1
|
Prince BC, Chan K, Rückert C. Elucidating the role of dsRNA sensing and Toll6 in antiviral responses of Culex quinquefasciatus cells. Front Cell Infect Microbiol 2023; 13:1251204. [PMID: 37712057 PMCID: PMC10499357 DOI: 10.3389/fcimb.2023.1251204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
The first step of any immune response is the recognition of foreign molecular structures inside the host organism. An important molecule that is generally foreign to eukaryotic cells is long double-stranded RNA (dsRNA), which can be generated during virus replication. The mechanisms of sensing viral dsRNA are well-studied in mammalian systems but are only poorly understood in insects, including disease vectors such as Culex quinquefasciatus mosquitoes. These mosquitoes are vectors for important arboviruses, such as West Nile virus, and Culex species mosquitoes are distributed across the globe in many temperate and tropical regions. The major antiviral response triggered by dsRNA in mosquitoes is RNA interference - a sequence-specific response which targets complementary viral RNA for degradation. However, here, we aimed to identify whether sequence-independent dsRNA sensing, mimicked by poly(I:C), can elicit an antiviral response. We observed a significant reduction in replication of La Crosse virus (LACV) in Cx. quinquefasciatus mosquito cells following poly(I:C) priming. We identified a number of antimicrobial peptides and Toll receptors that were upregulated at the transcript level by poly(I:C) stimulation. Notably, Toll6 was upregulated and we determined that a knockdown of Toll6 expression resulted also in increased LACV replication. Future efforts require genetic tools to validate whether the observed Toll6 antiviral activity is indeed linked to dsRNA sensing. However, large-scale functional genomic and proteomic approaches are also required to determine which downstream responses are part of the poly(I:C) elicited antiviral response.
Collapse
Affiliation(s)
| | | | - Claudia Rückert
- Department of Biochemistry and Molecular Biology, College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, NV, United States
| |
Collapse
|
2
|
Silva MJA, Silva CS, da Silva Vieira MC, dos Santos PAS, Frota CC, Lima KVB, Lima LNGC. The Relationship between TLR3 rs3775291 Polymorphism and Infectious Diseases: A Meta-Analysis of Case-Control Studies. Genes (Basel) 2023; 14:1311. [PMID: 37510216 PMCID: PMC10379146 DOI: 10.3390/genes14071311] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
As the host's first line of defense against pathogens, Toll-like receptors (TLRs), such as the TLR3, are genes encoding transmembrane receptors of the same name. Depending on their expression, TLRs cause a pro- or anti-inflammatory response. The purpose of the article was to determine whether there is an association between the Toll-like receptor 3 (TLR3) rs3775291 Single Nucleotide Polymorphism-SNP and susceptibility to infections. This review was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 guidelines and was registered in PROSPERO under the code CRD42023429533. A systematic search for relevant studies was performed using PubMed, Scopus, SciELO, Google Scholar, and Science Direct by the MeSH descriptors and the Boolean Operator "AND": "Infections"; "TLR3"; "SNP", between January 2005 and July 2022. Summary odds ratios (ORs) and corresponding 95% confidence intervals (CIs) were calculated for genotypic comparison assuming a dominant genetic model (CT + TT vs. CC). A meta-analysis of 18 studies consisting of 3118 cases and 4368 controls found a significant association for risk between the presence of the TLR3 SNP rs3775291 and infections as part of the general analysis (OR = 1.16, 95% CI = 1.04-1.28, p = 0.004). In the subgroups of continents, the SNP had a protective role in Europe for 1044 cases and 1471 controls (OR = 0.83, 95% CI = 0.70-0.99, p = 0.04); however, the Asian (for 1588 patients and 2306 controls) and American (for 486 patients and 591 controls) continents had an increase in infectious risk (OR = 1.37, 95% CI = 1.19-1.58, p < 0.001; OR = 1.42, 95% CI = 1.08-1.86, and p = 0.01, respectively). Heterogeneity between studies was detected (I2 = 58%) but was explained in meta-regression by the subgroup of continents itself and publication bias was not evident. The results of the meta-analysis suggest a significant association between the TLR3 rs3775291 polymorphism and susceptibility to infections. Thus, when analyzing subgroups, the Asian and American continents showed that this SNP confers a higher risk against infections in a dominant genotypic model. Therefore, more studies are necessary to fully elucidate the role of TLR3 rs3775291 in infections.
Collapse
Affiliation(s)
- Marcos Jessé Abrahão Silva
- Graduate Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ananindeua 67030-000, PA, Brazil;
| | - Caroliny Soares Silva
- Postgraduate Program in Parasitic Biology in the Amazon (PPGBPA), University of State of Pará (UEPA), Belém 66087-670, PA, Brazil; (C.S.S.); (M.C.d.S.V.); (P.A.S.d.S.)
| | - Marcelo Cleyton da Silva Vieira
- Postgraduate Program in Parasitic Biology in the Amazon (PPGBPA), University of State of Pará (UEPA), Belém 66087-670, PA, Brazil; (C.S.S.); (M.C.d.S.V.); (P.A.S.d.S.)
| | - Pabllo Antonny Silva dos Santos
- Postgraduate Program in Parasitic Biology in the Amazon (PPGBPA), University of State of Pará (UEPA), Belém 66087-670, PA, Brazil; (C.S.S.); (M.C.d.S.V.); (P.A.S.d.S.)
| | - Cristiane Cunha Frota
- Department of Pathology and Legal Medicine, Faculty of Medicine, Federal University of Ceará (UFC), Fortaleza 60441-750, CE, Brazil;
| | - Karla Valéria Batista Lima
- Bacteriology and Mycology Section of the Evandro Chagas Institute (IEC), Ananindeua 67030-000, PA, Brazil;
| | | |
Collapse
|
3
|
Wang J, Mai X, He Y, Zhu C, Zhou D. IgG1-Dominant Antibody Response Induced by Recombinant Trimeric SARS-CoV-2 Spike Protein with PIKA Adjuvant. Vaccines (Basel) 2023; 11:vaccines11040827. [PMID: 37112739 PMCID: PMC10144704 DOI: 10.3390/vaccines11040827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/19/2023] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Recombinant trimeric SARS-CoV-2 Spike protein with PIKA (polyI:C) adjuvant induces potent and durable neutralizing antibodies that protect against multiple SARS-CoV-2 variants. The immunoglobulin subclasses of viral-specific antibodies remain unknown, as do their glycosylation status on Fc regions. In this study, we analyzed immunoglobulins adsorbed by plate-bound recombinant trimeric SARS-CoV-2 Spike protein from serum of Cynomolgus monkey immunized by recombinant trimeric SARS-CoV-2 Spike protein with PIKA (polyI:C) adjuvant. The results showed that IgG1 was the dominant IgG subclass as revealed by ion mobility mass spectrometry. The average percentage of Spike protein-specific IgG1 increased to 88.3% as compared to pre-immunization. Core fucosylation for Fc glycopeptide of Spike protein-specific IgG1 was found to be higher than 98%. These results indicate that a unique Th1-biased, IgG1-dominant antibody response was responsible for the effectiveness of PIKA (polyI:C) adjuvant. Vaccine-induced core-fucosylation of IgG1 Fc region may reduce incidence of severe COVID-19 disease associated with overstimulation of FCGR3A by afucosylated IgG1.
Collapse
Affiliation(s)
- Jingxia Wang
- Department of Immunology and Pathogen Biology, Tongji University School of Medicine, 500 Zhennan Road, Shanghai 200331, China
| | - Xinjia Mai
- Department of Immunology and Pathogen Biology, Tongji University School of Medicine, 500 Zhennan Road, Shanghai 200331, China
| | - Yu He
- Department of Immunology and Pathogen Biology, Tongji University School of Medicine, 500 Zhennan Road, Shanghai 200331, China
| | - Chenxi Zhu
- Department of Immunology and Pathogen Biology, Tongji University School of Medicine, 500 Zhennan Road, Shanghai 200331, China
| | - Dapeng Zhou
- Department of Immunology and Pathogen Biology, Tongji University School of Medicine, 500 Zhennan Road, Shanghai 200331, China
| |
Collapse
|
4
|
Rehman MSU, Rehman SU, Yousaf W, Hassan FU, Ahmad W, Liu Q, Pan H. The Potential of Toll-Like Receptors to Modulate Avian Immune System: Exploring the Effects of Genetic Variants and Phytonutrients. Front Genet 2021; 12:671235. [PMID: 34512716 PMCID: PMC8427530 DOI: 10.3389/fgene.2021.671235] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/27/2021] [Indexed: 11/13/2022] Open
Abstract
Toll-like receptors (TLRs) are pathogen recognition receptors, and primitive sources of innate immune response that also play key roles in the defense mechanism against infectious diseases. About 10 different TLRs have been discovered in chicken that recognize ligands and participate in TLR signaling pathways. Research findings related to TLRs revealed new approaches to understand the fundamental mechanisms of the immune system, patterns of resistance against diseases, and the role of TLR-specific pathways in nutrient metabolism in chicken. In particular, the uses of specific feed ingredients encourage molecular biologists to exploit the relationship between nutrients (including different phytochemicals) and TLRs to modulate immunity in chicken. Phytonutrients and prebiotics are noteworthy dietary components to promote immunity and the production of disease-resistant chicken. Supplementations of yeast-derived products have also been extensively studied to enhance innate immunity during the last decade. Such interventions pave the way to explore nutrigenomic approaches for healthy and profitable chicken production. Additionally, single-nucleotide polymorphisms in TLRs have shown potential association with few disease outbreaks in chickens. This review aimed to provide insights into the key roles of TLRs in the immune response and discuss the potential applications of these TLRs for genomic and nutritional interventions to improve health, and resistance against different fatal diseases in chicken.
Collapse
Affiliation(s)
- Muhammad Saif-ur Rehman
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China
- Faculty of Animal Husbandry, Institute of Animal and Dairy Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Saif ur Rehman
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China
| | - Wasim Yousaf
- Faculty of Animal Husbandry, Institute of Animal and Dairy Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Faiz-ul Hassan
- Faculty of Animal Husbandry, Institute of Animal and Dairy Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Waqas Ahmad
- Department of Clinical Sciences, University College of Veterinary and Animal Sciences, Narowal, Pakistan
| | - Qingyou Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China
| | - Hongping Pan
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China
| |
Collapse
|
5
|
SUJATHA T, SUNDER J, DE AK, BHATTACHARYA D, BHOWMICK S, KUNDU A. Serum cytokine concentration in native Nicobari fowl of Andaman and Nicobar Islands. THE INDIAN JOURNAL OF ANIMAL SCIENCES 2020. [DOI: 10.56093/ijans.v90i7.106670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
6
|
Lomphithak T, Choksi S, Mutirangura A, Tohtong R, Tencomnao T, Usubuchi H, Unno M, Sasano H, Jitkaew S. Receptor-interacting protein kinase 1 is a key mediator in TLR3 ligand and Smac mimetic-induced cell death and suppresses TLR3 ligand-promoted invasion in cholangiocarcinoma. Cell Commun Signal 2020; 18:161. [PMID: 33036630 PMCID: PMC7545934 DOI: 10.1186/s12964-020-00661-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/10/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Toll-like receptor 3 (TLR3) ligand which activates TLR3 signaling induces both cancer cell death and activates anti-tumor immunity. However, TLR3 signaling can also harbor pro-tumorigenic consequences. Therefore, we examined the status of TLR3 in cholangiocarcinoma (CCA) cases to better understand TLR3 signaling and explore the potential therapeutic target in CCA. METHODS The expression of TLR3 and receptor-interacting protein kinase 1 (RIPK1) in primary CCA tissues was assayed by Immunohistochemical staining and their associations with clinicopathological characteristics and survival data were evaluated. The effects of TLR3 ligand, Poly(I:C) and Smac mimetic, an IAP antagonist on CCA cell death and invasion were determined by cell death detection methods and Transwell invasion assay, respectively. Both genetic and pharmacological inhibition of RIPK1, RIPK3 and MLKL and inhibitors targeting NF-κB and MAPK signaling were used to investigate the underlying mechanisms. RESULTS TLR3 was significantly higher expressed in tumor than adjacent normal tissues. We demonstrated in a panel of CCA cell lines that TLR3 was frequently expressed in CCA cell lines, but was not detected in a nontumor cholangiocyte. Subsequent in vitro study demonstrated that Poly(I:C) specifically induced CCA cell death, but only when cIAPs were removed by Smac mimetic. Cell death was also switched from apoptosis to necroptosis when caspases were inhibited in CCA cells-expressing RIPK3. In addition, RIPK1 was required for Poly(I:C) and Smac mimetic-induced apoptosis and necroptosis. Of particular interest, high TLR3 or low RIPK1 status in CCA patients was associated with more invasiveness. In vitro invasion demonstrated that Poly(I:C)-induced invasion through NF-κB and MAPK signaling. Furthermore, the loss of RIPK1 enhanced Poly(I:C)-induced invasion and ERK activation in vitro. Smac mimetic also reversed Poly(I:C)-induced invasion, partly mediated by RIPK1. Finally, a subgroup of patients with high TLR3 and high RIPK1 had a trend toward longer disease-free survival (p = 0.078, 28.0 months and 10.9 months). CONCLUSION RIPK1 plays a pivotal role in TLR3 ligand, Poly(I:C)-induced cell death when cIAPs activity was inhibited and loss of RIPK1 enhanced Poly(I:C)-induced invasion which was partially reversed by Smac mimetic. Our results suggested that TLR3 ligand in combination with Smac mimetic could provide therapeutic benefits to the patients with CCA. Video abstract.
Collapse
Affiliation(s)
- Thanpisit Lomphithak
- Graduate Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Swati Choksi
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892 USA
| | - Apiwat Mutirangura
- Department of Anatomy, Faculty of Medicine, Center of Excellence in Molecular Genetics of Cancer and Human Diseases, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Rutaiwan Tohtong
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, 10400 Thailand
| | - Tewin Tencomnao
- Age-Related Inflammation and Degeneration Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Hajime Usubuchi
- Department of Pathology, Tohoku University School of Medicine, Sendai, Miyagi 980-8575 Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University School of Medicine, Sendai, Miyagi 98-8075 Japan
| | - Hironobu Sasano
- Department of Pathology, Tohoku University School of Medicine, Sendai, Miyagi 980-8575 Japan
| | - Siriporn Jitkaew
- Age-Related Inflammation and Degeneration Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330 Thailand
| |
Collapse
|
7
|
Woelfl F, Léger P, Oreshkova N, Pahmeier F, Windhaber S, Koch J, Stanifer M, Roman Sosa G, Uckeley ZM, Rey FA, Boulant S, Kortekaas J, Wichgers Schreur PJ, Lozach PY. Novel Toscana Virus Reverse Genetics System Establishes NSs as an Antagonist of Type I Interferon Responses. Viruses 2020; 12:v12040400. [PMID: 32260371 PMCID: PMC7232479 DOI: 10.3390/v12040400] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/30/2020] [Accepted: 04/03/2020] [Indexed: 12/13/2022] Open
Abstract
The sand fly-borne Toscana virus (TOSV) is the major cause of human meningoencephalitis in the Mediterranean basin during the summer season. In this work, we have developed a T7 RNA polymerase-driven reverse genetics system to recover infectious particles of a lineage B strain of TOSV. The viral protein pattern and growth properties of the rescued virus (rTOSV) were found to be similar to those of the corresponding wild-type (wt) virus. Using this system, we genetically engineered a TOSV mutant lacking expression of the non-structural protein NSs (rTOSVɸNSs). Unlike rTOSV and the wt virus, rTOSVɸNSs was unable to (i) suppress interferon (IFN)-b messenger RNA induction; and (ii) grow efficiently in cells producing IFN-b. Together, our results highlight the importance of NSs for TOSV in evading the IFN response and provide a comprehensive toolbox to investigate the TOSV life cycle in mammalian and insect host cells, including several novel polyclonal antibodies.
Collapse
Affiliation(s)
- Franziska Woelfl
- CellNetworks Cluster of Excellence, University Hospital Heidelberg, 69120 Heidelberg, Germany; (F.W.); (P.L.); (F.P.); (S.W.); (J.K.); (Z.M.U.)
- Center for Integrative Infectious Diseases Research (CIID), Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Psylvia Léger
- CellNetworks Cluster of Excellence, University Hospital Heidelberg, 69120 Heidelberg, Germany; (F.W.); (P.L.); (F.P.); (S.W.); (J.K.); (Z.M.U.)
- Center for Integrative Infectious Diseases Research (CIID), Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Nadia Oreshkova
- Wageningen Bioveterinary Research, Department of Virology, 8221 RA Lelystad, The Netherlands; (N.O.); (J.K.)
| | - Felix Pahmeier
- CellNetworks Cluster of Excellence, University Hospital Heidelberg, 69120 Heidelberg, Germany; (F.W.); (P.L.); (F.P.); (S.W.); (J.K.); (Z.M.U.)
- Center for Integrative Infectious Diseases Research (CIID), Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Stefan Windhaber
- CellNetworks Cluster of Excellence, University Hospital Heidelberg, 69120 Heidelberg, Germany; (F.W.); (P.L.); (F.P.); (S.W.); (J.K.); (Z.M.U.)
- Center for Integrative Infectious Diseases Research (CIID), Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Jana Koch
- CellNetworks Cluster of Excellence, University Hospital Heidelberg, 69120 Heidelberg, Germany; (F.W.); (P.L.); (F.P.); (S.W.); (J.K.); (Z.M.U.)
- Center for Integrative Infectious Diseases Research (CIID), Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Megan Stanifer
- Center for Integrative Infectious Diseases Research (CIID), Molecular Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Gleyder Roman Sosa
- Structural Virology Unit, Pasteur Institute, 75015 Paris, France; (G.R.S.); (F.A.R.)
| | - Zina M. Uckeley
- CellNetworks Cluster of Excellence, University Hospital Heidelberg, 69120 Heidelberg, Germany; (F.W.); (P.L.); (F.P.); (S.W.); (J.K.); (Z.M.U.)
- Center for Integrative Infectious Diseases Research (CIID), Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Felix A. Rey
- Structural Virology Unit, Pasteur Institute, 75015 Paris, France; (G.R.S.); (F.A.R.)
| | - Steeve Boulant
- Center for Integrative Infectious Diseases Research (CIID), Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Jeroen Kortekaas
- Wageningen Bioveterinary Research, Department of Virology, 8221 RA Lelystad, The Netherlands; (N.O.); (J.K.)
- Laboratory of Virology, Wageningen University, 6708 PB Wageningen, The Netherlands
| | - Paul J. Wichgers Schreur
- Wageningen Bioveterinary Research, Department of Virology, 8221 RA Lelystad, The Netherlands; (N.O.); (J.K.)
- Correspondence: (P.J.W.S.); (P.-Y.L.)
| | - Pierre-Yves Lozach
- CellNetworks Cluster of Excellence, University Hospital Heidelberg, 69120 Heidelberg, Germany; (F.W.); (P.L.); (F.P.); (S.W.); (J.K.); (Z.M.U.)
- Center for Integrative Infectious Diseases Research (CIID), Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
- INRAE, EPHE, Viral Infections and Comparative Pathology (IVPC), University Claude Bernard Lyon1, University of Lyon, UMR754, 69007 Lyon, France
- Correspondence: (P.J.W.S.); (P.-Y.L.)
| |
Collapse
|
8
|
Duncan RS, Rohowetz L, Vogt A, Koulen P. Repeat exposure to polyinosinic:polycytidylic acid induces TLR3 expression via JAK-STAT signaling and synergistically potentiates NFκB-RelA signaling in ARPE-19 cells. Cell Signal 2019; 66:109494. [PMID: 31809875 DOI: 10.1016/j.cellsig.2019.109494] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/11/2019] [Accepted: 12/02/2019] [Indexed: 01/05/2023]
Abstract
Dry age-related macular degeneration (AMD), accounting for approximately 90% of AMD cases, is characterized by photoreceptor death, retinal pigment epithelium (RPE) dysfunction and, ultimately, geographic atrophy - the localized death of RPE leading to loss of the center of the visual field. The pathological etiology of AMD is multifactorial, but innate immune signaling and inflammation are involved in early stages of the disease. Although numerous single-nucleotide polymorphisms in innate immune genes are associated with dry AMD, no single gene appears to cause dry AMD. Here, we hypothesized that activation of TLR3 potentiates expression of TLR3 itself and the NFκB-p65 (RelA) subunit as part of pro-inflammatory RPE signaling. Furthermore, we hypothesized that TLR3 activation can 'prime' cells to future RelA stimulation, leading to enhanced, persistent RelA expression and signaling following a second TLR3 activation. We used the human RPE-derived cell line ARPE-19 as a model system for RPE signaling and measured NFκB expression and activity in response to TLR3 stimulation with its ligand, polyinosinic:polycytidylic acid (pI:C). Activation of TLR3 with pI:C led to increased TLR3 and RelA expression that was sustained for at least 24 h. Cells exposed for a second time to pI:C after an initial pI:C exposure displayed elevated RelA expression and RelA nuclear translocation above the level generated by individual primary or secondary exposures alone. Such an elevated response could also not be generated by a single application of higher concentrations of the agonist pI:C. Additionally, we determined the mechanism for TLR3 mediated TLR3 and RelA expression by using inhibitors of canonical TLR3-TBK1-IKKε and JAK-STAT signaling pathways. These data suggest that initial exposure of ARPE-19 cells to pI:C upregulates TLR3 and RelA signaling, leading to potentiated and persistent RelA signaling potentially generated by a positive feedback loop that may cause exacerbated inflammation in AMD. Furthermore, inhibition of JAK-STAT signaling may be a possible therapeutic treatment to prevent induction of TLR3 expression subsequent to pI:C exposure. Our results identify possible therapeutic targets to reduce the TLR3 positive feedback loop and subsequent overproduction of pro-inflammatory cytokines in RPE cells.
Collapse
Affiliation(s)
- R Scott Duncan
- Vision Research Center, Department of Ophthalmology, School of Medicine, University of Missouri - Kansas City, 2411 Holmes St., Kansas City, MO 64108, United States of America.
| | - Landon Rohowetz
- Vision Research Center, Department of Ophthalmology, School of Medicine, University of Missouri - Kansas City, 2411 Holmes St., Kansas City, MO 64108, United States of America
| | - Alex Vogt
- Vision Research Center, Department of Ophthalmology, School of Medicine, University of Missouri - Kansas City, 2411 Holmes St., Kansas City, MO 64108, United States of America
| | - Peter Koulen
- Vision Research Center, Department of Ophthalmology, School of Medicine, University of Missouri - Kansas City, 2411 Holmes St., Kansas City, MO 64108, United States of America; Department of Biomedical Sciences, School of Medicine, University of Missouri - Kansas City, 2411 Holmes St., Kansas City, MO 64108, United States of America
| |
Collapse
|
9
|
Ahmed-Hassan H, Abdul-Cader MS, Ahmed Sabry M, Hamza E, Sharif S, Nagy E, Abdul-Careem MF. Double-Stranded Ribonucleic Acid-Mediated Antiviral Response Against Low Pathogenic Avian Influenza Virus Infection. Viral Immunol 2018; 31:433-446. [PMID: 29813000 DOI: 10.1089/vim.2017.0142] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Toll-like receptor (TLR)3 signaling pathway is known to induce type 1 interferons (IFNs) and proinflammatory mediators leading to antiviral response against many viral infections. Double-stranded ribonucleic acid (dsRNA) has been shown to act as a ligand for TLR3 and, as such, has been a focus as a potential antiviral agent in many host-viral infection models. Yet, its effectiveness and involved mechanisms as a mediator against low pathogenic avian influenza virus (LPAIV) have not been investigated adequately. In this study, we used avian fibroblasts to verify whether dsRNA induces antiviral response against H4N6 LPAIV and clarify whether type 1 IFNs and proinflammatory mediators such as interleukin (IL)-1β are contributing to the dsRNA-mediated antiviral response against H4N6 LPAIV. We found that dsRNA induces antiviral response in avian fibroblasts against H4N6 LPAIV infection. The treatment of avian fibroblasts with dsRNA increases the expressions of TLR3, IFN-α, IFN-β, and IL-1β. We also confirmed that this antiviral response elicited against H4N6 LPAIV infection correlates, but is not attributable to type 1 IFNs or IL-1β. Our findings imply that the TLR3 ligand, dsRNA, can elicit antiviral response in avian fibroblasts against LPAIV infection, highlighting potential value of dsRNA as an antiviral agent against LPAIV infections. However, further investigations are required to determine the potential role of other innate immune mediators or combination of the tested cytokines in the dsRNA-mediated antiviral response against H4N6 LPAIV infection.
Collapse
Affiliation(s)
- Hanaa Ahmed-Hassan
- 1 Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, Health Research Innovation Center 2C53, University of Calgary , Calgary, Alberta, Canada .,2 Zoonoses Department, Faculty of Veterinary Medicine, Cairo University , Giza, Egypt
| | - Mohamed Sarjoon Abdul-Cader
- 1 Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, Health Research Innovation Center 2C53, University of Calgary , Calgary, Alberta, Canada
| | - Maha Ahmed Sabry
- 2 Zoonoses Department, Faculty of Veterinary Medicine, Cairo University , Giza, Egypt
| | - Eman Hamza
- 2 Zoonoses Department, Faculty of Veterinary Medicine, Cairo University , Giza, Egypt
| | - Shayan Sharif
- 3 Department of Pathobiology, University of Guelph , Guelph, Ontario, Canada
| | - Eva Nagy
- 3 Department of Pathobiology, University of Guelph , Guelph, Ontario, Canada
| | - Mohamed Faizal Abdul-Careem
- 1 Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, Health Research Innovation Center 2C53, University of Calgary , Calgary, Alberta, Canada
| |
Collapse
|
10
|
Duffney PF, McCarthy CE, Nogales A, Thatcher TH, Martinez-Sobrido L, Phipps RP, Sime PJ. Cigarette smoke dampens antiviral signaling in small airway epithelial cells by disrupting TLR3 cleavage. Am J Physiol Lung Cell Mol Physiol 2018; 314:L505-L513. [PMID: 29351447 PMCID: PMC5900359 DOI: 10.1152/ajplung.00406.2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 12/08/2017] [Accepted: 12/08/2017] [Indexed: 12/20/2022] Open
Abstract
Cigarette smokers and people exposed to second-hand smoke are at an increased risk for pulmonary viral infections, and yet the mechanism responsible for this heightened susceptibility is not understood. To understand the effect of cigarette smoke on susceptibility to viral infection, we used an air-liquid interface culture system and exposed primary human small airway epithelial cells (SAEC) to whole cigarette smoke, followed by treatment with the viral mimetic polyinosinic polycytidylic acid (poly I:C) or influenza A virus (IAV). We found that prior smoke exposure strongly inhibited production of proinflammatory (interleukin-6 and interleukin-8) and antiviral [interferon-γ-induced protein 10 (IP-10) and interferons] mediators in SAECs in response to poly I:C and IAV infection. Impaired antiviral responses corresponded to increased infection with IAV. This was associated with a decrease in phosphorylation of the key antiviral transcription factor interferon response factor 3 (IRF3). Here, we found that cigarette smoke exposure inhibited activation of Toll-like receptor 3 (TLR3) by impairing TLR3 cleavage, which was required for downstream phosphorylation of IRF3 and production of IP-10. These results identify a novel mechanism by which cigarette smoke exposure impairs antiviral responses in lung epithelial cells, which may contribute to increased susceptibility to respiratory infections.
Collapse
Affiliation(s)
- Parker F Duffney
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry , Rochester, New York
| | - Claire E McCarthy
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry , Rochester, New York
| | - Aitor Nogales
- Department of Microbiology and Immunology, University of Rochester , Rochester, New York
| | - Thomas H Thatcher
- Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry , Rochester, New York
- Division of Pulmonary and Critical Care Medicine, University of Rochester School of Medicine and Dentistry , Rochester, New York
| | - Luis Martinez-Sobrido
- Department of Microbiology and Immunology, University of Rochester , Rochester, New York
| | - Richard P Phipps
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry , Rochester, New York
- Department of Microbiology and Immunology, University of Rochester , Rochester, New York
- Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry , Rochester, New York
- Division of Pulmonary and Critical Care Medicine, University of Rochester School of Medicine and Dentistry , Rochester, New York
| | - Patricia J Sime
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry , Rochester, New York
- Department of Microbiology and Immunology, University of Rochester , Rochester, New York
- Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry , Rochester, New York
- Division of Pulmonary and Critical Care Medicine, University of Rochester School of Medicine and Dentistry , Rochester, New York
| |
Collapse
|
11
|
Ganesan P, Chandwani MN, Creisher PS, Bohn L, O'Donnell LA. The neonatal anti-viral response fails to control measles virus spread in neurons despite interferon-gamma expression and a Th1-like cytokine profile. J Neuroimmunol 2017; 316:80-97. [PMID: 29366594 PMCID: PMC6003673 DOI: 10.1016/j.jneuroim.2017.12.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 12/16/2017] [Accepted: 12/19/2017] [Indexed: 01/01/2023]
Abstract
Neonates are highly susceptible to viral infections in the periphery, potentially due to deviant cytokine responses. Here, we investigated the role of interferon-gamma (IFNγ), a key anti-viral in the neonatal brain. We found that (i) IFNγ, which is critical for viral control and survival in adults, delays mortality in neonates, (ii) IFNγ limits infiltration of macrophages, neutrophils, and T cells in the neonatal brain, (iii) neonates and adults differentially express pathogen recognition receptors and Type I interferons in response to the infection, (iv) both neonates and adults express IFNγ and other Th1-related factors, but expression of many cytokines/chemokines and IFNγ-responsive genes is age-dependent, and (v) administration of IFNγ extends survival and reduces CD4 T cell infiltration in the neonatal brain. Our findings suggest age-dependent expression of cytokine/chemokine profiles in the brain and distinct dynamic interplays between lymphocyte populations and cytokines/chemokines in MV-infected neonates. The role of the anti-viral cytokine interferon-gamma (IFNγ) is investigated during a neonatal viral infection in CNS neurons. IFNγ did not prevent mortality in neonates, but it slowed disease progression. IFNγ reduced infiltration of neutrophils, macrophages, and T cells in the neonatal CNS. Both adult and neonatal mice expressed Th1-like cytokines, including IFNγ and some IFNγ-stimulated genes, during infection. Despite a Th1-like cytokine profile in the neonatal CNS, the cytokine milieu is ineffective at controlling viral spread.
Collapse
Affiliation(s)
- Priya Ganesan
- Duquesne University, School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Pittsburgh, PA 15282, United States
| | - Manisha N Chandwani
- Duquesne University, School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Pittsburgh, PA 15282, United States
| | - Patrick S Creisher
- Duquesne University, School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Pittsburgh, PA 15282, United States
| | - Larissa Bohn
- Duquesne University, School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Pittsburgh, PA 15282, United States
| | - Lauren A O'Donnell
- Duquesne University, School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Pittsburgh, PA 15282, United States.
| |
Collapse
|
12
|
Liu Q, Xu D, Jiang S, Huang J, Zhou F, Yang Q, Jiang S, Yang L. Toll-receptor 9 gene in the black tiger shrimp (Penaeus monodon) induced the activation of the TLR-NF-κB signaling pathway. Gene 2017; 639:27-33. [PMID: 28982619 DOI: 10.1016/j.gene.2017.09.060] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 09/22/2017] [Accepted: 09/26/2017] [Indexed: 12/11/2022]
Abstract
Toll receptors are important pathogen recognition receptors (PRRs) in shrimps, which play a vital role in defending against virus and bacterial challenge. In this paper, the characterization and functional analysis of a Toll9 receptor gene from Penaeus monodon was performed in HEK293T cells. Data showed that PmToll9 can activate the NF-κB promoter activities of TLR pathway, while ISRE and IFN-β promoter cannot be activated obviously in HEK293T cells using dual-luciferase reporter system. The downstream immune factors of IL-8, IκB-α, and TRAF6 were activated by PmToll9 and IL-8 showed the most significant up-regulation in expression levels, indicating the activities of NF-κB can be mediated by PmToll9. Six LRRs-deletion mutants were constructed and results showed these mutants had obvious declines in luciferase activities, among which the mutant pCMV-DeLRR4 showed the most significant decline. qPCR data indicated LRRs-deletion mutants efficiently impaired the activities of the downstream immune factors IL-8, IκB-α, and TRAF6. It demonstrates that LRRs-deletion mutants could result in the weaken abilities of PmToll9 in signaling transduction. Overexpression of PmToll9-GFP fusion protein in Hela cells revealed the primary cellular localization of PmToll9 is in the cytoplasm.
Collapse
Affiliation(s)
- Qian Liu
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China; College of Aqua-life Science and Technology, Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, Guangzhou 510300, China
| | - Dan Xu
- College of Aqua-life Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Shigui Jiang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China; Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, Guangzhou 510300, China
| | - Jianhua Huang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China
| | - Falin Zhou
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China
| | - Qibin Yang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China
| | - Song Jiang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China
| | - Lishi Yang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China.
| |
Collapse
|
13
|
Bianchi F, Pretto S, Tagliabue E, Balsari A, Sfondrini L. Exploiting poly(I:C) to induce cancer cell apoptosis. Cancer Biol Ther 2017; 18:747-756. [PMID: 28881163 PMCID: PMC5678690 DOI: 10.1080/15384047.2017.1373220] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
TLR3 belong to the Toll-like receptors family, it is mainly expressed on immune cells where it senses pathogen-associated molecular patterns and initiates innate immune response. TLR3 agonist poly(I:C) was developed to mimic pathogens infection and boost immune system activation to promote anti-cancer therapy. Accordingly, TLR agonists were included in the National Cancer Institute list of immunotherapeutic agents with the highest potential to cure cancer. Besides well known effects on immune cells, poly(I:C) was also shown, in experimental models, to directly induce apoptosis in cancer cells expressing TLR3. This review presents the current knowledge on the mechanism of poly(I:C)-induced apoptosis in cancer cells. Experimental evidences on positive or negative regulators of TLR3-mediated apoptosis induced by poly(I:C) are reported and strategies are proposed to successfully promote this event in cancer cells. Cancer cells apoptosis is an additional arm offered by poly(I:C), besides activation of immune system, for the treatment of various type of cancer. A further dissection of TLR3 signaling would contribute to greater resolution of the critical steps that impede full exploitation of the poly(I:C)-induced apoptosis. Experimental evidences about negative regulator of poly(I:C)-induced apoptotic program should be considered in combinations with TLR3 agonists in clinical trials.
Collapse
Affiliation(s)
- Francesca Bianchi
- a Fondazione IRCCS Istituto Nazionale dei Tumori , Department of Research, Epidemiologia e Medicina Molecolare , via Amadeo 42, Milan , Italy.,b Università degli Studi di Milano , Dipartimento di Scienze Biomediche per la Salute , via Mangiagalli 31, Milan , Italy
| | - Samantha Pretto
- b Università degli Studi di Milano , Dipartimento di Scienze Biomediche per la Salute , via Mangiagalli 31, Milan , Italy
| | - Elda Tagliabue
- a Fondazione IRCCS Istituto Nazionale dei Tumori , Department of Research, Epidemiologia e Medicina Molecolare , via Amadeo 42, Milan , Italy
| | - Andrea Balsari
- a Fondazione IRCCS Istituto Nazionale dei Tumori , Department of Research, Epidemiologia e Medicina Molecolare , via Amadeo 42, Milan , Italy.,b Università degli Studi di Milano , Dipartimento di Scienze Biomediche per la Salute , via Mangiagalli 31, Milan , Italy
| | - Lucia Sfondrini
- b Università degli Studi di Milano , Dipartimento di Scienze Biomediche per la Salute , via Mangiagalli 31, Milan , Italy
| |
Collapse
|
14
|
Ye L, Jiang Y, Yang G, Yang W, Hu J, Cui Y, Shi C, Liu J, Wang C. Murine bone marrow-derived DCs activated by porcine rotavirus stimulate the Th1 subtype response in vitro. Microb Pathog 2017; 110:325-334. [PMID: 28710013 DOI: 10.1016/j.micpath.2017.07.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/18/2016] [Accepted: 07/10/2017] [Indexed: 11/22/2022]
Abstract
Rotavirus (RV) infection causes acute, watery dehydrating diarrhea and even death in infants and other young animals, resulting in a severe economic burden; however, little is known about the innate immune mechanisms associated with RV infection. Dendritic cells (DCs), which are professional antigen-presenting cells (APCs), serve as a bridge connecting the innate and adaptive immune system. In this study, the interaction between murine bone marrow-derived DCs (BMDCs) and porcine rotavirus (PRV) was investigated in vitro. Upon stimulation, the expression levels of MHC-II, CD40, CD80, CD86 and CD83 in BMDCs increased in a time-dependent manner, indicating activation and maturation by PRV. In addition, up-regulated Toll-like receptor 2 (TLR2), TLR3 and NF-κB increased the production of interleukin-12 and interferon-γ. The PRV-stimulated BMDCs also showed increased stimulatory capacity in mixed lymphocyte reactions and promoted the Th1 subtype response.
Collapse
Affiliation(s)
- Liping Ye
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Yanlong Jiang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Guilian Yang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Wentao Yang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Jingtao Hu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Yulin Cui
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Chunwei Shi
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Jing Liu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Chunfeng Wang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
15
|
Mumps Virus SH Protein Inhibits NF-κB Activation by Interacting with Tumor Necrosis Factor Receptor 1, Interleukin-1 Receptor 1, and Toll-Like Receptor 3 Complexes. J Virol 2017; 91:JVI.01037-17. [PMID: 28659487 DOI: 10.1128/jvi.01037-17] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 06/22/2017] [Indexed: 11/20/2022] Open
Abstract
The mumps virus (MuV) small hydrophobic protein (SH) is a type I membrane protein expressed in infected cells. SH has been reported to interfere with innate immunity by inhibiting tumor necrosis factor alpha (TNF-α)-mediated apoptosis and NF-κB activation. To elucidate the underlying mechanism, we generated recombinant MuVs (rMuVs) expressing the SH protein with an N-terminal FLAG epitope or lacking SH expression due to the insertion of three stop codons into the SH gene. Using these viruses, we were able to show that SH reduces the phosphorylation of IKKβ, IκBα, and p65 as well as the translocation of p65 into the nucleus of infected A549 cells. Reporter gene assays revealed that SH interferes not only with TNF-α-mediated NF-κB activation but also with IL-1β- and poly(I·C)-mediated NF-κB activation, and that this inhibition occurs upstream of the NF-κB pathway components TRAF2, TRAF6, and TAK1. Since SH coimmunoprecipitated with tumor necrosis factor receptor 1 (TNFR1), RIP1, and IRAK1, we hypothesize that SH exerts its inhibitory function by interacting with TNFR1, interleukin-1 receptor type 1 (IL-1R1), and TLR3 complexes in the plasma membrane of infected cells.IMPORTANCE The MuV SH has been shown to impede TNF-α-mediated NF-κB activation and is therefore thought to contribute to viral immune evasion. However, the mechanisms by which SH mediates NF-κB inhibition remained largely unknown. In this study, we show that SH interacts with TNFR1, IL-1R1, and TLR3 complexes in infected cells. We thereby not only shed light on the mechanisms of SH-mediated NF-κB inhibition but also reveal that SH interferes with NF-κB activation induced by interleukin-1β (IL-1β) and double-stranded RNA.
Collapse
|
16
|
Involvement of the Toll-Like Receptor/Nitric Oxide Signaling Pathway in the Pathogenesis of Cervical Cancer Caused by High-Risk Human Papillomavirus Infection. BIOMED RESEARCH INTERNATIONAL 2017. [PMID: 28626766 PMCID: PMC5463171 DOI: 10.1155/2017/7830262] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Human papillomavirus (HPV) can activate Toll-like receptor (TLR)/nitric oxide (NO) signaling pathways; however, whether the TLR/NO pathway is involved in cervical cancer caused by high-risk HPV (HR-HPV) remains unclear. In this study, 43 HR-HPV-positive patients with cervical cancer (CC group), 39 HR-HPV-positive patients with a healthy cervix (HR-HPV group), and 33 HR-HPV-negative controls were recruited. NO concentration in cervical canal and expression of inducible NO synthase (iNOS) in cervical tissues were detected. Expressions of key TLR/NO pathway genes (TLR3/4/7/8, NF-κB p65, and iNOS) in cervical epithelial cells were detected by quantitative reverse transcription PCR. Expressions of TLR4, NF-κB p65, and iNOS in CaSki, HeLa, and C33a cells were determined by Western blot. NO concentration in cervical canal of CC group was significantly higher than in other groups (P < 0.05). Positive rates of iNOS in cervical tissues were 72.1%, 28.2%, and 3.1% in the CC group, HR-HPV group, and controls, respectively (P < 0.05). Levels of TLR3, TLR4, TLR7, TLR8, NF-κB p65, and iNOS in cervical epithelial cells were higher in CC group than in other groups (P < 0.05). Both mRNA and protein levels of TLR4, NF-κB p65, and iNOS were higher in HPV-positive HeLa and CaSki cells than in HPV-negative C33a cells (P < 0.05). Together, these results suggest that TLR/NO signaling pathway may be involved in pathogenesis of cervical cancer caused by HR-HPV.
Collapse
|
17
|
Zayed RA, Omran D, Mokhtar DA, Zakaria Z, Ezzat S, Soliman MA, Mobarak L, El-Sweesy H, Emam G. Association of Toll-Like Receptor 3 and Toll-Like Receptor 9 Single Nucleotide Polymorphisms with Hepatitis C Virus Infection and Hepatic Fibrosis in Egyptian Patients. Am J Trop Med Hyg 2017; 96:720-726. [PMID: 28093541 DOI: 10.4269/ajtmh.16-0644] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Toll-like receptors (TLRs) are recognized as fundamental contributors to the immune system function against infections. Hepatitis C virus (HCV) infection represents a global health problem especially in Egypt having the highest HCV prevalence worldwide where HCV infection is a continuing epidemic. The aim of the present study was to investigate the possible association between genetic variation in TLR-3 and TLR-9 and HCV infection and hepatic fibrosis in chronic HCV-positive Egyptian patients. The present study included 100 naïve chronic HCV-positive patients and 100 age- and sex-matched healthy controls. Genotyping of TLR-3 (_7 C/A [rs3775296]), TLR-3 (c.1377C/T [rs3775290]) and TLR-9 (1237T/C [rs5743836]) were done by polymerase chain reaction restriction fragment length polymorphism technique. Frequency of polymorphic genotypes in TLR-3 (_7 C/A), TLR-3 (c.1377C/T) and TLR-9 (1237T/C) were not significantly different between studied HCV-positive patients and controls with P values 0.121, 0.112, and 0.683, respectively. TLR-3 c.1377 T-allele was associated with advanced stage of hepatic fibrosis (P = 0.003).
Collapse
Affiliation(s)
- Rania A Zayed
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Dalia Omran
- Department of Endemic Medicine and Hepato-gastroentrology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Doha A Mokhtar
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Zinab Zakaria
- Department of Endemic Medicine and Hepato-gastroentrology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Sameera Ezzat
- Community Medicine Department, National Liver Institute, Menofia University, Menofia, Egypt
| | - Mohamed A Soliman
- Specialized Liver Unit, Kasr Alainy Hospital, Cairo University, Cairo, Egypt
| | - Lamiaa Mobarak
- National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt
| | - Hossam El-Sweesy
- Tropical Medicine Department, Cairo Fatemic Hospital, Ministry of Health, Cairo, Egypt
| | - Ghada Emam
- Clinical Pathology Department, National Institute of Neuromotor System, Cairo, Egypt
| |
Collapse
|
18
|
Sunder J, Sujatha T, Kundu A. Effect of Morinda citrifolia in growth, production and immunomodulatory properties in livestock and poultry: a review. ACTA ACUST UNITED AC 2016. [DOI: 10.18006/2016.4(3s).249.265] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
19
|
Abdelwahab SF. Cellular immune response to hepatitis-C-virus in subjects without viremia or seroconversion: is it important? Infect Agent Cancer 2016; 11:23. [PMID: 27186234 PMCID: PMC4867533 DOI: 10.1186/s13027-016-0070-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 03/30/2016] [Indexed: 02/08/2023] Open
Abstract
Hepatitis C Virus (HCV) causes chronic infection and represents a global health burden. To date, there is no licensed vaccine for HCV. The high viral replication rate and the existence of several HCV genotypes and quasispecies hamper the development of an effective universal vaccine. In this regard, the current HCV vaccine candidates show genotype-specific protection or narrow cross reactivity against other genotypes. Importantly, HCV spontaneous clearance occurs in 15-50 % of infected subjects, indicating that natural resistance to chronic infection exists. This phenomenon was demonstrated among humans and chimpanzees and continues to motivate researchers attempting to develop an effective HCV vaccine. However, what constitutes a protective immune response or correlate of protection against HCV infection is still vague. Additionally, the mechanisms behind successful HCV clearance suggest the coordination of several arms of the immune system, with cell-mediated immunity (CMI) playing a crucial role in this process. By contrast, although neutralizing antibodies have been identified, they are isolate-specific and poorly correlate with viral clearance. Antigen-specific CD4 T cells, instead, correlate with transient decline in HCV viremia and long-lasting control of the infection. Unfortunately, HCV has been very successful in evading host immune mechanisms, leading to complications such as liver fibrosis, cirrhosis and hepatocellular carcinoma. Interestingly, CMI to HCV antigens were shown among exposed individuals without viremia or seroconversion, suggesting the clearance of prior HCV infection(s). These individuals include family members living with HCV-infected subjects, healthcare workers, IV drug users, and sexual contacts. The correlates of protection could be closely monitored among these individuals. This review provides a summary of HCV-specific immune responses in general and of CMI in particular in these cohorts. The importance of these CMI responses are discussed.
Collapse
Affiliation(s)
- Sayed F. Abdelwahab
- />Departement of Microbiology and Immunology, Faculty of Medicine, Minia University, Minia, 61511 Egypt
- />Department of Microbiology, College of Pharmacy, Taif University, Taif, 21974 Kingdom of Saudi Arabia
| |
Collapse
|
20
|
Toll-Like Receptor 3/TRIF-Dependent IL-12p70 Secretion Mediated by Streptococcus pneumoniae RNA and Its Priming by Influenza A Virus Coinfection in Human Dendritic Cells. mBio 2016; 7:e00168-16. [PMID: 26956584 PMCID: PMC4810485 DOI: 10.1128/mbio.00168-16] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
A functional immune response is crucial to prevent and limit infections with Streptococcus pneumoniae. Dendritic cells (DCs) play a central role in orchestrating the adaptive and innate immune responses by communicating with other cell types via antigen presentation and secretion of cytokines. In this study, we set out to understand how pneumococci activate human monocyte-derived DCs to produce interleukin-12 (IL-12) p70, an important cytokine during pneumococcal infections. We show that IL-12p70 production requires uptake of bacteria as well as the presence of the adaptor molecule TRIF, which is known to transfer signals of Toll-like receptor 3 (TLR3) or TLR4 from the endosome into the cell. While TLR4 is redundant for IL-12p70 production in DCs, we found that TLR3 is required to induce full IL-12p70 secretion. Influenza A virus (IAV) infection of DCs did not induce IL-12p70 but markedly upregulated TLR3 expression that during coinfection with S. pneumoniae significantly enhanced IL-12p70 secretion. Finally, we show that pneumococcal RNA can act as a bacterial stimulus for TLR3 and that it is a key signal to induce IL-12p70 production during challenge of DCs with pneumococci. Streptococcus pneumoniae, a common colonizer of the nose, is the causative agent of severe and deadly diseases. A well-orchestrated immune response is vital to prevent and limit these diseases. Dendritic cells (DCs) reside in the mucosal linings of the lungs and sample antigens. They are activated by pathogens to present antigens and secrete cytokines. While many studies focus on murine models, we focused our work on human monocyte-derived DCs. We found that pneumococcal RNA is an important stimulus in DCs to activate the endosomal receptor TLR3, a receptor previously not identified to sense pneumococci, and its adaptor molecule TRIF. This leads to secretion of the cytokine interleukin-12 (IL-12). Severe pneumococcal pneumonia occurs closely after influenza A virus (IAV) infection. We show that IAV infection upregulates TLR3 in DCs, which sensitizes the cells to endosomal pneumococcal RNA. This new insight contributes to unlock the interplay between pneumococci, IAV, and humans.
Collapse
|
21
|
Activation of TLR2 and TLR6 by Dengue NS1 Protein and Its Implications in the Immunopathogenesis of Dengue Virus Infection. PLoS Pathog 2015; 11:e1005053. [PMID: 26226614 PMCID: PMC4520596 DOI: 10.1371/journal.ppat.1005053] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 06/27/2015] [Indexed: 01/04/2023] Open
Abstract
Dengue virus (DV) infection is the most prevalent mosquito-borne viral disease and its manifestation has been shown to be contributed in part by the host immune responses. In this study, pathogen recognition receptors, Toll-like receptor (TLR) 2 and TLR6 were found to be up-regulated in DV-infected human PBMC using immunofluorescence staining, flow cytometry and Western blot analyses. Using ELISA, IL-6 and TNF-α, cytokines downstream of TLR2 and TLR6 signaling pathways were also found to be up-regulated in DV-infected PBMC. IL-6 and TNF-α production by PBMC were reduced when TLR2 and TLR6 were blocked using TLR2 and TLR6 neutralizing antibodies during DV infection. These results suggested that signaling pathways of TLR2 and TLR6 were activated during DV infection and its activation contributed to IL-6 and TNF-α production. DV NS1 protein was found to significantly increase the production of IL-6 and TNF-α when added to PBMC. The amount of IL-6 and TNF-α stimulated by DV NS1 protein was reduced when TLR2 and TLR6 were blocked, suggesting that DV NS1 protein is the viral protein responsible for the activation of TLR2 and TLR6 during DV infection. Secreted alkaline phosphatase (SEAP) reporter assay was used to further confirm activation of TLR2 and TLR6 by DV NS1 protein. In addition, DV-infected and DV NS1 protein-treated TLR6-/- mice have higher survivability compared to DV-infected and DV NS1 protein-treated wild-type mice. Hence, activation of TLR6 via DV NS1 protein could potentially play an important role in the immunopathogenesis of DV infection. Despite the prevalence of dengue virus infection and the heavy economic burden it puts on the endemic countries, the immunopathogenesis of dengue virus infection remains unclear. Plasma leakage in dengue hemorrhagic fever (DHF) develops not when the viremia is at its peak in infected patients but when viremia has been significantly reduced or cleared. This suggests that host immune response is responsible for the development DHF. The interactions of the viral factors with host factors which trigger the host immune responses are likely to play a significant role in the development of dengue diseases, thus are of great interests. In this study, we found that dengue NS1 protein activates TLR2 and TLR6, leading to increase proinflammatory cytokine production. In addition, the interaction of viral factor with TLR6 was found to play an important role in the manifestation of dengue virus infection. Our study provides new insights into the involvement of TLR6 in dengue virus infection and the potential of using TLR6 anatagonist in therapeutic treatment for DV infection.
Collapse
|
22
|
Blok BA, Arts RJW, van Crevel R, Benn CS, Netea MG. Trained innate immunity as underlying mechanism for the long-term, nonspecific effects of vaccines. J Leukoc Biol 2015; 98:347-56. [DOI: 10.1189/jlb.5ri0315-096r] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/19/2015] [Indexed: 12/31/2022] Open
|
23
|
Hu GB, Li XP, Liu DH, Liu QM, Zhang SC. A toll-like receptor 3 homologue that is up-regulated by poly I:C and DNA virus in turbot Scophthalmus maximus. JOURNAL OF FISH BIOLOGY 2015; 86:431-447. [PMID: 25631305 DOI: 10.1111/jfb.12559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 09/21/2014] [Indexed: 06/04/2023]
Abstract
In this study, the gene and promoter sequences of turbot Scophthalmus maximus (Sm) toll-like receptor 3 (Tlr3) were cloned and its mRNA tissue distribution and gene expression in response to polyinosinic:polycytidylic acid (poly I:C) and turbot reddish body iridovirus (TRBIV) challenges were studied in vivo. The smtlr3 gene spans over 4·4 kb with a structure of five exons-four introns and encodes a peptide of 916 amino acids. The putative protein shares the highest sequence identity of 52·8-78·5% with fish Tlr3 and contains a signal peptide sequence, 13 leucine-rich repeat (LRR) motifs, a transmembrane region and a toll/interleukin-1 receptor (TIR) domain. Phylogenetic analysis grouped it with other teleost Tlr3s. A number of transcription factor binding sites were identified in the 1538 bp 5' flanking region of smtlr3, including interferon-stimulated response element (ISRE) and those for interferon regulatory factors (IRF) and signal transducer and activator of transcriptions (STATs) smtlr3 transcripts were expressed ubiquitously with higher levels in the head kidney, heart and digestion organs. They were up-regulated by both poly I:C and TRBIV in immune and non-immune organs, but most strongly in the head kidney. Finally, the smtlr3 exhibited a two-wave induced expression during a five day time course when exposure of S. maximus to poly I:C. These findings provide insights into the role of SmTlr3 in antiviral response.
Collapse
Affiliation(s)
- G-B Hu
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
| | - X-P Li
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - D-H Liu
- First Institute of Oceanography, State Oceanic Administration of China, Qingdao 266061, China
| | - Q-M Liu
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - S-C Zhang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
24
|
Kinose D, Ogawa E, Kudo M, Marumo S, Kiyokawa H, Hoshino Y, Hirai T, Chin K, Muro S, Mishima M. Association of COPD exacerbation frequency with gene expression of pattern recognition receptors in inflammatory cells in induced sputum. CLINICAL RESPIRATORY JOURNAL 2014; 10:11-21. [PMID: 24902764 DOI: 10.1111/crj.12171] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 04/30/2014] [Accepted: 05/29/2014] [Indexed: 11/29/2022]
Abstract
BACKGROUND Bacteria and viruses are major causes of chronic obstructive pulmonary disease (COPD) exacerbations. Molecular components of these pathogens are recognized by pattern-recognition receptors (PRRs) expressed by various cells in the airway, which leads to initiation of inflammatory processes. Expression levels of PRRs in airway inflammatory cells are expected to affect susceptibility to COPD exacerbation. AIMS This prospective observational study was conducted to detect any association between exacerbation and PRR expression. METHODS Thirty-one male COPD patients were recruited. At baseline, clinical history, lung function measurements, peripheral blood samples and induced sputum were obtained. Using sputum samples, we performed gene expression analysis of TLR2, TLR3, TLR4, NOD1, NOD2, RIG-I and MDA-5 by quantitative reverse transcription-polymerase chain reaction in addition to quantitative bacterial culture. COPD exacerbations were assessed based on Anthonisen's criteria using symptom diaries for the following 1-year period. RESULTS During 1-year follow-up period, 13 patients experienced at least one exacerbation, but 18 patients did not. Those with exacerbations tended to be more severe COPD and showed larger neutrophil fraction in their induced sputum. Among PRRs, only TLR3 gene expression was increased in COPD patients with exacerbation compared with those without exacerbations. Multivariate logistic regression analysis including neutrophil fraction and TLR3 gene expression as predictor variables demonstrated that only an increase of neutrophil fraction, but not TLR3 gene expression, was a significant predictor for COPD exacerbation. CONCLUSION TLR3 expression in inflammatory cells might affect the susceptibility to COPD exacerbation.
Collapse
Affiliation(s)
- Daisuke Kinose
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Emiko Ogawa
- Health Administration Center and Division of Respiratory Medicine, Department of Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Megumi Kudo
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Marumo
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hirofumi Kiyokawa
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuma Hoshino
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuo Chin
- Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shigeo Muro
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Michiaki Mishima
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
25
|
MAVS-dependent IRF3/7 bypass of interferon β-induction restricts the response to measles infection in CD150Tg mouse bone marrow-derived dendritic cells. Mol Immunol 2013; 57:100-10. [PMID: 24096085 DOI: 10.1016/j.molimm.2013.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 08/07/2013] [Accepted: 08/15/2013] [Indexed: 01/14/2023]
Abstract
Measles virus (MV) infects CD150Tg/Ifnar (IFN alpha receptor)(-/-) mice but not CD150 (a human MV receptor)-transgenic (Tg) mice. We have shown that bone marrow-derived dendritic cells (BMDCs) from CD150Tg/Ifnar(-/-) mice are permissive to MV in contrast to those from simple CD150Tg mice, which reveals a crucial role of type I interferon (IFN) in natural tropism against MV. Yet, the mechanism whereby BMDCs produce initial type I IFN has not been elucidated in MV infection. RNA virus infection usually allows cells to generate double-stranded RNA and induce activation of IFN regulatory factor (IRF) 3/7 transcription factors, leading to the production of type I IFN through the retinoic acid-inducible gene I (RIG-I)/melanoma differentiation-associated gene 5 (MDA5)-mitochondrial antiviral signaling protein (MAVS) pathway. In mouse experimental BMDCs models, we found CD150Tg/Mavs(-/-)BMDCs, but not CD150Tg/Irf3(-/-)/Irf7(-/-)BMDCs, permissive to MV. IFN-α/β were not induced in MV-infected CD150Tg/Mavs(-/-)BMDCs, while IFN-β was subtly induced in CD150Tg/Irf3(-/-)/Irf7(-/-)BMDCs. In vivo systemic infection was therefore established by transfer of MV-infected CD150Tg/Mavs(-/-) BMDCs to CD150Tg/Ifnar(-/-) mice. These data indicate that MAVS-dependent, IRF3/7-independent IFN-β induction triggers the activation of the IFNAR pathway so as to restrict the spread of MV by infected BMDCs. Hence, MAVS participates in the initial induction of type I IFN in BMDCs and IFNAR protects against MV spreading. We also showed the importance of IL-10-producing CD4(+) T cells induced by MV-infected BMDCs in vitro, which may account for immune modulation due to the functional aberration of DCs.
Collapse
|
26
|
Multi-step regulation of interferon induction by hepatitis C virus. Arch Immunol Ther Exp (Warsz) 2013; 61:127-38. [PMID: 23292079 DOI: 10.1007/s00005-012-0214-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2012] [Accepted: 12/20/2012] [Indexed: 02/07/2023]
Abstract
Acute hepatitis C virus (HCV) infection evokes several distinct innate immune responses in host, but the virus usually propagates by circumventing these responses. Although a replication intermediate double-stranded RNA is produced in infected cells, type I interferon (IFN) induction and immediate cell death are largely blocked in infected cells. In vitro studies suggested that type I and III IFNs are mainly produced in HCV-infected hepatocytes if the MAVS pathway is functional, and dysfunction of this pathway may lead to cellular permissiveness to HCV replication and production. Cellular immunity, including natural killer cell activation and antigen-specific CD8 T-cell proliferation, occurs following innate immune activation in response to HCV, but is often ineffective for eradication of HCV. Constitutive dsRNA stimulation differs in output from type I IFN therapy, which has been an authentic therapy for patients with HCV. Host innate immune responses to HCV RNA/proteins may be associated with progressive hepatic fibrosis and carcinogenesis once persistent HCV infection is established in opposition to the IFN system. Hence, innate RNA sensing exerts pivotal functions against HCV genome replication and host pathogenesis through modulation of the IFN system. Molecules participating in the RIG-I and Toll-like receptor 3 pathways are the main targets for HCV, disabling the anti-viral functions of these IFN-inducing molecules. We discuss the mechanisms that abolish type I and type III IFN production in HCV-infected cells, which may contribute to understanding the mechanism of virus persistence and resistance to the IFN therapy.
Collapse
|
27
|
Zhou Y, Guo M, Wang X, Li J, Wang Y, Ye L, Dai M, Zhou L, Persidsky Y, Ho W. TLR3 activation efficiency by high or low molecular mass poly I:C. Innate Immun 2012; 19:184-92. [PMID: 23035017 PMCID: PMC3942089 DOI: 10.1177/1753425912459975] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Toll-like receptor 3 (TLR3) plays a critical role in initiating type I IFN-mediated innate immunity against viral infections. TLR3 recognizes various forms of double stranded (ds) RNA, including viral dsRNA and a synthetic mimic of dsRNA, poly I:C, which has been used extensively as a TLR3 ligand to induce antiviral immunity. The activation efficiency of TLR3 by poly I:C is influenced by various factors, including size of the ligands, delivery methods and cell types. In this study, we examined the stimulatory effect of two commercially-available poly I:Cs [high molecular mass (HMM) and low molecular mass (LMM)] on TLR3 activation in various human cell types by determining the induction of type I and type III IFNs, as well as the antiviral effect. We demonstrated that the direct addition of both HMM- and LMM-poly I:C to the cultures of primary macrophages or a neuroplastoma cell line could activate TLR3. However, the transfection of poly I:C was necessary to induce TLR3 activation in other cell types studied. In all the cell lines tested, the efficiency of TLR3 activation by HMM-poly I:C was significantly higher than that by LMM-poly I:C. These observations indicate the importance and necessity of developing effective TLR3 ligands for antiviral therapy.
Collapse
Affiliation(s)
- Yu Zhou
- Center for Animal Experiment/Animal Biosafety Level III Laboratory and State Key Laboratory of Virology, Wuhan University School of Medicine, Wuhan, Hubei, People's Republic of China
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Ming Guo
- Center for Animal Experiment/Animal Biosafety Level III Laboratory and State Key Laboratory of Virology, Wuhan University School of Medicine, Wuhan, Hubei, People's Republic of China
| | - Xu Wang
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Jielang Li
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Yizhong Wang
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Li Ye
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Ming Dai
- Center for Animal Experiment/Animal Biosafety Level III Laboratory and State Key Laboratory of Virology, Wuhan University School of Medicine, Wuhan, Hubei, People's Republic of China
| | - Li Zhou
- Center for Animal Experiment/Animal Biosafety Level III Laboratory and State Key Laboratory of Virology, Wuhan University School of Medicine, Wuhan, Hubei, People's Republic of China
| | - Yuri Persidsky
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Wenzhe Ho
- Center for Animal Experiment/Animal Biosafety Level III Laboratory and State Key Laboratory of Virology, Wuhan University School of Medicine, Wuhan, Hubei, People's Republic of China
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
28
|
Double-stranded RNA induces biphasic STAT1 phosphorylation by both type I interferon (IFN)-dependent and type I IFN-independent pathways. J Virol 2012; 86:12760-9. [PMID: 22973045 DOI: 10.1128/jvi.01881-12] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Upon viral infection, pattern recognition receptors sense viral nucleic acids, leading to the production of type I interferons (IFNs), which initiate antiviral activities. Type I IFNs bind to their cognate receptor, IFNAR, resulting in the activation of signal-transducing activators of transcription 1 (STAT1). Thus, it has long been thought that double-stranded RNA (dsRNA)-induced STAT1 phosphorylation is mediated by the transactivation of type I IFN signaling. Foreign RNA, such as viral RNA, in cells is sensed by the cytoplasmic sensors retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA-5). In this study, we explored the molecular mechanism responsible for STAT1 phosphorylation in response to the sensing of dsRNA by cytosolic RNA sensors. Polyinosinic-poly(C) [poly(I:C)], a synthetic dsRNA that is sensed by both RIG-I and MDA-5, induces STAT1 phosphorylation. We found that the poly(I:C)-induced initial phosphorylation of STAT1 is dependent on the RIG-I pathway and that MDA-5 is not involved in STAT1 phosphorylation. Furthermore, pretreatment of the cells with neutralizing antibody targeting the IFN receptor suppressed the initial STAT1 phosphorylation in response to poly(I:C), suggesting that this initial phosphorylation event is predominantly type I IFN dependent. In contrast, neither the known RIG-I pathway nor type I IFN is involved in the late phosphorylation of STAT1. In addition, poly(I:C) stimulated STAT1 phosphorylation in type I IFN receptor-deficient U5A cells with delayed kinetics. Collectively, our study provides evidence of a comprehensive regulatory mechanism in which dsRNA induces STAT1 phosphorylation, indicating the importance of STAT1 in maintaining very tight regulation of the innate immune system.
Collapse
|
29
|
Clifford HD, Hayden CM, Khoo SK, Naniche D, Mandomando IM, Zhang G, Richmond P, Le Souëf PN. Polymorphisms in key innate immune genes and their effects on measles vaccine responses and vaccine failure in children from Mozambique. Vaccine 2012; 30:6180-5. [PMID: 22871352 DOI: 10.1016/j.vaccine.2012.07.063] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2012] [Revised: 07/23/2012] [Accepted: 07/24/2012] [Indexed: 11/25/2022]
Abstract
Despite an effective vaccine, measles remains a major health problem globally, particularly in developing countries. More than 30% of children show primary vaccine failure and therefore remain vulnerable to measles. Genetic variation in key innate pathogen recognition receptors, such as the measles cell entry receptors CD46 and SLAM, measles attachment receptor DC-SIGN, the antiviral toll-like receptors (TLR)3, TLR7 and TLR8, and the cytosolic antiviral receptor RIG-I, may significantly affect measles IgG antibody responses. Measles is still highly prevalent in developing countries such as those in Africa however there is no previous data on the effect of these innate immune genes in a resident African population. Polymorphisms (n=29) in the candidate genes were genotyped in a cohort of vaccinated children (n=238) aged 6 months-14 years from Mozambique, Africa who either had vaccine failure and contracted measles (cases; n=66) or controls (n=172). Contrasting previous associations with measles responses in Caucasians and/or strong evidence for candidacy, we found little indication that these key innate immune genes affect measles IgG responses in our cohort of Mozambican children. We did however identify that CD46 and TLR8 variants may be involved in the occurrence of measles vaccine failure. This study highlights the importance of genetic studies in resident, non-Caucasian populations, from areas where determining the factors that may affect measles control is of a high priority.
Collapse
Affiliation(s)
- Holly D Clifford
- School of Paediatrics and Child Health, University of Western Australia, Perth, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Clifford HD, Yerkovich ST, Khoo SK, Zhang G, Upham J, Le Souëf PN, Richmond P, Hayden CM. TLR3 and RIG-I gene variants: associations with functional effects on receptor expression and responses to measles virus and vaccine in vaccinated infants. Hum Immunol 2012; 73:677-85. [PMID: 22504413 DOI: 10.1016/j.humimm.2012.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Revised: 02/14/2012] [Accepted: 03/19/2012] [Indexed: 01/08/2023]
Abstract
Measles virus causes severe morbidity and mortality, despite the availability of measles vaccines. Successful defence against viral pathogens requires early recognition of virus-specific patterns by innate receptors like Toll-like receptor (TLR)3 and the RNA helicase, retinoic acid inducible gene-I (RIG-I). Genetic differences in these receptors may influence the primary immune responses to measles and the efficacy of measles vaccine. In 1-year-old Australian infants after their first measles vaccine dose, we investigated functional effects of TLR3 and RIG-I polymorphisms on intracellular protein expression using flow cytometry, cytokine responses to receptor ligands and measles lysate, and post-vaccination measles IgG levels. We found that TLR3 Leu412Phe was significantly associated with IFN-α/β response after stimulation with TLR3 ligand, poly(I:C) (P=0.024). Downregulation of TLR3 protein expression in NK cells after poly(I:C) was also associated with this variant (P=0.011). In contrast, measles-specific expression, cytokine responses and antibody responses were not associated with TLR3 polymorphisms. No associations were found with RIG-I variants. These results suggest that a TLR3 polymorphism has functional effects on receptor expression and cytokine response. However, this did not translate to an effect on specific responses to measles virus or vaccine. We found no evidence that RIG-I polymorphisms were involved in measles immune responses.
Collapse
MESH Headings
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Cells, Cultured
- DEAD Box Protein 58
- DEAD-box RNA Helicases/genetics
- DEAD-box RNA Helicases/immunology
- Female
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/immunology
- Genetic Association Studies
- Humans
- Immunity, Cellular/drug effects
- Immunity, Humoral/drug effects
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Infant
- Interferon-beta/blood
- Interferon-beta/immunology
- Interferon-gamma/blood
- Interferon-gamma/immunology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Male
- Measles/immunology
- Measles/metabolism
- Measles/prevention & control
- Measles virus/immunology
- Measles-Mumps-Rubella Vaccine/administration & dosage
- Measles-Mumps-Rubella Vaccine/immunology
- Poly I-C/pharmacology
- Polymorphism, Single Nucleotide/genetics
- Polymorphism, Single Nucleotide/immunology
- Receptors, Immunologic
- Toll-Like Receptor 3/genetics
- Toll-Like Receptor 3/immunology
- Vaccination
Collapse
Affiliation(s)
- Holly D Clifford
- School of Paediatrics and Child Health, University of Western Australia, Perth, WA, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Griffin DE, Lin WH, Pan CH. Measles virus, immune control, and persistence. FEMS Microbiol Rev 2012; 36:649-62. [PMID: 22316382 DOI: 10.1111/j.1574-6976.2012.00330.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 01/16/2012] [Accepted: 01/17/2012] [Indexed: 12/31/2022] Open
Abstract
Measles remains one of the most important causes of child morbidity and mortality worldwide with the greatest burden in the youngest children. Most acute measles deaths are owing to secondary infections that result from a poorly understood measles-induced suppression of immune responses. Young children are also vulnerable to late development of subacute sclerosing panencephalitis, a progressive, uniformly fatal neurologic disease caused by persistent measles virus (MeV) infection. During acute infection, the rash marks the appearance of the adaptive immune response and CD8(+) T cell-mediated clearance of infectious virus. However, after clearance of infectious virus, MeV RNA persists and can be detected in blood, respiratory secretions, urine, and lymphoid tissue for many weeks to months. This prolonged period of virus clearance may help to explain measles immunosuppression and the development of lifelong immunity to re-infection, as well as occasional infection of the nervous system. Once MeV infects neurons, the virus can spread trans-synaptically and the envelope proteins needed to form infectious virus are unnecessary, accumulate mutations, and can establish persistent infection. Identification of the immune mechanisms required for the clearance of MeV RNA from multiple sites will enlighten our understanding of the development of disease owing to persistent infection.
Collapse
Affiliation(s)
- Diane E Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| | | | | |
Collapse
|
32
|
Ovsyannikova IG, Haralambieva IH, Vierkant RA, Pankratz VS, Jacobson RM, Poland GA. The role of polymorphisms in Toll-like receptors and their associated intracellular signaling genes in measles vaccine immunity. Hum Genet 2011; 130:547-61. [PMID: 21424379 PMCID: PMC3924423 DOI: 10.1007/s00439-011-0977-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 03/08/2011] [Indexed: 12/15/2022]
Abstract
Toll-like receptors (TLRs) and their intracellular signaling molecules play an important role in innate immunity. In this study, we examined associations between polymorphisms in TLR family genes and measles vaccine-specific immune responses. We genotyped 764 subjects (11-22 years old) after two doses of measles vaccine for TLR signaling SNP markers (n = 454). The major alleles of coding SNPs in the TLR2 (rs3804100) and TLR4 (rs5030710) genes were associated with a dose-related increase (660 vs. 892 mIU/ml, p = 0.002) and a dose-related decrease (2,209 vs. 830 mIU/ml, p = 0.001) in measles-specific antibodies, respectively. A significant association was found between lower measles antibody levels and the haplotype ACGGCGAGAAAAGAGAAGAGAGAGAA (p = 0.01) in the MAP3K7 gene. Furthermore, the minor allele of a SNP (rs702966) of the KIAA1542 (IRF7) gene was associated with a dose-related decrease in IFN-γ Elispot responses (38 vs. 26 spot-forming cells per 2 × 10(5) PBMCs, p = 0.00002). We observed an additional 12 associations (p < 0.01) between coding (nonsynonymous and synonymous) polymorphisms within the TLRs (TLR2, 7, and 8), IKBKE, TICAM1, NFKBIA, IRAK2, and KIAA1542 genes and variations in measles-specific IL-2, IL-6, IFN-α, IFN-γ, IFNλ-1, and TNF-α secretion levels. Our data demonstrate that polymorphisms in TLR and other related immune response signaling molecules have significant effects on measles vaccine-associated immune responses. These data help to establish the genetic foundation for immune response variation in response to measles immunization and provide important insights for the rational development of new measles vaccines.
Collapse
Affiliation(s)
- Inna G. Ovsyannikova
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905 USA
- Program in Translational Immunovirology and Biodefense, Mayo Clinic, Rochester, MN 55905 USA
| | | | - Robert A. Vierkant
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905 USA
| | - V. Shane Pankratz
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905 USA
| | - Robert M. Jacobson
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905 USA
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905 USA
- Program in Translational Immunovirology and Biodefense, Mayo Clinic, Rochester, MN 55905 USA
| | - Gregory A. Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905 USA
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905 USA
- Program in Translational Immunovirology and Biodefense, Mayo Clinic, Rochester, MN 55905 USA
| |
Collapse
|
33
|
Karpala AJ, Lowenthal JW, Bean AGD. Identifying innate immune pathways of the chicken may lead to new antiviral therapies. Vet Immunol Immunopathol 2011; 148:100-9. [PMID: 21715024 DOI: 10.1016/j.vetimm.2011.05.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 03/25/2011] [Accepted: 05/30/2011] [Indexed: 12/23/2022]
Abstract
Zoonotic viruses, such as highly pathogenic avian influenza (HPAI), present a significant threat to both the poultry industry and public health. The present method of controlling avian influenza (AI) relies on good farming practice with limited use of vaccination in some countries. However, new ways to control disease outbreaks might be possible with additional knowledge of the natural host response to virus. Moreover, manipulation of the innate immune system in mammals improves the outcomes following viral infection. A similar approach might be applied to the chicken, nevertheless, a greater knowledge of the chicken innate immune system is required. This review outlines important mammalian antiviral mechanisms that have been modulated to strengthen viral immunity and highlights the potential application of these strategies in the chicken, especially in regards, to AI.
Collapse
Affiliation(s)
- Adam J Karpala
- CSIRO, Livestock Industries, Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia.
| | | | | |
Collapse
|
34
|
Matsumoto M, Oshiumi H, Seya T. Antiviral responses induced by the TLR3 pathway. Rev Med Virol 2011; 21:67-77. [PMID: 21312311 DOI: 10.1002/rmv.680] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2010] [Revised: 12/21/2010] [Accepted: 12/21/2010] [Indexed: 12/24/2022]
Abstract
Antiviral responses are successively induced in virus-infected animals, and include primary innate immune responses such as type I interferon (IFN) and cytokine production, secondary natural killer (NK) cell responses, and final cytotoxic T lymphocyte (CTL) responses and antibody production. The endosomal Toll-like receptors (TLRs) and cytoplasmic RIG-I-like receptors (RLRs), which recognize viral nucleic acids, are responsible for virus-induced type I IFN production. RLRs are expressed in most tissues and cells and are primarily implicated in innate immune responses against various viruses through type I IFN production, whereas nucleic acid-sensing TLRs, TLRs 3, 7, 8 and 9, are expressed on the endosomal membrane of dendritic cells (DCs) and play distinct roles in antiviral immunity. TLR3 recognizes viral double-stranded RNA taken up into the endosome and serves to protect the host against viral infection by the induction of a range of responses including type I IFN production and DC-mediated activation of NK cells and CTLs, although the deteriorative role of TLR3 has also been reported in some virus infections. Here, we review the current knowledge on the role of TLR3 during viral infection, and the current understanding of the TLR3-signalling cascade that operates via the adaptor protein TICAM-1 (also called TRIF).
Collapse
Affiliation(s)
- Misako Matsumoto
- Department of Microbiology and Immunology, Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo, Japan.
| | | | | |
Collapse
|
35
|
Fragale A, Stellacci E, Ilari R, Remoli AL, Lanciotti A, Perrotti E, Shytaj I, Orsatti R, Lawrence HR, Lawrence NJ, Wu J, Rehli M, Ozato K, Battistini A. Critical role of IRF-8 in negative regulation of TLR3 expression by Src homology 2 domain-containing protein tyrosine phosphatase-2 activity in human myeloid dendritic cells. THE JOURNAL OF IMMUNOLOGY 2011; 186:1951-62. [PMID: 21220691 DOI: 10.4049/jimmunol.1000918] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Despite extensive studies that unraveled ligands and signal transduction pathways triggered by TLRs, little is known about the regulation of TLR gene expression. TLR3 plays a crucial role in the recognition of viral pathogens and induction of immune responses by myeloid DCs. IFN regulatory factor (IRF)-8, a member of the IRF family, is a transcriptional regulator that plays essential roles in the development and function of myeloid lineage, affecting different subsets of myeloid DCs. In this study, we show that IRF-8 negatively controls TLR3 gene expression by suppressing IRF-1- and/or polyinosinic-polycytidylic acid-stimulated TLR3 expression in primary human monocyte-derived DCs (MDDCs). MDDCs expressed TLR3 increasingly during their differentiation from monocytes to DCs with a peak at day 5, when TLR3 expression was further enhanced upon stimulation with polyinosinic-polycytidylic acid and then was promptly downregulated. We found that both IRF-1 and IRF-8 bind the human TLR3 promoter during MDDC differentiation in vitro and in vivo but with different kinetic and functional effects. We demonstrate that IRF-8-induced repression of TLR3 is specifically mediated by ligand-activated Src homology 2 domain-containing protein tyrosine phosphatase association. Indeed, Src homology 2 domain-containing protein tyrosine phosphatase-dephosphorylated IRF-8 bound to the human TLR3 promoter competing with IRF-1 and quashing its activity by recruitment of histone deacetylase 3. Our findings identify IRF-8 as a key player in the control of intracellular viral dsRNA-induced responses and highlight a new mechanism for negative regulation of TLR3 expression that can be exploited to block excessive TLR activation.
Collapse
Affiliation(s)
- Alessandra Fragale
- Dipartimento di Malattie Infettive, Parassitarie ed Immunomediate, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Khoo JJ, Forster S, Mansell A. Toll-like receptors as interferon-regulated genes and their role in disease. J Interferon Cytokine Res 2011; 31:13-25. [PMID: 21198355 DOI: 10.1089/jir.2010.0095] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The Toll-like receptors (TLRs) are innate sensors that recognize both microbial and endogenous ligands, initiating the host defense response. TLRs initiate the potent proinflammatory response to infection, are the target for adjuvants, and are essential for the establishment and maturation of adaptive immunity. As such they have been the interest of widespread research and the target of therapeutic intervention on multiple diseases. It has become apparent that expression of a subset of TLRs (TLR1, TLR2, TLR3, TLR5, and TLR7) is induced by Type I interferons (IFN). The role and impact of IFN expression on TLR responses is therefore critical in understanding the role of TLRs in disease, particularly as IFN itself is a downstream gene induced by specific TLRs. In this review we discuss the function and role of IFN-regulated TLRs in disease and how the role of IFN may impact upon TLR induction of the immune response in diseases, particularly in mouse models.
Collapse
Affiliation(s)
- Jing Jing Khoo
- Centre for Innate Immunity and Infectious Diseases, Monash Institute of Medical Research, Monash University, Victoria, Australia
| | | | | |
Collapse
|
37
|
Wood LG, Simpson JL, Wark PAB, Powell H, Gibson PG. Characterization of innate immune signalling receptors in virus-induced acute asthma. Clin Exp Allergy 2010; 41:640-8. [PMID: 21129050 DOI: 10.1111/j.1365-2222.2010.03669.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The role of toll-like receptors (TLRs) and innate immune activation in clinical asthma exacerbations and their relationship to virus infection are unclear. OBJECTIVE This study aimed to characterize TLR expression and innate immune activity during virus infection in acute asthma. METHODS Subjects with acute asthma, stable asthma and healthy controls were recruited and underwent spirometry and sputum induction with isotonic saline. Selected sputum was dispersed with dithiothreitol and total and differential leucocyte counts were performed. Selected sputum was also used for quantitative real-time PCR for TLR2, TLR3, TLR4, IL-10 and IP-10mRNA expression. Sputum supernatant was used for the measurement of innate immune markers, including IL-8, matrix metalloproteinase-9 and neutrophil elastase activity. Viruses were detected using real-time and gel-based PCR. RESULTS Sputum TLR2 mRNA expression was up-regulated in both acute and stable asthma compared with healthy controls and decreased 4-6 weeks after acute exacerbation. Sputum TLR2 mRNA expression was elevated in viral, compared with non-viral, acute asthma. Sputum TLR3 mRNA expression was similar in controls, stable and acute asthma. However, in acute asthma, subjects with virus-induced acute asthma had significantly higher sputum TLR3 mRNA expression. Induced sputum gene expression for IP-10 and IL-10 were increased in viral, compared with non-viral, acute asthma. In virus-induced acute asthma, levels of IP-10 and IL-10 mRNA expression were correlated with the mRNA expression of TLR2 and TLR3. CONCLUSIONS AND CLINICAL RELEVANCE Virus-induced acute asthma leads to specific induction of TLR2, TLR3, IP-10 and IL-10, suggesting that signalling via TLRs may play an important role in mediating airway inflammation, via both innate and adaptive pathways, in virus-induced exacerbations. These mediators may provide potential treatment targets for virus-induced asthma. They may also be useful in diagnosing the nature of acute asthma exacerbations and monitoring treatment responses, which would be useful in the clinical management of asthma exacerbations.
Collapse
Affiliation(s)
- L G Wood
- Centre for Asthma and Respiratory Diseases, University of Newcastle, Callaghan, NSW, Australia
| | | | | | | | | |
Collapse
|
38
|
Takaki H, Watanabe Y, Shingai M, Oshiumi H, Matsumoto M, Seya T. Strain-to-strain difference of V protein of measles virus affects MDA5-mediated IFN-β-inducing potential. Mol Immunol 2010; 48:497-504. [PMID: 21071089 DOI: 10.1016/j.molimm.2010.10.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 10/07/2010] [Accepted: 10/12/2010] [Indexed: 12/23/2022]
Abstract
Laboratory-adapted and vaccine strains of measles virus (MV) induce type I interferon (IFN) in infected cells to a far greater extent than wild-type strains. We investigated the mechanisms for this differential type I IFN production in cells infected with representative MV strains. The overexpression of the wild-type V protein suppressed melanoma differentiation-associated gene 5 (MDA5)-induced IFN-β promoter activity, while this was not seen in A549 cells expressing CD150 transfected with the V protein of the vaccine strain. The V proteins of the wild-type also suppressed poly I:C-induced IFN regulatory factor 3 (IRF-3) dimerization. The V proteins of the wild-type and vaccine strain did not affect retinoic acid-inducible gene 1 (RIG-I)- or toll-IL-1R homology domain-containing adaptor molecule 1 (TICAM-1)-induced IFN-β promoter activation. We identified an amino acid substitution of the cysteine residue at position 272 (which is conserved among paramyxoviruses) to an arginine residue in the V protein of the vaccine strain. Only the V protein possessing the 272C residue binds to MDA5. The mutation introduced into the wild-type V protein (C272R) was unable to suppress MDA5-induced IRF-3 nuclear translocation and IFN-β promoter activation as seen in the V proteins of the vaccine strain, whereas the mutation introduced in the vaccine strain V protein (R272C) was able to inhibit MDA5-induced IRF-3 and IFN-β promoter activation. The other 6 residues of the vaccine strain V sequence inconsistent with the authentic sequence of the wild-type V protein barely affected the IRF-3 nuclear translocation. These data suggested that the structural difference of laboratory-adapted [corrected] MV V protein hampers MDA5 blockade and acts as a nidus for the spread/amplification of type I IFN induction. Ultimately, measles vaccine strains have two modes of IFN-β-induction for their attenuation: V protein mutation and production of defective interference (DI) RNA.
Collapse
Affiliation(s)
- Hiromi Takaki
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Kita-ku, Sapporo 060-8638, Japan
| | | | | | | | | | | |
Collapse
|
39
|
Abstract
Measles is an important cause of child mortality that has a seemingly paradoxical interaction with the immune system. In most individuals, the immune response is successful in eventually clearing measles virus (MV) infection and in establishing life-long immunity. However, infection is also associated with persistence of viral RNA and several weeks of immune suppression, including loss of delayed type hypersensitivity responses and increased susceptibility to secondary infections. The initial T-cell response includes CD8+ and T-helper 1 CD4+ T cells important for control of infectious virus. As viral RNA persists, there is a shift to a T-helper 2 CD4+ T-cell response that likely promotes B-cell maturation and durable antibody responses but may suppress macrophage activation and T-helper 1 responses to new infections. Suppression of mitogen-induced lymphocyte proliferation can be induced by lymphocyte infection with MV or by lymphocyte exposure to a complex of the hemagglutinin and fusion surface glycoproteins without infection. Dendritic cells (DCs) are susceptible to infection and can transmit infection to lymphocytes. MV-infected DCs are unable to stimulate a mixed lymphocyte reaction and can induce lymphocyte unresponsiveness through expression of MV glycoproteins. Thus, multiple factors may contribute both to measles-induced immune suppression and to the establishment of durable protective immunity.
Collapse
Affiliation(s)
- Diane E Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| |
Collapse
|
40
|
Gauzzi M, Del Cornò M, Gessani S. Dissecting TLR3 signalling in dendritic cells. Immunobiology 2010; 215:713-23. [DOI: 10.1016/j.imbio.2010.05.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 05/20/2010] [Indexed: 01/10/2023]
|
41
|
Taura M, Fukuda R, Suico MA, Eguma A, Koga T, Shuto T, Sato T, Morino-Koga S, Kai H. TLR3 induction by anticancer drugs potentiates poly I:C-induced tumor cell apoptosis. Cancer Sci 2010; 101:1610-7. [PMID: 20367642 PMCID: PMC11159143 DOI: 10.1111/j.1349-7006.2010.01567.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Toll-like receptor 3 (TLR3) has gained recognition as a novel molecular target for cancer therapy because TLR3 activation by its synthetic ligand poly I:C directly causes tumor cell death. Recently, we reported that tumor suppressor p53 increases the expression of TLR3 in several tumor cell lines. Another study also showed that interferon-alpha (IFN-alpha) up-regulates TLR3 expression. We thus hypothesized that various anticancer drugs such as p53-activating reagents and IFNs may potentiate poly I:C-induced tumor cell death through the up-regulation of TLR3 expression. Here, we screened several anticancer drugs that, together with poly I:C, effectively cause tumor cell death in colon carcinoma HCT116 cells. We found that the DNA-damaging reagent 5-fluorouracil (5-FU) increased TLR3 mRNA expression and potentiated poly I:C-induced apoptosis in HCT116 p53(+/+) cells but had only minimal effect in p53(-/-) cells, indicating a p53-dependent pathway. On the other hand, IFN-alpha increased poly I:C-induced apoptosis and the TLR3 mRNA level in HCT116 p53(+/+) and p53(-/-) cell lines. Furthermore, the combination of poly I:C, 5-FU and IFN-alpha induced the highest apoptosis in HCT116 p53(+/+) and p53(-/-) cells. Taken together, these data suggest that the anticancer drugs increased TLR3 expression and subsequently potentiated poly I:C-induced apoptosis likely via p53-dependent and -independent pathways. Considering that the p53 status in malignant cells is heterogeneous, this combination approach may provide a highly effective tumor therapy.
Collapse
Affiliation(s)
- Manabu Taura
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Global COE Cell Fate Regulation Research and Education Unit, Kumamoto University, Kumamoto, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Friboulet L, Gourzones C, Tsao SW, Morel Y, Paturel C, Témam S, Uzan C, Busson P. Poly(I:C) induces intense expression of c-IAP2 and cooperates with an IAP inhibitor in induction of apoptosis in cancer cells. BMC Cancer 2010; 10:327. [PMID: 20576118 PMCID: PMC2928000 DOI: 10.1186/1471-2407-10-327] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Accepted: 06/24/2010] [Indexed: 12/31/2022] Open
Abstract
Background There is increasing evidence that the toll-like receptor 3 (TLR3) is an interesting target for anti-cancer therapy. Unfortunately, most laboratory investigations about the impact of TLR3 stimulation on human malignant cells have been performed with very high concentrations - 5 to 100 μg/ml - of the prototype TLR3 ligand, poly(I:C). In a previous study focused on a specific type of human carcinoma - nasopharyngeal carcinoma - we have shown that concentrations of poly(I:C) as low as 100 ng/ml are sufficient to induce apoptosis of malignant cells when combined to a pharmacological antagonist of the IAP family based on Smac mimicry. Methods This observation prompted us to investigate the contribution of the IAP family in cell response to poly(I:C) in a variety of human malignant cell types. Results We report a rapid, intense and selective increase in c-IAP2 protein expression observed under stimulation by poly(I:C)(500 ng/ml) in all types of human malignant cells. In most cell types, this change in protein expression is underlain by an increase in c-IAP2 transcripts and dependent on the TLR3/TRIF pathway. When poly(I:C) is combined to the IAP inhibitor RMT 5265, a cooperative effect in apoptosis induction and/or inhibition of clonogenic growth is obtained in a large fraction of carcinoma and melanoma cell lines. Conclusions Currently, IAP inhibitors like RMT 5265 and poly(I:C) are the subject of separate therapeutic trials. In light of our observations, combined use of both types of compounds should be considered for treatment of human malignancies including carcinomas and melanomas.
Collapse
Affiliation(s)
- Luc Friboulet
- Univ Paris-Sud, CNRS-UMR 8126 and Institut de Cancérologie Gustave Roussy, 39 rue Camille Desmoulins, F-94805 Villejuif, France
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Poland GA, Ovsyannikova IG, Jacobson RM. Application of pharmacogenomics to vaccines. Pharmacogenomics 2009; 10:837-52. [PMID: 19450131 DOI: 10.2217/pgs.09.25] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The field of pharmacogenomics and pharmacogenetics provides a promising science base for vaccine research and development. A broad range of phenotype/genotype data combined with high-throughput genetic sequencing and bioinformatics are increasingly being integrated into this emerging field of vaccinomics. This paper discusses the hypothesis of the 'immune response gene network' and genetic (and bioinformatic) strategies to study associations between immune response gene polymorphisms and variations in humoral and cellular immune responses to prophylactic viral vaccines, such as measles-mumps-rubella, influenza, HIV, hepatitis B and smallpox. Immunogenetic studies reveal promising new vaccine targets by providing a better understanding of the mechanisms by which gene polymorphisms may influence innate and adaptive immune responses to vaccines, including vaccine failure and vaccine-associated adverse events. Additional benefits from vaccinomic studies include the development of personalized vaccines, the development of novel vaccines and the development of novel vaccine adjuvants.
Collapse
Affiliation(s)
- Gregory A Poland
- Mayo Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | |
Collapse
|
44
|
Ciencewicki JM, Brighton LE, Jaspers I. Localization of type I interferon receptor limits interferon-induced TLR3 in epithelial cells. J Interferon Cytokine Res 2009; 29:289-97. [PMID: 19231996 DOI: 10.1089/jir.2008.0075] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Previous studies have shown that influenza infections increase Toll-like receptor 3 (TLR3) expression and that type I interferons (IFNs) may play a role in this response. This study aimed to expand on the role of type I IFNs in the influenza-induced upregulation of TLR3 and determine whether and how the localization of the IFN-alpha/beta receptor (IFNAR) in respiratory epithelial cells could modify IFN-induced responses. Using differentiated primary human airway epithelial cells this study demonstrates that soluble mediators secreted in response to influenza infection upregulate TLR3 expression in naive cells. This response was associated with an upregulation of type I IFNs and stimulation with type I, but not type II, IFNs enhanced TLR3 expression. Interestingly, although influenza infection results in IFN-beta release both toward the apical and basolateral sides of the epithelium, TLR3 expression is only enhanced in cells stimulated with IFN-beta from the basolateral side. Immunohistochemical analysis demonstrates that IFNAR expression is limited to the basolateral side of differentiated human airway epithelial cells. However, non- or poorly differentiated epithelial cells express IFNAR more toward the apical side. These data demonstrate that restricted expression of the IFNAR in the differentiated airway epithelium presents a potential mechanism of regulating type I IFN-induced TLR3 expression.
Collapse
Affiliation(s)
- Jonathan M Ciencewicki
- Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27599-7310, USA
| | | | | |
Collapse
|
45
|
Labreuche Y, O'Leary NA, de la Vega E, Veloso A, Gross PS, Chapman RW, Browdy CL, Warr GW. Lack of evidence for Litopenaeus vannamei Toll receptor (lToll) involvement in activation of sequence-independent antiviral immunity in shrimp. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2009; 33:806-810. [PMID: 19428481 DOI: 10.1016/j.dci.2009.02.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2009] [Revised: 02/19/2009] [Accepted: 02/19/2009] [Indexed: 05/27/2023]
Abstract
Injection of non-specific dsRNA initiates a broad-spectrum innate antiviral immune response in the Pacific white shrimp, Litopenaeus vannamei, however, the receptor involved in recognition of this by-product of viral infections remains unknown. In vertebrates, dsRNA sensing is mediated by a class of Toll-like receptors (TLRs) and results in activation of the interferon system. Because a TLR (lToll) was recently characterized in L. vannamei, we investigated its potential role in dsRNA recognition. We showed that injection of non-specific RNA duplexes did not modify lToll gene expression. A reverse genetic approach was therefore implemented to study its role in vivo. Silencing of lToll did not impair the ability of non-specific dsRNA to trigger protection from white spot syndrome virus and did not increase the shrimp susceptibility to viral infection, when compared to controls. In contrast, gene-specific dsRNA injected to specifically silence lToll expression activated an antiviral response. These data strongly suggest that shrimp lToll plays no role in dsRNA-induced antiviral immunity.
Collapse
Affiliation(s)
- Yannick Labreuche
- Marine Biomedicine and Environmental Sciences Center, Medical University of South Carolina, Charleston, SC 29412, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Karpala AJ, Morris KR, Broadway MM, McWaters PGD, O'Neil TE, Goossens KE, Lowenthal JW, Bean AGD. Molecular cloning, expression, and characterization of chicken IFN -lambda. J Interferon Cytokine Res 2009; 28:341-50. [PMID: 18593329 DOI: 10.1089/jir.2007.0117] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Interferons (IFN) provide a critical first line of defense against viral infection in vertebrates. Moreover, IFN-lambda, a recently identified group of mammalian IFN, has demonstrated antiviral potential in the treatment of mammalian viruses. With the growing concern over such diseases as avian influenza (AI), there is a pressing need for new antiviral strategies to manage problem viruses in poultry. Furthermore, the use of immune molecules, such as IFN-lambda, provides an attractive option for treating poultry by augmenting the host response to virus. With this in mind, we report here the first cloning, expression, and analysis of biologic activity of chicken IFN-lambda (ChIFN-lambda). We compared the similarity of ChIFN-lambda to those identified in other species and demonstrate that ChIFN-lambda has antiviral properties similar to those of human IFN-lambda (HuIFN-lambda). Our results demonstrate that in the chicken, as in human, the antiviral activity demonstrated by ChIFN-lambda supports its inclusion in therapeutic strategies directed against viral infections.
Collapse
Affiliation(s)
- Adam J Karpala
- CSIRO, Livestock Industries, Australian Animal Health Laboratory, Geelong, Victoria, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Hahm B. Hostile communication of measles virus with host innate immunity and dendritic cells. Curr Top Microbiol Immunol 2009; 330:271-87. [PMID: 19203114 DOI: 10.1007/978-3-540-70617-5_13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Following measles virus (MV) infection, host innate immune responses promptly operate to purge the virus. Detection of alerting measles viral components or replication intermediates by pattern-recognizing host machinery of Toll-like receptors and RNA helicases triggers signaling to synthesize array of anti-viral and immunoregulatory molecules, including type I interferon (IFN). Diverse subtypes of dendritic cells (DCs) play pivotal roles in both host innate immunity on the primary MV-infected site and initiating adaptive immune responses on secondary lymphoid tissues. Responding to the predictable host immune responses, MV appears to have devised multiple strategies to evade, suppress, or even utilize host innate immunity and DC responses. This review focuses on versatile actions of MV-induced type I IFNs causing beneficial or deleterious influence on host immunity and the interplay between MV and heterogeneous DCs at distinct locations.
Collapse
Affiliation(s)
- B Hahm
- Department of Surgery, Center for Cellular and Molecular Immunology, University of Missouri-Columbia School of Medicine, One Hospital Dr., Columbia, MO 65212, USA.
| |
Collapse
|
48
|
Oshiumi H, Matsumoto M, Hatakeyama S, Seya T. Riplet/RNF135, a RING finger protein, ubiquitinates RIG-I to promote interferon-beta induction during the early phase of viral infection. J Biol Chem 2009; 284:807-17. [PMID: 19017631 DOI: 10.1074/jbc.m804259200] [Citation(s) in RCA: 294] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
RIG-I (retinoic acid-inducible gene-I), a cytoplasmic RNA helicase, interacts with IPS-1/MAVS/Cardif/VISA, a protein on the outer membrane of mitochondria, to signal the presence of virus-derived RNA and induce type I interferon production. Activation of RIG-I requires the ubiquitin ligase, TRIM25, which mediates lysine 63-linked polyubiquitination of the RIG-I N-terminal CARD-like region. However, how this modification proceeds for activation of IPS-1 by RIG-I remains unclear. Here we identify an alternative factor, Riplet/RNF135, that promotes RIG-I activation independent of TRIM25. The Riplet/RNF135 protein consists of an N-terminal RING finger domain, C-terminal SPRY and PRY motifs, and shows sequence similarity to TRIM25. Immunoprecipitation analyses demonstrated that the C-terminal helicase and repressor domains of RIG-I interact with the Riplet/RNF135 C-terminal region, whereas the CARD-like region of RIG-I is dispensable for this interaction. Riplet/RNF135 promotes lysine 63-linked polyubiquitination of the C-terminal region of RIG-I, modification of which differs from the N-terminal ubiquitination by TRIM25. Overexpression and knockdown analyses revealed that Riplet/RNF135 promotes RIG-I-mediated interferon-beta promoter activation and inhibits propagation of the negative-strand RNA virus, vesicular stomatitis virus. Our data suggest that Riplet/RNF135 is a novel factor of the RIG-I pathway that is involved in the evoking of human innate immunity against RNA virus infection, and activates RIG-I through ubiquitination of its C-terminal region. We infer that a variety of RIG-I-ubiquitinating molecular complexes sustain RIG-I activation to modulate RNA virus replication in the cytoplasm.
Collapse
Affiliation(s)
- Hiroyuki Oshiumi
- Department of Microbiology and Immunology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | | | | | | |
Collapse
|
49
|
Poland GA, Ovsyannikova IG, Jacobson RM. Vaccine immunogenetics: bedside to bench to population. Vaccine 2008; 26:6183-8. [PMID: 18598732 PMCID: PMC2614670 DOI: 10.1016/j.vaccine.2008.06.057] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2008] [Accepted: 06/13/2008] [Indexed: 01/16/2023]
Abstract
The immunogenetic basis for variations in immune response to vaccines in humans remains largely unknown. Many factors can contribute to the heterogeneity of vaccine-induced immune responses, including polymorphisms of immune response genes. It is important to identify those genes involved directly or indirectly in the generation of the immune response to vaccines. Our previous work with measles reveals the impact of immune response gene polymorphisms on measles vaccine-induced humoral and cellular immune responses. We demonstrate associations between genetic variations (single nucleotide polymorphisms, SNPs) in HLA class I and class II genes, cytokine, cell surface receptor, and toll-like receptor genes and variations in immune responses to measles vaccine. Such information may provide further understanding of genetic restrictions that influence the generation of protective immune responses to vaccines, and eventually the development of new vaccines.
Collapse
Affiliation(s)
- Gregory A Poland
- Mayo Vaccine Research Group, The Program in Translational Immunovirology and Biodefense, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| | | | | |
Collapse
|
50
|
Ishizaki Y, Takemoto M, Kira R, Kusuhara K, Torisu H, Sakai Y, Sanefuji M, Yukaya N, Hara T. Association of toll-like receptor 3 gene polymorphism with subacute sclerosing panencephalitis. J Neurovirol 2008; 14:486-91. [PMID: 19016379 DOI: 10.1080/13550280802298120] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Innate immunity plays an important role in measles virus (MV) infection. MV-derived double-stranded RNA is recognized by toll-like receptor 3 (TLR3), retinoic acid-inducible protein I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5). We investigated whether genes encoding these molecules contributed to the development of subacute sclerosing panencephalitis (SSPE) in Japanese individuals. Four single nucleotide polymorphisms (SNPs) of the three genes (TLR3 rs3775291:Leu412Phe, RIG1 rs277729 and rs9695310, and MDA5 rs4664463) were assessed in 40 SSPE patients and 84 controls. Because the TLR3 SNP showed a positive association with SSPE, three additional SNPs were subjected to haplotype analysis. The frequency of 412Phe allele of TLR3 rs3775291 in SSPE patients was significantly higher than that in controls (P=.03). In haplotype analysis of four SNPs in the TLR3 gene, the frequency of -7C/IVS3+71C/Phe412/c.1377C haplotype was significantly increased in SSPE patients (P=.006, odds ration [OR]: 2.2). TLR3 gene may confer host genetic susceptibility to SSPE in Japanese individuals.
Collapse
Affiliation(s)
- Yoshito Ishizaki
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|