Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 509
Filter
1.
Cell Mol Life Sci ; 81(1): 433, 2024 Oct 12.
Article in English | MEDLINE | ID: mdl-39395053

ABSTRACT

Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is an enveloped, positive-sense RNA virus that emerged in 2012, causing sporadic cases and localized outbreaks of severe respiratory illness with high fatality rates. A characteristic feature of the immune response to MERS-CoV infection is low type I IFN induction, despite its importance in viral clearance. The non-structural proteins (nsps) of other coronaviruses have been shown to block IFN production. However, the role of nsp5 from MERS-CoV in IFN induction of human respiratory cells is unclear. In this study, we elucidated the role of MERS-CoV-nsp5, the viral main protease, in modulating the host's antiviral responses in human bronchial epithelial BEAS 2b cells. We found that overexpression of MERS-CoV-nsp5 had a dose-dependent inhibitory effect on IFN-ß promoter activation and cytokine production induced by HMW-poly(I:C). It also suppressed IFN-ß promoter activation triggered by overexpression of key components in the RIG-I-like receptor (RLR) pathway, including RIG-I, MAVS, IKK-ε and IRF3. Moreover, the overexpression of MERS-CoV-nsp5 did not impair expression or phosphorylation of IRF3, but suppressed the nuclear translocation of IRF3. Further investigation revealed that MERS-CoV-nsp5 specifically interacted with IRF3. Using docking and molecular dynamic (MD) simulations, we also found that amino acids on MERS-CoV-nsp5, IRF3, and KPNA4 may participate in protein-protein interactions. Additionally, we uncovered protein conformations that mask the nuclear localization signal (NLS) regions of IRF3 and KPNA4 when interacting with MERS-CoV-nsp5, suggesting a mechanism by which this viral protein blocks IRF3 nuclear translocation. Of note, the IFN-ß expression was restored after administration of protease inhibitors targeting nsp5, indicating this suppression of IFN-ß production was dependent on the enzyme activity of nsp5. Collectively, our findings elucidate a mechanism by which MERS-CoV-nsp5 disrupts the host's innate antiviral immunity and thus provides insights into viral pathogenesis.


Subject(s)
Epithelial Cells , Interferon Regulatory Factor-3 , Middle East Respiratory Syndrome Coronavirus , Viral Nonstructural Proteins , Humans , Interferon Regulatory Factor-3/metabolism , Middle East Respiratory Syndrome Coronavirus/immunology , Epithelial Cells/metabolism , Epithelial Cells/virology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/genetics , Interferon Type I/metabolism , Cell Line , Cell Nucleus/metabolism , Interferon-beta/metabolism , Signal Transduction/drug effects , Poly I-C/pharmacology , Promoter Regions, Genetic/genetics , alpha Karyopherins/metabolism , alpha Karyopherins/genetics , Active Transport, Cell Nucleus , DEAD Box Protein 58/metabolism , DEAD Box Protein 58/genetics
2.
Inflammation ; 2024 Oct 14.
Article in English | MEDLINE | ID: mdl-39400777

ABSTRACT

Sepsis and septic shock are life-threatening systemic inflammatory conditions and among the most frequent causes of morbidity and mortality globally. Preclinical evidence has identified a number of diazepine-based compounds with therapeutic potential in inflammatory diseases. However, the potential anti-inflammatory properties of diazepines in the overwhelming immune response during sepsis have been rarely examined. Thus, the current study aimed to identify a new diazepine compound with therapeutic potential in sepsis. Assessing the inflammatory response of macrophages to Lipopolysaccharides (LPS) in vitro identified 2-[7-(trifluoromethyl)-2,3-dihydro-1H-1,4-diazepin-5-yl]phenol (2-TDDP) as a potential anti-inflammatory agent. It reduced secretion of Interleukin-1ß (IL-1ß), IL-6, IL-12p70, IL-18, Tumor necrosis factor-α (TNF-α), Interferon-γ (IFN-γ), IFN-ß, and increased the secretion of IL-10. In a mouse model of LPS-induced endotoxin shock, 2-TDDP reduced mortality and attenuated inflammation-induced tissue injury in the spleen, liver, kidney, and lung. This was accompanied by reduced serum levels of IL-1ß, IL-6, IL-12p70, TNF-α, IFN-γ, IFN-ß, and increased levels of IL-10. Importantly, 2-TDDP suppressed the Toll-like receptor 4 (TLR4)/Nuclear factor-κB (NF-κB) and TLR4/Interferon regulatory factor 3 (IRF3) signaling pathways through a reduction in the expression of TLR4, Myeloid differentiation primary response 88 (MyD88), P65, and TNF receptor-associated factor 3 (Traf3). Moreover, 2-TDDP suppressed the expression of CD86, Programmed death-ligand 1 (PD-L1) and C5a receptor (C5aR), but not Major histocompatibility complex II (MHCII). Analysis of splenic lymphocyte populations revealed a decrease in the number of CD4+, CD8+, and B cells. Collectively, these findings introduced the dihydrodiazepine 2-TDDP as a new anti-inflammatory agent with potent therapeutic potential in endotoxin shock, paving an avenue for future clinical application.

3.
J Neurosci ; 44(41)2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39227159

ABSTRACT

Targeting altered expression and/or activity of GABA (γ-aminobutyric acid) transporters (GATs) provide therapeutic benefit for age-related impairments, including cognitive dysfunction. However, the mechanisms underlying the transcriptional regulation of GATs are unknown. In the present study, we demonstrated that the stimulator of interferon genes (STING) upregulates GAT1 and GAT3 expression in the brain, which resulted in cognitive dysfunction. Genetic and pharmacological intervention of STING suppressed the expression of both GAT1 and GAT3, increased the ambient GABA concentration, and therefore, enhanced tonic GABAA inhibition of principal hippocampal neurons, resulting in spatial learning and working memory deficits in mice in a type I interferon-independent manner. Stimulation of the STING→GAT pathway efficiently restored cognitive dysfunction in STING-deficient mice models. Our study uncovered for the first time that the STING signaling pathway regulates GAT expression in a cell autonomous manner and therefore could be a novel target for GABAergic cognitive deficits.


Subject(s)
GABA Plasma Membrane Transport Proteins , Homeostasis , Interferon Regulatory Factor-3 , Membrane Proteins , Mice, Inbred C57BL , Signal Transduction , gamma-Aminobutyric Acid , Animals , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice , gamma-Aminobutyric Acid/metabolism , GABA Plasma Membrane Transport Proteins/metabolism , GABA Plasma Membrane Transport Proteins/genetics , Signal Transduction/physiology , Homeostasis/physiology , Male , Interferon Regulatory Factor-3/metabolism , Cognition/physiology , Hippocampus/metabolism , Mice, Knockout
4.
J Nat Med ; 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39283364

ABSTRACT

Previously, we reported that azamollugin, an aza-derivative of mollugin, exhibited potent inhibitory activity on NO production in LPS-stimulated RAW 264.7 cells. Further investigations in this study revealed that azamollugin not only suppressed iNOS gene expression regulated by NF-κB, but also inhibited LPS-induced IFN-ß expression, which is known to be regulated by IRF3. Azamollugin exhibited an inhibitory activity on LPS-induced IRAK1 activation, suggesting inhibitory effect on the MyD88-dependent pathway. Furthermore, azamollugin inhibited LPS-induced phosphorylation of IRF3 and its upstream factor, TBK1/IKKε, suggesting an inhibitory effect on the TRIF-dependent pathway via TLR4. In addition, azamollugin also suppressed poly(I:C)-induced phosphorylation of TBK1 and IRF3, suggesting an inhibitory effect on the TRIF-dependent pathway via TLR3. These results suggest that azamollugin has inhibitory activity against both the MyD88-dependent and TRIF-dependent pathways, respectively.

5.
Acta Pharm Sin B ; 14(8): 3513-3527, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39220861

ABSTRACT

Bile acids (BAs) are natural metabolites in mammals and have the potential to function as drugs against viral infection. However, the limited understanding of chenodeoxycholic acid (CDCA) receptors and downstream signaling, along with its lower suppression efficiency in inhibiting virus infection limits its clinical application. In this study, we demonstrate that farnesoid X receptor (FXR), the receptor of CDCA, negatively regulates interferon signaling, thereby contributing to the reduced effectiveness of CDCA against virus replication. FXR deficiency or pharmacological inhibition enhances interferon signaling activation to suppress virus infection. Mechanistically, FXR impairs the DNA binding and transcriptional abilities of activated interferon regulatory factor 3 (IRF3) through interaction. Reduced IRF3 transcriptional activity by FXR-IRF3 interaction significantly undermines the expression of Interferon Beta 1 (IFNB1) and the antiviral response of cells, especially upon the CDCA treatment. In FXR-deficient cells, or when combined with Z-guggulsterone (GUGG) treatment, CDCA exhibits a more potent ability to restrict virus infection. Thus, these findings suggest that FXR serves as a limiting factor for CDCA in inhibiting virus replication, which can be attributed to the "signaling-brake" roles of FXR in interferon signaling. Targeting FXR inhibition represents a promising pharmaceutical strategy for the clinical application of BAs metabolites as antiviral drugs.

6.
Curr Cardiol Rev ; 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39229989

ABSTRACT

Thirty percent of deaths worldwide are caused by cardiovascular disorders (CVDs). As per the WHO data, the number of fatalities due to CVDs is 17.9 million years, and it is projected to cause 22.2 million deaths by 2030. In terms of gender, women die from CVD at a rate of 51% compared to 42% for males. Most people use phytochemicals, a type of traditional medicine derived from plants, either in addition to or instead of commercially available medications to treat and prevent CVD. Phytochemicals are useful in lowering cardiovascular risks, especially for lowering blood cholesterol, lowering obesity-related factors, controlling blood sugar and the consequences of type 2 diabetes, controlling oxidative stress factors and inflammation, and preventing platelet aggregation. Medicinal plants that are widely known for treating CVD include ginseng, ginkgo biloba, ganoderma lucidum, gynostemma pentaphyllum, viridis amaranthus, etc. Plant sterol, flavonoids, polyphenols, sulphur compound and terpenoid are the active phytochemicals present in these plants. The aim of this article is to cover more and more drugs that are used for cardiovascular diseases. In this article, we will learn about the use of different herbal drugs, mechanism of action, phytochemical compounds, side effects, etc. However, more research is required to comprehend the process and particular phytochemicals found in plants that treat CVD.

7.
J Ethnopharmacol ; 337(Pt 1): 118707, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39181282

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Mailuoning oral liquid (MLN O), one traditional Chinese patent medicine, has a good therapeutic effect on thromboangiitis obliterans (TAO) in clinical practice. However, the underlying mechanism remains unclear. AIM OF THE STUDY: This study aimed to explore the effects and potential mechanisms of MLN O against TAO based on network pharmacology and experimental verification. MATERIALS AND METHODS: Network pharmacology was used to identify the intersectional targets and signaling pathways of MLN O and TAO. In vivo, the TAO model was established by injecting sodium laurate and dihydrotestosterone (DHT) into the femoral arteries of Wistar rats. Rats were given the indicated drugs by intragastric administration (i.g.), intravenous injection (i.v.), or subcutaneous injection (s.c.) per day for 21 days since a week before surgery. In vitro, HUVECs, RAW264.7, and THP-1 cells were stimulated by LPS and DHT to simulate the pathological changes of TAO. The anti-inflammatory, anticoagulant, and immunomodulatory effects of MLN O were evaluated by histological observation, blood biochemical indexes detection, H&E staining, immunohistochemistry, enzyme-linked immunosorbent assay (ELISA), qRT-PCR, western blotting and immunofluorescence assays. Furthermore, the vascular ring test was applied to explore the vasodilatory activity of MLN O. RESULTS: MLN O significantly improved the pathological signs in TAO rats through its excellent anti-inflammatory, anticoagulant, immunomodulatory, and vasodilatory effects. Specifically, MLN O alleviated the gangrene and reduced the thrombosis in TAO rats, meanwhile, suppressed the expressions of inflammatory factors and clotting factors, which is related to the inactivations of cGAS-STING-IRF3 and TLR4-MAPKs/NF-κB signaling pathways. However, the superphysiological dose of DHT deteriorated the pathological development of TAO in vitro and in vivo. Moreover, the results of network pharmacology are consistent with the experimental verification. CONCLUSION: Collectively, this study indicates for the first time that MLN O could alleviate TAO by inhibiting cGAS-STING-IRF3 and TLR4-MAPKs/NF-κB signaling pathways, which sheds light on a novel clinical therapeutic strategy for TAO.

8.
J Biol Chem ; 300(9): 107645, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39127175

ABSTRACT

Epstein-Barr virus (EBV), the causative agent of infectious mononucleosis, persistently infects over 90% of the human adult population and is associated with several human cancers. To establish life-long infection, EBV tampers with the induction of type I interferon (IFN I)-dependent antiviral immunity in the host. How various EBV genes help orchestrate this crucial strategy is incompletely defined. Here, we reveal a mechanism by which the EBV nuclear antigen 3A (EBNA3A) may inhibit IFNß induction. Using proximity biotinylation we identify the histone acetyltransferase P300, a member of the IFNß transcriptional complex, as a binding partner of EBNA3A. We further show that EBNA3A also interacts with the activated IFN-inducing transcription factor interferon regulatory factor 3 that collaborates with P300 in the nucleus. Both events are mediated by the N-terminal domain of EBNA3A. We propose that EBNA3A limits the binding of interferon regulatory factor 3 to the IFNß promoter, thereby hampering downstream IFN I signaling. Collectively, our findings suggest a new mechanism of immune evasion by EBV, affected by its latency gene EBNA3A.


Subject(s)
E1A-Associated p300 Protein , Epstein-Barr Virus Nuclear Antigens , Herpesvirus 4, Human , Interferon Regulatory Factor-3 , Interferon-beta , Humans , Epstein-Barr Virus Nuclear Antigens/metabolism , Epstein-Barr Virus Nuclear Antigens/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Interferon-beta/metabolism , Interferon-beta/genetics , Herpesvirus 4, Human/metabolism , Herpesvirus 4, Human/genetics , E1A-Associated p300 Protein/metabolism , E1A-Associated p300 Protein/genetics , HEK293 Cells , Promoter Regions, Genetic , Gene Expression Regulation , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/genetics , Protein Binding , Signal Transduction , Cell Nucleus/metabolism
9.
Cell Rep ; 43(8): 114608, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39120972

ABSTRACT

Ubiquitination is essential for the proteasomal turnover of IRF3, the central factor mediating the antiviral innate immune response. However, the spatiotemporal regulation of IRF3 ubiquitination for the precise activation and timely resolution of innate immunity remains unclear. Here, we identified BRCA1-associated protein-1 (BAP1) and ubiquitin-protein ligase E3C (UBE3C) as the key deubiquitinase and ubiquitinase for temporal control of IRF3 stability during viral infection. In the early stage, BAP1 dominates and removes K48-linked ubiquitination of IRF3 in the nucleus, preventing its proteasomal degradation and facilitating efficient interferon (IFN)-ß production. In the late stage, E3 ligase UBE3C, induced by IFN-ß, specifically mediates IRF3 ubiquitination and promotes its proteasomal degradation. Overall, the sequential interactions with BAP1 and UBE3C govern IRF3 stability during innate response, ensuring effective viral clearance and inflammation resolution. Our findings provide insights into the temporal control of innate signaling and suggest potential interventions in viral infection.


Subject(s)
Immunity, Innate , Interferon Regulatory Factor-3 , Tumor Suppressor Proteins , Ubiquitin Thiolesterase , Ubiquitin-Protein Ligases , Ubiquitination , Interferon Regulatory Factor-3/metabolism , Ubiquitin-Protein Ligases/metabolism , Humans , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Tumor Suppressor Proteins/metabolism , Animals , HEK293 Cells , Mice , Interferon-beta/metabolism , Proteolysis , Mice, Inbred C57BL , Signal Transduction , Proteasome Endopeptidase Complex/metabolism
10.
J Neuroinflammation ; 21(1): 212, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39215356

ABSTRACT

The pathological role of interferon signaling is emerging in neuroinflammatory disorders, yet, the specific role of Interferon Regulatory Factor 3 (IRF3) in neuroinflammation remains poorly understood. Here, we show that global IRF3 deficiency delays TLR4-mediated signaling in microglia and attenuates the hallmark features of LPS-induced inflammation such as cytokine release, microglial reactivity, astrocyte activation, myeloid cell infiltration, and inflammasome activation. Moreover, expression of a constitutively active IRF3 (S388D/S390D: IRF3-2D) in microglia induces a transcriptional program reminiscent of the Activated Response Microglia and the expression of genes associated with Alzheimer's disease, notably apolipoprotein-e. Using bulk-RNAseq of IRF3-2D brain myeloid cells, we identified Z-DNA binding protein-1 (ZBP1) as a target of IRF3 that is relevant across various neuroinflammatory disorders. Lastly, we show IRF3 phosphorylation and IRF3-dependent ZBP1 induction in response to Aß in primary microglia cultures. Together, our results identify IRF3 as an important regulator of LPS and Aß -mediated neuroinflammatory responses and highlight IRF3 as a central regulator of disease-specific gene activation in different neuroinflammatory diseases.


Subject(s)
Alzheimer Disease , Interferon Regulatory Factor-3 , Microglia , Neuroinflammatory Diseases , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Animals , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Mice , Neuroinflammatory Diseases/genetics , Neuroinflammatory Diseases/metabolism , Microglia/metabolism , Lipopolysaccharides/pharmacology , Lipopolysaccharides/toxicity , Mice, Inbred C57BL , Gene Expression Regulation/drug effects , Cells, Cultured , Humans , Mice, Knockout
11.
J Virol ; 98(9): e0078424, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39194214

ABSTRACT

Porcine respiratory and reproductive syndrome (PRRS) is one of the most devastating infectious diseases of pigs, causing reproductive failures in sows and severe respiratory symptoms in piglets and growing pigs. MicroRNAs (miRNAs) are reported to play an essential role in virus-host interactions. In this study, we demonstrated that miR-451 enhanced type I interferon (IFN-I) production through targeting proteasome subunit ß8 (PSMB8), therefore restricting PRRS virus (PRRSV) replication. We showed that the expression of PSMB8 was upregulated by PRRSV infection, and knockdown of PSMB8 inhibited PRRSV replication by promoting IFN-I production. Moreover, we demonstrated that PSMB8 interacted with the regulatory domain of IRF3 to mediate K48-linked polyubiquitination and degradation of IRF3. Also, importantly, we showed that PSMB8, as a target gene of miR-451, negatively regulated IFN-I production by promoting IRF3 degradation, which is a previously unknown mechanism for PSMB8 to modulate innate immune responses. IMPORTANCE: Porcine respiratory and reproductive syndrome virus (PRRSV), as a huge threat to the swine industry, is a causative agent that urgently needs to be solved. The dissecting of PRRSV pathogenesis and understanding of the host-pathogen interaction will provide insights into developing effective anti-PRRSV strategies. In this study, we showed that miR-451 dramatically inhibited PRRSV replication by targeting proteasome subunit ß8 (PSMB8), a subunit of the immunoproteasome. Mutation of PSMB8 is often related to autoinflammatory diseases due to the elevated IFN production. We revealed that PSMB8 downregulated IFN production by promoting IRF3 degradation. In addition, we showed that PRRSV infection upregulated PSMB8 expression. Taken together, our findings reveal that miR-451 is a negative regulator of PRRSV replication, and PSMB8, a target gene of miR-451, negatively regulates IFN-I production by promoting IRF3 degradation, which is a previously unknown mechanism for PSMB8 to regulate innate immune responses.


Subject(s)
Interferon Regulatory Factor-3 , MicroRNAs , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Proteasome Endopeptidase Complex , Virus Replication , Animals , Porcine respiratory and reproductive syndrome virus/genetics , Swine , MicroRNAs/genetics , MicroRNAs/metabolism , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/genetics , Porcine Reproductive and Respiratory Syndrome/virology , Porcine Reproductive and Respiratory Syndrome/genetics , Porcine Reproductive and Respiratory Syndrome/metabolism , Porcine Reproductive and Respiratory Syndrome/immunology , Humans , Interferon Type I/metabolism , Ubiquitination , Immunity, Innate , Cell Line , HEK293 Cells , Host-Pathogen Interactions/genetics , Proteolysis
12.
J Virol ; 98(9): e0079624, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39115433

ABSTRACT

Host cells have evolved an intricate regulatory network to fine tune the type-I interferon responses. However, the full picture of this regulatory network remains to be depicted. In this study, we found that knock out of zinc-finger CCHC-type containing protein 8 (ZCCHC8) impairs the replication of influenza A virus (IAV), Sendai virus (Sev), Japanese encephalitis virus (JEV), and vesicular stomatitis virus (VSV). Further investigation unveiled that ZCCHC8 suppresses the type-I interferon responses by targeting the interferon regulatory factor 3 (IRF3) signaling pathway. Mechanistically, ZCCHC8 associates with phosphorylated IRF3 and disrupts the interaction of IRF3 with the co-activator CREB-binding protein (CBP). Additionally, the direct binding of ZCCHC8 with the IFN-stimulated response element (ISRE) impairs the ISRE-binding of IRF3. Our study contributes to the comprehensive understanding for the negative regulatory network of the type-I interferon responses and provides valuable insights for the control of multiple viruses from a host-centric perspective.IMPORTANCEThe innate immune responses serve as the initial line of defense against invading pathogens and harmful substances. Negative regulation of the innate immune responses plays an essential role in avoiding auto-immune diseases and over-activated immune responses. Hence, the comprehensive understanding of the negative regulation network for innate immune responses could provide novel therapeutic insights for the control of viral infections and immune dysfunction. In this study, we report that ZCCHC8 negatively regulates the type-I interferon responses. We illustrate that ZCCHC8 impedes the IRF3-CBP association by interacting with phosphorylated IRF3 and competes with IRF3 for binding to ISRE. Our study demonstrates the role of ZCCHC8 in the replication of multiple RNA viruses and contributes to a deeper understanding of the negative regulation system for the type-I interferon responses.


Subject(s)
CREB-Binding Protein , Immunity, Innate , Interferon Regulatory Factor-3 , Interferon Type I , Sendai virus , Signal Transduction , Virus Replication , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Humans , HEK293 Cells , Sendai virus/physiology , Sendai virus/genetics , CREB-Binding Protein/metabolism , CREB-Binding Protein/genetics , RNA Viruses/physiology , RNA Viruses/immunology , RNA Viruses/genetics , Animals , A549 Cells , Influenza A virus/physiology , Influenza A virus/immunology , Phosphorylation , Host-Pathogen Interactions , Vesiculovirus/physiology , Encephalitis Virus, Japanese/physiology , Encephalitis Virus, Japanese/immunology
13.
Adv Biol (Weinh) ; 8(10): e2400174, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38977406

ABSTRACT

Mycobacterium tuberculosis (M. tb) is a significant intracellular pathogen responsible for numerous infectious disease-related deaths worldwide. It uses ESX-1 T7SS to damage phagosomes and to enter the cytosol of host cells after phagocytosis. During infection, M. tb and host mitochondria release dsDNA, which activates the CGAS-STING1 pathway. This pathway leads to the production of type I interferons and proinflammatory cytokines and activates autophagy, which targets and degrades bacteria within autophagosomes. However, the role of type I IFNs in immunity against M. tb is controversial. While previous research has suggested a protective role, recent findings from cgas-sting1 knockout mouse studies have contradicted this. Additionally, a study using knockout mice and non-human primate models uncovered a new mechanism by which neutrophils recruited to lung infections form neutrophil extracellular traps. Activating plasmacytoid dendritic cells causes them to produce type I IFNs, which interfere with the function of interstitial macrophages and increase the likelihood of tuberculosis. Notably, M. tb uses its virulence proteins to disrupt the CGAS-STING1 signaling pathway leading to enhanced pathogenesis. Investigating the CGAS-STING1 pathway can help develop new ways to fight tuberculosis.


Subject(s)
Autophagy , Interferon Type I , Membrane Proteins , Mycobacterium tuberculosis , Nucleotidyltransferases , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/immunology , Interferon Type I/immunology , Interferon Type I/metabolism , Animals , Autophagy/immunology , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Membrane Proteins/metabolism , Membrane Proteins/immunology , Membrane Proteins/genetics , Humans , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/metabolism , Mice , Signal Transduction/immunology
14.
Adv Sci (Weinh) ; 11(35): e2308890, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39004913

ABSTRACT

Interferons (IFNs) activate JAK-STAT pathways to induce downstream effector genes for host defense against invaded pathogens and tumors. Here both type I (ß) and II (γ) IFNs are shown that can activate the transcription factor IRF3 in parallel with STAT1. IRF3-deficiency impairs transcription of a subset of downstream effector genes induced by IFN-ß and IFN-γ. Mechanistically, IFN-induced activation of IRF3 is dependent on the cGAS-STING-TBK1 axis. Both IFN-ß and IFN-γ cause mitochondrial DNA release into the cytosol. In addition, IFNs induce JAK1-mediated tyrosine phosphorylation of cGAS at Y214/Y215, which is essential for its DNA binding activity and signaling. Furthermore, deficiency of cGAS, STING, or IRF3 impairs IFN-ß- or IFN-γ-mediated antiviral and antitumor activities. The findings reveal a novel IRF3 activation pathway parallel with the canonical STAT1/2 activation pathways triggered by IFNs and provide an explanation for the pleiotropic roles of the cGAS-STING-IRF3 axis in host defense.


Subject(s)
Interferon Regulatory Factor-3 , Membrane Proteins , Nucleotidyltransferases , Signal Transduction , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Animals , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Humans , Interferon-gamma/metabolism , Interferon-gamma/immunology , Interferon-gamma/genetics , Interferon Type I/metabolism , Interferon Type I/genetics , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Interferon-beta/metabolism , Interferon-beta/genetics
15.
Dev Comp Immunol ; 158: 105208, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38834141

ABSTRACT

Interferon regulatory factors (IRFs) are transcription factors involved in immune responses, such as pathogen response regulation, immune cell growth, and differentiation. IRFs are necessary for the synthesis of type I interferons through a signaling cascade when pathogen recognition receptors identify viral DNA or RNA. We discovered that irf3 is expressed in the early embryonic stages and in all immune organs of adult zebrafish. We demonstrated the antiviral immune mechanism of Irf3 against viral hemorrhagic septicemia virus (VHSV) using CRISPR/Cas9-mediated knockout zebrafish (irf3-KO). In this study, we used a truncated Irf3 protein, encoded by irf3 with a 10 bp deletion, for further investigation. Upon VHSV injection, irf3-KO zebrafish showed dose-dependent high and early mortality compared with zebrafish with the wild-type Irf3 protein (WT), confirming the antiviral activity of Irf3. Based on the results of expression analysis of downstream genes upon VHSV challenge, we inferred that Irf3 deficiency substantially affects the expression of ifnphi1 and ifnphi2. However, after 5 days post infection (dpi), ifnphi3 expression was not significantly altered in irf3-KO compared to that in WT, and irf7 transcription showed a considerable increase in irf3-KO after 5 dpi, indicating irf7's control over ifnphi3 expression. The significantly reduced expression of isg15, viperin, mxa, and mxb at 3 dpi also supported the effect of Irf3 deficiency on the antiviral activity in the early stage of infection. The higher mortality in irf3-KO zebrafish than in WT might be due to an increased inflammation and tissue damage that occurs in irf3-KO because of delayed immune response. Our results suggest that Irf3 plays a role in antiviral immunity of zebrafish by modulating critical immune signaling molecules and regulating antiviral immune genes.


Subject(s)
CRISPR-Cas Systems , Gene Knockout Techniques , Hemorrhagic Septicemia, Viral , Interferon Regulatory Factor-3 , Novirhabdovirus , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/immunology , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Novirhabdovirus/physiology , Novirhabdovirus/immunology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Hemorrhagic Septicemia, Viral/immunology , Hemorrhagic Septicemia, Viral/genetics , Hemorrhagic Septicemia, Viral/virology , Animals, Genetically Modified , Fish Diseases/immunology , Fish Diseases/virology , Fish Diseases/genetics , Immunity, Innate/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/virology , Disease Models, Animal , Interferons
16.
Vet Sci ; 11(6)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38921999

ABSTRACT

African swine fever virus (ASFV) is a double-stranded DNA virus with an envelope. ASFV has almost the largest genome among all DNA viruses, and its mechanisms of immune evasion are complex. Better understanding of the molecular mechanisms of ASFV genes will improve vaccine design. A238L, a nonstructural protein of ASFV, inhibits NF-κB activation by suppressing the HAT activity of p300. Whether A238L also affects the transcriptional activity of IRF3 remains unexplored. Here we first confirmed the ability of A238L to suppress NF-κB-activity in L929 cells. A238L inhibits the expression of proinflammatory cytokine genes. In contrast, A238L increased the phosphorylation levels of TBK1 and IRF3 in three different cell lines. A238L increases the IRF3-driven promoter activity and induces IRF3 nuclear translocation. Furthermore, A238L enhanced innate antiviral immunity in the absence or presence of poly d (A:T) or poly (I:C) stimulation, or herpes simplex virus type 1 (HSV-1) or Sendai virus (SeV) infection. This study reveals a previously unrecognized role of A238L in promoting antiviral immune responses by TBK1-IRF3 pathway activation.

17.
Virol Sin ; 39(4): 587-599, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38823782

ABSTRACT

Herpesviruses antagonize host antiviral responses through a myriad of molecular strategies culminating in the death of the host cells. Pseudorabies virus (PRV) is a significant veterinary pathogen in pigs, causing neurological sequalae that ultimately lead to the animal's demise. PRV is known to trigger apoptotic cell death during the late stages of infection. The virion host shutdown protein (VHS) encoded by UL41 plays a crucial role in the PRV infection process. In this study, we demonstrate that UL41 inhibits PRV-induced activation of inflammatory cytokine and negatively regulates the cGAS-STING-mediated antiviral activity by targeting IRF3, thereby inhibiting the translocation and phosphorylation of IRF3. Notably, mutating the conserved amino acid sites (E192, D194, and D195) in the RNase domain of UL41 or knocking down UL41 inhibits the immune evasion of PRV, suggesting that UL41 may play a crucial role in PRV's evasion of the host immune response during infection. These results enhance our understanding of how PRV structural proteins assist the virus in evading the host immune response.


Subject(s)
Herpesvirus 1, Suid , Immune Evasion , Interferon Regulatory Factor-3 , NF-kappa B , Herpesvirus 1, Suid/immunology , Herpesvirus 1, Suid/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Animals , Swine , NF-kappa B/metabolism , NF-kappa B/genetics , NF-kappa B/immunology , Humans , Interferons/immunology , Interferons/metabolism , Interferons/genetics , Pseudorabies/virology , Pseudorabies/immunology , Cell Line , Host-Pathogen Interactions/immunology , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Proteins/immunology , HEK293 Cells , Phosphorylation , Protein Transport
18.
Vet Microbiol ; 295: 110148, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38851152

ABSTRACT

Water buffalo Hunnivirus (BufHuV) belongs to the family Picornaviridae and is a newly discovered member of the Hunnivirus A genus. It causes intestinal diseases in cattle, mainly lead to subclinical infections, thereby seriously threatening the health of cattle herds. In addition, it can also bring about various clinical disease syndromes which results in severe economic losses to the cattle industry. To date, there have been no reports worldwide on the study of Hunnivirus virus infecting host cells and causing innate immune responses. In this study, we found that interferon treatment effectively blocked BufHuV replication and infection with the virus weakened the host antiviral responses. Inhibiting the transcription of IFN-ß and ISGs induced by either Sendai virus (SeV) or poly(I:C) in MDBK and HCT-8 cells, were dependent on the IRF3 or NF-κB signaling pathways, and this inhibited the activation of IFN-ß promoter by TBK1 and its upstream molecules, RIGI and MDA5. By constructing and screening five BufHuV proteins, we found that VP2, 2 C, 3 C and 3D inhibited the activation of IFN-ß promoter induced by SeV. Subsequently, we showed that VP2 inhibited the activation of IRF3 induced by SeV or poly (I:C), and it inhibited IRF3 activation by inhibiting its phosphorylation and nuclear translocation. In addition, we confirmed that VP2 inhibited the activation of IFNß induced by signaling molecules, MDA5 and TBKI. In summary, these findings provide new insights into the pathogenesis of Hunnivirus and its mechanisms involved in evading host immune responses.


Subject(s)
Interferon Regulatory Factor-3 , Interferon-beta , Interferon-beta/genetics , Interferon-beta/immunology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Animals , Humans , Cell Line , Signal Transduction/drug effects , Viral Structural Proteins/genetics , Viral Structural Proteins/metabolism , Virus Replication/drug effects , Immunity, Innate , Cattle , Buffaloes/virology , NF-kappa B/metabolism
19.
Dev Comp Immunol ; 159: 105218, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38914152

ABSTRACT

Increasing evidence has been shown that OTUB1, a member of OTU deubiquitinases, is of importance in regulating the immune system. However, its molecular identification and functional characterization in teleosts are still rarely known. In this work, we cloned the otub1 of miiuy croaker (Miichthys miiuy), analyzed its sequence, structure, and evolution at genetic and protein levels, and determined its function in the antiviral immune response. The complete open reading frame (ORF) of miiuy croaker otub1 is 843 bp in length, encoding 280 amino acids. Miiuy croaker Otub1 has an OTU domain at the carboxyl terminus, which is a common functional domain that exists in OTU deubiquitinases. Molecular characteristics and evolution analysis results indicated that miiuy croaker Otub1, especially its functional domain, is highly conserved during evolution. The luciferase reporter assays showed that miiuy croaker Otub1 could significantly inhibit the poly(I:C) and Irf3-induced IFN1 and IFN-stimulated response element (ISRE) activation. Further experiments showed that miiuy croaker Otub1 decreases Irf3 protein abundance by promoting its proteasomal degradation. These data suggest that the evolutionarily conserved Otub1 acts as a suppressor in controlling antiviral immune response by promoting Irf3 proteasomal degradation in miiuy croaker.


Subject(s)
Fish Proteins , Interferon Regulatory Factor-3 , Perciformes , Proteasome Endopeptidase Complex , Proteolysis , Animals , Perciformes/immunology , Perciformes/genetics , Fish Proteins/genetics , Fish Proteins/metabolism , Fish Proteins/immunology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/genetics , Immunity, Innate , Evolution, Molecular , Poly I-C/immunology , Fish Diseases/immunology , Fish Diseases/virology , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/genetics , Cloning, Molecular , Phylogeny
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