Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
PLoS Pathog ; 18(7): e1010695, 2022 07.
Article in English | MEDLINE | ID: mdl-35788221

ABSTRACT

Chikungunya virus (CHIKV) is an emerging/re-emerging mosquito-borne pathogen responsible for explosive epidemics of febrile illness characterized by debilitating polyarthralgia and the risk of lethal infection among the most severe cases. Despite the public health risk posed by CHIKV, no vaccine is currently available. Using a site-directed hydrogen peroxide-based inactivation approach, we developed a new CHIKV vaccine, HydroVax-CHIKV. This vaccine technology was compared to other common virus inactivation approaches including ß-propiolactone (BPL), formaldehyde, heat, and ultraviolet (UV) irradiation. Heat, UV, and BPL were efficient at inactivating CHIKV-181/25 but caused substantial damage to neutralizing epitopes and failed to induce high-titer neutralizing antibodies in vaccinated mice. HydroVax-CHIKV and formaldehyde-inactivated CHIKV retained intact neutralizing epitopes similar to live virus controls but the HydroVax-CHIKV approach demonstrated a more rapid rate of virus inactivation. HydroVax-CHIKV vaccination induced high neutralizing responses to homologous and heterologous CHIKV clades as well as to other alphaviruses including Mayaro virus, O'nyong'nyong virus, and Una virus. Following heterologous infection with CHIKV-SL15649, HydroVax-CHIKV-immunized mice were protected against viremia, CHIKV-associated arthritic disease, and lethal CHIKV infection by an antibody-dependent mechanism. In contrast, animals vaccinated with Heat- or UV-inactivated virus showed no protection against viremia in addition to demonstrating significantly exacerbated CD4+ T cell-mediated footpad swelling after CHIKV infection. Together, these results demonstrate the risks associated with using suboptimal inactivation methods that fail to elicit protective neutralizing antibody responses and show that HydroVax-CHIKV represents a promising new vaccine candidate for prevention of CHIKV-associated disease.


Subject(s)
Chikungunya Fever , Chikungunya virus , Viral Vaccines , Animals , Antibodies, Neutralizing , Antibodies, Viral , Chikungunya Fever/prevention & control , Epitopes , Formaldehyde , Mice , Viremia
2.
Curr Top Microbiol Immunol ; 435: 81-106, 2022.
Article in English | MEDLINE | ID: mdl-31338593

ABSTRACT

Chikungunya is a clinically and economically important arbovirus that has spread globally in the twenty-first century. While uncommonly fatal, infection with the virus can lead to incapacitating arthralgia that can persist for months to years. The adverse impacts of viral spread are most severe in developing low- and middle-income countries in which medical infrastructure is insufficient and manual labor is an economic driver. Unfortunately, no prophylactic or therapeutic treatments are approved for human use to combat the virus. Historically, vaccination has proven to be the most efficient and successful strategy for protecting populations and eradicating infectious disease. A large and diverse range of promising vaccination approaches for use against Chikungunya has emerged in recent years and been shown to safely elicit protective immune responses in animal models and humans. Importantly, many of these are based on technologies that have been clinically approved for use against other pathogens. Furthermore, clinical trials are currently ongoing for a subset of these. The purpose of this review is to provide a description of the relevant immunobiology of Chikungunya infection, to present immune-stimulating technologies that have been successfully employed to protect against infection, and discuss priorities and challenges regarding the future development of a vaccine for clinical use.


Subject(s)
Chikungunya Fever , Chikungunya virus , Viral Vaccines , Animals , Chikungunya Fever/prevention & control , Humans , Vaccination
3.
PLoS Pathog ; 17(8): e1009380, 2021 08.
Article in English | MEDLINE | ID: mdl-34411201

ABSTRACT

Human cytomegalovirus (HCMV) microRNAs (miRNAs) significantly rewire host signaling pathways to support the viral lifecycle and regulate host cell responses. Here we show that SMAD3 expression is regulated by HCMV miR-UL22A and contributes to the IRF7-mediated induction of type I IFNs and IFN-stimulated genes (ISGs) in human fibroblasts. Addition of exogenous TGFß interferes with the replication of a miR-UL22A mutant virus in a SMAD3-dependent manner in wild type fibroblasts, but not in cells lacking IRF7, indicating that downregulation of SMAD3 expression to limit IFN induction is important for efficient lytic replication. These findings uncover a novel interplay between SMAD3 and innate immunity during HCMV infection and highlight the role of viral miRNAs in modulating these responses.


Subject(s)
Cytomegalovirus Infections/microbiology , Cytomegalovirus/physiology , Fibroblasts/microbiology , Immunity, Innate/immunology , Interferon Type I/metabolism , MicroRNAs/genetics , Transforming Growth Factor beta/metabolism , Fibroblasts/immunology , Fibroblasts/pathology , Host-Pathogen Interactions , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Interferon Type I/genetics , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/genetics , Virus Physiological Phenomena
4.
Front Immunol ; 11: 1430, 2020.
Article in English | MEDLINE | ID: mdl-32733475

ABSTRACT

The innate immune response to cytosolic DNA involves transcriptional activation of type I interferons (IFN-I) and proinflammatory cytokines. This represents the culmination of intracellular signaling pathways that are initiated by pattern recognition receptors that engage DNA and require the adaptor protein Stimulator of Interferon Genes (STING). These responses lead to the generation of cellular and tissue states that impair microbial replication and facilitate the establishment of long-lived, antigen-specific adaptive immunity. Ultimately this can lead to immune-mediated protection from infection but also to the cytotoxic T cell-mediated clearance of tumor cells. Intriguingly, pharmacologic activation of STING-dependent phenotypes is known to enhance both vaccine-associated immunogenicity and immune-based anti-tumor therapies. Unfortunately, the STING protein exists as multiple variant forms in the human population that exhibit differences in their reactivity to chemical stimuli and in the intensity of molecular signaling they induce. In light of this, STING-targeting drug discovery efforts require an accounting of protein variant-specific activity. Herein we describe a small molecule termed M04 that behaves as a novel agonist of human STING. Importantly, we find that the molecule exhibits a differential ability to activate STING based on the allelic variant examined. Furthermore, while M04 is inactive in mice, expression of human STING in mouse cells rescues reactivity to the compound. Using primary human cells in ex vivo assays we were also able to show that M04 is capable of simulating innate responses important for adaptive immune activation such as cytokine secretion, dendritic cell maturation, and T cell cross-priming. Collectively, this work demonstrates the conceivable utility of a novel agonist of human STING both as a research tool for exploring STING biology and as an immune potentiating molecule.


Subject(s)
Immunity, Innate/drug effects , Immunologic Factors/pharmacology , Membrane Proteins/agonists , Alleles , Animals , Drug Discovery , Humans , Immunity, Innate/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice
5.
Front Microbiol ; 11: 24, 2020.
Article in English | MEDLINE | ID: mdl-32117091

ABSTRACT

The dengue virus presents a serious threat to human health globally and can cause severe, even life-threatening, illness. Dengue virus (DENV) is endemic on all continents except Antarctica, and it is estimated that more than 100 million people are infected each year. Herein, we further mine the data from a previously described screen for microRNAs (miRNAs) that block flavivirus replication. We use miR-424, a member of the miR-15/16 family, as a tool to further dissect the role of host cell proteins during DENV infection. We observed that miR-424 suppresses expression of the E3 ubiquitin ligase SIAH1, which is normally induced during dengue virus 2 (DENV2) infection through activation of the unfolded protein response (UPR). Specific siRNA-mediated knockdown of SIAH1 also results in inhibition of DENV replication, demonstrating that this target is at least partly responsible for the antiviral activity of miR-424. We further show that SIAH1 binds to and ubiquitinates the innate immune adaptor protein MyD88 and that the antiviral effect of SIAH1 knockdown is reduced in cells in which MyD88 has been deleted by CRISPR/Cas9 gene editing. Additionally, MyD88-dependent signaling, triggered either by DENV2 infection or the Toll-like receptor 7 (TLR7) ligand imiquimod, is increased in cells in which SIAH1 has been knocked down by miR-424 or a SIAH1-specific siRNA. These observations suggest an additional pathway by which DENV2 harnesses aspects of the UPR to dampen the host innate immune response and promote viral replication.

6.
PLoS One ; 15(2): e0229570, 2020.
Article in English | MEDLINE | ID: mdl-32101570

ABSTRACT

The proinflammatory cytokines interleukin (IL)-1ß and IL-18 are products of activation of the inflammasome, an innate sensing system, and important in the pathogenesis of herpes simplex virus type 1 (HSV-1). The release of IL-18 and IL-1ß from monocytes/macrophages is critical for protection from HSV-1 based on animal models of encephalitis and genital infection, yet if and how HSV-1 activates inflammasomes in human macrophages is unknown. To investigate this, we utilized both primary human monocyte derived macrophages and human monocytic cell lines (THP-1 cells) with various inflammasome components knocked-out. We found that HSV-1 activates inflammasome signaling in proinflammatory primary human macrophages, but not in resting macrophages. Additionally, HSV-1 inflammasome activation in THP-1 cells is dependent on nucleotide-binding domain and leucine-rich repeat-containing receptor 3 (NLRP3), apoptosis-associated speck-like molecule containing a caspase recruitment domain (ASC), and caspase-1, but not on absent in melanoma 2 (AIM2), or gamma interferon-inducible protein 16 (IFI16). In contrast, HSV-1 activates non-canonical inflammasome signaling in proinflammatory macrophages that results in IL-1ß, but not IL-18, release that is independent of NLRP3, ASC, and caspase-1. Ultraviolet irradiation of HSV-1 enhanced inflammasome activation, demonstrating that viral replication suppresses inflammasome activation. These results confirm that HSV-1 is capable of activating the inflammasome in human macrophages through an NLRP3 dependent process and that the virus has evolved an NLRP3 specific mechanism to inhibit inflammasome activation in macrophages.


Subject(s)
Herpesvirus 1, Human/metabolism , Inflammasomes/metabolism , Macrophages/metabolism , CARD Signaling Adaptor Proteins/metabolism , Carrier Proteins/metabolism , Caspase 1/metabolism , Cytokines/metabolism , Humans , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Monocytes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Primary Cell Culture , Signal Transduction/drug effects , THP-1 Cells
7.
ACS Infect Dis ; 5(7): 1139-1149, 2019 07 12.
Article in English | MEDLINE | ID: mdl-31060350

ABSTRACT

Stimulator of interferon genes (STING) is an integral ER-membrane protein that can be activated by 2'3'-cGAMP synthesized by cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) upon binding of double-stranded DNA. It activates interferon (IFN) and inflammatory cytokine responses to defend against infection by microorganisms. Pharmacologic activation of STING has been demonstrated to induce an antiviral state and boost antitumor immunity. We previously reported a cell-based high-throughput-screening assay that allowed for identification of small-molecule cGAS-STING-pathway agonists. We report herein a compound, 6-bromo-N-(naphthalen-1-yl)benzo[d][1,3]dioxole-5-carboxamide (BNBC), that induces a proinflammatory cytokine response in a human-STING-dependent manner. Specifically, we showed that BNBC induced type I and III IFN dominant cytokine responses in primary human fibroblasts and peripheral-blood mononuclear cells (PBMCs). BNBC also induced cytokine response in PBMC-derived myeloid dendritic cells and promoted their maturation, suggesting that STING-agonist treatment could potentially regulate the activation of CD4+ and CD8+ T lymphocytes. As anticipated, treatment of primary human fibroblast cells with BNBC induced an antiviral state that inhibited the infection of several kinds of flaviviruses. Taken together, our results indicate that BNBC is a human-STING agonist that not only induces innate antiviral immunity against a broad spectrum of viruses but may also stimulate the activation of adaptive immune responses, which is important for the treatment of chronic viral infections and tumors.


Subject(s)
Antiviral Agents/chemical synthesis , Benzodioxoles/chemical synthesis , Flavivirus Infections/immunology , Interferons/metabolism , Membrane Proteins/agonists , Adaptive Immunity/drug effects , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Benzodioxoles/chemistry , Benzodioxoles/pharmacology , Cells, Cultured , Hep G2 Cells , High-Throughput Screening Assays , Humans , Immunity, Innate/drug effects , Membrane Proteins/chemistry , Molecular Structure , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
8.
Article in English | MEDLINE | ID: mdl-30917980

ABSTRACT

Alphaviruses are arthropod-transmitted RNA viruses that can cause arthralgia, myalgia, and encephalitis in humans. Since the role of cellular kinases in alphavirus replication is unknown, we profiled kinetic changes in host kinase abundance and phosphorylation following chikungunya virus (CHIKV) infection of fibroblasts. Based upon the results of this study, we treated CHIKV-infected cells with kinase inhibitors targeting the Src family kinase (SFK)-phosphatidylinositol 3-kinase (PI3K)-AKT-mTORC signaling pathways. Treatment of cells with SFK inhibitors blocked the replication of CHIKV as well as multiple other alphaviruses, including Mayaro virus, O'nyong-nyong virus, Ross River virus, and Venezuelan equine encephalitis virus. Dissecting the effect of SFK inhibition on alphavirus replication, we found that viral structural protein levels were significantly reduced, but synthesis of viral genomic and subgenomic RNAs was unaffected. By measuring the association of viral RNA with polyribosomes, we found that the SFK inhibitor dasatinib blocks alphavirus subgenomic RNA translation. Our results demonstrate a role for SFK signaling in alphavirus subgenomic RNA translation and replication. Targeting host factors involved in alphavirus replication represents an innovative, perhaps paradigm-shifting, strategy for exploring the replication of CHIKV and other alphaviruses while promoting antiviral therapeutic development.


Subject(s)
Alphavirus Infections/drug therapy , Alphavirus/drug effects , Protein Biosynthesis/drug effects , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , src-Family Kinases/genetics , Alphavirus/genetics , Alphavirus Infections/virology , Animals , Antiviral Agents/pharmacology , Cell Line , Chlorocebus aethiops , Genome, Viral/drug effects , Genome, Viral/genetics , Humans , Mice, Knockout , Phosphatidylinositol 3-Kinases/genetics , RNA, Viral/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Vero Cells , Viral Proteins/genetics , Virus Replication/drug effects , Virus Replication/genetics
9.
mBio ; 10(1)2019 02 12.
Article in English | MEDLINE | ID: mdl-30755509

ABSTRACT

Secretion of interleukin-1ß (IL-1ß) represents a fundamental innate immune response to microbial infection that, at the molecular level, occurs following activation of proteolytic caspases that cleave the immature protein into a secretable form. Human cytomegalovirus (HCMV) is the archetypal betaherpesvirus that is invariably capable of lifelong infection through the activity of numerous virally encoded immune evasion phenotypes. Innate immune pathways responsive to cytoplasmic double-stranded DNA (dsDNA) are known to be activated in response to contact between HCMV and host cells. Here, we used clustered regularly interspaced short palindromic repeat (CRISPR)-CRISPR-associated protein 9 (Cas9) genome editing to demonstrate that the dsDNA receptor absent in melanoma 2 (AIM2) is required for secretion of IL-1ß following HCMV infection. Furthermore, dsDNA-responsive innate signaling induced by HCMV infection that leads to activation of the type I interferon response is also shown, unexpectedly, to play a contributory role in IL-1ß secretion. Importantly, we also show that rendering virus particles inactive by UV exposure leads to substantially increased IL-1ß processing and secretion and that live HCMV can inhibit this, suggesting the virus encodes factors that confer an inhibitory effect on this response. Further examination revealed that ectopic expression of the immediate early (IE) 86-kDa protein (IE86) is actually associated with a block in transcription of the pro-IL-1ß gene and, independently, diminishment of the immature protein. Overall, these results reveal two new and distinct phenotypes conferred by the HCMV IE86 protein, as well as an unusual circumstance in which a single herpesviral protein exhibits inhibitory effects on multiple molecular processes within the same innate immune response.IMPORTANCE Persistent infection with HCMV is associated with the operation of diverse evasion phenotypes directed at antiviral immunity. Obstruction of intrinsic and innate immune responses is typically conferred by viral proteins either associated with the viral particle or expressed immediately after entry. In line with this, numerous phenotypes are attributed to the HCMV IE86 protein that involve interference with innate immune processes via transcriptional and protein-directed mechanisms. We describe novel IE86-mediated phenotypes aimed at virus-induced secretion of IL-1ß. Intriguingly, while many viruses target the function of the molecular scaffold required for IL-1ß maturation to prevent this response, we find that HCMV and IE86 target the IL-1ß protein specifically. Moreover, we show that IE86 impairs both the synthesis of the IL-1ß transcript and the stability of the immature protein. This indicates an unusual phenomenon in which a single viral protein exhibits two molecularly separate evasion phenotypes directed at a single innate cytokine.


Subject(s)
Cytomegalovirus/physiology , DNA-Binding Proteins/metabolism , Immediate-Early Proteins/metabolism , Immune Evasion , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Humans , Proteolysis , THP-1 Cells
10.
Methods Mol Biol ; 1656: 183-193, 2017.
Article in English | MEDLINE | ID: mdl-28808971

ABSTRACT

Modern drug discovery has embraced in vitro platforms that enable investigation of large numbers of compounds within tractable timeframes and for feasible costs. These endeavors have been greatly aided in recent years by advances in molecular and cell-based methods such as gene delivery and editing technology, advanced imaging, robotics, and quantitative analysis. As such, the examination of phenotypic impacts of novel molecules may only be limited by the size of the compound collection. Innate immune signaling processes in mammalian cells are especially amenable to high-throughput screening platforms since the cellular responses elicited by their activation often result in high level transcription that can be harnessed in the form of bioluminescent or fluorescent signal. In addition, targeted activation of innate immune pathways represents a valuable therapeutic strategy applicable to multiple chronic and acute human diseases. Herein, we describe the optimization and utilization of a high-throughput screening method using human reporter cells reactive to stimulation of the type I interferon response. Importantly, the principles and methods described can be applied to adherent reporter cells of diverse derivation and innate signaling pathway readouts.


Subject(s)
Cell Culture Techniques/methods , Drug Discovery/methods , Immunity, Innate/drug effects , Immunologic Factors , Animals , Humans , Immunologic Factors/chemistry , Immunologic Factors/pharmacokinetics , Immunologic Factors/pharmacology
11.
mBio ; 8(3)2017 05 02.
Article in English | MEDLINE | ID: mdl-28465426

ABSTRACT

The ongoing concurrent outbreaks of Zika, Chikungunya, and dengue viruses in Latin America and the Caribbean highlight the need for development of broad-spectrum antiviral treatments. The type I interferon (IFN) system has evolved in vertebrates to generate tissue responses that actively block replication of multiple known and potentially zoonotic viruses. As such, its control and activation through pharmacological agents may represent a novel therapeutic strategy for simultaneously impairing growth of multiple virus types and rendering host populations resistant to virus spread. In light of this strategy's potential, we undertook a screen to identify novel interferon-activating small molecules. Here, we describe 1-(2-fluorophenyl)-2-(5-isopropyl-1,3,4-thiadiazol-2-yl)-1,2-dihydrochromeno[2,3-c]pyrrole-3,9-dione, which we termed AV-C. Treatment of human cells with AV-C activates innate and interferon-associated responses that strongly inhibit replication of Zika, Chikungunya, and dengue viruses. By utilizing genome editing, we investigated the host proteins essential to AV-C-induced cellular states. This showed that the compound requires a TRIF-dependent signaling cascade that culminates in IFN regulatory factor 3 (IRF3)-dependent expression and secretion of type I interferon to elicit antiviral responses. The other canonical IRF3-terminal adaptor proteins STING and IPS-1/MAVS were dispensable for AV-C-induced phenotypes. However, our work revealed an important inhibitory role for IPS-1/MAVS, but not TRIF, in flavivirus replication, implying that TRIF-directed viral evasion may not occur. Additionally, we show that in response to AV-C, primary human peripheral blood mononuclear cells secrete proinflammatory cytokines that are linked with establishment of adaptive immunity to viral pathogens. Ultimately, synthetic innate immune activators such as AV-C may serve multiple therapeutic purposes, including direct antimicrobial responses and facilitation of pathogen-directed adaptive immunity.IMPORTANCE The type I interferon system is part of the innate immune response that has evolved in vertebrates as a first line of broad-spectrum immunological defense against an unknowable diversity of microbial, especially viral, pathogens. Here, we characterize a novel small molecule that artificially activates this response and in so doing generates a cellular state antagonistic to growth of currently emerging viruses: Zika virus, Chikungunya virus, and dengue virus. We also show that this molecule is capable of eliciting cellular responses that are predictive of establishment of adaptive immunity. As such, this agent may represent a powerful and multipronged therapeutic tool to combat emerging and other viral diseases.


Subject(s)
Adaptor Proteins, Vesicular Transport/agonists , Antiviral Agents/pharmacology , Benzopyrans/pharmacology , Chikungunya virus/physiology , Dengue Virus/physiology , Thiadiazoles/pharmacology , Virus Replication , Zika Virus/physiology , Adaptor Proteins, Vesicular Transport/metabolism , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Benzopyrans/chemistry , Benzopyrans/isolation & purification , Cell Line , Chikungunya Fever/drug therapy , Chikungunya virus/drug effects , Cytokines/biosynthesis , DNA Replication/drug effects , Dengue/drug therapy , Dengue Virus/drug effects , Dengue Virus/metabolism , Drug Discovery , Gene Editing , Host-Pathogen Interactions , Humans , Immune Evasion , Immunity, Innate/drug effects , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Type I/drug effects , Interferon Type I/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Thiadiazoles/chemistry , Thiadiazoles/isolation & purification , Zika Virus/drug effects
13.
PLoS Pathog ; 13(3): e1006219, 2017 03.
Article in English | MEDLINE | ID: mdl-28278237

ABSTRACT

Zika virus (ZIKV), an emerging flavivirus, has recently spread explosively through the Western hemisphere. In addition to symptoms including fever, rash, arthralgia, and conjunctivitis, ZIKV infection of pregnant women can cause microcephaly and other developmental abnormalities in the fetus. We report herein the results of ZIKV infection of adult rhesus macaques. Following subcutaneous infection, animals developed transient plasma viremia and viruria from 1-7 days post infection (dpi) that was accompanied by the development of a rash, fever and conjunctivitis. Animals produced a robust adaptive immune response to ZIKV, although systemic cytokine response was minimal. At 7 dpi, virus was detected in peripheral nervous tissue, multiple lymphoid tissues, joints, and the uterus of the necropsied animals. Notably, viral RNA persisted in neuronal, lymphoid and joint/muscle tissues and the male and female reproductive tissues through 28 to 35 dpi. The tropism and persistence of ZIKV in the peripheral nerves and reproductive tract may provide a mechanism of subsequent neuropathogenesis and sexual transmission.


Subject(s)
Zika Virus Infection/pathology , Zika Virus Infection/virology , Animals , Cell Separation , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , In Situ Hybridization , Macaca mulatta , Male , Neutralization Tests , Polymerase Chain Reaction , Viremia/virology , Zika Virus
14.
Elife ; 52016 11 22.
Article in English | MEDLINE | ID: mdl-27874833

ABSTRACT

Natural Killer (NK) cells are essential for control of viral infection and cancer. NK cells express NKG2D, an activating receptor that directly recognizes NKG2D ligands. These are expressed at low level on healthy cells, but are induced by stresses like infection and transformation. The physiological events that drive NKG2D ligand expression during infection are still poorly understood. We observed that the mouse cytomegalovirus encoded protein m18 is necessary and sufficient to drive expression of the RAE-1 family of NKG2D ligands. We demonstrate that RAE-1 is transcriptionally repressed by histone deacetylase inhibitor 3 (HDAC3) in healthy cells, and m18 relieves this repression by directly interacting with Casein Kinase II and preventing it from activating HDAC3. Accordingly, we found that HDAC inhibiting proteins from human herpesviruses induce human NKG2D ligand ULBP-1. Thus our findings indicate that virally mediated HDAC inhibition can act as a signal for the host to activate NK-cell recognition.


Subject(s)
Gene Expression Regulation , Histone Deacetylases/metabolism , Host-Pathogen Interactions , Muromegalovirus/physiology , Animals , Humans , Mice , NK Cell Lectin-Like Receptor Subfamily K , Nuclear Matrix-Associated Proteins , Nucleocytoplasmic Transport Proteins
15.
PLoS Pathog ; 12(10): e1005891, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27736984

ABSTRACT

Chikungunya virus (CHIKV) is a re-emerging global pathogen with pandemic potential, which causes fever, rash and debilitating arthralgia. Older adults over 65 years are particularly susceptible to severe and chronic CHIKV disease (CHIKVD), accounting for >90% of all CHIKV-related deaths. There are currently no approved vaccines or antiviral treatments available to limit chronic CHIKVD. Here we show that in old mice excessive, dysregulated TGFß production during acute infection leads to a reduced immune response and subsequent chronic disease. Humans suffering from CHIKV infection also exhibited high TGFß levels and a pronounced age-related defect in neutralizing anti-CHIKV antibody production. In vivo reduction of TGFß levels minimized acute joint swelling, restored neutralizing antibody production and diminished chronic joint pathology in old mice. This study identifies increased and dysregulated TGFß secretion as one key mechanism contributing to the age-related loss of protective anti-CHIKV-immunity leading to chronic CHIKVD.


Subject(s)
Aging/immunology , Chikungunya Fever/immunology , Transforming Growth Factor beta/immunology , Adult , Aged , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Chikungunya virus , Disease Models, Animal , Disease Susceptibility , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Male , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Transforming Growth Factor beta/biosynthesis
16.
PLoS Pathog ; 11(12): e1005324, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26646986

ABSTRACT

Pharmacologic stimulation of innate immune processes represents an attractive strategy to achieve multiple therapeutic outcomes including inhibition of virus replication, boosting antitumor immunity, and enhancing vaccine immunogenicity. In light of this we sought to identify small molecules capable of activating the type I interferon (IFN) response by way of the transcription factor IFN regulatory factor 3 (IRF3). A high throughput in vitro screen yielded 4-(2-chloro-6-fluorobenzyl)-N-(furan-2-ylmethyl)-3-oxo-3,4-dihydro-2H-benzo[b][1,4]thiazine-6-carboxamide (referred to herein as G10), which was found to trigger IRF3/IFN-associated transcription in human fibroblasts. Further examination of the cellular response to this molecule revealed expression of multiple IRF3-dependent antiviral effector genes as well as type I and III IFN subtypes. This led to the establishment of a cellular state that prevented replication of emerging Alphavirus species including Chikungunya virus, Venezuelan Equine Encephalitis virus, and Sindbis virus. To define cellular proteins essential to elicitation of the antiviral activity by the compound we employed a reverse genetics approach that utilized genome editing via CRISPR/Cas9 technology. This allowed the identification of IRF3, the IRF3-activating adaptor molecule STING, and the IFN-associated transcription factor STAT1 as required for observed gene induction and antiviral effects. Biochemical analysis indicates that G10 does not bind to STING directly, however. Thus the compound may represent the first synthetic small molecule characterized as an indirect activator of human STING-dependent phenotypes. In vivo stimulation of STING-dependent activity by an unrelated small molecule in a mouse model of Chikungunya virus infection blocked viremia demonstrating that pharmacologic activation of this signaling pathway may represent a feasible strategy for combating emerging Alphaviruses.


Subject(s)
Antiviral Agents/pharmacology , Chikungunya Fever/immunology , Membrane Proteins/agonists , Signal Transduction/immunology , Thiazines/pharmacology , Alphavirus/immunology , Alphavirus Infections/immunology , Animals , Cells, Cultured , Chikungunya virus/immunology , High-Throughput Screening Assays , Humans , Immunoblotting , Interferon Regulatory Factor-3/immunology , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
17.
J Virol ; 89(17): 8687-700, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26085158

ABSTRACT

UNLABELLED: Varicella-zoster virus (VZV) causes chickenpox upon primary infection and establishes latency in ganglia. Reactivation from latency causes herpes zoster, which may be complicated by postherpetic neuralgia. Innate immunity mediated by interferon and proinflammatory cytokines represents the first line of immune defense upon infection and reactivation. VZV is known to interfere with multiple innate immune signaling pathways, including the central transcription factor NF-κB. However, the role of these inhibitory mechanisms in vivo is unknown. Simian varicella virus (SVV) infection of rhesus macaques recapitulates key aspects of VZV pathogenesis, and this model thus permits examination of the role of immune evasion mechanisms in vivo. Here, we compare SVV and VZV with respect to interference with NF-κB activation. We demonstrate that both viruses prevent ubiquitination of the NF-κB inhibitor IκBα, whereas SVV additionally prevents IκBα phosphorylation. We show that the ORF61 proteins of VZV and SVV are sufficient to prevent IκBα ubiquitination upon ectopic expression. We further demonstrate that SVV ORF61 interacts with ß-TrCP, a subunit of the SCF ubiquitin ligase complex that mediates the degradation of IκBα. This interaction seems to inactivate SCF-mediated protein degradation in general, since the unrelated ß-TrCP target Snail is also stabilized by ORF61. In addition to ORF61, SVV seems to encode additional inhibitors of the NF-κB pathway, since SVV with ORF61 deleted still prevented IκBα phosphorylation and degradation. Taken together, our data demonstrate that SVV interferes with tumor necrosis factor alpha (TNF-α)-induced NF-κB activation at multiple levels, which is consistent with the importance of these countermechanisms for varicella virus infection. IMPORTANCE: The role of innate immunity during the establishment of primary infection, latency, and reactivation by varicella-zoster virus (VZV) is incompletely understood. Since infection of rhesus macaques by simian varicella virus (SVV) is used as an animal model of VZV infection, we characterized the molecular mechanism by which SVV interferes with innate immune activation. Specifically, we studied how SVV prevents activation of the transcription factor NF-κB, a central factor in eliciting proinflammatory responses. The identification of molecular mechanisms that counteract innate immunity might ultimately lead to better vaccines and treatments for VZV, since overcoming these mechanisms, either by small-molecule inhibition or by genetic modification of vaccine strains, is expected to reduce the pathogenic potential of VZV. Moreover, using SVV infection of rhesus macaques, it will be possible to study how increasing the vulnerability of varicella viruses to innate immunity will impact viral pathogenesis.


Subject(s)
Herpesvirus 3, Human/genetics , I-kappa B Proteins/metabolism , NF-kappa B/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Viral Proteins/metabolism , Animals , Cell Line , Chickenpox/immunology , Chickenpox/virology , Disease Models, Animal , Enzyme Activation , Gene Deletion , HEK293 Cells , Herpesvirus 3, Human/immunology , Humans , Immune Evasion/immunology , Immunity, Innate/immunology , Macaca mulatta , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Phosphorylation , SKP Cullin F-Box Protein Ligases/metabolism , Snail Family Transcription Factors , Transcription Factors/metabolism , Ubiquitination , Viral Load , Viral Proteins/genetics , Virus Activation/immunology , beta-Transducin Repeat-Containing Proteins/metabolism
18.
J Virol ; 89(15): 8011-25, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26018150

ABSTRACT

UNLABELLED: The cytosolic RIG-I (retinoic acid-inducible gene I) receptor plays a pivotal role in the initiation of the immune response against RNA virus infection by recognizing short 5'-triphosphate (5'ppp)-containing viral RNA and activating the host antiviral innate response. In the present study, we generated novel 5'ppp RIG-I agonists of varieous lengths, structures, and sequences and evaluated the generation of the antiviral and inflammatory responses in human epithelial A549 cells, human innate immune primary cells, and murine models of influenza and chikungunya viral pathogenesis. A 99-nucleotide, uridine-rich hairpin 5'pppRNA termed M8 stimulated an extensive and robust interferon response compared to other modified 5'pppRNA structures, RIG-I aptamers, or poly(I·C). Interestingly, manipulation of the primary RNA sequence alone was sufficient to modulate antiviral activity and inflammatory response, in a manner dependent exclusively on RIG-I and independent of MDA5 and TLR3. Both prophylactic and therapeutic administration of M8 effectively inhibited influenza virus and dengue virus replication in vitro. Furthermore, multiple strains of influenza virus that were resistant to oseltamivir, an FDA-approved therapeutic treatment for influenza, were highly sensitive to inhibition by M8. Finally, prophylactic M8 treatment in vivo prolonged survival and reduced lung viral titers of mice challenged with influenza virus, as well as reducing chikungunya virus-associated foot swelling and viral load. Altogether, these results demonstrate that 5'pppRNA can be rationally designed to achieve a maximal RIG-I-mediated protective antiviral response against human-pathogenic RNA viruses. IMPORTANCE: The development of novel therapeutics to treat human-pathogenic RNA viral infections is an important goal to reduce spread of infection and to improve human health and safety. This study investigated the design of an RNA agonist with enhanced antiviral and inflammatory properties against influenza, dengue, and chikungunya viruses. A novel, sequence-dependent, uridine-rich RIG-I agonist generated a protective antiviral response in vitro and in vivo and was effective at concentrations 100-fold lower than prototype sequences or other RNA agonists, highlighting the robust activity and potential clinical use of the 5'pppRNA against RNA virus infection. Altogether, the results identify a novel, sequence-specific RIG-I agonist as an attractive therapeutic candidate for the treatment of a broad range of RNA viruses, a pressing issue in which a need for new and more effective options persists.


Subject(s)
Chikungunya virus/immunology , DEAD-box RNA Helicases/immunology , Dengue Virus/immunology , Influenza A Virus, H1N1 Subtype/immunology , RNA, Viral/agonists , RNA, Viral/immunology , Virus Diseases/immunology , Animals , Cell Line , Chikungunya virus/chemistry , Chikungunya virus/genetics , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Dengue Virus/chemistry , Dengue Virus/genetics , Humans , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H1N1 Subtype/genetics , Mice , Mice, Inbred BALB C , Nucleic Acid Conformation , RNA, Viral/genetics , Receptors, Immunologic , Virus Diseases/genetics , Virus Diseases/virology
19.
PLoS Pathog ; 11(5): e1004901, 2015 May.
Article in English | MEDLINE | ID: mdl-25973608

ABSTRACT

Varicella zoster virus (VZV) causes chickenpox in humans and, subsequently, establishes latency in the sensory ganglia from where it reactivates to cause herpes zoster. Infection of rhesus macaques with simian varicella virus (SVV) recapitulates VZV pathogenesis in humans thus representing a suitable animal model for VZV infection. While the type I interferon (IFN) response has been shown to affect VZV replication, the virus employs counter mechanisms to prevent the induction of anti-viral IFN stimulated genes (ISG). Here, we demonstrate that SVV inhibits type I IFN-activated signal transduction via the JAK-STAT pathway. SVV-infected rhesus fibroblasts were refractory to IFN stimulation displaying reduced protein levels of IRF9 and lacking STAT2 phosphorylation. Since previous work implicated involvement of the VZV immediate early gene product ORF63 in preventing ISG-induction we studied the role of SVV ORF63 in generating resistance to IFN treatment. Interestingly, SVV ORF63 did not affect STAT2 phosphorylation but caused IRF9 degradation in a proteasome-dependent manner, suggesting that SVV employs multiple mechanisms to counteract the effect of IFN. Control of SVV ORF63 protein levels via fusion to a dihydrofolate reductase (DHFR)-degradation domain additionally confirmed its requirement for viral replication. Our results also show a prominent reduction of IRF9 and inhibition of STAT2 phosphorylation in VZV-infected cells. In addition, cells expressing VZV ORF63 blocked IFN-stimulation and displayed reduced levels of the IRF9 protein. Taken together, our data suggest that varicella ORF63 prevents ISG-induction both directly via IRF9 degradation and indirectly via transcriptional control of viral proteins that interfere with STAT2 phosphorylation. SVV and VZV thus encode multiple viral gene products that tightly control IFN-induced anti-viral responses.


Subject(s)
Herpesviridae Infections/metabolism , Host-Pathogen Interactions , Interferon Type I/metabolism , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Varicellovirus/physiology , Animals , Cell Line , Cercopithecinae , Chickenpox/immunology , Chickenpox/metabolism , Chickenpox/pathology , Chickenpox/virology , DNA, Recombinant/metabolism , Gene Expression Regulation, Viral , Herpesviridae Infections/immunology , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Herpesvirus 3, Human/immunology , Herpesvirus 3, Human/physiology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Immunity, Innate , Interferon Type I/antagonists & inhibitors , Interferon-Stimulated Gene Factor 3, gamma Subunit/antagonists & inhibitors , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Phosphorylation , Proteasome Endopeptidase Complex , Protein Processing, Post-Translational , Proteolysis , Recombinant Proteins/metabolism , STAT Transcription Factors/genetics , Varicellovirus/immunology
20.
J Clin Invest ; 124(5): 1928-44, 2014 May.
Article in English | MEDLINE | ID: mdl-24691437

ABSTRACT

The most abundantly produced virion protein in human cytomegalovirus (HCMV) is the immunodominant phosphoprotein 65 (pp65), which is frequently included in CMV vaccines. Although it is nonessential for in vitro CMV growth, pp65 displays immunomodulatory functions that support a potential role in primary and/or persistent infection. To determine the contribution of pp65 to CMV infection and immunity, we generated a rhesus CMV lacking both pp65 orthologs (RhCMVΔpp65ab). While deletion of pp65ab slightly reduced growth in vitro and increased defective particle formation, the protein composition of secreted virions was largely unchanged. Interestingly, pp65 was not required for primary and persistent infection in animals. Immune responses induced by RhCMVΔpp65ab did not prevent reinfection with rhesus CMV; however, reinfection with RhCMVΔUS2-11, which lacks viral-encoded MHC-I antigen presentation inhibitors, was prevented. Unexpectedly, induction of pp65b-specific T cells alone did not protect against RhCMVΔUS2-11 challenge, suggesting that T cells targeting multiple CMV antigens are required for protection. However, pp65-specific immunity was crucial for controlling viral dissemination during primary infection, as indicated by the marked increase of RhCMVΔpp65ab genome copies in CMV-naive, but not CMV-immune, animals. Our data provide rationale for inclusion of pp65 into CMV vaccines but also demonstrate that pp65-induced T cell responses alone do not recapitulate the protective effect of natural infection.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Phosphoproteins/immunology , Viral Matrix Proteins/immunology , Animals , Antigen Presentation/immunology , Cell Line , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/pathology , Cytomegalovirus Vaccines/genetics , Cytomegalovirus Vaccines/immunology , Gene Deletion , Humans , Macaca mulatta , Mice , Phosphoproteins/genetics , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Viral Matrix Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...