1
|
Huang M, Xu M, Han J, Ke E, Niu X, Zhang Y, Wang G, Yu H, Liu R, Wang S, Liu Y, Chen Y, Han J, Wu Z, Cui H, Zhang Y, Duan Y, Gao Y, Qi X. Enhancing MyD88 oligomerization is one important mechanism by which IBDV VP2 induces inflammatory response. PLoS Pathog 2025; 21:e1012985. [PMID: 40067802 PMCID: PMC11957393 DOI: 10.1371/journal.ppat.1012985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/31/2025] [Accepted: 02/16/2025] [Indexed: 04/02/2025] Open
Abstract
The inflammatory response is an essential component of innate immunity to defense against pathogens. Infectious bursal disease (IBD) is the most important immunosuppressive disease in chickens and is caused by the infectious bursal disease virus (IBDV). Acute inflammation is a typical pathogenic process for IBD, however, the underlying mechanism is not clear. Here, we report that IBDV induces obvious inflammatory response in vivo and in vitro. Furthermore, viral VP2 is identified as an important inflammatory stimulus. It is observed that IBDV VP2 can activate NF-κB signaling pathway and then increase IL-1β production. In detail, IBDV VP2 interacts with myeloid differentiation primary response gene 88 (MyD88), potentiates the oligomerization of MyD88 and assembly of MyD88 complex, which is one important element leading to NF-κB signaling pathway activation and IL-1β production increase. More meaningfully, residues 253/284 of viral VP2 are significantly involved in IBDV-induced inflammatory response through modulating the interaction strength between VP2 and MyD88 and the following MyD88-NF-κB-IL-1β signaling pathway. This study reveals one molecular mechanism that trigger inflammation during IBDV infection, which is of great significance for a deeper understanding of the pathogenic mechanisms of IBDV.
Collapse
Affiliation(s)
- Mengmeng Huang
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Mengmeng Xu
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Jingzhe Han
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Erjing Ke
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Xinxin Niu
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yulong Zhang
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Guodong Wang
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hangbo Yu
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Runhang Liu
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Suyan Wang
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Yongzhen Liu
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Yuntong Chen
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Jinze Han
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Ziwen Wu
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hongyu Cui
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Yanping Zhang
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Yulu Duan
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Yulong Gao
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| | - Xiaole Qi
- Avian Immunosuppressive Diseases Division, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- World Organization for Animal Health (WOAH) Reference Laboratory for Infectious Bursal Disease, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
| |
Collapse
|
2
|
Kumar R, Meena AS, Baraiya T, Swarnkar CP, Misra SS, Kumar A. Expression of Toll-like receptors in Haemonchus Contortus resistant sheep: An innate immune parameter for host defense against gastrointestinal nematode infection. Vet Immunol Immunopathol 2024; 275:110813. [PMID: 39142124 DOI: 10.1016/j.vetimm.2024.110813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/17/2024] [Accepted: 08/01/2024] [Indexed: 08/16/2024]
Abstract
Innate immune parameters, a first line of defense against invading pathogens like bacteria, parasites, fungi, etc, play a significant role in the prevention and elimination of aetiological agents primarily by recognition of invading pathogen-specific molecules by different pattern recognition receptors. Toll-like receptors (TLRs), a type-I transmembrane glycoprotein, cause innate immune responses mainly by produing inflammatory cytokines, chemokines and interferons. The objective of present study was to determine the role of TLRs in parasite resistance in Malpura sheep. In the current study, transcript variation of TLRs and its downstream signalling molecules namely MyD88, TRIF, IRF-3, TRAF, TGF-β, NFκB, and CD14 were ascertained by real-time PCR in Haemonchus contortus resistant (R) and susceptible (S) Malpura sheep. Results have shown significantly (P<0.05) up-regulated expression of TLR-2, TLR-4, TLR-5, TLR-8 and TLR-10 in July however down-regulated patterns were observed in August and September in R-line sheep compared to S-line sheep. This indicates that at more or less equal parasite load, the TLR genes in R sheep produce more transcripts, but after parasite loads have increased hugely in the S line, they easily surpass the levels seen in the S line. Result suggests that transcriptional activity of the TLR genes was related to parasite load and there were differences between the lines at different infection intensities. Three-point transcript expression observation of the signalling molecules namely TRIF, IRF-3, TRAF, a similar pattern was observed in R sheep compared with S sheep.
Collapse
Affiliation(s)
- Rajiv Kumar
- ICAR-Central Sheep and Wool Research Institute, Avikanagar. Rajasthan 304501, India.
| | - A S Meena
- ICAR-Central Sheep and Wool Research Institute, Avikanagar. Rajasthan 304501, India
| | - Trusha Baraiya
- ICAR-Central Sheep and Wool Research Institute, Avikanagar. Rajasthan 304501, India
| | - C P Swarnkar
- ICAR-Central Sheep and Wool Research Institute, Avikanagar. Rajasthan 304501, India
| | - S S Misra
- ICAR-Central Sheep and Wool Research Institute, Avikanagar. Rajasthan 304501, India
| | - Arun Kumar
- ICAR-Central Sheep and Wool Research Institute, Avikanagar. Rajasthan 304501, India
| |
Collapse
|
3
|
Liu Y, Yuan J, Zhang Y, Qin F, Bai X, Sun W, Chen T, Liu F, Zheng Y, Qi X, Zhao W, Liu B, Gao C. OTUD5 promotes the inflammatory immune response by enhancing MyD88 oligomerization and Myddosome formation. Cell Death Differ 2024; 31:753-767. [PMID: 38605168 PMCID: PMC11164869 DOI: 10.1038/s41418-024-01293-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/23/2024] [Accepted: 04/02/2024] [Indexed: 04/13/2024] Open
Abstract
Myddosome is an oligomeric complex required for the transmission of inflammatory signals from TLR/IL1Rs and consists of MyD88 and IRAK family kinases. However, the molecular basis for the self-assemble of Myddosome proteins and regulation of intracellular signaling remains poorly understood. Here, we identify OTUD5 acts as an essential regulator for MyD88 oligomerization and Myddosome formation. OTUD5 directly interacts with MyD88 and cleaves its K11-linked polyubiquitin chains at Lys95, Lys231 and Lys250. This polyubiquitin cleavage enhances MyD88 oligomerization after LPS stimulation, which subsequently promotes the recruitment of downstream IRAK4 and IRAK2 to form Myddosome and the activation of NF-κB and MAPK signaling and production of inflammatory cytokines. Consistently, Otud5-deficient mice are less susceptible to LPS- and CLP-induced sepsis. Taken together, our findings reveal a positive regulatory role of OTUD5 in MyD88 oligomerization and Myddosome formation, which provides new sights into the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Yaxing Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P.R. China
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Jiahua Yuan
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P.R. China
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Yuling Zhang
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P.R. China
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Fei Qin
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P.R. China
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Xuemei Bai
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P.R. China
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Wanwei Sun
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P.R. China
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Tian Chen
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P.R. China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Feng Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P.R. China
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Yi Zheng
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P.R. China
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Xiaopeng Qi
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Wei Zhao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P.R. China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Bingyu Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P.R. China.
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P.R. China.
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P.R. China.
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P.R. China.
| |
Collapse
|
4
|
Flow cytometric reporter assays provide robust functional analysis of signaling complexes. J Biol Chem 2022; 298:102666. [PMID: 36334634 PMCID: PMC9747584 DOI: 10.1016/j.jbc.2022.102666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 10/26/2022] [Accepted: 10/31/2022] [Indexed: 11/05/2022] Open
Abstract
Conventional assays to probe signaling protein interactions and function involve measurement of luciferase reporter expression within the bulk cell population, with lack of control over target-protein expression level. To address this issue, we have developed a rapid and robust flow cytometric assay for analysis of signaling protein function. A fluorescent reporter and fluorescent tagging of the target protein enables simultaneous assessment of protein expression and signaling within individual cells. We have applied our technique to the analysis of variants of the lipopolysaccharide receptor Toll-like receptor 4 (TLR4) and its adapter protein MyD88, using a NF-кB-responsive promoter driving mScarlet-I expression. The assay enables exclusion of nontransfected cells and overexpressing cells that signal spontaneously. Additionally, our assay allows the identification of protein variants that fail to express. We found that the assays were highly sensitive, with cells expressing an appropriate level of GFP-MyD88 showing approximately 200-fold induction of mScarlet-I by lipopolysaccharide, and we can detect subtle protein concentration-dependent effects of mutations. Importantly, the assay is adaptable to various signaling pathways.
Collapse
|
5
|
Gosu V, Won K, Oh JD, Shin D. Conformational Changes Induced by S34Y and R98C Variants in the Death Domain of Myd88. Front Mol Biosci 2020; 7:27. [PMID: 32266286 PMCID: PMC7106778 DOI: 10.3389/fmolb.2020.00027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 02/10/2020] [Indexed: 11/13/2022] Open
Abstract
Myeloid differentiating factor 88 (Myd88) is a universal adaptor protein that plays a critical role in innate immunity by mediating TLR downstream signaling. Myd88 death domain (DD) forms an oligomeric complex by association with other DD-containing proteins such as IRAK4. Despite its universal role, polymorphisms in Myd88 can result in several diseases. Previous studies have suggested that, out of several non-synonymous single-nucleotide polymorphisms (nsSNPs), the variants S34Y and R98C in the DD of Myd88 disrupt the formation of the Myddosome complex. Therefore, we performed molecular dynamics (MD) simulations on wild-type (Myd88WT) and mutant (Myd88S34Y, Myd88R98C) DDs to evaluate the subtle conformational changes induced by these mutations. Our results suggest that the S34Y variant induces large structural transitions compared to the R98C variant as evidenced by residual flexibility at the variable loop regions, particularly in the H1-H2 loop, and variations in the collective modes of motion observed for wild-type and mutant Myd88 DDs. The residue interaction network strongly suggests a distortion in the interaction pattern at the location of the mutated residue between the wild type and mutants. Moreover, betweenness centrality values indicate that variations in the distribution of functionally important residues may be reflected by distinct residue signal transductions in both wild-type and mutant Myd88 DDs, which may influence the interaction with other DDs in TLR downstream signaling.
Collapse
Affiliation(s)
- Vijayakumar Gosu
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju-si, South Korea
| | - KyeongHye Won
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju-si, South Korea
| | - Jae-Don Oh
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju-si, South Korea
| | - Donghyun Shin
- The Animal Molecular Genetics and Breeding Center, Jeonbuk National University, Jeonju-si, South Korea
| |
Collapse
|
6
|
Xiong MG, Xu ZS, Li YH, Wang SY, Wang YY, Ran Y. RNF152 positively regulates TLR/IL-1R signaling by enhancing MyD88 oligomerization. EMBO Rep 2020; 21:e48860. [PMID: 31930677 DOI: 10.15252/embr.201948860] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 11/09/2022] Open
Abstract
Toll-like receptors (TLRs) are important pattern recognition receptors (PRRs) that are critical for the defense against invading pathogens. IL-1β is an important pro-inflammatory cytokine that also plays pivotal roles in shaping the adaptive immune response. TLRs and interleukin-1 receptor (IL-1R) share similar cytosolic domains and signaling processes. In this study, we identify the E3 ubiquitin ligase RNF152 as a positive regulator of TLR/IL-1R-mediated signaling. Overexpression of RNF152 potentiates IL-1β- and LPS-induced NF-κB activation in an ubiquitination-independent manner, whereas knockdown of RNF152 has the opposite effects. RNF152-deficient mice produce less inflammatory cytokines in response to LPS and are more resistant to LPS-induced lethal endotoxemia. Mechanistically, RNF152 interacts with the adaptor protein MyD88 and enhances oligomerization of MyD88, which is essential for the recruitment of downstream signaling components and activation of TLR/IL-1R-mediated signal transduction. Our findings suggest that RNF152-mediated oligomerization of MyD88 is important for TLR/IL-1R-mediated inflammatory response.
Collapse
Affiliation(s)
- Mei-Guang Xiong
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhi-Sheng Xu
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yu-Hui Li
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Su-Yun Wang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yan-Yi Wang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yong Ran
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
7
|
Innate Immune Signaling Organelles Display Natural and Programmable Signaling Flexibility. Cell 2019; 177:384-398.e11. [PMID: 30853218 DOI: 10.1016/j.cell.2019.01.039] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 12/09/2018] [Accepted: 01/24/2019] [Indexed: 12/18/2022]
Abstract
The signaling organelles of the innate immune system consist of oligomeric protein complexes known as supramolecular organizing centers (SMOCs). Examples of SMOCs include myddosomes and inflammasomes, which respectively induce transcription-dependent and -independent inflammatory responses. The common use of oligomeric structures as signaling platforms suggests multifunctionality, but each SMOC has a singular biochemically defined function. Here, we report that the myddosome is a multifunctional organizing center. In addition to promoting inflammatory transcription factor activation, the myddosome drives the rapid induction of glycolysis. We identify the kinase TBK1 as a myddosome component that promotes glycolysis, but not nuclear factor κB (NF-κB) activation. Synthetic immunology approaches further diversified SMOC activities, as we created interferon- or necroptosis-inducing myddosomes, inflammasomes that induce interferon responses instead of pyroptosis, and a SMOC-like nanomachine that induces interferon expression in response to a chemical ligand. These discoveries demonstrate the flexibility of immune signaling organelles, which permits the design of user-defined innate immune responses.
Collapse
|
8
|
Zhou H, Harberts E, Fishelevich R, Gaspari AA. TLR4 acts as a death receptor for ultraviolet radiation (UVR) through IRAK-independent and FADD-dependent pathway in macrophages. Exp Dermatol 2018; 25:949-955. [PMID: 27676214 DOI: 10.1111/exd.13222] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2016] [Indexed: 12/15/2022]
Abstract
UVR-induced apoptosis in cutaneous antigen presenting cells (APC) causes systemic immune suppression and is dependent on TLR4/MyD88 signalling, but the apoptotic signalling pathways have not been defined. Macrophages pretreated with lipopolysaccharide (LPS) were unresponsive to subsequent LPS treatment, however, but were susceptible to UVR-induced apoptosis. Macrophage survival and apoptotic events after UVR were also unaffected by treatment with TLR4 antagonists, a blocking IgG or a TLR4 analog antagonist, suggesting that UVR cell death is independent of a soluble ligand. After UVR, IRAK4KDKI (catalytically inactive IRAK4) and wild-type (WT) macrophages show equivalent levels of survival, as measured by MTT assay, and apoptosis, as measured by cleaved caspase-3. Furthermore, in macrophages from both mice, UVR activated caspase-8 and PARP, while inactivating Rip3. This finding is supported by a lack of IRAK1 degradation after UVR, compared to treatment with TLR2 or TLR4 agonists. UVR induced association of MyD88 with FADD, an extrinsic apoptotic pathway protein, but not IRAK4. UVR-induced migration of FADD to the cell membrane of WT macrophages, but not MyD88-/- macrophages, was observed (confocal microscopy). Co-immunoprecipitation using an epitope-tagged MyD88 revealed that FADD, but not TRADD, was recruited to MyD88 within 30 minutes of UVR exposure. UVR engages TLR4/MyD88 as a death signalling complex, rather than the classical inflammatory signalling pathway triggered by PAMP recognition of TLR4. These studies provide the rationale for the future development of topical TLR4 modulating therapies to interfere with this UVB-mediated apoptosis and the associated negative consequences of immune suppression.
Collapse
Affiliation(s)
- Hua Zhou
- Department of Dermatology, University Maryland Baltimore, Baltimore, MD, USA
| | - Erin Harberts
- Department of Dermatology, University Maryland Baltimore, Baltimore, MD, USA.,Department of Microbiology and Immunology, University of Maryland Baltimore, Baltimore, MD, USA
| | - Rita Fishelevich
- Department of Dermatology, University Maryland Baltimore, Baltimore, MD, USA
| | - Anthony A Gaspari
- Department of Dermatology, University Maryland Baltimore, Baltimore, MD, USA.,Department of Microbiology and Immunology, University of Maryland Baltimore, Baltimore, MD, USA.,Research Service, Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| |
Collapse
|
9
|
Ali YB, Foad RM, Abdel-Wahed E. Lack of Associations between TLR9 and MYD88 Gene Polymorphisms and Risk of Chronic Lymphocytic Leukemia. Asian Pac J Cancer Prev 2017; 18:3245-3250. [PMID: 29286214 PMCID: PMC5980878 DOI: 10.22034/apjcp.2017.18.12.3245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Background: Genetic factors like single nucleotide polymorphisms (SNPs) may play an important role in the etiology of chronic lymphocytic leukemia (CLL). Mutations in Toll like receptor 9 (TLR9) and myeloid differentiation primary response 88 (MYD88) genes may lead to an abnormal immune response that may cause greater cell proliferation and thus alter an individual’s susceptibility to haematological malignancies including CLL. Objective: This work was designed to study any association of the TLR9 (rs2066807C/G and rs187084T/C) and MYD88 (L265P) single nucleotide polymorphism (SNPs) with risk of CLL in Egyptians. Materials and methods: One hundred patients with CLL and 100 healthy controls from the Egyptian population were genotyped by the polymerase chain reaction/restriction fragment length polymorphism (PCR/RFLP) method. Results: With TLR9 rs2066807C/G the CC genotype was more frequent in both control and patient groups while for TLR9 rs187084T/C the TT genotype was most common. There were no significant associations with CLL risk. With MYD88 (L265P) only the TT genotype was detected. Conclusion: Our preliminary data suggest that polymorphisms in the TLR9 and MYD88 genes may not contribute to CLL susceptibility. To the best of our knowledge, this study is the first dealing with TLR9 and MYD88 gene polymorphisms in CLL patients. Further studies with larger sample size should be conducted to validate these results in the Egyptian population.
Collapse
Affiliation(s)
- Yasser B.M Ali
- Molecular Biology Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), University of Sadat City, Egypt
| | | | | |
Collapse
|
10
|
TLR Specific Immune Responses against Helminth Infections. J Parasitol Res 2017; 2017:6865789. [PMID: 29225962 PMCID: PMC5684585 DOI: 10.1155/2017/6865789] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/21/2017] [Accepted: 10/03/2017] [Indexed: 01/07/2023] Open
Abstract
Despite marked improvement in the quality of lives across the globe, more than 2 million individuals in socioeconomically disadvantaged environments remain infected by helminth (worm) parasites. Owing to the longevity of the worms and paucity of immunologic controls, these parasites survive for long periods within the bloodstream, lymphatics, and gastrointestinal tract resulting in pathologic conditions such as anemia, cirrhosis, and lymphatic filariasis. Despite infection, an asymptomatic state may be maintained by the host immunoregulatory environment, which involves multiple levels of regulatory cells and cytokines; a breakdown of this regulation is observed in pathological disease. The role of TLR expression and function in relation to intracellular parasites has been documented but limited studies are available for multicellular helminth parasites. In this review, we discuss the unique and shared host effector mechanisms elicited by systemic helminth parasites and their derived products, including the role of TLRs and sphingolipids. Understanding and exploiting the interactions between these parasites and the host regulatory network are likely to highlight new strategies to control both infectious and immunological diseases.
Collapse
|
11
|
Structural basis of TIR-domain-assembly formation in MAL- and MyD88-dependent TLR4 signaling. Nat Struct Mol Biol 2017; 24:743-751. [PMID: 28759049 DOI: 10.1038/nsmb.3444] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022]
Abstract
Toll-like receptor (TLR) signaling is a key innate immunity response to pathogens. Recruitment of signaling adapters such as MAL (TIRAP) and MyD88 to the TLRs requires Toll/interleukin-1 receptor (TIR)-domain interactions, which remain structurally elusive. Here we show that MAL TIR domains spontaneously and reversibly form filaments in vitro. They also form cofilaments with TLR4 TIR domains and induce formation of MyD88 assemblies. A 7-Å-resolution cryo-EM structure reveals a stable MAL protofilament consisting of two parallel strands of TIR-domain subunits in a BB-loop-mediated head-to-tail arrangement. Interface residues that are important for the interaction are conserved among different TIR domains. Although large filaments of TLR4, MAL or MyD88 are unlikely to form during cellular signaling, structure-guided mutagenesis, combined with in vivo interaction assays, demonstrated that the MAL interactions defined within the filament represent a template for a conserved mode of TIR-domain interaction involved in both TLR and interleukin-1 receptor signaling.
Collapse
|
12
|
Imbert PR, Louche A, Luizet JB, Grandjean T, Bigot S, Wood TE, Gagné S, Blanco A, Wunderley L, Terradot L, Woodman P, Garvis S, Filloux A, Guery B, Salcedo SP. A Pseudomonas aeruginosa TIR effector mediates immune evasion by targeting UBAP1 and TLR adaptors. EMBO J 2017; 36:1869-1887. [PMID: 28483816 PMCID: PMC5494471 DOI: 10.15252/embj.201695343] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 03/29/2017] [Accepted: 04/05/2017] [Indexed: 12/11/2022] Open
Abstract
Bacterial pathogens often subvert the innate immune system to establish a successful infection. The direct inhibition of downstream components of innate immune pathways is particularly well documented but how bacteria interfere with receptor proximal events is far less well understood. Here, we describe a Toll/interleukin 1 receptor (TIR) domain‐containing protein (PumA) of the multi‐drug resistant Pseudomonas aeruginosa PA7 strain. We found that PumA is essential for virulence and inhibits NF‐κB, a property transferable to non‐PumA strain PA14, suggesting no additional factors are needed for PumA function. The TIR domain is able to interact with the Toll‐like receptor (TLR) adaptors TIRAP and MyD88, as well as the ubiquitin‐associated protein 1 (UBAP1), a component of the endosomal‐sorting complex required for transport I (ESCRT‐I). These interactions are not spatially exclusive as we show UBAP1 can associate with MyD88, enhancing its plasma membrane localization. Combined targeting of UBAP1 and TLR adaptors by PumA impedes both cytokine and TLR receptor signalling, highlighting a novel strategy for innate immune evasion.
Collapse
Affiliation(s)
- Paul Rc Imbert
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique, University of Lyon, Lyon, France
| | - Arthur Louche
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique, University of Lyon, Lyon, France
| | - Jean-Baptiste Luizet
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique, University of Lyon, Lyon, France
| | - Teddy Grandjean
- EA 7366 Recherche Translationelle Relations Hôte-Pathogènes, Faculté de Médecine Pôle Recherche, Université Lille 2, Lille, France
| | - Sarah Bigot
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique, University of Lyon, Lyon, France
| | - Thomas E Wood
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Stéphanie Gagné
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique, University of Lyon, Lyon, France
| | - Amandine Blanco
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique, University of Lyon, Lyon, France
| | - Lydia Wunderley
- School of Biological Sciences, Faculty of Biology Medicine and Health University of Manchester Manchester Academic Health Science Centre, Manchester, UK†
| | - Laurent Terradot
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique, University of Lyon, Lyon, France
| | - Philip Woodman
- School of Biological Sciences, Faculty of Biology Medicine and Health University of Manchester Manchester Academic Health Science Centre, Manchester, UK†
| | - Steve Garvis
- Laboratoire de Biologie et Modelisation, Ecole Normal Supérieur, UMR5239, Lyon, France
| | - Alain Filloux
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Benoit Guery
- EA 7366 Recherche Translationelle Relations Hôte-Pathogènes, Faculté de Médecine Pôle Recherche, Université Lille 2, Lille, France
| | - Suzana P Salcedo
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique, University of Lyon, Lyon, France
| |
Collapse
|
13
|
Perelman SS, Abrams ME, Eitson JL, Chen D, Jimenez A, Mettlen M, Schoggins JW, Alto NM. Cell-Based Screen Identifies Human Interferon-Stimulated Regulators of Listeria monocytogenes Infection. PLoS Pathog 2016; 12:e1006102. [PMID: 28002492 PMCID: PMC5176324 DOI: 10.1371/journal.ppat.1006102] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 11/29/2016] [Indexed: 12/15/2022] Open
Abstract
The type I interferon (IFN) activated transcriptional response is a critical antiviral defense mechanism, yet its role in bacterial pathogenesis remains less well characterized. Using an intracellular pathogen Listeria monocytogenes (Lm) as a model bacterial pathogen, we sought to identify the roles of individual interferon-stimulated genes (ISGs) in context of bacterial infection. Previously, IFN has been implicated in both restricting and promoting Lm growth and immune stimulatory functions in vivo. Here we adapted a gain-of-function flow cytometry based approach to screen a library of more than 350 human ISGs for inhibitors and enhancers of Lm infection. We identify 6 genes, including UNC93B1, MYD88, AQP9, and TRIM14 that potently inhibit Lm infection. These inhibitors act through both transcription-mediated (MYD88) and non-transcriptional mechanisms (TRIM14). Further, we identify and characterize the human high affinity immunoglobulin receptor FcγRIa as an enhancer of Lm internalization. Our results reveal that FcγRIa promotes Lm uptake in the absence of known host Lm internalization receptors (E-cadherin and c-Met) as well as bacterial surface internalins (InlA and InlB). Additionally, FcγRIa-mediated uptake occurs independently of Lm opsonization or canonical FcγRIa signaling. Finally, we established the contribution of FcγRIa to Lm infection in phagocytic cells, thus potentially linking the IFN response to a novel bacterial uptake pathway. Together, these studies provide an experimental and conceptual basis for deciphering the role of IFN in bacterial defense and virulence at single-gene resolution. While the type I interferon response is known to be activated by both viruses and bacteria, it has mostly been characterized in terms of its antiviral properties. Listeria monocytogenes, an opportunistic Gram-positive bacterial pathogen with up to 50% mortality rate and a variety of clinical manifestations, is a potent activator of interferon secretion. In mouse models, interferon has been previously implicated in both restricting and promoting L. monocytogenes infection. Here, we utilized a high-throughput flow-cytometry based approach to screen a library of human interferon I stimulated genes (ISGs) and identified regulators of L. monocytogenes infection. These include inhibitors that act through both transcriptional (MYD88) and transcription-independent (TRIM14) mechanisms. Strikingly, expression of the human high affinity immunoglobulin receptor FcγRIa (CD64) was found to potently enhance L. monocytogenes infection. Both biochemical and cellular studies indicate that FcγRIa increases primary invasion of L. monocytogenes through a previously uncharacterized IgG-independent internalization mechanism. Together, these studies provide an important insight into the complex role of interferon response in bacterial virulence and host defense.
Collapse
Affiliation(s)
- Sofya S. Perelman
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Michael E. Abrams
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Jennifer L. Eitson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Didi Chen
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Alyssa Jimenez
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Marcel Mettlen
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - John W. Schoggins
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- * E-mail: (JWS); (NMA)
| | - Neal M. Alto
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- * E-mail: (JWS); (NMA)
| |
Collapse
|
14
|
Wang J, Grishin AV, Ford HR. Experimental Anti-Inflammatory Drug Semapimod Inhibits TLR Signaling by Targeting the TLR Chaperone gp96. THE JOURNAL OF IMMUNOLOGY 2016; 196:5130-7. [PMID: 27194788 DOI: 10.4049/jimmunol.1502135] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 04/18/2016] [Indexed: 01/07/2023]
Abstract
Semapimod, a tetravalent guanylhydrazone, suppresses inflammatory cytokine production and has potential in a variety of inflammatory and autoimmune disorders. The mechanism of action of Semapimod is not well understood. In this study, we demonstrate that in rat IEC-6 intestinal epithelioid cells, Semapimod inhibits activation of p38 MAPK and NF-κB and induction of cyclooxygenase-2 by TLR ligands, but not by IL-1β or stresses. Semapimod inhibits TLR4 signaling (IC50 ≈0.3 μmol) and acts by desensitizing cells to LPS; it fails to block responses to LPS concentrations of ≥5 μg/ml. Inhibition of TLR signaling by Semapimod is almost instantaneous: the drug is effective when applied simultaneously with LPS. Semapimod blocks cell-surface recruitment of the MyD88 adapter, one of the earliest events in TLR signaling. gp96, the endoplasmic reticulum-localized chaperone of the HSP90 family critically involved in the biogenesis of TLRs, was identified as a target of Semapimod using ATP-desthiobiotin pulldown and mass spectroscopy. Semapimod inhibits ATP-binding and ATPase activities of gp96 in vitro (IC50 ≈0.2-0.4 μmol). On prolonged exposure, Semapimod causes accumulation of TLR4 and TLR9 in perinuclear space, consistent with endoplasmic reticulum retention, an anticipated consequence of impaired gp96 chaperone function. Our data indicate that Semapimod desensitizes TLR signaling via its effect on the TLR chaperone gp96. Fast inhibition by Semapimod is consistent with gp96 participating in high-affinity sensing of TLR ligands in addition to its role as a TLR chaperone.
Collapse
Affiliation(s)
- Jin Wang
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, CA 90027; and
| | - Anatoly V Grishin
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, CA 90027; and Department of Surgery, University of Southern California, Los Angeles, CA 90027
| | - Henri R Ford
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, CA 90027; and Department of Surgery, University of Southern California, Los Angeles, CA 90027
| |
Collapse
|
15
|
Wang H, El Maadidi S, Fischer J, Grabski E, Dickhöfer S, Klimosch S, Flannery SM, Filomena A, Wolz OO, Schneiderhan-Marra N, Löffler MW, Wiese M, Pichulik T, Müllhaupt B, Semela D, Dufour JF, Bochud PY, Bowie AG, Kalinke U, Berg T, Weber ANR. A frequent hypofunctional IRAK2 variant is associated with reduced spontaneous hepatitis C virus clearance. Hepatology 2015; 62:1375-87. [PMID: 26250868 DOI: 10.1002/hep.28105] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 08/03/2015] [Indexed: 12/29/2022]
Abstract
UNLABELLED Patients carrying very rare loss-of-function mutations in interleukin-1 receptor-associated kinase 4 (IRAK4), a critical signaling mediator in Toll-like receptor signaling, are severely immunodeficient, highlighting the paramount role of IRAK kinases in innate immunity. We discovered a comparatively frequent coding variant of the enigmatic human IRAK2, L392V (rs3844283), which is found homozygously in ∼15% of Caucasians, to be associated with a reduced ability to induce interferon-alpha in primary human plasmacytoid dendritic cells in response to hepatitis C virus (HCV). Cytokine production in response to purified Toll-like receptor agonists was also impaired. Additionally, rs3844283 was epidemiologically associated with a chronic course of HCV infection in two independent HCV cohorts and emerged as an independent predictor of chronic HCV disease. Mechanistically, IRAK2 L392V showed intact binding to, but impaired ubiquitination of, tumor necrosis factor receptor-associated factor 6, a vital step in signal transduction. CONCLUSION Our study highlights IRAK2 and its genetic variants as critical factors and potentially novel biomarkers for human antiviral innate immunity.
Collapse
Affiliation(s)
- Hui Wang
- Junior Research Group Toll-Like Receptors and Cancer, German Cancer Research Center, Heidelberg, Germany
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Souhayla El Maadidi
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Janett Fischer
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
- Section of Hepatology, Clinic for Gastroenterology and Rheumatology, University Hospital Leipzig, Leipzig, Germany
| | - Elena Grabski
- Institute for Experimental Infection Research, TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Sabine Dickhöfer
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Sascha Klimosch
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Sinead M Flannery
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Angela Filomena
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Olaf-Oliver Wolz
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | | | - Markus W Löffler
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
- Department of General, Visceral, and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Manfred Wiese
- Section of Hepatology, Clinic for Gastroenterology and Rheumatology, University Hospital Leipzig, Leipzig, Germany
| | - Tica Pichulik
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Beat Müllhaupt
- Gastroenterology and Hepatology Department, University Hospital Zurich, Zurich, Switzerland
| | - David Semela
- Department of Gastroenterology and Hepatology, Canton Hospital St. Gallen, St. Gallen, Switzerland
| | - Jean-François Dufour
- Hepatology Section, Department Visceral Surgery and Medicine, University Hospital Bern, Bern, Switzerland
| | | | - Andrew G Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Thomas Berg
- Section of Hepatology, Clinic for Gastroenterology and Rheumatology, University Hospital Leipzig, Leipzig, Germany
| | - Alexander N R Weber
- Junior Research Group Toll-Like Receptors and Cancer, German Cancer Research Center, Heidelberg, Germany
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
16
|
Du J, Nicolaes GA, Kruijswijk D, Versloot M, van der Poll T, van 't Veer C. The structure function of the death domain of human IRAK-M. Cell Commun Signal 2014; 12:77. [PMID: 25481771 PMCID: PMC4273448 DOI: 10.1186/s12964-014-0077-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 11/21/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND IRAK-M is an inhibitor of Toll-like receptor signaling that acts by re-directing IRAK-4 activity to TAK1 independent NF-κB activation and by inhibition of IRAK-1/IRAK-2 activity. IRAK-M is expressed in monocytes/macrophages and lung epithelial cells. Lack of IRAK-M in mice greatly improves the resistance to nosocomial pneumonia and lung tumors, which entices IRAK-M as a potential therapeutic target. IRAK-M consists of an N-terminal death domain (DD), a dysfunctional kinase domain and unstructured C-terminal domain. Little is known however on IRAK-M's structure-function relationships. RESULTS Since death domains provide the important interactions of IRAK-1, IRAK-2 and IRAK-4 molecules, we generated a 3D structure model of the human IRAK-M-DD (residues C5-G119) to guide mutagenesis studies and predict protein-protein interaction points. First we identified the DD residues involved in the endogenous capacity of IRAK-M to activate NF-κB that is displayed upon overexpression in 293T cells. W74 and R97, at distinct interfaces of the IRAK-M-DD, were crucial for this endogenous NF-κB activating capacity, as well as the C-terminal domain (S445-E596) of IRAK-M. Resulting anti-inflammatory A20 and pro-inflammatory IL-8 transcription in 293T cells was W74 dependent, while IL-8 protein expression was dependent on R97 and the TRAF6 binding motif at P478. The IRAK-M-DD W74 and R97 binding interfaces are predicted to interact with opposite sides of IRAK-4-DD's. Secondly we identified DD residues important for the inhibitory action of IRAK-M by stable overexpression of mutants in THP-1 macrophages and H292 lung epithelial cells. IRAK-M inhibited TLR2/4-mediated cytokine production in macrophages in a manner that is largely dependent on W74. R97 was not involved in inhibition of TNF production but was engaged in IL-6 down-regulation by IRAK-M. Protein-interactive residues D19-A23, located in between W74 and R97, were also observed to be crucial for inhibition of TLR2/4 mediated cytokine induction in macrophages. Remarkably, IRAK-M inhibited TLR5 mediated IL-8 production by lung epithelial cells independent of W74 and R97, but dependent on D19-A23 and R70, two surface-exposed regions that harbor predicted IRAK-2-DD interaction points of IRAK-M. CONCLUSION IRAK-M employs alternate residues of its DD to inhibit the different inflammatory mediators induced by varying TLRs and cells.
Collapse
|
17
|
Saba TG, Chung Y, Hong JY, Sajjan US, Bentley JK, Hershenson MB. Rhinovirus-induced macrophage cytokine expression does not require endocytosis or replication. Am J Respir Cell Mol Biol 2014; 50:974-84. [PMID: 24783958 DOI: 10.1165/rcmb.2013-0354oc] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Rhinovirus (RV) is responsible for the majority of virus-induced asthma exacerbations. We showed previously that RV infection of ovalbumin-sensitized and -challenged BALB/c mice induces production of type 2 cytokines from M2-polarized macrophages. In the present study, we sought to determine the mechanism of RV-induced cytokine expression. We infected bone marrow-derived macrophages (BMMs) from BALB/c mice with RV serotype 1B, a minor group virus that infects mouse cells. Selected cultures were pretreated with IL-4, a type 2 cytokine increased in allergic asthma. RV infection of untreated cells increased messenger RNA and protein expression of the M1 cytokines TNF-α, CXCL1, and IL-6 but failed to induce expression of the M2 cytokines CCL22 and CCL24. Cells pretreated with IL-4 showed decreased expression of M1 cytokines but increased expression of Ym-1, Arg-1 (M2 markers), CCL22, and CCL24. Infection with ultraviolet (UV)-irradiated, replication-deficient RV elicited similar cytokine responses, suggesting that the outcome is replication independent. Consistent with this, viral RNA copy number did not increase in RV-treated BMMs or bronchoalveolar macrophages. RV-induced cytokine expression was not affected when cells were pretreated with cytochalasin D, suggesting that viral endocytosis is not required for the response. Finally, RV-induced cytokine expression and viral attachment were abolished in BMMs from myeloid differentiation factor 88 and Toll-like receptor (TLR)2 KO mice, suggesting a specific requirement of TLR2. We conclude that RV elicits a proinflammatory cytokine response in BMMs through a cell-surface-mediated, TLR2-dependent mechanism that does not require viral endocytosis or replication.
Collapse
Affiliation(s)
- Thomas G Saba
- Departments of 1 Pediatrics and Communicable Diseases and
| | | | | | | | | | | |
Collapse
|
18
|
Wang H, Flannery SM, Dickhöfer S, Huhn S, George J, Kubarenko AV, Lascorz J, Bevier M, Willemsen J, Pichulik T, Schafmayer C, Binder M, Manoury B, Paludan SR, Alarcon-Riquelme M, Bowie AG, Försti A, Weber ANR. A coding IRAK2 protein variant compromises Toll-like receptor (TLR) signaling and is associated with colorectal cancer survival. J Biol Chem 2014; 289:23123-23131. [PMID: 24973222 DOI: 10.1074/jbc.m113.492934] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Within innate immune signaling pathways, interleukin-1 receptor-associated kinases (IRAKs) fulfill key roles downstream of multiple Toll-like receptors and the interleukin-1 receptor. Although human IRAK4 deficiency was shown to lead to severe immunodeficiency in response to pyogenic bacterial infection during childhood, little is known about the role of human IRAK2. We here identified a non-synonymous IRAK2 variant, rs35060588 (coding R214G), as hypofunctional in terms of NF-κB signaling and Toll-like receptor-mediated cytokine induction. This was due to reduced ubiquitination of TRAF6, a key step in signal transduction. IRAK2 rs35060588 occurs in 3-9% of individuals in different ethnic groups, and our studies suggested a genetic association of rs35060588 with colorectal cancer survival. This for the first time implicates human IRAK2 in a human disease and highlights the R214G IRAK2 variant as a potential novel and broadly applicable biomarker for disease or as a therapeutic intervention point.
Collapse
Affiliation(s)
- Hui Wang
- Junior Research Group Toll-like Receptors and Cancer and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany; Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Sinead M Flannery
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Sabine Dickhöfer
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Stefanie Huhn
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Julie George
- Junior Research Group Toll-like Receptors and Cancer and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Andriy V Kubarenko
- Junior Research Group Toll-like Receptors and Cancer and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Jesus Lascorz
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Melanie Bevier
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Joschka Willemsen
- Department of Infectious Diseases/Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, 69120 Heidelberg, Germany
| | - Tica Pichulik
- Junior Research Group Toll-like Receptors and Cancer and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Clemens Schafmayer
- Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, 24105 Kiel, Germany,; POPGEN Biobank Project, Christian-Albrechts University, 24105 Kiel, Germany
| | - Marco Binder
- Department of Infectious Diseases/Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, 69120 Heidelberg, Germany
| | - Bénédicte Manoury
- INSERM, Unité 1013 and Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, 75015 Paris, France
| | - Søren R Paludan
- Department of Biomedicine, Aarhus University, Bartholin Building, 8000 Aarhus, Denmark
| | - Marta Alarcon-Riquelme
- Pfizer-Universidad de Granada-Junta de Andalucía Centre for Genomics and Oncological Research, Avenida de la Ilustración 114, 18016 Granada, Spain,; Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104,; BIOLUPUS Network, European Science Foundation, F-67080 Strasbourg Cedex, France, and
| | - Andrew G Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Asta Försti
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany,; Center for Primary Health Care Research, Clinical Research Center, Lund University, 20502 Malmö, Sweden
| | - Alexander N R Weber
- Junior Research Group Toll-like Receptors and Cancer and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany; Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany,.
| |
Collapse
|
19
|
Functional assessment of the mutational effects of human IRAK4 and MyD88 genes. Mol Immunol 2013; 58:66-76. [PMID: 24316379 DOI: 10.1016/j.molimm.2013.11.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 11/08/2013] [Accepted: 11/09/2013] [Indexed: 01/08/2023]
Abstract
Human interleukin-1 receptor-associated kinase 4 (IRAK4) deficiency and myeloid differentiating factor 88 (MyD88) deficiency syndromes are two primary immune-deficiency disorders with innate immune defects. Although new genetic variations of IRAK4 and MyD88 have recently been deposited in the single nucleotide polymorphism (SNP) database, the clinical significance of these variants has not yet been established. Therefore, it is important to establish methods for assessing the association of each gene variation with human diseases. Because cell-based assays, western blotting and an NF-κB reporter gene assay, showed no difference in protein expression and NF-κB activity between R12C and wild-type IRAK4, we examined protein-protein interactions of purified recombinant IRAK4 and MyD88 proteins by analytical gel filtration and NMR titration. We found that the variant of IRAK4, R12C, as well as R20W, located in the death domain of IRAK4 and regarded as a SNP, caused a loss of interaction with MyD88. Our studies suggest that not only the loss of protein expression but also the defect of Myddosome formation could cause IRAK4 and MyD88 deficiency syndromes. Moreover a combination of in vitro functional assays is effective for confirming the pathogenicity of mutants found in IRAK4 and MyD88-deficiency patients.
Collapse
|
20
|
Fekonja O, Benčina M, Jerala R. Toll/interleukin-1 receptor domain dimers as the platform for activation and enhanced inhibition of Toll-like receptor signaling. J Biol Chem 2012; 287:30993-1002. [PMID: 22829600 DOI: 10.1074/jbc.m112.376186] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
TIR (Toll/IL-1 receptor) domains mediate interactions between TLR (Toll-like) or IL-1 family receptors and signaling adapters. While homotypic TIR domain interactions mediate receptor activation they are also usurped by microbial TIR domain containing proteins for immunosuppression. Here we show the role of a dimerized TIR domain platform for the suppression as well as for the activation of MyD88 signaling pathway. Coiled-coil dimerization domain, present in many bacterial TCPs, potently augments suppression of TLR/IL-1R signaling. The addition of a strong coiled-coil dimerization domain conferred the superior inhibition against the wide spectrum of TLRs and prevented the constitutive activation by a dimeric TIR platform. We propose a molecular model of MyD88-mediated signaling based on the dimerization of TIR domains as the limiting step.
Collapse
Affiliation(s)
- Ota Fekonja
- Laboratory of Biotechnology, National Institute of Chemistry, 1000 Ljubljana, Slovenia
| | | | | |
Collapse
|
21
|
Netea MG, Wijmenga C, O'Neill LAJ. Genetic variation in Toll-like receptors and disease susceptibility. Nat Immunol 2012; 13:535-42. [PMID: 22610250 DOI: 10.1038/ni.2284] [Citation(s) in RCA: 258] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Toll-like receptors (TLRs) are key initiators of the innate immune response and promote adaptive immunity. Much has been learned about the role of TLRs in human immunity from studies linking TLR genetic variation with disease. First, monogenic disorders associated with complete deficiency in certain TLR pathways, such as MyD88-IRAK4 or TLR3-Unc93b-TRIF-TRAF3, have demonstrated the specific roles of these pathways in host defense against pyogenic bacteria and herpesviruses, respectively. Second, common polymorphisms in genes encoding several TLRs and associated genes have been associated with both infectious and autoimmune diseases. The study of genetic variation in TLRs in various populations combined with information on infection has demonstrated complex interaction between genetic variation in TLRs and environmental factors. This interaction explains the differences in the effect of TLR polymorphisms on susceptibility to infection and autoimmune disease in various populations.
Collapse
Affiliation(s)
- Mihai G Netea
- Department of Internal Medicine, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | | | | |
Collapse
|
22
|
Bauerfeld CP, Rastogi R, Pirockinaite G, Lee I, Hüttemann M, Monks B, Birnbaum MJ, Franchi L, Nuñez G, Samavati L. TLR4-mediated AKT activation is MyD88/TRIF dependent and critical for induction of oxidative phosphorylation and mitochondrial transcription factor A in murine macrophages. THE JOURNAL OF IMMUNOLOGY 2012; 188:2847-57. [PMID: 22312125 DOI: 10.4049/jimmunol.1102157] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Mitochondria play a critical role in cell survival and death. Mitochondrial recovery during inflammatory processes such as sepsis is associated with cell survival. Recovery of cellular respiration, mitochondrial biogenesis, and function requires coordinated expression of transcription factors encoded by nuclear and mitochondrial genes, including mitochondrial transcription factor A (T-fam) and cytochrome c oxidase (COX, complex IV). LPS elicits strong host defenses in mammals with pronounced inflammatory responses, but also triggers activation of survival pathways such as AKT pathway. AKT/PKB is a serine/threonine protein kinase that plays an important role in cell survival, protein synthesis, and controlled inflammation in response to TLRs. Hence we investigated the role of LPS-mediated AKT activation in mitochondrial bioenergetics and function in cultured murine macrophages (B6-MCL) and bone marrow-derived macrophages. We show that LPS challenge led to increased expression of T-fam and COX subunits I and IV in a time-dependent manner through early phosphorylation of the PI3K/AKT pathway. PI3K/AKT pathway inhibitors abrogated LPS-mediated T-fam and COX induction. Lack of induction was associated with decreased ATP production, increased proinflammatory cytokines (TNF-α), NO production, and cell death. The TLR4-mediated AKT activation and mitochondrial biogenesis required activation of adaptor protein MyD88 and Toll/IL-1R domain-containing adaptor-inducing IFN-β. Importantly, using a genetic approach, we show that the AKT1 isoform is pivotal in regulating mitochondrial biogenesis in response to TLR4 agonist.
Collapse
Affiliation(s)
- Christian P Bauerfeld
- Department of Pediatrics, St. John Hospital and Medical Center, Detroit, MI 48236, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Theofilopoulos AN, Kono DH, Beutler B, Baccala R. Intracellular nucleic acid sensors and autoimmunity. J Interferon Cytokine Res 2011; 31:867-86. [PMID: 22029446 DOI: 10.1089/jir.2011.0092] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
A collection of molecular sensors has been defined by studies in the last decade that can recognize a diverse array of pathogens and initiate protective immune and inflammatory responses. However, if the molecular signatures recognized are shared by both foreign and self-molecules, as is the case of nucleic acids, then the responses initiated by these sensors may have deleterious consequences. Notably, this adverse occurrence may be of primary importance in autoimmune disease pathogenesis. In this case, microbe-induced damage or mishandled physiologic processes could lead to the generation of microparticles containing self-nucleic acids. These particles may inappropriately gain access to the cytosol or endolysosomes and, hence, engage resident RNA and DNA sensors. Evidence, as reviewed here, strongly indicates that these sensors are primary contributors to autoimmune disease pathogenesis, spearheading efforts toward development of novel therapeutics for these disorders.
Collapse
Affiliation(s)
- Argyrios N Theofilopoulos
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California 92137, USA.
| | | | | | | |
Collapse
|
24
|
Iliev DB, Sobhkhez M, Fremmerlid K, Jørgensen JB. MyD88 interacts with interferon regulatory factor (IRF) 3 and IRF7 in Atlantic salmon (Salmo salar): transgenic SsMyD88 modulates the IRF-induced type I interferon response and accumulates in aggresomes. J Biol Chem 2011; 286:42715-42724. [PMID: 21990356 DOI: 10.1074/jbc.m111.293969] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
MyD88 is an intracellular adaptor protein that transmits signals downstream of immune receptors such as the IL-1 receptor and the majority of the known mammalian toll-like receptors. Homologs of MyD88 have been identified in many vertebrate species; however, the adaptor has been studied mostly in mammals, and little is known about its function in lower vertebrates. The results presented in the current paper demonstrate, for the first time, that the teleost MyD88, through its Toll/Interleukin-1 receptor domain, interacts with SsIRF3 and two SsIRF7 paralogs: transcription factors that are critically involved in the virus-induced IFN responses. The data further highlight the potential of transgenic SsMyD88 to modulate the IRF-induced type I IFN response as the adaptor synergized with SsIRF3 to activate IRF-E/IFN-stimulated response element-containing reporter gene constructs and endogenous myxovirus resistance homolog expression. Microscopy analyses demonstrated that, similar to mammalian MyD88, both endogenous and transgenic SsMyD88 accumulated in intracellular aggregates. However, unlike the endogenous SsMyD88 clusters, which co-localized with endocytosed CpGs and probably represented myddosomes, overexpressed SsMyD88 accumulated in aggresomes. Although these structures accumulated ubiquitinated proteins, they did not associate with the autophagosome markers p62 and light chain 3-like protein, indicating that they are most likely classical aggresomes rather than aggresome-like induced structures, aggregates of ubiquitinated proteins induced by toll-like receptor/MyD88 signaling in antigen-presenting cells. The significance of the accumulation of transgenic MyD88 in aggresomes is currently unknown; nevertheless it is tempting to speculate that it might represent a defense mechanism against the potentially harmful effects of excessive MyD88 signaling.
Collapse
Affiliation(s)
- Dimitar B Iliev
- Faculty of Biosciences, Fisheries and Economics, University of Tromsø, N-9037 Tromsø, Norway
| | - Mehrdad Sobhkhez
- Faculty of Biosciences, Fisheries and Economics, University of Tromsø, N-9037 Tromsø, Norway
| | - Kjersti Fremmerlid
- Faculty of Biosciences, Fisheries and Economics, University of Tromsø, N-9037 Tromsø, Norway
| | - Jorunn B Jørgensen
- Faculty of Biosciences, Fisheries and Economics, University of Tromsø, N-9037 Tromsø, Norway
| |
Collapse
|
25
|
Sun S, Wang X, Wu X, Zhao Y, Wang F, Liu X, Song Y, Wu Z, Liu M. Toll-like receptor activation by helminths or helminth products to alleviate inflammatory bowel disease. Parasit Vectors 2011; 4:186. [PMID: 21943110 PMCID: PMC3199248 DOI: 10.1186/1756-3305-4-186] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2011] [Accepted: 09/27/2011] [Indexed: 12/11/2022] Open
Abstract
Helminth infection may modulate the expression of Toll like receptors (TLR) in dendritic cells (DCs) and modify the responsiveness of DCs to TLR ligands. This may regulate aberrant intestinal inflammation in humans with helminthes and may thus help alleviate inflammation associated with human inflammatory bowel disease (IBD). Epidemiological and experimental data provide further evidence that reducing helminth infections increases the incidence rate of such autoimmune diseases. Fine control of inflammation in the TLR pathway is highly desirable for effective host defense. Thus, the use of antagonists of TLR-signaling and agonists of their negative regulators from helminths or helminth products should be considered for the treatment of IBD.
Collapse
Affiliation(s)
- ShuMin Sun
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, Jilin University, Zoonosis Research Centre of State Key Laboratory for Molecular Virology and Genetic Engineering, Institute of Pathogen Biology, Chinese Academy of Medical Sciences, Changchun 130062, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|