population. Monkeypox virus remains a significant agent of morbidity and mortality in Africa. Furthermore, monkeypox virus's broad host-range and expanding environs make it of particular concern as an emerging human pathogen. Monkeypox virus and variola virus (the etiological agent of smallpox) are both potential agents of bioterrorism. The first line response to orthopoxvirus disease is through vaccination with first-generation and second-generation vaccines, such as Dryvax and ACAM2000. Although these vaccines provide excellent protection, their widespread use is impeded by the high level of adverse events associated with vaccination using live, attenuated virus. It is possible that vaccines could be used in combination with antiviral drugs to reduce the incidence and severity of vaccine-associated adverse events, or as a preventive in individuals with uncertain exposure status or contraindication to vaccination. We have used the intranasal mousepox (ectromelia) model to evaluate the efficacy of vaccination with Dryvax or ACAM2000 in conjunction with treatment using the broad spectrum antiviral, brincidofovir (BCV, CMX001). We found that co-treatment with BCV reduced the severity of vaccination-associated lesion development. Although the immune response to vaccination was quantifiably attenuated, vaccination combined with BCV treatment did not alter the development of full protective immunity, even when administered two days following ectromelia challenge. Studies with a non-replicating vaccine, ACAM3000 (MVA), confirmed that BCV's mechanism of attenuating the immune response following vaccination with live virus was, as expected, by limiting viral replication and not through inhibition of the immune system. These studies suggest that, in the setting of post-exposure prophylaxis, co-administration of BCV with vaccination should be considered a first response to a smallpox emergency in subjects of uncertain exposure status or as a means of reduction of the incidence and severity of vaccine-associated adverse events.


Asunto(s)
Antivirales/administración & dosificación , Citosina/análogos & derivados , Virus de la Ectromelia/fisiología , Ectromelia Infecciosa/prevención & control , Organofosfonatos/administración & dosificación , Vacuna contra Viruela/administración & dosificación , Animales , Citosina/administración & dosificación , Virus de la Ectromelia/inmunología , Ectromelia Infecciosa/inmunología , Ectromelia Infecciosa/virología , Femenino , Humanos , Inmunidad , Ratones , Ratones Endogámicos C57BL , Vacuna contra Viruela/inmunología , Vacunación , Replicación Viral
19.
Virology ; 456-457: 108-20, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24889230

RESUMEN

Currently, little is known about the ankyrin/F-box protein B4. Here, we report that B4R-null viruses exhibited reduced plaque size in tissue culture, and decreased ability to spread, as assessed by multiple-step growth analysis. Electron microscopy indicated that B4R-null viruses still formed mature and extracellular virions; however, there was a slight decrease of virions released into the media following deletion of B4R. Deletion of B4R did not affect the ability of the virus to rearrange actin; however, VACV811, a large vaccinia virus deletion mutant missing 55 open reading frames, had decreased ability to produce actin tails. Using ectromelia virus, a natural mouse pathogen, we demonstrated that virus devoid of EVM154, the B4R homolog, showed decreased spread to organs and was attenuated during infection. This initial characterization suggests that B4 may play a role in virus spread, and that other unidentified mediators of actin tail formation may exist in vaccinia virus.


Asunto(s)
Virus Vaccinia/crecimiento & desarrollo , Proteínas Virales/metabolismo , Estructuras Animales/virología , Animales , Virus de la Ectromelia/genética , Virus de la Ectromelia/patogenicidad , Ectromelia Infecciosa/patología , Ectromelia Infecciosa/virología , Femenino , Eliminación de Gen , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Virus Vaccinia/genética , Virus Vaccinia/ultraestructura , Carga Viral , Ensayo de Placa Viral , Proteínas Virales/genética , Virión/ultraestructura , Virulencia
20.
Virus Genes ; 48(1): 38-47, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24078045

RESUMEN

Poxviruses are dsDNA viruses with large genomes. Many genes in the genome remain uncharacterized, and recent studies have demonstrated that the poxvirus transcriptome includes numerous so-called anomalous transcripts not associated with open reading frames. Here, we characterize the expression and role of an apparently non-coding RNA in orthopoxviruses, which we call viral hairpin RNA (vhRNA). Using a bioinformatics approach, we predicted expression of a transcript not associated with an open reading frame that is likely to form a stem-loop structure due to the presence of a 21 nt palindromic sequence. Expression of the transcript as early as 2 h post-infection was confirmed by northern blot and analysis of publicly available vaccinia virus infected cell transcriptomes. The transcription start site was determined by RACE PCE and transcriptome analysis, and early and late promoter sequences were identified. Finally, to test the function of the transcript we generated an ectromelia virus knockout, which failed to form plaques in cell culture. The important role of the transcript in viral replication was further demonstrated using siRNA. Although the function of the transcript remains unknown, our work contributes to evidence of an increasingly complex poxvirus transcriptome, suggesting that transcripts such as vhRNA not associated with an annotated open reading frame can play an important role in viral replication.


Asunto(s)
Virus de la Ectromelia/crecimiento & desarrollo , Virus de la Ectromelia/genética , Regulación Viral de la Expresión Génica , Expresión Génica , ARN no Traducido/biosíntesis , Ensayo de Placa Viral , Animales , Northern Blotting , Línea Celular , Chlorocebus aethiops , Biología Computacional , Técnicas de Inactivación de Genes , Macaca mulatta , Conformación de Ácido Nucleico , Regiones Promotoras Genéticas , ARN no Traducido/genética , Sitio de Iniciación de la Transcripción , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...