1
|
Cui Q, Miao Y, Li M, Zheng H, Yuan Y. Viperin: A Multifunctional Protein in Antiviral Immunity and Disease Pathogenesis. Pathogens 2025; 14:510. [PMID: 40430829 PMCID: PMC12114427 DOI: 10.3390/pathogens14050510] [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: 04/26/2025] [Revised: 05/15/2025] [Accepted: 05/20/2025] [Indexed: 05/29/2025] Open
Abstract
Innate immunity is an important component of the immune system and serves as the first line of defense for the host against the invasion of foreign pathogens. Viperin (RSAD2), a core member of the interferon-stimulated gene (ISG) family, plays a key role in innate immunity through direct inhibition of viral replication and modulation of the host immune-metabolic network. The intracellular expression of Viperin rises markedly after viral infection or interferon-induced induction, showing a wide range of antiviral activities. In recent years, the versatility of Viperin in viral infections, autoimmune diseases, and tumor immune metabolism has been gradually revealed. Here, we summarize and discuss the gene regulatory network, molecular functions, and multi-dimensional roles of Viperin in diseases to provide a theoretical basis for the development of broad-spectrum antiviral strategies and immunometabolic therapies based on Viperin.
Collapse
Affiliation(s)
- Qun Cui
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China (M.L.)
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, China
| | - Ying Miao
- Department of Laboratory Medicine, Institute of Laboratory Medicine, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Min Li
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China (M.L.)
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, China
| | - Hui Zheng
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China (M.L.)
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, China
- Department of Laboratory Medicine, Institute of Laboratory Medicine, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Yukang Yuan
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China (M.L.)
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, China
- Department of Laboratory Medicine, Institute of Laboratory Medicine, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 611731, China
| |
Collapse
|
2
|
Woolls MK, Mott MD, Poole CS, Gregory JA, Ivester HM, Allen IC. Innate Immunity Never "NODs" Off: NLRs Regulate the Host Anti-Viral Immune Response. Immunol Rev 2025; 330:e13429. [PMID: 39878363 PMCID: PMC11776368 DOI: 10.1111/imr.13429] [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: 09/14/2024] [Revised: 11/22/2024] [Accepted: 12/02/2024] [Indexed: 01/31/2025]
Abstract
A robust innate immune response is essential in combating viral pathogens. However, it is equally critical to quell overzealous immune signaling to limit collateral damage and enable inflammation resolution. Pattern recognition receptors are critical regulators of these processes. The cytosolic nucleotide-binding domain leucine-rich repeat (NLR; NOD-like receptor) family of pattern recognition receptors plays essential roles in the sensing of viral pathogen-associated molecular patterns and is best characterized for itsr pro-inflammatory biological functions. Specifically, these include the formation of multi-protein complexes, defined as inflammasomes or NODosomes that regulate the production of IL-1beta, IL-18, and pyroptosis, or the induction of NF-ΚB signaling. While these biological effects are inherently pro-inflammatory, it is also important to recognize that other NLR family members conversely function to negatively regulate inflammation through modulating signaling initiated by other families of pattern recognition receptors. Mechanistically, these unique NLRs also form multiprotein complexes that act to attenuate a variety of biological signaling pathways, such as the inhibition of NF-ΚB. This inhibition facilitates inflammation resolution and functions to restore cellular homeostasis. Despite extensive characterization of individual NLR family members, the mechanisms of immune system regulation are highly nuanced and remain enigmatic. This is especially true for non-inflammasome-forming, regulatory NLRs. Here, we discuss recent findings associated with NLR family members that play essential roles in the host immune response to viruses and mechanisms by which these pattern recognition receptors may function to regulate antiviral immunity.
Collapse
Affiliation(s)
- Mackenzie K. Woolls
- Graduate Program in Translational Biology, Medicine, and HealthVirginia TechRoanokeVirginiaUSA
| | - Madeline D. Mott
- Graduate Program in Translational Biology, Medicine, and HealthVirginia TechRoanokeVirginiaUSA
| | - Cassandra S. Poole
- Biomedical and Veterinary Sciences Graduate Program, Virginia Maryland College of Veterinary MedicineVirginia TechBlacksburgVirginiaUSA
| | - Julia A. Gregory
- Department of Biological Sciences Undergraduate Program, College of Biological SciencesVirginia TechBlacksburgVirginiaUSA
| | - Hannah M. Ivester
- Graduate Program in Translational Biology, Medicine, and HealthVirginia TechRoanokeVirginiaUSA
| | - Irving Coy Allen
- Graduate Program in Translational Biology, Medicine, and HealthVirginia TechRoanokeVirginiaUSA
- Biomedical and Veterinary Sciences Graduate Program, Virginia Maryland College of Veterinary MedicineVirginia TechBlacksburgVirginiaUSA
- Department of Basic Science EducationVirginia Tech Carilion School of MedicineRoanokeVirginiaUSA
- The Virginia Tech Center for Emerging, Zoonotic and Arthropod‐Borne PathogensVirginia TechBlacksburgVirginiaUSA
| |
Collapse
|
3
|
Jing H, Song Y, Duan E, Liu J, Ke W, Tao R, Lv Y, Zhao P, Dong W, Li X, Guo Y, Li H. NLRP12 inhibits PRRSV-2 replication by promoting GP2a degradation via MARCH8. Vet Microbiol 2024; 298:110271. [PMID: 39362085 DOI: 10.1016/j.vetmic.2024.110271] [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: 07/30/2024] [Revised: 09/25/2024] [Accepted: 09/28/2024] [Indexed: 10/05/2024]
Abstract
NLRP12, a member of the NLR family, has been shown to exert a vital function in orchestrating immune responses. Here, using the immunosuppressive porcine reproductive and respiratory syndrome virus (PRRSV) as a model, the role of NLRP12 in virus infection was deciphered. We demonstrated that overexpression of NLRP12 significantly restrained PRRSV replication, while NLRP12 silencing resulted in increased viral titer. Mechanistically, NLRP12 interacts with glycoprotein 2a (GP2a) through its LRR domain and recruits the membrane-associated RING-CH E3 ubiquitin ligase 8 (MARCH8) via the PYD domain. NLRP12 facilitates the lysine-48 (K48)-linked polyubiquitination of GP2a at K128 and induces its lysosome degradation via the MARCH8-NDP52 (nuclear dot protein 52 kDa) pathway. To counteract this, PRRSV Nsp2 effectively prevented the polyubiquitination of GP2a induced by NLRP12 by its deubiquitinating activity. Meanwhile, the overexpression of Nsp4 decreased the mRNA of endogenous NLRP12 and cleaved NLRP12 in a 3C-like protease activity-dependent manner, which collaboratively counteracts the antiviral function of NLRP12. Collectively, this study revealed the mechanisms of the NLRP12-MARCH8-NDP52 axis in the host defense against PRRSV, which might be harnessed for the development of anti-PRRSV therapies.
Collapse
Affiliation(s)
- Huiyuan Jing
- Key Laboratory of Veterinary Biological Products, College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, China.
| | - Yuzhen Song
- Key Laboratory of Veterinary Biological Products, College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, China
| | - Erzhen Duan
- College of Biological Engineering, Henan university of technology, Zhengzhou, China
| | - Jie Liu
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu, China
| | - Wenting Ke
- College of Animal Science and Technology, Yangtze University, Jingzhou, China
| | - Ran Tao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Yujin Lv
- Key Laboratory of Veterinary Biological Products, College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, China
| | - Pandeng Zhao
- Key Laboratory of Veterinary Biological Products, College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, China
| | - Wang Dong
- Key Laboratory of Veterinary Biological Products, College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, China
| | - Xianghui Li
- Key Laboratory of Veterinary Biological Products, College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, China
| | - Yongbin Guo
- Key Laboratory of Veterinary Biological Products, College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, China
| | - Huawei Li
- Key Laboratory of Veterinary Biological Products, College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, China.
