Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Elife ; 122024 Mar 07.
Article in English | MEDLINE | ID: mdl-38451065

ABSTRACT

Investigating the human fallopian tube (FT) microbiota has significant implications for understanding the pathogenesis of ovarian cancer (OC). In this large prospective study, we collected swabs intraoperatively from the FT and other surgical sites as controls to profile the microbiota in the FT and to assess its relationship with OC. Eighty-one OC and 106 non-cancer patients were enrolled and 1001 swabs were processed for 16S rRNA gene PCR and sequencing. We identified 84 bacterial species that may represent the FT microbiota and found a clear shift in the microbiota of the OC patients when compared to the non-cancer patients. Of the top 20 species that were most prevalent in the FT of OC patients, 60% were bacteria that predominantly reside in the gastrointestinal tract, while 30% normally reside in the mouth. Serous carcinoma had higher prevalence of almost all 84 FT bacterial species compared to the other OC subtypes. The clear shift in the FT microbiota in OC patients establishes the scientific foundation for future investigation into the role of these bacteria in the pathogenesis of OC.


Subject(s)
Microbiota , Ovarian Neoplasms , Female , Humans , Fallopian Tubes , Prospective Studies , RNA, Ribosomal, 16S/genetics , Mouth
2.
Immunity ; 56(8): 1876-1893.e8, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37480848

ABSTRACT

Acute graft-versus-host disease (aGVHD) remains a major limitation of allogeneic stem cell transplantation (SCT), and severe intestinal manifestation is the major cause of early mortality. Intestinal microbiota control MHC class II (MHC-II) expression by ileal intestinal epithelial cells (IECs) that promote GVHD. Here, we demonstrated that genetically identical mice of differing vendor origins had markedly different intestinal microbiota and ileal MHC-II expression, resulting in discordant GVHD severity. We utilized cohousing and antibiotic treatment to characterize the bacterial taxa positively and negatively associated with MHC-II expression. A large proportion of bacterial MHC-II inducers were vancomycin sensitive, and peri-transplant oral vancomycin administration attenuated CD4+ T cell-mediated GVHD. We identified a similar relationship between pre-transplant microbes, HLA class II expression, and both GVHD and mortality in a large clinical SCT cohort. These data highlight therapeutically tractable mechanisms by which pre-transplant microbial taxa contribute to GVHD independently of genetic disparity.


Subject(s)
Gastrointestinal Microbiome , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Mice , Animals , Vancomycin , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Transplantation, Homologous/adverse effects
3.
mSystems ; 8(4): e0067822, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37350639

ABSTRACT

Metaproteomics, a method for untargeted, high-throughput identification of proteins in complex samples, provides functional information about microbial communities and can tie functions to specific taxa. Metaproteomics often generates less data than other omics techniques, but analytical workflows can be improved to increase usable data in metaproteomic outputs. Identification of peptides in the metaproteomic analysis is performed by comparing mass spectra of sample peptides to a reference database of protein sequences. Although these protein databases are an integral part of the metaproteomic analysis, few studies have explored how database composition impacts peptide identification. Here, we used cervicovaginal lavage (CVL) samples from a study of bacterial vaginosis (BV) to compare the performance of databases built using six different strategies. We evaluated broad versus sample-matched databases, as well as databases populated with proteins translated from metagenomic sequencing of the same samples versus sequences from public repositories. Smaller sample-matched databases performed significantly better, driven by the statistical constraints on large databases. Additionally, large databases attributed up to 34% of significant bacterial hits to taxa absent from the sample, as determined orthogonally by 16S rRNA gene sequencing. We also tested a set of hybrid databases which included bacterial proteins from NCBI RefSeq and translated bacterial genes from the samples. These hybrid databases had the best overall performance, identifying 1,068 unique human and 1,418 unique bacterial proteins, ~30% more than a database populated with proteins from typical vaginal bacteria and fungi. Our findings can help guide the optimal identification of proteins while maintaining statistical power for reaching biological conclusions. IMPORTANCE Metaproteomic analysis can provide valuable insights into the functions of microbial and cellular communities by identifying a broad, untargeted set of proteins. The databases used in the analysis of metaproteomic data influence results by defining what proteins can be identified. Moreover, the size of the database impacts the number of identifications after accounting for false discovery rates (FDRs). Few studies have tested the performance of different strategies for building a protein database to identify proteins from metaproteomic data and those that have largely focused on highly diverse microbial communities. We tested a range of databases on CVL samples and found that a hybrid sample-matched approach, using publicly available proteins from organisms present in the samples, as well as proteins translated from metagenomic sequencing of the samples, had the best performance. However, our results also suggest that public sequence databases will continue to improve as more bacterial genomes are published.


Subject(s)
Microbiota , Proteomics , Female , Humans , RNA, Ribosomal, 16S/genetics , Proteomics/methods , Microbiota/genetics , Bacterial Proteins/genetics , Peptides/metabolism , Bacteria
4.
Clin Vaccine Immunol ; 22(3): 354-6, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25589554

ABSTRACT

Vesicular stomatitis virus expressing Zaire Ebola virus (EBOV) glycoprotein (VSVΔG/EBOVgp) could be used as a vaccine to meet the 2014 Ebola virus outbreak. To characterize the host response to this vaccine, we used mRNA sequencing to analyze peripheral blood mononuclear cells (PBMCs) from cynomolgus macaques after VSVΔG/EBOVgp immunization and subsequent EBOV challenge. We found a controlled transcriptional response that transitioned to immune regulation as the EBOV was cleared. This observation supports the safety of the vaccine.


Subject(s)
Ebola Vaccines/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Leukocytes, Mononuclear/immunology , Vesiculovirus/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Viral/blood , Democratic Republic of the Congo , Ebolavirus/genetics , Ebolavirus/pathogenicity , Gene Expression , High-Throughput Nucleotide Sequencing , Leukocytes, Mononuclear/metabolism , Macaca fascicularis , Sequence Analysis, RNA , Time Factors , Transcriptome , Vaccines, Synthetic/immunology
5.
mBio ; 5(4)2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25118237

ABSTRACT

UNLABELLED: In March 2013, three fatal human cases of infection with influenza A virus (H7N9) were reported in China. Since then, human cases have been accumulating. Given the public health importance of this virus, we performed a pathogenicity study of the H7N9 virus in the cynomolgus macaque model, focusing on clinical aspects of disease, radiographic, histological, and gene expression profile changes in the upper and lower respiratory tracts, and changes in systemic cytokine and chemokine profiles during infection. Cynomolgus macaques developed transient, mild to severe disease with radiographic evidence of pulmonary infiltration. Virus replicated in the upper as well as lower respiratory tract, with sustained replication in the upper respiratory tract until the end of the experiment at 6 days after inoculation. Virus shedding occurred mainly via the throat. Histopathological changes in the lungs were similar to those observed in humans, albeit less severe, with diffuse alveolar damage, infiltration of polymorphonuclear cells, formation of hyaline membranes, pneumocyte hyperplasia, and fibroproliferative changes. Analysis of gene expression profiles in lung lesions identified pathways involved in tissue damage during H7N9 infection as well as leads for development of therapeutics targeting host responses rather than virus replication. Overall, H7N9 infection was not as severe in cynomolgus macaques as in humans, supporting the possible role of underlying medical complications in disease severity as discussed for human H7N9 infection (H. N. Gao et al., N. Engl. J. Med. 368:2277-2285, 2013, doi:10.1056/NEJMoa1305584). IMPORTANCE: Influenza A virus H7N9 emerged early in 2013, and human cases have continued to emerge since then. Although H7N9 virus-induced disease in humans is often very severe and even lethal, the majority of reported H7N9 cases occurred in older people and people with underlying medical conditions. To better understand the pathogenicity of this virus, healthy cynomolgus macaques were inoculated with influenza A virus H7N9. Cynomolgus macaques were used as a model because the receptor distribution for H7N9 virus in macaques was recently shown to be more similar to that in humans than that of other frequently used animal models. From comparison with previous studies, we conclude that the emerging H7N9 influenza virus was more pathogenic in cynomolgus macaques than seasonal influenza A viruses and most isolates of the pandemic H1N1 virus but less pathogenic than the 1918 Spanish influenza virus or highly pathogenic avian influenza (HPAI) H5N1 virus.


Subject(s)
Influenza A Virus, H7N9 Subtype/physiology , Influenza A Virus, H7N9 Subtype/pathogenicity , Influenza, Human/virology , Lung/virology , Orthomyxoviridae Infections/virology , Virus Replication , Animals , Bronchi/virology , Cytokines/blood , Dogs , Humans , Influenza A Virus, H5N1 Subtype , Influenza in Birds/physiopathology , Influenza, Human/genetics , Influenza, Human/pathology , Lung/diagnostic imaging , Lung/pathology , Macaca fascicularis , Madin Darby Canine Kidney Cells , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/pathology , Pharynx/virology , Poultry/virology , Radiography , Trachea/virology , Transcriptome , Turbinates/virology , Viral Load
6.
PLoS Pathog ; 9(2): e1003168, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23544010

ABSTRACT

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.


Subject(s)
Immunity, Cellular/genetics , Immunity, Innate/genetics , Viral Tropism/genetics , West Nile Fever/immunology , West Nile virus/immunology , West Nile virus/physiology , Animals , Gene Regulatory Networks/immunology , Genes/physiology , Humans , Interferon Type I/metabolism , Interferon Type I/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Immunologic/physiology , Signal Transduction/genetics , Signal Transduction/immunology , Systems Biology/methods , West Nile Fever/genetics
7.
Nat Med ; 19(4): 458-64, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23455712

ABSTRACT

Although susceptibility of neurons in the brain to microbial infection is a major determinant of clinical outcome, little is known about the molecular factors governing this vulnerability. Here we show that two types of neurons from distinct brain regions showed differential permissivity to replication of several positive-stranded RNA viruses. Granule cell neurons of the cerebellum and cortical neurons from the cerebral cortex have unique innate immune programs that confer differential susceptibility to viral infection ex vivo and in vivo. By transducing cortical neurons with genes that were expressed more highly in granule cell neurons, we identified three interferon-stimulated genes (ISGs; Ifi27, Irg1 and Rsad2 (also known as Viperin)) that mediated the antiviral effects against different neurotropic viruses. Moreover, we found that the epigenetic state and microRNA (miRNA)-mediated regulation of ISGs correlates with enhanced antiviral response in granule cell neurons. Thus, neurons from evolutionarily distinct brain regions have unique innate immune signatures, which probably contribute to their relative permissiveness to infection.


Subject(s)
Brain/virology , Immunity, Innate/immunology , Neurons/virology , RNA Virus Infections/immunology , Animals , Brain/cytology , Brain/immunology , Cerebellum/cytology , Cerebellum/immunology , Cerebellum/virology , Cerebral Cortex/cytology , Cerebral Cortex/immunology , Cerebral Cortex/virology , Disease Susceptibility/immunology , Epigenesis, Genetic/immunology , Female , Gene Expression Regulation, Viral/immunology , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/physiology , Neurons/immunology , RNA Virus Infections/virology , Tissue Array Analysis
8.
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
9.
Virology ; 429(1): 37-46, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22542004

ABSTRACT

Human immunodeficiency virus (HIV-1) depends upon host-encoded proteins to facilitate its replication while at the same time inhibiting critical components of innate and/or intrinsic immune response pathways. To characterize the host cell response on protein levels in CD4+ lymphoblastoid SUP-T1 cells after infection with HIV-1 strain LAI, we used mass spectrometry (MS)-based global quantitation with iTRAQ (isobaric tag for relative and absolute quantification). We found 266, 60 and 22 proteins differentially expressed (DE) (P-value ≤ 0.05) at 4, 8, and 20 hours post-infection (hpi), respectively, compared to time-matched mock-infected samples. The majority of changes in protein abundance occurred at an early stage of infection well before the de novo production of viral proteins. Functional analyses of these DE proteins showed enrichment in several biological pathways including protein synthesis, cell proliferation, and T-cell activation. Importantly, these early changes before the time of robust viral production have not been described before.


Subject(s)
CD4-Positive T-Lymphocytes/chemistry , HIV Infections/genetics , HIV-1/physiology , Proteomics , T-Lymphocytes/chemistry , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Cell Line , Cell Proliferation , Gene Regulatory Networks , HIV Infections/immunology , HIV Infections/physiopathology , HIV Infections/virology , HIV-1/genetics , Host-Pathogen Interactions , Humans , Protein Biosynthesis , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/virology
10.
J Virol ; 86(13): 7334-44, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22532686

ABSTRACT

There is a growing need for novel antiviral therapies that are broad spectrum, effective, and not subject to resistance due to viral mutations. Using high-throughput screening methods, including computational docking studies and an interferon-stimulated gene 54 (ISG54)-luciferase reporter assay, we identified a class of isoflavone compounds that act as specific agonists of innate immune signaling pathways and cause activation of the interferon regulatory factor (IRF-3) transcription factor. The isoflavone compounds activated the ISG54 promoter, mediated nuclear translocation of IRF-3, and displayed highly potent activity against hepatitis C virus (HCV) and influenza virus. Additionally, these agonists efficiently activated IRF-3 in the presence of the HCV protease NS3-4A, which is known to blunt the host immune response. Furthermore, genomic studies showed that discrete innate immune pathways centered on IRF signaling were regulated following agonist treatment without causing global changes in host gene expression. Following treatment, the expression of only 64 cellular genes was significantly induced. This report provides the first evidence that innate immune pathways dependent on IRF-3 can be successfully targeted by small-molecule drugs for the development of novel broad-spectrum antiviral compounds.


Subject(s)
Antiviral Agents/metabolism , Hepacivirus/immunology , Immunologic Factors/metabolism , Interferon Regulatory Factor-3/biosynthesis , Isoflavones/agonists , Orthomyxoviridae/immunology , Signal Transduction/drug effects , Hepacivirus/physiology , Humans , Immunity, Innate , Orthomyxoviridae/physiology , Protein Transport , Virus Replication
11.
Hepatology ; 56(1): 28-38, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22331615

ABSTRACT

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.


Subject(s)
Hepacivirus/metabolism , Hepatitis C/complications , Liver Cirrhosis/pathology , Liver Transplantation/pathology , Protein Array Analysis/methods , Proteome/metabolism , Adult , Aged , Biopsy, Needle , Chromatography, Liquid/methods , Cohort Studies , Diagnosis, Computer-Assisted/methods , Disease Progression , Female , Graft Rejection , Graft Survival , Hepacivirus/pathogenicity , Hepatitis C/pathology , Humans , Immunohistochemistry , Liver Cirrhosis/etiology , Liver Cirrhosis/surgery , Liver Transplantation/adverse effects , Male , Mass Spectrometry/methods , Middle Aged , Oxidative Stress/physiology , Proteome/genetics , Proteomics/methods , Recurrence , Reference Values , Risk Assessment , Sampling Studies , Sensitivity and Specificity
12.
Hepatology ; 56(1): 17-27, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22278598

ABSTRACT

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.


Subject(s)
Graft Rejection/genetics , Hepatitis C, Chronic/complications , Liver Failure/surgery , Liver Transplantation/adverse effects , Transcriptional Activation/genetics , Aged , Biopsy, Needle , Cohort Studies , Disease Progression , Female , Gene Expression Regulation , Hepacivirus/genetics , Hepacivirus/physiology , Hepatitis C, Chronic/pathology , Hepatitis C, Chronic/surgery , Humans , Immunohistochemistry , Liver Failure/etiology , Liver Failure/genetics , Liver Transplantation/methods , Longitudinal Studies , Male , Middle Aged , Postoperative Complications/genetics , Postoperative Complications/physiopathology , Prognosis , Recurrence , Risk Assessment , Severity of Illness Index , Tissue Donors
13.
J Gen Virol ; 92(Pt 9): 2093-2104, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21593271

ABSTRACT

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.


Subject(s)
Antigen Presentation , Influenza A Virus, H1N1 Subtype/immunology , Interferon-alpha/antagonists & inhibitors , Interferon-beta/antagonists & inhibitors , Proteasome Inhibitors , Viral Nonstructural Proteins/metabolism , Virulence Factors/metabolism , Cells, Cultured , Epithelial Cells/immunology , Epithelial Cells/virology , Gene Expression Profiling , Humans , Influenza A Virus, H1N1 Subtype/genetics , Mutant Proteins/genetics , Mutant Proteins/immunology , Mutant Proteins/metabolism , NF-kappa B/antagonists & inhibitors , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology , Virulence Factors/immunology
14.
mBio ; 2(1): e00325-10, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21285433

ABSTRACT

The twentieth century was marked by extraordinary advances in our understanding of microbes and infectious disease, but pandemics remain, food and waterborne illnesses are frequent, multidrug-resistant microbes are on the rise, and the needed drugs and vaccines have not been developed. The scientific approaches of the past-including the intense focus on individual genes and proteins typical of molecular biology-have not been sufficient to address these challenges. The first decade of the twenty-first century has seen remarkable innovations in technology and computational methods. These new tools provide nearly comprehensive views of complex biological systems and can provide a correspondingly deeper understanding of pathogen-host interactions. To take full advantage of these innovations, the National Institute of Allergy and Infectious Diseases recently initiated the Systems Biology Program for Infectious Disease Research. As participants of the Systems Biology Program, we think that the time is at hand to redefine the pathogen-host research paradigm.


Subject(s)
Communicable Diseases , Host-Pathogen Interactions , Systems Biology/methods , Communicable Diseases/genetics , Communicable Diseases/immunology , Communicable Diseases/metabolism , Humans
15.
J Virol ; 85(3): 1214-23, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21084481

ABSTRACT

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.


Subject(s)
Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H1N1 Subtype/pathogenicity , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Primate Diseases/pathology , Primate Diseases/virology , Animals , Child, Preschool , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Gene Expression Profiling , Genetic Variation , Humans , Influenza, Human/virology , Macaca , Male , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Respiratory Tract Infections/pathology , Respiratory Tract Infections/virology , Severity of Illness Index , Virulence
16.
PLoS Pathog ; 6(10): e1001139, 2010 Oct 07.
Article in English | MEDLINE | ID: mdl-20949022

ABSTRACT

Since the first recorded infection of humans with H5N1 viruses of avian origin in 1997, sporadic human infections continue to occur with a staggering mortality rate of >60%. Although sustained human-to-human transmission has not occurred yet, there is a growing concern that these H5N1 viruses might acquire this trait and raise the specter of a pandemic. Despite progress in deciphering viral determinants of pathogenicity, we still lack crucial information on virus/immune system interactions pertaining to severe disease and high mortality associated with human H5N1 influenza virus infections. Using two human isolates of H5N1 viruses that differ in their pathogenicity in mice, we have defined mechanistic links among the rate of viral replication, mortality, CD8 T cell responses, and immunopathology. The extreme pathogenicity of H5N1 viruses was directly linked to the ability of the virus to replicate rapidly, and swiftly attain high steady-state titers in the lungs within 48 hours after infection. The remarkably high replication rate of the highly pathogenic H5N1 virus did not prevent the induction of IFN-ß or activation of CD8 T cells, but the CD8 T cell response was ineffective in controlling viral replication in the lungs and CD8 T cell deficiency did not affect viral titers or mortality. Additionally, BIM deficiency ameliorated lung pathology and inhibited T cell apoptosis without affecting survival of mice. Therefore, rapidly replicating, highly lethal H5N1 viruses could simply outpace and overwhelm the adaptive immune responses, and kill the host by direct cytopathic effects. However, therapeutic suppression of early viral replication and the associated enhancement of CD8 T cell responses improved the survival of mice following a lethal H5N1 infection. These findings suggest that suppression of early H5N1 virus replication is key to the programming of an effective host response, which has implications in treatment of this infection in humans.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/physiology , Orthomyxoviridae Infections/diagnosis , Orthomyxoviridae Infections/immunology , Virus Replication/physiology , Animals , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Dogs , Humans , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza, Human/diagnosis , Influenza, Human/drug therapy , Influenza, Human/genetics , Influenza, Human/immunology , Lung Diseases/etiology , Lung Diseases/immunology , Lung Diseases/virology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/genetics , Oseltamivir/therapeutic use , Prognosis , Virus Replication/immunology
17.
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
18.
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
19.
PLoS Pathog ; 6(1): e1000719, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20062526

ABSTRACT

Proteomic and lipidomic profiling was performed over a time course of acute hepatitis C virus (HCV) infection in cultured Huh-7.5 cells to gain new insights into the intracellular processes influenced by this virus. Our proteomic data suggest that HCV induces early perturbations in glycolysis, the pentose phosphate pathway, and the citric acid cycle, which favor host biosynthetic activities supporting viral replication and propagation. This is followed by a compensatory shift in metabolism aimed at maintaining energy homeostasis and cell viability during elevated viral replication and increasing cellular stress. Complementary lipidomic analyses identified numerous temporal perturbations in select lipid species (e.g. phospholipids and sphingomyelins) predicted to play important roles in viral replication and downstream assembly and secretion events. The elevation of lipotoxic ceramide species suggests a potential link between HCV-associated biochemical alterations and the direct cytopathic effect observed in this in vitro system. Using innovative computational modeling approaches, we further identified mitochondrial fatty acid oxidation enzymes, which are comparably regulated during in vitro infection and in patients with histological evidence of fibrosis, as possible targets through which HCV regulates temporal alterations in cellular metabolic homeostasis.


Subject(s)
Hepacivirus/physiology , Lipids/analysis , Liver/metabolism , Liver/virology , Proteins/analysis , Cell Line, Tumor , Chromatography, Liquid , Energy Metabolism/physiology , Humans , Mass Spectrometry , Proteins/metabolism , Proteome , Virus Replication
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...