168 results on '"IFN-λ"'
Search Results
2. Swine acute diarrhea syndrome coronavirus Nsp1 suppresses IFN-λ1 production by degrading IRF1 via ubiquitin–proteasome pathway
- Author
-
Chunhui Zhong, Gaoli She, Yukun Zhao, Yufang Liu, Jingmin Li, Xiaona Wei, Zexin Chen, Keyu Zhao, Zhiqing Zhao, Zhichao Xu, Hao Zhang, Yongchang Cao, and Chunyi Xue
- Subjects
Swine acute diarrhea syndrome coronavirus ,Nsp1 ,IFN-λ ,IRF1 ,innate immune evasion ,Veterinary medicine ,SF600-1100 - Abstract
Abstract Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a novel porcine enteric coronavirus that causes acute watery diarrhea, vomiting, and dehydration in newborn piglets. The type III interferon (IFN-λ) response serves as the primary defense against viruses that replicate in intestinal epithelial cells. However, there is currently no information available on how SADS-CoV modulates the production of IFN-λ. In this study, we utilized IPI-FX cells (a cell line of porcine ileum epithelium) as an in vitro model to investigate the potential immune evasion strategies employed by SADS-CoV against the IFN-λ response. Our results showed that SADS-CoV infection suppressed the production of IFN-λ1 induced by poly(I:C). Through screening SADS-CoV-encoded proteins, nsp1, nsp5, nsp10, nsp12, nsp16, E, S1, and S2 were identified as antagonists of IFN-λ1 production. Specifically, SADS-CoV nsp1 impeded the activation of the IFN-λ1 promoter mediated by MAVS, TBK1, IKKε, and IRF1. Both SADS-CoV and nsp1 obstructed poly(I:C)-induced nuclear translocation of IRF1. Moreover, SADS-CoV nsp1 degraded IRF1 via the ubiquitin-mediated proteasome pathway without interacting with it. Overall, our study provides the first evidence that SADS-CoV inhibits the type III IFN response, shedding light on the molecular mechanisms employed by SADS-CoV to evade the host immune response.
- Published
- 2024
- Full Text
- View/download PDF
3. NS7a of SADS-CoV promotes viral infection via inducing apoptosis to suppress type III interferon production.
- Author
-
Xiaowei Wang, Wenjing Qiu, Guangli Hu, Xiaoyuan Diao, Yunfei Li, Yue Li, Peng Li, Yufang Liu, Yongtong Feng, Chunyi Xue, Yongchang Cao, and Zhichao Xu
- Subjects
- *
VIRUS diseases , *INTERFERON regulatory factors , *APOPTOSIS , *VIRAL proteins , *APOPTOSIS inducing factor , *INTERFERONS - Abstract
Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a newly discovered swine coronavirus with potential cross-species transmission risk. Although SADS-CoV-induced host cell apoptosis and innate immunity antagonization has been revealed, underlying signaling pathways remain obscure. Here, we demonstrated that infection of SADS-CoV induced apoptosis in vivo and in vitro, and that viral protein NS7a is mainly responsible for SADS-CoV-induced apoptosis in host cells. Furthermore, we found that NS7a interacted with apoptosis-inducing factor mitochondria associated 1 (AIFM1) to activate caspase-3 via caspase-6 in SADS-CoV-infected cells, and enhanced SADS-CoV replication. Importantly, NS7a suppressed poly(I:C)-induced expression of type III interferon (IFN-λ) via activating caspase-3 to cleave interferon regulatory factor 3 (IRF3), and caspase-3 inhibitor protects piglets against SADS-CoV infection in vivo. These findings reveal how SADS-CoV induced apoptosis to inhibit innate immunity and provide a valuable clue to the development of effective drugs for the clinical control of SADS-CoV infection. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. Swine acute diarrhea syndrome coronavirus Nsp1 suppresses IFN-λ1 production by degrading IRF1 via ubiquitin–proteasome pathway.
- Author
-
Zhong, Chunhui, She, Gaoli, Zhao, Yukun, Liu, Yufang, Li, Jingmin, Wei, Xiaona, Chen, Zexin, Zhao, Keyu, Zhao, Zhiqing, Xu, Zhichao, Zhang, Hao, Cao, Yongchang, and Xue, Chunyi
- Abstract
Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a novel porcine enteric coronavirus that causes acute watery diarrhea, vomiting, and dehydration in newborn piglets. The type III interferon (IFN-λ) response serves as the primary defense against viruses that replicate in intestinal epithelial cells. However, there is currently no information available on how SADS-CoV modulates the production of IFN-λ. In this study, we utilized IPI-FX cells (a cell line of porcine ileum epithelium) as an in vitro model to investigate the potential immune evasion strategies employed by SADS-CoV against the IFN-λ response. Our results showed that SADS-CoV infection suppressed the production of IFN-λ1 induced by poly(I:C). Through screening SADS-CoV-encoded proteins, nsp1, nsp5, nsp10, nsp12, nsp16, E, S1, and S2 were identified as antagonists of IFN-λ1 production. Specifically, SADS-CoV nsp1 impeded the activation of the IFN-λ1 promoter mediated by MAVS, TBK1, IKKε, and IRF1. Both SADS-CoV and nsp1 obstructed poly(I:C)-induced nuclear translocation of IRF1. Moreover, SADS-CoV nsp1 degraded IRF1 via the ubiquitin-mediated proteasome pathway without interacting with it. Overall, our study provides the first evidence that SADS-CoV inhibits the type III IFN response, shedding light on the molecular mechanisms employed by SADS-CoV to evade the host immune response. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. The extracellular polysaccharide inhibit porcine epidemic diarrhea virus with extract and gene editing Lacticaseibacillus
- Author
-
Shaojun Chen, Zida Nai, Ziliang Qin, Gang Li, Xinmiao He, Wentao Wang, Yaguang Tian, Di Liu, and Xinpeng Jiang
- Subjects
The extracellular polysaccharide ,The anti-PEDV infection ,IFN-λ ,Extract ,Genome editing lacticaseibacillus casei ,Microbiology ,QR1-502 - Abstract
Abstract Lacticaseibacillus is one of the predominant microorganisms in gut from human and animal, and the lacticaseibacillus have effective applications against the viral diarrhea of piglets in the farm. However, the function and the concrete cell single pathways of the active ingredient from lacticaseibacillus was not clear within anti-infection in the postbiotics research. Here, we compared the biological function of extracellular polysaccharides (EPS) purified from lacticaseibacillus casei (L. casei) and gene editing lacticaseibacillus casei with the CRISPER-Cas9 technology, which were with the ability of antioxidation and anti-inflammation, and the EPS could also inhibit the ROS production within the Porcine Small Intestinal Epithelial Cells-J2 (IPEC-J2). Interestingly, we found that both of EPS and genome editing lacticaseibacillus casei could specifically target the IFN-λ expression in the IPEC-J2, which was beneficial against the PEDV infection in the virus replication and production with the qRT-PCR and indirect immunofluorescence methods. Finally, the STAT3 cell single pathway was stimulated to transcribe IFN-λ with the EPS to elucidate the detailed mechanism of activating type III IFN signals receptor of IL-10R2, which play the function between anti-inflammation and anti-virus in the PEDV infection. Taken together, our research linked a postbiotics of EPS with the antiviral infection of PEDV, which suggest that the lacticaseibacillus itself still have displayed the potential immunomodulatory activities, and highlight the immunomodulatory potential of EPS-producing microbes. Graphical Abstract
- Published
- 2023
- Full Text
- View/download PDF
6. Enterovirus D68 vRNA induces type III IFN production via MDA5
- Author
-
Chi-Chong Chio, Hio-Wai Chan, Shih-Hsiang Chen, and Hsing-I Huang
- Subjects
Ev-D68 ,Calu-3 ,IFN-λ ,vRNA ,MDA5 ,Microbiology ,QR1-502 ,Infectious and parasitic diseases ,RC109-216 - Abstract
Enterovirus D68 (EV-D68) primarily spreads through the respiratory tract and causes respiratory symptoms in children and acute flaccid myelitis (AFM). Type III interferons (IFNs) play a critical role in inhibiting viral growth in respiratory epithelial cells. However, the mechanism by which EV-D68 induces type III IFN production is not yet fully understood. In this study, we show that EV-D68 infection stimulates Calu-3 cells to secrete IFN-λ. The transfection of EV-D68 viral RNA (vRNA) stimulated IFN-λ via MDA5. Furthermore, our findings provide evidence that EV-D68 infection also induces MDA5- IRF3/IRF7-mediated IFN-λ. In addition, we discovered that EV-D68 infection downregulated MDA5 expression. Knockdown of MDA5 increased EV-D68 replication in Calu-3 cells. Finally, we demonstrated that the IFN-λ1 and IFN-λ2/3 proteins effectively inhibit EV-D68 infection in respiratory epithelial cells. In summary, our study shows that EV-D68 induces type III IFN production via the activated MDA5-IRF3/IRF7 pathway and that type III IFNs inhibit EV-D68 replication in Calu-3 cells.
- Published
- 2024
- Full Text
- View/download PDF
7. The extracellular polysaccharide inhibit porcine epidemic diarrhea virus with extract and gene editing Lacticaseibacillus.
- Author
-
Chen, Shaojun, Nai, Zida, Qin, Ziliang, Li, Gang, He, Xinmiao, Wang, Wentao, Tian, Yaguang, Liu, Di, and Jiang, Xinpeng
- Subjects
- *
PORCINE epidemic diarrhea virus , *LACTOBACILLUS casei , *GENOME editing , *POLYSACCHARIDES , *VIRAL diarrhea - Abstract
Lacticaseibacillus is one of the predominant microorganisms in gut from human and animal, and the lacticaseibacillus have effective applications against the viral diarrhea of piglets in the farm. However, the function and the concrete cell single pathways of the active ingredient from lacticaseibacillus was not clear within anti-infection in the postbiotics research. Here, we compared the biological function of extracellular polysaccharides (EPS) purified from lacticaseibacillus casei (L. casei) and gene editing lacticaseibacillus casei with the CRISPER-Cas9 technology, which were with the ability of antioxidation and anti-inflammation, and the EPS could also inhibit the ROS production within the Porcine Small Intestinal Epithelial Cells-J2 (IPEC-J2). Interestingly, we found that both of EPS and genome editing lacticaseibacillus casei could specifically target the IFN-λ expression in the IPEC-J2, which was beneficial against the PEDV infection in the virus replication and production with the qRT-PCR and indirect immunofluorescence methods. Finally, the STAT3 cell single pathway was stimulated to transcribe IFN-λ with the EPS to elucidate the detailed mechanism of activating type III IFN signals receptor of IL-10R2, which play the function between anti-inflammation and anti-virus in the PEDV infection. Taken together, our research linked a postbiotics of EPS with the antiviral infection of PEDV, which suggest that the lacticaseibacillus itself still have displayed the potential immunomodulatory activities, and highlight the immunomodulatory potential of EPS-producing microbes. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
8. Interleukin-22 facilitates the interferon-λ-mediated production of tripartite motif protein 25 to inhibit replication of duck viral hepatitis A virus type 1
- Author
-
Hao An, Yumei Liu, Ming Shu, and Junhao Chen
- Subjects
TRIM25 ,IL-22 ,IFN-λ ,phosphorylation ,STAT3 ,Veterinary medicine ,SF600-1100 - Abstract
Abstract The innate immune system provides a defense against invading pathogens by inducing various interferon (IFN)-stimulated genes (ISGs). We recently reported that tripartite motif protein 25 (TRIM25), an important ISG, was highly upregulated in duck embryo hepatocyte cells (DEFs) after infection with duck viral hepatitis A virus type 1 (DHAV-1). However, the mechanism of upregulation of TRIM25 remains unknown. Here we reported that interleukin-22 (IL-22), whose expression was highly facilitated in DEFs and various organs of 1-day-old ducklings after DHAV-1 infection, highly enhanced the IFN-λ-induced production of TRIM25. The treatment with IL-22 neutralizing antibody or the overexpression of IL-22 highly suppressed or facilitated TRIM25 expression, respectively. The phosphorylation of signal transducer and activator of transcription 3 (STAT3) was crucial for the process of IL-22 enhancing IFN-λ-induced TRIM25 production, which was suppressed by WP1066, a novel inhibitor of STAT3 phosphorylation. The overexpression of TRIM25 in DEFs resulted in a high production of IFNs and reduced DHAV-1 replication, whereas the attenuated expression of IFNs and facilitated replication of DHAV-1 were observed in the RNAi group, implying that TRIM25 defended the organism against DHAV-1 propagation by inducing the production of IFNs. In summary, we reported that IL-22 activated the phosphorylation of STAT3 to enhance the IFN-λ-mediated TRIM25 expression and provide a defense against DHAV-1 by inducing IFN production.
- Published
- 2023
- Full Text
- View/download PDF
9. Anticancer Property of Alstonia scholaris Linn Leaf Extract in MCF -7 and MDA-MB -231 Breast Cancer Cell Lines.
- Author
-
Shanmugapriya, Dhanakodi, Jayanthi, Gopalan, and Rajeswari, Ranga Anantha Sayanam
- Subjects
CANCER cells ,CELL lines ,ALSTONIA ,BREAST cancer ,P53 antioncogene ,BREAST ,MAMMOGRAMS - Abstract
Breast cancer is the major risk factor for mortality globally. Medicinal plant extracts were investigated as a potential source for breast cancer therapy. The current work aims to assess the intracellular ROS production and apoptosis induction by Alstonia scholaris Linn (A. scholaris) leaf extract in breast cancer cell lines. Column chromatography was used to purify bioactive components from an A. scholaris Linn methanolic extract. Fraction with a significant concentration of phytochemicals was utilised for further investigation. The results of DCFH-DA staining revealed a substantial rise in intracellular ROS levels in A. scholaris Linn-treated cell lines. The dual staining technique (AO/EtBr) was used to examine apoptotic changes. Treatment with A. scholaris Linn substantially enhanced the rate of apoptosis in breast cancer cell lines. Furthermore, enhanced p53 gene expression, downregulation of Bcl-2 and BRAC1 genes, and reduced TNF-α and IFN-λ concentrations in A. scholaris Linn treated cancer cells demonstrated its anticancer activity. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
10. Epigenetic control of type III interferon expression by 8-oxoguanine and its reader 8-oxoguanine DNA glycosylase1.
- Author
-
Yaoyao Xue, Pan, Lang, Vlahopoulos, Spiros, Ke Wang, Xu Zheng, Radak, Zsolt, Bacsi, Attila, Tanner, Lloyd, Brasier, Allan R., Xueqing Ba, and Boldogh, Istvan
- Subjects
GENE expression ,RESPIRATORY syncytial virus infections ,SMALL molecules ,EPIGENETICS ,INTERFERONS ,METHYLGUANINE - Abstract
Interferons (IFNs) are secreted cytokines with the ability to activate expression of IFN stimulated genes that increase resistance of cells to virus infections. Activated transcription factors in conjunction with chromatin remodelers induce epigenetic changes that reprogram IFN responses. Unexpectedly, 8-oxoguanine DNA glycosylase1 (Ogg1) knockout mice show enhanced stimuli-driven IFN expression that confers increased resistance to viral and bacterial infections and allergen challenges. Here, we tested the hypothesis that the DNA repair protein OGG1 recognizes 8-oxoguanine (8-oxoGua) in promoters modulating IFN expression. We found that functional inhibition, genetic ablation, and inactivation by post-translational modification of OGG1 significantly augment IFN-λ expression in epithelial cells infected by human respiratory syncytial virus (RSV). Mechanistically, OGG1 bound to 8-oxoGua in proximity to interferon response elements, which inhibits the IRF3/IRF7 and NF-κB/RelA DNA occupancy, while promoting the suppressor NF-κB1/p50-p50 homodimer binding to the IFN-λ2/3 promoter. In a mouse model of bronchiolitis induced by RSV infection, functional ablation of OGG1 by a small molecule inhibitor (TH5487) enhances IFN-λ production, decreases immunopathology, neutrophilia, and confers antiviral protection. These findings suggest that the ROS-generated epigenetic mark 8-oxoGua via its reader OGG1 serves as a homeostatic thresholding factor in IFN-λ expression. Pharmaceutical targeting of OGG1 activity may have clinical utility in modulating antiviral response. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
11. The transcriptional foundations of interferon‐λ‐mediated endometrial cell to uterine receptivity.
- Author
-
Yao, Kezhen, Fang, Li, and Sun, Yu
- Subjects
- *
ENZYME-linked immunosorbent assay , *GENE expression profiling , *EMBRYO implantation , *IMMUNOLOGICAL tolerance , *MESSENGER RNA - Abstract
Problem: Interferon‐λ (IFN‐λ) is a novel non‐redundant regulator that participates in the fetal–maternal immune interaction, including immune regulation, uterine receptivity, cell migration and adhesion, and endometrium apoptosis. However, the exact transcriptional foundation for endometrial signaling of IFN‐λ is not completely understood, and studies regarding IFN‐λ to implantation failure in vivo are limited. Method of study: The gene expression profile of human endometrial Ishikawa cell line treated with IFN‐λ or IFN‐α (100 ng/mL) for 6 h was analyzed using RNA‐sequencing. Real‐time qPCR, western blotting, and enzyme‐linked immunosorbent assay (ELISA) tests were used to validate these sequencing data. An in vivo IFN‐λ knock‐down mouse pregnancy model was performed, and the phenotype analysis and the intrauterine biomarkers detection were applied with the uterus samples. Results: High levels of messenger RNA (mRNA) were detected for genes previously associated with endometrial receptivity, including LIF, AXL, CRYAB, EPHB2, CCL5, and DDX58, following IFN‐λ treatment. Moreover, the data indicated IFN‐λ reduced pro‐inflammatory gene activity compared with IFN‐α, including members of the ISG, TNF, SP100 and interleukin genes. The in vivo mouse pregnancy model showed that inhibition of intrauterine IFN‐λ results in aberrant epithelial phenotype and significantly decreases the embryo implantation rates and derails normal uterine receptivity. Conclusions: These findings demonstrate the antagonistic and agonistic roles of IFNs in the endometrial cell, suggesting a selective role of IFN‐λ in endometrial receptivity and immunological tolerance regulation. Moreover, the findings provide valuable insight into potential biomarkers related to endometrial receptivity and facilitate an understanding of the molecular changes observed during infertility treatment and contraception usage. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
12. Interleukin-22 facilitates the interferon-λ-mediated production of tripartite motif protein 25 to inhibit replication of duck viral hepatitis A virus type 1.
- Author
-
An, Hao, Liu, Yumei, Shu, Ming, and Chen, Junhao
- Abstract
The innate immune system provides a defense against invading pathogens by inducing various interferon (IFN)-stimulated genes (ISGs). We recently reported that tripartite motif protein 25 (TRIM25), an important ISG, was highly upregulated in duck embryo hepatocyte cells (DEFs) after infection with duck viral hepatitis A virus type 1 (DHAV-1). However, the mechanism of upregulation of TRIM25 remains unknown. Here we reported that interleukin-22 (IL-22), whose expression was highly facilitated in DEFs and various organs of 1-day-old ducklings after DHAV-1 infection, highly enhanced the IFN-λ-induced production of TRIM25. The treatment with IL-22 neutralizing antibody or the overexpression of IL-22 highly suppressed or facilitated TRIM25 expression, respectively. The phosphorylation of signal transducer and activator of transcription 3 (STAT3) was crucial for the process of IL-22 enhancing IFN-λ-induced TRIM25 production, which was suppressed by WP1066, a novel inhibitor of STAT3 phosphorylation. The overexpression of TRIM25 in DEFs resulted in a high production of IFNs and reduced DHAV-1 replication, whereas the attenuated expression of IFNs and facilitated replication of DHAV-1 were observed in the RNAi group, implying that TRIM25 defended the organism against DHAV-1 propagation by inducing the production of IFNs. In summary, we reported that IL-22 activated the phosphorylation of STAT3 to enhance the IFN-λ-mediated TRIM25 expression and provide a defense against DHAV-1 by inducing IFN production. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
13. Role of Innate Interferon Responses at the Ocular Surface in Herpes Simplex Virus-1-Induced Herpetic Stromal Keratitis.
- Author
-
Ren, Jiayi, Antony, Ferrin, Rouse, Barry T., and Suryawanshi, Amol
- Subjects
HERPES simplex ,HUMAN herpesvirus 1 ,HERPES labialis ,PATTERN perception receptors ,KERATITIS - Abstract
Herpes simplex virus type 1 (HSV-1) is a highly successful pathogen that primarily infects epithelial cells of the orofacial mucosa. After initial lytic replication, HSV-1 enters sensory neurons and undergoes lifelong latency in the trigeminal ganglion (TG). Reactivation from latency occurs throughout the host's life and is more common in people with a compromised immune system. HSV-1 causes various diseases depending on the site of lytic HSV-1 replication. These include herpes labialis, herpetic stromal keratitis (HSK), meningitis, and herpes simplex encephalitis (HSE). HSK is an immunopathological condition and is usually the consequence of HSV-1 reactivation, anterograde transport to the corneal surface, lytic replication in the epithelial cells, and activation of the host's innate and adaptive immune responses in the cornea. HSV-1 is recognized by cell surface, endosomal, and cytoplasmic pattern recognition receptors (PRRs) and activates innate immune responses that include interferons (IFNs), chemokine and cytokine production, as well as the recruitment of inflammatory cells to the site of replication. In the cornea, HSV-1 replication promotes type I (IFN-α/β) and type III (IFN-λ) IFN production. This review summarizes our current understanding of HSV-1 recognition by PRRs and innate IFN-mediated antiviral immunity during HSV-1 infection of the cornea. We also discuss the immunopathogenesis of HSK, current HSK therapeutics and challenges, proposed experimental approaches, and benefits of promoting local IFN-λ responses. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