| |
Collapse
|
4
|
Cheng X, Zeng T, Xu Y, Xiong Y. The emerging role of PANoptosis in viral infections disease. Cell Signal 2024; 125:111497. [PMID: 39489200 DOI: 10.1016/j.cellsig.2024.111497] [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: 09/11/2024] [Revised: 10/19/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
PANoptosis is a distinct inflammatory cell death mechanism that involves interactions between pyroptosis, apoptosis, and necroptosis. It can be regulated by diverse PANoptosome complexes built by integrating components from various cell death modalities. There is a rising interest in PANoptosis' process and functions. Viral infection is an important trigger of PANoptosis. Viruses invade host cells through their unique mechanisms and utilize host cell resources for replication and proliferation. In this process, viruses interfere with the normal physiological functions of host cells, including cell death mechanisms. A variety of viruses, such as influenza A virus (IAV), herpes simplex virus 1 (HSV1) and coronaviruses, have been found to induce PANoptosis in host cells. Given the importance of PANoptosis across the disease spectrum, this review briefly describes the relationships between pyroptosis, apoptosis, and necroptosis, highlights the key molecules in PANoptosome formation and activation, and outlines the multifaceted roles of PANoptosis in viral diseases, including potential therapeutic targets. We also talk about key principles and significant concerns for future PANoptosis research. Improved understanding of PANoptosis and its mechanisms is critical for discovering new treatment targets and methods.
Collapse
Affiliation(s)
- Xu Cheng
- Department of Pharmaceutics, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Taoyuan Zeng
- Department of Pharmaceutics, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Yingshu Xu
- Department of Pharmaceutics, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.
| | - Yongai Xiong
- Department of Pharmaceutics, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
5
|
Jiang J, Shen W, He Y, Liu J, Ouyang J, Zhang C, Hu K. Overexpression of NLRP12 enhances antiviral immunity and alleviates herpes simplex keratitis via pyroptosis/IL-18/IFN-γ signaling. Int Immunopharmacol 2024; 137:112428. [PMID: 38908077 DOI: 10.1016/j.intimp.2024.112428] [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: 02/07/2024] [Revised: 06/04/2024] [Accepted: 06/04/2024] [Indexed: 06/24/2024]
Abstract
Herpes simplex keratitis (HSK) is a blinding disease caused by herpes simplex virus type 1 (HSV-1) infection, and rapid eradication of the virus from the affected cornea is imperative. Nod-like receptors (NLRs) are intracellular innate immune sensors closely associated with cell death, inflammation and immune responses. In this study, we investigated the role of NLRP12 in the antiviral immunology in HSK and the underlying mechanisms. We found that NLRP12 expression was significantly decreased in HSV-1-infected human corneal epithelial cells (HCE-Ts) and HSK mouse corneas. Overexpression of NLRP12 significantly reduced viral replication in infected HCE-Ts and functioned through inflammasome-mediated pyroptosis and downstream IL-18-IFN-γ axis. In HSK mouse models, overexpression of NLRP12 reduced viral replication in the cornea and alleviated HSK symptoms. This resulted from enhanced antiviral immune responses including the activation of specific immune cells in both the cornea and the draining lymph nodes. Specifically, the NLRP12-IL-18-IFN-γ axis regulated the interaction between infected corneal epithelial cells and macrophages. In conclusion, our study identified a role of NLRP12 in mediating pyroptosis and regulating antiviral immune responses. This novel finding opens the possibilities of NLRP12 as a viable target in the therapeutic strategies for HSV-1 infection.
Collapse
Affiliation(s)
- Jiaxuan Jiang
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing 210008, China
| | - Wenhao Shen
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing 210008, China
| | - Yun He
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing 210008, China
| | - Junpeng Liu
- Department of Ophthalmology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing 210008, China
| | - Junwen Ouyang
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing 210008, China
| | - Chengxiao Zhang
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing 210008, China
| | - Kai Hu
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing 210008, China.
| |
Collapse
|
6
|
Jiang J, Zhang D, Liu W, Yang J, Yang F, Liu J, Hu K. Overexpression of NLRP12 enhances macrophage immune response and alleviates herpes simplex keratitis. Front Cell Infect Microbiol 2024; 14:1416105. [PMID: 39119293 PMCID: PMC11306119 DOI: 10.3389/fcimb.2024.1416105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/27/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction Herpes simplex keratitis (HSK) is a blinding disease caused by corneal infection of Herpes simplex virus type 1 (HSV-1). Effective clearance of HSV-1 from the infected cornea is crucial for HSK management. Macrophages play an important part in the innate immune defense against viral infections. This study investigates the immunomodulatory role of NLRP12 in macrophage immune response during HSV-1 infection. Methods NLRP12 expression post-infection was assessed in various macrophage cell lines. Overexpression of NLRP12 was achieved by lentiviral transfection, and its effect on HSV-1 replication and immune responses were examined. Mechanistic insights into the role of NLRP12 were explored using immunofluorescence and Western Blot. For in vivo studies, ocular adoptive transfer of NLRP12-overexpressing bone marrow derived macrophages (BMDMs) was performed. HSV-1 viral loads, HSK symptoms, and macrophage-mediated immune responses were investigated. Results A significant decrease in NLRP12 expression post-infection was observed in various macrophage cell lines. Overexpression of NLRP12 in macrophages reduced HSV-1 replication. Mechanistically, overexpression of NLRP12 triggered early and robust pyroptosis in response to HSV-1 infection, inducing interleukin (IL)-18 production and activating downstream antiviral responses through the JAK-STAT signaling pathway. In vivo, ocular adoptive transfer of NLRP12-overexpressing BMDMs to mouse corneas alleviated HSK damage and reduced HSV-1 viral loads. NLRP12-overexpressing BMDMs improved antiviral responses in the cornea and promoted the maturation of corneal-infiltrating macrophages and dendritic cells. Additionally, NLRP12-overexpressing BMDMs amplified the adaptive immune response in the submandibular draining lymph nodes. Discussion These findings highlight the role of NLRP12 in macrophage-mediated immune response against HSV-1 infection and suggest its potential for possible immunotherapy for HSK.
Collapse
Affiliation(s)
- Jiaxuan Jiang
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Di Zhang
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wei Liu
- Department of Ophthalmology, Linyi Bright Eye Hospital, Linyi, China
| | - Jingya Yang
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Fan Yang
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Junpeng Liu
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Kai Hu
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
7
|
Zhu L, Qi Z, Zhang H, Wang N. Nucleic Acid Sensor-Mediated PANoptosis in Viral Infection. Viruses 2024; 16:966. [PMID: 38932258 PMCID: PMC11209569 DOI: 10.3390/v16060966] [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: 04/29/2024] [Revised: 06/04/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Innate immunity, the first line of host defense against viral infections, recognizes viral components through different pattern-recognition receptors. Nucleic acids derived from viruses are mainly recognized by Toll-like receptors, nucleotide-binding domain leucine-rich repeat-containing receptors, absent in melanoma 2-like receptors, and cytosolic DNA sensors (e.g., Z-DNA-binding protein 1 and cyclic GMP-AMP synthase). Different types of nucleic acid sensors can recognize specific viruses due to their unique structures. PANoptosis is a unique form of inflammatory cell death pathway that is triggered by innate immune sensors and driven by caspases and receptor-interacting serine/threonine kinases through PANoptosome complexes. Nucleic acid sensors (e.g., Z-DNA-binding protein 1 and absent in melanoma 2) not only detect viruses, but also mediate PANoptosis through providing scaffold for the assembly of PANoptosomes. This review summarizes the structures of different nucleic acid sensors, discusses their roles in viral infections by driving PANoptosis, and highlights the crosstalk between different nucleic acid sensors. It also underscores the promising prospect of manipulating nucleic acid sensors as a therapeutic approach for viral infections.
Collapse
Affiliation(s)
- Lili Zhu
- Department of Pathology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410083, China;
| | - Zehong Qi
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha 410083, China;
- Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha 410083, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha 410083, China
| | - Huali Zhang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha 410083, China;
- Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha 410083, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha 410083, China
| | - Nian Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha 410083, China;
- Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha 410083, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha 410083, China
| |
Collapse
|
8
|
Li X, Zhou G, Sun X, Qu S, Lai H, Wu Y, Li D, Liu L, Zhang G, Yang J, Huang X. NLRP12 Senses the SARS-CoV-2 Membrane Protein and Promotes an Inflammatory Response. J Infect Dis 2024; 229:660-670. [PMID: 37976229 DOI: 10.1093/infdis/jiad458] [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: 01/12/2023] [Indexed: 11/19/2023] Open
Abstract
COVID-19 is an acute respiratory disorder that is caused by SARS-CoV-2, in which excessive systemic inflammation is associated with adverse patient clinical outcomes. Here, we observed elevated expression levels of NLRP12 (nucleotide-binding leucine-rich repeat-containing receptor 12) in human peripheral monocytes and lung tissue during infection with SARS-CoV-2. Co-immunoprecipitation analysis revealed that NLRP12 directly interacted with the M protein through its leucine-rich repeat domain. Moreover, in vitro studies demonstrated that NLRP12 interacted with TRAF3 and promoted its ubiquitination and degradation, which counteracted the inhibitory effect of TRAF3 on the NF-κB/MAPK signaling pathway and promoted the production of inflammatory cytokines. Furthermore, an in vivo study revealed that NLRP12 knockout mice displayed attenuated tissue injury and ameliorated inflammatory responses in the lungs when infected with a SARS-CoV-2 M protein-reconstituted pseudovirus and mouse coronavirus. Taken together, these findings suggest that NLRP12 mediates the inflammatory responses during coronavirus infection.
Collapse
Affiliation(s)
- Xingyu Li
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou
| | - Guangde Zhou
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen
| | - Xingzi Sun
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai
| | - Siying Qu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai
| | - Hongzhi Lai
- Medical Intensive Care Unit, The Third People's Hospital of Shantou, Shantou
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai
| | - Dechang Li
- Tuberculosis Prevention and Control Institution, Yuebei Second People's Hospital, Shaoguan
| | - Lei Liu
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen
| | - Guoliang Zhang
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen
| | - Jingwen Yang
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Xi Huang
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou
| |
Collapse
|
9
|
Kuang W, Gu Q, Zhou Y, Xiao X, He D, Deng Q. Inhibited Expression of NLRP12 Promotes the Development of Triple-Negative Breast Cancer by Activating the NF-κB Pathway. Cell Biochem Biophys 2023; 81:727-735. [PMID: 37658975 PMCID: PMC10611651 DOI: 10.1007/s12013-023-01166-9] [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] [Accepted: 08/18/2023] [Indexed: 09/05/2023]
Abstract
NLRP12 can affect the progression of different diseases, including hepatocellular carcinoma. However, no report on triple-negative breast cancer (TNBC) has been found. Thus, this study aimed to explore the role of NLRP12 in TNBC. In our study, immunohistochemistry, real-time quantitative PCR (qPCR), and Western blot assays were used to evaluate NLRP12 expression in TNBC tissues and cells. Then, NLRP12 lentivirus was constructed and infected into MDA-MB-231 and MDA-MB-157 cells with or without PTD-p65-P1 treatment. Next, cells were collected for cell function detection using the following procedures: colony formation assay for proliferation, Transwell for migration and invasion, and Western blot for NF-κB and MAPK pathway-associated proteins. Finally, a xenograft mouse model was applied; the tumor volume and weight were determined, and NLRP12, p-IκBb-α, and p-IκBb-α expressions were evaluated using qPCR and Western blot. Results indicated that NLRP12 was lowly expressed in TNBC tissues and cells. The inhibition of NLRP12 could induce the proliferation, migration, and invasion of TNBC cells, which also could be reversed by inhibiting the NF-κB pathway (PTD-p65-P1). Moreover, silencing of NLRP12 could upregulate p-IκBb-α, while IκBb-α, p-ERK, ERK, p-p38, p38, p-JNK, and JNK expressions remained unchanged, thereby indicating that only the NF-κB pathway could be activated by NLRP12 silencing. Furthermore, the xenograft mouse model confirmed the abovementioned findings. Therefore, the low expression of NLRP12 promoted the proliferation, migration, and invasion in TNBC cells by activating the NF-κB pathway. This study might provide insights into TNBC therapy.
Collapse
Affiliation(s)
- Wenbin Kuang
- Department of Clinical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, 518110, China
| | - Qingdan Gu
- Laboratory Medicine, Guangdong Medical University, Zhanjiang, 524023, China
| | - Ying Zhou
- Laboratory Medicine, Guangdong Medical University, Zhanjiang, 524023, China
| | - Xiaoqin Xiao
- Department of Pathology, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, 518110, China
| | - Dabao He
- Department of Clinical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, 518110, China
| | - Qiuchan Deng
- Department of Clinical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, 518110, China.
| |
Collapse
|
10
|
Wang Q, Yang J, Li X, Wang W, Wu Y, Li Z, Huang X. HSPA13 modulates type I interferon antiviral pathway and NLRP3 inflammasome to restrict dengue virus infection in macrophages. Int Immunopharmacol 2023; 124:110988. [PMID: 37776769 DOI: 10.1016/j.intimp.2023.110988] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/12/2023] [Accepted: 09/22/2023] [Indexed: 10/02/2023]
Abstract
Dengue virus (DENV) is a type of arthropod-borne Flavivirus, which leads to a series of serious diseases like dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). DENV has a devastating health and economic impact worldwide. However, there are no suitable drugs to combat the virus. Here we reported that HSPA13, also known as stress chaperone (STCH), is a member of the HSP70 family and is a key regulator of type I interferon (IFN-I) and pro-inflammatory responses during DENV infection. HSPA13 expression was increased in macrophages infected with DENV or other Flaviviruses like Zika virus (ZIKV), Yellow fever virus (YFV) and Japanese encephalitis virus (JEV). Further, HSPA13 suppressed the replication of DENV and other Flaviviruses (ZIKV, JEV, YFV), which exhibited broad-spectrum antiviral effects. On the one hand, HSPA13 promoted production of IFN-β and interferon-stimulated genes (ISGs, such as ISG15, OAS and IFIT3) by interacting with RIG-I and up-regulating RIG-I expression during DENV infection. On the other hand, HSPA13 enhanced NLRP3 inflammasome activation and IL-1β secretion by interacting with ASC in DENV infection. We identified HSPA13 as a potential anti-DENV target. Our results provide clues for the development of antiviral drugs against DENV based on HSPA13 and reveal novel drug target against Flaviviruses.
Collapse
Affiliation(s)
- Qiaohua Wang
- Foshan Fourth People's Hospital, Foshan, China; Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Jingwen Yang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Xingyu Li
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Wei Wang
- Foshan Fourth People's Hospital, Foshan, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Zhijian Li
- Foshan Fourth People's Hospital, Foshan, China.
| | - Xi Huang
- Foshan Fourth People's Hospital, Foshan, China; Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China.
| |
Collapse
|
11
|
Huang L, Tao Y, Wu X, Wu J, Shen M, Zheng Z. The role of NLRP12 in inflammatory diseases. Eur J Pharmacol 2023; 956:175995. [PMID: 37572944 DOI: 10.1016/j.ejphar.2023.175995] [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: 04/20/2023] [Revised: 08/04/2023] [Accepted: 08/10/2023] [Indexed: 08/14/2023]
Abstract
Nucleotide-binding leucine-rich repeat-containing receptor 12 (NLRP12), a highly conserved protein containing an N-terminal pyrin domain (PYD), a nucleotide-binding domain and a C-terminal leucine-rich repeat region, belongs to the nucleotide-binding oligomerization domain-like receptor-containing PYD (NLRP) family and is a cytoplasmic sensor that plays a negative role in inflammation. NLRP12 is involved in multiple disease processes, including formation of inflammasomes and regulation of both canonical and noncanonical inflammatory signaling pathways. NLRP12 and pathogenic infections are closely linked, and alterations in NLRP12 expression and activity are associated with inflammatory diseases. In this review, we begin with a summary of the mechanisms of negative regulation by NLRP12. We then underscore the important roles of NLRP12 in the onset and progression of inflammation, infectious disease, host defense, carcinogenesis and COVID-19. Finally, we highlight factors that influence NLRP12 activity, including synthetic and naturally derived agonists, and are regarded as potential therapeutic agents to overcome inflammatory diseases.
Collapse
Affiliation(s)
- Lili Huang
- Lihuili Hospital Affiliated to Ningbo University, Ningbo, 315100, Zhejiang, China
| | - Youli Tao
- Lihuili Hospital Affiliated to Ningbo University, Ningbo, 315100, Zhejiang, China
| | - Xiping Wu
- Lihuili Hospital Affiliated to Ningbo University, Ningbo, 315100, Zhejiang, China
| | - Jianzhang Wu
- The Eye Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Mengya Shen
- Affiliated Hospital of Jiaxing University, Jiaxing Maternity and Child Health Care Hospital in Zhejiang Province, Jiaxing, 314000, Zhejiang, China.
| | - Zhiwei Zheng
- School of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
12
|
Solstad A, Hogaboam O, Forero A, Hemann EA. RIG-I-like Receptor Regulation of Immune Cell Function and Therapeutic Implications. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:845-854. [PMID: 36130131 PMCID: PMC9512390 DOI: 10.4049/jimmunol.2200395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 06/30/2022] [Indexed: 01/04/2023]
Abstract
Retinoic acid-inducible gene I-like receptors (RLRs) are cytosolic RNA sensors critical for initiation of antiviral immunity. Activation of RLRs following RNA recognition leads to production of antiviral genes and IFNs for induction of broad antiviral immunity. Although the RLRs are ubiquitously expressed, much of our understanding of these molecules comes from their study in epithelial cells and fibroblasts. However, RLR activation is critical for induction of immune function and long-term protective immunity. Recent work has focused on the roles of RLRs in immune cells and their contribution to programming of effective immune responses. This new understanding of RLR function in immune cells and immune programming has led to the development of vaccines and therapeutics targeting the RLRs. This review covers recent advances in our understanding of the contribution of RLRs to immune cell function during infection and the emerging RLR-targeting strategies for induction of immunity against cancer and viral infection.
Collapse
Affiliation(s)
- Abigail Solstad
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH
| | - Octavia Hogaboam
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH
| | - Adriana Forero
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH
- Infectious Diseases Institute, The Ohio State University, Columbus, OH; and
| | - Emily A Hemann
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH;
- Infectious Diseases Institute, The Ohio State University, Columbus, OH; and
| |
Collapse
|
13
|
Li Y, Wei L, He L, Sun J, Liu N. Interferon-induced transmembrane protein 3 gene polymorphisms are associated with COVID-19 susceptibility and severity: A meta-analysis. J Infect 2022; 84:825-833. [PMID: 35461906 PMCID: PMC9022375 DOI: 10.1016/j.jinf.2022.04.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/01/2022] [Accepted: 04/15/2022] [Indexed: 01/17/2023]
Abstract
BACKGROUND Recent evidence has linked the interferon-induced transmembrane protein 3 gene (IFITM3) to coronavirus disease 2019 (COVID-19) outcomes, but the results are inconsistent. The purpose of this meta-analysis was to evaluate the association of IFITM3 gene polymorphisms with COVID-19 susceptibility and severity. METHOD A systematic search was performed with PubMed, Web of Science, Cochrane Library, and Embase from the date of inception to 20 December 2021. The results were analyzed with pooled odds ratios (ORs) and 95% confidence intervals (95% CIs). The robustness was performed using the method of sequential removal for each trial. RESULTS Four studies involving 1989 subjects were included, from which 1114 patients were positive for COVID-19. For IFITM3 rs12252, the pooled OR showed that there was a significant association between the genotype frequencies and infection with COVID-19 in any of the gene models, i.e., the allelic model (OR = 1.91, 95% CI, 1.36-2.68), the dominant model (OR = 1.80, 95% CI, 1.27-2.56), the recessive model (OR = 5.67, 95% CI, 1.01-31.77), the heterozygous model (OR = 1.65, 95% CI, 1.16-2.36) and the homozygous model (OR = 5.88, 95% CI, 1.05-32.98). The results stratified by severity showed that there was a significant correlation only between the allelic (OR = 0.69, 95% CI, 0.49-0.97) and recessive (OR = 0.43, 95% CI, 0.20-0.93) models. Our results did not support the associations between the IFITM3 rs34481144 gene polymorphism and COVID-19 susceptibility or severity in any of the gene models. CONCLUSIONS The findings indicated that IFITM3 rs12252 gene polymorphisms were associated with COVID-19 susceptibility and that the rs12252-C variant was particularly critical for severity. Genetic factors should be considered in future vaccine development.
Collapse
Affiliation(s)
- Yapeng Li
- Rehabilitation Therapy Center, Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Luoyang, China
| | - Lanlan Wei
- Inspection and Monitoring Center, Luoyang Center for Disease Control and Prevention, Luoyang, China
| | - Lanye He
- Department of Geratology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiahui Sun
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Nanyang Liu
- Department of Geratology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China,Corresponding author
| |
Collapse
|