1
|
Xu M, Tian N, Liu Y, Qian K, Shao H, Ye J, Qin A. Identification of host factors affecting Chicken Infectious Anemia Virus infection and pathogenicity through RNA-seq and LC-MS. Vet Microbiol 2025; 305:110524. [PMID: 40279723 DOI: 10.1016/j.vetmic.2025.110524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/09/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025]
Abstract
Chicken Infectious Anemia Virus (CIAV) infection causes severe anemia, hematopoietic disorders, and immunosuppression in poultry. However, the mechanisms underlying its pathogenicity remain poorly understood. In this study, RNA sequencing (RNA-seq) was employed to investigate the transcriptional response of MSB1 cells to CIAV infection at 12, 24, and 48 hours post-infection. The results revealed differential gene expression associated with immune response, inflammatory response, apoptosis, and metabolic pathways. Several immune-related factors, including IL18, TRAF1, MYD88, IRF2, and CD28, were found to be downregulated. VP1, the capsid protein of CIAV, plays a significant role in viral pathogenicity. To explore how VP1 affects CIAV replication and pathogenicity, co-immunoprecipitation (Co-IP) was used to identify host proteins interacting with VP1. Liquid chromatography-mass spectrometry (LC-MS) analysis revealed that VP1 interacts with a range of host proteins involved in metabolic pathways, the PI3K-AKT signaling pathway, and ribosomal functions. Based on RNA-seq and LC-MS findings, ANXA6 and HSP90aa1 were identified as key interacting partners, which co-localize in the cytoplasm of infected cells. Both proteins are involved in immune responses and metabolic regulation and were found to be downregulated in CIAV-infected cells. Notably, overexpression of ANXA6 or HSP90aa1 inhibited CIAV infection during the early stages of the viral lifecycle. These findings enhance our understanding of the host factors influencing CIAV infection and provide new insights into potential therapeutic targets for managing CIAV-induced diseases.
Collapse
Affiliation(s)
- Moru Xu
- Ministry of Education Key Lab for Avian Preventive Medicine, College of Veterinary Medicine, Yangzhou University, No.12 East Wenhui Road, Yangzhou, Jiangsu 225009, PR China.
| | - Nana Tian
- Ministry of Education Key Lab for Avian Preventive Medicine, College of Veterinary Medicine, Yangzhou University, No.12 East Wenhui Road, Yangzhou, Jiangsu 225009, PR China.
| | - Yupeng Liu
- Ministry of Education Key Lab for Avian Preventive Medicine, College of Veterinary Medicine, Yangzhou University, No.12 East Wenhui Road, Yangzhou, Jiangsu 225009, PR China.
| | - Kun Qian
- Ministry of Education Key Lab for Avian Preventive Medicine, College of Veterinary Medicine, Yangzhou University, No.12 East Wenhui Road, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, No.12 East Wenhui Road, Yangzhou, Jiangsu 225009, PR China.
| | - Hongxia Shao
- Ministry of Education Key Lab for Avian Preventive Medicine, College of Veterinary Medicine, Yangzhou University, No.12 East Wenhui Road, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, No.12 East Wenhui Road, Yangzhou, Jiangsu 225009, PR China.
| | - Jianqiang Ye
- Ministry of Education Key Lab for Avian Preventive Medicine, College of Veterinary Medicine, Yangzhou University, No.12 East Wenhui Road, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, No.12 East Wenhui Road, Yangzhou, Jiangsu 225009, PR China.
| | - Aijian Qin
- Ministry of Education Key Lab for Avian Preventive Medicine, College of Veterinary Medicine, Yangzhou University, No.12 East Wenhui Road, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, No.12 East Wenhui Road, Yangzhou, Jiangsu 225009, PR China.
| |
Collapse
|
2
|
Chen Y, Zhou T, Zhou R, Sun W, Li Y, Zhou Q, Xu D, Zhao Y, Hu P, Liang J, Zhang Y, Zhong B, Yao J, Jing D. TRAF7 knockdown induces cellular senescence and synergizes with lomustine to inhibit glioma progression and recurrence. J Exp Clin Cancer Res 2025; 44:112. [PMID: 40181456 PMCID: PMC11969748 DOI: 10.1186/s13046-025-03363-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 03/08/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND The progression and recurrence are the fatal prognostic factors in glioma patients. However, the therapeutic role and potential mechanism of TRAF7 in glioma patients remain largely unknown. METHODS TRAF7 RNA-seq was analysed with the TCGA and CGGA databases between glioma tissues and normal brain tissues. The expression of TRAF7, cellular senescence and cell cycle arrest pathways in glioma tissues and cell lines was detected by real-time quantitative PCR (RT-qPCR), western blotting and immunohistochemistry. The interaction between TRAF7 and KLF4 was determined by Co-immunoprecipitation (Co-IP) assays. The functions of TRAF7 combined with lomustine in glioma were assessed by both in vitro, in vivo and patient-derived primary and recurrent glioma stem cell (GSC) assays. RESULTS High TRAF7 expression is closely associated with a higher recurrence rate and poorer overall survival (OS). In vitro, TRAF7 knockdown significantly inhibits glioma cell proliferation, invasion, and migration. RNA-seq analysis revealed that TRAF7 inhibition activates pathways related to cellular senescence and cell cycle arrest. In both in vitro and patient-derived GSC assays, the combination of sh-TRAF7 and lomustine enhanced therapeutic efficacy by inducing senescence and G0/G1 cell cycle arrest, surpassing the effects of lomustine or TRAF7 inhibition alone. Mechanistically, TRAF7 interacts with KLF4, and a rescue assay demonstrated that KLF4 overexpression could reverse the effects of TRAF7 depletion on proliferation and cellular senescence. In vivo, TRAF7 knockdown combined with lomustine treatment effectively suppressed glioma growth. CONCLUSION TRAF7 could be used as a predictive biomarker and the potential therapeutic target among National Comprehensive Cancer Network (NCCN) treatment guidelines in the progression and recurrence of glioma. Lomustine, regulating cellular senescence and cell cycle could be the priority choice in glioma patients with high-level TRAF7 expression.
Collapse
Affiliation(s)
- Yu Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Animal Biological Products & Genetic Engineering, Ministry of Agriculture and Rural, Sinopharm Animal Health Corporation Ltd, Wuhan, 430023, China
- State Key Laboratory of Novel Vaccines for Emerging Infectious Diseases, China National Biotec Group Company Limited, Beijing, 100024, China
| | - Tongyu Zhou
- Department of Global Health and Social Medicine, King's College London, London, UK
| | - Rongrong Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wen Sun
- Key Laboratory of Animal Biological Products & Genetic Engineering, Ministry of Agriculture and Rural, Sinopharm Animal Health Corporation Ltd, Wuhan, 430023, China
- State Key Laboratory of Novel Vaccines for Emerging Infectious Diseases, China National Biotec Group Company Limited, Beijing, 100024, China
| | - Yan Li
- Key Laboratory of Animal Biological Products & Genetic Engineering, Ministry of Agriculture and Rural, Sinopharm Animal Health Corporation Ltd, Wuhan, 430023, China
- State Key Laboratory of Novel Vaccines for Emerging Infectious Diseases, China National Biotec Group Company Limited, Beijing, 100024, China
| | - Qiyi Zhou
- Center of PRaG Therapy, Center for Cancer Diagnosis and Treatment, Laboratory of Cancer Radioimmunotherapy, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Dongcheng Xu
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yuxin Zhao
- Key Laboratory of Animal Biological Products & Genetic Engineering, Ministry of Agriculture and Rural, Sinopharm Animal Health Corporation Ltd, Wuhan, 430023, China
- State Key Laboratory of Novel Vaccines for Emerging Infectious Diseases, China National Biotec Group Company Limited, Beijing, 100024, China
| | - Peihao Hu
- Key Laboratory of Animal Biological Products & Genetic Engineering, Ministry of Agriculture and Rural, Sinopharm Animal Health Corporation Ltd, Wuhan, 430023, China
- State Key Laboratory of Novel Vaccines for Emerging Infectious Diseases, China National Biotec Group Company Limited, Beijing, 100024, China
| | - Jingrui Liang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yumeng Zhang
- Key Laboratory of Animal Biological Products & Genetic Engineering, Ministry of Agriculture and Rural, Sinopharm Animal Health Corporation Ltd, Wuhan, 430023, China
- State Key Laboratory of Novel Vaccines for Emerging Infectious Diseases, China National Biotec Group Company Limited, Beijing, 100024, China
| | - Bin Zhong
- Department of Neurosurgery, Hunan University of Chinese Medicine Affiliated Yueyang Hospital, Yueyang, 414000, China
| | - Juncheng Yao
- Dalian Medical University, Dalian, 116041, China
| | - Di Jing
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
3
|
Han XF, Che MY, Su X, Tian JY, Liu LH, Nie P, Wang S. Molecular characterization of TRAF gene family in snakehead (Channa argus). FISH & SHELLFISH IMMUNOLOGY 2025; 158:110135. [PMID: 39837401 DOI: 10.1016/j.fsi.2025.110135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/16/2024] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
Tumor necrosis factor receptor-associated factors (TRAFs) act as signal transducers and are critical in many biological processes. However, in contrast to mammals, the function of TRAFs in teleost is still largely unknown. In this study, we identified and cloned eight TRAF genes in snakehead (Channa argus), namely CaTRAF2aa, CaTRAF2ab, CaTRAF2b, CaTRAF3, CaTRAF4a, CaTRAF5, CaTRAF6, and CaTRAF7. Bioinformatics analyses exhibited CaTRAF genes were evolutionarily conserved among teleost. Subcellular localization results demonstrated that eight CaTRAFs were all localized in cytoplasm. These CaTRAFs showed widespread but different expression profiles in various organs/tissues of snakehead and their expression could be induced by IHSV infection. Furthermore, almost all CaTRAFs can be impacted by poly (I:C) stimulation in HKLs. To sum up, this study provides a valuable foundation for further functional research on teleost TRAF genes.
Collapse
Affiliation(s)
- Xiu Feng Han
- School of Marine Science and Engineering, Qingdao Agricultural University, 700 Greatwall Road, Qingdao, Shandong Province, 266109, China
| | - Ming Yue Che
- School of Marine Science and Engineering, Qingdao Agricultural University, 700 Greatwall Road, Qingdao, Shandong Province, 266109, China
| | - Xue Su
- School of Marine Science and Engineering, Qingdao Agricultural University, 700 Greatwall Road, Qingdao, Shandong Province, 266109, China
| | - Jing Yun Tian
- Marine Science Research Institute of Shandong Province (National Oceanographic Center, Qingdao), Qingdao, Shandong Province, 266104, China
| | - Lan Hao Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, 700 Greatwall Road, Qingdao, Shandong Province, 266109, China
| | - Pin Nie
- School of Marine Science and Engineering, Qingdao Agricultural University, 700 Greatwall Road, Qingdao, Shandong Province, 266109, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, Shandong Province, 266237, China.
| | - Su Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, 700 Greatwall Road, Qingdao, Shandong Province, 266109, China.
| |
Collapse
|
4
|
Ammara M, Samiry I, Zaid Y, Oudghiri M, Naya A. A Pilot Study on the Role of TRAFs in the Development of SARS-CoV-2 Infection Before and After Immunization with AstraZeneca Chadox1 in Mice. Curr Issues Mol Biol 2025; 47:165. [PMID: 40136419 PMCID: PMC11941553 DOI: 10.3390/cimb47030165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025] Open
Abstract
The TRAF family of molecules are intracellular signaling adaptors that regulate various signaling pathways. These pathways are not only mediated by the TNFR superfamily and the Toll-like receptor/IL-1 receptor superfamily but also by unconventional cytokine receptors like IL-6 and IL-17 receptors. Overactive immune responses caused by TRAF signaling following the activation of these receptors frequently result in inflammatory and autoimmune diseases such as rheumatoid arthritis, inflammatory bowel disease, psoriasis, and autoinflammatory syndromes. Therefore, it is crucial to comprehend the signaling processes controlled by TRAFs, which have a significant influence on the determination of cell fate (life or death) and the functioning, specialization, and endurance of cells in the innate and adaptive immune systems. Our data indicate that the dysregulation of cellular expression and/or signaling of TRAFs leads to the excessive production of pro-inflammatory cytokines, hence promoting abnormal activation of immune cells. The objective of our investigation was to comprehend the function of these molecules in SARS-CoV-2 infection both prior to and during SARS-CoV-2 vaccination. Our results demonstrate a clear inactivation of the TRAF5 and TRAF6 genes when infection occurs after immunization, in contrast to infection without prior vaccination. This can bolster the belief that immunization is essential while also demonstrating the involvement of these molecules in the pathogenesis of SARS-CoV-2.
Collapse
Affiliation(s)
- Mounia Ammara
- Immunology and Biodiversity Laboratory, Department of Biology, Faculty of Sciences Ain Chock, Hassan II University, Casablanca 20000, Morocco; (M.A.); (I.S.); (Y.Z.); (M.O.)
| | - Inass Samiry
- Immunology and Biodiversity Laboratory, Department of Biology, Faculty of Sciences Ain Chock, Hassan II University, Casablanca 20000, Morocco; (M.A.); (I.S.); (Y.Z.); (M.O.)
| | - Younes Zaid
- Immunology and Biodiversity Laboratory, Department of Biology, Faculty of Sciences Ain Chock, Hassan II University, Casablanca 20000, Morocco; (M.A.); (I.S.); (Y.Z.); (M.O.)
- Materials, Nanotechnologies and Environment Laboratory, Department of Biology, Faculty of Sciences, Mohammed V University, Rabat 10000, Morocco
| | - Mounia Oudghiri
- Immunology and Biodiversity Laboratory, Department of Biology, Faculty of Sciences Ain Chock, Hassan II University, Casablanca 20000, Morocco; (M.A.); (I.S.); (Y.Z.); (M.O.)
| | - Abdallah Naya
- Immunology and Biodiversity Laboratory, Department of Biology, Faculty of Sciences Ain Chock, Hassan II University, Casablanca 20000, Morocco; (M.A.); (I.S.); (Y.Z.); (M.O.)
| |
Collapse
|
5
|
Zhang X, Zhang Z, Zheng Z, Yao D, Zhao Y, Liu Q, Lin Z, Zhang Y. Ubiquitination of Hemocyanin Mediated by a Mitochondrial E3 Ubiquitin Ligase Regulates Immune Response in Penaeus vannamei. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1746-1759. [PMID: 39513673 DOI: 10.4049/jimmunol.2400493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 09/24/2024] [Indexed: 11/15/2024]
Abstract
Ubiquitination is a critical posttranslational modification that regulates host immune responses to pathogens. In this study, we investigated the ubiquitination of hemocyanin (PvHMC [Penaeus vannamei hemocyanin]) mediated by the mitochondrial E3 ubiquitin ligase (PvMulan) in shrimp Penaeus vannamei. We characterized distinct ubiquitination patterns of PvHMC in response to different pathogen challenges, both in vitro and in vivo. Specifically, we found that Vibrio parahaemolyticus infection led to an increase in PvMulan, which resulted in K48-linked ubiquitination and subsequent proteasomal degradation of PvHMC. In contrast, PvMulan primarily enhanced the SUMOylation of PvHMC, bolstering its immune functions against white spot syndrome virus challenges. Inhibition of PvMulan-mediated PvHMC ubiquitination significantly affected the proliferation of V. parahaemolyticus and the survival rate of infected shrimps. This study sheds light on the role of hemocyanin ubiquitination in immune regulation, illustrating its dual function in response to distinct pathogens.
Collapse
Affiliation(s)
- Xiaojun Zhang
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou, China
| | - Zhaoxue Zhang
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou, China
| | - Zhihong Zheng
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou, China
| | - Defu Yao
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou, China
| | - Yongzhen Zhao
- Guangxi Academy of Fishery Sciences, Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Nanning, China
| | - Qingyun Liu
- Guangxi Academy of Fishery Sciences, Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Nanning, China
| | - Zhongyang Lin
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou, China
| | - Yueling Zhang
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou, China
| |
Collapse
|
6
|
Herrera CM, McMahon E, Swaney DL, Sherry J, Pha K, Adams-Boone K, Johnson JR, Krogan NJ, Stevers M, Solomon D, Elwell C, Engel J. The Chlamydia trachomatis Inc Tri1 interacts with TRAF7 to displace native TRAF7 interacting partners. Microbiol Spectr 2024; 12:e0045324. [PMID: 38814079 PMCID: PMC11218536 DOI: 10.1128/spectrum.00453-24] [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: 02/26/2024] [Accepted: 04/23/2024] [Indexed: 05/31/2024] Open
Abstract
Chlamydia trachomatis is the leading cause of bacterial sexually transmitted infections in the USA and of preventable blindness worldwide. This obligate intracellular pathogen replicates within a membrane-bound inclusion, but how it acquires nutrients from the host while avoiding detection by the innate immune system is incompletely understood. C. trachomatis accomplishes this in part through the translocation of a unique set of effectors into the inclusion membrane, the inclusion membrane proteins (Incs). Incs are ideally positioned at the host-pathogen interface to reprogram host signaling by redirecting proteins or organelles to the inclusion. Using a combination of co-affinity purification, immunofluorescence confocal imaging, and proteomics, we characterize the interaction between an early-expressed Inc of unknown function, Tri1, and tumor necrosis factor receptor-associated factor 7 (TRAF7). TRAF7 is a multi-domain protein with a RING finger ubiquitin ligase domain and a C-terminal WD40 domain. TRAF7 regulates several innate immune signaling pathways associated with C. trachomatis infection and is mutated in a subset of tumors. We demonstrate that Tri1 and TRAF7 specifically interact during infection and that TRAF7 is recruited to the inclusion. We further show that the predicted coiled-coil domain of Tri1 is necessary to interact with the TRAF7 WD40 domain. Finally, we demonstrate that Tri1 displaces the native TRAF7 binding partners, mitogen-activated protein kinase kinase kinase 2 (MEKK2), and MEKK3. Together, our results suggest that by displacing TRAF7 native binding partners, Tri1 has the capacity to alter TRAF7 signaling during C. trachomatis infection.IMPORTANCEChlamydia trachomatis is the leading cause of bacterial sexually transmitted infections in the USA and preventable blindness worldwide. Although easily treated with antibiotics, the vast majority of infections are asymptomatic and therefore go untreated, leading to infertility and blindness. This obligate intracellular pathogen evades the immune response, which contributes to these outcomes. Here, we characterize the interaction between a C. trachomatis-secreted effector, Tri1, and a host protein involved in innate immune signaling, TRAF7. We identified host proteins that bind to TRAF7 and demonstrated that Tri1 can displace these proteins upon binding to TRAF7. Remarkably, the region of TRAF7 to which these host proteins bind is often mutated in a subset of human tumors. Our work suggests a mechanism by which Tri1 may alter TRAF7 signaling and has implications not only in the pathogenesis of C. trachomatis infections but also in understanding the role of TRAF7 in cancer.
Collapse
Affiliation(s)
- Clara M. Herrera
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Eleanor McMahon
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Danielle L. Swaney
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California, USA
| | - Jessica Sherry
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Khavong Pha
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Kathleen Adams-Boone
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jeffrey R. Johnson
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California, USA
| | - Nevan J. Krogan
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California, USA
| | - Meredith Stevers
- Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - David Solomon
- Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - Cherilyn Elwell
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Joanne Engel
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
7
|
Wang L, Yang F, Ye J, Zhang L, Jiang X. Insight into the role of IRF7 in skin and connective tissue diseases. Exp Dermatol 2024; 33:e15083. [PMID: 38794808 DOI: 10.1111/exd.15083] [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: 01/06/2024] [Revised: 03/15/2024] [Accepted: 04/08/2024] [Indexed: 05/26/2024]
Abstract
Interferons (IFNs) are signalling proteins primarily involved in initiating innate immune responses against pathogens and promoting the maturation of immune cells. Interferon Regulatory Factor 7 (IRF7) plays a pivotal role in the IFNs signalling pathway. The activation process of IRF7 is incited by exogenous or abnormal nucleic acids, which is followed by the identification via pattern recognition receptors (PRRs) and the ensuing signalling cascades. Upon activation, IRF7 modulates the expression of both IFNs and inflammatory gene regulation. As a multifunctional transcription factor, IRF7 is mainly expressed in immune cells, yet its presence is also detected in keratinocytes, fibroblasts, and various dermal cell types. In these cells, IRF7 is critical for skin immunity, inflammation, and fibrosis. IRF7 dysregulation may lead to autoimmune and inflammatory skin conditions, including systemic scleroderma (SSc), systemic lupus erythematosus (SLE), Atopic dermatitis (AD) and Psoriasis. This comprehensive review aims to extensively elucidate the role of IRF7 and its signalling pathways in immune cells and keratinocytes, highlighting its significance in skin-related and connective tissue diseases.
Collapse
Affiliation(s)
- Lian Wang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Fengjuan Yang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Ye
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Zhang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xian Jiang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
8
|
Herrera CM, McMahon E, Swaney DL, Sherry J, Pha K, Adams-Boone K, Johnson JR, Krogan NJ, Stevers M, Solomon D, Elwell C, Engel J. The Chlamydia trachomatis Inc Tri1 interacts with TRAF7 to displace native TRAF7 interacting partners. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.581999. [PMID: 38464023 PMCID: PMC10925117 DOI: 10.1101/2024.02.26.581999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Chlamydia trachomatis is the leading cause of bacterial sexually transmitted infections in the US and of preventable blindness worldwide. This obligate intracellular pathogen replicates within a membrane-bound inclusion, but how it acquires nutrients from the host while avoiding detection by the innate immune system is incompletely understood. C. trachomatis accomplishes this in part through the translocation of a unique set of effectors into the inclusion membrane, the inc lusion membrane proteins (Incs). Incs are ideally positioned at the host-pathogen interface to reprogram host signaling by redirecting proteins or organelles to the inclusion. Using a combination of co-affinity purification, immunofluorescence confocal imaging, and proteomics, we characterize the interaction between an early-expressed Inc of unknown function, Tri1, and tumor necrosis factor receptor associated factor 7 (TRAF7). TRAF7 is a multi-domain protein with a RING finger ubiquitin ligase domain and a C-terminal WD40 domain. TRAF7 regulates several innate immune signaling pathways associated with C. trachomatis infection and is mutated in a subset of tumors. We demonstrate that Tri1 and TRAF7 specifically interact during infection and that TRAF7 is recruited to the inclusion. We further show that the predicted coiled-coil domain of Tri1 is necessary to interact with the TRAF7 WD40 domain. Finally, we demonstrate that Tri1 displaces the native TRAF7 binding partners, mitogen activated protein kinase kinase kinase 2 (MEKK2) and MEKK3. Together, our results suggest that by displacing TRAF7 native binding partners, Tri1 has the capacity to alter TRAF7 signaling during C. trachomatis infection. Importance Chlamydia trachomatis is the leading cause of bacterial sexually transmitted infections in the US and preventable blindness worldwide. Although easily treated with antibiotics, the vast majority of infections are asymptomatic and therefore go untreated, leading to infertility and blindness. This obligate intracellular pathogen evades the immune response, which contributes to these outcomes. Here, we characterize the interaction between a C. trachomatis secreted effector, Tri1, and a host protein involved in innate immune signaling, TRAF7. We identified host proteins that bind to TRAF7 and demonstrate that Tri1 can displace these proteins upon binding to TRAF7. Remarkably, the region of TRAF7 to which these host proteins bind is often mutated in a subset of human tumors. Our work suggests a mechanism by which Tri1 may alter TRAF7 signaling and has implications not only in the pathogenesis of C. trachomatis infections, but also in understanding the role of TRAF7 in cancer.
Collapse
|