1
|
Qian G, Wang Y, Yao H, Zhang Z, Wang W, Xu L, Li W, Huang L, Li X, Gao Y, Wang N, Wang S, Pan J, Lv H. Involvement of USP7 in aggravating Kawasaki disease by promoting TGFβ2 signaling mediated endothelial-mesenchymal transition and coronary artery remodeling. Int Immunopharmacol 2025; 146:113823. [PMID: 39674001 PMCID: PMC11799894 DOI: 10.1016/j.intimp.2024.113823] [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: 11/05/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/16/2024]
Abstract
Kawasaki disease (KD), characterized by systematic vasculitis, is a leading cause of pediatric heart disease. Although recent studies have highlighted the critical role of deubiquitinases in vascular pathophysiology, their specific contribution to KD remains largely unknown. Herein, we investigated the function of the deubiquitinase USP7 in both KD patients and a CAWS-induced KD murine model. USP7 expression level is increased both in HCAECs induced by KD sera and cardiac CD31+ endothelial cells of KD mice. Whereas knockout of USP7 increases the cellular proportion of endothelial cells and potentially attenuates the elevated EndoMT, fibrosis, and inflammation in cardiac tissue of KD mice, consistently with the in vitro experiment observed in HCAECs induced by TGF-β2. Mechanistically, USP7 interacts with SMAD2/3, enhancing their protein stability by removing the K48 ubiquitin chain from both proteins and preventing their proteasome degradation, thus increasing the p-SMAD2 levels and nuclear entry. Importantly, intraperitoneal injection of USP7 inhibitor, P22077 elicited a robust anti-EndoMT and anti-vascular inflammation effect in KD model mice. Therefore, our study uncovered a previously unrecognized function of increased USP7 in KD by augmenting TGFβ2/SMAD2/SMAD3 signaling, thus facilitating the transcription of genes implicated in the EndoMT, cardiac fibrosis, and vascular remodeling. Our finding suggests that USP7 could serve as a potential therapeutic target for the prevention and treatment of coronary artery lesions in KD and related vascular diseases.
Collapse
Affiliation(s)
- Guanghui Qian
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| | - Yan Wang
- Department of Cardiology, The Affiliated Xuzhou Children's Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province 221000, China.
| | - Hongwei Yao
- Providence VA Medical Center, Providence, RI, USA; Departments of Medicine, Molecular Biology, Cell Biology & Biochemistry, Brown University, Providence, RI, USA.
| | - Zimu Zhang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| | - Wang Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China
| | - Lei Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| | - Wenjie Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| | - Li Huang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| | - Xuan Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| | - Yang Gao
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China
| | - Nana Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| | - Shuhui Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| | - Haitao Lv
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025 Jiangsu, China.
| |
Collapse
|
2
|
Liu Y, Pei S, Wang X, Li X, Long Y, Sun S, Meng C, Feng F. Effect of the Deubiquitinating Peptidase 7 (USP7) on Hepatitis B Virus (HBV) Replication and the Antiviral Efficacy of Entecavir (ETV). Mol Biotechnol 2024:10.1007/s12033-024-01355-8. [PMID: 39715932 DOI: 10.1007/s12033-024-01355-8] [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: 03/19/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024]
Abstract
Hepatitis B is a viral infection of the liver caused by the hepatitis B virus (HBV). Entecavir (ETV) is considered the primary therapeutic option for HBV treatment, primarily functioning by inhibiting HBV replication. Ubiquitin-specific peptidase 7 (USP7), a deubiquitinating enzyme, plays a crucial role in regulating DNA repair mechanisms. This article aims to investigate the role of USP7 in HBV replication and its potential to enhance the antiviral efficacy of ETV, while exploring the underlying mechanisms involved. HBV infection is closely associated with the development of liver cancer. In this study, we selected the HepG2.2.15 cell line, which was stably HepG2 cell transfected with two complete HBV genomes. HepG2.2.15 supports HBV replication, assembly, and secretion. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot (WB) assays were subsequently employed to measure USP7 mRNA and protein levels in both cell lines. The USP7 gene was silenced using small interfering RNA (siRNA), cells were transfected with siRNA-USP7 using Lipo6000™ Transfection Reagent, after which we assessed HBV replication, the levels of HBsAg, and HBeAg following 24, 48, and 72 h of culture in HepG2.2.15 cells. Afterwards, HepG2.2.15 cells were divided into several groups: control, USP7 gene silencing by siRNA group (siRNA-USP7), USP7 silencing negative control group (siRNA-NC), ETV drug treatment (ETV), ETV drug treatment combined with USP7 gene silencing by siRNA group (ETV + siRNA-USP7), and ETV therapy alongside a negative control for siRNA silencing (ETV + siRNA-NC). HBV replication, the levels of HBsAg, and HBeAg in the cell supernatant were assessed after 24, 48, and 72 h of culture. Additionally, alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels were measured to evaluate cellular damage. Furthermore, qRT-PCR and Western blot techniques were utilized to analyze p53 mRNA and protein levels as potential downstream mechanisms of USP7, along with assessing Bax and Bcl-2 mRNA and protein levels within the p53 signaling pathway. Lastly, we investigated the interaction between USP7 and p53 proteins through co-immunoprecipitation. USP7 protein and mRNA levels were up-regulated in the HepG2.2.15 cell line, and silencing of USP7 inhibited HBV replication. More importantly, HBV replication, HBsAg, and HBeAg levels in the ETV + siRNA-USP7 group were significantly reduced compared to the other groups (P < 0.05), indicating that silencing USP7 enhances the antiviral effect of ETV. Additionally, ALT and AST levels were significantly decreased (P < 0.05), suggesting a reduction in cellular damage. Furthermore, an interaction between USP7 and p53 was observed. Both mRNA and protein levels of p53, as well as its downstream factors Bax and Bcl-2 in the ETV + siRNA-USP7 group, were significantly down-regulated (P < 0.05), implying that USP7 is involved in regulating the p53 pathway. Decreasing of deubiquitinating peptidase 7 expression in a human hepatoma model enhanced antiviral effect of entecavir and reduced cellular damage caused by the hepatitis B virus.
Collapse
Affiliation(s)
- Yue Liu
- School of Public Health, North China University of Science of Technology, Tangshan, 062310, Hebei, China
| | - Shengfei Pei
- School of Public Health, North China University of Science of Technology, Tangshan, 062310, Hebei, China
| | - Xue Wang
- School of Public Health, North China University of Science of Technology, Tangshan, 062310, Hebei, China
| | - Xueying Li
- School of Public Health, North China University of Science of Technology, Tangshan, 062310, Hebei, China
| | - Yifei Long
- School of Public Health, North China University of Science of Technology, Tangshan, 062310, Hebei, China
| | - Shufeng Sun
- College of Nursing and Rehabilitation, North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Chunyan Meng
- School of Public Health, North China University of Science of Technology, Tangshan, 062310, Hebei, China.
- Hebei Coordinated Innovation Center of Occupational Health and SafetySchool of Public Health, North China University of Science and Technology, Tangshan, 063210, China.
| | - Fumin Feng
- School of Public Health, North China University of Science of Technology, Tangshan, 062310, Hebei, China.
- Hebei Coordinated Innovation Center of Occupational Health and SafetySchool of Public Health, North China University of Science and Technology, Tangshan, 063210, China.
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, Tangshan, China.
| |
Collapse
|
3
|
Li Y, Liu X, Li Y, Wang J, Zhang M, Xue W, Zhang M. USP19 exerts a tumor-promoting role in diffuse large B cell lymphoma through stabilizing PARK7. FEBS J 2024; 291:4757-4774. [PMID: 39240655 DOI: 10.1111/febs.17259] [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: 11/02/2023] [Revised: 03/29/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of non-Hodgkin lymphoma and is associated with a poor prognosis. Data from the Gene Expression Profiling Interactive Analysis (GEPIA) database revealed dysregulated expression of several ubiquitin-specific proteases (USPs) in DLBCL tissues (DLBCL vs. non-DLBCL = 47 vs. 337), including USP19 (log2fold change = 1.17, P < 0.05). USP19 is closely linked to tumorigenesis, but its role in DLBCL progression remains largely unknown. Here, we investigated the role of USP19 in DLBCL development. Genetic manipulation of USP19 using adenovirus-based vectors was performed in two DLBCL cell lines, SUDHL4 and DB cells. The results showed that USP19 knockdown suppressed the proliferation, anchorage-independent growth and xenograft tumor formation of DLBCL cells and arrested the cell cycle at the G1 stage. In parallel, DLBCL cells overexpressing USP19 acquired a more malignant phenotype. Next, to explore USP19 interactors, we performed co-immunoprecipitation/liquid chromatography-mass spectrometry and identified potential interacting proteins. Among them, Parkinson disease protein 7 (PARK7), a member of the peptidase C56 family known to be involved in carcinogenesis, was further validated to bind with and be stabilized by USP19. Additionally, we found that USP19 induced PARK7 deubiquitylation in both DLBCL cell lines, and PARK7 acted as a downstream effector of USP19 in regulating the growth of DLBCL cells. Collectively, USP19 exerts a tumor-promoting role in DLBCL through interacting with and stabilizing PARK7.
Collapse
Affiliation(s)
- Yaqing Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, China
| | - Xiyang Liu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, China
| | - Yulai Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, China
| | - Jieting Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, China
| | - Mengqian Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, China
| | - Weili Xue
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, China
| |
Collapse
|
4
|
Sreepangi S, Baha H, Opoku LA, Jones NX, Konadu M, Alem F, Barrera MD, Narayanan A. Host-Driven Ubiquitination Events in Vector-Transmitted RNA Virus Infections as Options for Broad-Spectrum Therapeutic Intervention Strategies. Viruses 2024; 16:1727. [PMID: 39599842 PMCID: PMC11599102 DOI: 10.3390/v16111727] [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: 06/11/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
Many vector-borne viruses are re-emerging as public health threats, yet our understanding of the virus-host interactions critical for productive infection remains limited. The ubiquitination of proteins, including host- and pathogen-derived proteins is a highly prominent and consistent post-translational modification that regulates protein function through signaling and degradation. Viral proteins are documented to hijack the host ubiquitination machinery to modulate multiple host processes including antiviral defense mechanisms. The engagement of the host ubiquitination machinery in the post-translational modification of viral proteins to support aspects of the viral life cycle including assembly and egress is also well documented. Exploring the role ubiquitination plays in the life cycle of vector-transmitted viral pathogens will increase the knowledge base pertinent to the impact of host-enabled ubiquitination of viral and host proteins and the consequences on viral pathogenesis. In this review, we explore E3 ligase-regulated ubiquitination pathways functioning as proviral and viral restriction factors in the context of acutely infectious, vector-transmitted viral pathogens and the potential for therapeutically targeting them for countermeasures development.
Collapse
Affiliation(s)
- Sanskruthi Sreepangi
- School of Systems Biology, College of Science, George Mason University, Fairfax, VA 22030, USA; (S.S.); (H.B.); (L.A.O.); (N.X.J.); (M.K.); (M.D.B.)
| | - Haseebullah Baha
- School of Systems Biology, College of Science, George Mason University, Fairfax, VA 22030, USA; (S.S.); (H.B.); (L.A.O.); (N.X.J.); (M.K.); (M.D.B.)
| | - Lorreta Aboagyewa Opoku
- School of Systems Biology, College of Science, George Mason University, Fairfax, VA 22030, USA; (S.S.); (H.B.); (L.A.O.); (N.X.J.); (M.K.); (M.D.B.)
| | - Naomi X. Jones
- School of Systems Biology, College of Science, George Mason University, Fairfax, VA 22030, USA; (S.S.); (H.B.); (L.A.O.); (N.X.J.); (M.K.); (M.D.B.)
| | - Maame Konadu
- School of Systems Biology, College of Science, George Mason University, Fairfax, VA 22030, USA; (S.S.); (H.B.); (L.A.O.); (N.X.J.); (M.K.); (M.D.B.)
| | - Farhang Alem
- Institute of Biohealth Innovation, George Mason University, Fairfax, VA 22030, USA;
| | - Michael D. Barrera
- School of Systems Biology, College of Science, George Mason University, Fairfax, VA 22030, USA; (S.S.); (H.B.); (L.A.O.); (N.X.J.); (M.K.); (M.D.B.)
| | - Aarthi Narayanan
- Department of Biology, College of Science, George Mason University, Fairfax, VA 22030, USA
| |
Collapse
|
5
|
Tarasova O, Petrou A, Ivanov SM, Geronikaki A, Poroikov V. Viral Factors in Modulation of Host Immune Response: A Route to Novel Antiviral Agents and New Therapeutic Approaches. Int J Mol Sci 2024; 25:9408. [PMID: 39273355 PMCID: PMC11395507 DOI: 10.3390/ijms25179408] [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: 07/22/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Viruses utilize host cells at all stages of their life cycle, from the transcription of genes and translation of viral proteins to the release of viral copies. The human immune system counteracts viruses through a variety of complex mechanisms, including both innate and adaptive components. Viruses have an ability to evade different components of the immune system and affect them, leading to disruption. This review covers contemporary knowledge about the virus-induced complex interplay of molecular interactions, including regulation of transcription and translation in host cells resulting in the modulation of immune system functions. Thorough investigation of molecular mechanisms and signaling pathways that are involved in modulating of host immune response to viral infections can help to develop novel approaches for antiviral therapy. In this review, we consider new therapeutic approaches for antiviral treatment. Modern therapeutic strategies for the treatment and cure of human immunodeficiency virus (HIV) are considered in detail because HIV is a unique example of a virus that leads to host T lymphocyte deregulation and significant modulation of the host immune response. Furthermore, peculiarities of some promising novel agents for the treatment of various viral infections are described.
Collapse
Affiliation(s)
- Olga Tarasova
- Institute of Biomedical Chemistry, Moscow 119121, Russia
| | - Anthi Petrou
- School of Pharmacy, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | | | - Athina Geronikaki
- School of Pharmacy, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | | |
Collapse
|
6
|
Miao Y, Zhang T, Guan M, Zhao Q, Zhang R, Liu X, Ma T, Ren T, Zheng Z, He W, Tian W, Cui Q, Zhai X, Zuo Y, Zhu H, Zheng H, Yuan Y. DDX4 enhances antiviral activity of type I interferon by disrupting interaction of USP7/SOCS1 and promoting degradation of SOCS1. mBio 2024; 15:e0321323. [PMID: 38376239 PMCID: PMC10936202 DOI: 10.1128/mbio.03213-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/29/2024] [Indexed: 02/21/2024] Open
Abstract
DEAD-box helicase (DDX) family members play differential roles in regulating innate antiviral immune response. However, the physiological roles played by DDX4 in antiviral innate immunity remain unclear. In this study, we unveiled that DDX4 acts as a positive regulatory molecule of Type-I interferon (IFN-I)-mediated antiviral activity. Our findings demonstrate that IFN-I upregulates DDX4 protein levels, and subsequently, overexpression of DDX4 enhances the IFN-I-mediated signaling pathway. This creates a positive feedback loop that amplifies the antiviral response. DDX4 was found to bind with deubiquitinase ubiquitin-specific protease 7 (USP7), leading to the disruption of the interaction between USP7 and suppressor of cytokine signaling 1 (SOCS1) and the subsequent degradation of SOCS1. This process enhances the antiviral function of IFN-I. Our findings provide new insights into the regulatory role of DDX4 in the IFN-I response.IMPORTANCEDDX4, identified as a putative RNA helicase that modulates RNA secondary structure through RNA binding, is primarily acknowledged for its role in regulating mRNA translation within the germline. Nevertheless, the extent of DDX4's involvement in the antiviral innate immune response remains largely unexplored. This study presents evidence of a previously unrecognized positive feedback loop between DDX4 and the antiviral response, suggesting that disruption of this loop may serve as a novel mechanism for viral evasion. Furthermore, our findings elucidate a positive regulatory mechanism by which the DDX4/USP7/SOCS1 axis mediates the antiviral activity of Type-I interferon, which provides new insight into strategies for improving the efficacy of IFN-based antiviral therapy.
Collapse
Affiliation(s)
- Ying Miao
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Tingting Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Mingcheng Guan
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, China
| | - Qian Zhao
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Renxia Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Xuyi Liu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Tianrun Ma
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Tengfei Ren
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Zhijin Zheng
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Wei He
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Wanying Tian
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Qun Cui
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Xingyu Zhai
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Yibo Zuo
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Hong Zhu
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, China
| | - Hui Zheng
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Department/Institute of Laboratory Medicine, Sichuan Provincial People’s Hospital, Medical School, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yukang Yuan
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
7
|
Zhao Q, Zhang R, Qiao C, Miao Y, Yuan Y, Zheng H. Ubiquitination network in the type I IFN-induced antiviral signaling pathway. Eur J Immunol 2023; 53:e2350384. [PMID: 37194705 DOI: 10.1002/eji.202350384] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/14/2023] [Accepted: 05/16/2023] [Indexed: 05/18/2023]
Abstract
Type I IFN (IFN-I) is the body's first line of defense against pathogen infection. IFN-I can induce cellular antiviral responses and therefore plays a key role in driving antiviral innate and adaptive immunity. Canonical IFN-I signaling activates the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway, which induces the expression of IFN-stimulated genes and eventually establishes a complex antiviral state in the cells. Ubiquitin is a ubiquitous cellular molecule for protein modifications, and the ubiquitination modifications of protein have been recognized as one of the key modifications that regulate protein levels and/or signaling activation. Despite great advances in understanding the ubiquitination regulation of many signaling pathways, the mechanisms by which protein ubiquitination regulates IFN-I-induced antiviral signaling have not been explored until very recently. This review details the current understanding of the regulatory network of ubiquitination that critically controls the IFN-I-induced antiviral signaling pathway from three main levels, including IFN-I receptors, IFN-I-induced cascade signals, and effector IFN-stimulated genes.
Collapse
Affiliation(s)
- Qian Zhao
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Renxia Zhang
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Caixia Qiao
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Ying Miao
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Yukang Yuan
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Hui Zheng
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| |
Collapse
|
8
|
Huang S, Cui M, Huang J, Wu Z, Cheng A, Wang M, Zhu D, Chen S, Liu M, Zhao X, Wu Y, Yang Q, Zhang S, Ou X, Mao S, Gao Q, Tian B, Sun D, Yin Z, Jing B, Jia R. RNF123 Mediates Ubiquitination and Degradation of SOCS1 To Regulate Type I Interferon Production during Duck Tembusu Virus Infection. J Virol 2023; 97:e0009523. [PMID: 37014223 PMCID: PMC10134884 DOI: 10.1128/jvi.00095-23] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/14/2023] [Indexed: 04/05/2023] Open
Abstract
Many RING domain E3 ubiquitin ligases play critical roles in fine-tuning the innate immune response, yet little is known about their regulatory role in flavivirus-induced innate immunity. In previous studies, we found that the suppressor of cytokine signaling 1 (SOCS1) protein mainly undergoes lysine 48 (K48)-linked ubiquitination. However, the E3 ubiquitin ligase that promotes the K48-linked ubiquitination of SOCS1 is unknown. In the present study, we found that RING finger protein 123 (RNF123) binds to the SH2 domain of SOCS1 through its RING domain and facilitates the K48-linked ubiquitination of the K114 and K137 residues of SOCS1. Further studies found that RNF123 promoted the proteasomal degradation of SOCS1 and promoted Toll-like receptor 3 (TLR3)- and interferon (IFN) regulatory factor 7 (IRF7)-mediated type I IFN production during duck Tembusu virus (DTMUV) infection through SOCS1, ultimately inhibiting DTMUV replication. Overall, these findings demonstrate a novel mechanism by which RNF123 regulates type I IFN signaling during DTMUV infection by targeting SOCS1 degradation. IMPORTANCE In recent years, posttranslational modification (PTM) has gradually become a research hot spot in the field of innate immunity regulation, and ubiquitination is one of the critical PTMs. DTMUV has seriously endangered the development of the waterfowl industry in Southeast Asian countries since its outbreak in 2009. Previous studies have shown that SOCS1 is modified by K48-linked ubiquitination during DTMUV infection, but E3 ubiquitin ligase catalyzing the ubiquitination of SOCS1 has not been reported. Here, we identify for the first time that RNF123 acts as an E3 ubiquitin ligase that regulates TLR3- and IRF7-induced type I IFN signaling during DTMUV infection by targeting the K48-linked ubiquitination of the K114 and K137 residues of SOCS1 and the proteasomal degradation of SOCS1.
Collapse
Affiliation(s)
- Shanzhi Huang
- Research Centre of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
| | - Min Cui
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| | - Juan Huang
- Research Centre of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| | - Ziyu Wu
- Research Centre of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
| | - Anchun Cheng
- Research Centre of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| | - Mingshu Wang
- Research Centre of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| | - Dekang Zhu
- Research Centre of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| | - Shun Chen
- Research Centre of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| | - Mafeng Liu
- Research Centre of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| | - Xinxin Zhao
- Research Centre of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| | - Ying Wu
- Research Centre of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| | - Qiao Yang
- Research Centre of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| | - Shaqiu Zhang
- Research Centre of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| | - Xumin Ou
- Research Centre of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| | - Sai Mao
- Research Centre of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| | - Qun Gao
- Research Centre of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| | - Bin Tian
- Research Centre of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| | - Di Sun
- Research Centre of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| | - Zhongqiong Yin
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| | - Bo Jing
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| | - Renyong Jia
- Research Centre of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, People’s Republic of China
| |
Collapse
|
9
|
Low ZY, Wen Yip AJ, Chow VTK, Lal SK. The Suppressor of Cytokine Signalling family of proteins and their potential impact on COVID-19 disease progression. Rev Med Virol 2022; 32:e2300. [PMID: 34546610 PMCID: PMC8646547 DOI: 10.1002/rmv.2300] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 12/16/2022]
Abstract
The family of Suppressor of Cytokine Signalling (SOCS) proteins plays pivotal roles in cytokine and immune regulation. Despite their key roles, little attention has been given to the SOCS family as compared to other feedback regulators. To date, SOCS proteins have been found to be exploited by viruses such as herpes simplex virus (HSV), hepatitis B virus (HBV), hepatitis C virus (HCV), Zika virus, respiratory syncytial virus (RSV), Ebola virus, influenza A virus (IAV) and SARS-CoV, just to name a few. The hijacking and subsequent upregulation of the SOCS proteins upon viral infection, suppress the associated JAK-STAT signalling activities, thereby reducing the host antiviral response and promoting viral replication. Two SOCS protein family members, SOCS1 and SOCS3 are well-studied and their roles in the JAK-STAT signalling pathway are defined as attenuating interferon (IFN) signalling upon viral infection. The upregulation of SOCS protein by SARS-CoV during the early stages of infection implies strong similarity with SARS-CoV-2, given their closely related genomic organisation. Thus, this review aims to outline the plausibility of SOCS protein inhibitors as a potential therapeutic regimen for COVID-19 patients. We also discuss the antagonists against SOCS protein to offer an overview on the previous 'successes' of SOCS protein inhibition in various viral infections that may portray possible clues for COVID-19 disease management.
Collapse
Affiliation(s)
- Zheng Yao Low
- School of ScienceMonash UniversityBandar SunwaySelangorMalaysia
| | | | - Vincent T. K. Chow
- Infectious Diseases Translational Research ProgramDepartment of Microbiology and ImmunologyYong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Sunil K. Lal
- School of ScienceMonash UniversityBandar SunwaySelangorMalaysia
- Tropical Medicine and Biology PlatformMonash UniversityBandar SunwaySelangorMalaysia
| |
Collapse
|
10
|
High Expression of Ubiquitin-Specific Protease 39 and Its Roles in Prognosis in Patients with Hepatocellular Carcinoma. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2021:6233175. [PMID: 34987596 PMCID: PMC8723855 DOI: 10.1155/2021/6233175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 12/07/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Ubiquitin-specific protease 39 is mainly involved in mRNA splicing and multiple kinds of tumors. Accumulating evidence has shown that USP39 participated in the proliferation and metastasis of hepatocellular carcinoma (HCC). The present study aimed to demonstrate the association between USP39 expression and clinical features and the diagnostic value in HCC based on the Cancer Genome Atlas (TCGA). METHODS A comprehensive analysis for expression of USP39 in HCC was conducted by using multiple databases. The mRNA level of USP39, clinical features, survival rate, and diagnostic value in HCC were analyzed using data from TCGA. The Gene Set Enrichment Analysis (GSEA) was conducted to analyze signaling pathways correlated with USP39 expression in HCC. RESULTS The mRNA level of USP39 was significantly elevated in HCC. The expression of USP39 showed significant correlation with T stage, pathologic stage, tumor status, age, and histologic grade. Logistic analysis demonstrated that high expression of USP39 was significantly associated with older age, tumor status, advanced pathologic stage, T stage, and higher histologic grade. Univariate Cox regression analysis showed that high expression of USP39 was significantly associated with advanced T stage, pathological stage, and tumor status. Multivariate Cox analysis confirmed the result that USP39 expression was an independent prognostic factor for overall survival (OS) in HCC. Results of Kaplan-Meier curves showed that high expression of USP39 had a significant association with poor OS, disease-free survival (DSS), and progress-free interval (PFI) in HCC. ROC analysis indicated that USP39 could be regarded as a promising marker for distinguishing HCC from nontumor. CONCLUSION The increased USP39 might play roles in the progression, diagnosis, and prognosis of HCC.
Collapse
|
11
|
Qian G, Zhu L, Li G, Liu Y, Zhang Z, Pan J, Lv H. An Integrated View of Deubiquitinating Enzymes Involved in Type I Interferon Signaling, Host Defense and Antiviral Activities. Front Immunol 2021; 12:742542. [PMID: 34707613 PMCID: PMC8542838 DOI: 10.3389/fimmu.2021.742542] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/16/2021] [Indexed: 12/24/2022] Open
Abstract
Viral infectious diseases pose a great challenge to human health around the world. Type I interferons (IFN-Is) function as the first line of host defense and thus play critical roles during virus infection by mediating the transcriptional induction of hundreds of genes. Nevertheless, overactive cytokine immune responses also cause autoimmune diseases, and thus, tight regulation of the innate immune response is needed to achieve viral clearance without causing excessive immune responses. Emerging studies have recently uncovered that the ubiquitin system, particularly deubiquitinating enzymes (DUBs), plays a critical role in regulating innate immune responses. In this review, we highlight recent advances on the diverse mechanisms of human DUBs implicated in IFN-I signaling. These DUBs function dynamically to calibrate host defenses against various virus infections by targeting hub proteins in the IFN-I signaling transduction pathway. We also present a future perspective on the roles of DUB-substrate interaction networks in innate antiviral activities, discuss the promises and challenges of DUB-based drug development, and identify the open questions that remain to be clarified. Our review provides a comprehensive description of DUBs, particularly their differential mechanisms that have evolved in the host to regulate IFN-I-signaling-mediated antiviral responses.
Collapse
Affiliation(s)
- Guanghui Qian
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Liyan Zhu
- Department of Experimental Center, Medical College of Soochow University, Suzhou, China
| | - Gen Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Ying Liu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Zimu Zhang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Haitao Lv
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
12
|
Molecular Mechanisms of DUBs Regulation in Signaling and Disease. Int J Mol Sci 2021; 22:ijms22030986. [PMID: 33498168 PMCID: PMC7863924 DOI: 10.3390/ijms22030986] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
The large family of deubiquitinating enzymes (DUBs) are involved in the regulation of a plethora of processes carried out inside the cell by protein ubiquitination. Ubiquitination is a basic pathway responsible for the correct protein homeostasis in the cell, which could regulate the fate of proteins through the ubiquitin–proteasome system (UPS). In this review we will focus on recent advances on the molecular mechanisms and specificities found for some types of DUBs enzymes, highlighting illustrative examples in which the regulatory mechanism for DUBs has been understood in depth at the molecular level by structural biology. DUB proteases are responsible for cleavage and regulation of the multiple types of ubiquitin linkages that can be synthesized inside the cell, known as the ubiquitin-code, which are tightly connected to specific substrate functions. We will display some strategies carried out by members of different DUB families to provide specificity on the cleavage of particular ubiquitin linkages. Finally, we will also discuss recent progress made for the development of drug compounds targeting DUB proteases, which are usually correlated to the progress of many pathologies such as cancer and neurodegenerative diseases.
Collapse
|
13
|
Li X, Xia Q, Meng C, Wu H, Huang H, Qian J, Li A, Zhai A, Kao W, Song W, Zhang F. Downregulation of SOCS gene expression can inhibit the formation of acute and persistent BDV infections. Scand J Immunol 2020; 93:e12974. [PMID: 32910495 DOI: 10.1111/sji.12974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 08/30/2020] [Indexed: 01/18/2023]
Abstract
High expression of suppressors of cytokine signalling (SOCS) has been detected during various viral infections. As a negative feedback regulator, SOCS participates in the regulation of multiple signalling pathways. In this study, to study the related mechanism between SOCS and BDV and to explore the effect of SOCS on IFN pathways in nerve cells, downregulated of SOCS1/3 in oligodendroglial (OL) cells and OL cells persistently infected with BDV (OL/BDV) were constructed with RNA interference technology. An interferon inducer (poly I:C, PIC) and an IFN-α/β R1 antibody were used as stimulation in the SOCS1/3 low-expression cell models, qRT-PCR was used to detect type I IFN and BDV nucleic acid expression, Western blot was used to detect the expression of BDV P40 protein. After BDV acute infection with OL cells which with downregulated SOCS expression, the virus accounting was not detected, and the viral protein expression was lower than that of OL/BDV cells; the OL/BDV cells with downregulated SOCS expression had lower virus nucleic acid and protein expression than OL/BDV cells. Stimulated by IFN-α/β R1 antibody, the expression of type I interferon in OL/BDV cells decreased, and the content of BDV nucleic acid and protein increased, which was higher than that of OL/BDV cells. From the results, it was concluded that downregulating SOCS1/3 can inhibit the formation of acute BDV infection and virus replication in persistent BDV infection by promoting the expression of IFN-α/β and that SOCS can be used as a new target for antiviral therapy.
Collapse
Affiliation(s)
- Xuejiao Li
- Department of Microbiology, Wu Lien-Teh Institute, The Heilongjiang Key Laboratory of Immunity and Infection, The Key Laboratory of Pathogenic Biology, Heilongjiang Higher Education Institutions, Harbin Medical University, Harbin, China.,Department of Basic Medicine Science, Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Qing Xia
- Department of Microbiology, Wu Lien-Teh Institute, The Heilongjiang Key Laboratory of Immunity and Infection, The Key Laboratory of Pathogenic Biology, Heilongjiang Higher Education Institutions, Harbin Medical University, Harbin, China
| | - Caiyun Meng
- Department of Microbiology, Wu Lien-Teh Institute, The Heilongjiang Key Laboratory of Immunity and Infection, The Key Laboratory of Pathogenic Biology, Heilongjiang Higher Education Institutions, Harbin Medical University, Harbin, China
| | - Hao Wu
- Department of Microbiology, Wu Lien-Teh Institute, The Heilongjiang Key Laboratory of Immunity and Infection, The Key Laboratory of Pathogenic Biology, Heilongjiang Higher Education Institutions, Harbin Medical University, Harbin, China
| | - He Huang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jun Qian
- Department of Microbiology, Wu Lien-Teh Institute, The Heilongjiang Key Laboratory of Immunity and Infection, The Key Laboratory of Pathogenic Biology, Heilongjiang Higher Education Institutions, Harbin Medical University, Harbin, China
| | - Aimei Li
- Department of Microbiology, Wu Lien-Teh Institute, The Heilongjiang Key Laboratory of Immunity and Infection, The Key Laboratory of Pathogenic Biology, Heilongjiang Higher Education Institutions, Harbin Medical University, Harbin, China
| | - Aixia Zhai
- Department of Microbiology, Wu Lien-Teh Institute, The Heilongjiang Key Laboratory of Immunity and Infection, The Key Laboratory of Pathogenic Biology, Heilongjiang Higher Education Institutions, Harbin Medical University, Harbin, China
| | - Wenping Kao
- Department of Microbiology, Wu Lien-Teh Institute, The Heilongjiang Key Laboratory of Immunity and Infection, The Key Laboratory of Pathogenic Biology, Heilongjiang Higher Education Institutions, Harbin Medical University, Harbin, China
| | - Wuqi Song
- Department of Microbiology, Wu Lien-Teh Institute, The Heilongjiang Key Laboratory of Immunity and Infection, The Key Laboratory of Pathogenic Biology, Heilongjiang Higher Education Institutions, Harbin Medical University, Harbin, China
| | - Fengmin Zhang
- Department of Microbiology, Wu Lien-Teh Institute, The Heilongjiang Key Laboratory of Immunity and Infection, The Key Laboratory of Pathogenic Biology, Heilongjiang Higher Education Institutions, Harbin Medical University, Harbin, China
| |
Collapse
|