Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 9(2): e1003168, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23544010

RESUMEN

The actions of the RIG-I like receptor (RLR) and type I interferon (IFN) signaling pathways are essential for a protective innate immune response against the emerging flavivirus West Nile virus (WNV). In mice lacking RLR or IFN signaling pathways, WNV exhibits enhanced tissue tropism, indicating that specific host factors of innate immune defense restrict WNV infection and dissemination in peripheral tissues. However, the immune mechanisms by which the RLR and IFN pathways coordinate and function to impart restriction of WNV infection are not well defined. Using a systems biology approach, we defined the host innate immune response signature and actions that restrict WNV tissue tropism. Transcriptional profiling and pathway modeling to compare WNV-infected permissive (spleen) and nonpermissive (liver) tissues showed high enrichment for inflammatory responses, including pattern recognition receptors and IFN signaling pathways, that define restriction of WNV replication in the liver. Assessment of infected livers from Mavs(-/-) × Ifnar(-/-) mice revealed the loss of expression of several key components within the natural killer (NK) cell signaling pathway, including genes associated with NK cell activation, inflammatory cytokine production, and NK cell receptor signaling. In vivo analysis of hepatic immune cell infiltrates from WT mice demonstrated that WNV infection leads to an increase in NK cell numbers with enhanced proliferation, maturation, and effector action. In contrast, livers from Mavs(-/-) × Ifnar(-/-) infected mice displayed reduced immune cell infiltration, including a significant reduction in NK cell numbers. Analysis of cocultures of dendritic and NK cells revealed both cell-intrinsic and -extrinsic roles for the RLR and IFN signaling pathways to regulate NK cell effector activity. Taken together, these observations reveal a complex innate immune signaling network, regulated by the RLR and IFN signaling pathways, that drives tissue-specific antiviral effector gene expression and innate immune cellular processes that control tissue tropism to WNV infection.


Asunto(s)
Inmunidad Celular/genética , Inmunidad Innata/genética , Tropismo Viral/genética , Fiebre del Nilo Occidental/inmunología , Virus del Nilo Occidental/inmunología , Virus del Nilo Occidental/fisiología , Animales , Redes Reguladoras de Genes/inmunología , Genes/fisiología , Humanos , Interferón Tipo I/metabolismo , Interferón Tipo I/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/fisiología , Transducción de Señal/genética , Transducción de Señal/inmunología , Biología de Sistemas/métodos , Fiebre del Nilo Occidental/genética
2.
J Virol ; 86(13): 7192-206, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22532695

RESUMEN

While pandemic 2009 H1N1 influenza A viruses were responsible for numerous severe infections in humans, these viruses do not typically cause corresponding severe disease in mammalian models. However, the generation of a virulent 2009 H1N1 virus following serial lung passage in mice has allowed for the modeling of human lung pathology in this species. Genetic determinants of mouse-adapted 2009 H1N1 viral pathogenicity have been identified, but the molecular and signaling characteristics of the host response following infection with this adapted virus have not been described. Here we compared the gene expression response following infection of mice with A/CA/04/2009 (CA/04) or the virulent mouse-adapted strain (MA-CA/04). Microarray analysis revealed that increased pathogenicity of MA-CA/04 was associated with the following: (i) an early and sustained inflammatory and interferon response that could be driven in part by interferon regulatory factors (IRFs) and increased NF-κB activation, as well as inhibition of the negative regulator TRIM24, (ii) early and persistent infiltration of immune cells, including inflammatory macrophages, and (iii) the absence of activation of lipid metabolism later in infection, which may be mediated by inhibition of nuclear receptors, including PPARG and HNF1A and -4A, with proinflammatory consequences. Further investigation of these signatures in the host response to other H1N1 viruses of various pathogenicities confirmed their general relevance for virulence of influenza virus and suggested that lung response to MA-CA/04 virus was similar to that following infection with lethal H1N1 r1918 influenza virus. This study links differential activation of IRFs, nuclear receptors, and macrophage infiltration with influenza virulence in vivo.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Factores Reguladores del Interferón/inmunología , Macrófagos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Receptores Citoplasmáticos y Nucleares/inmunología , Adaptación Biológica , Animales , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Análisis por Micromatrices , Infecciones por Orthomyxoviridae/patología , Pase Seriado , Virulencia
3.
Hepatology ; 56(1): 28-38, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22331615

RESUMEN

UNLABELLED: Liver transplant tissues offer the unique opportunity to model the longitudinal protein abundance changes occurring during hepatitis C virus (HCV)-associated liver disease progression in vivo. In this study, our goal was to identify molecular signatures, and potential key regulatory proteins, representative of the processes influencing early progression to fibrosis. We performed global protein profiling analyses on 24 liver biopsy specimens obtained from 15 HCV(+) liver transplant recipients at 6 and/or 12 months posttransplantation. Differentially regulated proteins associated with early progression to fibrosis were identified by analysis of the area under the receiver operating characteristic curve. Analysis of serum metabolites was performed on samples obtained from an independent cohort of 60 HCV(+) liver transplant patients. Computational modeling approaches were applied to identify potential key regulatory proteins of liver fibrogenesis. Among 4,324 proteins identified, 250 exhibited significant differential regulation in patients with rapidly progressive fibrosis. Patients with rapid fibrosis progression exhibited enrichment in differentially regulated proteins associated with various immune, hepatoprotective, and fibrogenic processes. The observed increase in proinflammatory activity and impairment in antioxidant defenses suggests that patients who develop significant liver injury experience elevated oxidative stresses. This was supported by an independent study demonstrating the altered abundance of oxidative stress-associated serum metabolites in patients who develop severe liver injury. Computational modeling approaches further highlight a potentially important link between HCV-associated oxidative stress and epigenetic regulatory mechanisms impacting on liver fibrogenesis. CONCLUSION: Our proteome and metabolome analyses provide new insights into the role for increased oxidative stress in the rapid fibrosis progression observed in HCV(+) liver transplant recipients. These findings may prove useful in prognostic applications for predicting early progression to fibrosis.


Asunto(s)
Hepacivirus/metabolismo , Hepatitis C/complicaciones , Cirrosis Hepática/patología , Trasplante de Hígado/patología , Análisis por Matrices de Proteínas/métodos , Proteoma/metabolismo , Adulto , Anciano , Biopsia con Aguja , Cromatografía Liquida/métodos , Estudios de Cohortes , Diagnóstico por Computador/métodos , Progresión de la Enfermedad , Femenino , Rechazo de Injerto , Supervivencia de Injerto , Hepacivirus/patogenicidad , Hepatitis C/patología , Humanos , Inmunohistoquímica , Cirrosis Hepática/etiología , Cirrosis Hepática/cirugía , Trasplante de Hígado/efectos adversos , Masculino , Espectrometría de Masas/métodos , Persona de Mediana Edad , Estrés Oxidativo/fisiología , Proteoma/genética , Proteómica/métodos , Recurrencia , Valores de Referencia , Medición de Riesgo , Muestreo , Sensibilidad y Especificidad
4.
BMC Genomics ; 13: 627, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23153050

RESUMEN

BACKGROUND: The 2009 pandemic H1N1 influenza virus emerged in swine and quickly became a major global health threat. In mouse, non human primate, and swine infection models, the pH1N1 virus efficiently replicates in the lung and induces pro-inflammatory host responses; however, whether similar or different cellular pathways were impacted by pH1N1 virus across independent infection models remains to be further defined. To address this we have performed a comparative transcriptomic analysis of acute phase responses to a single pH1N1 influenza virus, A/California/04/2009 (CA04), in the lung of mice, macaques and swine. RESULTS: Despite similarities in the clinical course, we observed differences in inflammatory molecules elicited, and the kinetics of their gene expression changes across all three species. We found genes associated with the retinoid X receptor (RXR) signaling pathway known to control pro-inflammatory and metabolic processes that were differentially regulated during infection in each species, though the heterodimeric RXR partner, pathway associated signaling molecules, and gene expression patterns varied among the three species. CONCLUSIONS: By comparing transcriptional changes in the context of clinical and virological measures, we identified differences in the host transcriptional response to pH1N1 virus across independent models of acute infection. Antiviral resistance and the emergence of new influenza viruses have placed more focus on developing drugs that target the immune system. Underlying overt clinical disease are molecular events that suggest therapeutic targets identified in one host may not be appropriate in another.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/fisiología , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/veterinaria , Transcriptoma , Animales , Femenino , Perfilación de la Expresión Génica , Especificidad del Huésped , Interacciones Huésped-Patógeno , Pulmón/patología , Pulmón/virología , Macaca , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/virología , Pandemias , Multimerización de Proteína , Receptores X Retinoide/genética , Receptores X Retinoide/metabolismo , Transducción de Señal , Porcinos
5.
J Virol ; 85(22): 11626-37, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21900171

RESUMEN

There exists limited information about whether adaptation is needed for cross-species transmission of the 2009 pandemic H1N1 influenza virus (pH1N1). Here, we compare the pathogenesis of two pH1N1 viruses, one derived from a human patient (A/CA/04/09 [CA09]) and the other from swine (A/swine/Alberta/25/2009 [Alb09]), with that of the 1918-like classical swine influenza virus (A/swine/Iowa/1930 [IA30]) in the pig model. Both pH1N1 isolates induced clinical symptoms such as coughing, sneezing, decreased activity, fever, and labored breathing in challenged pigs, but IA30 virus did not cause any clinical symptoms except fever. Although both the pH1N1 viruses and the IA30 virus caused lung lesions, the pH1N1 viruses were shed from the nasal cavities of challenged pigs whereas the IA30 virus was not. Global gene expression analysis indicated that transcriptional responses of the viruses were distinct. pH1N1-infected pigs had an upregulation of genes related to inflammatory and immune responses at day 3 postinfection that was not seen in the IA30 infection, and expression levels of genes related to cell death and lipid metabolism at day 5 postinfection were markedly different from those of IA30 infection. These results indicate that both pH1N1 isolates are more virulent due in part to differences in the host transcriptional response during acute infection. Our study also indicates that pH1N1 does not need prior adaptation to infect pigs, has a high potential to be maintained in naïve swine populations, and might reassort with currently circulating swine influenza viruses.


Asunto(s)
Muerte Celular , Regulación de la Expresión Génica , Inflamación/patología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Metabolismo de los Lípidos , Redes y Vías Metabólicas/genética , Animales , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Inflamación/virología , Pulmón/patología , Pulmón/virología , Datos de Secuencia Molecular , Mucosa Nasal/virología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Análisis de Secuencia de ADN , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/patología , Enfermedades de los Porcinos/virología , Esparcimiento de Virus
6.
J Virol ; 85(22): 11646-54, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21917952

RESUMEN

We previously employed systems biology approaches to identify the mitochondrial fatty acid oxidation enzyme dodecenoyl coenzyme A delta isomerase (DCI) as a bottleneck protein controlling host metabolic reprogramming during hepatitis C virus (HCV) infection. Here we present the results of studies confirming the importance of DCI to HCV pathogenesis. Computational models incorporating proteomic data from HCV patient liver biopsy specimens recapitulated our original predictions regarding DCI and link HCV-associated alterations in cellular metabolism and liver disease progression. HCV growth and RNA replication in hepatoma cell lines stably expressing DCI-targeting short hairpin RNA (shRNA) were abrogated, indicating that DCI is required for productive infection. Pharmacologic inhibition of fatty acid oxidation also blocked HCV replication. Production of infectious HCV was restored by overexpression of an shRNA-resistant DCI allele. These findings demonstrate the utility of systems biology approaches to gain novel insight into the biology of HCV infection and identify novel, translationally relevant therapeutic targets.


Asunto(s)
Isomerasas de Doble Vínculo Carbono-Carbono/metabolismo , Hepacivirus/patogenicidad , Interacciones Huésped-Patógeno , Mitocondrias/enzimología , Replicación Viral , Biopsia , Línea Celular , Dodecenoil-CoA Isomerasa , Ácidos Grasos/metabolismo , Silenciador del Gen , Hepatocitos/enzimología , Hepatocitos/virología , Humanos , Hígado/química , Hígado/patología , Oxidación-Reducción , Proteoma
7.
J Virol ; 85(3): 1214-23, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21084481

RESUMEN

The first influenza pandemic of the new millennium was caused by a newly emerged swine-origin influenza virus (SOIV) (H1N1). This new virus is characterized by a previously unknown constellation of gene segments derived from North American and Eurasian swine lineages and the absence of common markers predictive of human adaptation. Overall, human infections appeared to be mild, but an alarming number of young individuals presented with symptoms atypical for seasonal influenza. The new SOIV also showed a sustained human-to-human transmissibility and higher reproduction ratio than common seasonal viruses, altogether indicating a higher pathogenic potential for this newly emerged virus. To study the virulence of the SOIV, we used a recently established cynomolgus macaque model and compared parameters of clinical disease, virology, host responses, and pathology/histopathology with a current seasonal H1N1 virus. We here show that infection of macaques with two genetically similar but clinically distinct SOIV isolates from the early stage of the pandemic (A/Mexico/4108/2009 and A/Mexico/InDRE4487/2009) resulted in upper and lower respiratory tract infections and clinical disease ranging from mild to severe pneumonia that was clearly advanced over the mild infection caused by A/Kawasaki/UTK-4/2009, a current seasonal strain. Unexpectedly, we observed heterogeneity among the two SOIV isolates in virus replication, host transcriptional and cytokine responses, and disease progression, demonstrating a higher pathogenic potential for A/Mexico/InDRE4487/2009. Differences in virulence may explain more severe disease, as was seen with certain individuals infected with the emerged pandemic influenza virus. Thus, the nonhuman primate model closely mimics influenza in humans.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/aislamiento & purificación , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Enfermedades de los Primates/patología , Enfermedades de los Primates/virología , Animales , Preescolar , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Variación Genética , Humanos , Gripe Humana/virología , Macaca , Masculino , Neumonía Viral/patología , Neumonía Viral/virología , Infecciones del Sistema Respiratorio/patología , Infecciones del Sistema Respiratorio/virología , Índice de Severidad de la Enfermedad , Virulencia
8.
J Gen Virol ; 92(Pt 9): 2093-2104, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21593271

RESUMEN

The NS1 protein of influenza virus counters host antiviral defences primarily by antagonizing the type I interferon (IFN) response. Both the N-terminal dsRNA-binding domain and the C-terminal effector domain are required for optimal suppression of host responses during infection. To better understand the regulatory role of the NS1 effector domain, we used an NS1-truncated mutant virus derived from human H1N1 influenza isolate A/Texas/36/91 (Tx/91) and assessed global transcriptional profiles from two independent human lung cell-culture models. Relative to the wild-type Tx/91-induced gene expression, the NS1 mutant virus induced enhanced expression of innate immune genes, specifically NF-κB signalling-pathway genes and IFN-α and -ß target genes. We queried an experimentally derived IFN gene set to gauge the proportion of IFN-responsive genes that are suppressed specifically by NS1. We show that the C-terminally truncated NS1 mutant virus is less efficient at suppressing IFN-regulated gene expression associated with activation of antigen-presentation and immune-proteasome pathways. This is the first report integrating genomic analysis from two independent human culture systems, including primary lung cells, using genetically similar H1N1 influenza viruses that differ only in the length of the NS1 protein.


Asunto(s)
Presentación de Antígeno , Subtipo H1N1 del Virus de la Influenza A/inmunología , Interferón-alfa/antagonistas & inhibidores , Interferón beta/antagonistas & inhibidores , Inhibidores de Proteasoma , Proteínas no Estructurales Virales/metabolismo , Factores de Virulencia/metabolismo , Células Cultivadas , Células Epiteliales/inmunología , Células Epiteliales/virología , Perfilación de la Expresión Génica , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Proteínas Mutantes/genética , Proteínas Mutantes/inmunología , Proteínas Mutantes/metabolismo , FN-kappa B/antagonistas & inhibidores , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Factores de Virulencia/inmunología
9.
J Virol ; 84(24): 12576-88, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20926563

RESUMEN

The influenza pandemic of 1918 to 1919 was one of the worst global pandemics in recent history. The highly pathogenic nature of the 1918 virus is thought to be mediated in part by a dysregulation of the host response, including an exacerbated proinflammatory cytokine response. In the present study, we compared the host transcriptional response to infection with the reconstructed 1918 virus in wild-type, tumor necrosis factor (TNF) receptor-1 knockout (TNFRKO), and interleukin-1 (IL-1) receptor-1 knockout (IL1RKO) mice as a means of further understanding the role of proinflammatory cytokine signaling during the acute response to infection. Despite reported redundancy in the functions of IL-1ß and TNF-α, we observed that reducing the signaling capacity of each of these molecules by genetic disruption of their key receptor genes had very different effects on the host response to infection. In TNFRKO mice, we found delayed or decreased expression of genes associated with antiviral and innate immune signaling, complement, coagulation, and negative acute-phase response. In contrast, in IL1RKO mice numerous genes were differentially expressed at 1 day postinoculation, including an increase in the expression of genes that contribute to dendritic and natural killer cell processes and cellular movement, and gene expression profiles remained relatively constant at later time points. We also observed a compensatory increase in TNF-α expression in virus-infected IL1RKO mice. Our data suggest that signaling through the IL-1 receptor is protective, whereas signaling through the TNF-α receptor increases the severity of 1918 virus infection. These findings suggest that manipulation of these pathways may have therapeutic benefit.


Asunto(s)
Biomarcadores/metabolismo , Perfilación de la Expresión Génica , Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/virología , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Transducción de Señal/fisiología , Animales , Western Blotting , Comunicación Celular , Movimiento Celular , Células Dendríticas/metabolismo , Células Dendríticas/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Pandemias , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Triazoles , Factor de Necrosis Tumoral alfa/fisiología
10.
J Nutr ; 139(10): 1855-60, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19710156

RESUMEN

Vitamin E has been shown to affect cytokine production. However, individual response to vitamin E supplementation varies. Previous studies indicate that cytokine production is heritable and common single nucleotide polymorphisms (SNP) may explain differences in cytokine production between individuals. We hypothesize that the differential response to the immunomodulatory actions of vitamin E reflects genetic differences among individuals, including SNP at cytokine genes that modulate cytokine production. We used data from a double-blind, placebo-controlled 1-y vitamin E (182 mg d,l-alpha-tocopherol) intervention study in elderly men and women (mean age 83 y) to test this hypothesis (vitamin E, n = 47; placebo, n = 63). We found that the effect of vitamin E on tumor necrosis factor (TNF)-alpha production in whole blood stimulated for 24 h with lipopolysaccharide (1.0 mg/L) is dependent on TNFalpha -308G > A. Participants with the A/A and A/G genotypes at TNFalpha -308G > A who were treated with vitamin E had lower TNFalpha production than those with the A allele treated with placebo. These observations suggest that individual immune responses to vitamin E supplementation are in part mediated by genetic factors. Because the A allele at TNFalpha has been previously associated with higher TNFalpha levels in whole blood and isolated immune cells, our observations suggest that the antiinflammatory effect of vitamin E is specific to those genetically predisposed to higher inflammation. Further studies are needed to determine the biological mechanism driving the interaction between vitamin E treatment and TNFalpha -308G > A and its implications for disease resistance.


Asunto(s)
Citocinas/genética , Citocinas/metabolismo , Polimorfismo Genético , Vitamina E/farmacología , Anciano , Anciano de 80 o más Años , Método Doble Ciego , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Genotipo , Humanos , Masculino
11.
Biofactors ; 33(3): 191-200, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19478423

RESUMEN

Vitamin E supplementation has been suggested to improve immune response in the aged in part by altering cytokine production. However, there is not a consensus regarding the effect of supplemental vitamin E on cytokine production in humans. There is evidence that baseline immune health can affect immune response to supplemental vitamin E in the elderly. Thus, the effect of vitamin E on cytokines may depend on their pre-supplementation cytokine response. Using data from a vitamin E intervention in elderly nursing home residents, we examined if the effect of vitamin E on ex vivo cytokine production of IL-1 beta, IL-6, TNF-alpha, and IFN-gamma depended on baseline cytokine production. We observed that the effect of vitamin E supplementation on cytokine production depended on pre-supplementation production of the respective cytokines. The interactions between vitamin E and baseline cytokine production were not explained by covariates known to impact cytokine production. Our results offer evidence that baseline cytokine production should be considered in studies that examine the effect of supplemental vitamin E on immune and inflammatory responses. Our results could have implications in designing clinical trials to determine the impact of vitamin E on conditions in which cytokines are implicated such as infections and atherosclerotic disease.


Asunto(s)
Citocinas/biosíntesis , Vitamina E/uso terapéutico , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Interferón gamma/biosíntesis , Interleucina-1beta/biosíntesis , Interleucina-6/biosíntesis , Masculino , Factor de Necrosis Tumoral alfa/biosíntesis
12.
mBio ; 4(4)2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-23919993

RESUMEN

UNLABELLED: Systems biology offers considerable promise in uncovering novel pathways by which viruses and other microbial pathogens interact with host signaling and expression networks to mediate disease severity. In this study, we have developed an unbiased modeling approach to identify new pathways and network connections mediating acute lung injury, using severe acute respiratory syndrome coronavirus (SARS-CoV) as a model pathogen. We utilized a time course of matched virologic, pathological, and transcriptomic data within a novel methodological framework that can detect pathway enrichment among key highly connected network genes. This unbiased approach produced a high-priority list of 4 genes in one pathway out of over 3,500 genes that were differentially expressed following SARS-CoV infection. With these data, we predicted that the urokinase and other wound repair pathways would regulate lethal versus sublethal disease following SARS-CoV infection in mice. We validated the importance of the urokinase pathway for SARS-CoV disease severity using genetically defined knockout mice, proteomic correlates of pathway activation, and pathological disease severity. The results of these studies demonstrate that a fine balance exists between host coagulation and fibrinolysin pathways regulating pathological disease outcomes, including diffuse alveolar damage and acute lung injury, following infection with highly pathogenic respiratory viruses, such as SARS-CoV. IMPORTANCE: Severe acute respiratory syndrome coronavirus (SARS-CoV) emerged in 2002 and 2003, and infected patients developed an atypical pneumonia, acute lung injury (ALI), and acute respiratory distress syndrome (ARDS) leading to pulmonary fibrosis and death. We identified sets of differentially expressed genes that contribute to ALI and ARDS using lethal and sublethal SARS-CoV infection models. Mathematical prioritization of our gene sets identified the urokinase and extracellular matrix remodeling pathways as the most enriched pathways. By infecting Serpine1-knockout mice, we showed that the urokinase pathway had a significant effect on both lung pathology and overall SARS-CoV pathogenesis. These results demonstrate the effective use of unbiased modeling techniques for identification of high-priority host targets that regulate disease outcomes. Similar transcriptional signatures were noted in 1918 and 2009 H1N1 influenza virus-infected mice, suggesting a common, potentially treatable mechanism in development of virus-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/virología , Interacciones Huésped-Patógeno , Pulmón/patología , Pulmón/virología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Coagulación Sanguínea , Modelos Animales de Enfermedad , Fibrinólisis , Perfilación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteoma/análisis , Factores de Tiempo , Activador de Plasminógeno de Tipo Uroquinasa/genética
13.
BMC Syst Biol ; 5: 190, 2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-22074594

RESUMEN

BACKGROUND: Understanding host response to influenza virus infection will facilitate development of better diagnoses and therapeutic interventions. Several different experimental models have been used as a proxy for human infection, including cell cultures derived from human cells, mice, and non-human primates. Each of these systems has been studied extensively in isolation, but little effort has been directed toward systematically characterizing the conservation of host response on a global level beyond known immune signaling cascades. RESULTS: In the present study, we employed a multivariate modeling approach to characterize and compare the transcriptional regulatory networks between these three model systems after infection with a highly pathogenic avian influenza virus of the H5N1 subtype. Using this approach we identified functions and pathways that display similar behavior and/or regulation including the well-studied impact on the interferon response and the inflammasome. Our results also suggest a primary response role for airway epithelial cells in initiating hypercytokinemia, which is thought to contribute to the pathogenesis of H5N1 viruses. We further demonstrate that we can use a transcriptional regulatory model from the human cell culture data to make highly accurate predictions about the behavior of important components of the innate immune system in tissues from whole organisms. CONCLUSIONS: This is the first demonstration of a global regulatory network modeling conserved host response between in vitro and in vivo models.


Asunto(s)
Redes Reguladoras de Genes , Subtipo H5N1 del Virus de la Influenza A , Gripe Humana/genética , Animales , Resistencia a la Enfermedad/genética , Humanos , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Gripe Humana/inmunología , Macaca , Ratones , Modelos Inmunológicos , Análisis Multivariante , Biología de Sistemas
14.
PLoS One ; 6(6): e19681, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21701683

RESUMEN

While HIV-1-specific cellular immunity is thought to be critical for the suppression of viral replication, the correlates of protection have not yet been determined. Rhesus macaques (RM) are an important animal model for the study and development of vaccines against HIV/AIDS. Our laboratory has helped to develop and study DNA-based vaccines in which recent technological advances, including genetic optimization and in vivo electroporation (EP), have helped to dramatically boost their immunogenicity. In this study, RMs were immunized with a DNA vaccine including individual plasmids encoding SIV gag, env, and pol alone, or in combination with a molecular adjuvant, plasmid DNA expressing the chemokine ligand 5 (RANTES), followed by EP. Along with standard immunological assays, flow-based activation analysis without ex vivo restimulation and high-throughput gene expression analysis was performed. Strong cellular immunity was induced by vaccination which was supported by all assays including PBMC microarray analysis that identified the up-regulation of 563 gene sequences including those involved in interferon signaling. Furthermore, 699 gene sequences were differentially regulated in these groups at peak viremia following SIVmac251 challenge. We observed that the RANTES-adjuvanted animals were significantly better at suppressing viral replication during chronic infection and exhibited a distinct pattern of gene expression which included immune cell-trafficking and cell cycle genes. Furthermore, a greater percentage of vaccine-induced central memory CD8+ T-cells capable of an activated phenotype were detected in these animals as measured by activation analysis. Thus, co-immunization with the RANTES molecular adjuvant followed by EP led to the generation of cellular immunity that was transcriptionally distinct and had a greater protective efficacy than its DNA alone counterpart. Furthermore, activation analysis and high-throughput gene expression data may provide better insight into mechanisms of viral control than may be observed using standard immunological assays.


Asunto(s)
Leucocitos Mononucleares/metabolismo , Vacunas contra el SIDAS/inmunología , Vacunas de ADN/inmunología , Animales , Citometría de Flujo , Perfilación de la Expresión Génica , Interferón gamma/metabolismo , Macaca mulatta , Análisis de Secuencia por Matrices de Oligonucleótidos
15.
J Clin Endocrinol Metab ; 96(5): E816-20, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21307135

RESUMEN

CONTEXT: IL1b (IL1B or IL1ß), a key modulator of the immune response, exerts its functions mainly via IL6 regulation. Fatty meals cause transient hypertriglyceridemia and are considered to be proinflammatory, but the extent of these responses shows high interindividual susceptibility. OBJECTIVE: We evaluated the influence of a genetic variant located in the promoter region of IL1B (-1473G/C) on fasting and postprandial lipids and IL6. DESIGN, SETTING, AND PARTICIPANTS: A total of 477 people over age 65 yr were genotyped for IL1B -1473G/C, and we evaluated fasting lipids depending on genotype. Then, 88 healthy young men were also genotyped and were fed a saturated fatty acid-rich meal. Serial blood samples were drawn for 11 h after the meal, and lipid fractions and IL6 were assayed. MAIN OUTCOME AND INTERVENTIONS: Fasting lipids were studied in the aged persons. Fasting and postprandial measurements of lipids and IL6 were performed in the healthy young men. RESULTS: In the aged persons, CC subjects (minor allele homozygotes) showed higher triglyceride (P = 0.002) and cholesterol (P = 0.011) levels. Healthy young male carriers of the minor C allele showed higher postprandial triglycerides (P = 0.037), and those carried into large triglyceride-rich lipoproteins (P = 0.004). In addition, they showed higher postprandial IL6 concentrations (P = 0.008). CONCLUSIONS: Our work shows that inflammatory genes may regulate fasting and postprandial lipids because the carriers of the minor allele of an IL gene variant have altered lipid metabolism. To reinforce these gene-phenotype findings, IL6 (the natural effector of IL1B) was increased in these persons.


Asunto(s)
Interleucina-1beta/genética , Interleucina-6/metabolismo , Triglicéridos/metabolismo , Adolescente , Adulto , Anciano , Estudios de Cohortes , Grasas de la Dieta/metabolismo , Ayuno/fisiología , Femenino , Variación Genética , Genotipo , Humanos , Lipoproteínas/análisis , Lipoproteínas/aislamiento & purificación , Lipoproteínas/metabolismo , Masculino , Periodo Posprandial/genética , Periodo Posprandial/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Adulto Joven
16.
Am J Clin Nutr ; 92(1): 106-14, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20484443

RESUMEN

BACKGROUND: Vitamin E supplementation may be a potential strategy to prevent respiratory tract infections (RIs) in the elderly. The efficacy of vitamin E supplementation may depend on individual factors including specific single nucleotide polymorphisms (SNPs) at immunoregulatory genes. OBJECTIVE: We examined whether the effect of vitamin E on RIs in the elderly was dependent on genetic backgrounds as indicated by SNPs at cytokine genes. DESIGN: We used data and DNA from a previous vitamin E intervention study (200 IU vitamin E or a placebo daily for 1 y) in elderly nursing home residents to examine vitamin E-gene interactions for incidence of RI. We determined the genotypes of common SNPs at IL-1beta, IL-2, IL-6, IL-10, TNF-alpha, and IFN-gamma in 500 participants. We used negative binomial regression to analyze the association between genotype and incidence of infection. RESULTS: The effect of vitamin E on lower RI depended on sex and the SNP at IL-10 -819G-->A (P = 0.03 for interaction for lower RI). Furthermore, we observed that subjects with the least prevalent genotypes at IL-2 -330A-->C (P = 0.02 for upper RI), IL-10 -819G-->A (P = 0.08 for upper RI), and IL-10 -1082C-->T (P < 0.001 for lower RI in men) had a lower incidence of RI independent of vitamin E supplementation. CONCLUSIONS: Studies that evaluate the effect of vitamin E on RIs should consider both genetic factors and sex because our results suggest that both may have a significant bearing on the efficacy of vitamin E. Furthermore, common SNPs at cytokine genes may contribute to the individual risk of RIs in the elderly. This trial was registered at clinicaltrials.gov as NCT00758914.


Asunto(s)
Interleucina-10/genética , Interleucina-2/genética , Polimorfismo Genético , Infecciones del Sistema Respiratorio/epidemiología , Vitamina E/uso terapéutico , Anciano , Anciano de 80 o más Años , Índice de Masa Corporal , ADN/genética , ADN/aislamiento & purificación , Suplementos Dietéticos , Femenino , Frecuencia de los Genes , Genotipo , Frecuencia Cardíaca , Humanos , Masculino , Casas de Salud , Placebos , Polimorfismo de Nucleótido Simple , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/genética , Factores de Riesgo , Caracteres Sexuales
17.
Future Virol ; 4(6): 553-561, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20352075

RESUMEN

Sporadic outbreaks of epizootics including SARS coronavirus and H5N1 avian influenza remind us of the potential for communicable diseases to quickly spread into worldwide epidemics. To confront emerging viral threats, nations have implemented strategies to prepare for pandemics and to control virus spread. Despite improved surveillance and quarantine measures, we find ourselves in the midst of a H1N1 influenza pandemic. Effective therapeutics and vaccines are essential to protect against current and future pandemics. The best route to effective therapeutics and vaccines is through a detailed and global view of virus-host interactions that can be achieved using a systems biology approach. Here, we provide our perspective on the role of systems biology in deepening our understanding of virus-host interactions and in improving drug and vaccine development. We offer examples from influenza virus research, as well as from research on other pandemics of our time - HIV/AIDS and HCV - to demonstrate that systems biology offers one possible key to stopping the cycle of viral pandemics.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA