1
|
Zhong H, Li M, Wu H, Ying H, Zhong M, Huang M. Silencing DDX3 Attenuates Interleukin-1β-Induced Intervertebral Disc Degeneration Through Inhibiting Pyroptosis. Inflammation 2025; 48:104-117. [PMID: 38735906 DOI: 10.1007/s10753-024-02042-1] [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: 02/26/2024] [Revised: 04/08/2024] [Accepted: 04/30/2024] [Indexed: 05/14/2024]
Abstract
Intervertebral disc degeneration (IVDD) is a common disorder associated with chronic inflammation and cell death. In this study, an IVDD rat model was created through Interleukin-1β (IL-1β) injection. The degeneration of intervertebral disc tissues was assessed using magnetic resonance imaging (MRI), followed by hematoxylin and eosin (H&E) and terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling (TUNEL) staining. RNA sequencing was performed to identify differentially expressed genes (DEGs) between the IVDD model and control rats. The expression levels of DEGs (DEAD-box polypeptide 3 (DDX3), lysine-specific demethylase 5D (KDM5D), interferon-induced gene-1 (IFIT1), ribosomal protein S10 (RPS10), tenomodulin (TNMD), and pentraxin 3 (PTX3)) were measured by real-time quantitative polymerase chain reaction (RT-qPCR). The regulatory effect of DDX3 on pyroptosis in IL-1β-treated nucleus pulpous (NP) cells was assessed after transfection with siRNA of DDX3. A total of 601 DEGs were identified from the IVDD model rat, and were abundant in extracellular matrix (ECM) organization, ECM-receptor interaction, and inflammatory pathways, including the PI3K-Akt, TNF, and AMPK signaling pathways. DDX3, KDM5D, and IFIT1 levels were notably elevated, whereas RPS10, TNMD, and PTX3 levels were decreased in the IL-1β-induced IVDD rat model. Moreover, silencing DDX3 promoted cell proliferation and abolished IL-1β-induced cell apoptosis and pyroptosis. This study revealed the role of DDX3 in IVDD pyroptosis, providing potential target for IVDD management.
Collapse
Affiliation(s)
- Hongfa Zhong
- Department of Orthopaedics, Ganzhou People's Hospital, No.16 Meiguang Avenue, Ganzhou City, Jiangxi Province, 341000, China.
| | - Mingheng Li
- Department of Orthopaedics, Ganzhou People's Hospital, No.16 Meiguang Avenue, Ganzhou City, Jiangxi Province, 341000, China
| | - Haijian Wu
- Department of Orthopaedics, Ganzhou People's Hospital, No.16 Meiguang Avenue, Ganzhou City, Jiangxi Province, 341000, China
| | - Hui Ying
- Department of Orthopaedics, Ganzhou People's Hospital, No.16 Meiguang Avenue, Ganzhou City, Jiangxi Province, 341000, China
| | - Mingliang Zhong
- Department of Orthopaedics, Ganzhou People's Hospital, No.16 Meiguang Avenue, Ganzhou City, Jiangxi Province, 341000, China
| | - Mouzhang Huang
- Department of Orthopaedics, Ganzhou People's Hospital, No.16 Meiguang Avenue, Ganzhou City, Jiangxi Province, 341000, China
| |
Collapse
|
2
|
Zhu L, Tong H, Ren C, Chen K, Luo S, Wang Q, Guo M, Xu Y, Hu M, Fang J, Xu J, Shi P. Inflammation unleashed: The role of pyroptosis in chronic liver diseases. Int Immunopharmacol 2024; 141:113006. [PMID: 39213865 DOI: 10.1016/j.intimp.2024.113006] [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: 06/12/2024] [Revised: 08/04/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Pyroptosis, a newly identified form of programmed cell death intertwined with inflammatory responses, is facilitated by the Gasdermin family's pore-forming activity, leading to cell lysis and the release of pro-inflammatory cytokines. This process is a double-edged sword in innate immunity, offering protection against pathogens while risking excessive inflammation and tissue damage when dysregulated. Specifically, pyroptosis operates through two distinct signaling pathways, namely the Caspase-1 pathway and the Caspase-4/5/11 pathway. In the context of chronic liver diseases like fibrosis and cirrhosis, inflammation emerges as a central contributing factor to their pathogenesis. The identification of inflammation is characterized by the activation of innate immune cells and the secretion of pro-inflammatory cytokines such as IL-1α, IL-1β, and TNF-α. This review explores the interrelationship between pyroptosis and the inflammasome, a protein complex located in liver cells that recognizes danger signals and initiates Caspase-1 activation, resulting in the secretion of IL-1β and IL-18. The article delves into the influence of the inflammasome and pyroptosis on various liver disorders, with a specific focus on their molecular and pathophysiological mechanisms. Additionally, the potential therapeutic implications of targeting pyroptosis for liver diseases are highlighted for future consideration.
Collapse
Affiliation(s)
- Lujian Zhu
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Hongjie Tong
- Department of Intensive Care Unit, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Chao Ren
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Kun Chen
- Department of Intensive Care Unit, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Shengnan Luo
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Qin Wang
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Maodong Guo
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Yichen Xu
- Department of Gerontology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Minli Hu
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jinyong Fang
- Department of Hematology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jinxian Xu
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Peifei Shi
- Department of Intensive Care Unit, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China.
| |
Collapse
|
3
|
Zhang Y, Li T, Liu Y, Wang C, Wang D, Xu L, Zhao H, Bai X, Li Z, Wang Y. GSDMD KNOCKOUT ALLEVIATES SEPSIS-ASSOCIATED SKELETAL MUSCLE ATROPHY BY INHIBITING IL18/AMPK SIGNALING. Shock 2024; 62:565-573. [PMID: 39227368 DOI: 10.1097/shk.0000000000002430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
ABSTRACT Background: Sepsis commonly leads to skeletal muscle atrophy, characterized by substantial muscle weakness and degeneration, ultimately contributing to an adverse prognosis. Studies have shown that programmed cell death is an important factor in the progression of muscle loss in sepsis. However, the precise role and mechanism of pyroptosis in skeletal muscle atrophy are not yet fully comprehended. Therefore, we aimed to examine the role and mechanism of action of the pyroptosis effector protein GSDMD in recognized cellular and mouse models of sepsis. Methods: The levels of GSDMD and N-GSDMD in skeletal muscle were evaluated 2, 4, and 8 days after cecal ligation and puncture. Sepsis was produced in mice that lacked the Gsdmd gene (Gsdmd knockout) and in mice with the normal Gsdmd gene (wild-type) using a procedure called cecal ligation and puncture. The degree of muscular atrophy in the gastrocnemius and tibialis anterior muscles was assessed 72 h after surgery in the septic mouse model. In addition, the architecture of skeletal muscles, protein expression, and markers associated with pathways leading to muscle atrophy were examined in mice from various groups 72 h after surgery. The in vitro investigations entailed the use of siRNA to suppress Gsdmd expression in C2C12 cells, followed by stimulation of these cells with lipopolysaccharide to evaluate the impact of Gsdmd downregulation on muscle atrophy and the related signaling cascades. Results: This study has demonstrated that the GSDMD protein, known as the "executive" protein of pyroptosis, plays a crucial role in the advancement of skeletal muscle atrophy in septic mice. The expression of N-GSDMD in the skeletal muscle of septic mice was markedly higher compared with the control group. The Gsdmd knockout mice exhibited notable enhancements in survival, muscle strength, and body weight compared with the septic mice. Deletion of the Gsdmd gene reduced muscular wasting in the gastrocnemius and tibialis anterior muscles caused by sepsis. Studies conducted in living organisms ( in vivo ) and in laboratory conditions ( in vitro ) have shown that the absence of the Gsdmd gene decreases indicators of muscle loss associated with sepsis by blocking the IL18/AMPK signaling pathway. Conclusion: The results of this study demonstrate that the lack of Gsdmd has a beneficial effect on septic skeletal muscle atrophy by reducing the activation of IL18/AMPK and inhibiting the ubiquitin-proteasome system and autophagy pathways. Therefore, our research provides vital insights into the role of pyroptosis in sepsis-related skeletal muscle wasting, which could potentially lead to the development of therapeutic and interventional approaches for preventing septic skeletal muscle atrophy.
Collapse
Affiliation(s)
| | | | - Yukun Liu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Cai J, Li C, Liu S, Tan M, Sun Y, Sun X, Yang M, He B. Angiogenin-mediated tsRNAs control inflammation and metabolic disorder by regulating NLRP3 inflammasome. Cell Death Differ 2024; 31:1057-1069. [PMID: 38740959 PMCID: PMC11303556 DOI: 10.1038/s41418-024-01311-8] [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: 10/25/2023] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/16/2024] Open
Abstract
The cellular stress response system in immune cells plays a crucial role in regulating the development of inflammatory diseases. In response to cellular damage or microbial infection, the assembly of the NLRP3 inflammasome induces pyroptosis and the release of inflammatory cytokines. Meanwhile, Angiogenin (Ang)-mediated transfer RNA-derived small RNAs (tsRNAs) promote cell survival under stressful conditions. While both tsRNAs and inflammasomes are induced under stress conditions, the interplay between these two systems and their implications in regulating inflammatory diseases remains poorly understood. In this study, it was demonstrated that Ang deficiency exacerbated sodium arsenite-induced activation of NLRP3 inflammasome and pyroptosis. Moreover, Ang-induced 5'-tsRNAs inhibited NLRP3 inflammasome activation and pyroptosis. Mechanistically, 5'-tsRNAs recruit DDX3X protein into stress granules (SGs), consequently inhibiting the interaction between DDX3X and NLRP3, thus leading to the suppression of NLRP3 inflammasome activation. Furthermore, in vivo results showed that Ang deficiency led to the downregulation of tsRNAs, ultimately leading to an exacerbation of NLRP3 inflammasome-dependent inflammation, including lipopolysaccharide-induced systemic inflammation and type-2 diabetes-related inflammation. Altogether, our study sheds a new light on the role of Ang-induced 5'-tsRNAs in regulating NLRP3 inflammasome activation via SGs, and highlights tsRNAs as a promising target for the treatment of NLRP3 inflammasome-related diseases.
Collapse
Affiliation(s)
- Jiangxue Cai
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Chenxuan Li
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Suyuan Liu
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Meiling Tan
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Yiran Sun
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Xiaoxiao Sun
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Miaoxin Yang
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Bin He
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China.
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, 210095, PR China.
| |
Collapse
|
5
|
Wen R, Zhang TN, Yang N. [Recent research on pyroptosis in sepsis-induced myocardial depression]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:774-781. [PMID: 39014956 PMCID: PMC11562036 DOI: 10.7499/j.issn.1008-8830.2312039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 05/30/2024] [Indexed: 07/18/2024]
Abstract
Sepsis-induced myocardial depression (SIMD), a common complication of sepsis, is one of the main causes of death in patients with sepsis. The pathogenesis of SIMD is complicated, and the process of SIMD remains incompletely understood, with no single or definitive mechanism fully elucidated. Notably, pyroptosis, as a pro-inflammatory programmed cell death, is characterized by Gasdermin-mediated formation of pores on the cell membrane, cell swelling, and cell rupture accompanied by the release of large amounts of inflammatory factors and other cellular contents. Mechanistically, pyroptosis is mainly divided into the canonical pathway mediated by caspase-1 and the non-canonical pathway mediated by caspase-4/5/11. Pyroptosis has been confirmed to participate in various inflammation-associated diseases. In recent years, more and more studies have shown that pyroptosis is also involved in the occurrence and development of SIMD. This article reviews the molecular mechanisms of pyroptosis and its research progress in SIMD, aiming to provide novel strategies and targets for the treatment of SIMD.
Collapse
Affiliation(s)
- Ri Wen
- Department of Pediatric Intensive Care Unit, Shengjing Hospital, China Medical University, Shenyang 110004, China
| | - Tie-Ning Zhang
- Department of Pediatric Intensive Care Unit, Shengjing Hospital, China Medical University, Shenyang 110004, China
| | - Ni Yang
- Department of Pediatric Intensive Care Unit, Shengjing Hospital, China Medical University, Shenyang 110004, China
| |
Collapse
|
6
|
Dong W, Liao R, Weng J, Du X, Chen J, Fang X, Liu W, Long T, You J, Wang W, Peng X. USF2 activates RhoB/ROCK pathway by transcriptional inhibition of miR-206 to promote pyroptosis in septic cardiomyocytes. Mol Cell Biochem 2024; 479:1093-1108. [PMID: 37347361 DOI: 10.1007/s11010-023-04781-5] [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: 04/07/2023] [Accepted: 06/03/2023] [Indexed: 06/23/2023]
Abstract
Septic cardiomyopathy (SCM) is one of the most serious complications of sepsis. The present study investigated the role and mechanism of upstream stimulatory factor 2 (USF2) in SCM. Serum samples were extracted from SCM patients and healthy individuals. A murine model of sepsis was induced by caecal ligation and puncture (CLP) surgery. Myocardial injury was examined by echocardiography and HE staining. ELISA assay evaluated myocardial markers (CK-MB, cTnI) and inflammatory cytokines (TNF-α, IL-1β, IL-18). Primary mouse cardiomyocytes were treated with lipopolysaccharide (LPS) to simulate sepsis in vitro. RT-qPCR and Western blot were used for analyzing gene and protein levels. CCK-8 assay assessed cell viability. NLRP3 was detected by immunofluorescence. ChIP, RIP and dual luciferase reporter assays were conducted to validate the molecular associations. USF2 was increased in serum from SCM patients, septic mice and primary cardiomyocytes. USF2 silencing improved the survival of septic mice and attenuated sepsis-induced myocardial pyroptosis and inflammation in vitro and in vivo. Mechanistically, USF2 could directly bind to the promoter of miR-206 to transcriptionally inhibit its expression. Moreover, RhoB was confirmed as a target of miR-206 and could promote ROCK activation and NLRP3 inflammasome formation. Moreover, overexpression of RhoB remarkably reversed the protection against LPS-induced inflammation and pyroptosis mediated by USF2 deletion or miR-206 overexpression in cardiomyocytes. The above findings elucidated that USF2 knockdown exerted a cardioprotective effect on sepsis by decreasing pyroptosis and inflammation via miR-206/RhoB/ROCK pathway, suggesting that USF2 may be a novel drug target in SCM.
Collapse
Affiliation(s)
- Wei Dong
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, No. 17, Yong Waizheng Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Ruichun Liao
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, No. 17, Yong Waizheng Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Junfei Weng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, No. 17, Yong Waizheng Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Xingxiang Du
- Department of Emergency, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Jin Chen
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, No. 17, Yong Waizheng Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Xu Fang
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, No. 17, Yong Waizheng Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Wenyu Liu
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, No. 17, Yong Waizheng Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Tao Long
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, No. 17, Yong Waizheng Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Jiaxiang You
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, No. 17, Yong Waizheng Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Wensheng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, No. 17, Yong Waizheng Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Xiaoping Peng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, No. 17, Yong Waizheng Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China.
| |
Collapse
|
7
|
Wang J, Hou J, Peng C. Phospholipid transfer protein ameliorates sepsis-induced cardiac dysfunction through NLRP3 inflammasome inhibition. Open Med (Wars) 2024; 19:20240915. [PMID: 38584827 PMCID: PMC10996989 DOI: 10.1515/med-2024-0915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 02/03/2024] [Accepted: 02/05/2024] [Indexed: 04/09/2024] Open
Abstract
Cardiomyocyte pyroptosis is a primary contributor to sepsis-induced cardiac dysfunction (SICD). Recombinant phospholipid transfer protein (PLTP) have been demonstrated to possess anti-inflammatory and antiseptic properties. However, the effect of PLTP on SICD remains unknown. In this study, we established the in vivo and in vitro sepsis model with the recombinant PLTP treatment. The survival rates of mice, mouse cardiac function, cell viability, the protein level of proinflammatory cytokine, and lactate dehydrogenase level were evaluated. The cardiomyocyte pyroptotic changes were observed. The distribution of PLTP and NOD-like receptor thermal protein domain associated protein 3 (NLRP3) in mouse myocardial tissue and expression of PLTP, apoptosis associated speck like protein containing a CARD (ASC), NLRP3, caspase-1, interleukin (IL)-1β, and Gasdermin D (GSDMD) were detected. PLTP ameliorated the cecal ligation and puncture-induced mouse survival rate decrease and cardiac dysfunction, inhibited the IL-1β, IL-18, and tumor necrosis factor (TNF)-α release, and blocked the NLRP3 inflammasome/GSDMD signaling pathway in septic mice. In vitro, PLTP reversed the lipopolysaccharide-induced cardiomyocyte pyroptosis, expression of IL-1β, IL-6, TNF-α, and activation of the NLRP3 inflammasome/GSDMD signal pathway. Moreover, PLTP could bind to NLRP3 and negatively regulate the activity of the NLRP3 inflammasome/GSDMD signal pathway. This study demonstrated that PLTP can ameliorate SICD by inhibiting inflammatory responses and cardiomyocyte pyroptosis by blocking the activation of the NLRP3 inflammasome/GSDMD signaling pathway.
Collapse
Affiliation(s)
- Jian Wang
- Emergency and Intensive Care Medicine Center, Guang’an People’s Hospital, Guang’an city, Sichuan 638500, PR China
| | - Jing Hou
- Emergency and Intensive Care Medicine Center, Guang’an People’s Hospital, Guang’an city, Sichuan 638500, PR China
| | - Chaohua Peng
- Emergency and Intensive Care Medicine Center, Guang’an People’s Hospital, Guang’an city, Sichuan 638500, PR China
| |
Collapse
|
8
|
Li M, Wu R, Wang L, Zhu D, Liu S, Wang R, Deng C, Zhang S, Chen M, Lu R, Zhu H, Mo M, He X, Luo Z. Usenamine A triggers NLRP3/caspase-1/GSDMD-mediated pyroptosis in lung adenocarcinoma by targeting the DDX3X/SQSTM1 axis. Aging (Albany NY) 2024; 16:1663-1684. [PMID: 38265972 PMCID: PMC10866397 DOI: 10.18632/aging.205450] [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: 09/08/2023] [Accepted: 11/21/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND Usenamine A (C18H17NO6) is a newly developed, natural anticancer drug that reportedly exerts low toxicity. The therapeutic efficacy and underlying mechanisms of usenamine A in lung adenocarcinoma (LUAD) remain poorly understood. We aimed to explore the therapeutic effects and molecular mechanisms through which usenamine A inhibits LUAD tumorigenesis. METHODS We used LUAD cell lines H1299 and A549 in the present study. CCK-8 and colony formation assays were performed to analyze cell proliferation. Cell migration, invasion, and apoptosis were evaluated using wound-healing, transwell, and flow cytometric assays, respectively. Levels of reactive oxygen species were measured using a DCFH-DA probe. Inflammatory factors (lactate dehydrogenase, interleukin [IL]-1β, and IL-18) were detected using enzyme-linked immunosorbent assays. Western blotting was performed to determine the expression of NOD-like receptor pyrin 3 (NLRP3)/caspase-1/gasdermin D (GSDMD) pathway-related proteins. Pyroptosis was detected using transmission electron microscopy. The interaction and co-localization of DDX3X and sequestosome 1 (SQSTM1) were identified using co-immunoprecipitation and immunofluorescence assays, respectively. For in vivo assessment, we established a xenograft model to validate the usenamine A-mediated effects and mechanisms of action in LUAD. RESULTS Usenamine A inhibited the proliferation, migration, and invasion of LUAD cells. Furthermore, usenamine A induced NLRP3/caspase-1/GSDMD-mediated pyroptosis in LUAD cells. Usenamine A upregulated DDX3X expression to trigger pyroptosis. DDX3X interacted with SQSTM1, which is responsible for inducing pyroptosis. In vivo, usenamine A suppressed LUAD tumorigenesis by triggering NLRP3/caspase-1/GSDMD-mediated pyroptosis via the upregulation of the DDX3X/SQSTM1 axis. CONCLUSIONS Usenamine A was found to induce NLRP3/caspase-1/GSDMD-mediated pyroptosis in LUAD by upregulating the DDX3X/SQSTM1 axis.
Collapse
Affiliation(s)
- Min Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Rongrong Wu
- Department of Radiology, The First People’s Hospital of Yunnan Province (Affiliated Hospital of Kunming University of Science and Technology), Kunming 650034, China
| | - Le Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Dongyi Zhu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Shinan Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Ruolan Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Chaowen Deng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Shenglin Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Min Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Ruojin Lu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Hongxing Zhu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Mengting Mo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Xiaoqiong He
- School of Public Health, Kunming Medical University, Kunming 650500, China
| | - Zhuang Luo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| |
Collapse
|
9
|
Fang T, Wang X, Huangfu N. Superfamily II helicases: the potential therapeutic target for cardiovascular diseases. Front Cardiovasc Med 2023; 10:1309491. [PMID: 38152606 PMCID: PMC10752008 DOI: 10.3389/fcvm.2023.1309491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/29/2023] [Indexed: 12/29/2023] Open
Abstract
Cardiovascular diseases (CVDs) still maintain high morbidity and mortality globally. Helicases, a unique class of enzymes, are extensively implicated in the processes of nucleic acid (NA) metabolism across various organisms. They play a pivotal role in gene expression, inflammatory response, lipid metabolism, and so forth. However, abnormal helicase expression has been associated with immune response, cancer, and intellectual disability in humans. Superfamily II (SFII) is one of the largest and most diverse of the helicase superfamilies. Increasing evidence has implicated SFⅡ helicases in the pathogenesis of multiple CVDs. In this review, we comprehensively review the regulation mechanism of SFⅡ helicases in CVDs including atherosclerosis, myocardial infarction, cardiomyopathies, and heart failure, which will contribute to the investigation of ideal therapeutic targets for CVDs.
Collapse
Affiliation(s)
- Tianxiang Fang
- Health Science Center, Ningbo University, Ningbo, China
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Department of Cardiology, Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, China
- Clinical Medicine Research Centre for Cardiovascular Disease of Ningbo, Ningbo, China
| | - Xizhi Wang
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Ning Huangfu
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Department of Cardiology, Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, China
- Clinical Medicine Research Centre for Cardiovascular Disease of Ningbo, Ningbo, China
| |
Collapse
|
10
|
Hedaya OM, Venkata Subbaiah KC, Jiang F, Xie LH, Wu J, Khor ES, Zhu M, Mathews DH, Proschel C, Yao P. Secondary structures that regulate mRNA translation provide insights for ASO-mediated modulation of cardiac hypertrophy. Nat Commun 2023; 14:6166. [PMID: 37789015 PMCID: PMC10547706 DOI: 10.1038/s41467-023-41799-1] [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: 10/17/2022] [Accepted: 09/19/2023] [Indexed: 10/05/2023] Open
Abstract
Translation of upstream open reading frames (uORFs) typically abrogates translation of main (m)ORFs. The molecular mechanism of uORF regulation in cells is not well understood. Here, we data-mined human and mouse heart ribosome profiling analyses and identified a double-stranded RNA (dsRNA) structure within the GATA4 uORF that cooperates with the start codon to augment uORF translation and inhibits mORF translation. A trans-acting RNA helicase DDX3X inhibits the GATA4 uORF-dsRNA activity and modulates the translational balance of uORF and mORF. Antisense oligonucleotides (ASOs) that disrupt this dsRNA structure promote mORF translation, while ASOs that base-pair immediately downstream (i.e., forming a bimolecular double-stranded region) of either the uORF or mORF start codon enhance uORF or mORF translation, respectively. Human cardiomyocytes and mice treated with a uORF-enhancing ASO showed reduced cardiac GATA4 protein levels and increased resistance to cardiomyocyte hypertrophy. We further show the broad utility of uORF-dsRNA- or mORF-targeting ASO to regulate mORF translation for other mRNAs. This work demonstrates that the uORF-dsRNA element regulates the translation of multiple mRNAs as a generalizable translational control mechanism. Moreover, we develop a valuable strategy to alter protein expression and cellular phenotypes by targeting or generating dsRNA downstream of a uORF or mORF start codon.
Collapse
Affiliation(s)
- Omar M Hedaya
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine & Dentistry, Rochester, NY, 14642, USA
- Department of Biochemistry & Biophysics, University of Rochester School of Medicine & Dentistry, Rochester, NY, 14642, USA
| | - Kadiam C Venkata Subbaiah
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine & Dentistry, Rochester, NY, 14642, USA
| | - Feng Jiang
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine & Dentistry, Rochester, NY, 14642, USA
- Department of Biochemistry & Biophysics, University of Rochester School of Medicine & Dentistry, Rochester, NY, 14642, USA
| | - Li Huitong Xie
- Department of Biomedical Genetics, University of Rochester School of Medicine & Dentistry, Rochester, NY, 14642, USA
| | - Jiangbin Wu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine & Dentistry, Rochester, NY, 14642, USA
| | - Eng-Soon Khor
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine & Dentistry, Rochester, NY, 14642, USA
| | - Mingyi Zhu
- Department of Biochemistry & Biophysics, University of Rochester School of Medicine & Dentistry, Rochester, NY, 14642, USA
- The Center for RNA Biology, University of Rochester School of Medicine & Dentistry, Rochester, NY, 14642, USA
| | - David H Mathews
- Department of Biochemistry & Biophysics, University of Rochester School of Medicine & Dentistry, Rochester, NY, 14642, USA
- The Center for RNA Biology, University of Rochester School of Medicine & Dentistry, Rochester, NY, 14642, USA
- The Center for Biomedical Informatics, University of Rochester School of Medicine & Dentistry, Rochester, NY, 14642, USA
| | - Chris Proschel
- Department of Biomedical Genetics, University of Rochester School of Medicine & Dentistry, Rochester, NY, 14642, USA
| | - Peng Yao
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine & Dentistry, Rochester, NY, 14642, USA.
- Department of Biochemistry & Biophysics, University of Rochester School of Medicine & Dentistry, Rochester, NY, 14642, USA.
- The Center for RNA Biology, University of Rochester School of Medicine & Dentistry, Rochester, NY, 14642, USA.
- The Center for Biomedical Informatics, University of Rochester School of Medicine & Dentistry, Rochester, NY, 14642, USA.
| |
Collapse
|
11
|
Wen Y, Liu Y, Liu W, Liu W, Dong J, Liu Q, Hao H, Ren H. Research progress on the activation mechanism of NLRP3 inflammasome in septic cardiomyopathy. Immun Inflamm Dis 2023; 11:e1039. [PMID: 37904696 PMCID: PMC10549821 DOI: 10.1002/iid3.1039] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 11/01/2023] Open
Abstract
Sepsis is an uncontrolled host response to infection, resulting in a clinical syndrome involving multiple organ dysfunctions. Cardiac damage is the most common organ damage in sepsis. Uncontrolled inflammatory response is an important mechanism in the pathogenesis of septic cardiomyopathy (SCM). NLRP3 inflammasome promotes inflammatory response by controlling the activation of caspase-1 and the release of pro-inflammatory cytokines interleukin IL-1β and IL-18. The role of NLRP3 inflammasome has received increasing attention, but its activation mechanism and regulation of inflammation in SCM remain to be investigated.
Collapse
Affiliation(s)
- Yuqi Wen
- Shandong University of Traditional Chinese MedicineJinanChina
| | - Yang Liu
- Affiliated Hospital of Shandong University of Traditional Chinese MedicineJinanChina
| | - Weihong Liu
- Affiliated Hospital of Shandong University of Traditional Chinese MedicineJinanChina
| | - Wenli Liu
- Shandong University of Traditional Chinese MedicineJinanChina
| | - Jinyan Dong
- Shandong University of Traditional Chinese MedicineJinanChina
| | - Qingkuo Liu
- Shandong University of Traditional Chinese MedicineJinanChina
| | - Hao Hao
- Affiliated Hospital of Shandong University of Traditional Chinese MedicineJinanChina
| | - Hongsheng Ren
- Department of Intensive Care UnitShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| |
Collapse
|
12
|
Hedaya OM, Subbaiah KCV, Jiang F, Xie LH, Wu J, Khor E, Zhu M, Mathews DH, Proschel C, Yao P. Secondary structures that regulate mRNA translation provide insights for ASO-mediated modulation of cardiac hypertrophy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.15.545153. [PMID: 37397986 PMCID: PMC10312771 DOI: 10.1101/2023.06.15.545153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Translation of upstream open reading frames (uORFs) typically abrogates translation of main (m)ORFs. The molecular mechanism of uORF regulation in cells is not well understood. Here, we identified a double-stranded RNA (dsRNA) structure residing within the GATA4 uORF that augments uORF translation and inhibits mORF translation. Antisense oligonucleotides (ASOs) that disrupt this dsRNA structure promote mORF translation, while ASOs that base-pair immediately downstream (i.e., forming a bimolecular double-stranded region) of either the uORF or mORF start codon enhance uORF or mORF translation, respectively. Human cardiomyocytes and mice treated with a uORF-enhancing ASO showed reduced cardiac GATA4 protein levels and increased resistance to cardiomyocyte hypertrophy. We further show the general utility of uORF-dsRNA- or mORF- targeting ASO to regulate mORF translation for other mRNAs. Our work demonstrates a regulatory paradigm that controls translational efficiency and a useful strategy to alter protein expression and cellular phenotypes by targeting or generating dsRNA downstream of a uORF or mORF start codon. Bullet points for discoveries dsRNA within GATA4 uORF activates uORF translation and inhibits mORF translation. ASOs that target the dsRNA can either inhibit or enhance GATA4 mORF translation. ASOs can be used to impede hypertrophy in human cardiomyocytes and mouse hearts.uORF- and mORF-targeting ASOs can be used to control translation of multiple mRNAs.
Collapse
Affiliation(s)
- Omar M. Hedaya
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine & Dentistry, Rochester, New York 14642
- Department of Biochemistry & Biophysics, University of Rochester School of Medicine & Dentistry, Rochester, New York 14642
| | - Kadiam C. Venkata Subbaiah
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine & Dentistry, Rochester, New York 14642
| | - Feng Jiang
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine & Dentistry, Rochester, New York 14642
- Department of Biochemistry & Biophysics, University of Rochester School of Medicine & Dentistry, Rochester, New York 14642
| | - Li Huitong Xie
- Department of Biomedical Genetics, University of Rochester School of Medicine & Dentistry, Rochester, New York 14642
| | - Jiangbin Wu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine & Dentistry, Rochester, New York 14642
| | - EngSoon Khor
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine & Dentistry, Rochester, New York 14642
| | - Mingyi Zhu
- Department of Biochemistry & Biophysics, University of Rochester School of Medicine & Dentistry, Rochester, New York 14642
- The Center for RNA Biology, University of Rochester School of Medicine & Dentistry, Rochester, New York 14642
| | - David H. Mathews
- Department of Biochemistry & Biophysics, University of Rochester School of Medicine & Dentistry, Rochester, New York 14642
- The Center for RNA Biology, University of Rochester School of Medicine & Dentistry, Rochester, New York 14642
- The Center for Biomedical Informatics, University of Rochester School of Medicine & Dentistry, Rochester, New York 14642
| | - Chris Proschel
- Department of Biomedical Genetics, University of Rochester School of Medicine & Dentistry, Rochester, New York 14642
| | - Peng Yao
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine & Dentistry, Rochester, New York 14642
- Department of Biochemistry & Biophysics, University of Rochester School of Medicine & Dentistry, Rochester, New York 14642
- The Center for RNA Biology, University of Rochester School of Medicine & Dentistry, Rochester, New York 14642
- The Center for Biomedical Informatics, University of Rochester School of Medicine & Dentistry, Rochester, New York 14642
| |
Collapse
|
13
|
Zhang Y, Lv X, Fan Q, Chen F, Wan Z, Nibaruta J, Wang H, Wang X, Yuan Y, Guo W, Leng Y. miRNA155-5P participated in DDX3X targeted regulation of pyroptosis to attenuate renal ischemia/reperfusion injury. Aging (Albany NY) 2023; 15:3586-3597. [PMID: 37142295 PMCID: PMC10449305 DOI: 10.18632/aging.204692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/18/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND Renal ischemia/reperfusion injury (IRI) induced pathological damage to renal microvessels and tubular epithelial cells through multiple factors. However, studies investigated whether miRNA155-5P targeted DDX3X to attenuate pyroptosis were scarce. RESULTS The expression of pyroptosis-related proteins (caspase-1, interleukin-1β (IL-1β), NOD-like receptor family pyrin domain containing 3 (NLRP3), and IL-18) were up-regulated in the IRI group. Additionally, miR-155-5p was higher in the IRI group comparing with the sham group. The DDX3X was inhibited by the miR-155-5p mimic more than in the other groups. DEAD-box Helicase 3 X-Linked (DDX3X), NLRP3, caspase-1, IL-1β, IL-18, LDH, and pyroptosis rates were higher in all H/R groups than in the control group. These indicators were higher in the miR-155-5p mimic group than in the H/R and the miR-155-5p mimic negative control (NC) group. CONCLUSIONS Current findings suggested that miR-155-5p decreased the inflammation involved in pyroptosis by downregulating the DDX3X/NLRP3/caspase-1 pathway. METHODS Using the models of IRI in mouse and the hypoxia-reoxygenation (H/R)-induced injury in human renal proximal tubular epithelial cells (HK-2 cells), we analyzed the changes in renal pathology and the expression of factors correlated with pyroptosis and DDX3X. Real-time reverse transcription polymerase chain reaction (RT-PCR) detected miRNAs and enzyme-linked immunosorbent assay (ELISA) was used to detect lactic dehydrogenase activity. The StarBase and luciferase assays examined the specific interplay of DDX3X and miRNA155-5P. In the IRI group, severe renal tissue damage, swelling, and inflammation were examined.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu Province, China
| | - Xinghua Lv
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Qian Fan
- Tianjin Eye Hospital and Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Nankai University Affiliated Eye Hospital, Nankai Eye Institute, Nankai University, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Feng Chen
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu Province, China
| | - Zhanhai Wan
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu Province, China
| | - Janvier Nibaruta
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu Province, China
| | - Hao Wang
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu Province, China
| | - Xiaoxia Wang
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yuan Yuan
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu Province, China
| | - Wenwen Guo
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu Province, China
| | - Yufang Leng
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu Province, China
| |
Collapse
|
14
|
Chen Y, Wu Y, Guo L, Yuan S, Sun J, Zhao K, Wang J, An R. Exosomal Lnc NEAT1 from endothelial cells promote bone regeneration by regulating macrophage polarization via DDX3X/NLRP3 axis. J Nanobiotechnology 2023; 21:98. [PMID: 36941678 PMCID: PMC10029245 DOI: 10.1186/s12951-023-01855-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/10/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Bone regeneration is a complex procedure that involves an interaction between osteogenesis and inflammation. Macrophages in the microenvironment are instrumental in bone metabolism. Amount evidence have revealed that exosomes transmitting lncRNA is crucial nanocarriers for cellular interactions in various biotic procedures, especially, osteogenesis. However, the underlying mechanisms of the regulatory relationship between the exosomes and macrophages are awaiting clarification. In the present time study, we aimed to explore the roles of human umbilical vein endothelial cells (HUVECs)-derived exosomes carrying nuclear enrichment enriched transcript 1 (NEAT1) in the osteogenesis mediated by M2 polarized macrophages and elucidate the underlying mechanisms. RESULTS We demonstrated HUVECs-derived exosomes expressing NEAT1 significantly enhanced M2 polarization and attenuated LPS-induced inflammation in vitro. Besides, the conditioned medium from macrophages induced by the exosomes indirectly facilitated the migration and osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs). Mechanically, Exos carrying NEAT1 decreased remarkably both expression of dead-box helicase 3X-linked (DDX3X) and nod-like receptor protein 3 (NLRP3). The level of NLRP3 protein increased significantly after RAW264.7 cells transfected with DDX3X overexpression plasmid. Additionally, the knockdown of NEAT1 in exosomes partially counteracted the aforementioned effect of Exos. The results of air pouch rat model demonstrated that HUVECs-derived exosomes increased anti-inflammatory cytokines (IL-10) and decreased pro-inflammatory cytokines (IL-1β and IL-6) significantly in vivo, contributing to amelioration of LPS-induced inflammation. Afterwards, we further confirmed that the HUVECs-derived exosomes encapsulated in alginate/gelatin methacrylate (GelMA) interpenetrating polymer network (IPN) hydrogels could promote the bone regeneration, facilitate the angiogenesis, increase the infiltration of M2 polarized macrophages as well as decrease NLRP3 expression in the rat calvarial defect model. CONCLUSIONS HUVECs-derived exosomes enable transmitting NEAT1 to alleviate inflammation by inducing M2 polarization of macrophages through DDX3X/NLRP3 regulatory axis, which finally contributes to osteogenesis with the aid of alginate/GelMA IPN hydrogels in vivo. Thus, our study provides insights in bone healing with the aid of HUVECs-derived exosomes-encapsulated composite hydrogels, which exhibited potential towards the use of bone tissue engineering in the foreseeable future.
Collapse
Affiliation(s)
- Yuxuan Chen
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yuanhao Wu
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Linlin Guo
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shijie Yuan
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiaming Sun
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kangcheng Zhao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiecong Wang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Ran An
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
15
|
Chi S, Li S, Xu Z, Yang G, Song Y, Liao Z, Yang C, Wu X. The involvement of DDX3X in compression-induced nucleus pulposus pyroptosis. Biochem Biophys Res Commun 2023; 655:1-10. [PMID: 36907112 DOI: 10.1016/j.bbrc.2023.02.074] [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: 02/12/2023] [Revised: 02/22/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023]
Abstract
OBJECTIVE A study has been conducted to investigate the relationship between DDX3X and nucleus pulposus (NP) pyroptosis. METHODS DDX3X and pyroptosis-related proteins (Caspase-1, Full-length GSDMD, Cleaved GSDMD) were measured in compression-induced human NP cells and tissue. DDX3X was overexpressed or knocked down by gene transfection. The expressions of NLRP3, ASC, and pyroptosis-related proteins were detected by Western blot assay. IL-1β and IL-18 were detected by ELISA. HE staining and immunohistochemistry were used to observe the expression of DDX3X, NLRP3, and Caspase-1 in the rat model of compression-induced disc degeneration. RESULTS DDX3X, NLRP3, and Caspase-1 were highly expressed in degenerated NP tissue. Overexpression of DDX3X induced pyroptosis in NP cells and increased levels of NLRP3, IL-1β, IL-18, and pyroptosis-related proteins. Knockdown of DDX3X showed an opposite trend to overexpression of DDX3X. The NLRP3 inhibitor CY-09 effectively prevented the up-regulation of the expression of IL-1β, IL-18, ASC, Pro-caspase-1, Full-length GSDMD, and Cleaved GSDMD. Increased expression of DDX3X, NLRP3, and Caspase-1 was observed in the rat model of compression-induced disc degeneration. CONCLUSION Our study showed that DDX3X mediates pyroptosis of NP cells by upregulating NLRP3 expression, which ultimately leads to intervertebral disc degeneration (IDD). This discovery deepens the understanding of IDD pathogenesis and provides a promising and novel therapeutic target for IDD.
Collapse
Affiliation(s)
- Shouyuan Chi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Suyun Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Zhiqiang Xu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Guoyu Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Yu Song
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Zhiwei Liao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Xinghuo Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| |
Collapse
|
16
|
Inhibition of DDX3X alleviates persistent inflammation, immune suppression and catabolism syndrome in a septic mice model. Int Immunopharmacol 2023; 117:109779. [PMID: 36806038 DOI: 10.1016/j.intimp.2023.109779] [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: 11/11/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 02/22/2023]
Abstract
OBJECTIVE DDX3X is involved in various pathological processes such as infection, immunity and cell death. This study aimed to investigate the effect of RK-33, a specific inhibitor of DDX3X, on the progression of sepsis to persistent inflammation, immune suppression and catabolism syndrome(PICS). METHODS The septic mice model was established using caecal ligation and perforation (CLP). The mice were randomly divided into four groups: sham group, sham + RK-33 group (20 mg/kg, intraperitoneal injection, once a day), CLP group and CLP + RK-33 group (20 mg/kg, intraperitoneal injection, once a day). The number of inflammatory cells in the peripheral blood, spleen and bone marrow was calculated, and inflammatory cytokines were detected using an enzyme-linked immunosorbent assay. The septic mice's body weight and skeletal muscle mass were measured, and skeletal muscle tissues were examined using eosin staining. Western blotting was performed to detect the expression levels of MuRF1, atrogin1 and NLRP3 in the skeletal muscle of septic mice. Additionally, reactive oxidative species, superoxide dismutase and malondialdehyde were measured using commercial kits. RESULTS RK-33 reduced inflammatory cell counts and cytokine levels in CLP mice, ameliorated the decline in CD4 and CD8 T cells and prevented the loss of body weight and skeletal muscle mass in septic mice. Additionally, RX-33 reduced oxidative stress in the skeletal muscle of septic mice. CONCLUSION In the established sepsis mouse model, RK-33 alleviated inflammation and oxidative stress, ameliorated CLP-induced immunosuppression and skeletal muscle atrophy and improved survival. These findings suggest that RK-33 could be a novel potential therapeutic agent for preventing the progression of sepsis to PICS.
Collapse
|
17
|
Liu Y, Zhang Y, Liu Q, Li T, Wang W, Li H, Yang F, Gao W, Li Z, Bai X, Wang Y. Inhibition of DDX3X ameliorated CD4 + T cells pyroptosis and improves survival in septic mice. Mol Immunol 2023; 154:54-60. [PMID: 36603305 DOI: 10.1016/j.molimm.2022.12.014] [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: 11/01/2022] [Revised: 12/18/2022] [Accepted: 12/27/2022] [Indexed: 01/05/2023]
Abstract
Over-expression of DDX3X mRNA is associated with T cell loss in septic patients. This study aimed to investigate the molecular mechanism of DDX3X on T cell reduction in sepsis. The sepsis model was established using lipopolysaccharide stimulation in vitro and cecal ligation and puncture (CLP) surgery in vivo. Results showed that the expression of DDX3X was significantly upregulated in CD4+ T cells in sepsis. RK-33, the inhibitor of DDX3X, was found to dramatically increase CD4+ T cell counts and prolong the survival rate of mice with sepsis. The results also showed that the expression of caspase-1/GSDMD in CD4+ T cells was significantly increased in vitro and in vivo, and RK-33 can substantially reduce CD4+ T cell pyroptosis through inhibiting NLRP3/caspase-1/GSDMD. Globally, our results suggest that DDX3X is involved in the loss of CD4+ T cells partly through activating the pyroptotic pathway during sepsis, which may provide potential targets for therapeutic interventions in this highly lethal disease.
Collapse
Affiliation(s)
- Yukun Liu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yongsheng Zhang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Qinxin Liu
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Tianyu Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Wei Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Hui Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Fan Yang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Wei Gao
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Zhanfei Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Xiangjun Bai
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Yuchang Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China.
| |
Collapse
|
18
|
Harris J, Borg NA. The multifaceted roles of NLRP3-modulating proteins in virus infection. Front Immunol 2022; 13:987453. [PMID: 36110852 PMCID: PMC9468583 DOI: 10.3389/fimmu.2022.987453] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/11/2022] [Indexed: 12/14/2022] Open
Abstract
The innate immune response to viruses is critical for the correct establishment of protective adaptive immunity. Amongst the many pathways involved, the NLRP3 [nucleotide-binding oligomerisation domain (NOD)-like receptor protein 3 (NLRP3)] inflammasome has received considerable attention, particularly in the context of immunity and pathogenesis during infection with influenza A (IAV) and SARS-CoV-2, the causative agent of COVID-19. Activation of the NLRP3 inflammasome results in the secretion of the proinflammatory cytokines IL-1β and IL-18, commonly coupled with pyroptotic cell death. While this mechanism is protective and key to host defense, aberrant NLRP3 inflammasome activation causes a hyperinflammatory response and excessive release of cytokines, both locally and systemically. Here, we discuss key molecules in the NLRP3 pathway that have also been shown to have significant roles in innate and adaptive immunity to viruses, including DEAD box helicase X-linked (DDX3X), vimentin and macrophage migration inhibitory factor (MIF). We also discuss the clinical opportunities to suppress NLRP3-mediated inflammation and reduce disease severity.
Collapse
Affiliation(s)
- James Harris
- Cell Biology Assays Team, Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, Australia
- Centre for Inflammatory diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Natalie A. Borg
- Immunity and Immune Evasion Laboratory, Chronic Infectious and Inflammatory Diseases Research, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
19
|
Habimana O, Modupe Salami O, Peng J, Yi GH. Therapeutic Implications of Targeting Pyroptosis in Cardiac-related Etiology of Heart Failure. Biochem Pharmacol 2022; 204:115235. [PMID: 36044938 DOI: 10.1016/j.bcp.2022.115235] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/26/2022]
Abstract
Heart failure remains a considerable clinical and public health problem, it is the dominant cause of death from cardiovascular diseases, besides, cardiovascular diseases are one of the leading causes of death worldwide. The survival of patients with heart failure continues to be low with 45-60% reported deaths within five years. Apoptosis, necrosis, autophagy, and pyroptosis mediate cardiac cell death. Acute cell death is the hallmark pathogenesis of heart failure and other cardiac pathologies. Inhibition of pyroptosis, autophagy, apoptosis, or necrosis reduces cardiac damage and improves cardiac function in cardiovascular diseases. Pyroptosis is a form of inflammatory deliberate cell death that is characterized by the activation of inflammasomes such as NOD-like receptors (NLR), absent in melanoma 2 (AIM2), interferon-inducible protein 16 (IFI-16), and their downstream effector cytokines: Interleukin IL-1β and IL-18 leading to cell death. Recent studies have shown that pyroptosis is also the dominant cell death process in cardiomyocytes, cardiac fibroblasts, endothelial cells, and immune cells. It plays a crucial role in the pathogenesis of cardiac diseases that contribute to heart failure. This review intends to summarize the therapeutic implications targeting pyroptosis in the main cardiac pathologies preceding heart failure.
Collapse
Affiliation(s)
- Olive Habimana
- International College, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China
| | | | - Jinfu Peng
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China; Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China
| | - Guang-Hui Yi
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China; Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, 28, W Changsheng Road, Hengyang, Hunan, 421001, China.
| |
Collapse
|
20
|
Gong X, Li Y, He Y, Zhou F. USP7-SOX9-miR-96-5p-NLRP3 network regulates myocardial injury and cardiomyocyte pyroptosis in sepsis. Hum Gene Ther 2022; 33:1073-1090. [PMID: 35686454 DOI: 10.1089/hum.2022.078] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Sepsis is a common life-threatening pathology. This study investigated the role of transcription factor sex-determining region Y (SRY)-box 9 (SOX9) in sepsis-induced cardiomyocyte pyroptosis. A murine model of sepsis was established, followed by detection of cardiac functions and myocardial injury. HL-1 cells were induced by lipopolysaccharides (LPS). The levels of IL-18, IL-1β, TNF-α, IL-6, MDA, and SOD in myocardial tissues and HL-1 cells were determined. SOX9 ubiquitination level was measured. The binding relationships between SOX9-miR-96-5p and miR-96-5p-NLRP3 were analyzed, and the interaction between ubiquitin-specific peptidase 7 (USP7) and SOX9 was measured. SOX9 was highly expressed in septic mice and LPS-induced HL-1 cells. SOX9 silencing improved cardiac function, alleviated myocardial injury, reduced the levels of IL-1β, IL-18, cleaved caspase-1, GSDMD-N, TNF-α, IL-6, and MDA in myocardial tissues and HL-1 cells, increased the level of SOD, and alleviated cardiomyocyte pyroptosis. USP7 upregulated SOX9 expression through deubiquitination. SOX9 inhibited miR-96-5p expression and miR-96-5p targeted NLRP3. miR-96-5p silencing or USP7 overexpression reversed the inhibitory effect of SOX9 silencing on cardiomyocyte pyroptosis. Collectively, USP7 upregulated SOX9 expression through deubiquitination, and SOX9 suppressed miR-96-5p expression by binding to the miR-96-5p promoter region, thereby promoting NLRP3 expression and then exacerbating sepsis-induced myocardial injury and cardiomyocyte pyroptosis.
Collapse
Affiliation(s)
- Xinran Gong
- Sichuan Academy of Medical Sciences • Sichuan Provincial People's Hospital, Department of Anesthesiology, Chengdu City, Sichuan Province, China;
| | - Yao Li
- Sichuan Academy of Medical Sciences • Sichuan Provincial People's Hospital, Department of Anesthesiology, Chengdu City, Sichuan Province, China;
| | - Yu He
- Sichuan Academy of Medical Sciences • Sichuan Provincial People's Hospital, Department of Anesthesiology, Chengdu City, Sichuan Province, China;
| | - Fang Zhou
- Sichuan Academy of Medical Sciences • Sichuan Provincial People's Hospital, Department of Anesthesiology, 32# W.Sec 2,1st Ring Rd, Qingyang District, Chengdu City, Sichuan Province, China, 610072;
| |
Collapse
|
21
|
Wang J, Zhang F, Xu H, Yang H, Shao M, Xu S, Lyu F. TLR4 aggravates microglial pyroptosis by promoting DDX3X-mediated NLRP3 inflammasome activation via JAK2/STAT1 pathway after spinal cord injury. Clin Transl Med 2022; 12:e894. [PMID: 35692100 PMCID: PMC9189419 DOI: 10.1002/ctm2.894] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/30/2022] [Accepted: 05/09/2022] [Indexed: 12/17/2022] Open
Abstract
Background Toll‐like receptor 4 (TLR4) participates in the initiation of neuroinflammation in various neurological diseases, including central nervous system injuries. NLR family pyrin domain containing 3 (NLRP3) inflammasome‐mediated microglial pyroptosis is crucial for the inflammatory response during secondary spinal cord injury (SCI). However, the underlying mechanism by which TLR4 regulates NLRP3 inflammasome activation and microglial pyroptosis after SCI remains uncertain. Methods We established an in vivo mouse model of SCI using TLR4‐knockout (TLR4‐KO) and wild‐type (WT) mice. The levels of pyroptosis, tissue damage and neurological function recovery were evaluated in the three groups (Sham, SCI, SCI‐TLR4‐KO). To identify differentially expressed proteins, tandem mass tag (TMT)‐based proteomics was conducted using spinal cord tissue between TLR4‐KO and WT mice after SCI. For our in vitro model, mouse microglial BV2 cells were exposed to lipopolysaccharides (1 µg/ml, 8 h) and adenosine triphosphate (ATP) (5 mM, 2 h) to induce pyroptosis. A series of molecular biological experiments, including Western blot (WB), real‐time quantitative polymerase chain reaction (RT‐qPCR), enzyme‐linked immunosorbent assay (ELISA), immunofluorescence (IF), immunohistochemical (IHC), chromatin immunoprecipitation (ChIP), Dual‐Luciferase Reporter assay (DLA) and co‐immunoprecipitation (Co‐IP), were performed to explore the specific mechanism of microglial pyroptosis in vivo and in vitro. Results Our results indicated that TLR4 promoted the expression of dead‐box helicase 3 X‐linked (DDX3X), which mediated NLRP3 inflammasome activation and microglial pyroptosis after SCI. Further analysis revealed that TLR4 upregulated the DDX3X/NLRP3 axis by activating the JAK2/STAT1 signalling pathway, and importantly, STAT1 was identified as a transcription factor promoting DDX3X expression. In addition, we found that biglycan was increased after SCI and interacted with TLR4 to jointly regulate microglial pyroptosis through the JAK2/STAT1/DDX3X/NLRP3 axis after SCI. Conclusion Our study preliminarily identified a novel mechanism by which TLR4 regulates NLRP3 inflammasome‐mediated microglial pyroptosis in response to SCI—providing a novel and promising therapeutic target for SCI.
Collapse
Affiliation(s)
- Jin Wang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, P. R. China
| | - Fan Zhang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, P. R. China
| | - Haocheng Xu
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, P. R. China
| | - Haiyuan Yang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, P. R. China
| | - Minghao Shao
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, P. R. China
| | - Shun Xu
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P. R. China
| | - Feizhou Lyu
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, P. R. China.,Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P. R. China
| |
Collapse
|