Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 325
1.
Nucleic Acids Res ; 51(13): 6819-6840, 2023 07 21.
Article En | MEDLINE | ID: mdl-37283074

The interferon inducible protein 16 (IFI16) is a prominent sensor of nuclear pathogenic DNA, initiating innate immune signaling and suppressing viral transcription. However, little is known about mechanisms that initiate IFI16 antiviral functions or its regulation within the host DNA-filled nucleus. Here, we provide in vitro and in vivo evidence to establish that IFI16 undergoes liquid-liquid phase separation (LLPS) nucleated by DNA. IFI16 binding to viral DNA initiates LLPS and induction of cytokines during herpes simplex virus type 1 (HSV-1) infection. Multiple phosphorylation sites within an intrinsically disordered region (IDR) function combinatorially to activate IFI16 LLPS, facilitating filamentation. Regulated by CDK2 and GSK3ß, IDR phosphorylation provides a toggle between active and inactive IFI16 and the decoupling of IFI16-mediated cytokine expression from repression of viral transcription. These findings show how IFI16 switch-like phase transitions are achieved with temporal resolution for immune signaling and, more broadly, the multi-layered regulation of nuclear DNA sensors.


Herpes Simplex , Immunity, Innate , Interferons , Cytokines/genetics , Cytokines/metabolism , Herpesvirus 1, Human/genetics , Immunity, Innate/immunology , Interferons/genetics , Interferons/immunology , Phosphorylation , Herpes Simplex/immunology , Herpes Simplex/virology , Embryo, Mammalian , Urochordata/genetics , Urochordata/immunology , Gene Expression Regulation, Viral/immunology , Cyclin-Dependent Kinase 2/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Animals
2.
Front Immunol ; 12: 700184, 2021.
Article En | MEDLINE | ID: mdl-34408749

Coronavirus disease 2019 (COVID-19), which has high incidence rates with rapid rate of transmission, is a pandemic that spread across the world, resulting in more than 3,000,000 deaths globally. Currently, several drugs have been used for the clinical treatment of COVID-19, such as antivirals (radecivir, baritinib), monoclonal antibodies (tocilizumab), and glucocorticoids (dexamethasone). Accumulating evidence indicates that long noncoding RNAs (lncRNAs) are essential regulators of virus infections and antiviral immune responses including biological processes that are involved in the regulation of COVID-19 and subsequent disease states. Upon viral infections, cellular lncRNAs directly regulate viral genes and influence viral replication and pathology through virus-mediated changes in the host transcriptome. Additionally, several host lncRNAs could help the occurrence of viral immune escape by inhibiting type I interferons (IFN-1), while others could up-regulate IFN-1 production to play an antiviral role. Consequently, understanding the expression and function of lncRNAs during severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection will provide insights into the development of lncRNA-based methods. In this review, we summarized the current findings of lncRNAs in the regulation of the strong inflammatory response, immune dysfunction and thrombosis induced by SARS-CoV-2 infection, discussed the underlying mechanisms, and highlighted the therapeutic challenges of COVID-19 treatment and its future research directions.


COVID-19/immunology , Host Microbial Interactions/genetics , Immunity, Innate/genetics , RNA, Long Noncoding/metabolism , Thrombosis/immunology , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Biomarkers/analysis , COVID-19/complications , COVID-19/genetics , COVID-19 Testing/methods , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation, Viral/drug effects , Gene Expression Regulation, Viral/immunology , Host Microbial Interactions/drug effects , Host Microbial Interactions/immunology , Humans , Immune Evasion/genetics , Pandemics/prevention & control , RNA, Long Noncoding/analysis , RNA, Long Noncoding/antagonists & inhibitors , SARS-CoV-2/drug effects , SARS-CoV-2/genetics , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Signal Transduction/genetics , Signal Transduction/immunology , Thrombosis/genetics , Thrombosis/virology , Virus Replication/drug effects , Virus Replication/genetics , Virus Replication/immunology , COVID-19 Drug Treatment
3.
Nat Commun ; 12(1): 4193, 2021 07 07.
Article En | MEDLINE | ID: mdl-34234122

Interplay between EBV infection and acquired genetic alterations during nasopharyngeal carcinoma (NPC) development remains vague. Here we report a comprehensive genomic analysis of 70 NPCs, combining whole-genome sequencing (WGS) of microdissected tumor cells with EBV oncogene expression to reveal multiple aspects of cellular-viral co-operation in tumorigenesis. Genomic aberrations along with EBV-encoded LMP1 expression underpin constitutive NF-κB activation in 90% of NPCs. A similar spectrum of somatic aberrations and viral gene expression undermine innate immunity in 79% of cases and adaptive immunity in 47% of cases; mechanisms by which NPC may evade immune surveillance despite its pro-inflammatory phenotype. Additionally, genomic changes impairing TGFBR2 promote oncogenesis and stabilize EBV infection in tumor cells. Fine-mapping of CDKN2A/CDKN2B deletion breakpoints reveals homozygous MTAP deletions in 32-34% of NPCs that confer marked sensitivity to MAT2A inhibition. Our work concludes that NPC is a homogeneously NF-κB-driven and immune-protected, yet potentially druggable, cancer.


Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/genetics , Nasopharyngeal Carcinoma/immunology , Nasopharyngeal Neoplasms/immunology , Tumor Escape/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/immunology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/therapy , Epstein-Barr Virus Infections/virology , Female , Gene Expression Regulation, Viral/immunology , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/pathogenicity , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Methionine Adenosyltransferase/antagonists & inhibitors , Methionine Adenosyltransferase/metabolism , Mice , NF-kappa B/metabolism , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/therapy , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/therapy , Nasopharyngeal Neoplasms/virology , Nasopharynx/immunology , Nasopharynx/pathology , Nasopharynx/surgery , Nasopharynx/virology , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Sequence Deletion , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Escape/drug effects , Whole Genome Sequencing , Xenograft Model Antitumor Assays
4.
Front Immunol ; 12: 644153, 2021.
Article En | MEDLINE | ID: mdl-33968035

During recent Zika epidemics, adults infected with Zika virus (ZIKV) have developed organ-specific inflammatory complications. The most serious Zika-associated inflammatory eye disease is uveitis, which is commonly anterior in type, affecting both eyes and responding to corticosteroid eye drops. Mechanisms of Zika-associated anterior uveitis are unknown, but ZIKV has been identified in the aqueous humor of affected individuals. The iris pigment epithelium is a target cell population in viral anterior uveitis, and it acts to maintain immune privilege within the anterior eye. Interactions between ZIKV and human iris pigment epithelial cells were investigated with infectivity assays and RNA-sequencing. Primary cell isolates were prepared from eyes of 20 cadaveric donors, and infected for 24 hours with PRVABC59 strain ZIKV or incubated uninfected as control. Cytoimmunofluorescence, RT-qPCR on total cellular RNA, and focus-forming assays of culture supernatant showed cell isolates were permissive to infection, and supported replication and release of infectious ZIKV. To explore molecular responses of cell isolates to ZIKV infection at the whole transcriptome level, RNA was sequenced on the Illumina NextSeq 500 platform, and results were aligned to the human GRCh38 genome. Multidimensional scaling showed clear separation between transcriptomes of infected and uninfected cell isolates. Differential expression analysis indicated a vigorous molecular response of the cell to ZIKV: 7,935 genes were differentially expressed between ZIKV-infected and uninfected cells (FDR < 0.05), and 99% of 613 genes that changed at least two-fold were up-regulated. Reactome and KEGG pathway and Gene Ontology enrichment analyses indicated strong activation of viral recognition and defense, in addition to biosynthesis processes. A CHAT network included 6275 molecular nodes and 24 contextual hubs in the cell response to ZIKV infection. Receptor-interacting serine/threonine kinase 1 (RIPK1) was the most significantly connected contextual hub. Correlation of gene expression with read counts assigned to the ZIKV genome identified a negative correlation between interferon signaling and viral load across isolates. This work represents the first investigation of mechanisms of Zika-associated anterior uveitis using an in vitro human cell model. The results suggest the iris pigment epithelium mounts a molecular response that limits intraocular pathology in most individuals.


Epithelial Cells , Gene Expression Regulation, Viral/immunology , Pigment Epithelium of Eye , RNA, Viral/immunology , Zika Virus Infection , Zika Virus/immunology , Cells, Cultured , Epithelial Cells/immunology , Epithelial Cells/pathology , Epithelial Cells/virology , Genome, Viral/immunology , Humans , Iris/immunology , Iris/pathology , Iris/virology , Pigment Epithelium of Eye/immunology , Pigment Epithelium of Eye/pathology , Pigment Epithelium of Eye/virology , Zika Virus Infection/immunology , Zika Virus Infection/pathology
5.
Front Immunol ; 12: 804387, 2021.
Article En | MEDLINE | ID: mdl-35082790

To explore the potential mechanism of cancer patients appearing more vulnerable to SARS-CoV-2 infection and poor COVID-19 outcomes, we conducted an integrative bioinformatics analysis for SARS-CoV-2-required genes and host genes and variants related to SARS-CoV-2 susceptibility and COVID-19 severity. BLCA, HNSC, KIRC, KIRP, LGG, PCPG, PRAD, TGCT, and THCA patients carrying rs10774671-A (OAS1) genotype may be more likely to have poor COVID-19 outcomes relative to those who carry rs10774671-G, because individuals carrying rs10774671-A will have lower expression of OAS1, which serves as a protective factor against SARS-CoV-2 processes and poor COVID-19 outcomes. SARS-CoV-2-required genes were correlated with TME, immune infiltration, overall survival, and anti-cancer drug sensitivity. CHOL patients may have a higher risk of SARS-CoV-2 infection than healthy subjects. SARS-CoV-2-induced ACE2 and NPC1 elevation may have a negative influence on the immune responses of LUSC and CD8+T infiltration of LUAD, and negatively affect the sensitivity of anti-lung cancer drugs. LUSC and LUAD patients may have a varying degree of adverse outcomes if they are infected with SARS-CoV-2. miR-760 may target and inhibit ACE2 expression. Cancer patients appearing vulnerable to SARS-CoV-2 infection and having poor COVID-19 outcomes may be partly due to host genetic factors and dysregulation of SARS-CoV-2-required genes. OAS1, ACE2, and miR-760 could serve as the treatment and intervention targets for SARS-CoV-2.


COVID-19/immunology , Computational Biology , Gene Expression Regulation, Neoplastic/immunology , Gene Expression Regulation, Viral/immunology , Neoplasms/immunology , SARS-CoV-2/immunology , COVID-19/mortality , COVID-19/pathology , Female , Humans , Male , Neoplasms/mortality , Neoplasms/pathology
6.
Biol Pharm Bull ; 43(11): 1715-1728, 2020.
Article En | MEDLINE | ID: mdl-33132317

SV40-encoded microRNA (miRNA), miR-S1, downregulates the large and small T antigens (LTag and STag), which promote viral replication and cellular transformation, thereby presumably impairing LTag and STag functions essential for the viral life cycle. To explore the functional significance of miR-S1-mediated downregulation of LTag and STag as well as the functional roles of miR-S1, we evaluated viral DNA replication and proinflammatory cytokine induction in cells transfected with simian virus 40 (SV40) genome plasmid and its mutated form lacking miR-S1 expression. The SV40 genome encodes two mature miR-S1s, miR-S1-3p and miR-S1-5p, of which miR-S1-3p is the predominantly expressed form. MiR-S1-3p exerted strong repressive effects on a reporter containing full-length sequence complementarity, but only marginal effect on one harboring a sequence complementary to its seed sequence. Consistently, miR-S1-3p downregulated LTag and STag transcripts with complete sequence complementarity through miR-S1-3p-Ago2-mediated mRNA decay. Transfection of SV40 plasmid induced higher DNA replication and lower LTag and STag transcripts in most of the examined cells compared to that miR-S1-deficient SV40 plasmid. However, miR-S1 itself did not affect DNA replication without the downregulation of LTag transcripts. Both LTag and STag induced the expression of tumor necrosis factor α (TNFα) and interleukin (IL)-17F, which was slightly reduced by miR-S1 due to miR-S1-mediated downregulation of LTag and STag. Forced miR-S1 expression did not affect TNFα expression, but increased IL-17F expression. Overall, our findings suggest that miR-S1-3p is a latent modifier of LTag and STag functions, ensuring efficient viral replication and attenuating cytokine expression detrimental to the viral life cycle.


Antigens, Viral, Tumor/genetics , Gene Expression Regulation, Viral/immunology , MicroRNAs/metabolism , RNA, Viral/metabolism , Simian virus 40/genetics , A549 Cells , DNA Replication/immunology , DNA, Viral/biosynthesis , HEK293 Cells , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology , Humans , Interleukin-17/metabolism , Interleukin-8/metabolism , Polyomavirus Infections/genetics , Polyomavirus Infections/immunology , Polyomavirus Infections/virology , Simian virus 40/immunology , Tumor Necrosis Factor-alpha/metabolism , Tumor Virus Infections/genetics , Tumor Virus Infections/immunology , Tumor Virus Infections/virology , Virus Replication/immunology
7.
EBioMedicine ; 62: 103132, 2020 Dec.
Article En | MEDLINE | ID: mdl-33232870

BACKGROUND: Due to the lack of protective immunity of humans towards the newly emerged SARS-CoV-2, this virus has caused a massive pandemic across the world resulting in hundreds of thousands of deaths. Thus, a vaccine is urgently needed to contain the spread of the virus. METHODS: Here, we describe Newcastle disease virus (NDV) vector vaccines expressing the spike protein of SARS-CoV-2 in its wild type format or a membrane-anchored format lacking the polybasic cleavage site. All described NDV vector vaccines grow to high titers in embryonated chicken eggs. In a proof of principle mouse study, the immunogenicity and protective efficacy of these NDV-based vaccines were investigated. FINDINGS: We report that the NDV vector vaccines elicit high levels of antibodies that are neutralizing when the vaccine is given intramuscularly in mice. Importantly, these COVID-19 vaccine candidates protect mice from a mouse-adapted SARS-CoV-2 challenge with no detectable viral titer and viral antigen in the lungs. INTERPRETATION: The results suggested that the NDV vector expressing either the wild type S or membrane-anchored S without the polybasic cleavage site could be used as live vector vaccine against SARS-CoV-2. FUNDING: This work is supported by an NIAID funded Center of Excellence for Influenza Research and Surveillance (CEIRS) contract, the Collaborative Influenza Vaccine Innovation Centers (CIVIC) contract, philanthropic donations and NIH grants.


COVID-19 Vaccines , COVID-19 , Gene Expression Regulation, Viral/immunology , Newcastle disease virus , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Animals , COVID-19/genetics , COVID-19/immunology , COVID-19/prevention & control , COVID-19 Vaccines/genetics , COVID-19 Vaccines/immunology , Chlorocebus aethiops , Female , Mice , Mice, Inbred BALB C , Newcastle disease virus/genetics , Newcastle disease virus/immunology , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Vaccines, Live, Unattenuated/genetics , Vaccines, Live, Unattenuated/immunology , Vero Cells
8.
PLoS Pathog ; 16(7): e1008671, 2020 07.
Article En | MEDLINE | ID: mdl-32614923

Viral infection outcomes are governed by the complex and dynamic interplay between the infecting virus population and the host response. It is increasingly clear that both viral and host cell populations are highly heterogeneous, but little is known about how this heterogeneity influences infection dynamics or viral pathogenicity. To dissect the interactions between influenza A virus (IAV) and host cell heterogeneity, we examined the combined host and viral transcriptomes of thousands of individual cells, each infected with a single IAV virion. We observed complex patterns of viral gene expression and the existence of multiple distinct host transcriptional responses to infection at the single cell level. We show that human H1N1 and H3N2 strains differ significantly in patterns of both viral and host anti-viral gene transcriptional heterogeneity at the single cell level. Our analyses also reveal that semi-infectious particles that fail to express the viral NS can play a dominant role in triggering the innate anti-viral response to infection. Altogether, these data reveal how patterns of viral population heterogeneity can serve as a major determinant of antiviral gene activation.


Gene Expression Regulation, Viral/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/immunology , Influenza, Human/virology , A549 Cells , Humans , Immunity, Innate/immunology , Viral Nonstructural Proteins/immunology
9.
PLoS Pathog ; 16(5): e1008537, 2020 05.
Article En | MEDLINE | ID: mdl-32365141

Promyelocytic leukemia (PML) bodies are nuclear organelles implicated in intrinsic and innate antiviral defense. The eponymous PML proteins, central to the self-organization of PML bodies, and other restriction factors found in these organelles are common targets of viral antagonism. The 72-kDa immediate-early protein 1 (IE1) is the principal antagonist of PML bodies encoded by the human cytomegalovirus (hCMV). IE1 is believed to disrupt PML bodies by inhibiting PML SUMOylation, while PML was proposed to act as an E3 ligase for IE1 SUMOylation. PML targeting by IE1 is considered to be crucial for hCMV replication at low multiplicities of infection, in part via counteracting antiviral gene induction linked to the cellular interferon (IFN) response. However, current concepts of IE1-PML interaction are largely derived from mutant IE1 proteins known or predicted to be metabolically unstable and globally misfolded. We performed systematic clustered charge-to-alanine scanning mutagenesis and identified a stable IE1 mutant protein (IE1cc172-176) with wild-type characteristics except for neither interacting with PML proteins nor inhibiting PML SUMOylation. Consequently, IE1cc172-176 does not associate with PML bodies and is selectively impaired for disrupting these organelles. Surprisingly, functional analysis of IE1cc172-176 revealed that the protein is hypermodified by mixed SUMO chains and that IE1 SUMOylation depends on nucleosome rather than PML binding. Furthermore, a mutant hCMV expressing IE1cc172-176 was only slightly attenuated compared to an IE1-null virus even at low multiplicities of infection. Finally, hCMV-induced expression of cytokine and IFN-stimulated genes turned out to be reduced rather than increased in the presence of IE1cc172-176 relative to wild-type IE1. Our findings challenge present views on the relationship of IE1 with PML and the role of PML in hCMV replication. This study also provides initial evidence for the idea that disruption of PML bodies upon viral infection is linked to activation rather than inhibition of innate immunity.


Cytomegalovirus Infections , Cytomegalovirus/physiology , Immediate-Early Proteins , Immunity, Innate , Promyelocytic Leukemia Protein , Virus Replication , Cell Line , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Gene Expression Regulation, Viral/immunology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/immunology , Mutation , Promyelocytic Leukemia Protein/genetics , Promyelocytic Leukemia Protein/immunology , Sumoylation/immunology , Virus Replication/genetics , Virus Replication/immunology
10.
Microbes Infect ; 22(4-5): 188-194, 2020.
Article En | MEDLINE | ID: mdl-32302675

Several research lines are currently ongoing to address the multitude of facets of the pandemic COVID-19. In line with the One-Health concept, extending the target of the studies to the animals which humans are continuously interacting with may favor a better understanding of the SARS-CoV-2 biology and pathogenetic mechanisms; thus, helping to adopt the most suitable containment measures. The last two decades have already faced severe manifestations of the coronavirus infection in both humans and animals, thus, circulating epitopes from previous outbreaks might confer partial protection from SARS-CoV-2 infections. In the present study, we provide an in-silico survey of the major nucleocapsid protein epitopes and compare them with the homologues of taxonomically-related coronaviruses with tropism for animal species that are closely inter-related with the human beings population all over the world. Protein sequence alignment provides evidence of high sequence homology for some of the investigated proteins. Moreover, structural epitope mapping by homology modelling revealed a potential immunogenic value also for specific sequences scoring a lower identity with SARS-CoV-2 nucleocapsid proteins. These evidence provide a molecular structural rationale for a potential role in conferring protection from SARS-CoV-2 infection and identifying potential candidates for the development of diagnostic tools and prophylactic-oriented strategies.


Betacoronavirus/metabolism , Coronavirus/classification , Coronavirus/genetics , Epitopes , Nucleocapsid Proteins/metabolism , Amino Acid Sequence , Animals , Betacoronavirus/genetics , Computational Biology , Computer Simulation , Coronavirus Nucleocapsid Proteins , Gene Expression Regulation, Viral/immunology , Humans , Models, Molecular , Nucleocapsid Proteins/genetics , Phosphoproteins , Phylogeny , Protein Conformation , Protein Domains , SARS-CoV-2 , Species Specificity
11.
Nat Commun ; 11(1): 1997, 2020 04 24.
Article En | MEDLINE | ID: mdl-32332742

Persistent viruses cause chronic disease, and threaten the lives of immunosuppressed individuals. Here, we elucidate a mechanism supporting the persistence of human adenovirus (AdV), a virus that can kill immunosuppressed patients. Cell biological analyses, genetics and chemical interference demonstrate that one of five AdV membrane proteins, the E3-19K glycoprotein specifically triggers the unfolded protein response (UPR) sensor IRE1α in the endoplasmic reticulum (ER), but not other UPR sensors, such as protein kinase R-like ER kinase (PERK) and activating transcription factor 6 (ATF6). The E3-19K lumenal domain activates the IRE1α nuclease, which initiates mRNA splicing of X-box binding protein-1 (XBP1). XBP1s binds to the viral E1A-enhancer/promoter sequence, and boosts E1A transcription, E3-19K levels and lytic infection. Inhibition of IRE1α nuclease interrupts the five components feedforward loop, E1A, E3-19K, IRE1α, XBP1s, E1A enhancer/promoter. This loop sustains persistent infection in the presence of the immune activator interferon, and lytic infection in the absence of interferon.


Adenoviridae Infections/immunology , Adenoviridae/pathogenicity , Adenovirus E3 Proteins/metabolism , Endoribonucleases/metabolism , Gene Expression Regulation, Viral/immunology , Protein Serine-Threonine Kinases/metabolism , A549 Cells , Adenoviridae/genetics , Adenoviridae/immunology , Adenoviridae Infections/genetics , Adenoviridae Infections/virology , Adenovirus E1A Proteins/genetics , Chronic Disease , Endoplasmic Reticulum/metabolism , Endoribonucleases/genetics , Gene Knockdown Techniques , Gene Knockout Techniques , HeLa Cells , Host-Pathogen Interactions/genetics , Humans , Immunocompromised Host , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Protein Serine-Threonine Kinases/genetics , RNA Splicing , Virus Latency , Virus Release/genetics , X-Box Binding Protein 1/genetics
12.
J Virol ; 94(11)2020 05 18.
Article En | MEDLINE | ID: mdl-32213610

Ebola virus (EBOV) continues to pose a significant threat to human health, as evidenced by the 2013-2016 epidemic in West Africa and the ongoing outbreak in the Democratic Republic of the Congo. EBOV causes hemorrhagic fever, organ damage, and shock culminating in death, with case fatality rates as high as 90%. This high lethality combined with the paucity of licensed medical countermeasures makes EBOV a critical human pathogen. Although EBOV infection results in significant damage to the liver and the adrenal glands, little is known about the molecular signatures of injury in these organs. Moreover, while changes in peripheral blood cells are becoming increasingly understood, the host responses within organs and lymphoid tissues remain poorly characterized. To address this knowledge gap, we tracked longitudinal transcriptional changes in tissues collected from EBOV-Makona-infected cynomolgus macaques. Following infection, both liver and adrenal glands exhibited significant and early downregulation of genes involved in metabolism, coagulation, hormone synthesis, and angiogenesis; upregulated genes were associated with inflammation. Analysis of lymphoid tissues showed early upregulation of genes that play a role in innate immunity and inflammation and downregulation of genes associated with cell cycle and adaptive immunity. Moreover, transient activation of innate immune responses and downregulation of humoral immune responses in lymphoid tissues were confirmed with flow cytometry. Together, these data suggest that the liver, adrenal gland, and lymphatic organs are important sites of EBOV infection and that dysregulating the function of these vital organs contributes to the development of Ebola virus disease.IMPORTANCE Ebola virus (EBOV) remains a high-priority pathogen since it continues to cause outbreaks with high case fatality rates. Although it is well established that EBOV results in severe organ damage, our understanding of tissue injury in the liver, adrenal glands, and lymphoid tissues remains limited. We begin to address this knowledge gap by conducting longitudinal gene expression studies in these tissues, which were collected from EBOV-infected cynomolgus macaques. We report robust and early gene expression changes within these tissues, indicating they are primary sites of EBOV infection. Furthermore, genes involved in metabolism, coagulation, and adaptive immunity were downregulated, while inflammation-related genes were upregulated. These results indicate significant tissue damage consistent with the development of hemorrhagic fever and lymphopenia. Our study provides novel insight into EBOV-host interactions and elucidates how host responses within the liver, adrenal glands, and lymphoid tissues contribute to EBOV pathogenesis.


Adrenal Glands , Ebolavirus , Gene Expression Regulation, Viral/immunology , Hemorrhagic Fever, Ebola , Liver , Lymphoid Tissue , Monkey Diseases , Transcription, Genetic/immunology , Adrenal Glands/immunology , Adrenal Glands/metabolism , Adrenal Glands/pathology , Adrenal Glands/virology , Animals , Ebolavirus/immunology , Ebolavirus/metabolism , Female , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/metabolism , Hemorrhagic Fever, Ebola/pathology , Hemorrhagic Fever, Ebola/veterinary , Liver/immunology , Liver/metabolism , Liver/pathology , Liver/virology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Lymphoid Tissue/pathology , Lymphoid Tissue/virology , Macaca fascicularis , Male , Monkey Diseases/immunology , Monkey Diseases/metabolism , Monkey Diseases/pathology , Monkey Diseases/virology
13.
Sci Rep ; 10(1): 2227, 2020 02 10.
Article En | MEDLINE | ID: mdl-32042107

HIV-1 viral transcription persists in patients despite antiretroviral treatment, potentially due to intermittent HIV-1 LTR activation. While several mathematical models have been explored in the context of LTR-protein interactions, in this work for the first time HIV-1 LTR model featuring repressed, intermediate, and activated LTR states is integrated with generation of long (env) and short (TAR) RNAs and proteins (Tat, Pr55, and p24) in T-cells and macrophages using both cell lines and infected primary cells. This type of extended modeling framework allows us to compare and contrast behavior of these two cell types. We demonstrate that they exhibit unique LTR dynamics, which ultimately results in differences in the magnitude of viral products generated. One of the distinctive features of this work is that it relies on experimental data in reaction rate computations. Two RNA transcription rates from the activated promoter states are fit by comparison of experimental data to model predictions. Fitting to the data also provides estimates for the degradation/exit rates for long and short viral RNA. Our experimentally generated data is in reasonable agreement for the T-cell as well macrophage population and gives strong evidence in support of using the proposed integrated modeling paradigm. Sensitivity analysis performed using Latin hypercube sampling method confirms robustness of the model with respect to small parameter perturbations. Finally, incorporation of a transcription inhibitor (F07#13) into the governing equations demonstrates how the model can be used to assess drug efficacy. Collectively, our model indicates transcriptional differences between latently HIV-1 infected T-cells and macrophages and provides a novel platform to study various transcriptional dynamics leading to latency or activation in numerous cell types and physiological conditions.


Anti-HIV Agents/pharmacology , Gene Expression Regulation, Viral/immunology , HIV Infections/drug therapy , HIV-1/genetics , Macrophages/immunology , T-Lymphocytes/immunology , Anti-HIV Agents/therapeutic use , Cell Line , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , Drug Resistance, Viral/immunology , HIV Infections/blood , HIV Infections/immunology , HIV Long Terminal Repeat/genetics , HIV-1/drug effects , HIV-1/immunology , Humans , Macrophages/virology , Models, Genetic , Models, Immunological , Primary Cell Culture , RNA, Viral/genetics , RNA, Viral/metabolism , T-Lymphocytes/virology , Transcription, Genetic/drug effects , Transcription, Genetic/immunology , Virus Replication/drug effects , Virus Replication/genetics , Virus Replication/immunology
14.
mBio ; 10(6)2019 12 03.
Article En | MEDLINE | ID: mdl-31796538

Human cytomegalovirus (HCMV) latency is an active process which remodels the latently infected cell to optimize latent carriage and reactivation. This is achieved, in part, through the expression of viral genes, including the G-protein-coupled receptor US28. Here, we use an unbiased proteomic screen to assess changes in host proteins induced by US28, revealing that interferon-inducible genes are downregulated by US28. We validate that major histocompatibility complex (MHC) class II and two pyrin and HIN domain (PYHIN) proteins, myeloid cell nuclear differentiation antigen (MNDA) and IFI16, are downregulated during experimental latency in primary human CD14+ monocytes. We find that IFI16 is targeted rapidly during the establishment of latency in a US28-dependent manner but only in undifferentiated myeloid cells, a natural site of latent carriage. Finally, by overexpressing IFI16, we show that IFI16 can activate the viral major immediate early promoter and immediate early gene expression during latency via NF-κB, a function which explains why downregulation of IFI16 during latency is advantageous for the virus.IMPORTANCE Human cytomegalovirus (HCMV) is a ubiquitous herpesvirus which infects 50 to 100% of humans worldwide. HCMV causes a lifelong subclinical infection in immunocompetent individuals but is a serious cause of mortality and morbidity in the immunocompromised and neonates. In particular, reactivation of HCMV in the transplant setting is a major cause of transplant failure and related disease. Therefore, a molecular understanding of HCMV latency and reactivation could provide insights into potential ways to target the latent viral reservoir in at-risk patient populations.


Cytomegalovirus Infections/genetics , Cytomegalovirus/immunology , Interferons/genetics , Virus Latency/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Down-Regulation/genetics , Down-Regulation/immunology , Gene Expression/genetics , Gene Expression/immunology , Gene Expression Regulation, Viral/genetics , Gene Expression Regulation, Viral/immunology , HEK293 Cells , Humans , Interferons/immunology , Monocytes/immunology , Monocytes/virology , Myeloid Cells/immunology , Myeloid Cells/virology , NF-kappa B/genetics , NF-kappa B/immunology , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Proteomics/methods , Receptors, G-Protein-Coupled/immunology , THP-1 Cells , Viral Proteins/genetics , Viral Proteins/immunology , Virus Activation/genetics , Virus Activation/immunology , Virus Latency/immunology
15.
Cell Rep ; 28(1): 245-256.e4, 2019 07 02.
Article En | MEDLINE | ID: mdl-31269444

Type I interferon (IFNα/ß) pathways are fine-tuned to elicit antiviral protection while minimizing immunopathology; however, the initiating stimuli, target tissues, and underlying mechanisms are unclear. Using models of physiological and dysregulated IFNα/ß receptor (IFNAR1) surface expression, we show here that IFNAR1-dependent signals set the steady-state IFN signature in both hematopoietic and stromal cells. Increased IFNAR1 levels promote a lung environment refractory to early influenza virus replication by elevating the baseline interferon signature. Commensal microbiota drive the IFN signature specifically in lung stroma, as shown by antibiotic treatment and fecal transplantation. Bone marrow chimera experiments identify lung stromal cells as crucially important for early antiviral immunity and stroma-immune cell interaction for late antiviral resistance. We propose that the microbiota-driven interferon signature in lung epithelia impedes early virus replication and that IFNAR1 surface levels fine-tune this signature. Our findings highlight the interplay between bacterial and viral exposure, with important implications for antibiotic use.


Anti-Bacterial Agents/pharmacology , Influenza A virus , Influenza, Human/immunology , Influenza, Human/microbiology , Lung/immunology , Microbiota/immunology , Receptor, Interferon alpha-beta/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Line , Chimera/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Fecal Microbiota Transplantation , Gene Expression Regulation, Viral/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/virology , Humans , Influenza A virus/growth & development , Influenza A virus/immunology , Influenza, Human/drug therapy , Influenza, Human/pathology , Interferon Type I/metabolism , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Lung/drug effects , Lung/microbiology , Lung/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA-Seq , Receptor, Interferon alpha-beta/genetics , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/microbiology , Stromal Cells/virology
16.
Front Immunol ; 10: 267, 2019.
Article En | MEDLINE | ID: mdl-30842775

Endogenous plasma levels of the immunomodulatory carbohydrate-binding protein galectin-9 (Gal-9) are elevated during HIV infection and remain elevated after antiretroviral therapy (ART) suppression. We recently reported that Gal-9 regulates HIV transcription and potently reactivates latent HIV. However, the signaling mechanisms underlying Gal-9-mediated viral transcription remain unclear. Given that galectins are known to modulate T cell receptor (TCR)-signaling, we hypothesized that Gal-9 modulates HIV transcriptional activity, at least in part, through inducing TCR signaling pathways. Gal-9 induced T cell receptor ζ chain (CD3ζ) phosphorylation (11.2 to 32.1%; P = 0.008) in the J-Lat HIV latency model. Lck inhibition reduced Gal-9-mediated viral reactivation in the J-Lat HIV latency model (16.8-0.9%; P < 0.0001) and reduced both Gal-9-mediated CD4+ T cell activation (10.3 to 1.65% CD69 and CD25 co-expression; P = 0.0006), and IL-2/TNFα secretion (P < 0.004) in primary CD4+ T cells from HIV-infected individuals on suppressive ART. Using phospho-kinase antibody arrays, we found that Gal-9 increased the phosphorylation of the TCR-downstream signaling molecules ERK1/2 (26.7-fold) and CREB (6.6-fold). ERK and CREB inhibitors significantly reduced Gal-9-mediated viral reactivation (16.8 to 2.6 or 12.6%, respectively; P < 0.0007). Given that the immunosuppressive rapamycin uncouples HIV latency reversal from cytokine-associated toxicity, we also investigated whether rapamycin could uncouple Gal-9-mediated latency reactivation from its concurrent pro-inflammatory cytokine production. Rapamycin reduced Gal-9-mediated secretion of IL-2 (4.4-fold, P = 0.001) and TNF (4-fold, P = 0.02) without impacting viral reactivation (16.8% compared to 16.1%; P = 0.2). In conclusion, Gal-9 modulates HIV transcription by activating the TCR-downstream ERK and CREB signaling pathways in an Lck-dependent manner. Our findings could have implications for understanding the role of endogenous galectin interactions in modulating TCR signaling and maintaining chronic immune activation during ART-suppressed HIV infection. In addition, uncoupling Gal-9-mediated viral reactivation from undesirable pro-inflammatory effects, using rapamycin, may increase the potential utility of recombinant Gal-9 within the reversal of HIV latency eradication framework.


Galectins/immunology , HIV Infections/immunology , HIV-1/immunology , MAP Kinase Signaling System/immunology , Receptors, Antigen, T-Cell/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cell Line, Tumor , Gene Expression Regulation, Viral/immunology , Humans , Jurkat Cells , Lymphocyte Activation/immunology , Signal Transduction/immunology , Transcription, Genetic/immunology , Tumor Necrosis Factor-alpha/immunology , Virus Activation/immunology , Virus Latency/immunology , Virus Replication/immunology
17.
J Infect Dis ; 218(12): 1911-1921, 2018 11 05.
Article En | MEDLINE | ID: mdl-30010906

Background: Several promising live attenuated dengue vaccines are in development, but information about innate immune responses and early correlates of protection is lacking. Methods: We characterized human genome-wide transcripts in whole blood from 10 volunteers at 11 time points after immunization with the dengue virus type 3 (DENV-3) component of the National Institutes of Health dengue vaccine candidate TV003 and from 30 hospitalized children with acute primary DENV-3 infection. We compared day-specific gene expression patterns with subsequent neutralizing antibody (NAb) titers. Results: The transcriptional response to vaccination was largely confined to days 5-20 and was dominated by an interferon-associated signature and a cell cycle signature that peaked on days 8 and 14, respectively. Changes in transcript abundance were much greater in magnitude and scope in symptomatic natural infection than following vaccination (maximum fold-change >200 vs 21 postvaccination; 3210 vs 286 transcripts with significant fold-change), but shared gene modules were induced in the same sequence. The abundances of 131 transcripts on days 8 and 9 postvaccination were strongly correlated with NAb titers measured 6 weeks postvaccination. Conclusions: Live attenuated dengue vaccination elicits early transcriptional responses that mirror those found in symptomatic natural infection and provide candidate early markers of protection against DENV infection. Clinical Trials Registration: NCT00831012.


Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Dengue Vaccines/immunology , Dengue/prevention & control , Gene Expression Regulation, Viral/immunology , Adolescent , Adult , Dengue/blood , Dengue/immunology , Humans , Male , Middle Aged , Time Factors , Transcription, Genetic/immunology , Vaccination , Vaccines, Attenuated/immunology , Young Adult
18.
J Infect Dis ; 218(8): 1238-1248, 2018 09 08.
Article En | MEDLINE | ID: mdl-29846612

Background: Avian influenza A (H7N9) viruses emerged in China in 2013 and caused zoonotic disease associated with a case-fatality ratio of over 30%. Transcriptional profiles in peripheral blood reflect host responses and can help to elucidate disease pathogenesis. Methods: We correlated serial blood transcriptomic profiles of patients with avian influenza A (H7N9) virus infection and determined the biological significances from the analysis. Results: We found that specific gene expression profiles in the blood were strongly correlated with the Pao 2/Fio 2 ratio and viral load in the lower respiratory tract. Cell cycle and leukocyte-related immunity were activated at the acute stage of the infection while T-cell functions and various metabolic processes were associated with the recovery phase of the illness. A transition from systemic innate to adaptive immunity was found. Conclusions: We developed a novel approach for transcriptomic analysis to identify key host responses that were strongly correlated with specific clinical and virologic parameters in patients with H7N9 infection.


Influenza A Virus, H7N9 Subtype , Influenza, Human/virology , Case-Control Studies , Gene Expression Regulation, Viral/immunology , Humans , Immunity, Cellular/genetics , Immunity, Cellular/physiology , Influenza, Human/metabolism , Leukocytes/metabolism , Lung , T-Lymphocytes/metabolism , Transcriptome , Viral Load
19.
Cell Rep ; 23(5): 1249-1258, 2018 05 01.
Article En | MEDLINE | ID: mdl-29719242

Post-translational modification of ribosomal subunit proteins (RPs) is emerging as an important means of regulating gene expression. Recently, regulatory ubiquitination of small RPs RPS10 and RPS20 by the ubiquitin ligase ZNF598 was found to function in ribosome sensing and stalling on internally polyadenylated mRNAs during ribosome quality control (RQC). Here, we reveal that ZNF598 and RPS10 negatively regulate interferon-stimulated gene (ISG) expression in primary cells, depletion of which induced ISG expression and a broad antiviral state. However, cell lines lacking interferon responses revealed that ZNF598 E3 ligase activity and ubiquitination of RPS20, but not RPS10, were specifically required for poxvirus replication and synthesis of poxvirus proteins whose encoding mRNAs contain unusual 5' poly(A) leaders. Our findings reveal distinct functions for ZNF598 and its downstream RPS targets, one that negatively regulates ISG expression and infection by a range of viruses while the other is positively exploited by poxviruses.


Carrier Proteins/immunology , Gene Expression Regulation, Viral/immunology , Poxviridae Infections/immunology , Poxviridae/immunology , Protein Biosynthesis/immunology , Viral Proteins/immunology , Carrier Proteins/genetics , HCT116 Cells , HEK293 Cells , Humans , Interferons/genetics , Interferons/immunology , Poxviridae/genetics , Poxviridae Infections/genetics , Ribosomal Proteins/genetics , Ribosomal Proteins/immunology , Viral Proteins/genetics
20.
Med Hypotheses ; 112: 47-50, 2018 Mar.
Article En | MEDLINE | ID: mdl-29447938

Human Herpes Virus type 6 (HHV-6) is a ubiquitous virus consisting of two viral species, HHV-6A and HHV-6B that have been associated with numerous and diverse pathologies. As many other viruses HHV-6 modulates the apoptotic machinery of its host to subvert immune response to infection, yet the exact mechanisms behind this process remain under investigation. The genes encoding the CTSS, PTX3, CHI3L1, Mx1, CXCL16, BIRC3 and BST2 proteins have been linked to HHV-6Α related neurologic diseases whilst also associated with apoptosis. This study aimed at the identification and functional analysis of the gene interaction network (interactome) of CTSS-PTX3-CHI3L1-Mx1-CXCL16-BIRC3-BST2 so as to evaluate the hypothesis of a probable link between the latter and host's immune response to HHV-6A infection.


Adaptive Immunity/genetics , Apoptosis Regulatory Proteins/genetics , Gene Regulatory Networks , Herpesvirus 6, Human/physiology , Host-Pathogen Interactions/immunology , Models, Genetic , Models, Immunological , Roseolovirus Infections/immunology , Antigens, CD/biosynthesis , Antigens, CD/genetics , Apoptosis Regulatory Proteins/biosynthesis , Baculoviral IAP Repeat-Containing 3 Protein/biosynthesis , Baculoviral IAP Repeat-Containing 3 Protein/genetics , C-Reactive Protein/biosynthesis , C-Reactive Protein/genetics , Cathepsins/biosynthesis , Cathepsins/genetics , Chemokine CXCL16/biosynthesis , Chemokine CXCL16/genetics , Chitinase-3-Like Protein 1/biosynthesis , Chitinase-3-Like Protein 1/genetics , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , Gene Expression Regulation, Viral/immunology , Gene Ontology , Gene Regulatory Networks/genetics , Gene Regulatory Networks/immunology , Host-Pathogen Interactions/genetics , Humans , Immune Evasion/genetics , Myxovirus Resistance Proteins/biosynthesis , Myxovirus Resistance Proteins/genetics , Roseolovirus Infections/genetics , Serum Amyloid P-Component/biosynthesis , Serum Amyloid P-Component/genetics
...