14. Fulminant Viral Hepatitis in Two Siblings with Inherited IL-10RB Deficiency.
- Author
-
Korol, Cecilia B., Belkaya, Serkan, Alsohime, Fahad, Lorenzo, Lazaro, Boisson-Dupuis, Stéphanie, Brancale, Joseph, Neehus, Anna-Lena, Vilarinho, Silvia, Zobaida, Alsum, Halwani, Rabih, Al-Muhsen, Saleh, Casanova, Jean-Laurent, and Jouanguy, Emmanuelle
- Subjects
- *
VIRAL hepatitis , *INFLAMMATORY bowel diseases , *HEPATITIS A virus , *HEPATITIS viruses , *SIBLINGS - Abstract
Fulminant viral hepatitis (FVH) caused by hepatitis A virus (HAV) is a life-threatening disease that typically strikes otherwise healthy individuals. The only known genetic etiology of FVH is inherited IL-18BP deficiency, which unleashes IL-18-dependent lymphocyte cytotoxicity and IFN-γ production. We studied two siblings who died from a combination of early-onset inflammatory bowel disease (EOIBD) and FVH due to HAV. The sibling tested was homozygous for the W100G variant of IL10RB previously described in an unrelated patient with EOIBD. We show here that the out-of-frame IL10RB variants seen in other EOIBD patients disrupt cellular responses to IL-10, IL-22, IL-26, and IFN-λs in overexpression conditions and in homozygous cells. By contrast, the impact of in-frame disease-causing variants varies between cases. When overexpressed, the W100G variant impairs cellular responses to IL-10, but not to IL-22, IL-26, or IFN-λ1, whereas cells homozygous for W100G do not respond to IL-10, IL-22, IL-26, or IFN-λ1. As IL-10 is a potent antagonist of IFN-γ in phagocytes, these findings suggest that the molecular basis of FVH in patients with IL-18BP or IL-10RB deficiency may involve excessive IFN-γ activity during HAV infections of the liver. Inherited IL-10RB deficiency, and possibly inherited IL-10 and IL-10RA deficiencies, confer a predisposition to FVH, and patients with these deficiencies should be vaccinated against HAV and other liver-tropic viruses. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
15. Cytosolic DNA sensor activation inhibits HIV infection of macrophages.
- Author
-
Zhang, Biao, Liu, Jin‐Biao, Zhou, Lina, Wang, Xu, Khan, Shazheb, Hu, Wen‐Hui, and Ho, Wen‐Zhe
- Subjects
HIV infections ,DNA ,HIV ,ARTIFICIAL chromosomes ,MACROPHAGE activation - Abstract
Cytosolic recognition of microbial DNA in macrophages results in the activation of the interferon (IFN)‐dependent antiviral innate immunity. Here, we examined whether activating DNA sensors in peripheral blood monocyte‐derived macrophages (MDMs) can inhibit human immunodeficiency virus (HIV). We observed that the stimulation of MDMs with poly(dA:dT) or poly(dG:dC) (synthetic ligands for the DNA sensors) inhibited HIV infection and replication. MDMs treated with poly(dA:dT) or poly(dG:dC) expressed higher levels of both type I and type III IFNs than untreated cells. Activation of the DNA sensors in MDMs also induced the expression of the multiple intracellular anti‐HIV factors, including IFN‐stimulated genes (ISGs: ISG15, ISG56, Viperin, OAS2, GBP5, MxB, and Tetherin) and the HIV restriction microRNAs (miR‐29c, miR‐138, miR‐146a, miR‐155, miR‐198, and miR‐223). In addition, the DNA sensor activation of MDM upregulated the expression of the CC chemokines (RANTES, MIP‐1α, MIP‐1β), the ligands for HIV entry coreceptor CCR5. These observations indicate that the cytosolic DNA sensors have a protective role in the macrophage intracellular immunity against HIV and that targeting the DNA sensors has therapeutic potential for immune activation‐based anti‐HIV treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
16. Role of Interferons in Mycobacterium tuberculosis Infection.
- Author
-
Shanmuganathan, Gaithrri, Orujyan, Davit, Narinyan, William, Poladian, Nicole, Dhama, Sanya, Parthasarathy, Arpitha, Ha, Alexandra, Tran, Daniel, Velpuri, Prathosh, Nguyen, Kevin H., and Venketaraman, Vishwanath
- Subjects
- *
MYCOBACTERIAL diseases , *MYCOBACTERIUM tuberculosis , *INTERFERONS , *TUBERCULOUS meningitis , *CENTRAL nervous system , *MIDDLE-income countries - Abstract
Considerable measures have been implemented in healthcare institutions to screen for and treat tuberculosis (TB) in developed countries; however, in low- and middle-income countries, many individuals still suffer from TB's deleterious effects. TB is caused by an infection from the Mycobacterium tuberculosis (M. tb) bacteria. Symptoms of TB may range from an asymptomatic latent-phase affecting the pulmonary tract to a devastating active and disseminated stage that can cause central nervous system demise, musculoskeletal impairments, and genitourinary compromise. Following M. tb infection, cytokines such as interferons (IFNs) are released as part of the host immune response. Three main classes of IFNs prevalent during the immune defense include: type I IFN (α and β), type II IFN (IFN-γ), and type III IFN (IFN-λ). The current literature reports that type I IFN plays a role in diminishing the host defense against M. tb by attenuating T-cell activation. In opposition, T-cell activation drives type II IFN release, which is the primary cytokine mediating protection from M. tb by stimulating macrophages and their oxidative defense mechanisms. Type III IFN has a subsidiary part in improving the Th1 response for host cell protection against M. tb. Based on the current evidence available, our group aims to summarize the role that each IFN serves in TB within this literature review. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
17. RpIFN-λ1 alleviates the clinical symptoms of porcine epidemic diarrhea.
- Author
-
Liu X, Xie E, Wang J, Yan L, Tian T, You J, Lu L, Qian Z, Tan Z, Xiong J, Gong L, Zhang G, Luo H, and Wang H
- Abstract
Porcine epidemic diarrhea (PED), caused by the porcine epidemic diarrhea virus (PEDV), primarily affects the jejunum and ileum of pigs. Interferons, glycoproteins with high species specificity and potent antiviral activity, are crucial in defending against viral infections. Unlike other interferons, interferon-lambda (IFN-λ) mainly acts on mucosal epithelial cells and exhibits robust antiviral activity at mucosal surfaces. However, the high cost limits the use of naturally extracted interferons in farming. In this study, we expressed recombinant porcine interferon-lambda 1 (rpIFN-λ1) in eukaryotic cells, demonstrating effective antiviral activity against PEDV in Vero E6 and IPI-FX cells. In vivo, rpIFN-λ1 alleviated clinical symptoms and intestinal damage, enhanced antioxidant capacity, reduced inflammation, and significantly improved the survival rate of piglets following PEDV infection. Both in vitro and in vivo studies confirmed that rpIFN-λ1 upregulated interferon-stimulated genes (ISGs) via the JAK-STAT pathway, thereby exerting antiviral effects. In conclusion, rpIFN-λ1 significantly inhibited PEDV replication and alleviated clinical symptoms. The selectivity of rpIFN-λ1 for intestinal cells and its ability to reduce viral shedding suggest that this agent is a promising antiviral for enteric viruses such as PEDV. Our findings highlight rpIFN-λ1 as a cost-effective, efficient, and novel strategy for antiviral treatment of PEDV., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
18. Association between Psoriasis Disease and IFN-λ Gene Polymorphisms.
- Author
-
Yilmaz, Büşra, Çakmak Genç, Güneş, Karakaş Çelik, Sevim, Solak Tekin, Nilgün, Can, Murat, and Dursun, Ahmet
- Subjects
- *
GENETIC polymorphisms , *DISTRIBUTION (Probability theory) , *PSORIASIS , *SINGLE nucleotide polymorphisms , *T helper cells , *PSORIATIC arthritis - Abstract
Psoriasis is one of the most common chronic immune-mediated skin diseases, having a strong genetic predisposition. Psoriasis is a T-cell-mediated disease with a mixed Th1/Th17 cytokines environment. IL-23/IL-17 axis hyperactivation is the primary pathogenesis. Psoriasis lesions have been known to exhibit high IFN-λ1 and IFN-stimulated genes (ISGs) expression, which appears to be driven by Th17 cells. However, the role and mechanism of IFN-λs in psoriasis disease remains unknown. The study aimed to investigate the relationship between IL-28B and IL-29 gene polymorphisms with psoriasis disease and clinical severity. We performed single-nucleotide polymorphisms (SNPs) of IL-28B rs12979860 (IL-28 C/T), rs8099917 (IL-28 T/G), and IL-29 rs30461 (IL-29 T/C) in 140 patients with psoriasis disease and 159 healthy controls using the polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. The genotype and allele frequency distributions of the rs12979860 (IL-28 C/T) and rs30461 (IL-29 T/C) polymorphisms were similar in the patient and control groups and were not statistically significant. The TG genotype of rs8099917 was statistically significantly different in patients from both groups. The TG genotype increased the risk of disease1.9-fold. The G allele may be associated with the pathogenesis of psoriasis. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
19. Targeted Modulation of Interferon Response-Related Genes with IFN-Alpha/Lambda Inhibition.
- Author
-
Sur, Debpali, Leonova, Katerina, Levi, Bar, Markowitz, Shany Ivon, Cohen-Harazi, Raichel, Gitlin, Ilya, Gurova, Katerina, Gudkov, Andrei, Pinhasov, Albert, Koman, Igor, and Nesher, Elimelech
- Subjects
- *
INTERFERONS , *DOUBLE-stranded RNA , *ANTI-inflammatory agents , *ANTINEOPLASTIC agents , *GENES , *INFLAMMATION , *TYPE I interferons - Abstract
Interferon (IFN) signaling resulting from external or internal inflammatory processes initiates the rapid release of cytokines and chemokines to target viral or bacterial invasion, as well as cancer and other diseases. Prolonged exposure to IFNs, or the overexpression of other cytokines, leads to immune exhaustion, enhancing inflammation and leading to the persistence of infection and promotion of disease. Hence, to control and stabilize an excessive immune response, approaches for the management of inflammation are required. The potential use of peptides as anti-inflammatory agents has been previously demonstrated. Our team discovered, and previously published, a 9-amino-acid cyclic peptide named ALOS4 which exhibits anti-cancer properties in vivo and in vitro. We suggested that the anti-cancer effect of ALOS4 arises from interaction with the immune system, possibly through the modulation of inflammatory processes. Here, we show that treatment with ALOS4 decreases basal cytokine levels in mice with chronic inflammation and prolongs the lifespan of mice with acute systemic inflammation induced by irradiation. We also show that pretreatment with ALOS4 reduces the expression of IFN alpha, IFN lambda, and selected interferon-response genes triggered by polyinosinic-polycytidylic acid (Poly I:C), a synthetic analog of viral double-stranded RNA, while upregulating the expression of other genes with antiviral activity. Hence, we conclude that ALOS4 does not prevent IFN signaling, but rather supports the antiviral response by upregulating the expression of interferon-response genes in an interferon-independent manner. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
20. De novo developed protein binders mimicking Interferon lambda signaling.
- Author
-
Kolářová, Lucie, Zahradník, Jiří, Huličiak, Maroš, Mikulecký, Pavel, Peleg, Yoav, Shemesh, Maya, Schreiber, Gideon, and Schneider, Bohdan
- Subjects
- *
CELL receptors , *INTERFERONS , *PROTEINS , *SCAFFOLD proteins - Abstract
We hereby describe the process of design and selection of nonantibody protein binders mimicking cytokine signaling. We chose to mimic signaling of IFN‐λ1, type 3 interferon (also known as IL‐29) for its novelty and the importance of its biological functions. All four known interferons λ signal through binding to the extracellular domains of IL‐28 receptor 1 (IL‐28R1) and IL‐10 receptor 2 (IL‐10R2). Our binders were therefore trained to bind both receptors simultaneously. The bifunctional binder molecules were developed by yeast display, a method of directed evolution. The signaling capacity of the bivalent binders was tested by measuring phosphorylation of the JAK/STAT signaling pathway and production of mRNA of six selected genes naturally induced by IFN‐ λ1 in human cell lines. The newly developed bivalent binders offer opportunities to study cytokine‐related biological functions and modulation of the cell behavior by receptor activation on the cell surfaces alternative to the use of natural IFN‐λ. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
21. Intestinal antiviral signaling is controlled by autophagy gene Epg5 independent of the microbiota.
- Author
-
Lee, Sanghyun, Kalugotla, Gowri, Ingle, Harshad, Rodgers, Rachel, Wu, Chunyan, Wang, Yating, Li, Yuhao, Yang, Xia, Zhang, Jin, Borella, Nicolette R., Deng, Hongju, Droit, Lindsay, Hill, Ryan, Peterson, Stefan T., Desai, Chandni, Lawrence, Dylan, Lu, Qun, and Baldridge, Megan T.
- Subjects
FISHER discriminant analysis ,AUTOPHAGY ,INTERFERON gamma ,ENTEROVIRUSES ,INTERFERON receptors ,TUMOR necrosis factors ,ANAPLASTIC lymphoma kinase ,INTERLEUKIN receptors - Abstract
Mutations in the macroautophagy/autophagy gene EPG5 are responsible for Vici syndrome, a human genetic disease characterized by combined immunodeficiency. Previously, we found that epg5
−/- mice exhibit hyperinflammation in the lungs mediated by IL1B/IL-1β and TNF/TNFα, resulting in resistance to influenza. Here, we find that disruption of Epg5 results in protection against multiple enteric viruses including norovirus and rotavirus. Gene expression analysis reveals IFNL/IFN-λ responsive genes as a key alteration. Further, mice lacking Epg5 exhibit substantial alterations of the intestinal microbiota. Surprisingly, germ-free mouse studies indicate Epg5-associated inflammation of both the intestine and lung is microbiota-independent. Genetic studies support IFNL signaling as the primary mediator of resistance to enteric viruses, but not of microbial dysbiosis, in epg5−/- mice. This study unveils an important role, unexpectedly independent of the microbiota, for autophagy gene Epg5 in host organism protection by modulating intestinal IFNL responses. Abbreviations: CTNNB1: catenin (cadherin associated protein), beta 1; DAPI: 4′,6-diamidino-2-phenylindole; EPG5: ectopic P-granules autophagy protein 5 homolog (C. elegans); FT: fecal transplant; IFI44: interferon-induced protein 44; IFIT1: interferon-induced protein with tetratricopeptide repeats 1; IFNG/IFN-γ: interferon gamma; IFNL/IFN-λ: interferon lambda; IFNLR1: interferon lambda receptor 1; IL1B/IL-1β: interleukin 1 beta; ISG: interferon stimulated gene; GF: germ-free; LEfSe: linear discriminant analysis effect size; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MNoV: murine norovirus; MX2: MX dynamin-like GTPase 2; OAS1A: 2'-5' oligoadenylate synthetase 1A; RV: rotavirus; SPF: specific-pathogen free; SQSTM1/p62: sequestosome 1; STAT1: signal transducer and activator of transcription 1; STING1: stimulator of interferon response cGAMP interactor 1; TBK1: TANK-binding kinase 1; TNF/TNFα: tumor necrosis factor [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
22. Interleukin-10 Family Cytokines Immunobiology and Structure
- Author
-
Wei, Huaxing, Li, Bofeng, Sun, Anyuan, Guo, Feng, Cohen, Irun R., Editorial Board Member, Lajtha, Abel, Editorial Board Member, Paoletti, Rodolfo, Editorial Board Member, Lambris, John D., Editorial Board Member, Rezaei, Nima, Editorial Board Member, Jin, Tengchuan, editor, and Yin, Qian, editor
- Published
- 2019
- Full Text
- View/download PDF
23. Interferon Lambda Signals in Maternal Tissues to Exert Protective and Pathogenic Effects in a Gestational Stage-Dependent Manner
- Author
-
Rebecca L. Casazza, Drake T. Philip, and Helen M. Lazear
- Subjects
IFN ,IFN-λ ,interferon ,Zika virus ,congenital infection ,pregnancy ,Microbiology ,QR1-502 - Abstract
ABSTRACT Interferon lambda (IFN-λ) (type III IFN) is constitutively secreted from human placental cells in culture and reduces Zika virus (ZIKV) transplacental transmission in mice. However, the roles of IFN-λ during healthy pregnancy and in restricting congenital infection remain unclear. Here, we used mice lacking the IFN-λ receptor (Ifnlr1−/−) to generate pregnancies lacking either maternal or fetal IFN-λ responsiveness and found that the antiviral effect of IFN-λ resulted from signaling exclusively in maternal tissues. This protective effect depended on gestational stage, as infection earlier in pregnancy (E7 rather than E9) resulted in enhanced transplacental transmission of ZIKV. In Ifnar1−/− dams, which sustain robust ZIKV infection, maternal IFN-λ signaling caused fetal resorption and intrauterine growth restriction. Pregnancy pathology elicited by poly(I·C) treatment also was mediated by maternal IFN-λ signaling, specifically in maternal leukocytes, and also occurred in a gestational stage-dependent manner. These findings identify an unexpected effect of IFN-λ signaling, specifically in maternal (rather than placental or fetal) tissues, which is distinct from the pathogenic effects of IFN-αβ (type I IFN) during pregnancy. These results highlight the complexity of immune signaling at the maternal-fetal interface, where disparate outcomes can result from signaling at different gestational stages. IMPORTANCE Pregnancy is an immunologically complex situation, which must balance protecting the fetus from maternal pathogens with preventing maternal immune rejection of non-self fetal and placental tissue. Cytokines, such as interferon lambda (IFN-λ), contribute to antiviral immunity at the maternal-fetal interface. We found in a mouse model of congenital Zika virus infection that IFN-λ can have either a protective antiviral effect or cause immune-mediated pathology, depending on the stage of gestation when IFN-λ signaling occurs. Remarkably, both the protective and pathogenic effects of IFN-λ occurred through signaling exclusively in maternal immune cells rather than in fetal or placental tissues or in other maternal cell types, identifying a new role for IFN-λ at the maternal-fetal interface.
- Published
- 2022
- Full Text
- View/download PDF
24. Role of Innate Interferon Responses at the Ocular Surface in Herpes Simplex Virus-1-Induced Herpetic Stromal Keratitis
- Author
-
Jiayi Ren, Ferrin Antony, Barry T. Rouse, and Amol Suryawanshi
- Subjects
HSV-1 ,HSK ,HSE ,IFN-α ,IFN-β ,IFN-λ ,Medicine - Abstract
Herpes simplex virus type 1 (HSV-1) is a highly successful pathogen that primarily infects epithelial cells of the orofacial mucosa. After initial lytic replication, HSV-1 enters sensory neurons and undergoes lifelong latency in the trigeminal ganglion (TG). Reactivation from latency occurs throughout the host’s life and is more common in people with a compromised immune system. HSV-1 causes various diseases depending on the site of lytic HSV-1 replication. These include herpes labialis, herpetic stromal keratitis (HSK), meningitis, and herpes simplex encephalitis (HSE). HSK is an immunopathological condition and is usually the consequence of HSV-1 reactivation, anterograde transport to the corneal surface, lytic replication in the epithelial cells, and activation of the host’s innate and adaptive immune responses in the cornea. HSV-1 is recognized by cell surface, endosomal, and cytoplasmic pattern recognition receptors (PRRs) and activates innate immune responses that include interferons (IFNs), chemokine and cytokine production, as well as the recruitment of inflammatory cells to the site of replication. In the cornea, HSV-1 replication promotes type I (IFN-α/β) and type III (IFN-λ) IFN production. This review summarizes our current understanding of HSV-1 recognition by PRRs and innate IFN-mediated antiviral immunity during HSV-1 infection of the cornea. We also discuss the immunopathogenesis of HSK, current HSK therapeutics and challenges, proposed experimental approaches, and benefits of promoting local IFN-λ responses.
- Published
- 2023
- Full Text
- View/download PDF
25. The immunobiology of corneal HSV-1 infection and herpetic stromal keratitis.
- Author
-
Antony F, Kinha D, Nowińska A, Rouse BT, and Suryawanshi A
- Subjects
- Humans, Antiviral Agents therapeutic use, Animals, Keratitis, Herpetic immunology, Keratitis, Herpetic virology, Herpesvirus 1, Human immunology, Herpesvirus 1, Human physiology
- Abstract
SUMMARYHuman alphaherpesvirus 1 (HSV-1) is a highly successful neurotropic pathogen that primarily infects the epithelial cells lining the orofacial mucosa. After primary lytic replication in the oral, ocular, and nasal mucosal epithelial cells, HSV-1 establishes life-long latency in neurons within the trigeminal ganglion. Patients with compromised immune systems experience frequent reactivation of HSV-1 from latency, leading to virus entry in the sensory neurons, followed by anterograde transport and lytic replication at the innervated mucosal epithelial surface. Although recurrent infection of the corneal mucosal surface is rare, it can result in a chronic immuno-inflammatory condition called herpetic stromal keratitis (HSK). HSK leads to gradual vision loss and can cause permanent blindness in severe untreated cases. Currently, there is no cure or successful vaccine to prevent latent or recurrent HSV-1 infections, posing a significant clinical challenge to managing HSK and preventing vision loss. The conventional clinical management of HSK primarily relies on anti-virals to suppress HSV-1 replication, anti-inflammatory drugs (such as corticosteroids) to provide symptomatic relief from pain and inflammation, and surgical interventions in more severe cases to replace damaged cornea. However, each clinical treatment strategy has limitations, such as local and systemic drug toxicities and the emergence of anti-viral-resistant HSV-1 strains. In this review, we summarize the factors and immune cells involved in HSK pathogenesis and highlight alternate therapeutic strategies for successful clinical management of HSK. We also discuss the therapeutic potential of immunoregulatory cytokines and immunometabolism modulators as promising HSK therapies against emerging anti-viral-resistant HSV-1 strains., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
26. PGAP3 regulates human bronchial epithelial cell mRNAs present in asthma and respiratory virus reference data sets.
- Author
-
Leslie E, Miller M, Lafuze A, Svyatskaya S, Choi GS, and Broide DH
- Abstract
PGAP3 is a glycosylphosphatidylinositol (GPI) phospholipase gene localized within chromosome 17q12-21, a region highly linked to asthma. Although much is known about the function of other chromosome 17q12-21 genes expressed at increased levels in bronchial epithelium such as ORMDL3 and GSDMB, little is known about the function of increased PGAP3 expression in bronchial epithelium in the context of asthma. The aim of this study was therefore to determine whether increased PGAP3 expression in human bronchial epithelial cells regulated expression of mRNA pathways important to the pathogenesis of asthma by utilizing RNA-sequencing and bioinformatic analysis. We performed RNA-sequencing on normal human bronchial epithelial cells transfected with PGAP3 for 24 and 48 hours. PGAP3 regulated genes were compared to asthma and respiratory virus (influenza A, rhinovirus, respiratory syncytial virus) reference data sets to identify PGAP3 target genes and pathways. Approximately 9% of the upregulated PGAP3-induced genes were found in an asthma reference data set, 41% in a rhinovirus reference data set, 33% in an influenza A reference data set, and 3% in a respiratory syncytial virus reference data set. PGAP3 significantly upregulated the expression of several genes associated with the innate immune response and viral signatures of respiratory viruses associated with asthma exacerbations. Two of the highest expressed genes induced by PGAP3 are RSAD2, OASL, and IFN-λ, which are anti-viral genes associated with asthma. PGAP3 also upregulated the antiviral gene BST2, which like PGAP3 is a GPI-anchored protein. We conclude that PGAP3 expression in human bronchial epithelial cells regulates expression of genes known to be linked to asthma, and also regulates the bronchial epithelial expression of genes pertinent to the pathogenesis of respiratory viral triggered asthma exacerbations., Competing Interests: Statement of Competing Interests Authors did not receive any third-party payments or services that influenced the submitted work.
- Published
- 2024
- Full Text
- View/download PDF
27. SARS-CoV-2 Infection Prompts IL-1β-Mediated Inflammation and Reduces IFN-λ Expression in Human Lung Tissue
- Author
-
Bianca Vezzani, Margherita Neri, Stefano D’Errico, Alberto Papi, Marco Contoli, and Carlotta Giorgi
- Subjects
SARS-CoV-2 ,COVID-19 ,IFN-λ ,IL-1β ,IL-6 ,lungs ,Medicine - Abstract
Two years after its spreading, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is still responsible for more than 2000 deaths per day worldwide, despite vaccines and monoclonal antibody countermeasures. Therefore, there is a need to understand the immune–inflammatory pathways that prompt the manifestation of the disease to identify a novel potential target for pharmacological intervention. In this context, the characterization of the main players in the SARS-CoV-2-induced cytokine storm is mandatory. To date, the most characterized have been IL-6 and the class I and II interferons, while less is known about the proinflammatory cytokine IL-1β and class III interferons. Here, we report a preliminary study aimed at the characterization of the lung inflammatory context in COVID-19 patients, with a special focus on IFN-λ and IL-1β. By investigating IFN and inflammatory cytokine patterns by IHC in 10 deceased patients due to COVID-19 infection, compared to 10 control subjects, we reveal that while IFN-β production was increased in COVID-19 patients, IFN-λ was almost abolished. At the same time, the levels of IL-1β were dramatically improved, while IL-6 lung levels seem to be unaffected by the infection. Our findings highlight a central role of IL-1β in prompting lung inflammation after SARS-CoV-2 infection. Together, we show that IFN-λ is negatively affected by viral infection, supporting the idea that IFN-λ administration together with the pharmaceutical blockage of IL-1β represents a promising approach to revert the COVID-19-induced cytokine storm.
- Published
- 2022
- Full Text
- View/download PDF
28. Herpes simplex virus-1 (HSV-1) infection induces a potent but ineffective IFN-λ production in immune cells of AD and PD patients
- Author
-
Francesca La Rosa, Simone Agostini, Anna Bianchi, Raffaello Nemni, Federica Piancone, Ivana Marventano, Roberta Mancuso, Marina Saresella, and Mario Clerici
- Subjects
Alzheimer’s disease ,HSV-1 ,Immune response ,IFN-λ ,Parkinson’s disease ,Medicine - Abstract
Abstract Background The sequential activation of immediate early (IE), early (E) and late (L) genes is required to allow productive herpes simplex virus type 1 (HSV-1) infection. Several evidences suggest that, together with inflammation, an immunological response incapable to counteract HSV-1 reactivation plays a role in the pathogenesis of Alzheimer’s (AD) and Parkinson’s (PD) diseases. IFN-lambda (IFN-λ), a cytokine endowed with a robust antiviral activity, contains HSV-1 reactivation. HSV-1-induced IFN-λ, IL-10 and IL-1β as well as the expression of viral IE, E and L genes were analyzed in vitro in peripheral blood mononuclear cells (PBMC) of AD and PD patients as well as of healthy controls (HC). Methods PBMC of AD, PD and HC were in vitro infected with one multiplicity of infection (1 MOI) HSV-1. IE, E, and L viral genes transcription as well as IFN-λ, IL-10 and IL-1β production were analyzed. Results In HSV-1-infected cells of AD and PD patients compared to HC: (1) transcription of IE (ICP0, ICP27) genes was reduced whereas that of E (UL41, UL29) and L (UL48, LAT) genes was increased; (2) IFN-λ mRNA expression was increased. IL-1β was augmented and IL-10 was reduced in unstimulated cells of AD and PD compared to HC; HSV-1 infection significantly increased IL-10 production in HC alone. Conclusions Data herein show that a proinflammatory condition is present in AD and PD, in whom attempts to obstacle viral replication via an initial, possibly more potent IFN-λ-mediated control of IE viral genes is unsuccessful.
- Published
- 2019
- Full Text
- View/download PDF
29. NS7a of SADS-CoV promotes viral infection via inducing apoptosis to suppress type III interferon production.
- Author
-
Wang X, Qiu W, Hu G, Diao X, Li Y, Li Y, Li P, Liu Y, Feng Y, Xue C, Cao Y, and Xu Z
- Subjects
- Animals, Swine, Humans, Virus Replication, Interferon Lambda, Coronavirus Infections virology, Coronavirus Infections metabolism, Alphacoronavirus metabolism, Caspase 3 metabolism, Swine Diseases virology, Swine Diseases metabolism, Vero Cells, Signal Transduction, Chlorocebus aethiops, HEK293 Cells, Apoptosis, Interferons metabolism, Immunity, Innate, Interferon Regulatory Factor-3 metabolism, Viral Nonstructural Proteins metabolism
- Abstract
Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a newly discovered swine coronavirus with potential cross-species transmission risk. Although SADS-CoV-induced host cell apoptosis and innate immunity antagonization has been revealed, underlying signaling pathways remain obscure. Here, we demonstrated that infection of SADS-CoV induced apoptosis in vivo and in vitro , and that viral protein NS7a is mainly responsible for SADS-CoV-induced apoptosis in host cells. Furthermore, we found that NS7a interacted with apoptosis-inducing factor mitochondria associated 1 (AIFM1) to activate caspase-3 via caspase-6 in SADS-CoV-infected cells, and enhanced SADS-CoV replication. Importantly, NS7a suppressed poly(I:C)-induced expression of type III interferon (IFN-λ) via activating caspase-3 to cleave interferon regulatory factor 3 (IRF3), and caspase-3 inhibitor protects piglets against SADS-CoV infection in vivo . These findings reveal how SADS-CoV induced apoptosis to inhibit innate immunity and provide a valuable clue to the development of effective drugs for the clinical control of SADS-CoV infection.IMPORTANCEOver the last 20 years, multiple animal-originated coronaviruses, including severe acute respiratory syndrome coronavirus (SARS-CoV), middle east respiratory syndrome coronavirus (MERS-CoV), and SARS-CoV-2, have caused millions of deaths, seriously jeopardized human health, and hindered social development, indicating that the study of animal-originated coronaviruses with potential for cross-species transmission is particularly important. Bat-originated swine acute diarrhea syndrome coronavirus (SADS-CoV), discovered in 2017, can not only cause fatal diarrhea in piglets, but also infect multiple human cells, with a potential risk of cross-species transmission, but its pathogenesis is unclear. In this study, we demonstrated that NS7a of SADS-CoV suppresses IFN-λ production via apoptosis-inducing factor mitochondria associated 1 (AIFM1)-caspase-6-caspase-3-interferon regulatory factor 3 (IRF3) pathway, and caspase-3 inhibitor (Z-DEVD-FMK) can effectively inhibit SADS-CoV replication and protect infected piglets. Our findings in this study contribute to a better understanding of SADS-CoV-host interactions as a part of the coronaviruses pathogenesis and using apoptosis-inhibitor as a drug as potential therapeutic approaches for prevention and control of SADS-CoV infection., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
30. Dectin-1 Promotes Type I and III Interferon Expression to Support Optimal Antifungal Immunity in the Lung
- Author
-
Orchi Dutta, Vanessa Espinosa, Keyi Wang, Samantha Avina, and Amariliz Rivera
- Subjects
Aspergillus ,dectin-1 ,β-glucan ,cell wall ,innate immunity ,IFN-λ ,Microbiology ,QR1-502 - Abstract
Pulmonary infections with Aspergillus fumigatus (Af) are a significant cause of invasive fungal disease and lead to high morbidity and mortality in diverse populations throughout the world. Currently available antifungal drugs are often ineffective, thus contributing to unacceptably high mortality rates in patients suffering from invasive fungal infections. The use of cytokines as adjunctive immune therapies holds the promise of significantly improving patient outcomes in the future. In recent studies, we identified an essential role for type I and III interferons as regulators of optimal antifungal responses by pulmonary neutrophils during infection with Af. Although various membrane and cytosolic nucleic acid sensors are known to regulate interferon production in response to viruses, the pathways that regulate the production of these cytokines during fungal infection remain uncovered. In the current study, we demonstrate that dectin-1-mediated recognition of β-glucan on the cell wall of the clinically relevant fungal pathogen Aspergillus fumigatus promotes the activation of a protective cascade of type I and III interferon expression. We further demonstrate that exogenous administration of type I and III interferons can rescue inadequate antifungal responses in dectin-1−/− mice, suggesting the potential therapeutic benefit of these cytokines as activators of antifungal defense in the context of innate defects.
- Published
- 2020
- Full Text
- View/download PDF
31. The Coronavirus PEDV Evades Type III Interferon Response Through the miR-30c-5p/SOCS1 Axis
- Author
-
Changlin Wang, Lingling Shan, Shuxin Qu, Mei Xue, Keliang Wang, Fang Fu, Lu Wang, Ziqi Wang, Li Feng, Wanhai Xu, and Pinghuang Liu
- Subjects
coronavirus ,PEDV ,IFN-λ ,SOCS1 ,microRNA ,miR-30c-5p ,Microbiology ,QR1-502 - Abstract
Porcine epidemic diarrhea virus (PEDV) is an economically important pathogen that has evolved several mechanisms to evade type I IFN responses. Type III interferon (IFN-λ), an innate cytokine that primarily targets the mucosal epithelia, is critical in fighting mucosal infection in the host and has been reported to potently inhibit PEDV infection in vitro. However, how PEDV escapes IFN-λ antiviral response remains unclear. In this study, we found that PEDV infection induced significant IFN-λ expression in type I IFN-defective Vero E6 cells, but virus-induced endogenous IFN-λ did not reduce PEDV titers. Moreover, we demonstrated that PEDV escaped IFN-λ responses by substantially upregulating the suppressor of cytokine signaling protein 1 (SOCS1) expression, which impaired the induction of IFN-stimulated genes (ISGs) and dampened the IFN-λ antiviral response and facilitated PEDV replication in Vero E6 cells. We further showed that PEDV infection increased SOCS1 expression by decreasing host miR-30c-5p expression. MiR-30c-5p suppressed SOCS1 expression through targeting the 3′ untranslated region (UTR) of SOCS1. The inhibition of IFN-λ elicited ISGs expression by SOCS1 was specifically rescued by overexpression of miR-30c-5p. Collectively, our findings identify a new strategy by PEDV to escape IFN-λ-mediated antiviral immune responses by engaging the SOCS1/miR-30c axis, thus improving our understanding of its pathogenesis.
- Published
- 2020
- Full Text
- View/download PDF
32. The Coronavirus PEDV Evades Type III Interferon Response Through the miR-30c-5p/SOCS1 Axis.
- Author
-
Wang, Changlin, Shan, Lingling, Qu, Shuxin, Xue, Mei, Wang, Keliang, Fu, Fang, Wang, Lu, Wang, Ziqi, Feng, Li, Xu, Wanhai, and Liu, Pinghuang
- Subjects
TYPE I interferons ,COVID-19 ,PORCINE epidemic diarrhea virus ,SUPPRESSORS of cytokine signaling ,INTERFERONS ,STARTLE reaction - Abstract
Porcine epidemic diarrhea virus (PEDV) is an economically important pathogen that has evolved several mechanisms to evade type I IFN responses. Type III interferon (IFN-λ), an innate cytokine that primarily targets the mucosal epithelia, is critical in fighting mucosal infection in the host and has been reported to potently inhibit PEDV infection in vitro. However, how PEDV escapes IFN-λ antiviral response remains unclear. In this study, we found that PEDV infection induced significant IFN-λ expression in type I IFN-defective Vero E6 cells, but virus-induced endogenous IFN-λ did not reduce PEDV titers. Moreover, we demonstrated that PEDV escaped IFN-λ responses by substantially upregulating the suppressor of cytokine signaling protein 1 (SOCS1) expression, which impaired the induction of IFN-stimulated genes (ISGs) and dampened the IFN-λ antiviral response and facilitated PEDV replication in Vero E6 cells. We further showed that PEDV infection increased SOCS1 expression by decreasing host miR-30c-5p expression. MiR-30c-5p suppressed SOCS1 expression through targeting the 3′ untranslated region (UTR) of SOCS1. The inhibition of IFN-λ elicited ISGs expression by SOCS1 was specifically rescued by overexpression of miR-30c-5p. Collectively, our findings identify a new strategy by PEDV to escape IFN-λ-mediated antiviral immune responses by engaging the SOCS1/miR-30c axis, thus improving our understanding of its pathogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
33. Enterovirus D68 vRNA induces type III IFN production via MDA5.
- Author
-
Chio, Chi-Chong, Chan, Hio-Wai, Chen, Shih-Hsiang, and Huang, Hsing-I
- Subjects
- *
TYPE I interferons , *EPITHELIAL cells , *RESPIRATORY infections , *MYELITIS , *INTERFERONS - Abstract
• EV-D68 infection promotes IFN-λ expression in Calu-3 cells. • MDA5 recognizes EV-D68 vRNA to stimulate the expression of IFN-λ. • EV-D68 infection induces the expression of IFN-λ and activates the MDA5-IRF3/IRF7 pathway. • IFN-λ triggers the expression of ISGs, which decreases EV-D68 infection. Enterovirus D68 (EV-D68) primarily spreads through the respiratory tract and causes respiratory symptoms in children and acute flaccid myelitis (AFM). Type III interferons (IFNs) play a critical role in inhibiting viral growth in respiratory epithelial cells. However, the mechanism by which EV-D68 induces type III IFN production is not yet fully understood. In this study, we show that EV-D68 infection stimulates Calu-3 cells to secrete IFN-λ. The transfection of EV-D68 viral RNA (vRNA) stimulated IFN-λ via MDA5. Furthermore, our findings provide evidence that EV-D68 infection also induces MDA5-IRF3/IRF7-mediated IFN-λ. In addition, we discovered that EV-D68 infection downregulated MDA5 expression. Knockdown of MDA5 increased EV-D68 replication in Calu-3 cells. Finally, we demonstrated that the IFN-λ1 and IFN-λ2/3 proteins effectively inhibit EV-D68 infection in respiratory epithelial cells. In summary, our study shows that EV-D68 induces type III IFN production via the activated MDA5-IRF3/IRF7 pathway and that type III IFNs inhibit EV-D68 replication in Calu-3 cells. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
34. Interleukin 28A.rs12980602 and interleukin 28B.rs8103142 genotypes could be protective against HCV infection among Egyptians.
- Author
-
Zakaria, Zainab A., Knapp, Susanne, Hashem, Mohamed, Zaghla, Hassan, Thursz, Mark, Waked, Imam, and Abdelwahab, Sayed
- Abstract
Previous studies showed that interleukin (IL)-28B gene polymorphisms were associated with hepatitis C Virus (HCV) infection and treatment outcomes. We tested whether single-nucleotide polymorphisms (SNPs) in IL-28A and IL-28B are associated with HCV infection among Egyptians with HCV genotype 4 infections. We enrolled 144 chronic HCV patients, 72 spontaneously resolved HCV subjects, and 69 healthy controls. Four SNPs in IL-28A and IL-28B genes (IL-28A.rs12980602, IL-28B.rs12979860, IL-28B.rs8099917, and IL-28B.rs8103142) were genotyped. The most frequent IL-28B haplotype "TCT" was significantly more frequent in HCV-infected subjects than in HCV negative subjects (62.2% vs. 48.6%, respectively; p = 0.005). The frequency of IL-28A.rs12980602 "T" allele was significantly higher than the "C" allele in healthy controls compared to HCV-infected subjects (p < 0.001) with the "TT" genotype significantly higher in healthy controls compared to HCV-infected subjects (p < 0.001) with no association with viral load (p = 0.11) among chronically infected subjects. The results, also, confirmed the previous role of IL-28B SNPs in predicting HCV infection outcome. Importantly, IL-28B.rs8099917 "TT" genotype was significantly associated with low viral load in HCV-infected subjects, while the remaining three SNPs did not. The three IL-28B SNPs were in linkage disequilibrium (D′ > 0.68; r
2 > 0.43) for all comparisons in HCV patients, while there was no linkage disequilibrium of IL-28A polymorphisms and the three IL-28B SNPs. In conclusion, IL-28A.rs12980602 and IL-28B.rs8103142 TT genotype could be protective against HCV infection. Also, IL-28B.rs12979860, IL-28B.rs8099917, and IL-28B.rs8103142 SNPs predicted the outcome of HCV infection among genotype-4-infected Egyptians. Moreover, IL-28B.rs8099917 SNP affected the viral load in chronic HCV patients. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
35. Interferon Lambda: Toward a Dual Role in Cancer.
- Author
-
Lasfar, Ahmed, Zloza, Andrew, Silk, Ann W., Lee, Leonard Y., and Cohen-Solal, Karine A.
- Subjects
- *
INTERFERONS , *CANCER treatment , *IMMUNOLOGY , *HOMEOSTASIS , *CELLULAR signal transduction , *JANUS kinases - Abstract
Interferon (IFN)-λ, a type III interferon (IFN), is a member of a new family of pleotropic cytokines that share high similarity with classical IFNs α and β (IFN-α/β), type I IFNs. IFN-λ acts as an antiviral agent and displays distinct biological functions, including tumor suppression. Although it activates the common Janus kinase (JAK) and signal transducer and activator of transcription (STAT) pathways, similar to IFN-α/β, IFN-λ differentially induces the expression of IFN-stimulated genes (ISGs). Novel evidence indicates that IFN-λ acts quite differently from IFN-α/β under both homeostasis and pathological situations. In contrast to IFN-α/β, IFN-λ is not involved in over-stimulation of the immune response or exacerbation of inflammation. However, the emergence of unexpected characteristics of IFN-λ, in the control of inflammation and promotion of immune suppression and cancer, reveals novel challenges and offers more strategic opportunities in the context of cancer and beyond. In this article, we discuss new evidence and potential consequences associated with the biology of IFN-λ and provide a different vision for building novel therapeutic strategies in oncology. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
36. Design and evaluation of novel interferon lambda analogs with enhanced antiviral activity and improved drug attributes
- Author
-
Yu D, Zhao M, Dong L, Zhao L, Zou M, Sun H, Zhang M, Liu H, and Zou Z
- Subjects
type III interferon ,IFN-λ ,IL-29 ,IL-28B ,analog ,antiviral ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Debin Yu,1 Mingzhi Zhao,2 Liwei Dong,1 Lu Zhao,1 Mingwei Zou,3 Hetong Sun,4 Mengying Zhang,4 Hongyu Liu,4 Zhihua Zou1 1National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, 2State Key Laboratory of Proteomics, National Engineering Research Center for Protein Drugs, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, People’s Republic of China; 3Department of Psychology, College of Liberal Arts and Social Sciences, University of Houston, Houston, TX, USA; 4Prosit Sole Biotechnology, Co., Ltd., Beijing, People’s Republic of China Abstract: Type III interferons (IFNs) (also called IFN-λ: IFN-λ1, IFN-λ2, IFN-λ3, and IFN-λ4) are critical players in the defense against viral infection of mucosal epithelial cells, where the activity of type I IFNs is weak, and unlike type I IFNs that are associated with severe and diverse side effects, type III IFNs cause minimal side effects due to the highly restricted expression of their receptors, and thus appear to be promising agents for the treatment and prevention of respiratory and gastrointestinal viral infection. However, the antiviral potency of natural type III IFNs is weak compared to type I and, although IFN-λ3 possesses the highest bioactivity among the type III IFNs, IFN-λ1, instead of IFN-λ3, is being developed as a therapeutic drug due to the difficulty to express IFN-λ3 in the prokaryotic expression system. Here, to develop optimal IFN-λ molecules with improved drug attributes, we designed a series of IFN-λ analogs by replacing critical amino acids of IFN-λ1 with the IFN-λ3 counterparts, and vice versa. Four of the designed analogs were successfully expressed in Escherichia coli with high yield and were easily purified from inclusion bodies. Interestingly, all four analogs showed potent activity in inducing the expression of the antiviral genes MxA and OAS and two of them, analog-6 and -7, displayed an unexpected high potency that is higher than that of type I IFN (IFN-α2a) in activating the IFN-stimulated response element (ISRE)-luciferase reporter. Importantly, both analog-6 and -7 effectively inhibited replication of hepatitis C virus in Huh-7.5.1 cells, with an IC50 that is comparable to that of IFN-α2a; and consistent with the roles of IFN-λ in mucosal epithelia, both analogs potently inhibited replication of H3N2 influenza A virus in A549 cells. Together, these studies identified two IFN-λ analogs as candidates to be developed as novel antiviral biologics. Keywords: type III interferon, IFN-λ, IL-29, IL-28B, analog, antiviral
- Published
- 2016
37. Fulminant viral hepatitis in two siblings with inherited IL-10RB deficiency
- Author
-
Cecilia B. Korol, Serkan Belkaya, Fahad Alsohime, Lazaro Lorenzo, Stéphanie Boisson-Dupuis, Joseph Brancale, Anna-Lena Neehus, Silvia Vilarinho, Alsum Zobaida, Rabih Halwani, Saleh Al-Muhsen, Jean-Laurent Casanova, Emmanuelle Jouanguy, and Belkaya, Serkan
- Subjects
Excessive IFN-gamma ,Inborn error of immunity ,IFN-λ ,Immunology ,Fulminant viral hepatitis ,IL-18BP ,Autosomal recessive disease ,Inherited IL-10RB deficiency ,IL-10 ,IL-22 ,Immunology and Allergy ,Hepatitis A virus ,Early-onset inflammatory bowel disease ,IL-18 - Abstract
Fulminant viral hepatitis (FVH) caused by hepatitis A virus (HAV) is a life-threatening disease that typically strikes otherwise healthy individuals. The only known genetic etiology of FVH is inherited IL-18BP deficiency, which unleashes IL-18-dependent lymphocyte cytotoxicity and IFN-γ production. We studied two siblings who died from a combination of early-onset inflammatory bowel disease (EOIBD) and FVH due to HAV. The sibling tested was homozygous for the W100G variant of IL10RB previously described in an unrelated patient with EOIBD. We show here that the out-of-frame IL10RB variants seen in other EOIBD patients disrupt cellular responses to IL-10, IL-22, IL-26, and IFN-λs in overexpression conditions and in homozygous cells. By contrast, the impact of in-frame disease-causing variants varies between cases. When overexpressed, the W100G variant impairs cellular responses to IL-10, but not to IL-22, IL-26, or IFN-λ1, whereas cells homozygous for W100G do not respond to IL-10, IL-22, IL-26, or IFN-λ1. As IL-10 is a potent antagonist of IFN-γ in phagocytes, these findings suggest that the molecular basis of FVH in patients with IL-18BP or IL-10RB deficiency may involve excessive IFN-γ activity during HAV infections of the liver. Inherited IL-10RB deficiency, and possibly inherited IL-10 and IL-10RA deficiencies, confer a predisposition to FVH, and patients with these deficiencies should be vaccinated against HAV and other liver-tropic viruses.
- Published
- 2022
38. Species Specificity of Type III Interferon Activity and Development of a Sensitive Luciferase-Based Bioassay for Quantitation of Mouse Interferon-λ.
- Author
-
Jacobs, Sophie, Wavreil, Fanny, Schepens, Bert, Gad, Hans Henrik, Hartmann, Rune, Rocha-Pereira, Joana, Neyts, Johan, Saelens, Xavier, and Michiels, Thomas
- Subjects
- *
INTERFERONS , *LUCIFERASES , *ENZYME-linked immunosorbent assay , *ANTINEOPLASTIC agents , *ANTIVIRAL agents - Abstract
The type III interferon (IFN-λ) family includes 4 IFN-λ subtypes in man. In the mouse, only the genes coding for IFN-λ2 and -λ3 are present. Unlike mouse and human type I IFNs (IFN-α/β), which exhibit strong species specificity, type III IFNs were reported to act in a cross-specific manner. We reexamined the cross-specificity and observed that mouse and human IFN-λ exhibit some species specificity, although much less than type I IFNs. Mouse IFN-λ3 displayed clear species specificity, being 25-fold less active in human cells than the closely related mouse IFN-λ2. This specificity likely depends on amino acids in α helices A and F that diverged from other IFN-λ sequences. Human IFN-λ4, in contrast, retained high activity in mouse cells. We next developed a firefly luciferase-based reporter cell line, named Fawa-λ-luc, to detect IFN-λ in biological fluids with high specificity and sensitivity. Fawa-λ-luc cells, derived from mouse epithelial cells that are responsive to IFN-λ, were made nonresponsive to type I IFNs by inactivation of the Ifnar2 gene and strongly responsive to IFN-λ by overexpression of the mouse IFNLR1. This bioassay was as sensitive as a commercially available enzyme-linked immunosorbent assay in detecting mouse IFN-λ in cell culture supernatant, as well as in serum and bronchoalveolar lavage samples of virus-infected mice. The assay also enabled the sensitive detection of human IFN-λ activity, including that of the divergent IFN-λ4 with a bias, however, due to variable activity of IFN-λ subtypes. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
39. Induction of innate immune response following introduction of infectious bronchitis virus (IBV) in the trachea and renal tissues of chickens.
- Author
-
Yang, Xin, Li, Jianan, Liu, Hui, Zhang, Peng, Chen, Danyu, Men, Shuai, Li, Xiaocheng, and Wang, Hongning
- Subjects
- *
BRONCHITIS , *BRONCHITIS treatment , *B cells , *GENE expression , *MESSENGER RNA , *DIAGNOSIS - Abstract
Infectious bronchitis (IB) is a highly contagious respiratory disease of chickens, which is caused by the infectious bronchitis virus (IBV). The innate immune response is crucial for antiviral infections and revealing the pathogenic mechanisms of IBV. In this study, we presents an evaluation of interferon (I, II and III IFNs) in renal and tracheal samples from chickens experimentally infected previously vaccinated or not. The results suggest differential expression of chicken interferon, among them type I IFN elaborate a major role in fighting off virus. And vaccine confers greater induction ability of innate immunity thereby vaccination prior infection occurs might be necessary. Above all, we found that IFN-λ also have an effect on IBV infection in trachea besides many other respiratory virus. This study provides the first comprehensive analysis of host-virus interactions of IBV with chicken innate immune response mediated by interferon in various groups. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
40. Ran binding protein 9 (RanBPM) binds IFN-λR1 in the IFN-λ signaling pathway.
- Author
-
Zhang, Junwen, Cong, Xiaojie, Zhaoqiao, Jiajie, Yang, Xia, Li, Meng, Chen, Hong, Mi, Ruifang, Jin, Guishan, Liu, Fusheng, and Huang, Bing-Ren
- Abstract
Like the type I interferons (IFNs), the recently discovered cytokine IFN-λ displays antiviral, antiproliferative, and proapoptotic activities, mediated by a heterodimeric IFN-λ receptor complex composed of a unique IFN-λR1 chain and the IL-10R2 chain. However, the molecular mechanism of the IFN-λ-regulated pathway remains unclear. In this study, we newly identified RAN-binding protein M (RanBPM) as a binding partner of IFN-λR1. The interaction between RanBPM and IFN-λR1 was identified with a glutathione S-transferase pull-down assay and coimmunoprecipitation experiments. IFN-λ1 stimulates this interaction and affects the cellular distribution of RanBPM. However, the interaction between RanBPM and IFN-λR1 does not correlate with their conserved TRAF6-binding sites. Furthermore, we also found that RanBPM is a scaffolding protein with a modulatory function that regulates the activities of IFN-stimulated response elements. Therefore, RanBPM plays a novel role in the IFN-λ-regulated signaling pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
41. Identification of Type III Interferon (IFN-λ) in Chinese Goose: Gene Structure, Age-Dependent Expression Profile, and Antiviral Immune Characteristics In Vivo and In Vitro.
- Author
-
Zhou, Qin, Zhang, Wei, Chen, Shun, Wang, Anqi, Sun, Lipei, Wang, Mingshu, Jia, Renyong, Zhu, Dekang, Liu, Mafeng, Sun, Kunfeng, Yang, Qiao, Wu, Ying, Chen, Xiaoyue, and Cheng, Anchun
- Subjects
- *
SWAN goose , *INTERFERONS , *AMINO acids , *HARDERIAN gland , *MESSENGER RNA , *DISEASES - Abstract
Type III interferons (IFN-λ1/λ2/λ3, also known as IL-29/28A/28B, and IFN-λ4) are a recently discovered interferon group. In this study, we first identified the Chinese goose IFN-λ (goIFN-λ). The full-length sequence of goIFN-λ was found to be 823 bp. There was only one open reading frame that contained 570 bp, and, encoded 189 amino acids. The predicted goIFN-λ protein showed 78%, 67%, and 40% amino acid identity with duIFN-λ, chIFN-λ, and hIFN-λ3, respectively. The tissue distribution of goIFN-λ existed as a parallel distribution with goIFNLR1 as its functional receptor, which was mainly expressed in epithelium-rich tissues, such as lung, gizzard, proventriculus, skin and pancreas, and immune tissues, such as harderian gland and thymus. Furthermore, the immunological characteristics studies of goIFN-λ showed that there was a significant increase in the mRNA at the transcriptional level of goIFN-λ after the peripheral blood mononuclear cells were stimulated with ploy (I:C) and ODN2006, and infected with Gosling plague virus (GPV). In vivo, the mRNA transcriptional level of goIFN-λ increased nearly 20 times in the lung tissue and nearly 40 times in the pancreatic tissue after being artificially infected with H9N2 AIV. It is suggested that goIFN-λ might play a pivotal role in the mucosal immune protection and antiviral defense. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
42. Interferon Gamma Prevents Infectious Entry of Human Papillomavirus 16 via an L2-Dependent Mechanism.
- Author
-
Day, Patricia M., Thompson, Cynthia D., Lowy, Douglas R., and Schiller, John T.
- Subjects
- *
PAPILLOMAVIRUS diseases , *INTERFERON gamma , *ANTIVIRAL agents , *LYSOSOMES , *VIRAL genomes , *THERAPEUTICS - Abstract
In this study, we report that gamma interferon (IFN-λ) treatment, but not IFN-α, -β, or -λ treatment, dramatically decreased infection of human papillomavirus 16 (HPV16) pseudovirus (PsV). In a survey of 20 additional HPV and animal papillomavirus types, we found that many, but not all, PsV types were also inhibited by IFN-λ. Microscopic and biochemical analyses of HPV16 PsV determined that the antiviral effect was exerted at the level of endosomal processing of the incoming capsid and depended on the JAK2/STAT1 pathway. In contrast to infection in the absence of IFN-λ, where L1 proteolytic products are produced during endosomal capsid processing and L2/DNA complexes segregate from L1 in the late endosome and travel to the nucleus, IFN-λ treatment led to decreased L1 proteolysis and retention of L2 and the viral genome in the late endosome/lysosome. PsV sensitivity or resistance to IFN-λ treatment was mapped to the L2 protein, as determined with infectious hybrid PsV, in which the L1 protein was derived from an IFN-λ-sensitive HPV type and the L2 protein from an IFN-λ-insensitive type or vice versa. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
43. Downregulation of autophagy-related gene ATG5 and GABARAP expression by IFN-λ1 contributes to its anti-HCV activity in human hepatoma cells.
- Author
-
Li, Xu, Li, Yu, Fang, Shoucai, Su, Jinming, Jiang, Junjun, Liang, Bingyu, Huang, Jiegang, Zhou, Bo, Zang, Ning, Ho, Wenzhe, Li, Jieliang, Li, Yiping, Chen, Hui, Ye, Li, and Liang, Hao
- Subjects
- *
HEPATITIS C treatment , *INTERFERONS , *ANTIVIRAL agents , *AUTOPHAGY , *GENE expression - Abstract
Type-III interferon (IFN-λ), the most recently discovered family of IFNs, shares common features with type I IFNs, but also has many distinctive activities. It is not clear that whether IFN-λ has additional antiviral mechanisms. In this study, we investigated the effects of IFN-λ on autophagy, a cellular process closely related to hepatitis C virus (HCV) infection in human hepatoma Huh7 cells. Our results showed that IFN-λ1 treatment inhibit autophagic activity in Huh7 cells, as evidenced by the decreased expression of microtubule-associated protein 1 light chain 3B (LC3B)-II and conversion of LC3B-I to LC3B-II, decreased formation of GFP-LC3 puncta and accumulation of autophagosomes. IFN-λ1 could also inhibit HCV-induced or tunicamycin (a known inducer of autophagy with similar mechanism to HCV infection) -induced LC3B-II expression and autophagosome formation. Through PCR array, real time RT PCR, and western blot, two autophagy-related genes, ATG5 and GABARAP, were identified and verified to be down-regulated by IFN-λ1 treatment, either in HCV-uninfected Huh7 cells or in HCV JFH-1-infected cells. Overexpression of ATG5 and/or GABARAP could partly recover the IFN-λ1-inhibited HCV replication. Mechanism research demonstrated that IFN-λ1 could induce the expression of miR-181a and miR-214 (targeting ATG5 and GABARAP respectively), by which down-regulates ATG5 and GABARAP expression. Taken together, our results indicate that suppression of the autophagy response by IFN-λ1 contributes to IFN-λ1 anti-HCV activity. The results also provide a theoretical basis for improving the effectiveness of IFN treatment of HCV infection through inhibition of the HCV-induced autophagy response. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
44. Type I and III Interferon in the Gut: Tight Balance between Host Protection and Immunopathology.
- Author
-
Pott, Johanna and Stockinger, Silvia
- Subjects
INTERFERONS ,MUCOUS membrane diseases ,INTESTINAL abnormalities - Abstract
The intestinal mucosa forms an active interface to the outside word, facilitating nutrient and water uptake and at the same time acts as a barrier toward the highly colonized intestinal lumen. A tight balance of the mucosal immune system is essential to tolerate harmless antigens derived from food or commensals and to effectively defend against potentially dangerous pathogens. Interferons (IFN) provide a first line of host defense when cells detect an invading organism. Whereas type I IFN were discovered almost 60 years ago, type III IFN were only identified in the early 2000s. It was initially thought that type I IFN and type III IFN performed largely redundant functions. However, it is becoming increasingly clear that type III IFN exert distinct and non-redundant functions compared to type I IFN, especially in mucosal tissues. Here, we review recent progress made in unraveling the role of type I/III IFN in intestinal mucosal tissue in the steady state, in response to mucosal pathogens and during inflammation. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
45. IFN-λ Inhibits Drug-Resistant HIV Infection of Macrophages.
- Author
-
Xu Wang, He Wang, Man-Qing Liu, Jie-Liang Li, Run-Hong Zhou, Yu Zhou, Yi-Zhong Wang, Wang Zhou, and Wen-Zhe Ho
- Subjects
INTERFERONS ,DRUG resistance ,HIV infection genetics - Abstract
Type III interferons (IFN-λs) have been demonstrated to inhibit a number of viruses, including HIV. Here, we further examined the anti-HIV effect of IFN-λs in macrophages. We found that IFN-λs synergistically enhanced anti-HIV activity of antiretrovirals [azidothymidine (AZT), efavirenz, indinavir, and enfuvirtide] in infected macrophages. Importantly, IFN-λs could suppress HIV infection of macrophages with the drug-resistant strains, including AZT-resistant virus (A012) and reverse transcriptase inhibitor-resistant virus (TC49). Mechanistically, IFN-λs were able to induce the expression of several important anti-HIV cellular factors, including myxovirus resistance 2 (Mx2), a newly identified HIV post-entry inhibitor and tetherin, a restriction factor that blocks HIV release from infected cells. These observations provide additional evidence to support the potential use of IFN-λs as therapeutics agents for the treatment of HIV infection. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
46. Epigenetic control of type III interferon expression by 8-oxoguanine and its reader 8-oxoguanine DNA glycosylase1.
- Author
-
Xue Y, Pan L, Vlahopoulos S, Wang K, Zheng X, Radak Z, Bacsi A, Tanner L, Brasier AR, Ba X, and Boldogh I
- Subjects
- Animals, Mice, Mice, Knockout, DNA, Epigenesis, Genetic, Interferon Lambda, DNA Glycosylases genetics
- Abstract
Interferons (IFNs) are secreted cytokines with the ability to activate expression of IFN stimulated genes that increase resistance of cells to virus infections. Activated transcription factors in conjunction with chromatin remodelers induce epigenetic changes that reprogram IFN responses. Unexpectedly, 8-oxoguanine DNA glycosylase1 (Ogg1) knockout mice show enhanced stimuli-driven IFN expression that confers increased resistance to viral and bacterial infections and allergen challenges. Here, we tested the hypothesis that the DNA repair protein OGG1 recognizes 8-oxoguanine (8-oxoGua) in promoters modulating IFN expression. We found that functional inhibition, genetic ablation, and inactivation by post-translational modification of OGG1 significantly augment IFN-λ expression in epithelial cells infected by human respiratory syncytial virus (RSV). Mechanistically, OGG1 bound to 8-oxoGua in proximity to interferon response elements, which inhibits the IRF3/IRF7 and NF-κB/RelA DNA occupancy, while promoting the suppressor NF-κB1/p50-p50 homodimer binding to the IFN-λ2/3 promoter. In a mouse model of bronchiolitis induced by RSV infection, functional ablation of OGG1 by a small molecule inhibitor (TH5487) enhances IFN-λ production, decreases immunopathology, neutrophilia, and confers antiviral protection. These findings suggest that the ROS-generated epigenetic mark 8-oxoGua via its reader OGG1 serves as a homeostatic thresholding factor in IFN-λ expression. Pharmaceutical targeting of OGG1 activity may have clinical utility in modulating antiviral response., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Xue, Pan, Vlahopoulos, Wang, Zheng, Radak, Bacsi, Tanner, Brasier, Ba and Boldogh.)
- Published
- 2023
- Full Text
- View/download PDF
47. Interferon-λ in HCV Infection and Therapy
- Author
-
Michael D. Robek and Nicole E. Pagliaccetti
- Subjects
IFN-λ ,IL-28 ,IL-29 ,type III interferon ,IL28B ,IL-28Rα ,Microbiology ,QR1-502 - Abstract
Chronic infection with hepatitis C virus (HCV) is associated with significant liver disease and is therefore an important public health problem. The current standard-of-care therapy for chronic HCV infection consists of a combination of pegylated (PEG) interferon (IFN)-α and ribavirin. Although this therapy effectively generates a sustained viral response in approximately half of treated individuals, it is associated with significant hematological and neurological side effects. A new family of IFN-related proteins (IFN-λ1, 2, and 3; or alternately, IL-29, 28A, 28B, respectively) possesses properties that may make these cytokines superior to PEG-IFN-α for HCV therapy. Genetic studies have also implicated these proteins in both the natural and therapy-induced resolution of HCV infection. This review summarizes the basic aspects of IFN-λ biology, the potential role of these cytokines in HCV infection, and the outlook for their therapeutic application.
- Published
- 2010
- Full Text
- View/download PDF
48. Influence of polymorphic variations of IFNL, HLA, and IL-6 genes in severe cases of COVID-19.
- Author
-
Araújo A, Sgorlon G, Aguiar LE, Cidrão MHMC, Teixeira KS, Villalobos Salcedo JM, Passos-Silva AM, and Vieira D
- Subjects
- Humans, Disease Progression, Genotype, HLA Antigens genetics, Interleukin-6 genetics, Interleukins genetics, Polymorphism, Single Nucleotide genetics, COVID-19 genetics, Liver Neoplasms
- Abstract
The administration of vaccination doses to the global population has led to a decrease in the incidence of COVID-19. However, the clinical picture developed by infected individuals remains extremely concerning due to the great variability in the severity of cases even in vaccinated individuals. The clinical progression of the pathology is characterized by various influential factors such as sex, age group, comorbidities, and the genetics of the individual. The immune response to viral infections can be strongly influenced by the genetics of individuals; nucleotide variations called single-nucleotide polymorphisms (SNPs) in structures involved in the innate and adaptive immune response such as interferon (IFN)-λ, human leukocyte antigen (HLA), and interleukin (IL)-6 are frequently associated with pathological progression. In this study, we conducted a review of the main SNPs of these structures that are associated with severity in COVID-19. Searches were conducted on some platforms of the National Center for Biotechnology and Information (NCBI), and 102 studies were selected for full reading according to the inclusion criteria. IFNs showed a strong association with antiviral function, specifically, IFN-λ3 (IL-28B) demonstrated genetic variants commonly related to clinical progression in various pathologies. For COVID-19, rs12979860 and rs1298275 presented frequently described unfavorable genotypes for pathological conditions of hepatitis C and hepatocellular carcinoma. The high genetic variability of HLA was reported in the studies as a crucial factor relevant to the late immune response, mainly due to its ability to recognize antigens, with the HLA-B*46:01 SNP being associated with susceptibility to COVID-19. For IL-6, rs1554606 showed a strong relationship with the clinical progression of COVID-19. In addition, rs2069837 was identified with possible host protection relationships when linked to this infection.
- Published
- 2023
- Full Text
- View/download PDF
49. IFN-λ: A New Inducer of Local Immunity against Cancer and Infections.
- Author
-
Lasfar, Ahmed, Zloza, Andrew, De La Torre, Andrew, and Cohen-Solal, Karine A.
- Subjects
THERAPEUTIC use of interferons ,CANCER immunotherapy ,VIRUS diseases ,IMMUNOLOGY - Abstract
IFN-λ is the newly established type III IFN with unique immunomodulatory functions. In contrast to the IFN-α/β family and to some extent IFN-γ, IFN-λ is apparently acting in specific areas of the body to activate resident immune cells and induces a local immunity, instrumental in preventing particular infections and also keeping transformed cells under control. Mucosal areas of lung and gastrointestinal tracts are now under scrutiny to elucidate the immune mechanisms triggered by IFN-λ and leading to viral protection. New evidence also indicates the crucial role of IFN-λ in promoting innate immunity in solid cancer models. Based on its unique biological activities among the IFN system, new immunotherapeutic approaches are now emerging for the treatment of cancer, infection, and autoimmune diseases. In the present review, we highlight the recent advances of IFN-λ immunomodulatory functions. We also discuss the perspectives of IFN-λ as a therapeutic agent. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
50. Evolution of IFN-λ in tetrapod vertebrates and its functional characterization in green anole lizard (Anolis carolinensis).
- Author
-
Chen, Shan Nan, Zhang, Xiao Wen, Li, Li, Ruan, Bai Ye, Huang, Bei, Huang, Wen Shu, Zou, Peng Fei, Fu, Jian Ping, Zhao, Li Juan, Li, Nan, and Nie, Pin
- Subjects
- *
INTERFERONS , *TETRAPODS , *GREEN anole , *PHYLOGENY , *AMNIOTES , *LOCUS (Genetics) - Abstract
IFN-λ (IFNL), i.e. type III IFN genes were found in a conserved gene locus in tetrapod vertebrates. But, a unique locus containing IFNL was found in avian. In turtle and crocodile, IFNL genes were distributed in these two separate loci. As revealed in phylogenetic trees, IFN-λs in these two different loci and other amniotes were grouped into two different clades. The conservation in gene presence and gene locus was also observed for the receptors of IFN-λ, IFN-λR1 and IL-10RB in tetrapods. It is further revealed that in North American green anole lizard Anolis carolinensis , a single IFNL gene was situated collinearly in the conserved locus as in other tetrapods, together with its receptors IFN-λR1 and IL-10RB also identified in this study. The IFN-λ and its receptors were expressed in all examined organs/tissues, and their expression was stimulated following the injection of polyI:polyC. The ISREs in promoter of IFN-λ in lizard were responsible to IRF3 as demonstrated using luciferase report system, and IFN-λ in lizard functioned through the receptors, IFN-λR1 and IL-10RB, as the up-regulation of ISGs was observed in ligand-receptor transfected, and also in recombinant IFN-λ stimulated, cell lines. Taken together, it is concluded that the mechanisms involved in type III IFN ligand-receptor system, and in its signalling pathway and its down-stream genes may be conserved in green anole lizard, and may even be so in tetrapods from xenopus to human. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.