202 results on '"IRF-3"'
Search Results
2. Herpes Simplex Virus Type 2 Blocks IFN-β Production through the Viral UL24 N-Terminal Domain-Mediated Inhibition of IRF-3 Phosphorylation.
- Author
-
Zhang, Binman, Li, Yuncheng, Yang, Ping, He, Siyu, Li, Weilin, Li, Miaomiao, Hu, Qinxue, and Zhang, Mudan
- Subjects
- *
HUMAN herpesvirus 2 , *HERPES genitalis , *CYTOTOXINS , *CELL survival , *CELL cycle - Abstract
Herpes simplex virus type 2 (HSV-2) is a sexually transmitted virus, the cause of genital herpes, and its infection can increase the risk of HIV-1 infection. After initial infection, HSV-2 can establish lifelong latency within the nervous system, which is likely associated with the virus-mediated immune evasion. In this study, we found that HSV-2 UL24 significantly inhibited the activation of the IFN-β promoter and the production of IFN-β at both mRNA and protein levels. Of importance, the inhibitory effect of HSV-2 on IFN-β production was significantly impaired in the context of HSV-2 infection when UL24 was knocked down. Additional studies revealed that, although the full-length HSV-2 UL24 affected cell cycle and viability to some extent, its N-terminal 1–202AA domain showed no obvious cytotoxicity while its C-terminal 201–281 AA domain had a minimal impact on cell viability. Further studies showed that the N-terminal 1–202 AA domain of HSV-2 UL24 (HSV-2 UL24-N) was the main functional region responsible for the inhibition of IFN-β production mediated by HSV-2 UL24. This domain significantly suppressed the activity of RIG-IN, MAVS, TBK-1, IKK-ε, or the IRF-3/5D-activated IFN-β promoter. Mechanistically, HSV-2 UL24-N suppressed IRF-3 phosphorylation, resulting in the inhibition of IFN-β production. The findings of this study highlight the significance of HSV-2 UL24 in inhibiting IFN-β production, revealing two potential roles of UL24 during HSV-2 infection: facilitating immune evasion and inducing cell cycle arrest. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Grainyhead-like-2, an epithelial master programmer, promotes interferon induction and suppresses breast cancer recurrence.
- Author
-
MacFawn, Ian, Farris, Joshua, Pifer, Phillip, Margaryan, Naira V., Akhter, Halima, Wang, Lei, Dziadowicz, Sebastian, Denvir, James, Hu, Gangqing, and Frisch, Steven M.
- Subjects
- *
CANCER relapse , *TYPE I interferons , *INTERFERONS , *BREAST cancer , *EMBRYONIC stem cells , *TUMOR suppressor genes , *INTERFERON receptors , *TUMOR suppressor proteins - Abstract
Type-I and -III interferons play a central role in immune rejection of pathogens and tumors, thus promoting immunogenicity and suppressing tumor recurrence. Double strand RNA is an important ligand that stimulates tumor immunity via interferon responses. Differentiation of embryonic stem cells to pluripotent epithelial cells activates the interferon response during development, raising the question of whether epithelial vs. mesenchymal gene signatures in cancer potentially regulate the interferon pathway as well. Here, using genomics and signaling approaches, we show that Grainyhead-like-2 (GRHL2), a master programmer of epithelial cell identity, promotes type-I and -III interferon responses to double-strand RNA. GRHL2 enhanced the activation of IRF3 and relA/NF-kB and the expression of IRF1; a functional GRHL2 binding site in the IFNL1 promoter was also identified. Moreover, time to recurrence in breast cancer correlated positively with GRHL2 protein expression, indicating that GRHL2 is a tumor recurrence suppressor, consistent with its enhancement of interferon responses. These observations demonstrate that epithelial cell identity supports interferon responses in the context of cancer. • This manuscript demonstrates that Grainyhead-like-2, a core master programmer of epithelial cell identity, promotes type-I/III interferon responses to double strand RNA and suppresses tumor recurrence. • The work is significant because it connects the epithelial gene expression program with interferon response capability. • These observations have important inplications for tumor immune surveillance and development [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. FHL2 Inhibits SARS-CoV-2 Replication by Enhancing IFN-β Expression through Regulating IRF-3.
- Author
-
Xu, Zhiqiang, Tian, Mingyao, Tan, Qihan, Hao, Pengfei, Gao, Zihan, Li, Chang, and Jin, Ningyi
- Subjects
- *
TYPE I interferons , *INTERFERONS , *SARS-CoV-2 , *COVID-19 pandemic - Abstract
SARS-CoV-2 triggered the global COVID-19 pandemic, posing a severe threat to public health worldwide. The innate immune response in cells infected by SARS-CoV-2 is primarily orchestrated by type I interferon (IFN), with IFN-β exhibiting a notable inhibitory impact on SARS-CoV-2 replication. FHL2, acting as a docking site, facilitates the assembly of multiprotein complexes and regulates the transcription of diverse genes. However, the association between SARS-CoV-2 and FHL2 remains unclear. In this study, we report for the first time that SARS-CoV-2 infection in Caco2 cells results in the upregulation of FHL2 expression, while the virus's N proteins can enhance FHL2 expression. Notably, the knockdown of FHL2 significantly amplifies SARS-CoV-2 replication in vitro. Conversely, the overexpression of FHL2 leads to a marked reduction in SARS-CoV-2 replication, with the antiviral property of FHL2 being independent of the cell or virus type. Subsequent experiments reveal that FHL2 supports IFN-β transcription by upregulating the expression and phosphorylation of IRF-3, thereby impeding SARS-CoV-2 replication in cells. These findings highlight FHL2 as a potential antiviral target for treating SARS-CoV-2 infections. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Comprehensive Mutagenesis of Herpes Simplex Virus 1 Genome Identifies UL42 as an Inhibitor of Type I Interferon Induction.
- Author
-
Chapon, Maxime, Parvatiyar, Kislay, Aliyari, Saba Roghiyh, Zhao, Jeffrey S, and Cheng, Genhong
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Biological Sciences ,Biotechnology ,Genetics ,Human Genome ,Sexually Transmitted Infections ,Infectious Diseases ,2.2 Factors relating to the physical environment ,Aetiology ,Infection ,A549 Cells ,Antiviral Agents ,DNA-Directed DNA Polymerase ,Exodeoxyribonucleases ,HEK293 Cells ,Herpesvirus 1 ,Human ,Humans ,Interferon Regulatory Factor-3 ,Interferon Type I ,Interferon-beta ,Mutagenesis ,Receptor ,Interferon alpha-beta ,Viral Proteins ,HSV-1 ,IRF-3 ,UL42 ,interferon ,Agricultural and Veterinary Sciences ,Medical and Health Sciences ,Virology ,Agricultural ,veterinary and food sciences ,Biological sciences ,Biomedical and clinical sciences - Abstract
In spite of several decades of research focused on understanding the biology of human herpes simplex virus 1 (HSV-1), no tool has been developed to study its genome in a high-throughput fashion. Here, we describe the creation of a transposon insertion mutant library of the HSV-1 genome. Using this tool, we aimed to identify novel viral regulators of type I interferon (IFN-I). HSV-1 evades the host immune system by encoding viral proteins that inhibit the type I interferon response. Applying differential selective pressure, we identified the three strongest viral IFN-I regulators in HSV-1. We report that the viral polymerase processivity factor UL42 interacts with the host transcription factor IFN regulatory factor 3 (IRF-3), inhibiting its phosphorylation and downstream beta interferon (IFN-β) gene transcription. This study represents a proof of concept for the use of high-throughput screening of the HSV-1 genome in investigating viral biology and offers new targets both for antiviral therapy and for oncolytic vector design.IMPORTANCE This work is the first to report the use of a high-throughput mutagenesis method to study the genome of HSV-1. We report three novel viral proteins potentially involved in regulating the host type I interferon response. We describe a novel mechanism by which the viral protein UL42 is able to suppress the production of beta interferon. The tool we introduce in this study can be used to study the HSV-1 genome in great detail to better understand viral gene functions.
- Published
- 2019
6. Interferon Regulatory Factor 3 Supports the Establishment of Chronic Gammaherpesvirus Infection in a Route- and Dose-Dependent Manner.
- Author
-
Johnson, K. E., Sylvester, P. A., Jondle, C. N., Aurubin, C. A., and Tarakanova, V. L.
- Subjects
- *
INTERFERON regulatory factors , *B cell differentiation , *B cells , *VIRAL nonstructural proteins , *VIRAL genetics , *PROTEIN kinases , *TYPE I interferons - Abstract
Gammaherpesviruses are ubiquitous pathogens that establish lifelong infections and are associated with several malignancies, including B cell lymphomas. Uniquely, these viruses manipulate B cell differentiation to establish long-term latency in memory B cells. This study focuses on the interaction between gammaherpesviruses and interferon regulatory factor 3 (IRF-3), a ubiquitously expressed transcription factor with multiple direct target genes, including beta interferon (IFN-β), a type I IFN. IRF-3 attenuates acute replication of a plethora of viruses, including gammaherpesvirus. Furthermore, IRF-3-driven IFN-β expression is antagonized by the conserved gammaherpesvirus protein kinase during lytic virus replication in vitro. In this study, we have uncovered an unexpected proviral role of IRF-3 during chronic gammaherpesvirus infection. In contrast to the antiviral activity of IRF-3 during acute infection, IRF-3 facilitated establishment of latent gammaherpesvirus infection in B cells, particularly, germinal center and activated B cells, the cell types critical for both natural infection and viral lymphomagenesis. This proviral role of IRF-3 was further modified by the route of infection and viral dose. Furthermore, using a combination of viral and host genetics, we show that IRF-3 deficiency does not rescue attenuated chronic infection of a protein kinase null gammaherpesvirus mutant, highlighting the multifunctional nature of the conserved gammaherpesvirus protein kinases in vivo. In summary, this study unveils an unexpected proviral nature of the classical innate immune factor, IRF-3, during chronic virus infection. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
7. Orostachys japonicus extract inhibits the lipopolysaccharide‐induced pro‐inflammatory factors by suppression of transcription factors.
- Author
-
Lee, Hyeong‐Seon
- Subjects
- *
TRANSCRIPTION factors , *DICHLOROMETHANE , *INFLAMMATORY mediators , *ETHYL acetate , *ORGANIC solvents , *PROTEIN expression - Abstract
Orostachys japonicus (O. japonicus) was extracted with ethanol (EtOH) and sequentially separated with organic solvents, including n‐hexane (Hex), dichloromethane (DCM), ethyl acetate (EtOAc), n‐butanol (BuOH), and water (H2O). All the fractions were confirmed for anti‐inflammatory activity in an inflammatory condition. The DCM fraction showed the highest anti‐inflammatory ability. Here, we examined the effect of DCM fraction and investigated the intracellular signaling pathways in LPS‐stimulated RAW 264.7 macrophage cells. The DCM fraction significantly inhibited the mRNA levels of pro‐inflammatory mediators and cytokines including iNOS, COX‐2, IL‐1β, IL‐2, IL‐6, and IP‐10 in LPS‐stimulated cells. Also, the treatment of DCM fraction excellently reduced the expression of the proteins of AP‐1 (phospho‐c‐Jun and phospho‐c‐Fos) and phospho‐IRF3 as transcription factors. As a result, it suppressed LPS‐induced inflammatory mediator and cytokines via inhibition of transcription factors. In conclusion, our data demonstrated that DCM fraction has a strong anti‐inflammatory activity that improves the inflammatory state. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
8. Priming Phosphorylation of TANK-Binding Kinase 1 by IκB Kinase β Is Essential in Toll-Like Receptor 3/4 Signaling.
- Author
-
Hiroto Abe, Junko Satoh, Yutaro Shirasaka, Amane Kogure, Hiroki Kato, Shinji Ito, and Takashi Fujita
- Subjects
- *
TOLL-like receptors , *INTERFERON regulatory factors , *ADAPTOR proteins , *DELETION mutation , *PHOSPHORYLATION , *DOUBLE-stranded RNA - Abstract
TRIF is an essential adaptor for Toll-like receptor 3/4 (TLR3/4) signaling to activate transcription factor interferon regulatory factor 3 (IRF-3). We examined the molecular mechanism of TLR3 signaling and found that TLR3 stimulation by double-stranded RNA (dsRNA) induces phosphorylation of TRIF at Ser210 and is required for IRF-3 recruitment. TANK-binding kinase 1 (TBK1) is known to be responsible for IRF-3 phosphorylation and activation. We found that TBK1 is also responsible for phosphorylation of Ser210 in TRIF. Unexpectedly, we discovered that IB kinase (IKKβ) plays an essential role in TLR3/4 signaling using a pharmacological inhibitor and gene deletion. Of note, IKKβ is essential in TLR3/4 but not in retinoic acidinducible gene I (RIG-I) signaling. Mechanistically, IKKβ transiently associates with and induces the phosphorylation of TBK1 upon TLR3 stimulation. These results suggest a phosphorylation cascade of IKKβ and TBK1, where priming phosphorylation of TBK1 by IKKβ is required to surpass the threshold to induce signaling, thereby activating IRF-3. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
9. Vaccinia Virus Expressing Interferon Regulatory Factor 3 Induces Higher Protective Immune Responses against Lethal Poxvirus Challenge in Atopic Organism
- Author
-
Hana Pilna, Vera Hajkova, Jarmila Knitlova, Jana Liskova, Jana Elsterova, and Zora Melkova
- Subjects
IRF-3 ,vaccinia virus ,smallpox ,atopic dermatitis ,eczema vaccinatum ,immunization ,Microbiology ,QR1-502 - Abstract
Vaccinia virus (VACV) is an enveloped DNA virus from the Orthopoxvirus family, various strains of which were used in the successful eradication campaign against smallpox. Both original and newer VACV-based replicating vaccines reveal a risk of serious complications in atopic individuals. VACV encodes various factors interfering with host immune responses at multiple levels. In atopic skin, the production of type I interferon is compromised, while VACV specifically inhibits the phosphorylation of the Interferon Regulatory Factor 3 (IRF-3) and expression of interferons. To overcome this block, we generated a recombinant VACV-expressing murine IRF-3 (WR-IRF3) and characterized its effects on virus growth, cytokine expression and apoptosis in tissue cultures and in spontaneously atopic Nc/Nga and control Balb/c mice. Further, we explored the induction of protective immune responses against a lethal dose of wild-type WR, the surrogate of smallpox. We demonstrate that the overexpression of IRF-3 by WR-IRF3 increases the expression of type I interferon, modulates the expression of several cytokines and induces superior protective immune responses against a lethal poxvirus challenge in both Nc/Nga and Balb/c mice. Additionally, the results may be informative for design of other virus-based vaccines or for therapy of different viral infections.
- Published
- 2021
- Full Text
- View/download PDF
10. Rotavirus NSP1 Inhibits Type I and Type III Interferon Induction
- Author
-
Gennaro Iaconis, Ben Jackson, Kay Childs, Mark Boyce, Stephen Goodbourn, Neil Blake, Miren Iturriza-Gomara, and Julian Seago
- Subjects
rotavirus ,type I interferon ,type III interferon ,NSP1 ,IRF-1 ,IRF-3 ,Microbiology ,QR1-502 - Abstract
Type I interferons (IFNs) are produced by most cells in response to virus infection and stimulate a program of anti-viral gene expression in neighboring cells to suppress virus replication. Type III IFNs have similar properties, however their effects are limited to epithelial cells at mucosal surfaces due to restricted expression of the type III IFN receptor. Rotavirus (RV) replicates in intestinal epithelial cells that respond predominantly to type III IFNs, and it has been shown that type III rather than type I IFNs are important for controlling RV infections in vivo. The RV NSP1 protein antagonizes the host type I IFN response by targeting IRF-3, IRF-5, IRF-7, or β-TrCP for proteasome-mediated degradation in a strain-specific manner. Here we provide the first demonstration that NSP1 proteins from several human and animal RV strains antagonize type III as well as type I IFN induction. We also show that NSP1 is a potent inhibitor of IRF-1, a previously undescribed property of NSP1 which is conserved among human and animal RVs. Interestingly, all NSP1 proteins were substantially more effective inhibitors of IRF-1 than either IRF-3 or IRF-7 which has significance for evasion of basal anti-viral immunity and type III IFN induction in the intestinal epithelium.
- Published
- 2021
- Full Text
- View/download PDF
11. The Intrinsically Disordered W Protein Is Multifunctional during Henipavirus Infection, Disrupting Host Signalling Pathways and Nuclear Import
- Author
-
Sofiya Tsimbalyuk, Emily M. Cross, Mikayla Hoad, Camilla M. Donnelly, Justin A. Roby, and Jade K. Forwood
- Subjects
intrinsically disordered ,W protein ,henipaviruses ,STAT ,IRF-3 ,Cytology ,QH573-671 - Abstract
Nipah and Hendra viruses are highly pathogenic, zoonotic henipaviruses that encode proteins that inhibit the host’s innate immune response. The W protein is one of four products encoded from the P gene and binds a number of host proteins to regulate signalling pathways. The W protein is intrinsically disordered, a structural attribute that contributes to its diverse host protein interactions. Here, we review the role of W in innate immune suppression through inhibition of both pattern recognition receptor (PRR) pathways and interferon (IFN)-responsive signalling. PRR stimulation leading to activation of IRF-3 and IFN release is blocked by henipavirus W, and unphosphorylated STAT proteins are sequestered within the nucleus of host cells by W, thereby inhibiting the induction of IFN stimulated genes. We examine the critical role of nuclear transport in multiple functions of W and how specific binding of importin-alpha (Impα) isoforms, and the 14-3-3 group of regulatory proteins suggests further modulation of these processes. Overall, the disordered nature and multiple functions of W warrant further investigation to understand henipavirus pathogenesis and may reveal insights aiding the development of novel therapeutics.
- Published
- 2020
- Full Text
- View/download PDF
12. Regulation of 8-Hydroxydaidzein in IRF3-Mediated Gene Expression in LPS-Stimulated Murine Macrophages
- Author
-
Nur Aziz, Young-Gyu Kang, Yong-Jin Kim, Won-Seok Park, Deok Jeong, Jongsung Lee, Donghyun Kim, and Jae Youl Cho
- Subjects
8-hydroxydaidzein ,irf-3 ,traf3 ,ikkε ,inflammation ,Microbiology ,QR1-502 - Abstract
Cytokines and chemokines are transcriptionally regulated by inflammatory transcription factors such as nuclear factor-κB (NF-κB), activator protein-1 (AP-1), and interferon regulatory factor (IRF)-3. A daidzein derivative compound, 8-hydroxydaidzein (8-HD), isolated from soy products, has recently gained attention due to various pharmacological benefits, including anti-inflammatory activities. However, regulation of the inflammatory signaling mechanism for 8-HD is still poorly understood, particularly with respect to the IRF-3 signaling pathway. In this study, we explored the molecular mechanism of 8-HD in regulating inflammatory processes, with a focus on the IRF-3 signaling pathway using a lipopolysaccharide (LPS) and polyinosinic:polycytidylic acid [Poly (I:C)] stimulated murine macrophage cell line (RAW264.7). The 8-HD downregulated the mRNA expression level of IRF-3-dependent genes by inhibiting phosphorylation of the IRF-3 transcription factor. The inhibitory mechanism of 8-HD in the IRF-3 signaling pathway was shown to inhibit the kinase activity of IKKε to phosphorylate IRF-3. This compound can also interfere with the TRIF-mediated complex formation composed of TRAF3, TANK, and IKKε leading to downregulation of AKT phosphorylation and reduction of IRF-3 activation, resulted in inhibition of IRF-3-dependent expression of genes including IFN-β, C-X-C motif chemokine 10 (CXCL10), and interferon-induced protein with tetratricopeptide repeats 1 (IFIT1). Therefore, these results strongly suggest that 8-HD can act as a promising compound with the regulatory function of IRF-3-mediated inflammatory responses.
- Published
- 2020
- Full Text
- View/download PDF
13. Hesperetin Prevents Bone Resorption by Inhibiting RANKL-Induced Osteoclastogenesis and Jnk Mediated Irf-3/c-Jun Activation
- Author
-
Qiang Zhang, Xinqiao Tang, Zhong Liu, Xiaoxia Song, Dan Peng, Wei Zhu, Zhengxiao Ouyang, and Wanchun Wang
- Subjects
osteoporosis ,hesperetin ,RANKL ,Irf-3 ,MAPK ,NFATc-1 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Bone homeostasis and resorption is regulated by the proper activation of osteoclasts, whose stimulation largely depends on the receptor activator of nuclear factor κB ligand (RANKL)-RANK signaling. Herein, for the first time, we showed that interferon regulatory factor (Irf)-3 was intimately involved in RANKL-induced osteoclast formation. In addition, hesperetin (Hes) derived from citrus fruit could inhibit RANKL-induced osteoclast differentiation and maturation among three types of osteoclast precursors with inhibited formation of F-actin rings and resorption pits on bone slices. More importantly, by using SP600125, a selective Jnk inhibitor, we showed that Hes was able to significantly attenuate the Jnk downstream expression of Irf-3 and c-Jun, thereby inactivating NF-κB/MAPK signaling and transcriptional factor NFATc-1, leading to suppression of osteoclast-specific genes, which resulted in impaired osteoclastogenesis and functionality. An ovariectomized (OVX) osteoporosis mouse model demonstrated that Hes could increase trabecular bone volume fractions (BV/TV), trabecular thickness, and trabecular number, whereas it decreased trabecular separation in OVX mice with well-preserved trabecular bone architecture and decreased levels of TRAP-positive osteoclasts. This is further evidenced by the diminished serum expression of bone resorption marker CTX and enhanced production of osteoblastic ALP in vivo. Taken together, these results suggested that Hes could inhibit Jnk-mediated Irf-3/c-Jun activation, thus attenuating RANKL-induced osteoclast formation and function both in vitro and in vivo.
- Published
- 2018
- Full Text
- View/download PDF
14. Azithromycin and glucosamine may amplify the type 1 interferon response to RNA viruses in a complementary fashion.
- Author
-
DiNicolantonio, James J., Barroso-Aranda, Jorge, and McCarty, Mark F.
- Subjects
- *
TYPE I interferons , *AZITHROMYCIN , *GLUCOSAMINE , *RNA virus infections , *RNA viruses - Abstract
• Azithromycin can boost the ability of RNA viruses to induce type1 interferon by amplifying the expression and virally-mediated activation of MD5. cc O-GlcNAcylation of MAVS, a downstream target of MDA5, renders it more effective for type 1 interferon induction. • High-dose glucosamine administration up-regulates O-GlcNAcylation by increasing the cellular pool of UDP-N-acetylglucosamine. • Joint administration of azithromycin and high-dose glucosamine, early in the course of RNA infection, may synergize to boost interferon induction. Previous research demonstrates that, in clinically relevant concentrations, azithromycin can boost the ability of RNA viruses to induce type 1 interferon by amplifying the expression and virally-mediated activation of MDA5. O-GlcNAcylation of MAVS, a down-stream target of MDA5, renders it more effective for type 1 interferon induction. High-dose glucosamine administration up-regulates O-GlcNAcylation by increasing the cellular pool of UDP -N- acetylglucosamine. Hence, it is proposed that joint administration of azithromycin and high-dose glucosamine, early in the course of RNA virus infections, may interact in a complementary fashion to aid their control by enhancing type 1 interferon induction [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
15. Hesperetin Prevents Bone Resorption by Inhibiting RANKL-Induced Osteoclastogenesis and Jnk Mediated Irf-3/c-Jun Activation.
- Author
-
Zhang, Qiang, Tang, Xinqiao, Liu, Zhong, Song, Xiaoxia, Peng, Dan, Zhu, Wei, Ouyang, Zhengxiao, and Wang, Wanchun
- Subjects
BONE resorption ,TRANCE protein ,OSTEOCLASTOGENESIS ,OVARIECTOMY ,CANCELLOUS bone - Abstract
Bone homeostasis and resorption is regulated by the proper activation of osteoclasts, whose stimulation largely depends on the receptor activator of nuclear factor κB ligand (RANKL)-RANK signaling. Herein, for the first time, we showed that interferon regulatory factor (Irf)-3 was intimately involved in RANKL-induced osteoclast formation. In addition, hesperetin (Hes) derived from citrus fruit could inhibit RANKL-induced osteoclast differentiation and maturation among three types of osteoclast precursors with inhibited formation of F-actin rings and resorption pits on bone slices. More importantly, by using SP600125, a selective Jnk inhibitor, we showed that Hes was able to significantly attenuate the Jnk downstream expression of Irf-3 and c-Jun, thereby inactivating NF-κB/MAPK signaling and transcriptional factor NFATc-1, leading to suppression of osteoclast-specific genes, which resulted in impaired osteoclastogenesis and functionality. An ovariectomized (OVX) osteoporosis mouse model demonstrated that Hes could increase trabecular bone volume fractions (BV/TV), trabecular thickness, and trabecular number, whereas it decreased trabecular separation in OVX mice with well-preserved trabecular bone architecture and decreased levels of TRAP-positive osteoclasts. This is further evidenced by the diminished serum expression of bone resorption marker CTX and enhanced production of osteoblastic ALP in vivo. Taken together, these results suggested that Hes could inhibit Jnk-mediated Irf-3/c-Jun activation, thus attenuating RANKL-induced osteoclast formation and function both in vitro and in vivo. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
16. Defining the molecular role of RNA helicase DDX3 in antiviral signaling pathways
- Author
-
SAIKRUANG, WILAIPORN and SAIKRUANG, WILAIPORN
- Published
- 2022
17. RNAヘリカーゼDDX3の抗ウイルス性シグナル伝達経路における分子的役割の解明
- Author
-
SAIKRUANG, WILAIPORN, 野田, 岳志, 鈴木, 淳, and 高田, 穣
- Subjects
IRF-3 ,DDX3 ,virus infection ,type I interferon ,innate immunity - Published
- 2022
18. Oral cancer-derived exosomal NAP1 enhances cytotoxicity of natural killer cells via the IRF-3 pathway.
- Author
-
Wang, Yingnan, Qin, Xing, Zhu, Xueqin, Chen, Wanjun, Zhang, Jianjun, and Chen, Wantao
- Subjects
- *
ORAL cancer risk factors , *EXOSOMES , *KILLER cells , *INTERFERON regulatory factors , *TRANSCRIPTION factors , *TYPE I interferons - Abstract
Objective: To examine the effects of oral cancer-derived exosomes (OCEXs) on natural killer (NK) cells and to explore the underlying mechanism.Materials and Methods: OCEXs were isolated from the cell culture supernatant of oral cancer (OC) cells using ultrafiltration and affinity chromatography and were identified using electron microscopy, nanoparticle tracking analysis (NTA) and immunoblotting. The effects of OCEXs on NK cells were analyzed using laser scanning confocal microscopy and several functional assays of NK cells. To explore the mechanism of their effects, antibody array, protein mass spectrometry and RNA interference were adopted.Results: The particles isolated from the OC cells were identified as exosomes with satisfactory morphology, concentration and purity. The OCEXs were internalized by NK cells and then promoted the biological functions of NK cells, including proliferation, release of perforin and granzyme M and cytotoxicity. Furthermore, OCEXs increased the expression of interferon regulatory factor 3 (IRF-3) and its phosphorylation, which drove the expression of the type I interferon (IFN) gene and the chemokine (C-X-C motif) ligand (CXCL) genes, thereby promoting the functions of NK cells. In addition, NF-κB-activating kinase-associated protein 1 (NAP1), an upstream activator of IRF-3, was enriched in OCEXs, and treatment with OCEXs increased the expression of NAP1 in NK cells. Importantly, NAP1-depleted OCEXs obtained from OC cells had a dramatically weakened influence on NK cells.Conclusion: Our findings reveal a previously unrecognized function of exosomal NAP1 derived from OC cells in enhancing the cytotoxicity of NK cells via the IRF-3 pathway. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
19. Activation of IRF3 contributes to IFN-γ and ISG54 expression during the immune responses to B16F10 tumor growth.
- Author
-
Guinn, Zachary, Brown, Deborah M., and Petro, Thomas M.
- Subjects
- *
INTERFERON regulatory factors , *TUMOR growth , *TRANSFORMING growth factors , *TRANSCRIPTION factors , *LUCIFERASES - Abstract
Interferon Regulatory Factor (IRF-3) has been shown to contribute to immune control of B16 melanoma tumor growth. We have shown previously that IRF-3 has a role in IFN-γ-induced expression of pro-apoptotic interferon stimulated gene 54 (ISG54) in macrophages and IFN-γ in T cells. To investigate the IRF3-IFN-γ-ISG54 nexus, we injected C57Bl/6 (B6) and IRF3KO mice s.c. with luciferase-producing B16-F10 tumor cells. Tumor growth as measured by luciferase levels was similar between B6 and IRF3KO mice at days 2 and 6, but was significantly greater at day 9 in IRF3KO mice compared with B6 mice. Transcription factor assays on splenic protein extracts after tumor inoculation revealed peak activation of IRF3 and IRF7 at day 6 in B6 tumor-bearing mice but not in IRF3KO tumor-bearing mice. Likewise, significant induction of IFN-γ occurred in spleens and tumors in B6 mice from days 6–9 but failed to occur in tumor-bearing IRF3KO mice. Previous reports from other labs showed that the anti-tumor properties of IFN-γ are the result of cell cycle arrest. Using B16F1 cells or B16F1 cells deficient in IFN-γ receptor (B16-IRFGRKO), we found that IFN-γ alone and in synergy with the TLR3/IRF3 agonists, poly I:C, decreased B16F1 cell growth in significant correlation with increased ISG54 expression. Moreover, IFN-γ alone increased expression of the cell cycle inhibitor, p27Kip while IFN-γ plus poly I:C increased cleaved Caspase-3 in B16 cells. Thus, it is likely that an IFN-γ/IRF3/ISG54 nexus can significantly contribute to tumor cell control during anti-tumor immune responses. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
20. Hydroquinone suppresses IFN-β expression by targeting AKT/IRF3 pathway.
- Author
-
Yong Kim, Han Gyung Kim, Sang Yun Han, Deok Jeong, Woo Seok Yang, Jung-Il Kim, Ji Hye Kim, Young-Su Yi, and Jae Youl Cho
- Subjects
- *
HYDROQUINONE , *BENZENE compounds , *IMMUNE response , *INTERFERONS , *PROTEIN kinase B , *PHOSPHORYLATION - Abstract
Previous studies have demonstrated the role of hydroquinone (HQ), a hydroxylated benzene metabolite, in modulating various immune responses; however, its role in macrophage-mediated inflammatory responses is not fully understood. In this study, the role of HQ in inflammatory responses and the underlying molecular mechanism were explored in macrophages. HQ down-regulated the expression of interferon (IFN)-β mRNA in LPS-stimulated RAW264.7 cells without any cytotoxicity and suppressed interferon regulatory factor (IRF)-3-mediated luciferase activity induced by TIR-domain-containing adapter-inducing interferon-β (TRIF) and TANK-binding kinase 1 (TBK1). A mechanism study revealed that HQ inhibited IRF-3 phosphorylation induced by lipopolysaccharide (LPS), TRIF, and AKT by suppressing phosphorylation of AKT, an upstream kinase of the IRF-3 signaling pathway. IRF-3 phosphorylation is highly induced by wild-type AKT and poorly induced by an AKT mutant, AKT C310A, which is mutated at an inhibitory target site of HQ. We also showed that HQ inhibited IRF-3 phosphorylation by targeting all three AKT isoforms (AKT1, AKT2, and AKT3) in RAW264.7 cells and suppressed IRF-3-mediated luciferase activities induced by AKT in HEK293 cells. Taken together, these results strongly suggest that HQ inhibits the production of a type I IFN, IFN-β, by targeting AKTs in the IRF-3 signaling pathway during macrophage-mediated inflammation. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
21. SARS-CoV-2 Variants Show Different Host Cell Proteome Profiles With Delayed Immune Response Activation in Omicron-Infected Cells.
- Author
-
Metzler M, Tharyan RG, Klann K, Grikscheit K, Bojkova D, Cinatl J, Tascher G, Ciesek S, and Münch C
- Subjects
- Humans, Proteome, Pandemics, Antiviral Agents, Antibodies, Neutralizing, SARS-CoV-2, COVID-19
- Abstract
The ancestral SARS-CoV-2 strain that initiated the Covid-19 pandemic at the end of 2019 has rapidly mutated into multiple variants of concern with variable pathogenicity and increasing immune escape strategies. However, differences in host cellular antiviral responses upon infection with SARS-CoV-2 variants remain elusive. Leveraging whole-cell proteomics, we determined host signaling pathways that are differentially modulated upon infection with the clinical isolates of the ancestral SARS-CoV-2 B.1 and the variants of concern Delta and Omicron BA.1. Our findings illustrate alterations in the global host proteome landscape upon infection with SARS-CoV-2 variants and the resulting host immune responses. Additionally, viral proteome kinetics reveal declining levels of viral protein expression during Omicron BA.1 infection when compared to ancestral B.1 and Delta variants, consistent with its reduced replication rates. Moreover, molecular assays reveal deferral activation of specific host antiviral signaling upon Omicron BA.1 and BA.2 infections. Our study provides an overview of host proteome profile of multiple SARS-CoV-2 variants and brings forth a better understanding of the instigation of key immune signaling pathways causative for the differential pathogenicity of SARS-CoV-2 variants., Competing Interests: Conflict of interest The authors declare no competing interests., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
22. Scoparone Inhibits LPS-Simulated Inflammatory Response by Suppressing IRF3 and ERK in BV-2 Microglial Cells.
- Author
-
Duk-Yeon Cho, Hyun Myung Ko, Joonsoo Kim, Byung-Wook Kim, Yo-Sep Yun, Jeong-In Park, Ganesan, Palanivel, Jin-Tae Lee, and Dong-Kug Choi
- Subjects
- *
ALZHEIMER'S disease treatment , *PHYSIOLOGICAL effects of lipopolysaccharides , *CELL analysis , *PHOSPHORYLATION kinetics , *BIOACTIVE compounds - Abstract
Microglia activation and the release of various inflammatory cytokines are largely related to neurological diseases, including Parkinson's, Alzheimer's, and other brain diseases. The suppression of microglial cells using natural bioactive compounds has become increasingly important for brain therapy owing to the expected beneficial effect of lower toxicity. Scoparone (6,7-dimethoxycoumarin), a major bioactive compound found in various plant parts, including the inner shell of chestnut (Castanea crenata), was evaluated on lipopolysaccharide (LPS)-activated BV-2 microglia cells. The results indicated that scoparone suppresses the LPS-stimulated increase of neuroinflammatory responses and inhibited the pro-inflammatory cytokine production in the BV-2 microglial cells. A mechanistic study showed that scoparone specifically inhibited the LPS-stimulated activation via a major regulation of IRF-3 and a regulation of ERK, whereby the phosphorylation in the BV-2 microglial cells is blocked. These data suggest that scoparone has anti-neuroinflammatory effects in LPS-activated BV-2 microglial cells, and could possibly be used in the development of novel drugs for the prevention and treatment of neuroinflammatory diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
23. Clarithromycin prevents human respiratory syncytial virus-induced airway epithelial responses by modulating activation of interferon regulatory factor-3.
- Author
-
Yamamoto, Keisuke, Yamamoto, Soh, Ogasawara, Noriko, Takano, Kenichi, Shiraishi, Tsukasa, Sato, Toyotaka, Miyata, Ryo, Kakuki, Takuya, Kamekura, Ryuta, Kojima, Takashi, Tsutsumi, Hiroyuki, Himi, Tetsuo, and Yokota, Shin-ichi
- Subjects
- *
CLARITHROMYCIN , *RESPIRATORY syncytial virus infections , *INTERFERON regulatory factors , *IMMUNOMODULATORS , *MACROLIDE antibiotics , *TOLL-like receptors , *PREVENTION , *THERAPEUTICS - Abstract
Macrolide antibiotics exert immunomodulatory activity by reducing pro-inflammatory cytokine production by airway epithelial cells, fibroblasts, vascular endothelial cells, and immune cells. However, the underlying mechanism of action remains unclear. Here, we examined the effect of clarithromycin (CAM) on pro-inflammatory cytokine production, including interferons (IFNs), by primary human nasal epithelial cells and lung epithelial cell lines (A549 and BEAS-2B cells) after stimulation by Toll-like receptor (TLR) and RIG-I-like receptor (RLR) agonists and after infection by human respiratory syncytial virus (RSV). CAM treatment led to a significant reduction in poly I:C- and RSV-mediated IL-8, CCL5, IFN-β and -λ production. Furthermore, IFN-β promoter activity (activated by poly I:C and RSV infection) was significantly reduced after treatment with CAM. CAM also inhibited IRF-3 dimerization and subsequent translocation to the nucleus. We conclude that CAM acts a crucial modulator of the innate immune response, particularly IFN production, by modulating IRF-3 dimerization and subsequent translocation to the nucleus of airway epithelial cells. This newly identified immunomodulatory action of CAM will facilitate the discovery of new macrolides with an anti-inflammatory role. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
24. Tick-borne encephalitis virus induces chemokine RANTES expression via activation of IRF-3 pathway.
- Author
-
Xiaowei Zhang, Zhenhua Zheng, Xijuan Liu, Bo Shu, Panyong Mao, Bingke Bai, Qinxue Hu, Minhua Luo, Xiaohe Ma, Zongqiang Cui, Hanzhong Wang, Zhang, Xiaowei, Zheng, Zhenhua, Liu, Xijuan, Shu, Bo, Mao, Panyong, Bai, Bingke, Hu, Qinxue, Luo, Minhua, and Ma, Xiaohe
- Subjects
- *
TICK-borne encephalitis viruses , *CHEMOKINES , *CYTOKINES , *PROTEIN microarrays , *MONOCLONAL antibodies , *CCL5 (Chemokine) , *LABORATORY mice , *PROTEIN metabolism , *ANIMAL experimentation , *ANIMALS , *BRAIN , *CELL lines , *CELLULAR signal transduction , *EPIDEMIC encephalitis , *FLAVIVIRUSES , *GENE expression , *MICE , *PROTEINS , *VIRAL load ,BRAIN metabolism - Abstract
Background: Tick-borne encephalitis virus (TBEV) is one of the most important flaviviruses that targets the central nervous system (CNS) and causes encephalitides in humans. Although neuroinflammatory mechanisms may contribute to brain tissue destruction, the induction pathways and potential roles of specific chemokines in TBEV-mediated neurological disease are poorly understood.Methods: BALB/c mice were intracerebrally injected with TBEV, followed by evaluation of chemokine and cytokine profiles using protein array analysis. The virus-infected mice were treated with the CC chemokine antagonist Met-RANTES or anti-RANTES mAb to determine the role of RANTES in affecting TBEV-induced neurological disease. The underlying signaling mechanisms were delineated using RANTES promoter luciferase reporter assay, siRNA-mediated knockdown, and pharmacological inhibitors in human brain-derived cell culture models.Results: In a mouse model, pathological features including marked inflammatory cell infiltrates were observed in brain sections, which correlated with a robust up-regulation of RANTES within the brain but not in peripheral tissues and sera. Antagonizing RANTES within CNS extended the survival of mice and reduced accumulation of infiltrating cells in the brain after TBEV infection. Through in vitro studies, we show that virus infection up-regulated RANTES production at both mRNA and protein levels in human brain-derived cell lines and primary progenitor-derived astrocytes. Furthermore, IRF-3 pathway appeared to be essential for TBEV-induced RANTES production. Site mutation of an IRF-3-binding motif abrogated the RANTES promoter activity in virus-infected brain cells. Moreover, IRF-3 was activated upon TBEV infection as evidenced by phosphorylation of TBK1 and IRF-3, while blockade of IRF-3 activation drastically reduced virus-induced RANTES expression.Conclusions: Our findings together provide insights into the molecular mechanism underlying RANTES production induced by TBEV, highlighting its potential importance in the process of neuroinflammatory responses to TBEV infection. [ABSTRACT FROM AUTHOR]- Published
- 2016
- Full Text
- View/download PDF
25. Nonstructural protein p39 of feline calicivirus suppresses host innate immune response by preventing IRF-3 activation.
- Author
-
Yumiketa, Yo, Narita, Takanori, Inoue, Yosuke, Sato, Go, Kamitani, Wataru, Oka, Tomoichiro, Katayama, Kazuhiko, Sakaguchi, Takemasa, and Tohya, Yukinobu
- Subjects
- *
VIRAL nonstructural proteins , *CALICIVIRUSES , *NATURAL immunity , *HOSTS (Biology) , *IMPULSE response - Abstract
Feline calicivirus (FCV) is an important veterinary pathogen that causes acute upper respiratory tract diseases and, occasionally, highly contagious febrile hemorrhagic syndrome in cats. Many viruses have adopted mechanisms for evading IFN-α/β signaling, particularly by directly or indirectly suppressing activation of IRF-3. In this study, we investigated whether nonstructural proteins of FCV possess these mechanisms. When p39, a nonstructural protein of FCV, was transiently expressed in 293T cells, it suppressed IFN-β and ISG15 mRNA production induced by dsRNA. Expression of p39 also suppressed phosphorylation and dimerization of IRF-3 induced by dsRNA. These results suggest that p39 suppresses type 1 IFN production by preventing IRF-3 activation. This may become an important factor in understanding the pathogenesis and virulence of FCV. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
26. Targeting 7-dehydrocholesterol reductase against EV-A71 replication by upregulating interferon response.
- Author
-
Wang, Huiqiang, Cui, Boming, Yan, Haiyan, Wu, Shuo, Wang, Kun, Yang, Ge, Jiang, Jiandong, and Li, Yuhuan
- Subjects
- *
INTERFERONS - Published
- 2023
- Full Text
- View/download PDF
27. Proteolysis of MDA5 and IPS-1 is not required for inhibition of the type I IFN response by poliovirus.
- Author
-
Kotla, Swathi and Gustin, Kurt E.
- Subjects
- *
PROTEOLYSIS , *POLIOVIRUS , *INTERFERONS , *CYTOPLASM , *IMMUNOFLUORESCENCE , *IMMUNOBLOTTING - Abstract
Background: The type I interferon (IFN) response is a critical component of the innate immune response to infection by RNA viruses and is initiated via recognition of viral nucleic acids by RIG-like receptors (RLR). Engagement of these receptors in the cytoplasm initiates a signal transduction pathway leading to activation of the transcription factors NF-κB, ATF-2 and IRF-3 that coordinately upregulate transcription of type I IFN genes, such as that encoding IFN-β. In this study the impact of poliovirus infection on the type I interferon response has been examined. Methods: The type I IFN response was assessed by measuring IFN-β mRNA levels using qRT-PCR and normalizing to levels of β-actin mRNA. The status of host factors involved in activation of the type I IFN response was examined by immunoblot, immunofluorescence microcopy and qRT-PCR. Results: The results show that poliovirus infection results in induction of very low levels of IFN-β mRNA despite clear activation of NF-κB and ATF-2. In contrast, analysis of IRF-3 revealed no transcriptional induction of an IRF-3-responsive promoter or homodimerization of IRF-3 indicating it is not activated in poliovirus-infected cells. Exposure of poliovirus-infected cells to poly(I:C) results in lower levels of IFN-β mRNA synthesis and IRF-3 activation compared to mock-infected cells. Analysis of MDA-5 and IPS-1 revealed that these components of the RLR pathway were largely intact at times when the type I IFN response was suppressed. Conclusions: Collectively, these results demonstrate that poliovirus infection actively suppresses the host type I interferon response by blocking activation of IRF-3 and suggests that this is not mediated by cleavage of MDA-5 or IPS-1. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
28. TLR3 engagement induces IRF-3-dependent apoptosis in androgen-sensitive prostate cancer cells and inhibits tumour growth in vivo.
- Author
-
Gambara, Guido, Desideri, Marianna, Stoppacciaro, Antonella, Padula, Fabrizio, De Cesaris, Paola, Starace, Donatella, Tubaro, Andrea, Bufalo, Donatella, Filippini, Antonio, Ziparo, Elio, and Riccioli, Anna
- Subjects
CANCER treatment ,TUMOR growth ,MEMBRANE proteins ,TOLL-like receptors ,PROTEIN expression ,APOPTOSIS - Abstract
Toll-like receptors ( TLRs) are a family of highly conserved transmembrane proteins expressed in epithelial and immune cells that recognize pathogen associated molecular patterns. Besides their role in immune response against infections, numerous studies have shown an important role of different TLRs in cancer, indicating these receptors as potential targets for cancer therapy. We previously demonstrated that the activation of TLR3 by the synthetic double-stranded RNA analogue poly I:C induces apoptosis of androgen-sensitive prostate cancer ( PCa) LNCaP cells and, much less efficiently, of the more aggressive PC3 cell line. Therefore, in this study we selected LNCaP cells to investigate the mechanism of TLR3-mediated apoptosis and the in vivo efficacy of poly I:C-based therapy. We show that interferon regulatory factor-3 ( IRF-3) signalling plays an essential role in TLR3-mediated apoptosis in LNCaP cells through the activation of the intrinsic and extrinsic apoptotic pathways. Interestingly, hardly any apoptosis was induced by poly I:C in normal prostate epithelial cells RWPE-1. We also demonstrate for the first time the direct anticancer effect of poly I:C as a single therapeutic agent in a well-established human androgen-sensitive PCa xenograft model, by showing that tumour growth is highly impaired in poly I:C-treated immunodeficient mice. Immunohistochemical analysis of PCa xenografts highlights the antitumour role of poly I:C in vivo both on cancer cells and, indirectly, on endothelial cells. Notably, we show the presence of TLR3 and IRF-3 in both human normal and PCa clinical samples, potentially envisaging poly I:C-based therapy for PCa. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
29. Hantaan virus can infect human keratinocytes and activate an interferon response through the nuclear translocation of IRF-3.
- Author
-
Ye, Wei, Xu, Yongni, Wang, Yuan, Dong, Yangchao, Xi, Qianqian, Cao, Mengyuan, Yu, Lan, Zhang, Liang, Cheng, Linfeng, Wu, Xingan, Xu, Zhikai, Lei, Yingfeng, and Zhang, Fanglin
- Subjects
- *
KERATINOCYTES , *INTERFERONS , *CHROMOSOMAL translocation , *BUNYAVIRUSES , *HEMORRHAGIC fever with renal syndrome - Abstract
Hantaan virus (HTNV) is a rodent-borne virus that causes hemorrhagic fever with renal syndrome (HFRS) in Asia and can be transmitted to humans through bites or the inhalation of aerosolized urine, droppings, or saliva of infected rodents. Keratinocytes predominate in the epidermis and reportedly serve as a replication site for multiple vector-borne viruses, little is known about the susceptibility of human skin cells to HTNV infection. Therefore, we aimed to evaluate whether human keratinocytes support HTNV replication and elicit an immune response against HTNV infection. We found that a human keratinocyte cell line, HaCaT, supports HTNV replication. In addition, retinoic acid inducible gene-I (RIG-I) and melanoma differentiation associated gene-5 (MDA5) play key roles in the detection of HTNV infection in HaCaT cells and in the up-regulation of interferon (IFN)-β expression, which subsequently leads to the production of a large amount of antiviral interferon-stimulated genes (ISGs) and other chemokines used for immune cell recruitment. Furthermore, we suggest that interferon regulatory factor (IRF)-3, as opposed to NF-κB/p65 or IRF-7, is translocated to the nucleus to induce IFN-β. However, the early induction of chemokine CXCL10 was a direct result of HaCaT cells counteracting HTNV infection and was not due to the induction of IFN. Overall, our data demonstrate, for the first time, the permissiveness of human keratinocytes to HTNV infection. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
30. ATF-2/CREB/IRF-3-targeted anti-inflammatory activity of Korean red ginseng water extract.
- Author
-
Yang, Yanyan, Yang, Woo Seok, Yu, Tao, Sung, Gi-Ho, Park, Kye Won, Yoon, Keejung, Son, Young-Jin, Hwang, Hyunsik, Kwak, Yi-Seong, Lee, Chang-Muk, Rhee, Man Hee, Kim, Jong-Hoon, and Cho, Jae Youl
- Subjects
- *
GASTRITIS , *ENZYME metabolism , *ALTERNATIVE medicine , *ANIMAL experimentation , *ANTI-inflammatory agents , *APOPTOSIS , *BIOLOGICAL models , *BIOPHYSICS , *CARRIER proteins , *ENZYME inhibitors , *GINSENG , *INTERFERONS , *MACROPHAGES , *RESEARCH methodology , *MICE , *MOLECULAR biology , *NITRIC oxide , *PROTEIN kinases , *PLANT extracts , *DESCRIPTIVE statistics , *IN vitro studies , *PHARMACODYNAMICS , *PREVENTION - Abstract
Abstract: Ethnopharmacological relevance: Korean Red Ginseng (KRG) is one of the representative traditional herbal medicines prepared from Panax ginseng Meyer (Araliaceae) in Korea. It has been reported that KRG exhibits a lot of different biological actions such as anti-aging, anti-fatigue, anti-stress, anti-atherosclerosis, anti-diabetic, anti-cancer, and anti-inflammatory activities. Although systematic studies have investigated how KRG is able to ameliorate various inflammatory diseases, its molecular inhibitory mechanisms had not been carried out prior to this study. Materials and methods: In order to investigate these mechanisms, we evaluated the effects of a water extract of Korean Red Ginseng (KRG-WE) on the in vitro inflammatory responses of activated RAW264.7 cells, and on in vivo gastritis and peritonitis models by analyzing the activation events of inflammation-inducing transcription factors and their upstream kinases. Results: KRG-WE reduced the production of nitric oxide (NO), protected cells against NO-induced apoptosis, suppressed mRNA levels of inducible NO synthase (iNOS), cyclooxygenase (COX)-2, and interferon (IFN)-β, ameliorated EtOH/HCl-induced gastritis, and downregulated peritoneal exudate-derived NO production from lipopolysaccharide (LPS)-injected mice. The inhibition of these inflammatory responses by KRG-WE was regulated through the suppression of p38, c-Jun N-terminal kinase (JNK), and TANK-binding kinase 1 (TBK1) and by subsequent inhibition of activating transcription factor (ATF)-2, cAMP response element-binding protein (CREB), and IRF-3 activation. Of ginsensides included in this extract, interestingly, G-Rc showed the highest inhibitory potency on IRF-3-mediated luciferase activity. Conclusion: These results strongly suggest that the anti-inflammatory activities of KRG-WE could be due to its inhibition of the p38/JNK/TBK1 activation pathway. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
31. Regulatory role of TRIM21 in the type-I interferonpathway in Japanese encephalitis virus-infected human microglial cells.
- Author
-
Manocha, Gunjan Dhawan, Mishra, Ritu, Sharma, Nikhil, Kumawat, Kanhaiya Lal, Basu, Anirban, and Singh, Sunit K.
- Subjects
- *
JAPANESE encephalitis viruses , *MICROGLIA , *ZOONOSES , *INTERFERONS , *GLYCOPROTEINS - Abstract
Background Japanese encephalitis virus (JEV) infection leads to Japanese encephalitis (JE) in humans. JEV is transmitted through mosquitoes and maintained in a zoonotic cycle. This cycle involves pigs as the major reservoir, water birds as carriers and mosquitoes as vectors. JEV invasion into the central nervous system (CNS) may occur via antipodal transport of virions or through the vascular endothelial cells. Microglial cells get activated in response to pathogenic insults. JEV infection induces the innate immune response and triggers the production of type I interferons. The signaling pathway of type I interferon production is regulated by a number of molecules. TRIM proteins are known to regulate the expression of interferons; however, the involvement of TRIM genes and their underlying mechanism during JEV infection are not known. Methods Human microglial cells (CHME3) were infected with JEV to understand the role of TRIM21 in JEV infection and its effect on type I interferon (IFN-β) production. Cells were infected in presence and absence of exogenous TRIM21 as well as after knocking down the TRIM21 mRNA. Levels of activated IRF3 expression were measured through Western blot analyses of anti-p-IRF3 antibody, and IFN-β production was measured by using IFN-β real-time PCR and luciferase activity analyses. Results JEV infection increased expression of TRIM21 in CHME3 cells. JEV induced an innate immune response by increasing production of IFN-β via IRF3 activation and phosphorylation. Overexpression of TRIM21 resulted in downregulation of p-IRF3 and IFN- β, while silencing led to increased production of p-IRF3 and IFN-β in JEV-infected CHME3 cells. Conclusion This report demonstrates TRIM21 as a negative regulator of interferon-β (IFN- β) production mediated by IRF-3 during JEV infection in human microglial cells. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
32. Azithromycin and Glucosamine May Amplify the Type 1 Interferon Response to RNA Viruses in a Complementary Fashion
- Author
-
Mark F. McCarty, James J. DiNicolantonio, and Jorge Barroso-Aranda
- Subjects
0301 basic medicine ,MDA5 ,Immunology ,Biology ,Azithromycin ,Antiviral Agents ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,RNA Virus Infections ,Glucosamine ,medicine ,Immunology and Allergy ,Animals ,Humans ,RNA Viruses ,Type 1 interferon ,azithromycin ,IRF-3 ,RNA ,type 1 interferon ,MAVS ,Virology ,030104 developmental biology ,chemistry ,Host-Pathogen Interactions ,Interferon Type I ,glucosamine ,Drug Therapy, Combination ,030215 immunology ,medicine.drug - Abstract
Highlights • Previous research demonstrates that, in clinically relevant concentrations, azithromycin can boost the ability of RNA viruses to induce type 1 interferon by amplifying the expression and virally-mediated activation of MDA5. • O-GlcNAcylation of MAVS, a down-stream target of MDA5, renders it more effective for type 1 interferon induction. High-dose glucosamine administration up-regulates O-GlcNAcylation by increasing the cellular pool of UDP-N-acetylglucosamine. • Hence, it is proposed that joint administration of azithromycin and high-dose glucosamine, early in the course of RNA virus infections, may act synergistically to aid their control by enhancing type 1 interferon induction, Previous research demonstrates that, in clinically relevant concentrations, azithromycin can boost the ability of RNA viruses to induce type 1 interferon by amplifying the expression and virally-mediated activation of MDA5. O-GlcNAcylation of MAVS, a down-stream target of MDA5, renders it more effective for type 1 interferon induction. High-dose glucosamine administration up-regulates O-GlcNAcylation by increasing the cellular pool of UDP-N-acetylglucosamine. Hence, it is proposed that joint administration of azithromycin and high-dose glucosamine, early in the course of RNA virus infections, may interact in a complementary fashion to aid their control by enhancing type 1 interferon induction
- Published
- 2020
33. The Intrinsically Disordered W Protein Is Multifunctional during Henipavirus Infection, Disrupting Host Signalling Pathways and Nuclear Import
- Author
-
Camilla M Donnelly, Mikayla Hoad, Emily M. Cross, Sofiya Tsimbalyuk, Justin A Roby, and Jade K. Forwood
- Subjects
0301 basic medicine ,Nuclear Envelope ,henipaviruses ,Active Transport, Cell Nucleus ,Review ,intrinsically disordered ,stat ,Hendra Virus ,Viral Proteins ,03 medical and health sciences ,0302 clinical medicine ,Interferon ,medicine ,Humans ,lcsh:QH301-705.5 ,Gene ,Henipavirus Infections ,Innate immune system ,Host Microbial Interactions ,biology ,IRF-3 ,STAT ,Nipah Virus ,Pattern recognition receptor ,General Medicine ,biology.organism_classification ,Immunity, Innate ,Cell biology ,Intrinsically Disordered Proteins ,030104 developmental biology ,lcsh:Biology (General) ,Receptors, Pattern Recognition ,STAT protein ,Interferons ,Nuclear transport ,W protein ,Signal Transduction ,030215 immunology ,Henipavirus ,medicine.drug - Abstract
Nipah and Hendra viruses are highly pathogenic, zoonotic henipaviruses that encode proteins that inhibit the host’s innate immune response. The W protein is one of four products encoded from the P gene and binds a number of host proteins to regulate signalling pathways. The W protein is intrinsically disordered, a structural attribute that contributes to its diverse host protein interactions. Here, we review the role of W in innate immune suppression through inhibition of both pattern recognition receptor (PRR) pathways and interferon (IFN)-responsive signalling. PRR stimulation leading to activation of IRF-3 and IFN release is blocked by henipavirus W, and unphosphorylated STAT proteins are sequestered within the nucleus of host cells by W, thereby inhibiting the induction of IFN stimulated genes. We examine the critical role of nuclear transport in multiple functions of W and how specific binding of importin-alpha (Impα) isoforms, and the 14-3-3 group of regulatory proteins suggests further modulation of these processes. Overall, the disordered nature and multiple functions of W warrant further investigation to understand henipavirus pathogenesis and may reveal insights aiding the development of novel therapeutics.
- Published
- 2020
34. 自然免疫アダプター分子TRIFを介した抗ウイルスシグナル伝達経路の機能解析
- Author
-
Abe, Hiroto, 藤田, 尚志, 松田, 道行, and 杉田, 昌彦
- Subjects
自然免疫 ,IRF-3 ,TBK1 ,IKKβ ,TRIF - Published
- 2020
35. This title is unavailable for guests, please login to see more information.
- Author
-
Abe, Hiroto and Abe, Hiroto
- Published
- 2020
36. Classical swine fever virus triggers RIG-I and MDA5-dependent signaling pathway to IRF-3 and NF-κB activation to promote secretion of interferon and inflammatory cytokines in porcine alveolar macrophages.
- Author
-
Xiao-Ying Dong, Wen-Jun Liu, Ming-Qiu Zhao, Jia-Ying Wang, Jing-Jing Pei, Yong-Wen Luo, Chun-Mei Ju, and Jin-Ding Chen
- Subjects
- *
SWINE diseases , *CLASSICAL swine fever , *COMMUNICABLE diseases , *ANTIVIRAL agents , *RNA viruses , *GLYCOPROTEINS - Abstract
Background: Classical swine fever (CSF) caused by CSF virus (CSFV) is a highly contagious disease of pigs. The RNA helicases retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA-5) are differentially involved in the detection of various RNA viruses. In present study, we investigated the roles of RIG-I and MDA-5 in eliciting antiviral and inflammatory responses to CSFV shimen strain in Porcine alveolar macrophages (PAMs). Methods: CSFV Shimen strain was used as challenge virus in this study and PAMs were cultured in vitro. Interferon regulatory factor (IRF)-3 and nuclear factor-kappa B (NF-κB) translocation was detected using immunofluorescent staining; RIG-I, MDA5, interferon promoter-stimulating factor 1 (IPS-1), IRF-3 and NF-κB expression was measured by Western Blotting; Interferon beta (IFN-β), IFN-α, interleukin-1beta (IL-1β), IL-6 and tumor necrosis factor (TNF-α) expression was tested by Enzyme-linked immunosorbent assays (ELISA) and shRNA-mediated knockdown of MDA5 or RIG-I was performed. Results: The findings suggested that the initial response to CSFV infection resulted in the higher expression of RIG-I and MDA5 leading to the activation of IPS-1, IRF-3 and NF-κB in a dose-dependent manner. Evaluation of IFN-α, IFN-β, IL-1β, IL-6 or TNF-α expressed by PAMs showed significant differences between infected and uninfected cells. CSFV infected cells induced to express high levels of IFN-α, IFN-β, IL-1β, IL-6 and TNF-α in a dose-dependent way within 24 h post-infection (hpi). At the same time, CSFV improved the nuclear translocation of IRF-3 and NF-κB. We also directly compared and assessed the roles of RIG-I and MDA5 in triggering innate immune actions during CSFV infection through shRNA-mediated knockdown of MDA5 or RIG-I. We found that, compared to the control, the production of IFN-α, IFN-β, IL-1β, IL-6 and TNF-α in response to CSFV infection was heavily reduced in RIG-I knockdown cells while it was moderately decreased in MDA5 knockdown cells. PAMs derived from knockdown of both RIG-I and MDA5 almost failed to produce IFNs and inflammatory cytokines. Conclusions: It indicates that CSFV can be recognized by both RIG-I and MDA5 to initiate the RIG-I signaling pathway to trigger innate defenses against infection [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
37. Japanese encephalitis virus non-coding RNA inhibits activation of interferon by blocking nuclear translocation of interferon regulatory factor 3.
- Author
-
Chang, Ruey-Yi, Hsu, Ta-Wen, Chen, Yen-Lin, Liu, Shu-Fan, Tsai, Yi-Jer, Lin, Yun-Tong, Chen, Yi-Shiuan, and Fan, Yi-Hsin
- Subjects
- *
JAPANESE encephalitis viruses , *NON-coding RNA , *INTERFERONS , *CHROMOSOMAL translocation , *INTERFERON regulatory factors , *ARBOVIRUSES , *FLAVIVIRUSES , *VIRAL genomes - Abstract
Abstract: Noncoding RNA (ncRNA) plays a critical role in modulating a broad range of diseases. All arthropod-borne flaviviruses produce short fragment ncRNA (sfRNA) collinear with highly conserved regions of the 3′-untranslated region (UTR) in the viral genome. We show that the molar ratio of sfRNA to genomic RNA in Japanese encephalitis virus (JEV) persistently infected cells is greater than that in acutely infected cells, indicating an sfRNA role in establishing persistent infection. Transfecting excess quantities of sfRNA into JEV-infected cells reduced interferon-β (IFN-β) promoter activity by 57% and IFN-β mRNA levels by 52%, compared to mock-transfected cells. Transfection of sfRNA into JEV-infected cells also reduced phosphorylation of interferon regulatory factor-3 (IRF-3), the IFN-β upstream regulator, and blocked roughly 30% of IRF-3 nuclear localization. Furthermore, JEV-infected sfRNA transfected cells produced 23% less IFN-β-stimulated apoptosis than mock-transfected groups did. Taken together, these results suggest that sfRNA plays a role against host-cell antiviral responses, prevents cells from undergoing apoptosis, and thus contributes to viral persistence. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
38. Synergistic control of herpes simplex virus pathogenesis by IRF-3, and IRF-7 revealed through non-invasive bioluminescence imaging.
- Author
-
Murphy, Aisling A., Rosato, Pamela C., Parker, Zachary M., Khalenkov, Alexey, and Leib, David A.
- Subjects
- *
IMPULSE response , *NONINVASIVE diagnostic tests , *VIRAL disease diagnosis , *BIOLUMINESCENCE , *INTERFERON regulatory factors , *ANTIVIRAL agents , *LABORATORY mice - Abstract
Abstract: Interferon regulatory factors IRF-3 and IRF-7 are central to the establishment of the innate antiviral response. This study examines HSV-1 pathogenesis in IRF-3−/−, IRF-7−/− and double-deleted IRF3/7−/− (DKO) mice. Bioluminescence imaging of infection revealed that DKO mice developed visceral infection following corneal inoculation, along with increased viral burdens in all tissues relative to single knockout mice. While all DKO mice synchronously reached endpoint criteria 5 days post infection, the IRF-7−/− mice survived longer, indicating that although IRF-7 is dominant, IRF-3 also plays a role in controlling disease. Higher levels of systemic pro-inflammatory cytokines were found in IRF7−/− and DKO mice relative to wild-type and IRF-3−/− mice, and IL-6 and G-CSF, indicative of sepsis, were increased in the DKO mice relative to wild-type or single-knockout mice. In addition to controlling viral replication, IRF-3 and -7 therefore play coordinating roles in modulation of inflammation during HSV infection. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
39. Two new monoclonal antibodies for biochemical and flow cytometric analyses of human interferon regulatory factor-3 activation, turnover, and depletion
- Author
-
Rustagi, Arjun, Doehle, Brian P., McElrath, M. Juliana, and Gale, Michael
- Subjects
- *
MONOCLONAL antibodies , *BIOCHEMISTRY , *CYTOMETRY , *INTERFERON regulatory factors , *TRANSCRIPTION factors , *IMMUNE response , *VIRUS diseases - Abstract
Abstract: Interferon regulatory factor-3 (IRF-3) is a master transcription factor that drives the host intracellular innate immune response to virus infection. The importance of IRF-3 in innate immune responses is highlighted by the fact that pathogenic viruses have developed strategies for antagonism of IRF-3. Several tools exist for evaluation of viral regulation of IRF-3 activation and function, but high-quality monoclonal antibodies that mark the differential activation states of human IRF-3 are lacking. To study IRF-3 activation, turnover, and depletion in a high-throughput manner in the context of virus infection, we have developed two new monoclonal antibodies to human IRF-3. These antibodies detect IRF-3 in virus-infected cells in a wide variety of assays and provide a new tool to study virus-host interactions and innate immune signaling. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
40. Molecular Biology of Hepatitis C Virus: Interactions with the IFN-Betta Signalling Pathway
- Author
-
M Sabourighannad, C McCormick, A MacDonald, D Rowlands, and M Harris
- Subjects
HCV ,IFN betta ,HCV replicon ,IRF-3 ,NFκB ,Public aspects of medicine ,RA1-1270 - Abstract
The induction of IFN-β expression is the first stage in the innate anti-viral response. In order to investigate the possible effects of HCV proteins on IFN-β signalling, a baculovirus delivery system was developed to introduce the whole genome of HCV genotype 1b into hepatoma cells. The construct used in this study lacks the 3’UTR which is required for HCV replication, thus enabling us to look at the effects of HCV proteins on the IFN-β signalling pathway without inducing IFN-β expression by virtue of the presence of replicating (double-stranded) viral RNA. To facilitate this analysis the expression of the HCV polyprotein was under the control of a tetracycline–responsive promoter coupled to the HCV 5’UTR. As a comparison, we have also generated a recombinant baculovirus containing the culture adapted sub-genomic replicon (FK5.1) also derived from HCV genotype 1b, and a mutant form thereof containing an inactivating mutation within the NS5B (RdRp) coding sequence (termed GND). We first confirmed that HepG2 cells were able to mount an effective IFN-β response. As expected, the baculovirus carrying the FK5.1 replicon induced the production of IFN-β as judged by the use of an IFN-β-promoter luciferase reporter construct, whereas the GND baculovirus and the full-length 3’UTR deletant failed to induce luciferase expression. We then proceeded to analyse the effect of the HCV polyprotein on exogenous induction of the IFN-β promoter (by transfecting cells with poly I/C). These studies demonstrated that neither the HCV polyprotein nor the non-structural proteins of HCV (expressed from the replicon) had any effect on the dsRNA-mediated induction of IFN-β promoter. Secondly we analysed potential effects on the inhibition of the IFN-β response, using an ISRE-luciferase construct. Again we observed no effect of either the complete polyprotein or the sub-genomic replicon. Lastly, we examined the activation of both IRF-3 and NFκB, two transcription factors induced by dsRNA signalling that are key to the activation of transcription from the IFN-β promoter. Intriguingly, both the wild type or GND-mutant replicon blocked the dsRNA-induced activation of IRF-3 and NFκB. In contrast the full-length 3’UTR deletant had no significant effect on either transcription factor. These results suggest a complex interplay between HCV and the IFN-β system that is dependent both on the context of polyprotein expression (full length compared with sub-genomic replicon) and the primary amino acid sequence (culture adapted compared with infectious clone).
- Published
- 2005
41. Impairment of interferon regulatory factor-3 activation by hepatitis C virus core protein basic amino acid region 1
- Author
-
Inoue, Kazuaki, Tsukiyama-Kohara, Kyoko, Matsuda, Chiho, Yoneyama, Mitsutoshi, Fujita, Takashi, Kuge, Shusuke, Yoshiba, Makoto, and Kohara, Michinori
- Subjects
- *
INTERFERONS , *HEPATITIS C virus , *NEWCASTLE disease virus , *GENOMICS , *IMMUNOFLUORESCENCE , *WESTERN immunoblotting , *POLYMERASE chain reaction - Abstract
Abstract: Interferon regulatory factor-3 (IRF-3), a key transcriptional factor in the type I interferon system, is frequently impaired by hepatitis C virus (HCV), in order to establish persistent infection. However, the exact mechanism by which the virus establishes persistent infection has not been fully understood yet. The present study aimed to investigate the effects of various HCV proteins on IRF-3 activation, and elucidate the underlying mechanisms. To achieve this, full-length HCV and HCV subgenomic constructs corresponding to structural and each of the nonstructural proteins were transiently transfected into HepG2 cells. IFN-β induction, plaque formation, and IRF-3 dimerization were elicited by Newcastle disease virus (NDV) infection. The expressions of IRF-3 homodimer and its monomer, Ser386-phosphorylated IRF-3, and HCV core protein were detected by immunofluorescence and western blotting. IFN-β mRNA expression was quantified by real-time PCR (RT-PCR), and IRF-3 activity was measured by the levels of IRF-3 dimerization and phosphorylation, induced by NDV infection or polyriboinosinic:polyribocytidylic acid [poly(I:C)]. Switching of the expression of the complete HCV genome as well as the core proteins, E1, E2, and NS2, suppressed IFN-β mRNA levels and IRF-3 dimerization, induced by NDV infection. Our study revealed a crucial region of the HCV core protein, basic amino acid region 1 (BR1), to inhibit IRF-3 dimerization as well as its phosphorylation induced by NDV infection and poly (I:C), thus interfering with IRF-3 activation. Therefore, our study suggests that rescue of the IRF-3 pathway impairment may be an effective treatment for HCV infection. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
42. Translocase of outer mitochondrial membrane 70 induces interferon response and is impaired by hepatitis C virus NS3
- Author
-
Kasama, Yuri, Saito, Makoto, Takano, Takashi, Nishimura, Tomohiro, Satoh, Masaaki, Wang, Zhongzhi, Ali, Salem Nagla Elwy Salem, Harada, Shinji, Kohara, Michinori, and Tsukiyama-Kohara, Kyoko
- Subjects
- *
HEPATITIS C virus , *MITOCHONDRIAL membranes , *INTERFERONS , *LIVER cells , *GENE expression , *PHOSPHORYLATION , *CELLULAR signal transduction - Abstract
Abstract: Hepatitis C virus (HCV) elevated expression of the translocase of outer mitochondrial membrane 70 (Tom70). Interestingly, overexpression of Tom70 induces interferon (IFN) synthesis in hepatocytes, and it was impaired by HCV. Here, we addressed the mechanism of this impairment. The HCV NS3/4A protein induced Tom70 expression. The HCV NS3 protein interacted in cells, and cleaved the adapter protein mitochondrial anti-viral signaling (MAVS). Ectopic overexpression of Tom70 could not inhibit this cleavage. As a result, IRF-3 phosphorylation was impaired and IFN-β induction was suppressed. These results indicate that MAVS works upstream of Tom70 and the cleavage of MAVS by HCV NS3 protease suppresses signaling of IFN induction. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
43. An IRF-3 homolog that is up-regulated by DNA virus and poly I:C in turbot, Scophthalmus maximus
- Author
-
Hu, Guo-Bin, Xia, Jun, Lou, Hui-Min, Chen, Xiao-Ling, Li, Jing, and Liu, Qiu-Ming
- Subjects
- *
PSETTA maxima , *HOMOLOGY (Biology) , *DNA viruses , *MESSENGER RNA , *GENETIC regulation , *FISH genetics , *EXONS (Genetics) - Abstract
Abstract: In this study, we described the structure, mRNA tissue distribution and regulation of an IRF-3 gene from turbot, Scophthalmus maximus (SmIRF-3). The gene sequence of SmIRF-3 is 6077 bp long, composed of 11 exons and 10 introns similar to known IRF-3 genes of fish, and encodes a peptide of 466 amino acids. The deduced protein sequence shares the highest identity of 56.0–81.2% with fish IRF-3 and possesses a DNA-binding domain (DBD), an IRF association domain (IAD) and a serine-rich domain (SRD) known to be important for the functions of IRF-3 in vertebrates. Phylogenetic analysis grouped SmIRF-3 with other IRF3s of vertebrates. SmIRF-3 transcripts were detectable in limited tissue types of healthy fish, with higher expression observed in head, kidney, spleen and kidney,. The SmIRF-3 was transcriptionally up-regulated by turbot reddish body iridovirus (TRBIV) and polyinosinic:polycytidylic acid (poly I:C) in the head kidney, spleen and gills, with showing a two wave induced expression during a 7-day time course in all cases. The highest inducibility and the likely earliest increase of SmIRF-3 expression were observed in the spleen, and poly I:C was a stronger inducer. In addition, the maximal expression level of SmIRF-3 arose prior to that of the Mx in all the cases. [Copyright &y& Elsevier]
- Published
- 2011
- Full Text
- View/download PDF
44. IRF3 polymorphisms induce different innate anti-Theiler's virus immune responses in RAW264.7 macrophages
- Author
-
Moore, Tyler C., Al-Salleeh, Fahd M., Brown, Deborah M., and Petro, Thomas M.
- Subjects
- *
GENETIC polymorphisms , *IMMUNE response , *MACROPHAGES , *IMPULSE response , *APOPTOSIS , *VIRUS diseases , *MYOCARDITIS , *ENCEPHALOMYELITIS , *DISEASE risk factors - Abstract
Abstract: Persistent viral infections can lead to disease such as myocarditis. Theiler''s murine encephalomyelitis virus (TMEV) infects macrophages of SJL/J (H-2s) mice establishing persistent infections leading to demyelinating disease. In contrast macrophages from B10.S (H-2s) mice clear TMEV. Activation of the transcription factor IRF3 induces IFNβ, ISG56, and apoptosis for viral clearance, but also inflammatory cytokines, such as IL-23 and IL6, which contribute to disease. Here we identify polymorphisms in the IRF3 of SJL/J versus B10.S mice that are located in DNA binding, nuclear localization, and autoinhibitory domains. SJL-IRF3 expression in RAW264.7 macrophage cells with or without TMEV infection decreased IL-23p19 promoter activity compared with B10S-IRF3. In contrast SJL-IRF3 increased IL-6, ISG56 and IFNβ in response to TMEV. B10S-IRF3 expression augmented apoptotic caspase activation and decreased viral RNA in TMEV-infected macrophages while SJL-IRF3 increased viral replication with less caspase activation. Therefore IRF3 polymorphisms contribute to viral persistence and altered cytokine expression. [Copyright &y& Elsevier]
- Published
- 2011
- Full Text
- View/download PDF
45. A conventional protein kinase C inhibitor targeting IRF-3-dependent genes differentially regulates IL-12 family members
- Author
-
Johnson, Jolyn, Molle, Céline, Aksoy, Ezra, Goldman, Michel, Goriely, Stanislas, and Willems, Fabienne
- Subjects
- *
PROTEIN kinase C , *ENZYME inhibitors , *IMMUNOREGULATION , *INTERFERONS , *GENE expression , *DENDRITIC cells , *ENZYME-linked immunosorbent assay , *BONE marrow - Abstract
Abstract: Protein kinase C (PKC) isoforms play a critical role in the regulation of innate immune responses. We have previously demonstrated that conventional PKC (cPKC) α is involved in interferon regulatory factor 3 (IRF-3) activation and IFN-β synthesis. Herein, we investigated the role of cPKCs in the regulation of IL-12 family members expression mediated by the Toll-like receptor 3 (TLR3) and TLR4. First, inhibition of cPKCs activity in human DCs by a cPKC-specific inhibitor, Gö6976 downregulated the expression of IL-12p70 and IL-27p28 but not IL-12/IL-23p40, IL-23, IL-27EBI3 induced by LPS or poly(I:C). Furthermore, reporter gene assays in RAW 264.7 macrophages showed that cPKCs regulate IL-12p35 and IL-27p28 promoter activities since Gö6976 repressed LPS and poly(I:C)-mediated transcriptional activities of IL-12p35 and IL-27p28. In contrast, no effect was observed with IL-12/IL-23p40 and IL-23p19 reporter constructs. These results prompted us to study the role of IRF-3 on IL-23 expression. Bone marrow-derived DC (BMDCs) from IRF-3−/− mice produced comparable levels of IL-23 induced by both LPS and poly(I:C) as compared to wild type BMDCs, indicating that IRF-3 is not involved in IL-23 production. Finally, BMDCs from PKCα−/− mice displayed a reduced synthesis of IL-27 induced by poly(I:C). Collectively, these data identify cPKCs as critical components that control IRF-3-dependent IL-12p35 and IL-27p28 gene expression downstream of TLR3 and TLR4. [Copyright &y& Elsevier]
- Published
- 2011
- Full Text
- View/download PDF
46. Identification of novel alternative splicing variants of interferon regulatory factor 3.
- Author
-
Li, Yong, Hu, Xiuhua, Song, Yuqin, Lu, Zheming, Ning, Tao, Cai, Hong, and Ke, Yang
- Subjects
IMMUNE system ,INTERFERONS ,COMPETITIVE exclusion (Microbiology) ,NUCLEAR nonproliferation ,CERVICAL cancer treatment ,GENETIC engineering ,CHROMOSOMAL translocation ,CARCINOGENESIS - Abstract
Abstract: Interferon regulatory factor 3 (IRF-3) plays a crucial role in host defense against viral and microbial infection as well as in cell growth regulation. IRF-3a is the only structurally and functionally characterized IRF-3 splicing variant and has been established to antagonize IRF-3 activity. Here, five novel splicing variants of IRF-3, referred to as IRF-3b, -3c, -3d, -3e, and -3f, were identified and shown to be generated by deletion of exons 2, 3, or 6 or some combination thereof. RT-PCR examination revealed that these novel splicing variants were more frequently expressed in human liver, esophagus, and cervical tumor tissues than in their normal counterparts. Additionally, electrophoretic mobility shift assay and subcellular localization showed only IRF-3 and IRF-3e were capable of binding the PRDI/III element of interferon-beta (IFNβ) promoter in vitro and underwent cytoplasm-to-nucleus translocation following Poly(I:C) stimulation. Coimmunoprecipitation assay revealed that only IRF-3c (3f) of novel splicing variants associated with IRF-3 in vivo. Further luciferase assay showed IRF-3c (3f) and IRF-3e failed to transactivate PRDI/III-containing promoter but appeared to inhibit transactivation potential of IRF-3 to varying degrees. Taken together, our findings suggest novel splicing variants may function as negative modulators of IRF-3 and may be correlated with pathogenesis of human tumors. [Copyright &y& Elsevier]
- Published
- 2011
- Full Text
- View/download PDF
47. Interferon regulatory factor 3 (IRF-3) in Japanese flounder, Paralichthys olivaceus: Sequencing, limited tissue distribution, inducible expression and induction of fish type I interferon promoter
- Author
-
Hu, Guobin, Yin, Xiangyan, Lou, Huimin, Xia, Jun, Dong, Xianzhi, Zhang, Jianyie, and Liu, Qiuming
- Subjects
- *
GENETIC regulation , *PARALICHTHYS , *NUCLEOTIDE sequence , *GENE expression , *INTERFERON inducers , *REVERSE transcriptase polymerase chain reaction , *CIRCULAR DNA - Abstract
Abstract: Two cDNAs with different 3′-untranslated region (UTR) encoding an interferon regulatory factor 3 (IRF-3) were cloned from head kidney of Japanese flounder, Paralichthys olivaceus, by reverse transcription polymerase chain reaction (RT-PCR) and rapid amplification of cDNA ends (RACE) methods. Sequence analysis reveals that they were generated by alternative polyadenylation. The predicted protein consists of 467 amino acid residues which shares the highest identity of 50.7–57.6% to fish IRF-3 and possesses a DNA-binding domain (DBD), an IRF association domain (IAD) and a serine-rich domain (SRD) of vertebrate IRF-3. The presence of these domains along with phylogenetic analysis places it into the IRF-3 group of the IRF-3 subfamily. RT-PCR analysis revealed that flounder IRF-3 was expressed constitutively in limited tissue types including head kidney, spleen, kidney, heart, gill, intestine and liver. A quantitative real time PCR assay was employed to monitor expression of IRF-3, type I interferon (IFN) and Mx in flounder head kidney and gill. All three genes were up-regulated by polyinosinic:polycytidylic acid (polyI:C) and lymphocystis disease virus (LCDV) with an earlier but slight and less persistent increase in transcription levels seen for the IRF-3. Finally, flounder IRF-3 was proved to induce fish type I IFN promoter in FG9307 cells, a flounder gill cell line, by a luciferase assay. These results provide insights into the roles of fish IRF-3 in the antiviral immunity. [Copyright &y& Elsevier]
- Published
- 2011
- Full Text
- View/download PDF
48. Strain-to-strain difference of V protein of measles virus affects MDA5-mediated IFN-β-inducing potential
- Author
-
Takaki, Hiromi, Watanabe, Yumi, Shingai, Masashi, Oshiumi, Hiroyuki, Matsumoto, Misako, and Seya, Tsukasa
- Subjects
- *
MEASLES virus , *VIRUS diseases , *INTERFERONS , *STRAINS & stresses (Mechanics) , *MELANOMA , *TRETINOIN , *PARAMYXOVIRUSES , *GENE amplification , *RNA , *DISEASE risk factors - Abstract
Abstract: Laboratory-adapted and vaccine strains of measles virus (MV) induce type I interferon (IFN) in infected cells to a far greater extent than wild-type strains. We investigated the mechanisms for this differential type I IFN production in cells infected with representative MV strains. The overexpression of the wild-type V protein suppressed melanoma differentiation-associated gene 5 (MDA5)-induced IFN-β promoter activity, while this was not seen in A549 cells expressing CD150 transfected with the V protein of the vaccine strain. The V proteins of the wild-type also suppressed poly I:C-induced IFN regulatory factor 3 (IRF-3) dimerization. The V proteins of the wild-type and vaccine strain did not affect retinoic acid-inducible gene 1 (RIG-I)- or toll-IL-1R homology domain-containing adaptor molecule 1 (TICAM-1)-induced IFN-β promoter activation. We identified an amino acid substitution of the cysteine residue at position 272 (which is conserved among paramyxoviruses) to an arginine residue in the V protein of the vaccine strain. Only the V protein possessing the 272C residue binds to MDA5. The mutation introduced into the wild-type V protein (C272R) was unable to suppress MDA5-induced IRF-3 nuclear translocation and IFN-β promoter activation as seen in the V proteins of the vaccine strain, whereas the mutation introduced in the vaccine strain V protein (R272C) was able to inhibit MDA5-induced IRF-3 and IFN-β promoter activation. The other 6 residues of the vaccine strain V sequence inconsistent with the authentic sequence of the wild-type V protein barely affected the IRF-3 nuclear translocation. These data suggested that the structural difference of vaccine MV V protein hampers MDA5 blockade and acts as a nidus for the spread/amplification of type I IFN induction. Ultimately, measles vaccine strains have two modes of IFN-β-induction for their attenuation: V protein mutation and production of defective interference (DI) RNA. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
49. Hyperglycemia, oxidative and nitrosative stress affect antiviral, inflammatory and apoptotic signaling of cultured thymocytes.
- Author
-
Kocic, G., Sokolovic, D., Jevtovic, T., Veljkovic, A., Kocic, R., Nikolic, G., Basic, J., Stojanovic, D., Cencic, A., and Stojanovic, S.
- Subjects
- *
DISEASE prevalence , *COMMUNICABLE diseases , *NATURAL immunity , *PEOPLE with diabetes , *HYPERGLYCEMIA , *OXIDATIVE stress , *NITRIC oxide , *LABORATORY mice - Abstract
A high prevalence of various infectious diseases is reported in diabetic patients, which may suggest impaired innate immunity against different pathogen-associated molecular patterns. This study investigated the effects of hyperglycemia, oxidative stress (H2O2), nitric oxide (NO) and peroxynitrite (ONOO–) on the modulation of antiviral (MDA-5, IRF-3 and phospho-IRF-3), inflammatory (NF-κB) and pro/anti-apoptotic molecules (Bax and Bcl-2) in BALB/c mice thymocytes. Each of the experimental conditions, except the weakest NO concentration, resulted in down-regulation of MDA-5, IRF-3 and phospho-IRF-3. In contrast, each of the experimental conditions elicited up-regulation of NF-κB, Bcl-2 and Bax. These results suggest that hyperglycemia, oxidative and nitrosative stress may contribute to the reduced immunity of the host by altering the MDA-5/IRF-3/phosphoIRF-3 axis, as well as contributing to the mechanisms of inflammatory reaction via increased NF-κB, and to augmented turnover rate of thymocyte cells via Bcl2/Bax up-regulation. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
50. Viral apoptosis is induced by IRF-3-mediated activation of Bax.
- Author
-
Chattopadhyay, Saurabh, Marques, Joao T., Yamashita, Michifumi, Peters, Kristi L., Smith, Kevin, Desai, Avanti, Williams, Bryan R. G., and Sen, Ganes C.
- Subjects
- *
APOPTOSIS , *CELL death , *TRETINOIN , *TRANSCRIPTION factors , *ANTINEOPLASTIC agents - Abstract
Upon infection with many RNA viruses, the cytoplasmic retinoic acid inducible gene-I (RIG-I) pathway activates the latent transcription factor IRF-3, causing its nuclear translocation and the induction of many antiviral genes, including those encoding interferons. Here, we report a novel and distinct activity of IRF-3, in virus-infected cells, that induces apoptosis. Using genetically defective mouse and human cell lines, we demonstrated that, although both pathways required the presence of RIG-I, IPS1, TRAF3 and TBK1, only the apoptotic pathway required the presence of TRAF2 and TRAF6 in addition. More importantly, transcriptionally inactive IRF-3 mutants, such as the one missing its DNA-binding domain, could efficiently mediate apoptosis. Apoptosis was triggered by the direct interaction of IRF-3, through a newly identified BH3 domain, with the pro-apoptotic protein Bax, their co-translocation to the mitochondria and the resulting activation of the mitochondrial apoptotic pathway. Thus, IRF-3 is a dual-action cytoplasmic protein that, upon activation, translocates to the nucleus or to the mitochondrion and triggers two complementary antiviral responses of the infected cell. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.