Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 43(6): 1112-24, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26680205

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Matadoras Naturais/imunologia , Subfamília C de Receptores Semelhantes a Lectina de Células NK/imunologia , Infecções por Poxviridae/imunologia , Animais , Citotoxicidade Imunológica/imunologia , Modelos Animais de Doenças , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real
2.
Am J Physiol Heart Circ Physiol ; 323(6): H1091-H1107, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36269647

RESUMO

Many anticancer therapies cause serious cardiovascular complications that degrade quality of life and cause early mortality in treated patients. Specifically, doxorubicin is known as an effective anticancer agent that causes cardiomyopathy in treated patients. There has been growing interest in defining the role of endothelial cells in cardiac damage by doxorubicin. We have shown in the present study that endothelial nuclei accumulate more intravenously administered doxorubicin than other cardiac cell types. Doxorubicin enhanced cardiac production of the transforming growth factor-ß (TGF-ß) ligands and nuclear translocation of phospho-Smad3 in both cultured and in vivo cardiac endothelial cells. To examine the role of the TGF-ß/mothers against decapentaplegic homolog 3 (Smad3) pathway in cardiac damage by doxorubicin, we used both Smad3 shRNA stable endothelial cell lines and Smad3-knockout mice. We demonstrated using endothelial transcriptome analysis that upregulation of the TGF-ß and inflammatory cytokine/cytokine receptor pathways, as well as suppression of cell cycle and angiogenesis by doxorubicin, were alleviated in Smad3-deficient endothelial cells. The results of transcriptomic analysis were validated using qPCR, immunoblotting, and ex vivo aortic ring sprouting assays. Similarly, increased cardiac expression of cytokines and chemokines observed in treated wild-type mice was diminished in treated Smad3-knockout animals. We also detected increased end-diastolic diameter and depressed systolic function in doxorubicin-treated wild-type but not Smad3-knockout mice. This work provides evidence for the critical role of the canonical TGF-ß/Smad3 pathway in cardiac damage by doxorubicin.NEW & NOTEWORTHY Microvascular endothelial cells in the heart accumulate more intravenously administered doxorubicin than nonendothelial cardiac cell types. The treatment enhanced the TGF-ß/Smad3 pathway and elicited endothelial cell senescence and inflammatory responses followed by adverse cardiac remodeling and dysfunction in wild-type but not Smad3-deficient animals. Our study suggests that the TGF-ß/Smad3 pathway contributes to the development of doxorubicin cardiomyopathy and the potential value of novel approaches to ameliorate cardiotoxicity by targeting the Smad3 transcription factor.


Assuntos
Cardiomiopatias , Células Endoteliais , Camundongos , Animais , Células Endoteliais/metabolismo , Qualidade de Vida , Proteína Smad3/genética , Proteína Smad3/metabolismo , Doxorrubicina/toxicidade , Fator de Crescimento Transformador beta/metabolismo , Camundongos Knockout
3.
J Biol Chem ; 291(1): 447-61, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26546677

RESUMO

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.


Assuntos
Citocinas/imunologia , Fragmentos de Imunoglobulinas/imunologia , Receptores de Citocinas/imunologia , Transdução de Sinais , Antivirais/química , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Fragmentos de Imunoglobulinas/farmacologia , Interferon-alfa/farmacologia , Cinética , Mutação/genética , Fosforilação , Conformação Proteica , Receptor de Interferon alfa e beta/química , Receptor de Interferon alfa e beta/metabolismo , Reprodutibilidade dos Testes , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/efeitos dos fármacos
4.
J Mol Cell Cardiol ; 90: 129-38, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26686989

RESUMO

Elevated ALK4/5 ligands including TGF-ß and activins have been linked to cardiovascular remodeling and heart failure. Doxorubicin (Dox) is commonly used as a model of cardiomyopathy, a condition that often precedes cardiovascular remodeling and heart failure. In 7-8-week-old C57Bl/6 male mice treated with Dox we found decreased capillary density, increased levels of ALK4/5 ligand and Smad2/3 transcripts, and increased expression of Smad2/3 transcriptional targets. Human cardiac microvascular endothelial cells (HCMVEC) treated with Dox also showed increased levels of ALK4/5 ligands, Smad2/3 transcriptional targets, a decrease in proliferation and suppression of vascular network formation in a HCMVEC and human cardiac fibroblasts co-culture assay. Our hypothesis is that the deleterious effects of Dox on endothelial cells are mediated in part by the activation of the TGF-ß pathway. We used the inhibitor of ALK4/5 kinases SB431542 (SB) in concert with Dox to ascertain the role of TGF-ß pathway activation in doxorubicin induced endothelial cell defects. SB prevented the suppression of HCMVEC proliferation in the presence of TGF-ß2 and activin A, and alleviated the inhibition of HCMVEC proliferation by Dox. SB also prevented the suppression of vascular network formation in co-cultures of HCMVEC and human cardiac fibroblasts treated with Dox. Our results show that the inhibition of the TGF-ß pathway alleviates the detrimental effects of Dox on endothelial cells in vitro.


Assuntos
Doxorrubicina/farmacologia , Células Endoteliais/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fator de Crescimento Transformador beta2/farmacologia , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/metabolismo , Ativinas/genética , Ativinas/metabolismo , Ativinas/farmacologia , Animais , Benzamidas/farmacologia , Linhagem Celular , Técnicas de Cocultura , Dioxóis/farmacologia , Doxorrubicina/antagonistas & inibidores , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/citologia , Miocárdio/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad3/genética , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta2/antagonistas & inibidores , Fator de Crescimento Transformador beta2/genética , Fator de Crescimento Transformador beta2/metabolismo
5.
PLoS Pathog ; 10(8): e1004326, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25122471

RESUMO

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.


Assuntos
Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/metabolismo , NF-kappa B/metabolismo , Proteínas Virais/metabolismo , Animais , Vírus da Ectromelia/imunologia , Vírus da Ectromelia/metabolismo , Ectromelia Infecciosa/imunologia , Citometria de Fluxo , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , NF-kappa B/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Virais/imunologia , Virulência/fisiologia
6.
Nature ; 468(7322): 452-6, 2010 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-21085181

RESUMO

Cellular messenger RNA (mRNA) of higher eukaryotes and many viral RNAs are methylated at the N-7 and 2'-O positions of the 5' guanosine cap by specific nuclear and cytoplasmic methyltransferases (MTases), respectively. Whereas N-7 methylation is essential for RNA translation and stability, the function of 2'-O methylation has remained uncertain since its discovery 35 years ago. Here we show that a West Nile virus (WNV) mutant (E218A) that lacks 2'-O MTase activity was attenuated in wild-type primary cells and mice but was pathogenic in the absence of type I interferon (IFN) signalling. 2'-O methylation of viral RNA did not affect IFN induction in WNV-infected fibroblasts but instead modulated the antiviral effects of IFN-induced proteins with tetratricopeptide repeats (IFIT), which are interferon-stimulated genes (ISGs) implicated in regulation of protein translation. Poxvirus and coronavirus mutants that lacked 2'-O MTase activity similarly showed enhanced sensitivity to the antiviral actions of IFN and, specifically, IFIT proteins. Our results demonstrate that the 2'-O methylation of the 5' cap of viral RNA functions to subvert innate host antiviral responses through escape of IFIT-mediated suppression, and suggest an evolutionary explanation for 2'-O methylation of cellular mRNA: to distinguish self from non-self RNA. Differential methylation of cytoplasmic RNA probably serves as an example for pattern recognition and restriction of propagation of foreign viral RNA in host cells.


Assuntos
Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica/imunologia , Imunidade Inata/imunologia , Interferons/imunologia , Proteínas/metabolismo , Capuzes de RNA/metabolismo , RNA Viral/metabolismo , Células 3T3 , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/genética , Células Cultivadas , Coronavirus/enzimologia , Coronavirus/genética , Coronavirus/imunologia , Coronavirus/fisiologia , Fibroblastos , Regulação da Expressão Gênica/genética , Humanos , Imunidade Inata/genética , Interferons/deficiência , Interferons/genética , Metilação , Metiltransferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Genéticos , Modelos Imunológicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Poxviridae/enzimologia , Poxviridae/genética , Poxviridae/imunologia , Poxviridae/fisiologia , Biossíntese de Proteínas/imunologia , Proteínas/genética , Capuzes de RNA/genética , Capuzes de RNA/imunologia , RNA Viral/genética , RNA Viral/imunologia , Proteínas de Ligação a RNA , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Taxa de Sobrevida , Replicação Viral , Vírus do Nilo Ocidental/enzimologia , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/fisiologia
7.
J Virol ; 87(9): 4846-60, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23408632

RESUMO

Rift Valley fever virus (RVFV) is an emerging RNA virus with devastating economic and social consequences. Clinically, RVFV induces a gamut of symptoms ranging from febrile illness to retinitis, hepatic necrosis, hemorrhagic fever, and death. It is known that type I interferon (IFN) responses can be protective against severe pathology; however, it is unknown which innate immune receptor pathways are crucial for mounting this response. Using both in vitro assays and in vivo mucosal mouse challenge, we demonstrate here that RNA helicases are critical for IFN production by immune cells and that signaling through the helicase adaptor molecule MAVS (mitochondrial antiviral signaling) is protective against mortality and more subtle pathology during RVFV infection. In addition, we demonstrate that Toll-like-receptor-mediated signaling is not involved in IFN production, further emphasizing the importance of the RNA cellular helicases in type I IFN responses to RVFV.


Assuntos
RNA Helicases DEAD-box/imunologia , Interferon beta/imunologia , Mucosa/virologia , Febre do Vale de Rift/enzimologia , Febre do Vale de Rift/imunologia , Vírus da Febre do Vale do Rift/fisiologia , Animais , Linhagem Celular , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , Células Dendríticas/imunologia , Células Dendríticas/virologia , Feminino , Humanos , Interferon beta/genética , Macrófagos/imunologia , Macrófagos/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout/genética , Mucosa/imunologia , Febre do Vale de Rift/prevenção & controle , Febre do Vale de Rift/virologia , Transdução de Sinais , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia
8.
Virus Genes ; 48(1): 38-47, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24078045

RESUMO

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.


Assuntos
Vírus da Ectromelia/crescimento & desenvolvimento , Vírus da Ectromelia/genética , Regulação Viral da Expressão Gênica , Expressão Gênica , RNA não Traduzido/biossíntese , Ensaio de Placa Viral , Animais , Northern Blotting , Linhagem Celular , Chlorocebus aethiops , Biologia Computacional , Técnicas de Inativação de Genes , Macaca mulatta , Conformação de Ácido Nucleico , Regiões Promotoras Genéticas , RNA não Traduzido/genética , Sítio de Iniciação de Transcrição , Transcrição Gênica
9.
Appl Environ Microbiol ; 79(4): 1333-41, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23263945

RESUMO

Protection of the human lung from infectious agents, allergens, and ultrafine particles is difficult with current technologies. High-efficiency particulate air (HEPA) filters remove airborne particles of >0.3 µm with 99.97% efficiency, but they are expensive to maintain. Electrostatic precipitation has been used as an inexpensive approach to remove large particles from airflows, but it has a collection efficiency minimum in the submicrometer size range, allowing for a penetration window for some allergens and ultrafine particles. Incorporating soft X-ray irradiation as an in situ component of the electrostatic precipitation process greatly improves capture efficiency of ultrafine particles. Here we demonstrate the removal and inactivation capabilities of soft-X-ray-enhanced electrostatic precipitation technology targeting infectious agents (Bacillus anthracis, Mycobacterium bovis BCG, and poxviruses), allergens, and ultrafine particles. Incorporation of in situ soft X-ray irradiation at low-intensity corona conditions resulted in (i) 2-fold to 9-fold increase in capture efficiency of 200- to 600-nm particles and (ii) a considerable delay in the mean day of death as well as lower overall mortality rates in ectromelia virus (ECTV) cohorts. At the high-intensity corona conditions, nearly complete protection from viral and bacterial respiratory infection was afforded to the murine models for all biological agents tested. When optimized for combined efficient particle removal with limited ozone production, this technology could be incorporated into stand-alone indoor air cleaners or scaled for installation in aircraft cabin, office, and residential heating, ventilating, and air-conditioning (HVAC) systems.


Assuntos
Microbiologia do Ar , Alérgenos/efeitos da radiação , Bactérias/efeitos da radiação , Precipitação Química , Material Particulado/efeitos da radiação , Vírus/efeitos da radiação , Raios X , Alérgenos/química , Animais , Bactérias/química , Modelos Animais de Doenças , Camundongos , Material Particulado/química , Infecções Respiratórias/microbiologia , Infecções Respiratórias/prevenção & controle , Infecções Respiratórias/virologia , Eletricidade Estática , Vírus/química
10.
J Virol ; 84(8): 3909-20, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20130052

RESUMO

Monkeypox virus (MPXV) is an orthopoxvirus closely related to variola, the etiological agent of smallpox. In humans, MPXV causes a disease similar to smallpox and is considered to be an emerging infectious disease. Moreover, the use of MPXV for bioterroristic/biowarfare activities is of significant concern. Available small animal models of human monkeypox have been restricted to mammals with poorly defined biologies that also have limited reagent availability. We have established a murine MPXV model utilizing the STAT1-deficient C57BL/6 mouse. Here we report that a relatively low-dose intranasal (IN) infection induces 100% mortality in the stat1(-)(/)(-) model by day 10 postinfection with high infectious titers in the livers, spleens, and lungs of moribund animals. Vaccination with modified vaccinia virus Ankara (MVA) followed by a booster vaccination is sufficient to protect against an intranasal MPXV challenge and induces an immune response more robust than that of a single vaccination. Furthermore, antiviral treatment with CMX001 (HDP-cidofovir) and ST-246 protects when administered as a regimen initiated on the day of infection. Thus, the stat1(-)(/)(-) model provides a lethal murine platform for evaluating therapeutics and for investigating the immunological and pathological responses to MPXV infection.


Assuntos
Modelos Animais de Doenças , Monkeypox virus/efeitos dos fármacos , Monkeypox virus/patogenicidade , Mpox/tratamento farmacológico , Mpox/prevenção & controle , Animais , Antivirais/uso terapêutico , Benzamidas , Citosina/análogos & derivados , Citosina/uso terapêutico , Feminino , Humanos , Isoindóis , Fígado/virologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mpox/mortalidade , Organofosfonatos/uso terapêutico , Fator de Transcrição STAT1/deficiência , Vacina Antivariólica/imunologia , Baço/virologia , Análise de Sobrevida , Resultado do Tratamento , Vaccinia virus/imunologia , Carga Viral
11.
Proc Natl Acad Sci U S A ; 105(6): 1861-6, 2008 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-18252829

RESUMO

Ectromelia virus (ECTV) encodes an IFN-gamma-binding protein (IFN-gammaBP(ECTV)) that disrupts IFN-gamma signaling and its ability to induce an antiviral state within cells. IFN-gammaBP(ECTV) is an important virulence factor that is highly conserved (>90%) in all orthopoxviruses, including variola virus, the causative agent of smallpox. The 2.2-A crystal structure of the IFN-gammaBP(ECTV)/IFN-gamma complex reveals IFN-gammaBP(ECTV) consists of an IFN-gammaR1 ligand-binding domain and a 57-aa helix-turn-helix (HTH) motif that is structurally related to the transcription factor TFIIA. The HTH motif forms a tetramerization domain that results in an IFN-gammaBP(ECTV)/IFN-gamma complex containing four IFN-gammaBP(ECTV) chains and two IFN-gamma dimers. The structure, combined with biochemical and cell-based assays, demonstrates that IFN-gammaBP(ECTV) tetramers are required for efficient IFN-gamma antagonism.


Assuntos
Interferon gama/antagonistas & inibidores , Orthopoxvirus/metabolismo , Proteínas Virais/metabolismo , Animais , Cromatografia de Afinidade , Ligação de Hidrogênio , Interferon gama/metabolismo , Camundongos , Ligação Proteica , Conformação Proteica , Proteínas Virais/química , Proteínas Virais/isolamento & purificação
12.
Vaccine ; 37(36): 5371-5381, 2019 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-31331771

RESUMO

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.


Assuntos
Vacinas contra Influenza/uso terapêutico , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Animais , Biologia Computacional , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/metabolismo , Feminino , Humanos , Imunidade Celular/imunologia , Imunidade Celular/fisiologia , Vírus da Influenza A/imunologia , Vírus da Influenza A/patogenicidade , Masculino , Camundongos , Linfócitos T/imunologia , Linfócitos T/metabolismo
13.
Antivir Ther ; 13(7): 863-73, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19043920

RESUMO

BACKGROUND: The emergence of human monkeypox and the potential use of recombinant variola and monkeypox viruses as biological terrorist agents have necessitated the development of therapeutic and prophylactic therapies. The primary, or index, cases of smallpox and/or human monkeypox will likely be identified by a characteristic rash. Effective biomarkers will be required to monitor disease progression, guide the choice of therapeutic intervention strategies and evaluate their efficacies. To address this we have evaluated several biomarkers of disease in a lethal mousepox model. METHODS: The efficacy of a single dose of a hexadecyloxypropyl ester of cidofovir (CMX001) at 20, 25 and 30 mg/kg doses administered on days 4, 5, 6 and 7 post-infection was evaluated in A/Ncr mice intranasally infected with low doses of ectromelia virus (<20 plaque-forming units). Mice were monitored for weight loss, blood interferon-gamma levels, alanine aminotransferase (ALT), aspartate aminotransferase, viral DNA copies and neutrophilia levels to stage disease progression. RESULTS: We have used these biomarkers to establish the optimal dosing regimen for treatment and reveal that a single dose of 25 mg/kg of CMX001 can be efficacious at treating lethal mousepox when administered on days 4 or 5 post-infection. This dose significantly reduces ALT, interferon-gamma and DNA copies found in the blood of infected animals. CONCLUSIONS: A single dose regimen of CMX001 is efficacious at treating mousepox. Disease progression and antiviral efficacy can be monitored using several biomarkers that could readily be used in the case of a human monkeypox or smallpox outbreak.


Assuntos
Antivirais/uso terapêutico , Citosina/análogos & derivados , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/tratamento farmacológico , Ectromelia Infecciosa/fisiopatologia , Organofosfonatos/uso terapêutico , Alanina Transaminase/sangue , Animais , Antivirais/administração & dosagem , Aspartato Aminotransferases/sangue , Biomarcadores/análise , Linhagem Celular , Citosina/administração & dosagem , Citosina/uso terapêutico , DNA Viral/sangue , Modelos Animais de Doenças , Progressão da Doença , Ectromelia Infecciosa/virologia , Feminino , Humanos , Interferon gama/sangue , Camundongos , Organofosfonatos/administração & dosagem , Resultado do Tratamento , Redução de Peso
14.
Antiviral Res ; 73(3): 212-8, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17123638

RESUMO

Hexadecyloxypropyl-cidofovir (HDP-CDV) has been shown to be orally active against lethal infection with orthopoxviruses including, mousepox, cowpox, vaccinia and rabbitpox. The alkoxyalkyl group provides oral absorption and reduces greatly the amount of drug reaching the kidney, the site of CDV's dose limiting toxicity. However, the amount of HDP-CDV detected in lung, an important site of early poxvirus replication, is low and the reduction of viral titers in surviving animals is reduced moderately compared with the liver where poxvirus titers are virtually undetectable. We synthesized a novel glycerol ester of CDV, 1-O-octadecyl-2-O-benzyl-sn-glycero-3-CDV (ODBG-CDV), and compared its oral pharmacokinetics with that of HDP-CDV. Surprisingly, ODBG-CDV levels in lung are much higher and liver levels are reduced, suggesting that the compound is transported in small intestinal lymph instead the portal vein. ODBG-CDV has excellent in vitro activity in cells infected with ectromelia virus (ECTV). In mice infected with a lethal aerosol or intranasal challenge of ECTV, HDP-CDV and ODBG-CDV are equally effective in preventing death from disease. Other drugs esterified to 1-O-octadecyl-2-O-benzyl-sn-glycerol or 1-O-octadecyl-2-O-benzyl-sn-glycerol-3-phosphate may provide lung targeting for treatment of microbial or neoplastic diseases while reducing first pass removal by the liver during oral absorption.


Assuntos
Citosina/análogos & derivados , Vírus da Ectromelia/efeitos dos fármacos , Ectromelia Infecciosa/tratamento farmacológico , Pulmão/metabolismo , Organofosfonatos/farmacologia , Organofosfonatos/farmacocinética , Infecções Respiratórias/tratamento farmacológico , Administração Oral , Animais , Cidofovir , Citosina/farmacocinética , Citosina/farmacologia , Ectromelia Infecciosa/metabolismo , Ectromelia Infecciosa/virologia , Feminino , Fígado/metabolismo , Camundongos , Infecções Respiratórias/metabolismo , Infecções Respiratórias/virologia
15.
Methods Mol Biol ; 269: 289-308, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15114022

RESUMO

Concern regarding the use of variola and monkeypox viruses as bioterrorist agents has led to an increased study of orthopoxviruses to understand the molecular and cellular basis of pathogenesis and develop safe and effective antivirals and vaccines against smallpox. Crucial to these efforts is the availability of animal models, which are inexpensive, genetically homogeneous, and recapitulate the human disease. The popular small-animal orthopoxvirus models employ the inbred mouse as the host, the respiratory tract as the site of virus inoculation, and orthopoxviruses-vaccinia, cowpox, and ectromelia viruses-as surrogates for variola virus. Ectromelia virus is likely the best surrogate for variola virus in a mouse model, as it is infectious at very low doses of virus, and the mousepox disease is associated with high mortality in the susceptible A, BALB/c, and DBA/2 stains of mice, but causes an unapparent infection in the C57BL/6 mouse strain. This chapter describes an ectromelia virus respiratory infection model in the mouse.


Assuntos
Modelos Animais de Doenças , Orthopoxvirus/imunologia , Infecções por Poxviridae/imunologia , Infecções Respiratórias/virologia , Animais , Orthopoxvirus/genética , Orthopoxvirus/fisiologia , Infecções por Poxviridae/prevenção & controle , Infecções por Poxviridae/transmissão , Infecções Respiratórias/imunologia
16.
Microbes Infect ; 16(1): 73-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24120457

RESUMO

A better understanding of mucosal immunity is required to develop more protective vaccines against Mycobacterium tuberculosis. We developed a murine aerosol challenge model to investigate responses capable of protecting against mucosal infection. Mice received vaccinations intranasally with CpG-adjuvanted antigen 85B (Ag85B/CpG) and/or Bacillus Calmette-Guerin (BCG). Protection against aerosol challenge with a recombinant GFP-expressing BCG was assessed. Mucosal prime/boost vaccinations with Ag85B/CpG and BCG were protective, but did not prevent lung infection indicating more efficacious mucosal vaccines are needed. Our novel finding that protection correlated with increased airway dendritic cells early post-challenge could help guide the development of enhanced mucosal vaccines.


Assuntos
Imunidade nas Mucosas , Mycobacterium tuberculosis/imunologia , Vacinas contra a Tuberculose/imunologia , Tuberculose/prevenção & controle , Animais , Vacina BCG/administração & dosagem , Vacina BCG/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Camundongos , Mucosa Respiratória/imunologia , Mucosa Respiratória/microbiologia
17.
Antiviral Res ; 111: 42-52, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25128688

RESUMO

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.


Assuntos
Antivirais/administração & dosagem , Citosina/análogos & derivados , Vírus da Ectromelia/fisiologia , Ectromelia Infecciosa/prevenção & controle , Organofosfonatos/administração & dosagem , Vacina Antivariólica/administração & dosagem , Animais , Citosina/administração & dosagem , Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/imunologia , Ectromelia Infecciosa/virologia , Feminino , Humanos , Imunidade , Camundongos , Camundongos Endogâmicos C57BL , Vacina Antivariólica/imunologia , Vacinação , Replicação Viral
18.
Virology ; 456-457: 108-20, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24889230

RESUMO

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.


Assuntos
Vaccinia virus/crescimento & desenvolvimento , Proteínas Virais/metabolismo , Estruturas Animais/virologia , Animais , Vírus da Ectromelia/genética , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/patologia , Ectromelia Infecciosa/virologia , Feminino , Deleção de Genes , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Vaccinia virus/genética , Vaccinia virus/ultraestrutura , Carga Viral , Ensaio de Placa Viral , Proteínas Virais/genética , Vírion/ultraestrutura , Virulência
19.
PLoS One ; 8(2): e52408, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23383295

RESUMO

Autoimmune destruction of insulin producing pancreatic ß-cells is the hallmark of type I diabetes. One of the key molecules implicated in the disease onset is the immunoproteasome, a protease with multiple proteolytic sites that collaborates with the constitutive 19S and the inducible 11S (PA28) activators to produce immunogenic peptides for presentation by MHC class I molecules. Despite its importance, little is known about the function and regulation of the immunoproteasome in pancreatic ß-cells. Of special interest to immunoproteasome activation in ß-cells are the effects of IFNß, a type I IFN secreted by virus-infected cells and implicated in type I diabetes onset, compared to IFNγ, the classic immunoproteasome inducer secreted by cells of the immune system. By qPCR analysis, we show that mouse insulinoma MIN6 cells and mouse islets accumulate the immune proteolytic ß1(i), ß2(i) and ß5(i), and 11S mRNAs upon exposure to IFNß or IFNγ. Higher concentrations of IFNß than IFNγ are needed for similar expression, but in each case the expression is transient, with maximal mRNA accumulation in 12 hours, and depends primarily on Interferon Regulatory Factor 1. IFNs do not alter expression of regular proteasome genes, and in the time frame of IFNß-mediated response, the immune and regular proteolytic subunits co-exist in the 20S particles. In cell extracts with ATP, these particles have normal peptidase activities and degrade polyubiquitinated proteins with rates typical of the regular proteasome, implicating normal regulation by the 19S activator. However, ATP depletion rapidly stimulates the catalytic rates in a manner consistent with levels of the 11S activator. These findings suggest that stochastic combination of regular and immune proteolytic subunits may increase the probability with which unique immunogenic peptides are produced in pancreatic ß-cells exposed to IFNß, but primarily in cells with reduced ATP levels that stimulate the 11S participation in immunoproteasome function.


Assuntos
Trifosfato de Adenosina/metabolismo , Células Secretoras de Insulina/imunologia , Interferon beta/metabolismo , Proteínas Musculares/metabolismo , Complexo de Endopeptidases do Proteassoma/imunologia , Animais , Western Blotting , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Primers do DNA/genética , Imunoprecipitação , Células Secretoras de Insulina/virologia , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Octoxinol , Reação em Cadeia da Polimerase , Complexo de Endopeptidases do Proteassoma/metabolismo
20.
PLoS One ; 8(10): e77879, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24147092

RESUMO

Post-exposure vaccination with vaccinia virus (VACV) has been suggested to be effective in minimizing death if administered within four days of smallpox exposure. While there is anecdotal evidence for efficacy of post-exposure vaccination this has not been definitively studied in humans. In this study, we analyzed post-exposure prophylaxis using several attenuated recombinant VACV in a mouse model. A recombinant VACV expressing murine interferon gamma (IFN-γ) was most effective for post-exposure protection of mice infected with VACV and ectromelia virus (ECTV). Untreated animals infected with VACV exhibited severe weight loss and morbidity leading to 100% mortality by 8 to 10 days post-infection. Animals treated one day post-infection had milder symptoms, decreased weight loss and morbidity, and 100% survival. Treatment on days 2 or 3 post-infection resulted in 40% and 20% survival, respectively. Similar results were seen in ECTV-infected mice. Despite the differences in survival rates in the VACV model, the viral load was similar in both treated and untreated mice while treated mice displayed a high level of IFN-γ in the serum. These results suggest that protection provided by IFN-γ expressed by VACV may be mediated by its immunoregulatory activities rather than its antiviral effects. These results highlight the importance of IFN-γ as a modulator of the immune response for post-exposure prophylaxis and could be used potentially as another post-exposure prophylaxis tool to prevent morbidity following infection with smallpox and other orthopoxviruses.


Assuntos
Vírus da Ectromelia/imunologia , Vaccinia virus/metabolismo , Vacínia/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Modelos Animais de Doenças , Feminino , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Vacínia/imunologia , Vaccinia virus/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA