Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
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
2.
Immunity ; 38(1): 3-5, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23352217

ABSTRACT

In this issue of Immunity, studies by Blanc et al. (2013) and Liu et al. (2013) reveal how interferon induction of cholesterol-25-hydroxylase mediates innate immunity against multiple diverse viruses.

3.
PLoS Pathog ; 15(8): e1007899, 2019 08.
Article in English | MEDLINE | ID: mdl-31415679

ABSTRACT

West Nile Virus (WNV), an emerging and re-emerging RNA virus, is the leading source of arboviral encephalitic morbidity and mortality in the United States. WNV infections are acutely controlled by innate immunity in peripheral tissues outside of the central nervous system (CNS) but WNV can evade the actions of interferon (IFN) to facilitate CNS invasion, causing encephalitis, encephalomyelitis, and death. Recent studies indicate that STimulator of INterferon Gene (STING), canonically known for initiating a type I IFN production and innate immune response to cytosolic DNA, is required for host defense against neurotropic RNA viruses. We evaluated the role of STING in host defense to control WNV infection and pathology in a murine model of infection. When challenged with WNV, STING knock out (-/-) mice displayed increased morbidity and mortality compared to wild type (WT) mice. Virologic analysis and assessment of STING activation revealed that STING signaling was not required for control of WNV in the spleen nor was WNV sufficient to mediate canonical STING activation in vitro. However, STING-/- mice exhibited a clear trend of increased viral load and virus dissemination in the CNS. We found that STING-/- mice exhibited increased and prolonged neurological signs compared to WT mice. Pathological examination revealed increased lesions, mononuclear cellular infiltration and neuronal death in the CNS of STING-/- mice, with sustained pathology after viral clearance. We found that STING was required in bone marrow derived macrophages for early control of WNV replication and innate immune activation. In vivo, STING-/- mice developed an aberrant T cell response in both the spleen and brain during WNV infection that linked with increased and sustained CNS pathology compared to WT mice. Our findings demonstrate that STING plays a critical role in immune programming for the control of neurotropic WNV infection and CNS disease.


Subject(s)
Central Nervous System/immunology , Central Nervous System/pathology , Immunity, Innate/immunology , Membrane Proteins/physiology , Virus Replication , West Nile Fever/immunology , West Nile virus/immunology , Animals , Central Nervous System/metabolism , Central Nervous System/virology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Viral Load , West Nile Fever/metabolism , West Nile Fever/virology
4.
J Immunol ; 201(10): 3036-3050, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30297339

ABSTRACT

We examined the signaling pathways and cell type-specific responses of IFN regulatory factor (IRF) 5, an immune-regulatory transcription factor. We show that the protein kinases IKKα, IKKß, IKKε, and TANK-binding kinase 1 each confer IRF5 phosphorylation/dimerization, thus extending the family of IRF5 activator kinases. Among primary human immune cell subsets, we found that IRF5 is most abundant in plasmacytoid dendritic cells (pDCs). Flow cytometric cell imaging revealed that IRF5 is specifically activated by endosomal TLR signaling. Comparative analyses revealed that IRF3 is activated in pDCs uniquely through RIG-I-like receptor (RLR) signaling. Transcriptomic analyses of pDCs show that the partitioning of TLR7/IRF5 and RLR/IRF3 pathways confers differential gene expression and immune cytokine production in pDCs, linking IRF5 with immune regulatory and proinflammatory gene expression. Thus, TLR7/IRF5 and RLR-IRF3 partitioning serves to polarize pDC response outcome. Strategies to differentially engage IRF signaling pathways should be considered in the design of immunotherapeutic approaches to modulate or polarize the immune response for specific outcome.


Subject(s)
Dendritic Cells/immunology , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factors/immunology , Signal Transduction/immunology , Cells, Cultured , Dendritic Cells/metabolism , Gene Expression Regulation/immunology , Humans , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factors/metabolism
5.
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
6.
Proc Natl Acad Sci U S A ; 111(5): 1909-14, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24449862

ABSTRACT

In response to viral infection, the host induces over 300 IFN-stimulated genes (ISGs), which are the central component of intracellular antiviral innate immunity. Inefficient induction of ISGs contributes to poor control and persistence of hepatitis C virus infection. Therefore, further understanding of the hepatocytic ISG regulation machinery will guide us to an improved management strategy against hepatitis C virus infection. In this study, comprehensive genome-wide, high-throughput cDNA screening for genes regulating ISG expression identified a tyrosine kinase nonreceptor 1 (TNK1) as a unique player in the ISG induction pathway. The immune-modulatory function of TNK1 has never been studied, and this study characterizes its significance in antiviral innate immunity. TNK1 is abundantly expressed in hepatocytes and maintains basal ISG expression. More importantly, TNK1 plays a critical role in type I IFN-mediated ISG induction. We discovered that the activated IFN receptor complex recruits TNK1 from the cytoplasm. TNK1 is then phosphorylated to enhance its kinase activity. The activated TNK1 potentiates JAK-STAT signaling through dual phosphorylation of STAT1 at tyrosine 701 and serine 727 amino acid positions. Our loss-of-function approach demonstrated that TNK1 governs a cluster of ISG expression that defines the TNK1 pathway effector genes. More importantly, TNK1 abundance is inversely correlated to viral replication efficiency and is also a determinant factor for the hepatocytic response to antiviral treatment. Taken together, our studies found a critical but unidentified integrated component of the IFN-JAK-STAT signaling cascade.


Subject(s)
Antiviral Agents/metabolism , Fetal Proteins/metabolism , Interferons/metabolism , Phosphoserine/metabolism , Protein-Tyrosine Kinases/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , Animals , Cell Line, Tumor , DNA, Complementary/genetics , Disease Susceptibility , Gene Deletion , Gene Expression Regulation , Genetic Testing , Genome, Human/genetics , Hepacivirus/physiology , Hepatitis C/enzymology , Hepatitis C/genetics , Hepatitis C/pathology , Hepatitis C/virology , Hepatocytes/enzymology , Hepatocytes/pathology , Hepatocytes/virology , Humans , Immunity, Innate/genetics , Janus Kinase 1/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation
7.
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
8.
PLoS Pathog ; 9(1): e1003118, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23300459

ABSTRACT

Although the transcription factors IRF-3 and IRF-7 are considered master regulators of type I interferon (IFN) induction and IFN stimulated gene (ISG) expression, Irf3(-/-)×Irf7(-/-) double knockout (DKO) myeloid dendritic cells (mDC) produce relatively normal levels of IFN-ß after viral infection. We generated Irf3(-/-)×Irf5(-/-)×Irf7(-/-) triple knockout (TKO) mice to test whether IRF-5 was the source of the residual induction of IFN-ß and ISGs in mDCs. In pathogenesis studies with two unrelated positive-sense RNA viruses (West Nile virus (WNV) and murine norovirus), TKO mice succumbed at rates greater than DKO mice and equal to or approaching those of mice lacking the type I IFN receptor (Ifnar(-/-)). In ex vivo studies, after WNV infection or exposure to Toll-like receptor agonists, TKO mDCs failed to produce IFN-ß or express ISGs. In contrast, this response was sustained in TKO macrophages following WNV infection. To define IRF-regulated gene signatures, we performed microarray analysis on WNV-infected mDC from wild type (WT), DKO, TKO, or Ifnar(-/-) mice, as well as from mice lacking the RIG-I like receptor adaptor protein MAVS. Whereas the gene induction pattern in DKO mDC was similar to WT cells, remarkably, almost no ISG induction was detected in TKO or Mavs(-/-) mDC. The relative equivalence of TKO and Mavs(-/-) responses suggested that MAVS dominantly regulates ISG induction in mDC. Moreover, we showed that MAVS-dependent induction of ISGs can occur through an IRF-5-dependent yet IRF-3 and IRF-7-independent pathway. Our results establish IRF-3, -5, and -7 as the key transcription factors responsible for mediating the type I IFN and ISG response in mDC during WNV infection and suggest a novel signaling link between MAVS and IRF-5.


Subject(s)
Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-7/immunology , Interferon Regulatory Factors/immunology , Interferon-beta/immunology , West Nile virus/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Receptor, Interferon alpha-beta/genetics , Signal Transduction , Toll-Like Receptors/immunology , Viral Load , West Nile Fever/genetics , West Nile Fever/immunology , West Nile Fever/virology , West Nile virus/genetics
9.
Hepatology ; 57(2): 461-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22996292

ABSTRACT

UNLABELLED: Type 1 interferon (IFN) continues to be the foundation for the current standard of care combination therapy for chronic hepatitis C virus (HCV) infection, yet the component interferon-stimulated genes (ISGs) that mediate the antiviral actions of IFN are not fully defined. Interferon-induced transmembrane protein 1 (IFITM1) is an ISG product that suppresses early stage infection by a number of viruses through an unknown mechanism of action. Moreover, the actions of IFITM1 on HCV infection are not fully elucidated. Here we identify IFITM1 as a hepatocyte tight junction protein and a potent anti-HCV effector molecule. IFITM1 expression is induced early during IFN treatment of hepatocytes and accumulates at hepatic tight junctions in HCV-infected human patient liver during IFN therapy. Additionally, we found that IFITM1 interacts with HCV coreceptors, including CD81 and occludin, to disrupt the process of viral entry. Thus, IFITM1 is an anti-HCV ISG whose actions impart control of HCV infection through interruption of viral coreceptor function. CONCLUSION: This study defines IFITM1 as an ISG effector with action against HCV entry. Design of therapy regimens to enhance IFITM1 expression should improve the virologic response among HCV patients undergoing treatment with type I IFN.


Subject(s)
Antigens, Differentiation/therapeutic use , Antiviral Agents/therapeutic use , Hepatitis C/immunology , Interferon Type I/therapeutic use , Receptors, Virus/drug effects , Tight Junction Proteins/physiology , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/metabolism , Cells, Cultured , Hepacivirus/drug effects , Hepacivirus/physiology , Humans , Tetraspanin 28/metabolism , Virus Replication/drug effects
10.
Cell Chem Biol ; 30(11): 1377-1389.e8, 2023 11 16.
Article in English | MEDLINE | ID: mdl-37586370

ABSTRACT

TruAB Discovery is an approach that integrates cellular immunology, high-throughput immunosequencing, bioinformatics, and computational biology in order to discover naturally occurring human antibodies for prophylactic or therapeutic use. We adapted our previously described pairSEQ technology to pair B cell receptor heavy and light chains of SARS-CoV-2 spike protein-binding antibodies derived from enriched antigen-specific memory B cells and bulk antibody-secreting cells. We identified approximately 60,000 productive, in-frame, paired antibody sequences, from which 2,093 antibodies were selected for functional evaluation based on abundance, isotype and patterns of somatic hypermutation. The exceptionally diverse antibodies included RBD-binders with broad neutralizing activity against SARS-CoV-2 variants, and S2-binders with broad specificity against betacoronaviruses and the ability to block membrane fusion. A subset of these RBD- and S2-binding antibodies demonstrated robust protection against challenge in hamster and mouse models. This high-throughput approach can accelerate discovery of diverse, multifunctional antibodies against any target of interest.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Mice , Humans , Antibodies, Neutralizing , Broadly Neutralizing Antibodies , Antibodies, Viral
11.
PLoS Pathog ; 6(2): e1000764, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20169186

ABSTRACT

We have used multiplexed high-throughput sequencing to characterize changes in small RNA populations that occur during viral infection in animal cells. Small RNA-based mechanisms such as RNA interference (RNAi) have been shown in plant and invertebrate systems to play a key role in host responses to viral infection. Although homologs of the key RNAi effector pathways are present in mammalian cells, and can launch an RNAi-mediated degradation of experimentally targeted mRNAs, any role for such responses in mammalian host-virus interactions remains to be characterized. Six different viruses were examined in 41 experimentally susceptible and resistant host systems. We identified virus-derived small RNAs (vsRNAs) from all six viruses, with total abundance varying from "vanishingly rare" (less than 0.1% of cellular small RNA) to highly abundant (comparable to abundant micro-RNAs "miRNAs"). In addition to the appearance of vsRNAs during infection, we saw a number of specific changes in host miRNA profiles. For several infection models investigated in more detail, the RNAi and Interferon pathways modulated the abundance of vsRNAs. We also found evidence for populations of vsRNAs that exist as duplexed siRNAs with zero to three nucleotide 3' overhangs. Using populations of cells carrying a Hepatitis C replicon, we observed strand-selective loading of siRNAs onto Argonaute complexes. These experiments define vsRNAs as one possible component of the interplay between animal viruses and their hosts.


Subject(s)
Invertebrates/virology , RNA Virus Infections/genetics , RNA, Viral/genetics , Vertebrates/genetics , Vertebrates/virology , Animals , Invertebrates/genetics , MicroRNAs , RNA Viruses , RNA, Small Interfering
12.
Nature ; 436(7053): 1044-7, 2005 Aug 18.
Article in English | MEDLINE | ID: mdl-16107852

ABSTRACT

RNA interference (RNAi) is an evolutionarily conserved sequence-specific post-transcriptional gene silencing mechanism that is well defined genetically in Caenorhabditis elegans. RNAi has been postulated to function as an adaptive antiviral immune mechanism in the worm, but there is no experimental evidence for this. Part of the limitation is that there are no known natural viral pathogens of C. elegans. Here we describe an infection model in C. elegans using the mammalian pathogen vesicular stomatitis virus (VSV) to study the role of RNAi in antiviral immunity. VSV infection is potentiated in cells derived from RNAi-defective worm mutants (rde-1; rde-4), leading to the production of infectious progeny virus, and is inhibited in mutants with an enhanced RNAi response (rrf-3; eri-1). Because the RNAi response occurs in the absence of exogenously added VSV small interfering RNAs, these results show that RNAi is activated during VSV infection and that RNAi is a genuine antiviral immune defence mechanism in the worm.


Subject(s)
Antiviral Agents , Caenorhabditis elegans/genetics , Caenorhabditis elegans/virology , RNA Interference , Animals , Caenorhabditis elegans/immunology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cells, Cultured , Disease Models, Animal , Mutation/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Substrate Specificity , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/growth & development , Vesicular stomatitis Indiana virus/immunology
13.
Disabil Rehabil Assist Technol ; 16(8): 821-830, 2021 Nov.
Article in English | MEDLINE | ID: mdl-32189537

ABSTRACT

PURPOSE: Rates of prosthetic device abandonment are dramatically high; however, the reasons behind abandonment are less understood. A scoping review was conducted to explore the current state of the literature on why individuals abandon upper limb prosthetic devices and consider how these reasons have evolved historically. MATERIALS AND METHODS: A systematic search of the literature identified 123 articles. After reviewing the articles using predetermined inclusion and exclusion criteria, nine relevant articles were included in the final review. The included articles covered passive, body-powered and myoelectric prosthetic devices. RESULTS: Across time, reasons for abandonment could be broadly categorized into comfort and function. Weight, temperature and perspiration were among the most common and persistent comfort-related reasons for abandonment. Regarding function, studies-reported abandonment was attributed to key concerns about control and sensory feedback, whereby participants may feel more functional without their device. CONCLUSIONS: In agreement with the previous literature, lack of comfort and function remain persistent reasons for upper limb prosthesis abandonment. Up-to-date research on reasons for abandonment of upper limb prosthetic devices is lacking, and recent prosthesis advancements have not been included in studies of device use, adoption and abandonment. Therefore, future work should explore reasons for abandonment in contemporary upper limb prosthetic devices. By understanding the reasons for prosthetic device abandonment, clinicians, therapists and researchers can use this information to proactively mitigate future upper limb prosthetic device abandonment. Findings from this review can be used to guide future prosthetic device development to improve these areas of concern and satisfy user needs.IMPLICATIONS FOR REHABILITATIONBy understanding the reasons for prosthetic device abandonment, clinicians, therapists and researchers can use this information to proactively mitigate future upper limb prosthetic device abandonment.The findings from this review can be used to guide future prosthetic device development to improve areas of concern and satisfy user needs.


Subject(s)
Artificial Limbs , Humans , Prosthesis Design , Upper Extremity
14.
Nat Commun ; 10(1): 3649, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31409781

ABSTRACT

RIG-I-Like Receptors (RLRs) RIG-I, MDA5, and LGP2, are vital pathogen recognition receptors in the defense against RNA viruses. West Nile Virus (WNV) infections continue to grow in the US. Here, we use a systems biology approach to define the contributions of each RLR in the innate immune response to WNV. Genome-wide RNAseq and bioinformatics analyses of macrophages from mice lacking either RLR reveal that the RLRs drive distinct immune gene activation and response polarization to mediate an M1/inflammatory signature while suppressing the M2/wound healing phenotype. While LGP2 functions to modulate inflammatory signaling, RIG-I and MDA5 together are essential for M1 macrophage polarization in vivo and the control of WNV infection through potential downstream control of ATF4 and SMAD4 to regulate target gene expression for cell polarization. These analyses reveal the RLR-driven signature of macrophage polarization, innate immune protection, and immune programming against WNV infection.


Subject(s)
DEAD Box Protein 58/immunology , Macrophages/immunology , West Nile Fever/immunology , West Nile virus/physiology , Animals , Cell Polarity , DEAD Box Protein 58/genetics , Female , Humans , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/immunology , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , West Nile Fever/genetics , West Nile Fever/physiopathology , West Nile Fever/virology
15.
Sci Transl Med ; 11(519)2019 11 20.
Article in English | MEDLINE | ID: mdl-31748228

ABSTRACT

Activated CD4 T cells are a major target of HIV infection. Results from the STEP HIV vaccine trial highlighted a potential role for total activated CD4 T cells in promoting HIV acquisition. However, the influence of vaccine insert-specific CD4 T cell responses on HIV acquisition is not known. Here, using the data obtained from four macaque studies, we show that the DNA prime/modified vaccinia Ankara boost vaccine induced interferon γ (IFNγ+) CD4 T cells [T helper 1 (TH1) cells] rapidly migrate to multiple tissues including colon, cervix, and vaginal mucosa. These mucosal TH1 cells persisted at higher frequencies and expressed higher density of CCR5, a viral coreceptor, compared to cells in blood. After intravaginal or intrarectal simian immunodeficiency virus (SIV)/simian-human immunodeficiency virus (SHIV) challenges, strong vaccine protection was evident only in animals that had lower frequencies of vaccine-specific TH1 cells but not in animals that had higher frequencies of TH1 cells, despite comparable vaccine-induced humoral and CD8 T cell immunity in both groups. An RNA transcriptome signature in blood at 7 days after priming immunization from one study was associated with induction of fewer TH1-type CD4 cells and enhanced protection. These results demonstrate that high and persisting frequencies of HIV vaccine-induced TH1-biased CD4 T cells in the intestinal and genital mucosa can mitigate beneficial effects of protective antibodies and CD8 T cells, highlighting a critical role of priming immunization and vaccine adjuvants in modulating HIV vaccine efficacy.


Subject(s)
SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antibodies, Viral/immunology , Antibody Formation/immunology , CD8-Positive T-Lymphocytes/immunology , Colon/pathology , Female , Gene Expression Profiling , Interferon-gamma/metabolism , Lymphocyte Count , Macaca mulatta , Male , Mucous Membrane/pathology , Receptors, CCR5/metabolism , Simian Acquired Immunodeficiency Syndrome/blood , Simian Acquired Immunodeficiency Syndrome/genetics , Treatment Outcome , Vaccination , Vagina/immunology , Vagina/virology
16.
mBio ; 9(2)2018 03 20.
Article in English | MEDLINE | ID: mdl-29559569

ABSTRACT

Induction of interferon beta (IFN-ß), IFN-stimulated genes (ISGs), and inflammatory responses is critical for control of viral infection. We recently identified an essential linkage of stimulation of the inflammatory cytokine interleukin-1ß (IL-1ß) and induction of ISGs that function as host restriction pathways against the emerging flavivirus West Nile virus (WNV) in vivo Here we utilized ex vivo global transcriptome analysis of primary dendritic cells, known targets of WNV replication, to define gene signatures required for this IL-1ß-driven antiviral response. Dendritic cells that were deficient in IL-1 receptor signaling showed dysregulation of cell-intrinsic defense genes and loss of viral control during WNV infection. Surprisingly, we found that in wild-type cells, IL-1ß treatment, in the absence of infection, drove the transcription of IFN-ß and ISGs at late times following treatment. Expression of these antiviral innate immune genes was dependent on the transcription factor IFN regulatory factor 3 (IRF3) and appears to reflect a general shift in IL-1ß signaling from an early inflammatory response to a late IFN-mediated response. These data demonstrate that inflammatory and antiviral signals integrate to control viral infection in myeloid cells through a process of IL-1ß-to-IRF3 signaling crosstalk. Strategies to exploit these cytokines in the activation of host defense programs should be investigated as novel therapeutic approaches against individual pathogens.IMPORTANCE West Nile virus is an emerging mosquito-borne flavivirus that can result in serious illness, neuropathology, and death in infected individuals. Currently, there are no vaccines or therapies for human use against West Nile virus. Immune control of West Nile virus infection requires inflammatory and antiviral responses, though the effect that each arm of this response has on the other is unclear. The significance of our research is in defining how virus-induced inflammatory responses regulate critical antiviral immune programs for effective control of West Nile virus infection. These data identify essential mechanisms of immune control that can inform therapeutic efforts against West Nile virus, with potential efficacy against other neuroinvasive viruses.


Subject(s)
Dendritic Cells/metabolism , Interleukin-1beta/metabolism , Animals , Cell Line , Cells, Cultured , Cricetinae , Enzyme-Linked Immunosorbent Assay , Flavivirus/pathogenicity , Immunity, Innate/physiology , Inflammasomes/metabolism , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interleukin-1beta/genetics , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Myeloid Cells/virology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , West Nile virus/pathogenicity
17.
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
18.
Methods Mol Biol ; 1488: 251-263, 2017.
Article in English | MEDLINE | ID: mdl-27933528

ABSTRACT

The Collaborative Cross (CC) is a large panel of inbred mouse strains currently being developed for multiple areas of research. Scientists are taking integrated omics-style approaches to collecting data in order to obtain a deeper understanding of the biological mechanisms underlying a number of diverse disease phenotypes. As the cost of the next generation sequencing (NGS) decreases, RNA-sequencing (RNA-seq) has become the favored approach to transcriptomic analyses versus microarrays due to increases in sensitivity and resolution. This is particularly the case with newly defined genomes, where experimental annotation has not caught up to the new microarray platforms. Traditional RNA-seq approaches are not ideal when working with results from collaborative cross studies, as the genomes across individual strains differ considerably. In this chapter we will provide an overview of how to effectively perform RNA-seq analysis from data obtained from the CC mice.


Subject(s)
Computational Biology/methods , Crosses, Genetic , High-Throughput Nucleotide Sequencing , Sequence Analysis, RNA , Software , Animals , Chromosome Mapping , Genome , Genomics/methods , Mice , Mice, Inbred Strains , Pseudogenes , Web Browser
19.
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
20.
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.

SELECTION OF CITATIONS
SEARCH DETAIL