Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
J Infect Dis ; 228(9): 1263-1273, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37466213

ABSTRACT

BACKGROUND: Remdesivir is approved for treatment of coronavirus disease 2019 (COVID-19) in nonhospitalized and hospitalized adult and pediatric patients. Here we present severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) resistance analyses from the phase 3 ACTT-1 randomized placebo-controlled trial conducted in adult participants hospitalized with COVID-19. METHODS: Swab samples were collected at baseline and longitudinally through day 29. SARS-CoV-2 genomes were sequenced using next-generation sequencing. Phenotypic analysis was conducted directly on participant virus isolates and/or using SARS-CoV-2 subgenomic replicons expressing mutations identified in the Nsp12 target gene. RESULTS: Among participants with both baseline and postbaseline sequencing data, emergent Nsp12 substitutions were observed in 12 of 31 (38.7%) and 12 of 30 (40.0%) participants in the remdesivir and placebo arms, respectively. No emergent Nsp12 substitutions in the remdesivir arm were observed in more than 1 participant. Phenotyping showed low to no change in susceptibility to remdesivir relative to wild-type Nsp12 reference for the substitutions tested: A16V (0.8-fold change in EC50), P323L + V792I (2.2-fold), C799F (2.5-fold), K59N (1.0-fold), and K59N + V792I (3.4-fold). CONCLUSIONS: The similar rate of emerging Nsp12 substitutions in the remdesivir and placebo arms and the minimal change in remdesivir susceptibility among tested substitutions support a high barrier to remdesivir resistance development in COVID-19 patients. Clinical Trials Registration. NCT04280705.


Subject(s)
COVID-19 , Adult , Humans , Child , SARS-CoV-2/genetics , COVID-19 Drug Treatment , Adenosine Monophosphate/therapeutic use , Alanine/therapeutic use , Antiviral Agents/therapeutic use
2.
J Clin Virol ; 161: 105420, 2023 04.
Article in English | MEDLINE | ID: mdl-36913789

ABSTRACT

BACKGROUND: Guidelines for SARS-CoV-2 have relied on limited data on duration of viral infectiousness and correlation with COVID-19 symptoms and diagnostic testing. METHODS: We enrolled ambulatory adults with acute SARS-CoV-2 infection and performed serial measurements of COVID-19 symptoms, nasal swab viral RNA, nucleocapsid (N) and spike (S) antigens, and replication-competent SARS-CoV-2 by viral growth in culture. We determined average time from symptom onset to a first negative test result and estimated risk of infectiousness, as defined by positive viral growth in culture. RESULTS: Among 95 adults, median [interquartile range] time from symptom onset to first negative test result was 9 [5] days, 13 [6] days, 11 [4] days, and >19 days for S antigen, N antigen, culture growth, and viral RNA by RT-PCR, respectively. Beyond two weeks, virus growth and N antigen titers were rarely positive, while viral RNA remained detectable among half (26/51) of participants tested 21-30 days after symptom onset. Between 6-10 days from symptom onset, N antigen was strongly associated with culture positivity (relative risk=7.61, 95% CI: 3.01-19.22), whereas neither viral RNA nor symptoms were associated with culture positivity. During the 14 days following symptom onset, the presence of N antigen remained strongly associated (adjusted relative risk=7.66, 95% CI: 3.96-14.82) with culture positivity, regardless of COVID-19 symptoms. CONCLUSIONS: Most adults have replication-competent SARS-CoV-2 for 10-14 after symptom onset. N antigen testing is a strong predictor of viral infectiousness and may be a more suitable biomarker, rather than absence of symptoms or viral RNA, to discontinue isolation within two weeks from symptom onset.


Subject(s)
COVID-19 , Adult , Humans , COVID-19/diagnosis , SARS-CoV-2 , Longitudinal Studies , Diagnostic Techniques and Procedures , RNA, Viral , COVID-19 Testing
3.
J Immunol ; 210(9): 1247-1256, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36939421

ABSTRACT

Retinoic acid-inducible gene I (RIG-I) is essential for activating host cell innate immunity to regulate the immune response against many RNA viruses. We previously identified that a small molecule compound, KIN1148, led to the activation of IFN regulatory factor 3 (IRF3) and served to enhance protection against influenza A virus (IAV) A/California/04/2009 infection. We have now determined direct binding of KIN1148 to RIG-I to drive expression of IFN regulatory factor 3 and NF-κB target genes, including specific immunomodulatory cytokines and chemokines. Intriguingly, KIN1148 does not lead to ATPase activity or compete with ATP for binding but activates RIG-I to induce antiviral gene expression programs distinct from type I IFN treatment. When administered in combination with a vaccine against IAV, KIN1148 induces both neutralizing Ab and IAV-specific T cell responses compared with vaccination alone, which induces comparatively poor responses. This robust KIN1148-adjuvanted immune response protects mice from lethal A/California/04/2009 and H5N1 IAV challenge. Importantly, KIN1148 also augments human CD8+ T cell activation. Thus, we have identified a small molecule RIG-I agonist that serves as an effective adjuvant in inducing noncanonical RIG-I activation for induction of innate immune programs that enhance adaptive immune protection of antiviral vaccination.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza A virus , Influenza Vaccines , Influenza, Human , Humans , Animals , Mice , DEAD Box Protein 58/metabolism , Influenza A Virus, H5N1 Subtype/metabolism , Interferon Regulatory Factor-3/metabolism , Adjuvants, Immunologic , Antiviral Agents/pharmacology , Immunity, Innate
4.
J Infect Dis ; 219(7): 1162-1171, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30371803

ABSTRACT

BACKGROUND: A challenge to the design of improved therapeutic agents and prevention strategies for neuroinvasive infection and associated disease is the lack of known natural immune correlates of protection. A relevant model to study such correlates is offered by the Collaborative Cross (CC), a panel of recombinant inbred mouse strains that exhibit a range of disease manifestations upon infection. METHODS: We performed an extensive screen of CC-F1 lines infected with West Nile virus (WNV), including comprehensive immunophenotyping, to identify groups of lines that exhibited viral neuroinvasion or neuroinvasion with disease and lines that remained free of WNV neuroinvasion and disease. RESULTS: Our data reveal that protection from neuroinvasion and disease is multifactorial and that several immune outcomes can contribute. Immune correlates identified include decreased suppressive activity of regulatory T cells at steady state, which correlates with peripheral restriction of the virus. Further, a rapid contraction of WNV-specific CD8+ T cells in the brain correlated with protection from disease. CONCLUSIONS: These immune correlates of protection illustrate additional networks and pathways of the WNV immune response that cannot be observed in the C57BL/6 mouse model. Additionally, correlates of protection exhibited before infection, at baseline, provide insight into phenotypic differences in the human population that may predict clinical outcomes upon infection.


Subject(s)
Collaborative Cross Mice/immunology , Nervous System Diseases/immunology , West Nile Fever/immunology , West Nile virus/immunology , 2',5'-Oligoadenylate Synthetase/genetics , Adaptive Immunity , Animals , Brain/immunology , Brain/pathology , CD4-CD8 Ratio , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Collaborative Cross Mice/genetics , Disease Models, Animal , Heterozygote , Immunity, Innate , Immunophenotyping , Male , Mice , Mice, Inbred C57BL , Nervous System Diseases/microbiology , Polymorphism, Genetic , Spleen/immunology , Spleen/pathology , T-Lymphocytes, Regulatory/immunology , West Nile Fever/complications , West Nile Fever/genetics
5.
Mamm Genome ; 29(7-8): 593-602, 2018 08.
Article in English | MEDLINE | ID: mdl-29982912

ABSTRACT

Interferon-stimulated genes (ISGs) are the effectors of interferon (IFN) actions and play major roles in innate immune defense against microbial infection. During virus infection, ISGs impart antiviral actions to control virus replication and spread but can also contribute to disease pathology if their expression is unchecked. Antiviral ISGs have been identified by a variety of biochemical, genetic, and virologic methods. New computational approaches are expanding and redefining ISGs as responders to a variety of stimuli beyond IFNs, including virus infection, stress, and other events that induce cytokines. These studies reveal that the expression of ISG subsets link to interferon regulatory factors (IRF)s, NF-kB, and other transcription factors that impart gene expression in specific cell types independently of IFNs, including stem cells and other cell types where ISGs are constitutively expressed. Here, we provide a broad overview of ISGs, define virus-induced genes (VSG)s, and discuss the application of computational approaches and bioinformatics platforms to evaluate the functional role of ISGs in epigenetics, immune programming, and vaccine responses.


Subject(s)
Gene Expression Regulation , Interferons/metabolism , Quantitative Trait Loci , Animals , Computational Biology/methods , Genome-Wide Association Study/methods , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/genetics , Machine Learning , Molecular Sequence Annotation , Protein Binding , Response Elements , Signal Transduction , Vaccines/genetics , Vaccines/immunology
6.
Methods Mol Biol ; 1656: 119-129, 2017.
Article in English | MEDLINE | ID: mdl-28808965

ABSTRACT

Pathogen recognition receptors (PRR)s and their cognate pathogen-associated molecular pattern (PAMP) represent the basis of innate immune activation and immune response induction driven by the host-pathogen interaction that occurs during microbial infection in humans and other animals. For RNA virus infection such as hepatitis C virus (HCV) and others, specific motifs within viral RNA mark it as nonself and visible to the host as a PAMP through interaction with RIG-I-like receptors including retinoic inducible gene-I (RIG-I). Here, we present methods for producing and using HCV PAMP RNA as a molecular tool to study RIG-I and its signaling pathway, both in vitro and in vivo, in innate immune regulation.


Subject(s)
DEAD Box Protein 58 , Hepacivirus , Hepatitis C , Immunity, Innate , RNA, Viral , Receptors, Pattern Recognition , Animals , Cell Line , DEAD Box Protein 58/genetics , DEAD Box Protein 58/immunology , Hepacivirus/genetics , Hepacivirus/immunology , Hepatitis C/genetics , Hepatitis C/immunology , Humans , RNA, Viral/chemistry , RNA, Viral/genetics , RNA, Viral/immunology , Receptors, Immunologic , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/immunology
7.
G3 (Bethesda) ; 7(6): 1665-1682, 2017 06 07.
Article in English | MEDLINE | ID: mdl-28592649

ABSTRACT

The oligoadenylate-synthetase (Oas) gene locus provides innate immune resistance to virus infection. In mouse models, variation in the Oas1b gene influences host susceptibility to flavivirus infection. However, the impact of Oas variation on overall innate immune programming and global gene expression among tissues and in different genetic backgrounds has not been defined. We examined how Oas1b acts in spleen and brain tissue to limit West Nile virus (WNV) susceptibility and disease across a range of genetic backgrounds. The laboratory founder strains of the mouse Collaborative Cross (CC) (A/J, C57BL/6J, 129S1/SvImJ, NOD/ShiLtJ, and NZO/HlLtJ) all encode a truncated, defective Oas1b, whereas the three wild-derived inbred founder strains (CAST/EiJ, PWK/PhJ, and WSB/EiJ) encode a full-length OAS1B protein. We assessed disease profiles and transcriptional signatures of F1 hybrids derived from these founder strains. F1 hybrids included wild-type Oas1b (F/F), homozygous null Oas1b (N/N), and heterozygous offspring of both parental combinations (F/N and N/F). These mice were challenged with WNV, and brain and spleen samples were harvested for global gene expression analysis. We found that the Oas1b haplotype played a role in WNV susceptibility and disease metrics, but the presence of a functional Oas1b allele in heterozygous offspring did not absolutely predict protection against disease. Our results indicate that Oas1b status as wild-type or truncated, and overall Oas1b gene dosage, link with novel innate immune gene signatures that impact specific biological pathways for the control of flavivirus infection and immunity through both Oas1b-dependent and independent processes.


Subject(s)
2',5'-Oligoadenylate Synthetase/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Transcription, Genetic , West Nile Fever/genetics , West Nile Fever/immunology , West Nile virus/immunology , Animals , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Immunity, Innate/genetics , Immunomodulation/genetics , Immunomodulation/immunology , Male , Mice , Quantitative Trait Loci , Transcriptome , West Nile Fever/virology
8.
Vaccine ; 35(15): 1964-1971, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28279563

ABSTRACT

Vaccine adjuvants are essential to drive a protective immune response in cases where vaccine antigens are weakly immunogenic, where vaccine antigen is limited, or where an increase in potency is needed for a specific population, such as the elderly. To discover novel vaccine adjuvants, we used a high-throughput screen (HTS) designed to identify small-molecule agonists of the RIG-I-like receptor (RLR) pathway leading to interferon regulatory factor 3 (IRF3) activation. RLRs are a group of cytosolic pattern-recognition receptors that are essential for the recognition of viral nucleic acids during infection. Upon binding of viral nucleic acid ligands, the RLRs become activated and signal to transcription factors, including IRF3, to initiate an innate immune transcriptional program to control virus infection. Among our HTS hits were a series of benzothiazole compounds from which we designed the lead analog, KIN1148. KIN1148 induced dose-dependent IRF3 nuclear translocation and specific activation of IRF3-responsive promoters. Prime-boost immunization of mice with a suboptimal dose of a monovalent pandemic influenza split virus H1N1 A/California/07/2009 vaccine plus KIN1148 protected against a lethal challenge with mouse-adapted influenza virus (A/California/04/2009) and induced an influenza virus-specific IL-10 and Th2 response by T cells derived from lung and lung-draining lymph nodes. Prime-boost immunization with vaccine plus KIN1148, but not prime immunization alone, induced antibodies capable of inhibiting influenza virus hemagglutinin and neutralizing viral infectivity. Nevertheless, a single immunization with vaccine plus KIN1148 provided increased protection over vaccine alone and reduced viral load in the lungs after challenge. These findings suggest that protection was at least partially mediated by a cellular immune component and that the induction of Th2 and immunoregulatory cytokines by a KIN1148-adjuvanted vaccine may be particularly beneficial for ameliorating the immunopathogenesis that is associated with influenza viruses.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Benzothiazoles/administration & dosage , DEAD Box Protein 58/metabolism , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Interferon Regulatory Factor-3/metabolism , Adjuvants, Immunologic/isolation & purification , Animals , Benzothiazoles/isolation & purification , Cell Line , Disease Models, Animal , Drug Evaluation, Preclinical , Female , High-Throughput Screening Assays , Humans , Mice, Inbred C57BL , Orthomyxoviridae Infections/prevention & control , Receptors, Immunologic , Survival Analysis
9.
PLoS Pathog ; 12(11): e1005996, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27806117

ABSTRACT

Infection with West Nile virus (WNV) leads to a range of disease outcomes, including chronic infection, though lack of a robust mouse model of chronic WNV infection has precluded identification of the immune events contributing to persistent infection. Using the Collaborative Cross, a population of recombinant inbred mouse strains with high levels of standing genetic variation, we have identified a mouse model of persistent WNV disease, with persistence of viral loads within the brain. Compared to lines exhibiting no disease or marked disease, the F1 cross CC(032x013)F1 displays a strong immunoregulatory signature upon infection that correlates with restraint of the WNV-directed cytolytic response. We hypothesize that this regulatory T cell response sufficiently restrains the immune response such that a chronic infection can be maintained in the CNS. Use of this new mouse model of chronic neuroinvasive virus will be critical in developing improved strategies to prevent prolonged disease in humans.


Subject(s)
T-Lymphocytes, Regulatory/immunology , West Nile Fever/immunology , Animals , Chronic Disease , Disease Models, Animal , Female , Flow Cytometry , Humans , Male , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , West Nile virus/immunology
10.
Genom Data ; 10: 114-117, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27843766

ABSTRACT

Flaviviruses are hematophagous arthropod-viruses that pose global challenges to human health. Like Zika virus, West Nile Virus (WNV) is a flavivirus for which no approved vaccine exists [1]. The role host genetics play in early detection and response to WNV still remains largely unexplained. In order to capture the impact of genetic variation on innate immune responses, we studied gene expression following WNV infection using the collaborative cross (CC). The CC is a mouse genetics resource composed of hundreds of independently bred, octo-parental recombinant inbred mouse lines [2]. To accurately capture the host immune gene expression signatures of West Nile infection, we used the nanostring platform to evaluate expression in spleen tissue isolated from CC mice infected with WNV over a time course of 4, 7, and 12 days' post-infection [3]. Nanostring is a non-amplification based digital method to quantitate gene expression that uses color-coded molecular barcodes to detect hundreds of transcripts in a sample. Using this approach, we identified unique gene signatures in spleen tissue at days 4, 7, and 12 following WNV infection, which delineated distinct differences between asymptomatic and symptomatic CC lines. We also identified novel immune genes. Data was deposited into the Gene Expression Omnibus under accession GSE86000.

11.
Genom Data ; 10: 137-140, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27872814

ABSTRACT

West Nile Virus (WNV) is a mosquito-transmitted virus from the Flaviviridae family that causes fever in 1 in 5 infected people. WNV can also become neuro-invasive and cross the blood-brain barrier leading to severe neurological symptoms in a subset of WNV infected individuals [1]. WNV neuro-invasion is believed to be influenced by a number of factors including host genetics. In order to explore these effects and recapitulate the complex immune genetic differences among individuals, we studied gene expression following WNV infection in the Collaborative Cross (CC) model. The CC is a mouse genetics resource composed of > 70 independently bred, octo-parental recombinant inbred mouse lines [2]. To identify the individual host gene expression signatures influencing protection or susceptibility to WNV disease and WNV neuroinvasion, we used the nanostring nsolver platform to quantify gene expression in brain tissue isolated from WNV-infected CC mice at days 4, 7 and 12 post-infection [3]. This nanostring technology provided a high throughput, non-amplification based mRNA quantitation method to detect immune genes involved in neuro-invasion. Data was deposited into the Gene Expression Omnibus (GEO) under accession GSE85999.

12.
J Virol ; 90(5): 2372-87, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26676770

ABSTRACT

UNLABELLED: The cellular response to virus infection is initiated when pathogen recognition receptors (PRR) engage viral pathogen-associated molecular patterns (PAMPs). This process results in induction of downstream signaling pathways that activate the transcription factor interferon regulatory factor 3 (IRF3). IRF3 plays a critical role in antiviral immunity to drive the expression of innate immune response genes, including those encoding antiviral factors, type 1 interferon, and immune modulatory cytokines, that act in concert to restrict virus replication. Thus, small molecule agonists that can promote IRF3 activation and induce innate immune gene expression could serve as antivirals to induce tissue-wide innate immunity for effective control of virus infection. We identified small molecule compounds that activate IRF3 to differentially induce discrete subsets of antiviral genes. We tested a lead compound and derivatives for the ability to suppress infections caused by a broad range of RNA viruses. Compound administration significantly decreased the viral RNA load in cultured cells that were infected with viruses of the family Flaviviridae, including West Nile virus, dengue virus, and hepatitis C virus, as well as viruses of the families Filoviridae (Ebola virus), Orthomyxoviridae (influenza A virus), Arenaviridae (Lassa virus), and Paramyxoviridae (respiratory syncytial virus, Nipah virus) to suppress infectious virus production. Knockdown studies mapped this response to the RIG-I-like receptor pathway. This work identifies a novel class of host-directed immune modulatory molecules that activate IRF3 to promote host antiviral responses to broadly suppress infections caused by RNA viruses of distinct genera. IMPORTANCE: Incidences of emerging and reemerging RNA viruses highlight a desperate need for broad-spectrum antiviral agents that can effectively control infections caused by viruses of distinct genera. We identified small molecule compounds that can selectively activate IRF3 for the purpose of identifying drug-like molecules that can be developed for the treatment of viral infections. Here, we report the discovery of a hydroxyquinoline family of small molecules that can activate IRF3 to promote cellular antiviral responses. These molecules can prophylactically or therapeutically control infection in cell culture by pathogenic RNA viruses, including West Nile virus, dengue virus, hepatitis C virus, influenza A virus, respiratory syncytial virus, Nipah virus, Lassa virus, and Ebola virus. Our study thus identifies a class of small molecules with a novel mechanism to enhance host immune responses for antiviral activity against a variety of RNA viruses that pose a significant health care burden and/or that are known to cause infections with high case fatality rates.


Subject(s)
Antiviral Agents/pharmacology , Immunity, Innate/drug effects , Immunologic Factors/pharmacology , RNA Viruses/immunology , RNA Viruses/physiology , Virus Replication/drug effects , Animals , Antiviral Agents/isolation & purification , Cell Line , Gene Expression Profiling , Humans , Immunologic Factors/isolation & purification , Viral Load , Virus Cultivation
13.
Antiviral Res ; 108: 156-64, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24907428

ABSTRACT

Hepatitis C virus (HCV) causes 350,000 deaths and infects at least 3million people worldwide every year. Currently no vaccine has been developed. Direct-acting antiviral (DAA) drugs with high efficacy for suppressing HCV infection have recently been introduced into the clinic. While DAAs initially required combination therapy with type-1 interferon (IFN) administration for full efficacy and to avoid viral resistance to treatment, new DAA combinations show promise as an IFN-free regimen. However, IFN-free DAA therapy is in its infancy, still to be proven and today is cost-prohibitive for the patient. A major goal in HCV therapy to remove or replace IFN with DAAs or an alternative therapeutic to render virologic response with continued virus sensitivity to DAAs, thus facilitating a cure for infection. Recent advances in our understanding of innate immune responses to HCV have identified new therapeutic targets to combat HCV infection. We discuss how the targeting of innate immune response factors can be harnessed with DAAs to produce new generations of DAA-based HCV therapeutics. This article forms part of a symposium in Antiviral Research on "Hepatitis C: next steps toward global eradication."


Subject(s)
Antiviral Agents/therapeutic use , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Immunity, Innate , Immunologic Factors/therapeutic use , Biomedical Research/trends , Drug Discovery/trends , Hepacivirus/immunology , Hepatitis C, Chronic/immunology , Humans
14.
Curr Top Microbiol Immunol ; 363: 143-67, 2013.
Article in English | MEDLINE | ID: mdl-22903567

ABSTRACT

While 170 million people worldwide are chronically infected with HCV, the response rate to the current treatment regimens of pegylated IFN-α (IFN) in combination with ribavirin is only approximately 55 % of all HCV patients undergoing therapy. This IFN-based therapy is now slated to serve as the backbone for future combination therapeutics involving direct-acting antiviral compounds, including HCV protease inhibitors, viral polymerase inhibitors, and other small molecules. It is essential that the application of IFN be improved for overall enhancement of therapy outcome to effectively cure HCV infection. Systems approaches, including genomics and network modeling, are particularly powerful tools that are now being used to dissect the underlying mechanisms of successful or failed treatment response in an effort to design improved IFN-based therapeutic regimens. Furthermore, systems applications can be used to define virus-host interactions and map their variation within viral and host genomes, leading to identification of targets for novel therapy strategies. Using these approaches, we have defined distinct hepatic expression and tissue distribution of innate immune signaling molecules and gene networks that associate with IFN-based treatment outcome for HCV infection. This chapter will focus on using systems approaches to understand the host response to both HCV infection and therapy to drive the development of improved HCV therapeutics.


Subject(s)
Hepatitis C/drug therapy , Systems Biology/methods , Animals , Hepatitis C/immunology , Humans , Immune Evasion , Treatment Outcome
15.
Viruses ; 3(6): 906-19, 2011 06.
Article in English | MEDLINE | ID: mdl-21994761

ABSTRACT

The RNA helicase family of RIG-I-like receptors (RLRs) is a key component of host defense mechanisms responsible for detecting viruses and triggering innate immune signaling cascades to control viral replication and dissemination. As cytoplasm-based sensors, RLRs recognize foreign RNA in the cell and activate a cascade of antiviral responses including the induction of type I interferons, inflammasome activation, and expression of proinflammatory cytokines and chemokines. This review provides a brief overview of RLR function, ligand interactions, and downstream signaling events with an expanded discussion on the therapeutic potential of targeting RLRs for immune stimulation and treatment of virus infection.


Subject(s)
DEAD-box RNA Helicases/immunology , Immunity, Innate , Virus Diseases/enzymology , Virus Diseases/immunology , Animals , Antiviral Agents/therapeutic use , DEAD-box RNA Helicases/genetics , Enzyme Activators/therapeutic use , Humans , Virus Diseases/drug therapy , Virus Diseases/virology , Virus Physiological Phenomena , Viruses/drug effects
16.
PLoS One ; 4(11): e7916, 2009 Nov 19.
Article in English | MEDLINE | ID: mdl-19936259

ABSTRACT

BACKGROUND: The complexity of the human plasma proteome represents a substantial challenge for biomarker discovery. Proteomic analysis of genetically engineered mouse models of cancer and isolated cancer cells and cell lines provide alternative methods for identification of potential cancer markers that would be detectable in human blood using sensitive assays. The goal of this work is to evaluate the utility of an integrative strategy using these two approaches for biomarker discovery. METHODOLOGY/PRINCIPAL FINDINGS: We investigated a strategy that combined quantitative plasma proteomics of an ovarian cancer mouse model with analysis of proteins secreted or shed by human ovarian cancer cells. Of 106 plasma proteins identified with increased levels in tumor bearing mice, 58 were also secreted or shed from ovarian cancer cells. The remainder consisted primarily of host-response proteins. Of 25 proteins identified in the study that were assayed, 8 mostly secreted proteins common to mouse plasma and human cancer cells were significantly upregulated in a set of plasmas from ovarian cancer patients. Five of the eight proteins were confirmed to be upregulated in a second independent set of ovarian cancer plasmas, including in early stage disease. CONCLUSIONS/SIGNIFICANCE: Integrated proteomic analysis of cancer mouse models and human cancer cell populations provides an effective approach to identify potential circulating protein biomarkers.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/blood , Ovarian Neoplasms/metabolism , Proteomics/methods , Animals , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoblotting , Mass Spectrometry/methods , Mice , Models, Statistical , Neoplasm Transplantation , Proteome
17.
PLoS Med ; 5(6): e123, 2008 Jun 10.
Article in English | MEDLINE | ID: mdl-18547137

ABSTRACT

BACKGROUND: The complexity and heterogeneity of the human plasma proteome have presented significant challenges in the identification of protein changes associated with tumor development. Refined genetically engineered mouse (GEM) models of human cancer have been shown to faithfully recapitulate the molecular, biological, and clinical features of human disease. Here, we sought to exploit the merits of a well-characterized GEM model of pancreatic cancer to determine whether proteomics technologies allow identification of protein changes associated with tumor development and whether such changes are relevant to human pancreatic cancer. METHODS AND FINDINGS: Plasma was sampled from mice at early and advanced stages of tumor development and from matched controls. Using a proteomic approach based on extensive protein fractionation, we confidently identified 1,442 proteins that were distributed across seven orders of magnitude of abundance in plasma. Analysis of proteins chosen on the basis of increased levels in plasma from tumor-bearing mice and corroborating protein or RNA expression in tissue documented concordance in the blood from 30 newly diagnosed patients with pancreatic cancer relative to 30 control specimens. A panel of five proteins selected on the basis of their increased level at an early stage of tumor development in the mouse was tested in a blinded study in 26 humans from the CARET (Carotene and Retinol Efficacy Trial) cohort. The panel discriminated pancreatic cancer cases from matched controls in blood specimens obtained between 7 and 13 mo prior to the development of symptoms and clinical diagnosis of pancreatic cancer. CONCLUSIONS: Our findings indicate that GEM models of cancer, in combination with in-depth proteomic analysis, provide a useful strategy to identify candidate markers applicable to human cancer with potential utility for early detection.


Subject(s)
Biomarkers, Tumor/blood , Pancreatic Neoplasms/diagnosis , Proteome/metabolism , Animals , Humans , Mass Spectrometry , Mice , Pancreatic Neoplasms/blood , Proteomics/methods , RNA, Messenger/metabolism
18.
PLoS One ; 3(6): e2425, 2008 Jun 18.
Article in English | MEDLINE | ID: mdl-18560578

ABSTRACT

BACKGROUND: Elucidation of the repertoire of secreted and cell surface proteins of tumor cells is relevant to molecular diagnostics, tumor imaging and targeted therapies. We have characterized the cell surface proteome and the proteins released into the extra-cellular milieu of three ovarian cancer cell lines, CaOV3, OVCAR3 and ES2 and of ovarian tumor cells enriched from ascites fluid. METHODOLOGY AND FINDINGS: To differentiate proteins released into the media from protein constituents of media utilized for culture, cells were grown in the presence of [(13)C]-labeled lysine. A biotinylation-based approach was used to capture cell surface associated proteins. Our general experimental strategy consisted of fractionation of proteins from individual compartments followed by proteolytic digestion and LC-MS/MS analysis. In total, some 6,400 proteins were identified with high confidence across all specimens and fractions. CONCLUSIONS AND SIGNIFICANCE: Protein profiles of the cell lines had substantial similarity to the profiles of human ovarian cancer cells from ascites fluid and included protein markers known to be associated with ovarian cancer. Proteomic analysis indicated extensive shedding from extra-cellular domains of proteins expressed on the cell surface, and remarkably high secretion rates for some proteins (nanograms per million cells per hour). Cell surface and secreted proteins identified by in-depth proteomic profiling of ovarian cancer cells may provide new targets for diagnosis and therapy.


Subject(s)
Neoplasm Proteins/metabolism , Ovarian Neoplasms/metabolism , Proteomics , Cell Line, Tumor , Chromatography, Liquid , Extracellular Space/metabolism , Female , Humans , Tandem Mass Spectrometry
19.
J Cell Sci ; 121(Pt 3): 358-68, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18198190

ABSTRACT

EPHA2 receptor tyrosine kinase is overexpressed in several human cancer types and promotes malignancy. However, the mechanisms by which EPHA2 promotes tumor progression are not completely understood. Here we report that overexpression of a wild-type EPHA2, but not a signaling-defective cytoplasmic truncation mutant (DeltaC), in human mammary epithelial cells weakens E-cadherin-mediated cell-cell adhesion. Interestingly, the total level of cadherins and the composition of the adherens junction complexes were not affected, nor was the tyrosine phosphorylation of the cadherin complex components changed. By contrast, RhoA GTPase activity was significantly affected by modulating the EPHA2 activity in MCF-10A cells. Treatment with a ROCK kinase inhibitor rescued cell-cell adhesion defects in EPHA2-overexpressing cells, whereas expression of constitutively activated Rho disrupted adherens junctions in DeltaC-expressing cells. EPHA2-dependent Rho activation and destabilization of adherens junctions appeared to be regulated via a signaling pathway involving Src kinase, low molecular weight phosphotyrosine phosphatase (LMW-PTP) and p190 RhoGAP. EPHA2 interacted with both Src and LMW-PTP, and the interactions increased in EPHA2-overexpressing cells. In addition, LMW-PTP phosphatase activity was elevated, and this elevation was accompanied by a decrease in tyrosine phosphorylation of p190 RhoGAP and destabilization of cell-cell adhesion. Expression of either a dominant negative LMW-PTP mutant, C12S, or a wild-type p190 RhoGAP rescued adhesion defects in EPHA2-overexpressing cells. Together, these data suggest that EPHA2 promotes tumor malignancy through a mechanism involving RhoA-dependent destabilization of adherens junctions.


Subject(s)
Adherens Junctions/metabolism , Receptor, EphA2/metabolism , rhoA GTP-Binding Protein/metabolism , Cadherins/metabolism , Catenins/metabolism , Cell Adhesion/physiology , Cell Line , Female , Gene Expression , Guanine Nucleotide Exchange Factors/metabolism , Humans , Models, Biological , Mutation , Phosphorylation , Protein Tyrosine Phosphatases/metabolism , Receptor, EphA2/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/metabolism , Sequence Deletion , Signal Transduction , src-Family Kinases/metabolism
20.
Curr Cancer Drug Targets ; 5(3): 149-57, 2005 May.
Article in English | MEDLINE | ID: mdl-15892616

ABSTRACT

Eph receptors are a unique family of receptor tyrosine kinases (RTK) that play critical roles in embryonic patterning, neuronal targeting, and vascular development during normal embryogenesis. Eph RTKs and their ligands, the ephrins, are also frequently overexpressed in a variety of cancers and tumor cell lines. In particular, one family member, EphA2, is overexpressed in breast, prostate, lung, and colon cancers. Unlike traditional oncogenes that often function only in tumor cells, recent data show that Eph receptors mediate cell-cell interactions both in tumor cells and in the tumor microenvironment, namely the tumor stroma and tumor vasculature. Thus, EphA2 receptors are attractive targets for drug design, as targeting these molecules could simultaneously inhibit several aspects of tumor progression. This review focuses on the multiple roles of EphA2 in cancer progression, the mechanisms by which EphA2 inhibition may halt this progression, and the pre-clinical results of EphA2 inhibition in various cancer model systems.


Subject(s)
Antineoplastic Agents/pharmacology , Receptor, EphA2/drug effects , Animals , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Receptor, EphA2/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...