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

Bases de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
PLoS Pathog ; 14(12): e1007439, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30576397

RESUMO

Arenaviruses are a significant cause of hemorrhagic fever, an often-fatal disease for which there is no approved antiviral therapy. Lassa fever in particular generates high morbidity and mortality in West Africa, where the disease is endemic, and a recent outbreak in Nigeria was larger and more geographically diverse than usual. We are developing LHF-535, a small-molecule viral entry inhibitor that targets the arenavirus envelope glycoprotein, as a therapeutic candidate for Lassa fever and other hemorrhagic fevers of arenavirus origin. Using a lentiviral pseudotype infectivity assay, we determined that LHF-535 had sub-nanomolar potency against the viral envelope glycoproteins from all Lassa virus lineages, with the exception of the glycoprotein from the LP strain from lineage I, which was 100-fold less sensitive than that of other strains. This reduced sensitivity was mediated by a unique amino acid substitution, V434I, in the transmembrane domain of the envelope glycoprotein GP2 subunit. This position corresponds to the attenuation determinant of Candid#1, a live-attenuated Junín virus vaccine strain used to prevent Argentine hemorrhagic fever. Using a virus-yield reduction assay, we determined that LHF-535 potently inhibited Junín virus, but not Candid#1, and the Candid#1 attenuation determinant, F427I, regulated this difference in sensitivity. We also demonstrated that a daily oral dose of LHF-535 at 10 mg/kg protected mice from a lethal dose of Tacaribe virus. Serial passage of Tacaribe virus in LHF-535-treated Vero cells yielded viruses that were resistant to LHF-535, and the majority of drug-resistant viruses exhibited attenuated pathogenesis. These findings provide a framework for the clinical development of LHF-535 as a broad-spectrum inhibitor of arenavirus entry and provide an important context for monitoring the emergence of drug-resistant viruses.


Assuntos
Antivirais/farmacologia , Febre Lassa , Vírus Lassa/genética , Virulência/efeitos dos fármacos , Virulência/genética , Animais , Chlorocebus aethiops , Farmacorresistência Viral/efeitos dos fármacos , Farmacorresistência Viral/genética , Células HEK293 , Humanos , Vírus Lassa/efeitos dos fármacos , Camundongos , Mutação , Células Vero , Proteínas do Envelope Viral/genética
2.
Semin Immunol ; 25(3): 228-39, 2013 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-23218769

RESUMO

Influenza virus research has recently undergone a shift from a virus-centric perspective to one that embraces the full spectrum of virus-host interactions and cellular signaling events that determine disease outcome. This change has been brought about by the increasing use and expanding scope of high-throughput molecular profiling and computational biology, which together fuel discovery in systems biology. In this review, we show how these approaches have revealed an uncontrolled inflammatory response as a contributor to the extreme virulence of the 1918 pandemic and avian H5N1 viruses, and how this response differs from that induced by the 2009 H1N1 viruses responsible for the most recent influenza pandemic. We also discuss how new animal models, such as the Collaborative Cross mouse systems genetics platform, are key to the necessary systematic investigation of the impact of host genetics on infection outcome, how genome-wide RNAi screens have identified hundreds of cellular factors involved in viral replication, and how systems biology approaches are making possible the rational design of new drugs and vaccines against an ever-evolving respiratory virus.


Assuntos
Interações Hospedeiro-Patógeno , Influenza Humana/imunologia , Infecções por Orthomyxoviridae/imunologia , Orthomyxoviridae/imunologia , Biologia de Sistemas/métodos , Animais , Biologia Computacional , Modelos Animais de Doenças , Interação Gene-Ambiente , Ensaios de Triagem em Larga Escala , Humanos , Imunidade/genética , Vacinas contra Influenza/imunologia , Influenza Humana/epidemiologia , Influenza Humana/genética , Camundongos , Orthomyxoviridae/patogenicidade , Infecções por Orthomyxoviridae/genética , Pandemias , Virulência
3.
J Virol ; 89(5): 2543-52, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25520505

RESUMO

UNLABELLED: To identify host factors associated with arenavirus virulence, we used a cynomolgus macaque model to evaluate the pathogenesis of Lujo virus (LUJV), a recently emerged arenavirus that caused an outbreak of severe viral hemorrhagic fever in southern Africa. In contrast to human cases, LUJV caused mild, nonlethal illness in macaques. We then compared this to contrasting clinical outcomes during arenavirus infection, specifically to samples obtained from macaques infected with three highly pathogenic lines of Lassa virus (LASV), the causative agent of Lassa fever (LF). We assessed gene expression in peripheral blood mononuclear cells (PBMC) and determined genes that significantly changed expression relative to that in uninfected animals over the course of infection. We detected a 72-h delay in the induction of host responses to infection during LUJV infection compared to that of the animals infected with LASV. This included genes associated with inflammatory and antiviral responses and was particularly apparent among groups of genes promoting cell death. We also observed early differential expression of a subset of genes specific to LUJV infection that accounts for the delayed inflammatory response. Cell type enrichment analysis suggested that host response induction delay and an LUJV-specific profile are due to a different proportion of natural killer cells responding in LUJV infection than that in the LASV-infected animals. Together, these data indicate that delayed proinflammatory and proapoptotic host responses to arenavirus infection could ameliorate disease severity. This conclusion provides insight into the cellular and molecular mechanisms of arenaviral hemorrhagic fever and suggests potential strategies for therapeutic development. IMPORTANCE: Old World arenaviruses are significant human pathogens that often are associated with high mortality. However, mechanisms underlying disease severity and virulence in arenavirus hemorrhagic fever are largely unknown, particularly regarding host responses that contribute to pathogenicity. This study describes a comparison between Lujo and Lassa virus infection in cynomolgus macaques. Lujo virus-infected macaques developed only mild illness, while Lassa virus-infected macaques developed severe illness consistent with Lassa fever. We determined that mild disease is associated with a delay in host expression of genes linked to virulence, such as those causing inflammation and cell death, and with distinct cell types that may mediate this delay. This is the first study to associate the timing and directionality of gene expression with arenaviral pathogenicity and disease outcome and evokes new potential approaches for developing effective therapeutics for treating these deadly emerging pathogens.


Assuntos
Infecções por Arenaviridae/patologia , Infecções por Arenaviridae/virologia , Febres Hemorrágicas Virais/patologia , Febres Hemorrágicas Virais/virologia , Lujo virus/patogenicidade , Animais , Infecções por Arenaviridae/imunologia , Morte Celular , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Febres Hemorrágicas Virais/imunologia , Inflamação/patologia , Células Matadoras Naturais/imunologia , Febre Lassa/patologia , Febre Lassa/virologia , Vírus Lassa/patogenicidade , Leucócitos Mononucleares/imunologia , Macaca fascicularis , Fatores de Tempo
4.
J Virol ; 88(16): 8768-82, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24850744

RESUMO

UNLABELLED: Polyadenylated mature mRNAs are the focus of standard transcriptome analyses. However, the profiling of nascent transcripts, which often include nonpolyadenylated RNAs, can unveil novel insights into transcriptional regulation. Here, we separately sequenced total RNAs (Total RNAseq) and mRNAs (mRNAseq) from the same HIV-1-infected human CD4(+) T cells. We found that many nonpolyadenylated RNAs were differentially expressed upon HIV-1 infection, and we identified 8 times more differentially expressed genes at 12 h postinfection by Total RNAseq than by mRNAseq. These expression changes were also evident by concurrent changes in introns and were recapitulated by later mRNA changes, revealing an unexpectedly significant delay between transcriptional initiation and mature mRNA production early after HIV-1 infection. We computationally derived and validated the underlying regulatory programs, and we predicted drugs capable of reversing these HIV-1-induced expression changes followed by experimental confirmation. Our results show that combined total and mRNA transcriptome analysis is essential for fully capturing the early host response to virus infection and provide a framework for identifying candidate drugs for host-directed therapy against HIV/AIDS. IMPORTANCE: In this study, we used mass sequencing to identify genes differentially expressed in CD4(+) T cells during HIV-1 infection. To our surprise, we found many differentially expressed genes early after infection by analyzing both newly transcribed unprocessed pre-mRNAs and fully processed mRNAs, but not by analyzing mRNAs alone, indicating a significant delay between transcription initiation and mRNA production early after HIV-1 infection. These results also show that important findings could be missed by the standard practice of analyzing mRNAs alone. We then derived the regulatory mechanisms driving the observed expression changes using integrative computational analyses. Further, we predicted drugs that could reverse the observed expression changes induced by HIV-1 infection and showed that one of the predicted drugs indeed potently inhibited HIV-1 infection. This shows that it is possible to identify candidate drugs for host-directed therapy against HIV/AIDS using our genomics-based approach.


Assuntos
Infecções por HIV/genética , HIV-1/genética , Transcrição Gênica/genética , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/genética , Infecções por HIV/virologia , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , RNA/genética , RNA Mensageiro/genética , Replicação Viral/genética
5.
J Virol ; 86(5): 2437-46, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22190720

RESUMO

Understanding the role of host factors during lethal influenza virus infection is critical to deciphering the events that determine the fate of the host. One such factor is encoded by the Mx1 gene, which confers resistance to influenza virus infection. Here, we compared pathology and global gene expression profiles in lung tissue from BALB/c (Mx1(-)) and BALB · A2G-Mx1 mice (Mx1(+/+)) infected with the fully reconstructed 1918 pandemic influenza virus. Mx1(+/+) mice showed less tissue damage than Mx(-) animals, and pathology and mortality were further reduced by treating the mice with interferon prior to infection. Using global transcriptional profiling, we identified distinct molecular signatures associated with partial protection, complete protection, and the contribution of interferon to the host response. In the absence of interferon treatment, partial protection was characterized by the generation of an acute response with the upregulation of genes associated with apoptosis, reactive oxygen species, and cell migration. Complete protection was characterized by the downregulation of cytokine and chemokine genes previously associated with influenza virus pathogenesis. The contribution of interferon treatment to total protection in virus-infected Mx1(+/+) mice was characterized by the altered regulation of cell cycle genes. These genes were upregulated in Mx1(+/+) mice treated with interferon but downregulated in the absence of interferon treatment. Our results suggest that Mx1(+/+) mice generate a protective antiviral response by controlling the expression of key modulator molecules associated with influenza virus lethality.


Assuntos
Proteínas de Ligação ao GTP/imunologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Influenza Humana/imunologia , Influenza Humana/mortalidade , Animais , Resistência à Doença , Feminino , Proteínas de Ligação ao GTP/genética , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/patogenicidade , Influenza Humana/patologia , Influenza Humana/virologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteínas de Resistência a Myxovirus , Sobrevida
6.
Hepatology ; 56(1): 17-27, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22278598

RESUMO

UNLABELLED: Liver failure resulting from chronic hepatitis C virus (HCV) infection is a major cause for liver transplantation worldwide. Recurrent infection of the graft is universal in HCV patients after transplant and results in a rapid progression to severe fibrosis and end-stage liver disease in one third of all patients. No single clinical variable, or combination thereof, has, so far, proven accurate in identifying patients at risk of hepatic decompensation in the transplant setting. A combination of longitudinal, dimensionality reduction and categorical analysis of the transcriptome from 111 liver biopsy specimens taken from 57 HCV-infected patients over time identified a molecular signature of gene expression of patients at risk of developing severe fibrosis. Significantly, alterations in gene expression occur before histologic evidence of liver disease progression, suggesting that events that occur during the acute phase of infection influence patient outcome. Additionally, a common precursor state for different severe clinical outcomes was identified. CONCLUSION: Based on this patient cohort, incidence of severe liver disease is a process initiated early during HCV infection of the donor organ. The probable cellular network at the basis of the initial transition to severe liver disease was identified and characterized.


Assuntos
Rejeição de Enxerto/genética , Hepatite C Crônica/complicações , Falência Hepática/cirurgia , Transplante de Fígado/efeitos adversos , Ativação Transcricional/genética , Idoso , Biópsia por Agulha , Estudos de Coortes , Progressão da Doença , Feminino , Regulação da Expressão Gênica , Hepacivirus/genética , Hepacivirus/fisiologia , Hepatite C Crônica/patologia , Hepatite C Crônica/cirurgia , Humanos , Imuno-Histoquímica , Falência Hepática/etiologia , Falência Hepática/genética , Transplante de Fígado/métodos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Complicações Pós-Operatórias/genética , Complicações Pós-Operatórias/fisiopatologia , Prognóstico , Recidiva , Medição de Risco , Índice de Gravidade de Doença , Doadores de Tecidos
7.
J Virol ; 85(17): 9060-8, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21734050

RESUMO

Ebola virus is the etiologic agent of a lethal hemorrhagic fever in humans and nonhuman primates with mortality rates of up to 90%. Previous studies with Zaire Ebola virus (ZEBOV), mouse-adapted virus (MA-ZEBOV), and mutant viruses (ZEBOV-NP(ma), ZEBOV-VP24(ma), and ZEBOV-NP/VP24(ma)) allowed us to identify the mutations in viral protein 24 (VP24) and nucleoprotein (NP) responsible for acquisition of high virulence in mice. To elucidate specific molecular signatures associated with lethality, we compared global gene expression profiles in spleen samples from mice infected with these viruses and performed an extensive functional analysis. Our analysis showed that the lethal viruses (MA-ZEBOV and ZEBOV-NP/VP24(ma)) elicited a strong expression of genes 72 h after infection. In addition, we found that although the host transcriptional response to ZEBOV-VP24(ma) was nearly the same as that to ZEBOV-NP/VP24(ma), the contribution of a mutation in the NP gene was required for a lethal phenotype. Further analysis indicated that one of the most relevant biological functions differentially regulated by the lethal viruses was the inflammatory response, as was the induction of specific metalloproteinases, which were present in our newly identify functional network that was associated with Ebola virus lethality. Our results suggest that this dysregulated proinflammatory response increased the severity of disease. Consequently, the newly discovered molecular signature could be used as the starting point for the development of new drugs and therapeutics. To our knowledge, this is the first study that clearly defines unique molecular signatures associated with Ebola virus lethality.


Assuntos
Ebolavirus/patogenicidade , Perfilação da Expressão Gênica , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/patologia , Inflamação/imunologia , Inflamação/patologia , Metaloproteases/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Doença pelo Vírus Ebola/mortalidade , Camundongos , Camundongos Endogâmicos BALB C , Doenças dos Roedores/imunologia , Doenças dos Roedores/mortalidade , Doenças dos Roedores/patologia , Baço/imunologia , Baço/patologia
8.
J Virol ; 85(2): 1099-116, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21068249

RESUMO

We have employed global transcriptional profiling of whole blood to identify biologically relevant changes in cellular gene expression in response to alternative AIDS vaccine strategies with subsequent viral challenge in a rhesus macaque vaccine model. Samples were taken at day 0 (prechallenge), day 14 (peak viremia), and week 12 (set point) from animals immunized with replicating adenovirus type 5 host range (Ad5hr) recombinant viruses expressing human immunodeficiency virus HIV(env)(89.6P), simian immunodeficiency virus SIV(gag)(239), or SIV(nef)(239) alone or in combination with two intramuscular boosts with HIV(89.6P)gp140ΔCFI protein (L. J. Patterson et al., Virology 374:322-337, 2008), and each treatment resulted in significant control of viremia following simian-human immunodeficiency virus SHIV(89.6P) challenge (six animals per group plus six controls). At day 0, 8 weeks after the last treatment, the microarray profiles revealed significant prechallenge differences between treatment groups; data from the best-protected animals led to identification of a network of genes related to B cell development and lymphocyte survival. At peak viremia, expression profiles of the immunized groups were extremely similar, and comparisons to control animals reflected immunological differences other than effector T cell functions. Suggested protective mechanisms for vaccinated animals included upregulation of interleukin-27, a cytokine known to inhibit lentivirus replication, and increased expression of complement components, which may synergize with vaccine-induced antibodies. Divergent expression profiles at set point for the immunized groups implied distinct immunological responses despite phenotypic similarities in viral load and CD4(+) T cell levels. Data for the gp140-boosted group provided evidence for antibody-dependent, cell-mediated viral control, whereas animals immunized with only the replicating Ad5hr recombinants exhibited a different evolution of the B cell compartment even at 3 months postchallenge. This study demonstrates the sensitivity and discrimination of gene expression profiling of whole blood as an analytical tool in AIDS vaccine trials, providing unique insights into in vivo mechanisms and potential correlates of protection.


Assuntos
Vacinas contra a AIDS/imunologia , Perfilação da Expressão Gênica , HIV/imunologia , Vacinas contra a SAIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Vacinas contra a AIDS/genética , Adenoviridae/genética , Animais , Modelos Animais de Doenças , Vetores Genéticos , HIV/genética , Imunização Secundária/métodos , Injeções Intramusculares , Macaca mulatta , Masculino , Análise em Microsséries , Recombinação Genética , Vacinas contra a SAIDS/genética , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética , Vacinação/métodos , Carga Viral , Viremia
9.
J Virol ; 85(22): 11646-54, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21917952

RESUMO

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.


Assuntos
Isomerases de Ligação Dupla Carbono-Carbono/metabolismo , Hepacivirus/patogenicidade , Interações Hospedeiro-Patógeno , Mitocôndrias/enzimologia , Replicação Viral , Biópsia , Linhagem Celular , Dodecenoil-CoA Isomerase , Ácidos Graxos/metabolismo , Inativação Gênica , Hepatócitos/enzimologia , Hepatócitos/virologia , Humanos , Fígado/química , Fígado/patologia , Oxirredução , Proteoma
10.
J Virol ; 85(3): 1214-23, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21084481

RESUMO

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.


Assuntos
Vírus da Influenza A Subtipo H1N1/isolamento & purificação , Vírus da Influenza A Subtipo H1N1/patogenicidade , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Doenças dos Primatas/patologia , Doenças dos Primatas/virologia , Animais , Pré-Escolar , Citocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Perfilação da Expressão Gênica , Variação Genética , Humanos , Influenza Humana/virologia , Macaca , Masculino , Pneumonia Viral/patologia , Pneumonia Viral/virologia , Infecções Respiratórias/patologia , Infecções Respiratórias/virologia , Índice de Gravidade de Doença , Virulência
11.
Sci Rep ; 12(1): 19911, 2022 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-36402782

RESUMO

LHF-535 is a small molecule antiviral currently in development for the treatment of Lassa fever, a zoonotic disease endemic in West Africa that generates significant morbidity and mortality. Current treatment options are inadequate, and there are no approved therapeutics or vaccines for Lassa fever. LHF-535 was evaluated in a lethal guinea pig model of Lassa pathogenesis, using once-daily administration of a fixed dose (50 mg/kg/day) initiating either 1 or 3 days after inoculation with a lethal dose of Lassa virus. LHF-535 reduced viremia and clinical signs and protected all animals from lethality. A subset of surviving animals was rechallenged four months later with a second lethal challenge of Lassa virus and were found to be protected from disease. LHF-535 pharmacokinetics at the protective dose in guinea pigs showed plasma concentrations well within the range observed in clinical trials in healthy volunteers, supporting the continued development of LHF-535 as a Lassa therapeutic.


Assuntos
Febre Lassa , Cobaias , Animais , Febre Lassa/tratamento farmacológico , Febre Lassa/prevenção & controle , Antivirais/farmacologia , Antivirais/uso terapêutico , Vírus Lassa , Viremia/tratamento farmacológico , Vacinação
12.
J Gen Virol ; 92(Pt 9): 2093-2104, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21593271

RESUMO

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.


Assuntos
Apresentação de Antígeno , Vírus da Influenza A Subtipo H1N1/imunologia , Interferon-alfa/antagonistas & inibidores , Interferon beta/antagonistas & inibidores , Inibidores de Proteassoma , Proteínas não Estruturais Virais/metabolismo , Fatores de Virulência/metabolismo , Células Cultivadas , Células Epiteliais/imunologia , Células Epiteliais/virologia , Perfilação da Expressão Gênica , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Proteínas Mutantes/genética , Proteínas Mutantes/imunologia , Proteínas Mutantes/metabolismo , NF-kappa B/antagonistas & inibidores , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Fatores de Virulência/imunologia
13.
J Virol ; 84(21): 11297-309, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20702617

RESUMO

Severe acute respiratory syndrome coronavirus (SARS-CoV) infection can cause the development of severe end-stage lung disease characterized by acute respiratory distress syndrome (ARDS) and pulmonary fibrosis. The mechanisms by which pulmonary lesions and fibrosis are generated during SARS-CoV infection are not known. Using high-throughput mRNA profiling, we examined the transcriptional response of wild-type (WT), type I interferon receptor knockout (IFNAR1-/-), and STAT1 knockout (STAT1-/-) mice infected with a recombinant mouse-adapted SARS-CoV (rMA15) to better understand the contribution of specific gene expression changes to disease progression. Despite a deletion of the type I interferon receptor, strong expression of interferon-stimulated genes was observed in the lungs of IFNAR1-/- mice, contributing to clearance of the virus. In contrast, STAT1-/- mice exhibited a defect in the expression of interferon-stimulated genes and were unable to clear the infection, resulting in a lethal outcome. STAT1-/- mice exhibited dysregulation of T-cell and macrophage differentiation, leading to a TH2-biased immune response and the development of alternatively activated macrophages that mediate a profibrotic environment within the lung. We propose that a combination of impaired viral clearance and T-cell/macrophage dysregulation causes the formation of prefibrotic lesions in the lungs of rMA15-infected STAT1-/- mice.


Assuntos
Fibrose/etiologia , Perfilação da Expressão Gênica , Fator de Transcrição STAT1/deficiência , Síndrome Respiratória Aguda Grave/patologia , Animais , Fibrose/genética , Pneumopatias/etiologia , Pneumopatias/genética , Macrófagos/patologia , Camundongos , Camundongos Knockout , Fenótipo , RNA Mensageiro/análise , Receptor de Interferon alfa e beta/deficiência , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave , Síndrome Respiratória Aguda Grave/imunologia , Células Th2/imunologia
14.
J Virol ; 84(4): 2027-37, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19939913

RESUMO

The innate immune response provides the first line of defense against foreign pathogens by responding to molecules that are a signature of a pathogenic infection. Certain RNA viruses, such as influenza virus, produce double-stranded RNA as an intermediate during the replication life cycle, which activates pathogen recognition receptors capable of inducing interferon production. By engaging interferon receptors, interferon activates the JAK-STAT pathway and results in the positive feedback of interferon production, amplifying the response to viral infection. To examine how deficiencies in interferon signaling affect the cellular response to infection, we performed influenza virus infections of mouse embryonic fibroblasts lacking the alpha/beta interferon receptor, the gamma interferon receptor, or both. In the absence of the alpha/beta interferon receptor, we observed increased viral replication but decreased activation of PKR, Stat1, and NF-kappaB; the presence or absence of the gamma interferon receptor did not exhibit discernible differences in these readouts. Analysis of gene expression profiles showed that while cells lacking the alpha/beta interferon receptor exhibited decreased levels of transcription of antiviral genes, genes related to inflammatory and apoptotic responses were transcribed to levels similar to those of cells containing the receptor. These results indicate that while the alpha/beta interferon receptor is needed to curb viral replication, it is dispensable for the induction of certain inflammatory and apoptotic genes. We have identified potential pathways, via interferon regulatory factor 3 (IRF3) activation or Hoxa13, Polr2a, Nr4a1, or Ing1 induction, that contribute to this redundancy. This study illustrates another way in which the host has evolved to establish several overlapping mechanisms to respond to viral infections.


Assuntos
Vírus da Influenza A/imunologia , Vírus da Influenza A/fisiologia , Receptor de Interferon alfa e beta/fisiologia , Replicação Viral/imunologia , Animais , Apoptose/genética , Sequência de Bases , Células Cultivadas , DNA Viral/genética , Cães , Interações Hospedeiro-Patógeno/imunologia , Inflamação/imunologia , Inflamação/virologia , Vírus da Influenza A/genética , Vírus da Influenza A/patogenicidade , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/imunologia , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Receptor 3 Toll-Like/metabolismo , Virulência , eIF-2 Quinase/metabolismo
15.
J Virol ; 84(24): 12576-88, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20926563

RESUMO

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.


Assuntos
Biomarcadores/metabolismo , Perfilação da Expressão Gênica , Vírus da Influenza A/patogenicidade , Infecções por Orthomyxoviridae/virologia , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Transdução de Sinais/fisiologia , Animais , Western Blotting , Comunicação Celular , Movimento Celular , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Pandemias , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Triazóis , Fator de Necrose Tumoral alfa/fisiologia
16.
J Virol ; 84(15): 7613-24, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20504916

RESUMO

Periodic outbreaks of highly pathogenic avian H5N1 influenza viruses and the current H1N1 pandemic highlight the need for a more detailed understanding of influenza virus pathogenesis. To investigate the host transcriptional response induced by pathogenic influenza viruses, we used a functional-genomics approach to compare gene expression profiles in lungs from 129S6/SvEv mice infected with either the fully reconstructed H1N1 1918 pandemic virus (1918) or the highly pathogenic avian H5N1 virus Vietnam/1203/04 (VN/1203). Although the viruses reached similar titers in the lung and caused lethal infections, the mean time of death was 6 days for VN/1203-infected animals and 9 days for mice infected with the 1918 virus. VN/1203-infected animals also exhibited an earlier and more potent inflammatory response. This response included induction of genes encoding components of the inflammasome. VN/1203 was also able to disseminate to multiple organs, including the brain, which correlated with changes in the expression of genes associated with hematological functions and lipoxin biogenesis and signaling. Both viruses elicited expression of type I interferon (IFN)-regulated genes in wild-type mice and to a lesser extent in mice lacking the type I IFN receptor, suggesting alternative or redundant pathways for IFN signaling. Our findings suggest that VN/1203 is more pathogenic in mice as a consequence of several factors, including the early and sustained induction of the inflammatory response, the additive or synergistic effects of upregulated components of the immune response, and inhibition of lipoxin-mediated anti-inflammatory responses, which correlated with the ability of VN/1203 to disseminate to extrapulmonary organs.


Assuntos
Inflamação/patologia , Virus da Influenza A Subtipo H5N1/imunologia , Virus da Influenza A Subtipo H5N1/patogenicidade , Lipoxinas/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Transdução de Sinais , Animais , Feminino , Regulação da Expressão Gênica , Inflamação/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N1/patogenicidade , Pulmão/patologia , Pulmão/virologia , Camundongos , Infecções por Orthomyxoviridae/mortalidade , Infecções por Orthomyxoviridae/virologia , Análise de Sobrevida , Virulência
17.
PLoS Pathog ; 5(10): e1000604, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19798428

RESUMO

The enormous toll on human life during the 1918-1919 Spanish influenza pandemic is a constant reminder of the potential lethality of influenza viruses. With the declaration by the World Health Organization of a new H1N1 influenza virus pandemic, and with continued human cases of highly pathogenic H5N1 avian influenza virus infection, a better understanding of the host response to highly pathogenic influenza viruses is essential. To this end, we compared pathology and global gene expression profiles in bronchial tissue from macaques infected with either the reconstructed 1918 pandemic virus or the highly pathogenic avian H5N1 virus A/Vietnam/1203/04. Severe pathology was observed in respiratory tissues from 1918 virus-infected animals as early as 12 hours after infection, and pathology steadily increased at later time points. Although tissues from animals infected with A/Vietnam/1203/04 also showed clear signs of pathology early on, less pathology was observed at later time points, and there was evidence of tissue repair. Global transcriptional profiles revealed that specific groups of genes associated with inflammation and cell death were up-regulated in bronchial tissues from animals infected with the 1918 virus but down-regulated in animals infected with A/Vietnam/1203/04. Importantly, the 1918 virus up-regulated key components of the inflammasome, NLRP3 and IL-1beta, whereas these genes were down-regulated by A/Vietnam/1203/04 early after infection. TUNEL assays revealed that both viruses elicited an apoptotic response in lungs and bronchi, although the response occurred earlier during 1918 virus infection. Our findings suggest that the severity of disease in 1918 virus-infected macaques is a consequence of the early up-regulation of cell death and inflammatory related genes, in which additive or synergistic effects likely dictate the severity of tissue damage.


Assuntos
Inflamação/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/virologia , Animais , Brônquios/patologia , Brônquios/virologia , Surtos de Doenças , Expressão Gênica , Perfilação da Expressão Gênica , Marcação In Situ das Extremidades Cortadas , Inflamação/virologia , Macaca , Análise de Sequência com Séries de Oligonucleotídeos , Infecções por Orthomyxoviridae/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
PLoS Pathog ; 5(5): e1000438, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19461876

RESUMO

To support their replication, viruses take advantage of numerous cellular factors and processes. Recent large-scale screens have identified hundreds of such factors, yet little is known about how viruses exploit any of these. Influenza virus infection post-translationally activates P58(IPK), a cellular inhibitor of the interferon-induced, dsRNA-activated eIF2alpha kinase, PKR. Here, we report that infection of P58(IPK) knockout mice with influenza virus resulted in increased lung pathology, immune cell apoptosis, PKR activation, and mortality. Analysis of lung transcriptional profiles, including those induced by the reconstructed 1918 pandemic virus, revealed increased expression of genes associated with the cell death, immune, and inflammatory responses. These experiments represent the first use of a mammalian infection model to demonstrate the role of P58(IPK) in the antiviral response. Our results suggest that P58(IPK) represents a new class of molecule, a cellular inhibitor of the host defense (CIHD), as P58(IPK) is activated during virus infection to inhibit virus-induced apoptosis and inflammation to prolong host survival, even while prolonging viral replication.


Assuntos
Proteínas de Choque Térmico HSP40/metabolismo , Vírus da Influenza A/fisiologia , Infecções por Orthomyxoviridae/imunologia , Animais , Apoptose/genética , Fator de Iniciação 2 em Eucariotos/metabolismo , Proteínas de Choque Térmico HSP40/genética , Imunidade Inata , Inflamação , Vírus da Influenza A/patogenicidade , Interferon beta/genética , Interferon beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/mortalidade , Infecções por Orthomyxoviridae/virologia , Fosforilação , Replicação Viral/genética , eIF-2 Quinase/metabolismo
19.
J Virol ; 83(20): 10557-70, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19706713

RESUMO

The "Spanish influenza" of 1918 claimed an unprecedented number of lives, yet the determinants of virulence for this virus are still not fully understood. Here, we used functional genomics and an in vitro human lung epithelial cell infection model to define the global host transcriptional response to the eight-gene 1918 virus. To better understand the role of the 1918 virus NS1 gene, we also evaluated the host response to a reassortant 1918 virus containing the NS1 gene from A/Texas/36/91 (a seasonal isolate of human influenza virus), as well as the host response to a reassortant of A/Texas/36/91 containing the 1918 NS1 gene. Genomic analyses revealed that the 1918 virus blocked the transcription of multiple interferon-stimulated genes and also downregulated a network of genes associated with lipid metabolism. In contrast, the expression of genes encoding chemokines and cytokines, which serve to attract infiltrating immune cells, was upregulated. Viruses containing the NS1 gene from A/Texas/36/91 induced a significant increase in type I interferon signaling but did not repress lipid metabolism. The 1918 NS1 gene may therefore have contributed to the virulence of the 1918 pandemic virus by disrupting the innate immune response, inducing hypercytokinemia, and by blocking the transcription of certain lipid-based proinflammatory mediators that function as part of the host antiviral response.


Assuntos
Células Epiteliais/virologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Vírus da Influenza A Subtipo H1N1/patogenicidade , Interferons/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Pulmão/virologia , Proteínas não Estruturais Virais/metabolismo , Linhagem Celular , Surtos de Doenças , Células Epiteliais/citologia , Perfilação da Expressão Gênica , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A/genética , Vírus da Influenza A/imunologia , Vírus da Influenza A/patogenicidade , Influenza Humana/epidemiologia , Influenza Humana/imunologia , Influenza Humana/virologia , Interferons/metabolismo , Pulmão/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas/genética , Proteínas/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Ensaio de Placa Viral
20.
Cytokine Growth Factor Rev ; 18(5-6): 441-50, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17681845

RESUMO

We have always wanted to save the world from the scourges of virus infection by developing better drugs and vaccines. But fully understanding the intricacies of virus-host interactions, the first step in achieving this goal, requires the ability to view the process on a grand scale. The advent of high-throughput technologies, such as DNA microarrays and mass spectrometry, provided the first opportunities to obtain such a view. Here, we describe our efforts to use these tools to focus on the changes in cellular gene expression and protein abundance that occur in response to virus infection. By examining these changes in a comprehensive manner, we have been able to discover exciting new insights into innate immunity, interferon and cytokine signaling, and the strategies used by viruses to overcome these cellular defenses. Functional genomics may yet save the world from killer viruses.


Assuntos
Genômica , Interações Hospedeiro-Patógeno/genética , Vírus/patogenicidade , Síndrome da Imunodeficiência Adquirida/genética , Animais , Distinções e Prêmios , Biologia Computacional , Modelos Animais de Doenças , Expressão Gênica , Hepatite C/genética , Hepatite C/virologia , Humanos , Influenza Humana/genética , Fígado/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA