Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.257
Filter
1.
Nat Commun ; 15(1): 5842, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992037

ABSTRACT

Activating interferon responses with STING agonists (STINGa) is a current cancer immunotherapy strategy, and therapeutic modalities that enable tumor-targeted delivery via systemic administration could be beneficial. Here we demonstrate that tumor cell-directed STING agonist antibody-drug-conjugates (STINGa ADCs) activate STING in tumor cells and myeloid cells and induce anti-tumor innate immune responses in in vitro, in vivo (in female mice), and ex vivo tumor models. We show that the tumor cell-directed STINGa ADCs are internalized into myeloid cells by Fcγ-receptor-I in a tumor antigen-dependent manner. Systemic administration of STINGa ADCs in mice leads to STING activation in tumors, with increased anti-tumor activity and reduced serum cytokine elevations compared to a free STING agonist. Furthermore, STINGa ADCs induce type III interferons, which contribute to the anti-tumor activity by upregulating type I interferon and other key chemokines/cytokines. These findings reveal an important role for type III interferons in the anti-tumor activity elicited by STING agonism and provide rationale for the clinical development of tumor cell-directed STINGa ADCs.


Subject(s)
Immunity, Innate , Immunoconjugates , Interferons , Membrane Proteins , Animals , Membrane Proteins/agonists , Membrane Proteins/immunology , Immunity, Innate/drug effects , Female , Humans , Mice , Cell Line, Tumor , Immunoconjugates/pharmacology , Immunoconjugates/administration & dosage , Interferons/metabolism , Interferon Lambda , Neoplasms/immunology , Neoplasms/drug therapy , Interferon Type I/immunology , Cytokines/metabolism , Myeloid Cells/immunology , Myeloid Cells/drug effects , Immunotherapy/methods , Mice, Inbred C57BL , Receptors, IgG/agonists , Receptors, IgG/metabolism , Receptors, IgG/immunology
2.
Front Cell Infect Microbiol ; 14: 1415695, 2024.
Article in English | MEDLINE | ID: mdl-39035358

ABSTRACT

Histone deacetylates family proteins have been studied for their function in regulating viral replication by deacetylating non-histone proteins. RIG-I (Retinoic acid-inducible gene I) is a critical protein in RNA virus-induced innate antiviral signaling pathways. Our previous research showed that HDAC8 (histone deacetylase 8) involved in innate antiviral immune response, but the underlying mechanism during virus infection is still unclear. In this study, we showed that HDAC8 was involved in the regulation of vesicular stomatitis virus (VSV) replication. Over-expression of HDAC8 inhibited while knockdown promoted VSV replication. Further exploration demonstrated that HDAC8 interacted with and deacetylated RIG-I, which eventually lead to enhance innate antiviral immune response. Collectively, our data clearly demonstrated that HDAC8 inhibited VSV replication by promoting RIG-I mediated interferon production and downstream signaling pathway.


Subject(s)
DEAD Box Protein 58 , Histone Deacetylases , Immunity, Innate , Receptors, Immunologic , Signal Transduction , Vesiculovirus , Virus Replication , DEAD Box Protein 58/metabolism , DEAD Box Protein 58/genetics , Humans , Histone Deacetylases/metabolism , Vesiculovirus/immunology , Receptors, Immunologic/metabolism , Repressor Proteins/metabolism , Repressor Proteins/genetics , Acetylation , HEK293 Cells , Interferons/metabolism , Interferons/immunology , Cell Line , Host-Pathogen Interactions/immunology , Animals , Vesicular stomatitis Indiana virus/immunology
3.
J Exp Med ; 221(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-38953896

ABSTRACT

Gain-of-function mutations in STING cause STING-associated vasculopathy with onset in infancy (SAVI) characterized by early-onset systemic inflammation, skin vasculopathy, and interstitial lung disease. Here, we report and characterize a novel STING variant (F269S) identified in a SAVI patient. Single-cell transcriptomics of patient bone marrow revealed spontaneous activation of interferon (IFN) and inflammatory pathways across cell types and a striking prevalence of circulating naïve T cells was observed. Inducible STING F269S expression conferred enhanced signaling through ligand-independent translocation of the protein to the Golgi, protecting cells from viral infections but preventing their efficient immune priming. Additionally, endothelial cell activation was promoted and further exacerbated by cytokine secretion by SAVI immune cells, resulting in inflammation and endothelial damage. Our findings identify STING F269S mutation as a novel pathogenic variant causing SAVI, highlight the importance of the crosstalk between endothelial and immune cells in the context of lung disease, and contribute to a better understanding of how aberrant STING activation can cause pathology.


Subject(s)
Endothelial Cells , Membrane Proteins , Humans , Infant , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gain of Function Mutation , Golgi Apparatus/metabolism , Interferons/metabolism , Interferons/genetics , Lung Diseases, Interstitial/genetics , Lung Diseases, Interstitial/pathology , Lung Diseases, Interstitial/immunology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Signal Transduction , Vascular Diseases/genetics , Vascular Diseases/pathology , Infant, Newborn , Child, Preschool , Female
4.
JCI Insight ; 9(13)2024 May 21.
Article in English | MEDLINE | ID: mdl-38973611

ABSTRACT

Optimization of protective immune responses against SARS-CoV-2 remains an urgent worldwide priority. In this regard, type III IFN (IFN-λ) restricts SARS-CoV-2 infection in vitro, and treatment with IFN-λ limits infection, inflammation, and pathogenesis in murine models. Furthermore, IFN-λ has been developed for clinical use to limit COVID-19 severity. However, whether endogenous IFN-λ signaling has an effect on SARS-CoV-2 antiviral immunity and long-term immune protection in vivo is unknown. In this study, we identified a requirement for IFN-λ signaling in promoting viral clearance and protective immune programming in SARS-CoV-2 infection of mice. Expression of both IFN and IFN-stimulated gene (ISG) in the lungs were minimally affected by the absence of IFN-λ signaling and correlated with transient increases in viral titers. We found that IFN-λ supported the generation of protective CD8 T cell responses against SARS-CoV-2 by facilitating accumulation of CD103+ DC in lung draining lymph nodes (dLN). IFN-λ signaling specifically in DCs promoted the upregulation of costimulatory molecules and the proliferation of CD8 T cells. Intriguingly, antigen-specific CD8 T cell immunity to SARS-CoV-2 was independent of type I IFN signaling, revealing a nonredundant function of IFN-λ. Overall, these studies demonstrate a critical role for IFN-λ in protective innate and adaptive immunity upon infection with SARS-CoV-2 and suggest that IFN-λ serves as an immune adjuvant to support CD8 T cell immunity.


Subject(s)
CD8-Positive T-Lymphocytes , COVID-19 , Interferon Type I , SARS-CoV-2 , Animals , CD8-Positive T-Lymphocytes/immunology , SARS-CoV-2/immunology , Mice , COVID-19/immunology , COVID-19/virology , Interferon Type I/immunology , Interferon Type I/metabolism , Lung/immunology , Lung/virology , Signal Transduction/immunology , Disease Models, Animal , Interferon Lambda , Interferons/immunology , Interferons/metabolism , Mice, Inbred C57BL , Mice, Knockout , Dendritic Cells/immunology , Humans
5.
Int J Mol Sci ; 25(13)2024 Jun 24.
Article in English | MEDLINE | ID: mdl-39000027

ABSTRACT

Cytokines, chemokines, and interferons are released in response to viral infection with the ultimate aim of viral clearance. However, in SARS-CoV-2 infection, there is an imbalanced immune response, with raised cytokine levels but only a limited interferon response with inefficient viral clearance. Furthermore, the inflammatory response can be exaggerated, which risks both acute and chronic sequelae. Several observational studies have suggested a reduced risk of progression to severe COVID-19 in subjects with a higher omega-3 index. However, randomized studies of omega-3 supplementation have failed to replicate this benefit. Omega-3 fats provide important anti-inflammatory effects; however, fatty fish contains many other fatty acids that provide health benefits distinct from omega-3. Therefore, the immune health benefit of whole salmon oil (SO) was assessed in adults with mild to moderate COVID-19. Eleven subjects were randomized to best supportive care (BSC) with or without a full spectrum, enzymatically liberated SO, dosed at 4g daily, for twenty-eight days. Nasal swabs were taken to measure the change in gene expression of markers of immune response and showed that the SO provided both broad inflammation-resolving effects and improved interferon response. The results also suggest improved lung barrier function and enhanced immune memory, although the clinical relevance needs to be assessed in longer-duration studies. In conclusion, the salmon oil was well tolerated and provided broad inflammation-resolving effects, indicating a potential to enhance immune health.


Subject(s)
COVID-19 , Chemokines , Cytokines , Fish Oils , Interferons , SARS-CoV-2 , Humans , Fish Oils/pharmacology , Fish Oils/therapeutic use , COVID-19/immunology , COVID-19/virology , Male , Interferons/metabolism , Interferons/genetics , SARS-CoV-2/immunology , Cytokines/metabolism , Female , Middle Aged , Chemokines/metabolism , Chemokines/genetics , Adult , COVID-19 Drug Treatment , Fatty Acids, Omega-3/pharmacology
6.
Cell Rep Med ; 5(7): 101631, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38986623

ABSTRACT

Ovarian cancer (OC) manifests as a complex disease characterized by inter- and intra-patient heterogeneity. Despite enhanced biological and genetic insights, OC remains a recalcitrant malignancy with minimal survival improvement. Based on multi-site sampling and a multi-lineage patient-derived xenograft (PDX) establishment strategy, we present herein the establishment of a comprehensive PDX biobank from histologically and molecularly heterogeneous OC patients. Comprehensive profiling of matched PDX and patient samples demonstrates that PDXs closely recapitulate parental tumors. By leveraging multi-lineage models, we reveal that the previously reported genomic disparities of PDX could be mainly attributed to intra-patient spatial heterogeneity instead of substantial model-independent genomic evolution. Moreover, DNA damage response pathway inhibitor (DDRi) screening uncovers heterogeneous responses across models. Prolonged iterative drug exposure recapitulates acquired drug resistance in initially sensitive models. Meanwhile, interrogation of induced drug-resistant (IDR) models reveals that suppressed interferon (IFN) response and activated Wnt/ß-catenin signaling contribute to acquired DDRi drug resistance.


Subject(s)
Ovarian Neoplasms , Humans , Female , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Animals , Mice , Xenograft Model Antitumor Assays , Wnt Signaling Pathway/genetics , Drug Resistance, Neoplasm/genetics , Genomics/methods , Biological Specimen Banks , Genetic Heterogeneity , DNA Damage/genetics , Interferons/metabolism , Interferons/genetics , Cell Lineage/genetics
7.
Molecules ; 29(13)2024 Jul 07.
Article in English | MEDLINE | ID: mdl-38999177

ABSTRACT

A short 19 bp dsRNA with 3'-trinucleotide overhangs acting as immunostimulating RNA (isRNA) demonstrated strong antiproliferative action against cancer cells, immunostimulatory activity through activation of cytokines and Type-I IFN secretion, as well as anti-tumor and anti-metastatic effects in vivo. The aim of this study was to determine the tolerance of chemical modifications (2'-F, 2'-OMe, PS, cholesterol, and amino acids) located at different positions within this isRNA to its ability to activate the innate immune system. The obtained duplexes were tested in vivo for their ability to activate the synthesis of interferon-α in mice, and in tumor cell cultures for their ability to inhibit their proliferation. The obtained data show that chemical modifications in the composition of isRNA have different effects on its individual functions, including interferon-inducing and antiproliferative effects. The effect of modifications depends not only on the type of modification but also on its location and the surrounding context of the modifications. This study made it possible to identify leader patterns of modifications that enhance the properties of isRNA: F2/F2 and F2_S/F2 for interferon-inducing activity, as well as F2_S5/F2_S5, F2-NH2/F2-NH2, and Ch-F2/Ch-F2 for antiproliferative action. These modifications can improve the pharmacokinetic and pharmacodynamic properties, as well as increase the specificity of isRNA action to obtain the desired effect.


Subject(s)
Cell Proliferation , RNA, Double-Stranded , RNA, Double-Stranded/pharmacology , RNA, Double-Stranded/chemistry , Animals , Cell Proliferation/drug effects , Mice , Humans , Cell Line, Tumor , Interferon-alpha/metabolism , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/chemistry , Interferons/metabolism
8.
Mol Cell ; 84(13): 2436-2454.e10, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38925114

ABSTRACT

Signal transduction proteins containing a pLxIS motif induce interferon (IFN) responses central to antiviral immunity. Apart from their established roles in activating the IFN regulator factor (IRF) transcription factors, the existence of additional pathways and functions associated with the pLxIS motif is unknown. Using a synthetic biology-based platform, we identified two orphan pLxIS-containing proteins that stimulate IFN responses independent of all known pattern-recognition receptor pathways. We further uncovered a diversity of pLxIS signaling mechanisms, where the pLxIS motif represents one component of a multi-motif signaling entity, which has variable functions in activating IRF3, the TRAF6 ubiquitin ligase, IκB kinases, mitogen-activated protein kinases, and metabolic activities. The most diverse pLxIS signaling mechanisms were associated with the highest antiviral activities in human cells. The flexibility of domains that regulate IFN signaling may explain their prevalence in nature.


Subject(s)
Interferon Regulatory Factor-3 , Interferons , Signal Transduction , TNF Receptor-Associated Factor 6 , Humans , Interferons/metabolism , HEK293 Cells , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , TNF Receptor-Associated Factor 6/metabolism , TNF Receptor-Associated Factor 6/genetics , I-kappa B Kinase/metabolism , I-kappa B Kinase/genetics , Protein Domains , Animals , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Amino Acid Motifs , Mitogen-Activated Protein Kinases/metabolism
9.
Vet Res ; 55(1): 83, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38943190

ABSTRACT

Migratory birds are important vectors for virus transmission, how migratory birds recognize viruses and viruses are sustained in birds is still enigmatic. As an animal model for waterfowl among migratory birds, studying and dissecting the antiviral immunity and viral evasion in duck cells may pave a path to deciphering these puzzles. Here, we studied the mechanism of antiviral autophagy mediated by duck STING in DEF cells. The results collaborated that duck STING could significantly enhance LC3B-II/I turnover, LC3B-EGFP puncta formation, and mCherry/EGFP ratio, indicating that duck STING could induce autophagy. The autophagy induced by duck STING is not affected by shRNA knockdown of ATG5 expression, deletion of the C-terminal tail of STING, or TBK1 inhibitor BX795 treatment, indicating that duck STING activated non-classical selective autophagy is independent of interaction with TBK1, TBK1 phosphorylation, and interferon (IFN) signaling. The STING R235A mutant and Sar1A/B kinase mutant abolished duck STING induced autophagy, suggesting binding with cGAMP and COPII complex mediated transport are the critical prerequisite. Duck STING interacted with LC3B through LIR motifs to induce autophagy, the LIR 4/7 motif mutants of duck STING abolished the interaction with LC3B, and neither activated autophagy nor IFN expression, indicating that duck STING associates with LC3B directed autophagy and dictated innate immunity activation. Finally, we found that duck STING mediated autophagy significantly inhibited duck plague virus (DPV) infection via ubiquitously degraded viral proteins. Our study may shed light on one scenario about the control and evasion of diseases transmitted by migratory birds.


Subject(s)
Autophagy , Ducks , Signal Transduction , Animals , Mardivirus/physiology , Interferons/metabolism , Alphaherpesvirinae/physiology , Immunity, Innate , Membrane Proteins/metabolism , Membrane Proteins/genetics , Poxviridae Infections/veterinary , Poxviridae Infections/immunology , Poxviridae Infections/virology
10.
Curr Opin Immunol ; 87: 102430, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38824869

ABSTRACT

Lambda interferons (IFNλs), also termed type III interferons (IFNs) or interleukins-28/29, have been in the shadow of type I IFNs for a long time. Their common induction mechanisms and signalling cascades with type I IFNs have made difficult the unwinding of their unique nonredundant functions. However, this is now changing with mounting evidence supporting a major role of IFNλs as a specialized antiviral defense system in the body, mediating protection at mucosal barrier surfaces while limiting immunopathology. Here, we review the latest progress on the complex activities of IFNλs in the respiratory tract, focusing on their multiple effects in IFNλ receptor-expressing cells, the modulation of innate and adaptive immune responses in the context of infections and respiratory diseases, and their similarities and differences with type I IFNs. We also discuss their potential in therapeutic applications and the most recent developments in that direction.


Subject(s)
Adaptive Immunity , Immunity, Innate , Interferon Lambda , Interferons , Respiratory System , Humans , Animals , Interferons/metabolism , Interferons/immunology , Respiratory System/immunology , Respiratory System/metabolism , Signal Transduction/immunology , Interferon Type I/metabolism , Interferon Type I/immunology
11.
Viruses ; 16(6)2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38932225

ABSTRACT

The innate immune system, particularly the interferon (IFN) system, constitutes the initial line of defense against viral infections. IFN signaling induces the expression of interferon-stimulated genes (ISGs), and their products frequently restrict viral infection. Retroviruses like the human immunodeficiency viruses and the human T-lymphotropic viruses cause severe human diseases and are targeted by ISG-encoded proteins. Here, we discuss ISGs that inhibit the translation of retroviral mRNAs and thereby retrovirus propagation. The Schlafen proteins degrade cellular tRNAs and rRNAs needed for translation. Zinc Finger Antiviral Protein and RNA-activated protein kinase inhibit translation initiation factors, and Shiftless suppresses translation recoding essential for the expression of retroviral enzymes. We outline common mechanisms that underlie the antiviral activity of multifunctional ISGs and discuss potential antiretroviral therapeutic approaches based on the mode of action of these ISGs.


Subject(s)
Interferons , Protein Biosynthesis , Retroviridae , Humans , Interferons/immunology , Interferons/metabolism , Interferons/genetics , Retroviridae/genetics , Retroviridae/physiology , Immunity, Innate , Animals , Signal Transduction , Retroviridae Infections/virology , Retroviridae Infections/immunology , Retroviridae Infections/genetics
12.
Viruses ; 16(6)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38932230

ABSTRACT

Type I interferons (IFN-Is) are pivotal in innate immunity against human immunodeficiency virus I (HIV-1) by eliciting the expression of IFN-stimulated genes (ISGs), which encompass potent host restriction factors. While ISGs restrict the viral replication within the host cell by targeting various stages of the viral life cycle, the lesser-known IFN-repressed genes (IRepGs), including RNA-binding proteins (RBPs), affect the viral replication by altering the expression of the host dependency factors that are essential for efficient HIV-1 gene expression. Both the host restriction and dependency factors determine the viral replication efficiency; however, the understanding of the IRepGs implicated in HIV-1 infection remains greatly limited at present. This review provides a comprehensive overview of the current understanding regarding the impact of the RNA-binding protein families, specifically the two families of splicing-associated proteins SRSF and hnRNP, on HIV-1 gene expression and viral replication. Since the recent findings show specifically that SRSF1 and hnRNP A0 are regulated by IFN-I in various cell lines and primary cells, including intestinal lamina propria mononuclear cells (LPMCs) and peripheral blood mononuclear cells (PBMCs), we particularly discuss their role in the context of the innate immunity affecting HIV-1 replication.


Subject(s)
HIV Infections , HIV-1 , Immunity, Innate , Virus Replication , HIV-1/genetics , HIV-1/physiology , Humans , HIV Infections/virology , HIV Infections/genetics , HIV Infections/immunology , Gene Expression Regulation, Viral , RNA Splicing Factors/metabolism , RNA Splicing Factors/genetics , Interferon Type I/metabolism , Interferon Type I/genetics , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , Interferons/metabolism , Interferons/genetics , Interferons/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
13.
Viruses ; 16(6)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38932231

ABSTRACT

Porcine hemagglutinating encephalomyelitis virus (PHEV) replicates in the upper respiratory tract and tonsils of pigs. Using an air-liquid interface porcine respiratory epithelial cells (ALI-PRECs) culture system, we demonstrated that PHEV disrupts respiratory epithelia homeostasis by impairing ciliary function and inducing antiviral, pro-inflammatory cytokine, and chemokine responses. This study explores the mechanisms driving early innate immune responses during PHEV infection through host transcriptome analysis. Total RNA was collected from ALI-PRECs at 24, 36, and 48 h post inoculation (hpi). RNA-seq analysis was performed using an Illumina Hiseq 600 to generate 100 bp paired-end reads. Differential gene expression was analyzed using DeSeq2. PHEV replicated actively in ALI-PRECs, causing cytopathic changes and progressive mucociliary disruption. Transcriptome analysis revealed downregulation of cilia-associated genes such as CILK1, DNAH11, LRRC-23, -49, and -51, and acidic sialomucin CD164L2. PHEV also activated antiviral signaling pathways, significantly increasing the expression of interferon-stimulated genes (RSAD2, MX1, IFIT, and ISG15) and chemokine genes (CCL5 and CXCL10), highlighting inflammatory regulation. This study contributes to elucidating the molecular mechanisms of the innate immune response to PHEV infection of the airway epithelium, emphasizing the critical roles of the mucociliary, interferon, and chemokine responses.


Subject(s)
Betacoronavirus 1 , Epithelial Cells , Gene Expression Profiling , Interferons , Animals , Swine , Epithelial Cells/virology , Epithelial Cells/immunology , Interferons/genetics , Interferons/metabolism , Interferons/immunology , Betacoronavirus 1/immunology , Betacoronavirus 1/genetics , Immunity, Innate , Virus Replication , Coronavirus Infections/immunology , Coronavirus Infections/virology , Coronavirus Infections/veterinary , Cytokines/metabolism , Cytokines/genetics , Cytokines/immunology , Transcriptome , Respiratory Mucosa/virology , Respiratory Mucosa/immunology , Swine Diseases/virology , Swine Diseases/immunology , Swine Diseases/genetics , Cells, Cultured , Deltacoronavirus
15.
Nat Commun ; 15(1): 4920, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858353

ABSTRACT

The differentiation of the stroma is a hallmark event during postnatal uterine development. However, the spatiotemporal changes that occur during this process and the underlying regulatory mechanisms remain elusive. Here, we comprehensively delineated the dynamic development of the neonatal uterus at single-cell resolution and characterized two distinct stromal subpopulations, inner and outer stroma. Furthermore, single-cell RNA sequencing revealed that uterine ablation of Pr-set7, the sole methyltransferase catalyzing H4K20me1, led to a reduced proportion of the inner stroma due to massive cell death, thus impeding uterine development. By combining RNA sequencing and epigenetic profiling of H4K20me1, we demonstrated that PR-SET7-H4K20me1 either directly repressed the transcription of interferon stimulated genes or indirectly restricted the interferon response via silencing endogenous retroviruses. Declined H4K20me1 level caused viral mimicry responses and ZBP1-mediated apoptosis and necroptosis in stromal cells. Collectively, our study provides insight into the epigenetic machinery governing postnatal uterine stromal development mediated by PR-SET7.


Subject(s)
Epigenesis, Genetic , Histone-Lysine N-Methyltransferase , Stromal Cells , Uterus , Female , Animals , Uterus/metabolism , Stromal Cells/metabolism , Mice , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Interferons/metabolism , Interferons/genetics , Endogenous Retroviruses/genetics , Apoptosis/genetics , Mice, Inbred C57BL , Cell Death/genetics , Necroptosis/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Histones/metabolism , Single-Cell Analysis , Mice, Knockout , Cell Differentiation/genetics
16.
Elife ; 132024 Jun 28.
Article in English | MEDLINE | ID: mdl-38941138

ABSTRACT

SARS-CoV-2 induces delayed type-I/III interferon production, allowing it to escape the early innate immune response. The delay has been attributed to a deficiency in the ability of cells to sense viral replication upon infection, which in turn hampers activation of the antiviral state in bystander cells. Here, we introduce a cellular automaton model to investigate the spatiotemporal spreading of viral infection as a function of virus and host-dependent parameters. The model suggests that the considerable person-to-person heterogeneity in SARS-CoV-2 infections is a consequence of high sensitivity to slight variations in biological parameters near a critical threshold. It further suggests that within-host viral proliferation can be curtailed by the presence of remarkably few cells that are primed for IFN production. Thus, the observed heterogeneity in defense readiness of cells reflects a remarkably cost-efficient strategy for protection.


Subject(s)
COVID-19 , SARS-CoV-2 , SARS-CoV-2/physiology , Humans , COVID-19/virology , COVID-19/immunology , Virus Replication , Immunity, Innate , Epithelial Cells/virology , Interferons/metabolism
17.
Elife ; 132024 Jun 11.
Article in English | MEDLINE | ID: mdl-38860651

ABSTRACT

The autoimmune disease lupus erythematosus (lupus) is characterized by photosensitivity, where even ambient ultraviolet radiation (UVR) exposure can lead to development of inflammatory skin lesions. We have previously shown that Langerhans cells (LCs) limit keratinocyte apoptosis and photosensitivity via a disintegrin and metalloprotease 17 (ADAM17)-mediated release of epidermal growth factor receptor (EGFR) ligands and that LC ADAM17 sheddase activity is reduced in lupus. Here, we sought to understand how the lupus skin environment contributes to LC ADAM17 dysfunction and, in the process, differentiate between effects on LC ADAM17 sheddase function, LC ADAM17 expression, and LC numbers. We show through transcriptomic analysis a shared IFN-rich environment in non-lesional skin across human lupus and three murine models: MRL/lpr, B6.Sle1yaa, and imiquimod (IMQ) mice. IFN-I inhibits LC ADAM17 sheddase activity in murine and human LCs, and IFNAR blockade in lupus model mice restores LC ADAM17 sheddase activity, all without consistent effects on LC ADAM17 protein expression or LC numbers. Anti-IFNAR-mediated LC ADAM17 sheddase function restoration is associated with reduced photosensitive responses that are dependent on EGFR signaling and LC ADAM17. Reactive oxygen species (ROS) is a known mediator of ADAM17 activity; we show that UVR-induced LC ROS production is reduced in lupus model mice, restored by anti-IFNAR, and is cytoplasmic in origin. Our findings suggest that IFN-I promotes photosensitivity at least in part by inhibiting UVR-induced LC ADAM17 sheddase function and raise the possibility that anifrolumab ameliorates lupus skin disease in part by restoring this function. This work provides insight into IFN-I-mediated disease mechanisms, LC regulation, and a potential mechanism of action for anifrolumab in lupus.


Subject(s)
ADAM17 Protein , Langerhans Cells , Lupus Erythematosus, Systemic , Skin , ADAM17 Protein/metabolism , ADAM17 Protein/genetics , Animals , Humans , Langerhans Cells/metabolism , Mice , Skin/metabolism , Skin/pathology , Skin/radiation effects , Lupus Erythematosus, Systemic/metabolism , Ultraviolet Rays/adverse effects , Female , Disease Models, Animal , Photosensitivity Disorders/metabolism , Interferons/metabolism , Mice, Inbred MRL lpr
18.
Int J Mol Sci ; 25(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38891862

ABSTRACT

RNA processing is a highly conserved mechanism that serves as a pivotal regulator of gene expression. Alternative processing generates transcripts that can still be translated but lead to potentially nonfunctional proteins. A plethora of respiratory viruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), strategically manipulate the host's RNA processing machinery to circumvent antiviral responses. We integrated publicly available omics datasets to systematically analyze isoform-level expression and delineate the nascent peptide landscape of SARS-CoV-2-infected human cells. Our findings explore a suggested but uncharacterized mechanism, whereby SARS-CoV-2 infection induces the predominant expression of unproductive splicing isoforms in key IFN signaling, interferon-stimulated (ISGs), class I MHC, and splicing machinery genes, including IRF7, HLA-B, and HNRNPH1. In stark contrast, cytokine and chemokine genes, such as IL6 and TNF, predominantly express productive (protein-coding) splicing isoforms in response to SARS-CoV-2 infection. We postulate that SARS-CoV-2 employs an unreported tactic of exploiting the host splicing machinery to bolster viral replication and subvert the immune response by selectively upregulating unproductive splicing isoforms from antigen presentation and antiviral response genes. Our study sheds new light on the molecular interplay between SARS-CoV-2 and the host immune system, offering a foundation for the development of novel therapeutic strategies to combat COVID-19.


Subject(s)
Alternative Splicing , COVID-19 , Interferons , Protein Isoforms , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , COVID-19/virology , COVID-19/genetics , COVID-19/immunology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Interferons/metabolism , Interferons/genetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism
19.
J Virol ; 98(7): e0068624, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38888343

ABSTRACT

Nervous necrosis virus (NNV), an aquatic RNA virus belonging to Betanodavirus, infects a variety of marine and freshwater fishes, leading to massive mortality of cultured larvae and juveniles and substantial economic losses. The enzyme cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) is widely recognized as a central component in the innate immune response to cytosolic DNA derived from different pathogens. However, little is known about the response of cGAS to aquatic RNA viruses. This study found that Epinephelus coioides cGAS (EccGAS) overexpression inhibited NNV replication, whereas EccGAS silencing promoted NNV replication. The anti-NNV activity of EccGAS was involved in interferon (IFN) signaling activation including tumor necrosis factor receptor-associated factor family member-associated NF-kappa-B activator-binding kinase 1 (TBK1) phosphorylation, interferon regulatory factor 3 (IRF3) nuclear translocation, and the subsequent induction of IFNc and ISGs. Interestingly, NNV employed its capsid protein (CP) or Protein A (ProA) to negatively or positively modulate EccGAS-mediated IFN signaling by simultaneously targeting EccGAS. CP interacted with EccGAS via the arm-P, S-P, and SD structural domains and promoted its polyubiquitination with K48 and K63 linkages in an EcUBE3C (the ubiquitin ligase)-dependent manner, ultimately leading to EccGAS degradation. Conversely, ProA bound to EccGAS and inhibited its ubiquitination and degradation. In regulating EccGAS protein content, CP's inhibitory action was more pronounced than ProA's protective effect, allowing successful NNV replication. These novel findings suggest that NNV CP and ProA dynamically modulate the EccGAS-mediated IFN signaling pathway to facilitate the immune escape of NNV. Our findings shed light on a novel mechanism of virus-host interaction and provide a theoretical basis for the prevention and control of NNV.IMPORTANCEAs a well-known DNA sensor, cGAS is a pivotal component in innate anti-viral immunity to anti-DNA viruses. Although there is growing evidence regarding the function of cGAS in the resistance to RNA viruses, the mechanisms by which cGAS participates in RNA virus-induced immune responses in fish and how aquatic viruses evade cGAS-mediated immune surveillance remain elusive. Here, we investigated the detailed mechanism by which EccGAS positively regulates the anti-NNV response. Furthermore, NNV CP and ProA interacted with EccGAS, regulating its protein levels through ubiquitin-proteasome pathways, to dynamically modulate the EccGAS-mediated IFN signaling pathway and facilitate viral evasion. Notably, NNV CP was identified to promote the ubiquitination of EccGAS via ubiquitin ligase EcUBE3C. These findings unveil a novel strategy for aquatic RNA viruses to evade cGAS-mediated innate immunity, enhancing our understanding of virus-host interactions.


Subject(s)
Capsid Proteins , Fish Diseases , Immune Evasion , Immunity, Innate , Nodaviridae , Nucleotidyltransferases , RNA Virus Infections , Signal Transduction , Virus Replication , Animals , Fish Diseases/virology , Fish Diseases/immunology , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Capsid Proteins/metabolism , Capsid Proteins/immunology , RNA Virus Infections/immunology , RNA Virus Infections/metabolism , Interferons/metabolism , Interferons/immunology , Bass/immunology , Bass/virology , Bass/metabolism , Fish Proteins/metabolism , Fish Proteins/genetics , Fish Proteins/immunology
20.
Emerg Microbes Infect ; 13(1): 2372344, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38916407

ABSTRACT

The Orthopoxvirus (OPXV) genus of the Poxviridae includes human pathogens variola virus (VARV), monkeypox virus (MPXV), vaccinia virus (VACV), and a number of zoonotic viruses. A number of Bcl-2-like proteins of VACV are involved in escaping the host innate immunity. However, little work has been devoted to the evolution and function of their orthologues in other OPXVs. Here, we found that MPXV protein P2, encoded by the P2L gene, and P2 orthologues from other OPXVs, such as VACV protein N2, localize to the nucleus and antagonize interferon (IFN) production. Exceptions to this were the truncated P2 orthologues in camelpox virus (CMLV) and taterapox virus (TATV) that lacked the nuclear localization signal (NLS). Mechanistically, the NLS of MPXV P2 interacted with karyopherin α-2 (KPNA2) to facilitate P2 nuclear translocation, and competitively inhibited KPNA2-mediated IRF3 nuclear translocation and downstream IFN production. Deletion of the NLS in P2 or orthologues significantly enhanced IRF3 nuclear translocation and innate immune responses, thereby reducing viral replication. Moreover, deletion of NLS from N2 in VACV attenuated viral replication and virulence in mice. These data demonstrate that the NLS-mediated translocation of P2 is critical for P2-induced inhibition of innate immunity. Our findings contribute to an in-depth understanding of the mechanisms of OPXV P2 orthologue in innate immune evasion.


Subject(s)
Immunity, Innate , Interferon Regulatory Factor-3 , Monkeypox virus , Nuclear Localization Signals , Viral Proteins , Animals , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Mice , Humans , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Proteins/immunology , Nuclear Localization Signals/genetics , Monkeypox virus/genetics , Monkeypox virus/immunology , HEK293 Cells , alpha Karyopherins/genetics , alpha Karyopherins/metabolism , Immune Evasion , Cell Nucleus/metabolism , Interferons/genetics , Interferons/immunology , Interferons/metabolism , Poxviridae Infections/immunology , Poxviridae Infections/virology , Poxviridae Infections/veterinary , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL