Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.932
Filter
1.
Mol Cell ; 84(15): 2870-2881.e5, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39013473

ABSTRACT

The human silencing hub (HUSH) preserves genome integrity through the epigenetic repression of invasive genetic elements. However, despite our understanding of HUSH as an obligate complex of three subunits, only loss of MPP8 or Periphilin, but not TASOR, triggers interferon signaling following derepression of endogenous retroelements. Here, we resolve this paradox by characterizing a second HUSH complex that shares MPP8 and Periphilin but assembles around TASOR2, an uncharacterized paralog of TASOR. Whereas HUSH represses LINE-1 retroelements marked by the repressive histone modification H3K9me3, HUSH2 is recruited by the transcription factor IRF2 to repress interferon-stimulated genes. Mechanistically, HUSH-mediated retroelement silencing sequesters the limited pool of the shared subunits MPP8 and Periphilin, preventing TASOR2 from forming HUSH2 complexes and hence relieving the HUSH2-mediated repression of interferon-stimulated genes. Thus, competition between two HUSH complexes intertwines retroelement silencing with the induction of an immune response, coupling epigenetic and immune aspects of genome defense.


Subject(s)
Gene Silencing , Humans , HEK293 Cells , Histones/metabolism , Histones/genetics , Retroelements/genetics , Epigenesis, Genetic , Long Interspersed Nucleotide Elements/genetics , Signal Transduction , Interferons/metabolism , Interferons/immunology , Interferons/genetics , HeLa Cells
2.
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
3.
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
4.
Zool Res ; 45(5): 972-982, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39085753

ABSTRACT

Type IV interferon (IFN-υ) is a recently discovered cytokine crucial for host defense against viral infections. However, the role and mechanisms of IFN-υ in bacterial infections remain unexplored. This study investigated the antibacterial and antiviral functions and mechanisms of grass carp ( Ctenopharyngodon idella) IFN-υ (CiIFN-υ) both in vivo and in vitro. The CiIFN-υ gene was first identified and characterized in grass carp. Subsequently, the immune expression of CiIFN-υ significantly increased following bacterial challenge, indicating its response to bacterial infections. The eukaryotic recombinant expression plasmid of CiIFN-υ was then constructed and transfected into fathead minnow (FHM) cells. Supernatants were collected and incubated with four bacterial strains, followed by plate spreading and colony counting. Results indicated that CiIFN-υ exhibited more potent antibacterial activity against gram-negative bacteria compared to gram-positive bacteria and aggregated gram-negative bacteria but not gram-positive bacteria. In vivo experiments further confirmed the antibacterial function, showing high survival rates, low tissue edema and damage, reduced tissue bacterial load, and elevated proinflammatory response at the early stages of bacterial infection. In addition, the antiviral function of CiIFN-υ was confirmed through in vitro and in vivo experiments, including crystal violet staining, survival rates, tissue viral burden, and RT-qPCR. This study highlights the antibacterial function and preliminary mechanism of IFN-υ, demonstrating that IFN-υ possesses dual functions against bacterial and viral infections.


Subject(s)
Carps , Fish Diseases , Animals , Carps/immunology , Fish Diseases/immunology , Fish Diseases/virology , Antiviral Agents/pharmacology , Gene Expression Regulation/drug effects , Interferons/metabolism , Interferons/genetics , Anti-Bacterial Agents/pharmacology , Bacterial Infections/veterinary , Bacterial Infections/immunology , Fish Proteins/genetics , Fish Proteins/metabolism , Amino Acid Sequence , Phylogeny
5.
Fish Shellfish Immunol ; 152: 109781, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39029718

ABSTRACT

IkappaB kinase beta (IKKß) is a key member of IκB kinases and functions importantly in interferon (IFN) signaling. Phosphorylation and ubiquitination are involved in the activation of IKKß. A20 is a de-ubiquitin enzyme and functions as a suppressor in inflammation signaling, which has been reported to be phosphorylated and activated by IKKß. However, the role and relationship of IKKß and A20 in teleost remains unclear. In this study, IKKß (bcIKKß) and A20 (bcA20) of black carp (Mylopharyngodon piceus) have been cloned and characterized. Overexpressed bcIKKß in EPC cells showed strong anti-viral ability by activating both NF-κB and IFN signaling. EPC cells stable expressing bcIKKß presented improved anti-viral activity as well. The interaction between bcA20 and bcIKKß was identified, and overexpression of bcA20 was able to suppress bcIKKß-mediated activation of NF-κB and IFN signaling. Meanwhile, knock-down of A20 increased host the antiviral ability of host cells. Importantly, it has been identified that bcA20 was able to remove K27-linked ubiquitination and decrease the phosphorylation of bcIKKß. Thus, our data conclude that bcA20 suppresses the anti-viral activity of bcIKKß and removes its K27-linked ubiquitination, which presents a new mechanism of IKKß regulation.


Subject(s)
Carps , Fish Proteins , I-kappa B Kinase , Signal Transduction , Ubiquitination , Animals , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , I-kappa B Kinase/metabolism , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/chemistry , Carps/immunology , Carps/genetics , Signal Transduction/immunology , Interferons/genetics , Interferons/immunology , Interferons/metabolism , Fish Diseases/immunology , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/immunology , Immunity, Innate/genetics , Gene Expression Regulation/immunology , Sequence Alignment/veterinary , Phylogeny , Gene Expression Profiling/veterinary , Amino Acid Sequence
6.
Viruses ; 16(7)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-39066259

ABSTRACT

Viruses often pose a significant threat to the host through the exploitation of cellular machineries for their own benefit. In the context of immune responses, myriad host factors are deployed to target viral RNAs and inhibit viral protein translation, ultimately hampering viral replication. Understanding how "non-self" RNAs interact with the host translation machinery and trigger immune responses would help in the development of treatment strategies for viral infections. In this review, we explore how interferon-stimulated gene products interact with viral RNA and the translation machinery in order to induce either global or targeted translation inhibition.


Subject(s)
Interferons , Protein Biosynthesis , RNA, Viral , Virus Diseases , Animals , Humans , Host-Pathogen Interactions , Interferons/immunology , Interferons/metabolism , Interferons/genetics , RNA, Viral/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Diseases/immunology , Virus Diseases/virology , Virus Diseases/genetics , Virus Replication , Viruses/immunology , Viruses/genetics , Viruses/drug effects
7.
J Exp Med ; 221(9)2024 09 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
8.
Fish Shellfish Immunol ; 151: 109718, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38909635

ABSTRACT

Receptors of type I interferon (IFNR) play a vital role in the antiviral immune response. However, little is known about the negative regulatory role of the IFNR. Nervous necrosis virus (NNV) is one of the most significant viruses in cultured fish, resulting in great economic losses for the aquaculture industry. In this study, two orange-spotted grouper (Epinephelus coioides) cytokine receptor family B (CRFB) members, EcCRFB3 and EcCRFB4 were cloned and characterized from NNV infected grouper brain (GB) cells. The open reading frame (ORF) of EcCRFB3 consists of 852 bp encoding 283 amino acids, while EcCRFB4 has an ORF of 990 bp encoding 329 amino acids. The mRNA levels of EcCRFB3 or EcCRFB4 were significantly upregulated after NNV infection and the stimulation of poly (I:C) or NNV-encoded Protein A. In addition, EcCRFB3 or EcCRFB4 overexpression facilitated NNV replication, whereas EcCRFB3 or EcCRFB4 silencing resisted NNV replication. Overexpressed EcCRFB3 or EcCRFB4 inhibited the expression of IFN-I-induced ISGs. Taken together, our research provides the first evidence in fish demonstrating the role of IFNRs to regulate the IFN signaling pathway negatively. Our findings enrich the understanding of the functions of IFNRs and reveal a novel escape mechanism of NNV.


Subject(s)
Amino Acid Sequence , Bass , Fish Diseases , Fish Proteins , Gene Expression Regulation , Immunity, Innate , Nodaviridae , RNA Virus Infections , Virus Replication , Animals , Nodaviridae/physiology , Fish Diseases/immunology , Fish Diseases/virology , RNA Virus Infections/immunology , RNA Virus Infections/veterinary , Fish Proteins/genetics , Fish Proteins/immunology , Bass/immunology , Bass/genetics , Immunity, Innate/genetics , Gene Expression Regulation/immunology , Phylogeny , Sequence Alignment/veterinary , Receptors, Cytokine/genetics , Receptors, Cytokine/immunology , Gene Expression Profiling/veterinary , Interferons/immunology , Interferons/genetics
9.
Virulence ; 15(1): 2359470, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38918890

ABSTRACT

Influenza A virus (IAV) is the leading cause of highly contagious respiratory infections, which poses a serious threat to public health. The non-structural protein 1 (NS1) is encoded by segment 8 of IAV genome and is expressed in high levels in host cells upon IAV infection. It is the determinant of virulence and has multiple functions by targeting type Ι interferon (IFN-I) and type III interferon (IFN-III) production, disrupting cell apoptosis and autophagy in IAV-infected cells, and regulating the host fitness of influenza viruses. This review will summarize the current research on the NS1 including the structure and related biological functions of the NS1 as well as the interaction between the NS1 and host cells. It is hoped that this will provide some scientific basis for the prevention and control of the influenza virus.


Subject(s)
Influenza A virus , Influenza, Human , Viral Nonstructural Proteins , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Humans , Influenza A virus/genetics , Influenza A virus/pathogenicity , Influenza A virus/physiology , Influenza, Human/virology , Animals , Autophagy , Virulence , Host-Pathogen Interactions , Apoptosis , Interferons/metabolism , Interferons/immunology , Interferons/genetics
10.
Fish Shellfish Immunol ; 151: 109684, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38852788

ABSTRACT

Singapore grouper iridovirus (SGIV) is a large double-stranded DNA virus that has caused significant economic losses to the grouper aquaculture industry. So far, the structure and function of SGIV proteins have been successively reported. In the present paper, the protein of SGIV VP146 was cloned and identified. VP146 was whole-cell distributed in GS cells. VP146 promoted SGIV replication and inhibited the transcription of interferon-related genes as well as pro-inflammatory cytokines in GS cells. In addition, VP146 was involved in the regulation of the cGAS-STING signaling pathway, and decreased cGAS-STING induced the promoter of ISRE and NF-κB. VP146 interacted with the proteins of cGAS, STING, TBK1, and IRF3 from grouper, but did not affect the binding of grouper STING to grouper TBK1 and grouper IRF3. Interestingly, grouper STING was able to affect the intracellular localization of VP146. Four segment structural domains of grouper STING were constructed, and grouper STING-CTT could affect the intracellular localization of VP146. VP146 had no effect on the self-binding of EcSITNG, nor on the binding of EcSTING to EcTBK1 and EcIRF3. Together, the results demonstrated that SGIV VP146 modulated the cGAS-STING signaling pathway to escape the interferon immune response.


Subject(s)
Adaptor Proteins, Signal Transducing , Bass , Iridovirus , Nucleotidyltransferases , Signal Transduction , Iridovirus/immunology , Nucleotidyltransferases/genetics , Nucleotidyltransferases/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Signal Transduction/immunology , Bass/genetics , Bass/immunology , Bass/virology , Cell Line , Spleen/cytology , Gene Expression Regulation/immunology , Virus Replication/immunology , Interferons/genetics , Interferons/immunology , Fish Proteins/immunology , Animals
11.
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
12.
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
13.
Am J Hum Genet ; 111(7): 1352-1369, 2024 07 11.
Article in English | MEDLINE | ID: mdl-38866022

ABSTRACT

Primary proteasomopathies have recently emerged as a new class of rare early-onset neurodevelopmental disorders (NDDs) caused by pathogenic variants in the PSMB1, PSMC1, PSMC3, or PSMD12 proteasome genes. Proteasomes are large multi-subunit protein complexes that maintain cellular protein homeostasis by clearing ubiquitin-tagged damaged, misfolded, or unnecessary proteins. In this study, we have identified PSMD11 as an additional proteasome gene in which pathogenic variation is associated with an NDD-causing proteasomopathy. PSMD11 loss-of-function variants caused early-onset syndromic intellectual disability and neurodevelopmental delay with recurrent obesity in 10 unrelated children. Our findings demonstrate that the cognitive impairment observed in these individuals could be recapitulated in Drosophila melanogaster with depletion of the PMSD11 ortholog Rpn6, which compromised reversal learning. Our investigations in subject samples further revealed that PSMD11 loss of function resulted in impaired 26S proteasome assembly and the acquisition of a persistent type I interferon (IFN) gene signature, mediated by the integrated stress response (ISR) protein kinase R (PKR). In summary, these data identify PSMD11 as an additional member of the growing family of genes associated with neurodevelopmental proteasomopathies and provide insights into proteasomal biology in human health.


Subject(s)
Drosophila melanogaster , Intellectual Disability , Neurodevelopmental Disorders , Obesity , Proteasome Endopeptidase Complex , Adolescent , Animals , Child , Child, Preschool , Female , Humans , Male , Drosophila melanogaster/genetics , Intellectual Disability/genetics , Interferons/metabolism , Interferons/genetics , Loss of Function Mutation , Neurodevelopmental Disorders/genetics , Obesity/genetics , Phenotype , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism
14.
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
15.
mBio ; 15(7): e0113024, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38934617

ABSTRACT

Type III interferon signaling contributes to the pathogenesis of the important human pathogen Staphylococcus aureus in the airway. Little is known of the cellular factors important in this response. Using Ifnl2-green fluorescent protein reporter mice combined with flow cytometry and cellular depletion strategies, we demonstrate that the alveolar macrophage is the primary producer of interferon lambda (IFN-λ) in response to S. aureus in the airway. Bone marrow chimeras showed reduced bacterial burden in IFN-λ receptor (IFNLR1)-deficient recipient mice, indicative that non-hematopoietic cells were important for pathogenesis, in addition to significant reductions in pulmonary inflammation. These observations were confirmed through the use of an airway epithelial-specific IFNLR knockout mouse. Our data suggest that upon entry to the airway, S. aureus activates alveolar macrophages to produce type III IFN that is subsequently sensed by the airway epithelium. Future steps will determine how signaling from the epithelium then exerts its influence on bacterial clearance. These results highlight the important, yet sometimes detrimental, role of type III IFN signaling during infection and the impact the airway epithelium plays during host-pathogen interactions.IMPORTANCEThe contribution of type III interferon signaling to the control of bacterial infections is largely unknown. We have previously demonstrated that it contributes to the pathogenesis of acute Staphylococcus aureus respiratory infection. In this report, we document the importance of two cell types that underpin this pathogenesis. We demonstrate that the alveolar macrophage is the cell that is responsible for the production of type III interferon and that this molecule is sensed by airway epithelial cells, which impacts both bacterial clearance and induction of inflammation. This work sheds light on the first two aspects of this important pathogenic cascade.


Subject(s)
Interferons , Macrophages, Alveolar , Mice, Knockout , Staphylococcal Infections , Staphylococcus aureus , Animals , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/genetics , Mice , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/immunology , Staphylococcal Infections/microbiology , Interferons/metabolism , Interferons/genetics , Interferons/immunology , Mice, Inbred C57BL , Host-Pathogen Interactions , Signal Transduction , Respiratory Mucosa/microbiology , Interferon Lambda , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Virulence
16.
JCI Insight ; 9(14)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38885315

ABSTRACT

IFN-signaling gene (ISG) expression scores are potential markers of inflammation with significance from cancer to genetic syndromes. In Aicardi Goutières Syndrome (AGS), a disorder of abnormal DNA and RNA metabolism, this score has potential as a diagnostic biomarker, although the approach to ISG calculation has not been standardized or validated. To optimize ISG calculation and validate ISG as a diagnostic biomarker, mRNA levels of 36 type I IFN response genes were quantified from 997 samples (including 334 AGS), and samples were randomized into training and test data sets. An independent validation cohort (n = 122) was also collected. ISGs were calculated using all potential combinations up to 6 genes. A 4-gene approach (IFI44L, IFI27, USP18, IFI6) was the best-performing model (AUC of 0.8872 [training data set], 0.9245 [test data set]). The majority of top-performing gene combinations included IFI44L. Performance of IFI44L alone was 0.8762 (training data set) and 0.9580 (test data set) by AUC. The top approaches were able to discriminate individuals with genetic interferonopathy from control samples. This study validates the context of use for the ISG score as a diagnostic biomarker and underscores the importance of IFI44L in diagnosis of genetic interferonopathies.


Subject(s)
Autoimmune Diseases of the Nervous System , Biomarkers , Nervous System Malformations , Signal Transduction , Humans , Biomarkers/metabolism , Male , Female , Nervous System Malformations/genetics , Nervous System Malformations/diagnosis , Signal Transduction/genetics , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/diagnosis , Child , Interferons/genetics , Interferons/metabolism , Ubiquitin Thiolesterase/genetics , Child, Preschool , Interferon Type I/genetics , Interferon Type I/metabolism , Membrane Proteins/genetics , Adult , Adolescent , RNA, Messenger/metabolism , RNA, Messenger/genetics , Tumor Suppressor Proteins
17.
Mol Cell ; 84(13): 2423-2435.e5, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38917796

ABSTRACT

The innate immune cGAS-STING pathway is activated by cytosolic double-stranded DNA (dsDNA), a ubiquitous danger signal, to produce interferon, a potent anti-viral and anti-cancer cytokine. However, STING activation must be tightly controlled because aberrant interferon production leads to debilitating interferonopathies. Here, we discover PELI2 as a crucial negative regulator of STING. Mechanistically, PELI2 inhibits the transcription factor IRF3 by binding to phosphorylated Thr354 and Thr356 on the C-terminal tail of STING, leading to ubiquitination and inhibition of the kinase TBK1. PELI2 sets a threshold for STING activation that tolerates low levels of cytosolic dsDNA, such as that caused by silenced TREX1, RNASEH2B, BRCA1, or SETX. When this threshold is reached, such as during viral infection, STING-induced interferon production temporarily downregulates PELI2, creating a positive feedback loop allowing a robust immune response. Lupus patients have insufficient PELI2 levels and high basal interferon production, suggesting that PELI2 dysregulation may drive the onset of lupus and other interferonopathies.


Subject(s)
Interferon Regulatory Factor-3 , Membrane Proteins , Protein Serine-Threonine Kinases , Signal Transduction , Ubiquitination , Humans , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Phosphorylation , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Animals , HEK293 Cells , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/virology , Immunity, Innate , Host-Pathogen Interactions , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Mice , Interferons/metabolism , Interferons/immunology , Interferons/genetics , Feedback, Physiological , Mice, Inbred C57BL , Exodeoxyribonucleases , Phosphoproteins
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL