Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.984
Filter
1.
Mol Med Rep ; 30(3)2024 Sep.
Article in English | MEDLINE | ID: mdl-38963029

ABSTRACT

Viral infections in the respiratory tract are common, and, in recent years, severe acute respiratory syndrome coronavirus 2 outbreaks have highlighted the effect of viral infections on antiviral innate immune and inflammatory reactions. Specific treatments for numerous viral respiratory infections have not yet been established and they are mainly treated symptomatically. Therefore, understanding the details of the innate immune system underlying the airway epithelium is crucial for the development of new therapies. The present study aimed to investigate the function and expression of interferon (IFN)­stimulated gene (ISG)60 in non­cancerous bronchial epithelial BEAS­2B cells exposed to a Toll­like receptor 3 agonist. BEAS­2B cells were treated with a synthetic TLR3 ligand, polyinosinic­polycytidylic acid (poly IC). The mRNA and protein expression levels of ISG60 were analyzed using reverse transcription­quantitative PCR and western blotting, respectively. The levels of C­X­C motif chemokine ligand 10 (CXCL10) were examined using an enzyme­linked immunosorbent assay, and the effects of knockdown of IFN­ß, ISG60 and ISG56 were examined using specific small interfering RNAs. Notably, ISG60 expression was increased in proportion to poly IC concentration, and recombinant human IFN­ß also induced ISG60 expression. By contrast, knockdown of IFN­ß and ISG56 decreased ISG60 expression, and ISG60 knockdown reduced CXCL10 and ISG56 expression. These findings suggested that ISG60 is partly implicated in CXCL10 expression and that ISG60 may serve a role in the innate immune response of bronchial epithelial cells. The present study highlights ISG60 as a potential target for new therapeutic strategies against viral infections in the airway.


Subject(s)
Bronchi , Chemokine CXCL10 , Epithelial Cells , Poly I-C , Signal Transduction , Toll-Like Receptor 3 , Humans , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 3/genetics , Chemokine CXCL10/metabolism , Chemokine CXCL10/genetics , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Bronchi/cytology , Bronchi/metabolism , Poly I-C/pharmacology , Signal Transduction/drug effects , Cell Line , Immunity, Innate , Interferon-beta/metabolism , Interferon-beta/genetics , Gene Expression Regulation/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , RNA-Binding Proteins , Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins
2.
Viruses ; 16(6)2024 May 30.
Article in English | MEDLINE | ID: mdl-38932169

ABSTRACT

Repression of human cytomegalovirus (HCMV) immediate-early (IE) gene expression is a key regulatory step in the establishment and maintenance of latent reservoirs. Viral IE transcription and protein accumulation can be elevated during latency by treatment with histone deacetylase inhibitors such as valproic acid (VPA), rendering infected cells visible to adaptive immune responses. However, the latency-associated viral protein UL138 inhibits the ability of VPA to enhance IE gene expression during infection of incompletely differentiated myeloid cells that support latency. UL138 also limits the accumulation of IFNß transcripts by inhibiting the cGAS-STING-TBK1 DNA-sensing pathway. Here, we show that, in the absence of UL138, the cGAS-STING-TBK1 pathway promotes both IFNß accumulation and VPA-responsive IE gene expression in incompletely differentiated myeloid cells. Inactivation of this pathway by either genetic or pharmacological inhibition phenocopied UL138 expression and reduced VPA-responsive IE transcript and protein accumulation. This work reveals a link between cytoplasmic pathogen sensing and epigenetic control of viral lytic phase transcription and suggests that manipulation of pattern recognition receptor signaling pathways could aid in the refinement of MIEP regulatory strategies to target latent viral reservoirs.


Subject(s)
Cytomegalovirus , Membrane Proteins , Myeloid Cells , Nucleotidyltransferases , Protein Serine-Threonine Kinases , Signal Transduction , Valproic Acid , Humans , Valproic Acid/pharmacology , Myeloid Cells/virology , Myeloid Cells/metabolism , Myeloid Cells/drug effects , Signal Transduction/drug effects , Membrane Proteins/metabolism , Membrane Proteins/genetics , Cytomegalovirus/physiology , Cytomegalovirus/drug effects , Cytomegalovirus/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/genetics , Virus Latency/drug effects , Transcription, Genetic/drug effects , Cell Differentiation/drug effects , Gene Expression Regulation, Viral/drug effects , Genes, Immediate-Early , Interferon-beta/metabolism , Interferon-beta/genetics
3.
Nat Commun ; 15(1): 5423, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926338

ABSTRACT

Oncogene-induced senescence (OIS) arrests cell proliferation in response to replication stress (RS) induced by oncogenes. OIS depends on the DNA damage response (DDR), but also on the cGAS-STING pathway, which detects cytosolic DNA and induces type I interferons (IFNs). Whether and how RS and IFN responses cooperate to promote OIS remains unknown. Here, we show that the induction of OIS by the H-RASV12 oncogene in immortalized human fibroblasts depends on the MRE11 nuclease. Indeed, treatment with the MRE11 inhibitor Mirin prevented RS, micronuclei formation and IFN response induced by RASV12. Overexpression of the cytosolic nuclease TREX1 also prevented OIS. Conversely, overexpression of a dominant negative mutant of TREX1 or treatment with IFN-ß was sufficient to induce RS and DNA damage, independent of RASV12 induction. These data suggest that the IFN response acts as a positive feedback loop to amplify DDR in OIS through a process regulated by MRE11 and TREX1.


Subject(s)
Cellular Senescence , DNA Damage , DNA Replication , Exodeoxyribonucleases , MRE11 Homologue Protein , Phosphoproteins , Signal Transduction , Humans , Exodeoxyribonucleases/metabolism , Exodeoxyribonucleases/genetics , Phosphoproteins/metabolism , Phosphoproteins/genetics , MRE11 Homologue Protein/metabolism , MRE11 Homologue Protein/genetics , Cellular Senescence/genetics , Fibroblasts/metabolism , Interferon-beta/metabolism , Interferon-beta/genetics
4.
Virol Sin ; 39(3): 501-512, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38789039

ABSTRACT

The infection caused by porcine epidemic diarrhea virus (PEDV) is associated with high mortality in piglets worldwide. Host factors involved in the efficient replication of PEDV, however, remain largely unknown. Our recent proteomic study in the virus-host interaction network revealed a significant increase in the accumulation of CALML5 (EF-hand protein calmodulin-like 5) following PEDV infection. A further study unveiled a biphasic increase of CALML5 in 2 and 12 â€‹h after viral infection. Similar trends were observed in the intestines of piglets in the early and late stages of the PEDV challenge. Moreover, CALML5 depletion reduced PEDV mRNA and protein levels, leading to a one-order-of-magnitude decrease in virus titer. At the early stage of PEDV infection, CALML5 affected the endosomal trafficking pathway by regulating the expression of endosomal sorting complex related cellular proteins. CALML5 depletion also suppressed IFN-ß and IL-6 production in the PEDV-infected cells, thereby indicating its involvement in negatively regulating the innate immune response. Our study reveals the biological function of CALML5 in the virology field and offers new insights into the PEDV-host cell interaction.


Subject(s)
Calmodulin , Endosomes , Immunity, Innate , Porcine epidemic diarrhea virus , Virus Replication , Animals , Porcine epidemic diarrhea virus/immunology , Porcine epidemic diarrhea virus/physiology , Swine , Calmodulin/metabolism , Calmodulin/genetics , Endosomes/metabolism , Endosomes/virology , Host-Pathogen Interactions/immunology , Swine Diseases/virology , Swine Diseases/immunology , Vero Cells , Chlorocebus aethiops , Coronavirus Infections/immunology , Coronavirus Infections/virology , Coronavirus Infections/veterinary , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-6/immunology , Interferon-beta/genetics , Interferon-beta/immunology , Interferon-beta/metabolism
5.
Cancer Lett ; 596: 216988, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38797234

ABSTRACT

Type I interferons exhibit anti-proliferative and anti-cancer activities, but their detailed regulatory mechanisms in cancer have not been fully elucidated yet. RNA binding proteins are master orchestrators of gene regulation, which are closely related to tumor progression. Here we show that the upregulated RNA binding protein RBM45 correlates with poor prognosis in breast cancer. Depletion of RBM45 suppresses breast cancer progression both in cultured cells and xenograft mouse models. Mechanistically, RBM45 ablation inhibits breast cancer progression through regulating type I interferon signaling, particularly by elevating IFN-ß production. Importantly, RBM45 recruits TRIM28 to IRF7 and stimulates its SUMOylation, thereby repressing IFNB1 transcription. Loss of RBM45 reduced the SUMOylation of IRF7 by reducing the interaction between TRIM28 and IRF7 to promote IFNB1 transcription, leading to the inhibition of breast cancer progression. Taken together, our finding uncovers a vital role of RBM45 in modulating type I interferon signaling and cancer aggressive progression, implicating RBM45 as a potential therapeutic target in breast cancer.


Subject(s)
Breast Neoplasms , Disease Progression , Gene Expression Regulation, Neoplastic , Interferon Regulatory Factor-7 , RNA-Binding Proteins , Sumoylation , Humans , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Female , Animals , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Mice , Transcription, Genetic , Cell Line, Tumor , Interferon-beta/metabolism , Interferon-beta/genetics , Signal Transduction , Mice, Nude , Cell Proliferation , Tripartite Motif-Containing Protein 28/metabolism , Tripartite Motif-Containing Protein 28/genetics , Xenograft Model Antitumor Assays , Mice, Inbred BALB C
6.
PLoS Pathog ; 20(5): e1012125, 2024 May.
Article in English | MEDLINE | ID: mdl-38696536

ABSTRACT

Major 5'-terminally deleted (5'TD) RNA forms of group-B coxsackievirus (CVB-5'TD) has been associated with myocarditis in both mice and humans. Although it is known that interferon-ß (IFN-ß) signaling is critical for an efficient innate immune response against CVB-induced myocarditis, the link between CVB-5'TD RNA forms and type I IFN signaling in cardiomyocytes remains to be explored. In a mouse model of CVB3/28-induced myocarditis, major early-emerging forms of CVB-5'TD RNA have been characterized as replicative viral populations that impair IFN-ß production in the heart. Synthetic CVB3/28 RNA forms mimicking each of these major 5'TD virus populations were transfected in mice and have been shown to modulate innate immune responses in the heart and to induce myocarditis in mice. Remarkably, transfection of synthetic viral RNA with deletions in the secondary structures of the 5'-terminal CVB3 RNA domain I, modifying stem-loops "b", "c" or "d", were found to impair IFN-ß production in human cardiomyocytes. In addition, the activation of innate immune response by Poly(I:C), was found to restore IFN-ß production and to reduce the burden of CVB-5'TD RNA-forms in cardiac tissues, thereby reducing the mortality rate of infected mice. Overall, our results indicate that major early-emerging CVB3 populations deleted in the domain I of genomic RNA, in the 5' noncoding region, modulate the activation of the type I IFN pathway in cardiomyocytes and induce myocarditis in mice. These findings shed new light on the role of replicative CVB-5'TD RNA forms as key pathophysiological factors in CVB-induced human myocarditis.


Subject(s)
Coxsackievirus Infections , Enterovirus B, Human , Interferon Type I , Myocarditis , Myocytes, Cardiac , RNA, Viral , Myocarditis/virology , Myocarditis/immunology , Myocarditis/genetics , Animals , Myocytes, Cardiac/virology , Myocytes, Cardiac/metabolism , Mice , Enterovirus B, Human/immunology , Coxsackievirus Infections/immunology , Coxsackievirus Infections/virology , Coxsackievirus Infections/genetics , Interferon Type I/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Humans , Immunity, Innate , Signal Transduction , Interferon-beta/metabolism , Interferon-beta/genetics , Interferon-beta/immunology , Male , 5' Untranslated Regions
7.
Front Immunol ; 15: 1380220, 2024.
Article in English | MEDLINE | ID: mdl-38799458

ABSTRACT

African swine fever (ASF) is an acute hemorrhagic and devastating infectious disease affecting domestic pigs and wild boars. It is caused by the African swine fever virus (ASFV), which is characterized by genetic diversity and sophisticated immune evasion strategies. To facilitate infection, ASFV encodes multiple proteins to antagonize host innate immune responses, thereby contributing to viral virulence and pathogenicity. The molecular mechanisms employed by ASFV-encoded proteins to modulate host antiviral responses have not been comprehensively elucidated. In this study, it was observed that the ASFV MGF505-6R protein, a member of the multigene family 505 (MGF505), effectively suppressed the activation of the interferon-beta (IFN-ß) promoter, leading to reduced mRNA levels of antiviral genes. Additional evidence has revealed that MGF505-6R antagonizes the cGAS-STING signaling pathway by interacting with the stimulator of interferon genes (STING) for degradation in the autophagy-lysosomal pathway. The domain mapping revealed that the N-terminal region (1-260aa) of MGF505-6R is the primary domain responsible for interacting with STING, while the CTT domain of STING is crucial for its interaction with MGF505-6R. Furthermore, MGF505-6R also inhibits the activation of STING by reducing the K63-linked polyubiquitination of STING, leading to the disruption of STING oligomerization and TANK binding kinase 1 (TBK1) recruitment, thereby impairing the phosphorylation and nuclear translocation of interferon regulatory factor 3 (IRF3). Collectively, our study elucidates a novel strategy developed by ASFV MGF505-6R to counteract host innate immune responses. This discovery may offer valuable insights for further exploration of ASFV immune evasion mechanisms and antiviral strategies.


Subject(s)
African Swine Fever Virus , African Swine Fever , Membrane Proteins , Viral Proteins , Animals , African Swine Fever Virus/immunology , African Swine Fever Virus/genetics , Swine , Membrane Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/immunology , African Swine Fever/immunology , African Swine Fever/virology , African Swine Fever/metabolism , Viral Proteins/immunology , Viral Proteins/metabolism , Viral Proteins/genetics , Humans , Immunity, Innate , Interferon Type I/metabolism , Interferon Type I/immunology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/immunology , Signal Transduction , Proteolysis , HEK293 Cells , Host-Pathogen Interactions/immunology , Immune Evasion , Interferon-beta/metabolism , Interferon-beta/immunology , Interferon-beta/genetics
8.
Exp Mol Pathol ; 137: 104897, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38691979

ABSTRACT

BACKGROUND: Signaling by toll-like receptors (TLRs) initiates important immune responses against viral infection. The role of TLRs in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is not well elucidated. Thus, we investigated the interaction of TLRs agonists and SARS-COV-2 antigens with immune cells in vitro. MATERIAL & METHODS: 30 coronavirus disease 2019 (COVID-19) patients (15 severe and 15 moderate) and 10 age and sex-matched healthy control (HC) were enrolled. Peripheral blood mononuclear cells (PBMCs) were isolated and activated with TLR3, 7, 8, and 9 agonists, the spike protein (SP) of SARS-CoV-2, and the receptor binding domain (RBD) of SP. Frequencies of CD3+IFN-ß+ T cells, and CD3+IFN-γ+ T cells were evaluated by flow cytometry. Interferon (IFN)-ß gene expression was assessed by qRT-PCR. RESULTS: The frequency of CD3+IFN-ß+ T cells was higher in PBMCs from moderate (p < 0.0001) and severe (p = 0.009) patients at baseline in comparison with HCs. The highest increase in the frequency of CD3+IFN-ß+ T cells in cell from moderate patients was induced by TLR8 agonist and SP (p < 0.0001 for both) when compared to HC, while, the highest increase of the frequency of CD3+IFN-ß+ T cells in sample of severe patients was seen with TLR8 and TLR7 agonists (both p = 0.002). The frequency of CD3+IFN-γ+ T cells was significantly increased upon stimulation with TLR agonists in cell from patients with moderate and severe COVID-19, compared with HC (all p < 0.01), except with TLR7 and TLR8 agonists. The TLR8 agonist did not significantly increase the frequency of CD3+IFN-γ+ T cells in PBMCs of severe patients, but did so in cells from patients with moderate disease (p = 0.01). Moreover, IFN-ß gene expression was significantly upregulated in CD3+T cells from moderate (p < 0.0001) and severe (p = 0.002) COVID-19 patients, compared to HC after stimulation with the TLR8 agonist, while, stimulation of T cells with SP, significantly up-regulated IFN-ß mRNA expression in cells from patients with moderate (p = 0.0003), but not severe disease. CONCLUSION: Stimulation of PBMCs from COVID-19 patients, especially patients with moderate disease, with TLR8 agonist and SP increased the frequency of IFN-ß-producing T cells and IFN-ß gene expression.


Subject(s)
CD3 Complex , COVID-19 , SARS-CoV-2 , T-Lymphocytes , Toll-Like Receptors , Humans , COVID-19/immunology , COVID-19/virology , SARS-CoV-2/immunology , Male , Female , Middle Aged , Toll-Like Receptors/agonists , Toll-Like Receptors/genetics , CD3 Complex/immunology , CD3 Complex/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/drug effects , Adult , Interferon-gamma/metabolism , Interferon-gamma/genetics , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Interferon-beta/genetics , Interferon-beta/immunology , Aged , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/drug effects , Toll-Like Receptor Agonists
9.
J Transl Med ; 22(1): 463, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750559

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) have garnered significant interest for their tumor-tropic property, making them potential therapeutic delivery vehicles for cancer treatment. We have previously shown the significant anti-tumour activity in mice preclinical models and companion animals with naturally occurring cancers using non-virally engineered MSCs with a therapeutic transgene encoding cytosine deaminase and uracil phosphoribosyl transferase (CDUPRT) and green fluorescent protein (GFP). Clinical studies have shown improved response rate with combinatorial treatment of 5-fluorouracil and Interferon-beta (IFNb) in peritoneal carcinomatosis (PC). However, high systemic toxicities have limited the clinical use of such a regime. METHODS: In this study, we evaluated the feasibility of intraperitoneal administration of non-virally engineered MSCs to co-deliver CDUPRT/5-Flucytosine prodrug system and IFNb to potentially enhance the cGAS-STING signalling axis. Here, MSCs were engineered to express CDUPRT or CDUPRT-IFNb. Expression of CDUPRT and IFNb was confirmed by flow cytometry and ELISA, respectively. The anti-cancer efficacy of the engineered MSCs was evaluated in both in vitro and in vivo model. ES2, HT-29 and Colo-205 were cocultured with engineered MSCs at various ratio. The cell viability with or without 5-flucytosine was measured with MTS assay. To further compare the anti-cancer efficacy of the engineered MSCs, peritoneal carcinomatosis mouse model was established by intraperitoneal injection of luciferase expressing ES2 stable cells. The tumour burden was measured through bioluminescence tracking. RESULTS: Firstly, there was no changes in phenotypes of MSCs despite high expression of the transgene encoding CDUPRT and IFNb (CDUPRT-IFNb). Transwell migration assays and in-vivo tracking suggested the co-expression of multiple transgenes did not impact migratory capability of the MSCs. The superiority of CDUPRT-IFNb over CDUPRT expressing MSCs was demonstrated in ES2, HT-29 and Colo-205 in-vitro. Similar observations were observed in an intraperitoneal ES2 ovarian cancer xenograft model. The growth of tumor mass was inhibited by ~ 90% and 46% in the mice treated with MSCs expressing CDUPRT-IFNb or CDUPRT, respectively. CONCLUSIONS: Taken together, these results established the effectiveness of MSCs co-expressing CDUPRT and IFNb in controlling and targeting PC growth. This study lay the foundation for the development of clinical trial using multigene-armed MSCs for PC.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Pentosyltransferases , Peritoneal Neoplasms , Transgenes , Animals , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Peritoneal Neoplasms/therapy , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/pathology , Humans , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Cell Line, Tumor , Interferon-beta/metabolism , Interferon-beta/genetics , Xenograft Model Antitumor Assays , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , Mice , Female
10.
Virol J ; 21(1): 107, 2024 05 08.
Article in English | MEDLINE | ID: mdl-38720392

ABSTRACT

Natural immunity is the first defense line of the host immune system, which plays a significant role in combating foreign pathogenic microorganisms. The IFN-ß (interferon-beta) signaling pathway, being a typical example of innate immunity, plays a vital function. This study aimed to elucidate the function of pseudorabies virus (PRV) UL38 protein (unique long region 38) in suppressing the activation of the IFN-ß signaling pathway. The findings from our study indicate that the PRV UL38 protein effectively hampers the activation of IFN-ß by poly (dA: dT) (poly(deoxyadenylic-deoxythymidylic)) and 2'3'-cGAMP (2'-3'-cyclic GMP-AMP). Furthermore, UL38 exhibits spatial co-localization with STING (stimulator of interferon genes) and effectively hinders STING dimerization. Subsequently, STING was downgraded to suppress the production of IFN-ß and ISGs (interferon stimulated genes). Immunoprecipitation analysis revealed that the interaction between UL38 and STING, which subsequently initiated the degradation of STING via selective autophagy mediated by TOLLIP (toll interacting protein). To summarize, this research elucidates the function of UL38 in counteracting the cGAS (cGAMP synthase)-STING-induced IFN-ß pathway. The PRV UL38 protein may attenuate the activation of IFN-ß as a means of regulating the virus's persistence in the host.


Subject(s)
Autophagy , Herpesvirus 1, Suid , Interferon-beta , Membrane Proteins , Nucleotidyltransferases , Signal Transduction , Animals , Humans , Cell Line , HEK293 Cells , Herpesvirus 1, Suid/physiology , Herpesvirus 1, Suid/immunology , Host-Pathogen Interactions , Immunity, Innate , Interferon-beta/metabolism , Interferon-beta/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Pseudorabies/virology , Pseudorabies/metabolism , Pseudorabies/immunology , Viral Proteins/metabolism , Viral Proteins/genetics , Swine , Mesocricetus
11.
Virol J ; 21(1): 109, 2024 05 11.
Article in English | MEDLINE | ID: mdl-38734674

ABSTRACT

BACKGROUND: Syndrome coronavirus-2 (SARS-CoV-2) has developed various strategies to evade the antiviral impact of type I IFN. Non-structural proteins and auxiliary proteins have been extensively researched on their role in immune escape. Nevertheless, the detailed mechanisms of structural protein-induced immune evasion have not been well elucidated. METHODS: Human alveolar basal epithelial carcinoma cell line (A549) was stimulated with polyinosinic-polycytidylic acid (PIC) and independently transfected with four structural proteins expression plasmids, including nucleocapsid (N), spike (S), membrane (M) and envelope (E) proteins. By RT-qPCR and ELISA, the structural protein with the most pronounced inhibitory effects on IFN-ß induction was screened. RNA-sequencing (RNA-Seq) and two differential analysis strategies were used to obtain differentially expressed genes associated with N protein inhibition of IFN-ß induction. Based on DIANA-LncBase and StarBase databases, the interactive competitive endogenous RNA (ceRNA) network for N protein-associated genes was constructed. By combining single-cell sequencing data (GSE158055), lncRNA-miRNA-mRNA axis was further determined. Finally, RT-qPCR was utilized to illustrate the regulatory functions among components of the ceRNA axis. RESULTS: SARS-CoV-2 N protein inhibited IFN-ß induction in human alveolar epithelial cells most significantly compared with other structural proteins. RNA-Seq data analysis revealed genes related to N protein inhibiting IFNs induction. The obtained 858 differentially expressed genes formed the reliable ceRNA network. The function of LINC01002-miR-4324-FRMD8 axis in the IFN-dominated immune evasion was further demonstrated through integrating single-cell sequencing data. Moreover, we validated that N protein could reverse the effect of PIC on LINC01002, FRMD8 and miR-4324 expression, and subsequently on IFN-ß expression level. And LINC01002 could regulate the production of FRMD8 by inhibiting miR-4324. CONCLUSION: SARS-CoV-2 N protein suppressed the induction of IFN-ß by regulating LINC01002 which was as a ceRNA, sponging miR-4324 and participating in the regulation of FRMD8 mRNA. Our discovery provides new insights into early intervention therapy and drug development on SARS-CoV-2 infection.


Subject(s)
COVID-19 , MicroRNAs , RNA, Long Noncoding , SARS-CoV-2 , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , COVID-19/virology , COVID-19/immunology , SARS-CoV-2/genetics , A549 Cells , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Interferon-beta/genetics , Interferon-beta/metabolism , Immune Evasion , Coronavirus Nucleocapsid Proteins/genetics , Coronavirus Nucleocapsid Proteins/metabolism , RNA, Competitive Endogenous , Phosphoproteins
12.
J Clin Invest ; 134(9)2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38690737

ABSTRACT

Inflammation and pain are intertwined responses to injury, infection, or chronic diseases. While acute inflammation is essential in determining pain resolution and opioid analgesia, maladaptive processes occurring during resolution can lead to the transition to chronic pain. Here we found that inflammation activates the cytosolic DNA-sensing protein stimulator of IFN genes (STING) in dorsal root ganglion nociceptors. Neuronal activation of STING promotes signaling through TANK-binding kinase 1 (TBK1) and triggers an IFN-ß response that mediates pain resolution. Notably, we found that mice expressing a nociceptor-specific gain-of-function mutation in STING exhibited an IFN gene signature that reduced nociceptor excitability and inflammatory hyperalgesia through a KChIP1-Kv4.3 regulation. Our findings reveal a role of IFN-regulated genes and KChIP1 downstream of STING in the resolution of inflammatory pain.


Subject(s)
Membrane Proteins , Nociceptors , Animals , Mice , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nociceptors/metabolism , Ganglia, Spinal/metabolism , Interferon-beta/genetics , Interferon-beta/metabolism , Inflammation/genetics , Inflammation/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pain/metabolism , Pain/genetics , Signal Transduction , Male
13.
EMBO J ; 43(11): 2233-2263, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38658796

ABSTRACT

Type I interferons (IFN-I, including IFNß) and IFNγ produce overlapping, yet clearly distinct immunological activities. Recent data show that the distinctness of global transcriptional responses to the two IFN types is not apparent when comparing their immediate effects. By analyzing nascent transcripts induced by IFN-I or IFNγ over a period of 48 h, we now show that the distinctiveness of the transcriptomes emerges over time and is based on differential employment of the ISGF3 complex as well as of the second-tier transcription factor IRF1. The distinct transcriptional properties of ISGF3 and IRF1 correspond with a largely diverse nuclear protein interactome. Mechanistically, we describe the specific input of ISGF3 and IRF1 into enhancer activation and the regulation of chromatin accessibility at interferon-stimulated genes (ISG). We further report differences between the IFN types in altering RNA polymerase II pausing at ISG 5' ends. Our data provide insight how transcriptional regulators create immunological identities of IFN-I and IFNγ.


Subject(s)
Gene Expression Regulation , Interferon Regulatory Factor-1 , Interferon-beta , Interferon-gamma , Signal Transduction , Interferon-gamma/metabolism , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/genetics , Interferon-beta/metabolism , Interferon-beta/genetics , Humans , Interferon-Stimulated Gene Factor 3/metabolism , Interferon-Stimulated Gene Factor 3/genetics , Animals , Mice , RNA Polymerase II/metabolism , RNA Polymerase II/genetics
14.
J Virol ; 98(5): e0192523, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38624230

ABSTRACT

Recurrent respiratory papillomatosis (RRP) is a rare benign tumor caused mainly by the infection of the respiratory tract epithelial cells by the human papillomavirus (HPV) type 6/11. However, the specific mechanisms underlying the inhibition of the host's innate immune response by HPV remain unclear. For this purpose, we employed single-cell RNA sequencing to analyze the states of various immune cells in RRP samples post-HPV infection and utilized a cellular model of HPV infection to elucidate the mechanisms by which HPV evades the innate immune system in RRP. The results revealed distinct immune cell heterogeneity in RRP and demonstrated that HPV11 E7 can inhibit the phosphorylation of the stimulator of interferon genes protein, thereby circumventing the body's antiviral response. In vitro co-culture experiments demonstrated that stimulation of macrophages to produce interferon-beta induced the death of HPV-infected epithelial cells, also reducing HPV viral levels. In summary, our study preliminarily identifies the potential mechanisms by which HPV evades the host's antiviral immune response, as well as the latent antiviral functions exhibited by activated macrophages. This research serves as an initial exploration of antiviral immune evasion in RRP, laying a solid foundation for investigating immunotherapeutic approaches for the disease.IMPORTANCESurgical tumor reduction is the most common treatment for recurrent respiratory papillomatosis (RRP). One of the characteristics of RRP is its persistent recurrence, and multiple surgeries are usually required to control the symptoms. Recently, some adjuvant therapies have shown effectiveness, but none of them can completely clear human papillomavirus (HPV) infection, and thus, a localized antiviral immune response is significant for disease control; after all, HPV infection is limited to the epithelium. Inhibition of interferon-beta (IFN-ß) secretion by HPV11 E7 viral proteins in epithelial cells by affecting stimulator of interferon genes phosphorylation may account for the persistence of low-risk HPV replication in the RRP. Moreover, suppression of the IFN-I pathway in RRP cell types might provide clues regarding the hyporeactive function of local immune cells. However, activation of macrophage groups to produce IFN-ß can still destroy HPV-infected cells.


Subject(s)
Human papillomavirus 11 , Papillomavirus E7 Proteins , Papillomavirus Infections , Respiratory Tract Infections , Adult , Female , Humans , Male , Epithelial Cells/virology , Epithelial Cells/immunology , Human papillomavirus 11/genetics , Human papillomavirus 11/immunology , Immune Evasion , Immunity, Innate , Interferon-beta/metabolism , Interferon-beta/immunology , Interferon-beta/genetics , Macrophages/immunology , Macrophages/virology , Membrane Proteins/metabolism , Membrane Proteins/genetics , Papillomavirus E7 Proteins/metabolism , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Respiratory Tract Infections/virology , Respiratory Tract Infections/immunology
15.
Biochem Biophys Res Commun ; 712-713: 149915, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38663038

ABSTRACT

Viral infections pose a significant threat to public health, and the production of interferons represents one of the most critical antiviral innate immune responses of the host. Consequently, the screening and identification of compounds or reagents that induce interferon production are of paramount importance. This study commenced with the cultivation of host bacterium 15,597, followed by the infection of Escherichia coli with the MS2 bacteriophage. Utilizing the J2 capture technique, a class of dsRNA mixtures (MS2+15,597) was isolated from the E. coli infected with the MS2 bacteriophage. Subsequent investigations were conducted on the immunostimulatory activity of the MS2+15,597 mixture. The results indicated that the dsRNA mixtures (MS2+15,597) extracted from E. coli infected with the MS2 bacteriophage possess the capability to activate innate immunity, thereby inducing the production of interferon-ß. These dsRNA mixtures can activate the RIG-I and TLR3 pattern recognition receptors, stimulating the expression of interferon stimulatory factors 3/7, which in turn triggers the NF-κB signaling pathway, culminating in the cellular production of interferon-ß to achieve antiviral effects. This study offers novel insights and strategies for the development of broad-spectrum antiviral drugs, potentially providing new modalities for future antiviral therapies.


Subject(s)
Escherichia coli , Levivirus , RNA, Double-Stranded , Escherichia coli/virology , Escherichia coli/genetics , Escherichia coli/metabolism , RNA, Double-Stranded/metabolism , Humans , Levivirus/genetics , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 3/genetics , Immunity, Innate , Interferon-beta/metabolism , Interferon-beta/genetics , NF-kappa B/metabolism , DEAD Box Protein 58/metabolism , DEAD Box Protein 58/genetics , Signal Transduction , Interferon Regulatory Factor-7/metabolism , Interferon Regulatory Factor-7/genetics , Receptors, Immunologic , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics
16.
Vet Microbiol ; 292: 110050, 2024 May.
Article in English | MEDLINE | ID: mdl-38484578

ABSTRACT

The role of host factors in the replication of emerging senecavirus A (SVA) which induced porcine idiopathic vesicular disease (PIVD) distributed worldwide remains obscure. Here, interferon-induced transmembrane (IFITM) protein 1 and 2 inhibit SVA replication by positive feedback with RIG-I signaling pathway was reported. The expression levels of IFITM1 and IFITM2 increased significantly in SVA infected 3D4/21 cells. Infection experiments of cells with over and interference expression of IFITM1 and IFITM2 showed that these two proteins inhibit SVA replication by regulating the expression of interferon beta (IFN-ß), IFN-stimulated gene 15 (ISG-15), interleukin 6 (IL-6), IL-8, tumor necrosis factor alpha (TNF-α), IFN regulatory factor-3 (IRF3), and IRF7. Further results showed that antiviral responses of IFITM1 and IFITM2 were achieved by activating retinoic acid-inducible gene I (RIG-I) signaling pathway which in turn enhanced the expression of IFITM1 and IFITM2. It is noteworthy that conserved domains of these two proteins also paly the similar role. These findings provide new data on the role of host factors in infection and replication of SVA and help to develop new agents against the virus.


Subject(s)
Antigens, Differentiation , Interferon-beta , Membrane Proteins , Picornaviridae , Signal Transduction , Animals , Feedback , Interferon-beta/genetics , Swine , Virus Replication/genetics , Antigens, Differentiation/metabolism , Membrane Proteins/metabolism
17.
Autoimmunity ; 57(1): 2332340, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38545756

ABSTRACT

Interferon-beta (IFN-ß) is one of the classical drugs for immunomodulatory therapy in relapsing-remitting multiple sclerosis (RRMS) patients, but the drug responsiveness of different patients varies. Currently, there is no valid model to predict IFN-ß responsiveness. This research attempted to develop an IFN-ß responsiveness prediction model based on mRNA expression in RRMS patient peripheral blood mononuclear cells. Peripheral blood mononuclear cell mRNA expression datasets including 50 RRMS patients receiving IFN-ß treatment were obtained from GEO. Among the datasets, 24 cases from GSE24427 were included in a training set, and 18 and 9 cases from GSE19285 and GSE33464, respectively, were adopted as two independent test sets. In the training set, blood samples were collected immediately before first, second, month 1, 12, and 24 IFN-ß injection, and the mRNA expression data at four time points, namely, two days, one month, one year and two years after the onset of IFN-ß treatment, were compared with pre-treatment data to identify IFN-stimulated genes (ISGs). The ISGs at the one-month time point were used to construct the drug responsiveness prediction model. Next, the drug responsiveness model was verified in the two independent test sets to examine the performance of the model in predicting drug responsiveness. Finally, we used CIBERSORTx to estimate the content of cell subtypes in samples and evaluated whether differences in the proportions of cell subtypes were related to differences in IFN-ß responsiveness. Among the four time points, one month was the time point when the training set GSE24427 and test set GSE33464 had the highest number of ISGs. Functional analysis showed that these one-month ISGs were enriched in biological functions such as the innate immune response, type-I interferon signalling pathway, and other IFN-ß-associated functions. Based on these ISGs, we obtained a four-factor prediction model for IFN-ß responsiveness including MX1, MX2, XAF1, and LAMP3. In addition, the model demonstrated favourable predictive performance within the training set and two external test sets. A higher proportion of activated NK cells and lower naive CD4/total CD4 ratio might indicate better drug responsiveness. This research developed a polygene-based biomarker model that could predict RRMS patient IFN-ß responsiveness in the early treatment period. This model could probably help doctors screen out patients who would not benefit from IFN-ß treatment early and determine whether a current treatment plan should be continued.


Subject(s)
Multiple Sclerosis, Relapsing-Remitting , Multiple Sclerosis , Humans , RNA, Messenger/genetics , Leukocytes, Mononuclear , Interferon-beta/therapeutic use , Interferon-beta/genetics , Multiple Sclerosis, Relapsing-Remitting/diagnosis , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/genetics
18.
Virus Res ; 343: 199342, 2024 05.
Article in English | MEDLINE | ID: mdl-38408646

ABSTRACT

African swine fever virus is known to suppress type-I interferon (IFN) responses. The main objective of this study was to screen early-expressed viral genes for their ability to suppress IFN production. Out of 16 early genes examined, I73R exhibited robust suppression of cGAS-STING-induced IFN-ß promoter activities, impeding the function of both IRF3 and NF-κB transcription factors. As a result, I73R obstructed IRF3 nuclear translocation following the treatment of cells with poly(dA:dT), a strong inducer of the cGAS-STING signaling pathway. Although the I73R protein exhibits structural homology with the Zα domain binding to the left-handed helical form of DNA known as Z-DNA, its ability to suppress cGAS-STING induction of IFN-ß was independent of Z-DNA binding activity. Instead, the α3 and ß1 domains of I73R played a significant role in suppressing cGAS-STING induction of IFN-ß. These findings offer insights into the protein's functions and support its role as a virulence factor.


Subject(s)
African Swine Fever Virus , African Swine Fever , DNA, Z-Form , Interferon Type I , Animals , Swine , African Swine Fever Virus/genetics , Interferon-beta/genetics , Interferon-beta/metabolism , Signal Transduction/genetics , Immunity, Innate/genetics , DNA, Z-Form/metabolism , Membrane Proteins/metabolism , Interferon Type I/metabolism , Nucleotidyltransferases/genetics
19.
Arch Virol ; 169(2): 26, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38214770

ABSTRACT

Pigeons can be infected with various RNA viruses, and their innate immune system responds to viral infection to establish an antiviral response. Mitochondrial antiviral signaling protein (MAVS), an important adaptor protein in signal transduction, plays a pivotal role in amplifying the innate immune response. In this study, we successfully cloned pigeon MAVS (piMAVS) and performed a bioinformatics analysis. The results showed that the caspase recruitment domain (CARD) and transmembrane (TM) domain are highly conserved in poultry and mammals but poorly conserved in other species. Furthermore, we observed that MAVS expression is upregulated both in pigeons and pigeon embryonic fibroblasts (PEFs) upon RNA virus infection. Overexpression of MAVS resulted in increased levels of ß-interferon (IFN-ß), IFN-stimulated genes (ISGs), and interleukin (ILs) mRNA and inhibited Newcastle disease virus (NDV) replication. We also found that piMAVS and human MAVS (huMAVS) induced stronger expression of IFN-ß and ISGs when compared to chicken MAVS (chMAVS), and this phenomenon was also reflected in the degree of inhibition of NDV replication. Our findings demonstrate that piMAVS plays an important role in repressing viral replication by regulating the activation of the IFN signal pathway in pigeons. This study not only sheds light on the function of piMAVS in innate immunity but also contributes to a more comprehensive understanding of the innate immunity system in poultry. Our data also provide unique insights into the differences in innate immunity between poultry and mammal.


Subject(s)
Columbidae , Immunity, Innate , Signal Transduction , Animals , Humans , Antiviral Agents , Interferon-beta/genetics , Interferon-beta/metabolism , Mammals , Newcastle disease virus
20.
Vet Microbiol ; 290: 109973, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38211361

ABSTRACT

Newcastle disease virus (NDV) is an RNA virus that can promote its own replication through the inhibition of cellular mitochondrial fusion. The proteins involved in mitochondrial fusion, namely mitofusin 1 (Mfn1) and optic atrophy 1 (OPA1) are associated with interferon-beta (IFN-ß) secretion during NDV infection. However, the precise mechanism by which NDV modulates the Mfn1-mediated or OPA1-mediated fusion of mitochondria, thereby impacting IFN-ß, remains elusive. This study revealed that the downregulation of the mitochondrial protein known as coiled-coil-helix-coiled-coil-helix domain containing 10 (CHCHD10) exerts a negative regulatory effect on OPA1 and Mfn1 in human lung adenocarcinoma (A549) cells during the late stage of NDV infection. This reduction in CHCHD10 expression impeded cellular mitochondrial fusion, subsequently leading to a decline in the activation of interferon regulatory factor 3 (IRF3) and nuclear factor kappa B (NF-κB), ultimately resulting in diminished secretion of IFN-ß. In contrast, the overexpression of CHCHD10 alleviated infection-induced detrimental effect in mitochondrial fusion, thereby impeding viral proliferation. In summary, NDV enhances its replication by inhibiting the CHCHD10 protein, which impedes mitochondrial fusion and suppresses IFN-ß production through the activation of IRF3 and NF-κB.


Subject(s)
NF-kappa B , Newcastle disease virus , Humans , Animals , Newcastle disease virus/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Mitochondrial Dynamics , Interferon-beta/genetics , Interferon-beta/metabolism , Cell Proliferation , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...