Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
J Virol ; 89(5): 2543-52, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25520505

ABSTRACT

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.


Subject(s)
Arenaviridae Infections/pathology , Arenaviridae Infections/virology , Hemorrhagic Fevers, Viral/pathology , Hemorrhagic Fevers, Viral/virology , Lujo virus/pathogenicity , Animals , Arenaviridae Infections/immunology , Cell Death , Disease Models, Animal , Gene Expression Profiling , Hemorrhagic Fevers, Viral/immunology , Inflammation/pathology , Killer Cells, Natural/immunology , Lassa Fever/pathology , Lassa Fever/virology , Lassa virus/pathogenicity , Leukocytes, Mononuclear/immunology , Macaca fascicularis , Time Factors
2.
Science ; 346(6212): 987-91, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25359852

ABSTRACT

Existing mouse models of lethal Ebola virus infection do not reproduce hallmark symptoms of Ebola hemorrhagic fever, neither delayed blood coagulation and disseminated intravascular coagulation nor death from shock, thus restricting pathogenesis studies to nonhuman primates. Here we show that mice from the Collaborative Cross panel of recombinant inbred mice exhibit distinct disease phenotypes after mouse-adapted Ebola virus infection. Phenotypes range from complete resistance to lethal disease to severe hemorrhagic fever characterized by prolonged coagulation times and 100% mortality. Inflammatory signaling was associated with vascular permeability and endothelial activation, and resistance to lethal infection arose by induction of lymphocyte differentiation and cellular adhesion, probably mediated by the susceptibility allele Tek. These data indicate that genetic background determines susceptibility to Ebola hemorrhagic fever.


Subject(s)
Disease Models, Animal , Genetic Predisposition to Disease , Hemorrhagic Fever, Ebola/genetics , Hemorrhagic Fever, Ebola/immunology , Host-Pathogen Interactions/genetics , Mice , Receptor, TIE-2/genetics , Alleles , Animals , Blood Coagulation/genetics , Capillary Permeability/genetics , Endothelium, Vascular/physiopathology , Hemorrhagic Fever, Ebola/blood , Liver/blood supply , Liver/metabolism , Liver/pathology , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Neovascularization, Physiologic/genetics
3.
mBio ; 4(3): e00165-13, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23631916

ABSTRACT

UNLABELLED: A novel human coronavirus (HCoV-EMC) was recently identified in the Middle East as the causative agent of a severe acute respiratory syndrome (SARS) resembling the illness caused by SARS coronavirus (SARS-CoV). Although derived from the CoV family, the two viruses are genetically distinct and do not use the same receptor. Here, we investigated whether HCoV-EMC and SARS-CoV induce similar or distinct host responses after infection of a human lung epithelial cell line. HCoV-EMC was able to replicate as efficiently as SARS-CoV in Calu-3 cells and similarly induced minimal transcriptomic changes before 12 h postinfection. Later in infection, HCoV-EMC induced a massive dysregulation of the host transcriptome, to a much greater extent than SARS-CoV. Both viruses induced a similar activation of pattern recognition receptors and the interleukin 17 (IL-17) pathway, but HCoV-EMC specifically down-regulated the expression of several genes within the antigen presentation pathway, including both type I and II major histocompatibility complex (MHC) genes. This could have an important impact on the ability of the host to mount an adaptive host response. A unique set of 207 genes was dysregulated early and permanently throughout infection with HCoV-EMC, and was used in a computational screen to predict potential antiviral compounds, including kinase inhibitors and glucocorticoids. Overall, HCoV-EMC and SARS-CoV elicit distinct host gene expression responses, which might impact in vivo pathogenesis and could orient therapeutic strategies against that emergent virus. IMPORTANCE: Identification of a novel coronavirus causing fatal respiratory infection in humans raises concerns about a possible widespread outbreak of severe respiratory infection similar to the one caused by SARS-CoV. Using a human lung epithelial cell line and global transcriptomic profiling, we identified differences in the host response between HCoV-EMC and SARS-CoV. This enables rapid assessment of viral properties and the ability to anticipate possible differences in human clinical responses to HCoV-EMC and SARS-CoV. We used this information to predict potential effective drugs against HCoV-EMC, a method that could be more generally used to identify candidate therapeutics in future disease outbreaks. These data will help to generate hypotheses and make rapid advancements in characterizing this new virus.


Subject(s)
Coronavirus/pathogenicity , Epithelial Cells/immunology , Epithelial Cells/virology , Host-Pathogen Interactions , Severe Acute Respiratory Syndrome/virology , Cell Line , Coronavirus/isolation & purification , Coronavirus/physiology , Humans , Immune Tolerance , Transcriptome , Virus Replication
4.
Virology ; 430(1): 43-52, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22608059

ABSTRACT

Hepatitis C virus/human immunodeficiency virus (HCV/HIV) coinfected patients demonstrate accelerated progression to severe liver injury in comparison to HCV monoinfected patients, although the underlying mechanisms are unclear owing to infection of separate tissue compartments with two distinct viral pathogens. Microarray analysis of paired liver biopsy and peripheral blood mononuclear cell (PBMC) specimens from HCV/HIV coinfected and HCV monoinfected patients identified a gene expression signature associated with increased inflammation and immune activation that was present only in liver and PBMC samples from coinfected patients. We also identified in these samples liver- and PBMC-specific signatures enriched with fibrogenic/hepatic stellate activation and proinflammatory genes, respectively. Finally, Bayesian networks were constructed by assimilating these data with existing data from liver and PBMC samples from other cohorts, augmenting enrichment of biologically important pathways and further indicating that chronic immune activation in HCV/HIV coinfection may exacerbate liver disease progression in coinfected patients.


Subject(s)
HIV Infections/complications , HIV Infections/immunology , Hepatitis C/complications , Hepatitis C/immunology , Leukocytes, Mononuclear/immunology , Liver/immunology , Lymphocyte Activation , Adult , Biopsy , Cytokines/biosynthesis , Female , Gene Expression Profiling , Hepatic Stellate Cells/immunology , Humans , Liver/pathology , Male , Microarray Analysis , Middle Aged
5.
J Virol ; 86(5): 2437-46, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22190720

ABSTRACT

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.


Subject(s)
GTP-Binding Proteins/immunology , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/immunology , Influenza, Human/mortality , Animals , Disease Resistance , Female , GTP-Binding Proteins/genetics , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/pathology , Influenza, Human/virology , Lung/immunology , Lung/pathology , Lung/virology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Myxovirus Resistance Proteins , Survival
6.
J Virol ; 85(24): 12972-81, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21994441

ABSTRACT

Innate immune deficiencies result in a spectrum of severe clinical outcomes following infection. In particular, there is a strong association between loss of the signal transducer and activator of transcription (Stat) pathway, breach of the blood-brain barrier (BBB), and virus-induced neuropathology. The gene signatures that characterize resistance, disease, and mortality in the virus-infected nervous system have not been defined. Herpes simplex virus type 1 (HSV-1) is commonly associated with encephalitis in humans, and humans and mice lacking Stat1 display increased susceptibility to HSV central nervous system (CNS) infections. In this study, two HSV-1 strains were used, KOS (wild type [WT]), and Δvhs, an avirulent recombinant lacking the virion host shutoff (vhs) function. In addition, two mouse strains were used: strain 129 (control) and a Stat1-deficient (Stat1(-/-)) strain. Using combinations of these virus and mouse strains, we established a model of infection resulting in three different outcomes: viral clearance without neurological disease (Δvhs infection of control mice), neurological disease followed by viral clearance (Δvhs infection of Stat1(-/-) mice and WT infection of control mice), or neurological disease followed by death (WT infection of Stat1(-/-) mice). Through the use of functional genomics on the infected brain stems, we determined gene signatures that were representative of the three infection outcomes. We demonstrated a pathological signature in the brain stem of Stat1-deficient mice characterized by upregulation of transcripts encoding chemokine receptors, inflammatory markers, neutrophil chemoattractants, leukocyte adhesion proteins, and matrix metalloproteases. Additionally, there was a greater than 100-fold increase in the inflammatory markers interleukin 1ß (IL-1ß) and IL-6. Consistent with this gene signature, we demonstrated profound CNS inflammation with a concomitant lethal breach of the BBB. Taken together, our results indicated an essential role for normal Stat1-dependent signaling in mediating a nonpathological immune response to viral CNS infection.


Subject(s)
Encephalitis, Herpes Simplex/immunology , Encephalitis, Herpes Simplex/prevention & control , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/pathogenicity , Keratitis, Herpetic/immunology , Keratitis, Herpetic/virology , STAT1 Transcription Factor/immunology , Animals , Brain Stem/pathology , Brain Stem/virology , Cytokines/biosynthesis , Gene Expression Profiling , Histocytochemistry , Keratitis, Herpetic/complications , Mice , Mice, Knockout , Microarray Analysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis
7.
J Virol ; 85(22): 11646-54, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21917952

ABSTRACT

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.


Subject(s)
Carbon-Carbon Double Bond Isomerases/metabolism , Hepacivirus/pathogenicity , Host-Pathogen Interactions , Mitochondria/enzymology , Virus Replication , Biopsy , Cell Line , Dodecenoyl-CoA Isomerase , Fatty Acids/metabolism , Gene Silencing , Hepatocytes/enzymology , Hepatocytes/virology , Humans , Liver/chemistry , Liver/pathology , Oxidation-Reduction , Proteome
8.
J Virol ; 84(24): 12576-88, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20926563

ABSTRACT

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.


Subject(s)
Biomarkers/metabolism , Gene Expression Profiling , Influenza A virus/pathogenicity , Orthomyxoviridae Infections/virology , Receptors, Interleukin-1 Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Signal Transduction/physiology , Animals , Blotting, Western , Cell Communication , Cell Movement , Dendritic Cells/metabolism , Dendritic Cells/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Pandemics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Triazoles , Tumor Necrosis Factor-alpha/physiology
9.
J Virol ; 84(15): 7613-24, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20504916

ABSTRACT

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.


Subject(s)
Inflammation/pathology , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/pathogenicity , Lipoxins/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Signal Transduction , Animals , Female , Gene Expression Regulation , Inflammation/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/pathogenicity , Lung/pathology , Lung/virology , Mice , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Survival Analysis , Virulence
10.
J Virol ; 84(4): 2027-37, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19939913

ABSTRACT

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.


Subject(s)
Influenza A virus/immunology , Influenza A virus/physiology , Receptor, Interferon alpha-beta/physiology , Virus Replication/immunology , Animals , Apoptosis/genetics , Base Sequence , Cells, Cultured , DNA, Viral/genetics , Dogs , Host-Pathogen Interactions/immunology , Inflammation/immunology , Inflammation/virology , Influenza A virus/genetics , Influenza A virus/pathogenicity , Mice , Mice, Knockout , NF-kappa B/metabolism , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction , Toll-Like Receptor 3/metabolism , Virulence , eIF-2 Kinase/metabolism
11.
PLoS Pathog ; 5(10): e1000604, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19798428

ABSTRACT

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.


Subject(s)
Inflammation/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/virology , Animals , Bronchi/pathology , Bronchi/virology , Disease Outbreaks , Gene Expression , Gene Expression Profiling , In Situ Nick-End Labeling , Inflammation/virology , Macaca , Oligonucleotide Array Sequence Analysis , Orthomyxoviridae Infections/immunology , Reverse Transcriptase Polymerase Chain Reaction
12.
PLoS Pathog ; 5(5): e1000438, 2009 May.
Article in English | MEDLINE | ID: mdl-19461876

ABSTRACT

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.


Subject(s)
HSP40 Heat-Shock Proteins/metabolism , Influenza A virus/physiology , Orthomyxoviridae Infections/immunology , Animals , Apoptosis/genetics , Eukaryotic Initiation Factor-2/metabolism , HSP40 Heat-Shock Proteins/genetics , Immunity, Innate , Inflammation , Influenza A virus/pathogenicity , Interferon-beta/genetics , Interferon-beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lung/metabolism , Lung/pathology , Lung/virology , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Phosphorylation , Virus Replication/genetics , eIF-2 Kinase/metabolism
13.
J Virol ; 80(15): 7600-12, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16840339

ABSTRACT

Herpes simplex virus type 1 (HSV-1) mutants lacking the ICP34.5 gene are severely attenuated in mouse models and have a significant growth defect in confluent mouse embryo fibroblasts. Previously, ICP34.5 was demonstrated to have a crucial role in evading the innate immune response to infection by mediating the dephosphorylation of eIF2alpha, a translation initiation factor phosphorylated by PKR during the antiviral response. To further understand the role of ICP34.5 in evasion of the antiviral response, we used transcriptional profiling to examine host cell gene expression in both wild-type and ICP34.5-null virus-infected mouse embryo fibroblasts over a time course of infection. Our study revealed that cells responded to infection within 3 h through PKR-dependent eIF2alpha phosphorylation and that the majority of up-regulated genes at 3 h postinfection were involved in the antiviral response. HSV-1 counters this response through early expression of ICP34.5 and dephosphorylation of eIF2alpha. By 12 h postinfection, the differences between the number and functional classification of genes differentially up- and down-regulated between wild-type and ICP34.5-null virus-infected cells were maximal. Specifically, in wild-type virus-infected cells, the majority of changed genes were involved in metabolic and biosynthetic processes, while in ICP34.5-null virus-infected cells, mostly antiviral genes were up-regulated. Further, ICP34.5-null virus-infected cells produced greater amounts of beta interferon than wild-type virus-infected cells. These results indicate that ICP34.5 expression and function at early times postinfection have a pivotal role in the ability of HSV-1 to gain control of the host cell and maintain an environment for successful viral replication.


Subject(s)
Fibroblasts/virology , Genomics , Herpes Simplex , Herpesvirus 1, Human/growth & development , Viral Proteins/physiology , Animals , Chlorocebus aethiops , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Profiling , Herpes Simplex/immunology , Herpes Simplex/metabolism , Herpes Simplex/virology , Humans , Mice , Mice, Knockout , Mutation , Oligonucleotide Array Sequence Analysis , Phosphorylation , Signal Transduction , Vero Cells/metabolism , Vero Cells/virology , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/physiology , eIF-2 Kinase/genetics , eIF-2 Kinase/physiology
14.
J Virol ; 78(19): 10420-32, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15367608

ABSTRACT

For most severe viral pandemics such as influenza and AIDS, the exact contribution of individual viral genes to pathogenicity is still largely unknown. A necessary step toward that understanding is a systematic comparison of different influenza virus strains at the level of transcriptional regulation in the host as a whole and interpretation of these complex genetic changes in the context of multifactorial clinical outcomes and pathology. We conducted a study by infecting pigtailed macaques (Macaca nemestrina) with a genetically reconstructed strain of human influenza H1N1 A/Texas/36/91 virus and hypothesized not only that these animals would respond to the virus similarly to humans, but that gene expression patterns in the lungs and tracheobronchial lymph nodes would fit into a coherent and complete picture of the host-virus interactions during infection. The disease observed in infected macaques simulated uncomplicated influenza in humans. Clinical signs and an antibody response appeared with induction of interferon and B-cell activation pathways, respectively. Transcriptional activation of inflammatory cells and apoptotic pathways coincided with gross and histopathological signs of inflammation, with tissue damage and concurrent signs of repair. Additionally, cDNA microarrays offered new evidence of the importance of cytotoxic T cells and natural killer cells throughout infection. With this experiment, we confirmed the suitability of the nonhuman primate model in the quest for understanding the individual and joint contributions of viral genes to influenza virus pathogenesis by using cDNA microarray technology and a reverse genetics approach.


Subject(s)
Disease Models, Animal , Gene Expression Profiling , Influenza A virus/pathogenicity , Macaca nemestrina , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Animals , Antigen Presentation , Apoptosis , B-Lymphocytes/immunology , Female , Immunity, Innate , Inflammation , Interferons/immunology , Killer Cells, Natural/immunology , Lung/chemistry , Lung/immunology , Lung/pathology , Lung/virology , Lymph Nodes/chemistry , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymph Nodes/virology , Lymphocyte Activation , Macrophages/immunology , Neutrophils/immunology , Oligonucleotide Array Sequence Analysis , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/virology , Oxidative Stress , RNA, Messenger/analysis , RNA, Messenger/isolation & purification , T-Lymphocytes, Cytotoxic/immunology
15.
J Virol ; 77(11): 6367-75, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12743294

ABSTRACT

Alpha/beta interferons (IFN-alpha/beta) induce potent antiviral and antiproliferative responses and are used to treat a wide range of human diseases, including chronic hepatitis C virus (HCV) infection. However, for reasons that remain poorly understood, many HCV isolates are resistant to IFN therapy. To better understand the nature of the cellular IFN response, we examined the effects of IFN treatment on global gene expression by using several types of human cells, including HeLa cells, liver cell lines, and primary fetal hepatocytes. In response to IFN, 50 of the approximately 4,600 genes examined were consistently induced in each of these cell types and another 60 were induced in a cell type-specific manner. A search for IFN-stimulated response elements (ISREs) in genomic DNA located upstream of IFN-stimulated genes revealed both previously identified and novel putative ISREs. To determine whether HCV can alter IFN-regulated gene expression, we performed microarray analyses on IFN-treated HeLa cells expressing the HCV nonstructural 5A (NS5A) protein and on IFN-treated Huh7 cells containing an HCV subgenomic replicon. NS5A partially blocked the IFN-mediated induction of 14 IFN-stimulated genes, an effect that may play a role in HCV resistance to IFN. This block may occur through repression of ISRE-mediated transcription, since NS5A also inhibited the IFN-mediated induction of a reporter gene driven from an ISRE-containing promoter. In contrast, the HCV replicon had very little effect on IFN-regulated gene expression. These differences highlight the importance of comparing results from multiple model systems when investigating complex phenomena such as the cellular response to IFN and viral mechanisms of IFN resistance.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation/drug effects , Hepacivirus/pathogenicity , Interferons/pharmacology , Proteins/metabolism , Viral Nonstructural Proteins/pharmacology , Cell Line , Computational Biology , HeLa Cells , Hepacivirus/genetics , Hepacivirus/metabolism , Hepatocytes , Humans , Interferon-alpha/pharmacology , Interferon-beta/pharmacology , Oligonucleotide Array Sequence Analysis , Proteins/genetics , Transcription, Genetic , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
16.
Cancer Res ; 63(4): 859-64, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12591738

ABSTRACT

Hepatocellular carcinoma (HCC) is a common primary cancer associated frequently with hepatitis C virus (HCV). To gain insight into the molecular mechanisms of hepatocarcinogenesis, and to identify potential HCC markers, we performed cDNA microarray analysis on surgical liver samples from 20 HCV-infected patients. RNA from individual tumors was compared with RNA isolated from adjacent nontumor tissue that was cirrhotic in all of the cases. Gene expression changes related to cirrhosis were filtered out using experiments in which pooled RNA from HCV-infected cirrhotic liver without tumors was compared with pooled RNA from normal liver. Expression of approximately 13,600 genes was analyzed using the advanced analysis tools of the Rosetta Resolver System. This analysis revealed a set of 50 potential HCC marker genes, which were up-regulated in the majority of the tumors analyzed, much more widely than common clinical markers such as cell proliferation-related genes. This HCC marker set contained several cancer-related genes, including serine/threonine kinase 15 (STK15), which has been implicated in chromosome segregation abnormalities but which has not been linked previously with liver cancer. In addition, a set of genes encoding secreted or plasma proteins was identified, including plasma glutamate carboxypeptidase (PGCP) and two secreted phospholipases A2 (PLA2G13 and PLA2G7). These genes may provide potential HCC serological markers because of their strong up-regulation in more than half of the tumors analyzed. Thus, high throughput methods coupled with high-order statistical analyses may result in the development of new diagnostic tools for liver malignancies.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/virology , Hepatitis C/complications , Hepatitis C/genetics , Liver Neoplasms/genetics , Liver Neoplasms/virology , Aged , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/metabolism , Cell Division/genetics , Cohort Studies , Female , Gene Expression Regulation, Neoplastic , Genetic Markers , Hepacivirus , Hepatitis C/metabolism , Humans , Liver Neoplasms/metabolism , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Phospholipases A/biosynthesis , Phospholipases A/genetics , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
17.
Proc Natl Acad Sci U S A ; 99(16): 10736-41, 2002 Aug 06.
Article in English | MEDLINE | ID: mdl-12149435

ABSTRACT

The NS1 protein of influenza A virus contributes to viral pathogenesis, primarily by enabling the virus to disarm the host cell type IFN defense system. We examined the downstream effects of NS1 protein expression during influenza A virus infection on global cellular mRNA levels by measuring expression of over 13,000 cellular genes in response to infection with wild-type and mutant viruses in human lung epithelial cells. Influenza A/PR/8/34 virus infection resulted in a significant induction of genes involved in the IFN pathway. Deletion of the viral NS1 gene increased the number and magnitude of expression of cellular genes implicated in the IFN, NF-kappaB, and other antiviral pathways. Interestingly, different IFN-induced genes showed different sensitivities to NS1-mediated inhibition of their expression. A recombinant virus with a C-terminal deletion in its NS1 gene induced an intermediate cellular mRNA expression pattern between wild-type and NS1 knockout viruses. Most significantly, a virus containing the 1918 pandemic NS1 gene was more efficient at blocking the expression of IFN-regulated genes than its parental influenza A/WSN/33 virus. Taken together, our results suggest that the cellular response to influenza A virus infection in human lung cells is significantly influenced by the sequence of the NS1 gene, demonstrating the importance of the NS1 protein in regulating the host cell response triggered by virus infection.


Subject(s)
Influenza, Human/immunology , Transcription, Genetic , Viral Nonstructural Proteins/immunology , Cell Line , Epithelial Cells/cytology , Gene Expression Profiling , Humans , Immunity, Innate , Influenza A virus/genetics , Influenza A virus/immunology , Influenza, Human/epidemiology , Influenza, Human/genetics , Interferon-beta/genetics , Lung , NF-kappa B/immunology , Viral Nonstructural Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL