1
|
Uusi-Mäkelä M, Harjula SKE, Junno M, Sillanpää A, Nätkin R, Niskanen MT, Saralahti AK, Nykter M, Rämet M. The inflammasome adaptor pycard is essential for immunity against Mycobacterium marinum infection in adult zebrafish. Dis Model Mech 2025; 18:dmm052061. [PMID: 39916610 DOI: 10.1242/dmm.052061] [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: 08/14/2024] [Accepted: 02/03/2025] [Indexed: 03/25/2025] Open
Abstract
Inflammasomes regulate the host response to intracellular pathogens including mycobacteria. We have previously shown that the course of Mycobacterium marinum infection in adult zebrafish (Danio rerio) mimics the course of tuberculosis in human. To investigate the role of the inflammasome adaptor pycard in zebrafish M. marinum infection, we produced two zebrafish knockout mutant lines for the pycard gene with CRISPR/Cas9 mutagenesis. Although the zebrafish larvae lacking pycard developed normally and had unaltered resistance against M. marinum, the loss of pycard led to impaired survival and increased bacterial burden in the adult zebrafish. Based on histology, immune cell aggregates, granulomas, were larger in pycard-deficient fish than in wild-type controls. Transcriptome analysis with RNA sequencing of a zebrafish haematopoietic tissue, kidney, suggested a role for pycard in neutrophil-mediated defence, haematopoiesis and myelopoiesis during infection. Transcriptome analysis of fluorescently labelled, pycard-deficient kidney neutrophils identified genes that are associated with compromised resistance, supporting the importance of pycard for neutrophil-mediated immunity against M. marinum. Our results indicate that pycard is essential for resistance against mycobacteria in adult zebrafish.
Collapse
Affiliation(s)
- Meri Uusi-Mäkelä
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
| | | | - Maiju Junno
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
| | - Alina Sillanpää
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
| | - Reetta Nätkin
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, FI-33521 Tampere, Finland
| | | | | | - Matti Nykter
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, FI-33521 Tampere, Finland
| | - Mika Rämet
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
| |
Collapse
|
2
|
Lim EY, Kim GD, Kim HJ, Eom JE, Song HJ, Shin DU, Kim YI, Kim HJ, Lee SY, Shin HS. Cirsium japonicum leaf extract attenuated lipopolysaccharide-induced acute respiratory distress syndrome in mice via suppression of the NLRP3 and HIF1α pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156601. [PMID: 40064116 DOI: 10.1016/j.phymed.2025.156601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/20/2024] [Accepted: 03/01/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a severe inflammatory disorder characterized by acute respiratory failure, alveolar barrier dysfunction, edema, and dysregulated alveolar macrophage-mediated pulmonary inflammation. Despite advancements in treatment strategies, the mortality rate in patients with ARDS remains high, ranging from 40-60 %. Current approaches are limited to supportive care, necessitating the exploration of effective therapeutic options such as suppressing broad inflammatory responses. Although Cirsium japonicum leaves possess anti-inflammatory properties, their specific effects on ARDS have not yet been investigated. METHODS The anti-inflammatory activity of Cirsium japonicum extract (CJE) was investigated in a lipopolysaccharide (LPS)-induced ARDS model. RESULTS CJE significantly attenuated LPS-induced lung injury, including reduced alveolar wall thickness, inflammatory cell infiltration, proteinaceous debris, and hyaline membranes. Moreover, CJE repressed infiltration of inflammatory cells and pro-inflammatory gene expression in bronchoalveolar lavage fluid. Concordantly, CJE mitigated alveolar macrophage activation, which consequently reduced neutrophil chemoattractic infiltration. Additionally, CJE suppressed NLRP3 and HIF1α expression in the lungs of the ARDS mouse. Similarly, LPS-induced NLRP3 and HIF1α pathway-associated inflammatory and glycolytic gene expressions significantly diminished by CJE in murine alveolar macrophage cell line, MH-S cells, and bone marrow-derived macrophages. CONCLUSION CJE suppressed multiple inflammatory responses through the regulation of NLRP3 and HIF1α signaling-related gene expression in macrophages of LPS-induced ARDS mice. These results suggest that CJE has therapeutic potential for treating patients with ARDS via macrophage regulation.
Collapse
Affiliation(s)
- Eun Yeong Lim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea
| | - Gun-Dong Kim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea
| | - Ha-Jung Kim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea
| | - Ji-Eun Eom
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea
| | - Hyeon-Ji Song
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea; Department of Food Science and Technology, Jeonbuk National University, Jeonju 54896, South Korea
| | - Dong-Uk Shin
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea
| | - Young In Kim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea
| | - Hyun-Jin Kim
- Department of Food Science and Technology, Gyeongsang National University, Jinju 52828, South Korea
| | - So-Young Lee
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea; Department of Food Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, South Korea
| | - Hee Soon Shin
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea; Department of Food Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, South Korea.
| |
Collapse
|
3
|
Xu X, Wang Y, Pei K, Mao C, Fang F, Zhou T, Zhang M, Meng PN, Wei Z, Liu C, Dai Y, Yin R, Chen Z, Wang X. Shengmai-Yin resists myocardial ischemia reperfusion injury by inhibiting K27 ubiquitination of absent in melanoma 2. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119553. [PMID: 40010555 DOI: 10.1016/j.jep.2025.119553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/11/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Myocardial ischemia-reperfusion (I/R) injury stands as a significant contributor to cardiovascular disease. Shengmai-Yin (SMY), a traditional Chinese medicine, is widely used in myocardial infarct treatment. However, the specific mechanism of SMY in treating myocardial I/R injury is currently limited. AIM OF STUDY The study aimed to investigate the therapeutic efficacy of SMY in addressing myocardial I/R injury and elucidate its specific mechanisms. MATERIALS AND METHODS The active components of SMY were quantified using Ultra-high performance liquid chromatography-MS/MS (UPLC-MS/MS). Sprague-Dawley (SD) rats were treated with SMY post-I/R model establishment. Cardiac injury was assessed by heart weight to body weight ratio. Left ventricular function and infarct volume were evaluated using ultrasound cardiography and TTC staining. Tissue lesions were examined via hematoxylin-eosin (HE) and Sirius Red staining. Co-Immunoprecipitation (Co-IP) technology explored absent in melanoma 2 (AIM2) and K27 Ubiquitination Modification (K27-Ub) interactions. Immunofluorescence staining detected Apoptosis-associated Speck-like Protein containing a CARD (ASC) and AIM2 co-localization. Adeno-associated Virus (AAV) was used to upregulate AIM2 levels, while Shikonin was used to downregulate AIM2, to explore its roles in SMY's therapeutic effects on I/R injury. RESULTS SMY can reduce infarct size and enhance cardiac function. Furthermore, SMY can inhibit tissue fibrosis. Fibrosis markers and proinflammatory factors were reduced after SMY treatment. Serum levels of Lactate Dehydrogenase (LDH) and Creatine Kinase -MB (CK-MB) were also decreased. Mechanistically, SMY inhibits the activation of the AIM2 inflammasome by downregulating the K27 ubiquitination of AIM2. Overexpression of AIM2 reversed the anti-I/R effect of SMY, suggesting that AIM2 plays a crucial role in I/R injury. The AIM2 inhibitor counteracts the therapeutic effect of SMY. CONCLUSION SMY inhibits the K27 ubiquitination modification of AIM2 and inhibits the activation of AIM2 inflammasomes after myocardial I/R injury.
Collapse
Affiliation(s)
- Xiaojin Xu
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China; Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine, Shanghai, 201203, China.
| | - Yuanyi Wang
- School of Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China.
| | - Ke Pei
- School of Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China.
| | - Chenhan Mao
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China.
| | - Fei Fang
- School of Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China.
| | - Tiantong Zhou
- Acupuncture and Moxibustion Massage College Health and Rehabilitation College, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China.
| | - Meng Zhang
- Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine, Shanghai, 201203, China.
| | - Pei-Na Meng
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Zilun Wei
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute Cardiovascular Diseases, Shanghai, China.
| | - Chang Liu
- School of Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China.
| | - Yang Dai
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China.
| | - Rui Yin
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China.
| | - Zhaoyang Chen
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China.
| | - Xindong Wang
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China.
| |
Collapse
|
4
|
Liang JY, Yuan XL, Jiang JM, Zhang P, Tan K. Targeting the NLRP3 inflammasome in Parkinson's disease: From molecular mechanism to therapeutic strategy. Exp Neurol 2025; 386:115167. [PMID: 39884329 DOI: 10.1016/j.expneurol.2025.115167] [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: 12/07/2024] [Revised: 01/13/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Parkinson's disease is the second most common neurodegenerative disease, characterized by substantial loss of dopaminergic (DA) neurons, the formation of Lewy bodies (LBs) in the substantia nigra, and pronounced neuroinflammation. The nucleotide-binding domain like leucine-rich repeat- and pyrin domain-containing protein 3 (NLRP3) inflammasome is one of the pattern recognition receptors (PRRs) that function as intracellular sensors in response to both pathogenic microbes and sterile triggers associated with Parkinson's disease. These triggers include reactive oxygen species (ROS), misfolding protein aggregation, and potassium ion (K+) efflux. Upon activation, it recruits and activates caspase-1, then processes the pro-inflammatory cytokines interleukin-1β (IL-1β) and IL-18, which mediate neuroinflammation in Parkinson's disease. In this review, we provide a comprehensive overview of NLRP3 inflammasome, detailing its structure, activation pathways, and the factors that trigger its activation. We also explore the pathological mechanisms by which NLRP3 contributes to Parkinson's disease and discuss potential strategies for targeting NLRP3 as a therapeutic approach.
Collapse
Affiliation(s)
- Jin-Yu Liang
- Department of Clinical Laboratory Medicine, Zhuzhou Kind Cardiovascular Disease Hospital, Hunan Province, China
| | - Xiao-Lei Yuan
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Jia-Mei Jiang
- Institute of Neurology, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan, PR China
| | - Ping Zhang
- Department of Neurology, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan, PR China
| | - Kuang Tan
- Department of Clinical Laboratory Medicine, Zhuzhou Kind Cardiovascular Disease Hospital, Hunan Province, China.
| |
Collapse
|
5
|
Huang Q, Shan Q, Ma F, Li S, Sun P. Chlorogenic acid mitigates heat stress-induced oxidative damage in bovine mammary epithelial cells by inhibiting NF-κB-mediated NLRP3 inflammasome activation via upregulating the Nrf2 signaling pathway. Int J Biol Macromol 2025; 301:140133. [PMID: 39842566 DOI: 10.1016/j.ijbiomac.2025.140133] [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: 08/22/2024] [Revised: 01/16/2025] [Accepted: 01/19/2025] [Indexed: 01/24/2025]
Abstract
Chlorogenic acid (CGA), a polyphenolic bioactive molecule derived from medicinal plants, is known for its strong antioxidant and anti-inflammatory properties. Previous studies have demonstrated that dietary supplementation with Lonicera japonica extract, rich in CGA, effectively enhances the production performance of lactating dairy cows under heat stress (HS) conditions. However, the molecular mechanisms underlying CGA's protective effects remain unclear. This study aims to elucidate the mechanisms by which CGA alleviates HS-induced oxidative damage in bovine mammary epithelial cells (bMECs), focusing on its pharmacological activity and potential application as a natural therapeutic agent for bovine mammary disorders. The results demonstrated that HS activates the NF-κB and NLRP3 signaling pathways by increasing ROS generation, leading to oxidative stress and inflammatory response in bMECs. CGA mitigates these effects by scavenging intracellular ROS, activating the Nrf2 signaling pathway, and inhibiting key molecules in the NF-κB and NLRP3 signaling pathways. This study provides new insights into the underlying molecular mechanisms of CGA's protective effects, highlighting its potential as a natural antioxidant for bovine mammary health and contributing to the broader application of polyphenolic compounds in managing oxidative stress and inflammation.
Collapse
Affiliation(s)
- Qi Huang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Qiang Shan
- State Key Laboratory of Animal Nutrition and Feeding, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Fengtao Ma
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; State Key Laboratory of Animal Nutrition and Feeding, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shengli Li
- State Key Laboratory of Animal Nutrition and Feeding, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Peng Sun
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
6
|
Zhang XJ, Yang PY, Ding L, Wang J, Li XL, Xiao WL. Isolicoflavonol alleviates UVB-induced photodamage via protecting mitochondria and blocking the activation of NLRP3 inflammasome. Toxicol Appl Pharmacol 2025; 497:117262. [PMID: 39929282 DOI: 10.1016/j.taap.2025.117262] [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: 12/02/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 02/20/2025]
Abstract
Photodamage, a type of skin inflammation caused by excessive exposure to solar radiation, leads to skin redness, inflammation, and even the development of skin cancer, posing a severe threat to individuals living at high altitudes. UVB radiation is considered the primary factor contributing to photodamage. It stimulates macrophages within the epidermis, triggers inflammasome activation, and increases the inflammatory cytokine interleukin-1β (IL-1β) production. This study examined the protective effects of the compound isolicoflavonol (ILF) and its mechanism against UVB-induced photodamage. We irradiated UVB to create a photodamage model in mice and macrophages. Next, we assessed ILF's ability to protect the skin and cells from UVB photodamage and its inhibitory effects on UVB-mediated NLRP3 inflammasome. Our findings indicated that ILF reduced UVB-induced skin injury and inflammation in mouse skin, decreased cell death, NLRP3 inflammasome activation, ROS production, and mitochondrial dysfunction. These results suggest that ILF may be a potent agent for protecting the skin against UVB-induced photodamage.
Collapse
Affiliation(s)
- Xing-Jie Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China
| | - Peng-Yun Yang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China
| | - Ling Ding
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China
| | - Jun Wang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China
| | - Xiao-Li Li
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China.
| | - Wei-Lie Xiao
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China; Southwest United Graduate School, Kunming 650500, Yunnan, China.
| |
Collapse
|
7
|
Huang X, Niu M, Sun T, Li M, Jiang X, Duan H, Zhang T, Zhang J, Xie F, Song R, Yu A. X-ray irradiation reduces ATP-dependent activation of NLRP3 inflammasome by inhibiting TWIK2 activity in macrophages. Immunol Lett 2025; 272:106967. [PMID: 39732203 DOI: 10.1016/j.imlet.2024.106967] [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: 01/18/2024] [Revised: 11/30/2024] [Accepted: 12/25/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND The spleen, as the body's largest peripheral immune organ and a crucial source of circulating monocytes, plays a significant role in the acute inflammatory response of spleen-derived macrophages to diseases. Therefore, studying the impact and mechanism of X-ray irradiation on spleen-derived macrophages' inflammatory responses is of great importance. METHOD Extracted and identified mice splenic macrophages were divided into four groups: control group, LPS and ATP co-stimulated non-irradiated group, LPS and ATP co-stimulated group irradiated after 6 h, and LPS and ATP co-stimulated group irradiated after 12 h In the LPS and ATP co-stimulated groups, LPS (1μg/ml) and ATP (5mmol/L) were added to establish an inflammatory model in mice splenic macrophages. The irradiated groups were exposed to a medical linear accelerator (Elekta Synergy), while the non-irradiated groups were placed under the light source for the same duration without irradiation. Protein extraction was performed in each group at 6 h and 12 h post-treatment for subsequent analysis using Western blot, ELISA, RT-qPCR and other relevant methods. RESULTS (1) Compared with the non-irradiated group, the cell activity in the groups irradiated for 6 h and 12 h at 8 Gy showed a significant increase (P<0.01). (2) In the LPS and ATP co-stimulated groups irradiated after 6 h and 12 h, the expression of NLRP3 mRNA and protein, IL-18 and IL-1β showed a notable decrease compared to the LPS and ATP co-stimulated non-irradiated group (P<0.05). Additionally, caspase-1 expression of caspase-1 mRNA and protein in the 12 h post-irradiation group also decreased considerably when compared with the LPS and ATP co-stimulated non-irradiated group (P < 0.05). In the groups irradiated after 6 h and 12 h, (3) there was a remarkable decrease in the expression of TWIK mRNA and TWIK2, (4) as well as Gq mRNA and protein, when compared to the LPS and ATP co-stimulated non-irradiated group (P < 0.05). Particularly, the 12 h post-irradiation group exhibited a notable reduction in PKC expression (P < 0.05). CONCLUSION X-ray irradiation is capable of inhibiting the activation of ATP-dependent NLRP3 inflammasomes in splenic macrophages.
Collapse
Affiliation(s)
- Xiaofei Huang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Man Niu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China; Department of Emergency, Fourth Hospital of Shijiazhuang, 050035 Shijiazhuang, Hebei, China
| | - Tianjing Sun
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Mo Li
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Xuheng Jiang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Haizhen Duan
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Tianxi Zhang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Ji Zhang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Fangke Xie
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Renjie Song
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Anyong Yu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China.
| |
Collapse
|
8
|
Bai Y, Pan Y, Liu X. Mechanistic insights into gasdermin-mediated pyroptosis. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00837-0. [PMID: 40128620 DOI: 10.1038/s41580-025-00837-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2025] [Indexed: 03/26/2025]
Abstract
Pyroptosis, a novel mode of inflammatory cell death, is executed by membrane pore-forming gasdermin (GSDM) family members in response to extracellular or intracellular injury cues and is characterized by a ballooning cell morphology, plasma membrane rupture and the release of inflammatory mediators such as interleukin-1β (IL-1β), IL-18 and high mobility group protein B1 (HMGB1). It is a key effector mechanism for host immune defence and surveillance against invading pathogens and aberrant cancerous cells, and contributes to the onset and pathogenesis of inflammatory and autoimmune diseases. Manipulating the pore-forming activity of GSDMs and pyroptosis could lead to novel therapeutic strategies. In this Review, we discuss the current knowledge regarding how GSDM-mediated pyroptosis is initiated, executed and regulated, its roles in physiological and pathological processes, and the crosstalk between different modes of programmed cell death. We also highlight the development of drugs that target pyroptotic pathways for disease treatment.
Collapse
Affiliation(s)
- Yang Bai
- Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Youdong Pan
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xing Liu
- Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China.
- Shanghai Academy of Natural Sciences (SANS), Shanghai, China.
| |
Collapse
|
9
|
Liu Y, Wang Q, Ma J, Li J, Li C, Xie X, Xiao Q, Xie C, Liu H, Hong Y, Wang J. Discovery of Novel Sulfonylurea NLRP3 Inflammasome Inhibitor for the Treatment of Multiple Inflammatory Diseases. J Med Chem 2025. [PMID: 40112040 DOI: 10.1021/acs.jmedchem.4c02813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
NLRP3 inflammasome is critical in innate immunity and inflammatory responses. A series of novel sulfonylurea-based NLRP3 inflammasome inhibitors was designed and synthesized. Notably, compound 15 exhibited the potent NLRP3 inhibitory activity, effectively suppressing IL-1β secretion in THP-1 (IC50 = 23 nM), demonstrating better efficacy compared to MCC950. It selectively inhibits NLRP3 activation by disrupting inflammasome assembly, with no effect on NLRC4 or AIM2 inflammasomes. Molecular docking showed that the 1-methyl-4-(methylamino)piperidine moiety forms a novel hydrogen bond with Asp662 in the hydrophilic region of NLRP3. Additionally, compound 15 displayed excellent pharmacokinetic properties with 99.6% oral bioavailability in mice. It exhibited superior efficacy in acute peritonitis and diabetic kidney disease models, surpassing MCC950. Tissue distribution studies confirmed that compound 15 specifically targeted the gut and showed efficacy in an IBD model, comparable to MCC950. These findings highlight compound 15 as a promising lead for novel oral NLRP3 inflammasome inhibitors.
Collapse
Affiliation(s)
- Yiting Liu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Qinxue Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | | | - Jiyuan Li
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Cuina Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiong Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiannan Xiao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Cen Xie
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hong Liu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Hong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiang Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Lingang Laboratory, Shanghai 200031, China
| |
Collapse
|
10
|
Beesetti S. Ubiquitin Ligases in Control: Regulating NLRP3 Inflammasome Activation. FRONT BIOSCI-LANDMRK 2025; 30:25970. [PMID: 40152367 DOI: 10.31083/fbl25970] [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: 08/02/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 03/29/2025]
Abstract
Ubiquitin ligases play pivotal roles in the regulation of NLR family pyrin domain containing 3 (NLRP3) inflammasome activation, a critical process in innate immunity and inflammatory responses. This review explores the intricate mechanisms by which various E3 ubiquitin ligases exert both positive and negative influences on NLRP3 inflammasome activity through diverse post-translational modifications. Negative regulation of NLRP3 inflammasome assembly is mediated by several E3 ligases, including F-box and leucine-rich repeat protein 2 (FBXL2), tripartite motif-containing protein 31 (TRIM31), and Casitas B-lineage lymphoma b (Cbl-b), which induce K48-linked ubiquitination of NLRP3, targeting it for proteasomal degradation. Membrane-associated RING-CH 7 (MARCH7) similarly promotes K48-linked ubiquitination leading to autophagic degradation, while RING finger protein (RNF125) induces K63-linked ubiquitination to modulate NLRP3 function. Ariadne homolog 2 (ARIH2) targets the nucleotide-binding domain (NBD) domain of NLRP3, inhibiting its activation, and tripartite motif-containing protein (TRIM65) employs dual K48 and K63-linked ubiquitination to suppress inflammasome assembly. Conversely, Pellino2 exemplifies a positive regulator, promoting NLRP3 inflammasome activation through K63-linked ubiquitination. Additionally, ubiquitin ligases influence other components critical for inflammasome function. TNF receptor-associated factor 3 (TRAF3) mediates K63 polyubiquitination of apoptosis-associated speck-like protein containing a CARD (ASC), facilitating its degradation, while E3 ligases regulate caspase-1 activation and DEAH-box helicase 33 (DHX33)-NLRP3 complex formation through specific ubiquitination events. Beyond direct inflammasome regulation, ubiquitin ligases impact broader innate immune signaling pathways, modulating pattern-recognition receptor responses and dendritic cell maturation. Furthermore, they intricately control NOD1/NOD2 signaling through K63-linked polyubiquitination of receptor-interacting protein 2 (RIP2), crucial for nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) activation. Furthermore, we explore how various pathogens, including bacteria, viruses, and parasites, have evolved sophisticated strategies to hijack the host ubiquitination machinery, manipulating NLRP3 inflammasome activation to evade immune responses. This comprehensive analysis provides insights into the molecular mechanisms underlying inflammasome regulation and their implications for inflammatory diseases, offering potential avenues for therapeutic interventions targeting the NLRP3 inflammasome. In conclusion, ubiquitin ligases emerge as key regulators of NLRP3 inflammasome activation, exhibiting a complex array of functions that finely tune immune responses. Understanding these regulatory mechanisms not only sheds light on fundamental aspects of inflammation but also offers potential therapeutic avenues for inflammatory disorders and infectious diseases.
Collapse
Affiliation(s)
- Swarna Beesetti
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
11
|
Seo HY, Park JY, Lee SH, Lee HW, Han E, Hwang JS, Kim MK, Jang BK. Clusterin deficiency exacerbates cholestatic liver disease through ER stress and NLRP3 inflammasome activation. Cell Biosci 2025; 15:36. [PMID: 40089787 PMCID: PMC11909925 DOI: 10.1186/s13578-025-01376-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/04/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Cholestatic liver disease, characterized by impaired bile flow, leads to the accumulation of harmful metabolites and toxins, resulting in liver damage. Inflammatory cytokines are crucial for the progression of this condition. Clusterin is a glycoprotein with roles in cell death, lipid transport, and cellular protection. We previously demonstrated that clusterin protects against hepatic steatosis and hepatic fibrosis. This study explored the roles of clusterin in cholestatic liver injury induced by a DDC (3,5-diethoxycarbonyl-1,4-dihydrocollidine) diet. METHODS The study evaluated the impact of clusterin on liver injury in C57BL/6 mice and clusterin-knockout (KO) mice fed a DDC diet for 10-20 days. Primary Kupffer cells (KCs) and hepatocytes (HCs) of these mice were analyzed. Techniques such as Sirius red staining, immunohistochemistry, real-time RT-PCR, enzyme-linked immunosorbent assays, and western blotting were performed to assess the effects of clusterin. RESULTS Clusterin expression was upregulated in the cholestatic liver. Clusterin-KO mice exhibited elevated levels of alanine aminotransferase, aspartate aminotransferase, collagen, and αSMA upon DDC diet-induced liver injury. They also had increased levels of markers of endoplasmic reticulum (ER) stress (CHOP, ATF6, and p-eIF2α) and inflammasome activity (NLRP3, ASC, caspase-1, and interleukin 1 beta (IL1β) protein expression, and IL1β and interleukin 18 secretion). Thapsigargin, an ER stress inducer, heightened NLRP3 inflammasome activation in primary KCs and HCs, which was mitigated by overexpression of clusterin. CONCLUSIONS The absence of clusterin exacerbates ER stress and NLRP3 inflammasome activation in mice fed a DDC diet. Conversely, overexpression of clusterin suppresses these stress responses. Thus, clusterin deficiency is associated with an enhanced inflammasome response in the liver that is linked to upregulation of ER stress.
Collapse
Affiliation(s)
- Hye-Young Seo
- Department of Internal Medicine, School of Medicine, Institute for Medical Science, Keimyung University, Daegu, Korea
| | - Ji Yeon Park
- Department of Internal Medicine, School of Medicine, Institute for Medical Science, Keimyung University, Daegu, Korea
| | - So-Hee Lee
- Department of Internal Medicine, School of Medicine, Institute for Medical Science, Keimyung University, Daegu, Korea
| | - Hye Won Lee
- Department of Pathology, Keimyung University School of Medicine, Daegu, Korea
| | - Eugene Han
- Department of Internal Medicine, School of Medicine, Institute for Medical Science, Keimyung University, Daegu, Korea
| | - Jae Seok Hwang
- Department of Internal Medicine, School of Medicine, Institute for Medical Science, Keimyung University, Daegu, Korea
| | - Mi Kyung Kim
- Department of Internal Medicine, School of Medicine, Institute for Medical Science, Keimyung University, Daegu, Korea.
| | - Byoung Kuk Jang
- Department of Internal Medicine, School of Medicine, Institute for Medical Science, Keimyung University, Daegu, Korea.
| |
Collapse
|
12
|
Wāng Y, Wang C, Jiang Y, Wang T, Wu T, Tang M. Carbonaceous cores serve as surrogates for environmental particulate matter inducing vascular endothelial inflammation via inflammasome activation. JOURNAL OF HAZARDOUS MATERIALS 2025; 486:137011. [PMID: 39736255 DOI: 10.1016/j.jhazmat.2024.137011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/01/2025]
Abstract
Ambient particulate matter (PM) exposure is a known risk factor for cardiovascular diseases. Epidemiological studies have shown the association between PM exposure and vascular complications, including vasculitis, embolism, hypertension, stroke, and atherosclerosis. However, the exact mechanisms underlying its vascular toxicity, especially in relation to short-term exposures, remain incompletely understood. This study investigates the role of PM and its carbonaceous cores in driving vascular endothelial inflammation via inflammasome activation. We hypothesized that PM SRM1648a exposure induces vascular endothelial inflammation through oxidative stress and inflammasome activation. Short-term exposure to PM SRM1648a was assessed in BALB/c mice for systemic inflammation and oxidative stress biomarkers, alongside in vitro studies in HUVECs and EA.hy926 endothelial cells to elucidate inflammasome activation pathways. PM SRM1648a exposure significantly altered redox balance and cytokine profiles in mice and upregulated NLRP3/NLRC4 inflammasomes in vascular endothelial cells, leading to caspase-1/IL-1β activation. Intriguingly, pyroptosis was not the primary mode of cell death. In vitro studies demonstrated that antioxidants glutathione monoethyl ester effectively mitigated oxidative stress and inflammasome activation in endothelial cells. This study highlights the critical role of ROS-mediated inflammasome activation in vascular inflammation induced by PM SRM1648a, with carbon-based cores as key contributors.
Collapse
Affiliation(s)
- Yán Wāng
- Key laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China.
| | - Chunzhi Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Yang Jiang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Tian Wang
- School of Public Health, Anhui University of Science and Technology, Hefei, Anhui 231100, China
| | - Tianshu Wu
- Key laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Meng Tang
- Key laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
13
|
Shen DM, Byth KF, Bertheloot D, Braams S, Bradley S, Dean D, Dekker C, El-Kattan AF, Franchi L, Glick GD, Ghosh S, Hinniger A, Katz JD, Kitanovic A, Lu X, Olhava EJ, Opipari AW, Sanchez B, Seidel HM, Stunden J, Stutz A, Telling A, Venkatraman S, Winkler DG, Roush WR. Discovery of DFV890, a Potent Sulfonimidamide-Containing NLRP3 Inflammasome Inhibitor. J Med Chem 2025; 68:5529-5550. [PMID: 40036600 DOI: 10.1021/acs.jmedchem.4c02759] [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: 03/06/2025]
Abstract
The discovery of DFV890 ((R)-1), a potent and selective NLRP3 antagonist, is described. Replacement of the sulfonyl urea core from the first-generation NLRP3 antagonist CRID3 with a sulfonimidamide core afforded a novel and potent series of NLRP3 antagonists. The (R)-enantiomers of the sulfonimidamide series were found to be consistently more potent than structurally related sulfonyl ureas. Replacement of the furan unit of CRID3 with a 5-substituted thiazole unit led to DFV890 ((R)-1), which potently inhibited IL-1β production in THP-1 cells and in primary human cells, blocked multiple downstream effectors of NLRP3 activation, and substantially improved PK properties and significantly lowered the predicted human dose compared to that for CRID3. DFV890 ((R)-1) was also effective in an air pouch model of gout.
Collapse
Affiliation(s)
- Dong-Ming Shen
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | - Kate F Byth
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | | | | | - Sarah Bradley
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | - Dennis Dean
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | - Carien Dekker
- Novartis Biomedical Research, Basel CH-4002, Switzerland
| | | | - Luigi Franchi
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | - Gary D Glick
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | - Shomir Ghosh
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | | | - Jason D Katz
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | | | - Xiaokang Lu
- IFM Therapeutics, Ann Arbor, Michigan 48108, United States
| | - Edward J Olhava
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | | | - Brian Sanchez
- IFM Therapeutics, Ann Arbor, Michigan 48108, United States
| | - H Martin Seidel
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | | | | | - Alissa Telling
- IFM Therapeutics, Ann Arbor, Michigan 48108, United States
| | | | - David G Winkler
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | - William R Roush
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| |
Collapse
|
14
|
Coats SR, Su TH, Luderman Miller Z, King AJ, Ortiz J, Reddy A, Alaei SR, Jain S. Porphyromonas gingivalis outer membrane vesicles divert host innate immunity and promote inflammation via C4' monophosphorylated lipid A. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkae050. [PMID: 40131356 DOI: 10.1093/jimmun/vkae050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/05/2024] [Indexed: 03/27/2025]
Abstract
Porphyromonas gingivalis (Pg) is a prevalent pathogen that promotes human periodontal disease (PD) and exacerbates systemic comorbidities such as atherosclerosis, rheumatoid arthritis, and Alzheimer's disease. Pg produces nonphosphorylated tetra-acylated lipid A (NPLA) in its outer membrane (OM) that evades host Toll-like receptor 4 (TLR4), inflammasome pathways, and cationic peptides, enhancing bacterial survival. Here, we show that Pg also releases outer membrane vesicles (OMVs) that engage and divert host cell TLR4, inflammasome, and LL-37 responses away from the microbe. We determined that Pg OMVs are enriched for C4' monophosphoryl lipid A (C4'-MPLA), an established agonist for TLR4-TRIF-IFNβ and inflammasome-IL-1β responses. Comparisons of Pg 381 and Pg 33277 stationary phase cultures revealed higher OMV production by Pg 381, which correlates with its higher proinflammatory pathogenicity. The cationic peptide, polymyxin B (PMB), which selectively binds lipid A C4'-phosphate, reduces OMV-stimulated HEK cell TLR4 activation and THP-1 cell IL-1β production, confirming the proinflammatory role for OMV-C4'-MPLA. Similar to PMB, the host defense peptide, LL-37, inhibits OMV-C4'-MPLA-dependent HEK cell TLR4 activation. PMB and LL-37 also blocked OMV-C4'-MPLA-driven TLR4 activation in human umbilical vein endothelial cells. Finally, wild-type Pg-containing OM-NPLA is highly resistant to LL-37 antimicrobial activity, whereas the ΔlpxF mutant bacterium, retaining OM-C4'-MPLA, is killed by the peptide. In summary, Pg escapes host TLR4 signaling, inflammasome activation, and LL-37 interaction by retaining immunoevasive OM-NPLA. Moreover, Pg dispenses proinflammatory OMV-C4'-MPLA, which engages and redirects those host defenses. We suggest that OMV-C4'-MPLA triggers elevated IFNβ and IL-1β cytokines, which typify PD comorbidities, and drive PD-related alveolar bone loss.
Collapse
Affiliation(s)
- Stephen R Coats
- Department of Periodontics, University of Washington School of Dentistry, Seattle, WA, United States
| | - Thet Hnin Su
- Department of Periodontics, University of Washington School of Dentistry, Seattle, WA, United States
| | - Zoe Luderman Miller
- Department of Periodontics, University of Washington School of Dentistry, Seattle, WA, United States
| | - Alisa J King
- Sciences and Mathematics Division, School of Interdisciplinary Arts and Sciences, University of Washington Tacoma, Tacoma, WA, United States
| | - Joshua Ortiz
- Sciences and Mathematics Division, School of Interdisciplinary Arts and Sciences, University of Washington Tacoma, Tacoma, WA, United States
| | - Angel Reddy
- Sciences and Mathematics Division, School of Interdisciplinary Arts and Sciences, University of Washington Tacoma, Tacoma, WA, United States
| | - Sarah R Alaei
- Sciences and Mathematics Division, School of Interdisciplinary Arts and Sciences, University of Washington Tacoma, Tacoma, WA, United States
| | - Sumita Jain
- Department of Periodontics, University of Washington School of Dentistry, Seattle, WA, United States
| |
Collapse
|
15
|
Barrett TJ, Schlamp F, Muller M, Lee AH, Cornwell MG, Luttrell Williams E, Smilowitz NR, Hochman J, Ruggles KV, Reynolds HR, Berger JS. Myocardial Infarction Platelet Gene Expression Signatures in Women. JACC Basic Transl Sci 2025; 10:307-322. [PMID: 40139873 DOI: 10.1016/j.jacbts.2024.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 03/29/2025]
Abstract
Although platelets play a critical pathogenic role in myocardial infarction (MI), few studies have characterized the MI platelet transcriptome in the acute or chronic setting in women. We report that transcripts associated with the actin cytoskeleton, Rho family GTPases, mitochondrial dysfunction, and inflammatory signaling are enriched in platelets from MI patients in the acute setting (n = 40, MI; n = 38, control) and do not significantly change over time. Furthermore, 79 platelet genes chronically elevated or suppressed after MI are associated with future cardiovascular events in an independent high-risk cohort (n = 135). Compared with women with MI with nonobstructive coronary arteries, platelets from women with MI and obstructive coronary artery disease were enriched in neutrophil activation and proinflammatory signaling pathways driven by increased tumor necrosis factor (TNF)-α signaling. Hierarchic clustering of the MI transcriptomic profile identified 3 subgroups with distinctive biological pathways and MI correlates. Our data demonstrate that platelets from MI patients are phenotypically different from MI-naïve patients in the acute and chronic settings and reveal a platelet transcriptomic signature with distinct clinical features.
Collapse
Affiliation(s)
- Tessa J Barrett
- New York University Grossman School of Medicine, New York, New York, USA; Sarah Ross Soter Center for Women's Cardiovascular Research, NYU Grossman School of Medicine, New York, New York, USA.
| | - Florencia Schlamp
- New York University Grossman School of Medicine, New York, New York, USA; Sarah Ross Soter Center for Women's Cardiovascular Research, NYU Grossman School of Medicine, New York, New York, USA
| | - Matthew Muller
- New York University Grossman School of Medicine, New York, New York, USA; Institute for Systems Genetics, New York University Grossman School of Medicine, New York, New York, USA
| | - Angela H Lee
- New York University Grossman School of Medicine, New York, New York, USA
| | - Macintosh G Cornwell
- New York University Grossman School of Medicine, New York, New York, USA; Institute for Systems Genetics, New York University Grossman School of Medicine, New York, New York, USA
| | - Elliot Luttrell Williams
- New York University Grossman School of Medicine, New York, New York, USA; Sarah Ross Soter Center for Women's Cardiovascular Research, NYU Grossman School of Medicine, New York, New York, USA
| | - Nathaniel R Smilowitz
- New York University Grossman School of Medicine, New York, New York, USA; Sarah Ross Soter Center for Women's Cardiovascular Research, NYU Grossman School of Medicine, New York, New York, USA
| | - Judith Hochman
- New York University Grossman School of Medicine, New York, New York, USA; Sarah Ross Soter Center for Women's Cardiovascular Research, NYU Grossman School of Medicine, New York, New York, USA
| | - Kelly V Ruggles
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, New York, USA
| | - Harmony R Reynolds
- New York University Grossman School of Medicine, New York, New York, USA; Sarah Ross Soter Center for Women's Cardiovascular Research, NYU Grossman School of Medicine, New York, New York, USA
| | - Jeffrey S Berger
- New York University Grossman School of Medicine, New York, New York, USA; Sarah Ross Soter Center for Women's Cardiovascular Research, NYU Grossman School of Medicine, New York, New York, USA.
| |
Collapse
|
16
|
Feng S, Wierzbowski MC, Hrovat-Schaale K, Dumortier A, Zhang Y, Zyulina M, Baker PJ, Reygaerts T, Steiner A, De Nardo D, Narayanan DL, Milhavet F, Pinzon-Charry A, Arostegui JI, Khubchandani RP, Geyer M, Boursier G, Masters SL. Mechanisms of NLRP3 activation and inhibition elucidated by functional analysis of disease-associated variants. Nat Immunol 2025; 26:511-523. [PMID: 39930093 PMCID: PMC11876074 DOI: 10.1038/s41590-025-02088-9] [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] [Received: 02/27/2024] [Accepted: 01/13/2025] [Indexed: 02/16/2025]
Abstract
The NLRP3 inflammasome is a multiprotein complex that mediates caspase-1 activation and the release of proinflammatory cytokines, including interleukin (IL)-1β and IL-18. Gain-of-function variants in the gene encoding NLRP3 (also called cryopyrin) lead to constitutive inflammasome activation and excessive IL-1β production in cryopyrin-associated periodic syndromes (CAPS). Here we present functional screening and automated analysis of 534 NLRP3 variants from the international INFEVERS registry and the ClinVar database. This resource captures the effect of NLRP3 variants on ASC speck formation spontaneously, at low temperature, after inflammasome stimulation and with the specific NLRP3 inhibitor MCC950. Most notably, our analysis facilitated the updated classification of NLRP3 variants in INFEVERS. Structural analysis suggested multiple mechanisms by which CAPS variants activate NLRP3, including enhanced ATP binding, stabilizing the active NLRP3 conformation, destabilizing the inactive NLRP3 complex and promoting oligomerization of the pyrin domain. Furthermore, we identified pathogenic variants that can hypersensitize the activation of NLRP3 in response to nigericin and cold temperature exposure. We also found that most CAPS-related NLRP3 variants can be inhibited by MCC950; however, NLRP3 variants with changes to proline affecting helices near the inhibitor binding site are resistant to MCC950, as are variants in the pyrin domain, which likely trigger activation directly with the pyrin domain of ASC. Our findings could help stratify the CAPS population for NLRP3 inhibitor clinical trials and our automated methodologies can be implemented for molecules with a different mechanism of activation and in laboratories worldwide that are interested in adding new functionally validated NLRP3 variants to the resource. Overall, our study provides improved diagnosis for patients with CAPS, mechanistic insight into the activation of NLRP3 and stratification of patients for the future application of targeted therapeutics.
Collapse
Affiliation(s)
- Shouya Feng
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Matthew C Wierzbowski
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Katja Hrovat-Schaale
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Andreas Dumortier
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Yaoyuan Zhang
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Maria Zyulina
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- Institute of Structural Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Paul J Baker
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Thomas Reygaerts
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Annemarie Steiner
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Dominic De Nardo
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Dhanya Lakshmi Narayanan
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Florian Milhavet
- Department of Molecular genetics and Cytogenomics, CHU Montpellier, Rare and Autoinflammatory Diseases Unit, University of Montpellier, CEREMAIA, Institute for Regenerative Medicine and Biotherapy, INSERMU1183, Montpellier, France
| | - Alberto Pinzon-Charry
- Queensland Paediatric Immunology and Allergy Service, Children's Health Queensland, Brisbane, Queensland, Australia
| | - Juan Ignacio Arostegui
- Department of Immunology, Hospital Clínic, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- School of Medicine, University of Barcelona, Barcelona, Spain
| | - Raju P Khubchandani
- Sectional Head Pediatric Rheumatology, SRCC Children's Hospital, Mumbai, India
| | - Matthias Geyer
- Institute of Structural Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Guilaine Boursier
- Department of Molecular genetics and Cytogenomics, CHU Montpellier, Rare and Autoinflammatory Diseases Unit, University of Montpellier, CEREMAIA, Institute for Regenerative Medicine and Biotherapy, INSERMU1183, Montpellier, France
| | - Seth L Masters
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia.
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
17
|
Khawas S, Sharma N. Cell death crosstalk in respiratory diseases: unveiling the relationship between pyroptosis and ferroptosis in asthma and COPD. Mol Cell Biochem 2025; 480:1305-1326. [PMID: 39112808 DOI: 10.1007/s11010-024-05062-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/29/2024] [Indexed: 02/21/2025]
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) are heterogeneous obstructive diseases characterized by airflow limitations and are recognized as significant contributors to fatality all over the globe. Asthma accounts for about 4, 55,000 deaths, and COPD is the 3rd leading contributor of mortality worldwide. The pathogenesis of these two obstructive disorders is complex and involves numerous mechanistic pathways, including inflammation-mediated and non-inflammation-mediated pathways. Among all the pathological categorizations, programmed cell deaths (PCDs) play a dominating role in the progression of these obstructive diseases. The two major PCDs that are involved in structural and functional remodeling in the progression of asthma and COPD are Pyroptosis and Ferroptosis. Pyroptosis is a PCD mechanism mediated by the activation of the Nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome, leading to the maturation and release of Interleukin-1β and Interleukin-18, whereas ferroptosis is a lipid peroxidation-associated cell death. In this review, the major molecular pathways contributing to these multifaceted cell deaths have been discussed, and crosstalk among them regarding the pathogenesis of asthma and COPD has been highlighted. Further, the possible therapeutic approaches that can be utilized to mitigate both cell deaths at once have also been illustrated.
Collapse
Affiliation(s)
- Sayak Khawas
- Department of Pharmaceutical Science & Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Neelima Sharma
- Department of Pharmaceutical Science & Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India.
| |
Collapse
|
18
|
Bianchi F, Roccabianca P, Vianello E, Gentile G, La Sala L, Bandera F, Tacchini L, Zoia R, Corsi Romanelli MM, Dozio E. Inhibition of DPP-4 Attenuates Endotoxemia-Induced NLRC4 Inflammasome and Inflammation in Visceral Adipose Tissue of Mice Fed a High-Fat Diet. Biomolecules 2025; 15:333. [PMID: 40149869 DOI: 10.3390/biom15030333] [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: 01/22/2025] [Revised: 02/12/2025] [Accepted: 02/23/2025] [Indexed: 03/29/2025] Open
Abstract
Inflammasomes are protein complexes that trigger pro-inflammatory responses and promote many diseases, including adipose tissue dysfunction. Linagliptin (L), a DPP-4 inhibitor used for type 2 diabetes therapy, has putative anti-inflammatory effects. This work explores L effects on inflammasome regulation, inflammation, and adipose tissue dysfunction in obese mice. Male C57BL/6N mice were fed a normal chow (NC) diet, high-fat (HF) diet, or HF diet with L (HFL) for 15 weeks. Gene expression and histological examinations were performed on visceral (VAT) and subcutaneous (SAT) adipose tissue samples. Biomarkers were quantified on sera. Murine macrophages were utilized for in vitro analyses. L decreased HF-induced endotoxemia and circulating inflammatory indicators. Despite having no effect on body weight, L reduced VAT inflammation by decreasing endotoxemia-induced NLRC4 inflammasome, inflammation severity, and fat cell hypertrophy. Although SAT response differed from VAT, inflammation was slightly reduced in this tissue too. In vitro, L modulated inflammation by directly reducing the pro-inflammatory macrophage phenotype. In obesity, increased NLRC4 inflammasome expression links endotoxemia and VAT inflammation. L protected against endotoxemia, maybe by affecting gut permeability and VAT responses. The decreased polarization of macrophages toward a pro-inflammatory phenotype and the reduction in adipocyte hypertrophy are involved in the response to L.
Collapse
Affiliation(s)
- Francesca Bianchi
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy
- Laboratorio di Morfologia Umana Applicata, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy
| | - Paola Roccabianca
- Dipartimento di Medicina Veterinaria e Scienze Animali, Università degli Studi di Milano, 26900 Lodi, Italy
| | - Elena Vianello
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy
- Laboratorio Sperimentale Ricerche Biomarcatori di Danno d'Organo, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| | - Guendalina Gentile
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy
| | - Lucia La Sala
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy
- IRCCS MultiMedica, 20138 Milan, Italy
| | - Francesco Bandera
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy
- IRCCS MultiMedica, 20138 Milan, Italy
| | - Lorenza Tacchini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy
- Laboratorio Sperimentale Ricerche Biomarcatori di Danno d'Organo, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| | - Riccardo Zoia
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy
| | - Massimiliano M Corsi Romanelli
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy
- Dipartimento di Patologia Clinica e Sperimentale, IRCCS Istituto Auxologico, 20149 Milan, Italy
| | - Elena Dozio
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy
- Laboratorio Sperimentale Ricerche Biomarcatori di Danno d'Organo, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| |
Collapse
|
19
|
Zisis M, Chondrogianni ME, Androutsakos T, Rantos I, Oikonomou E, Chatzigeorgiou A, Kassi E. Linking Cardiovascular Disease and Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): The Role of Cardiometabolic Drugs in MASLD Treatment. Biomolecules 2025; 15:324. [PMID: 40149860 DOI: 10.3390/biom15030324] [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: 12/27/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
The link between cardiovascular disease (CVD) and metabolic dysfunction-associated steatotic liver disease (MASLD) is well-established at both the epidemiological and pathophysiological levels. Among the common pathophysiological mechanisms involved in the development and progression of both diseases, oxidative stress and inflammation, insulin resistance, lipid metabolism deterioration, hepatokines, and gut dysbiosis along with genetic factors have been recognized to play a pivotal role. Pharmacologic interventions with drugs targeting common modifiable cardiometabolic risk factors, such as T2DM, dyslipidemia, and hypertension, are a reasonable strategy to prevent CVD development and progression of MASLD. Recently, a novel drug for metabolic dysfunction-associated steatohepatitis (MASH), resmetirom, has shown positive effects regarding CVD risk, opening new opportunities for the therapeutic approach of MASLD and CVD. This review provides current knowledge on the epidemiologic association of MASLD to CVD morbidity and mortality and enlightens the possible underlying pathophysiologic mechanisms linking MASLD with CVD. The role of cardiometabolic drugs such as anti-hypertensive drugs, hypolipidemic agents, glucose-lowering medications, acetylsalicylic acid, and the thyroid hormone receptor-beta agonist in the progression of MASLD is also discussed. Metformin failed to prove beneficial effects in MASLD progression. Studies on the administration of thiazolinediones in MASLD suggest effectiveness in improving steatosis, steatohepatitis, and fibrosis, while newer categories of glucose-lowering agents such as GLP-1Ra and SGLT-2i are currently being tested for their efficacy across the whole spectrum of MASLD. Statins alone or in combination with ezetimibe have yielded promising results. The conduction of long-duration, large, high-quality, randomized-controlled trials aiming to assess by biopsy the efficacy of cardiometabolic drugs to reverse MASLD progression is of great importance.
Collapse
Affiliation(s)
- Marios Zisis
- Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, 11527 Athens, Greece
| | - Maria Eleni Chondrogianni
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Endocrine Unit, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Theodoros Androutsakos
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece
| | - Ilias Rantos
- Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, 11527 Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, "Sotiria" Thoracic Diseases Hospital of Athens, University of Athens Medical School, 11527 Athens, Greece
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Endocrine Unit, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
20
|
Zhang J, Li R, Wang L, Ni S. Dexmedetomidine activates mitophagy and protects against pyroptosis in oxygen-glucose deprivation/reperfusion-induced brain damage via PINK1/Parkin pathway activation. J Bioenerg Biomembr 2025:10.1007/s10863-025-10051-4. [PMID: 39985625 DOI: 10.1007/s10863-025-10051-4] [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: 09/19/2024] [Accepted: 01/18/2025] [Indexed: 02/24/2025]
Abstract
Accumulating studies have unraveled that dexmedetomidine (DEX) is neuroprotective against brain damage. However, it remains largely unknown about the mechanism involved in the neuroprotective effect of DEX. Therefore, this study explored whether DEX could affect mitophagy and pyroptosis in hypoxic-ischemic brain damage. We established a hippocampal neuron model of oxygen glucose-deprivation (OGD) and a rat model of cerebral ischemia/reperfusion (I/R) injury, which were then intervened with DEX and the autophagy inhibitor (3-MA). It was found that DEX intervention significantly increased neuron viability and mitophagy. Additionally, DEX intervention reversed increased oxidative stress and pyroptosis caused by OGD. DEX intervention further maintained the activation of the PINK1/Parkin pathway, while 3-MA treatment partly counteracted the protective effect of DEX on OGD-induced hippocampal neurons, suggesting that the inhibition of the PINK1/Parkin pathway reversed the function of DEX to increase cell viability and mitophagy and inhibit oxidative stress, pyroptosis, and apoptosis. Animal experiments also revealed that DEX intervention induced PINK1/Parkin pathway activation, reduced cerebral infarction and mitochondrial damage, promoted mitophagy, and inhibited pyroptosis, which was nullified by 3-MA treatment. Conclusively, DEX protects against pyroptosis and activates mitophagy in OGD/R-induced brain damage by activating the PINK1/Parkin pathway.
Collapse
Affiliation(s)
- Jieru Zhang
- Department of Anesthesiology, Yantaishan Hospital, 91 Jiefang Road, Yantai City, Shandong Province, 264001, China
| | - Ruxia Li
- Department of Anesthesiology, Yantaishan Hospital, 91 Jiefang Road, Yantai City, Shandong Province, 264001, China
| | - Luyong Wang
- Department of Anesthesiology, Yantaishan Hospital, 91 Jiefang Road, Yantai City, Shandong Province, 264001, China
| | - Shuqin Ni
- Department of Anesthesiology, Yantaishan Hospital, 91 Jiefang Road, Yantai City, Shandong Province, 264001, China.
| |
Collapse
|
21
|
Jing J, Yang F, Wang K, Cui M, Kong N, Wang S, Qiao X, Kong F, Zhao D, Ji J, Tang L, Gao J, Cong YS, Ding D, Chen K. UFMylation of NLRP3 Prevents Its Autophagic Degradation and Facilitates Inflammasome Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2406786. [PMID: 39985286 DOI: 10.1002/advs.202406786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 01/29/2025] [Indexed: 02/24/2025]
Abstract
NLRP3 (NOD, LRR and pyrin domain-containing protein 3) inflammasome is important for host defense against infections and maintaining homeostasis. Aberrant activation of NLRP3 inflammasome is closely related to various inflammatory diseases. Post-translational modifications are critical for NLRP3 inflammasome regulation. However, the mechanism of NLRP3 inflammasome activation remains incompletely understood. Here, it is demonstrated that the Ufm1 E3 ligase Ufl1 mediated UFMylation is essential for NLRP3 inflammasome activation. Mechanistically, Ufl1 binds and UFMylates NLRP3 in the priming stage of NLRP3 activation, thereby sustaining the stability of NLRP3 by preventing NLRP3 K63-linked ubiquitination and the subsequent autophagic degradation. It is further demonstrated that myeloid cell-specific Ufl1 or Ufm1 deficiency in mice significantly alleviated inflammatory responses and tissue damage following lipopolysaccharide (LPS)-induced endotoxemia and alum-induced peritonitis. Thus, the findings offer new insights into potential therapeutic targets for NLRP3 inflammasome-related diseases by targeting the UFMylation system.
Collapse
Affiliation(s)
- Jiongjie Jing
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200127, China
| | - Fan Yang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200127, China
| | - Ke Wang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Mintian Cui
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200127, China
| | - Ni Kong
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200127, China
| | - Shixi Wang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200127, China
| | - Xiaoyue Qiao
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200127, China
| | - Fanyu Kong
- Department of Internal Emergency Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Dongyang Zhao
- Department of Internal Emergency Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Jinlu Ji
- Department of Internal Emergency Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Lunxian Tang
- Department of Internal Emergency Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Jiaxin Gao
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yu-Sheng Cong
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou, 311121, China
| | - Deqiang Ding
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Kun Chen
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200127, China
- Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| |
Collapse
|
22
|
Zariņa KZ, Pilmane M, Pētersons A. Immunomodulatory Tissue Factors in the Gallbladder Walls of Pediatric Patients with Chronic Calculous Cholecystitis. CHILDREN (BASEL, SWITZERLAND) 2025; 12:205. [PMID: 40003307 PMCID: PMC11854828 DOI: 10.3390/children12020205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/03/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND The rising rates of gallstones and cholecystectomy in pediatric populations underscore the increasing concern regarding chronic cholecystitis. However, the morphopathogenesis of pediatric calculous cholecystitis is still not well understood. This study aimed to determine the expression and distribution of immunomodulatory factors interleukin-12 (IL-12), interleukin-13 (IL-13), interleukin-1β (IL-1β), sonic hedgehog protein (SHH), nuclear factor NF-kappa-B p65 subunit (NFkBp65), and heat shock protein 60 (HSP60) in the gallbladder walls of pediatric patients with chronic calculous cholecystitis. METHODS In total, 11 gallbladder samples were collected from pediatric patients with calculous cholecystitis during cholecystectomy, while 5 healthy gallbladder samples served as controls. IL-12, IL-13, IL-1β, SHH, NFkBp65, and HSP60 were detected by immunohistochemistry. The number of positive structures in gallbladder wall epithelium, vasculature, and inflammatory infiltrate was assessed semi-quantitatively by microscopy. A Mann-Whitney U test and Spearman's rank-order correlation coefficient were calculated. RESULTS Statistically significant differences were observed between patient and control samples in the expression of IL-1β, SHH, and NFkBp65 in the epithelium, as well as in the expression of IL-12, SHH, and HSP60 in the blood vessels. The expression of IL-1β was stronger in the epithelium of controls, while other markers were more prominent in patient samples. CONCLUSIONS An increased number of NFkBp65, IL-12, and HSP60 positive cells in patient gallbladder tissue suggests a significant role of these tissue factors in driving immune modulation and sustaining the inflammation in pediatric chronic calculous cholecystitis. The noticeable expression of SHH in patient gallbladder tissue indicates its part in tissue regeneration and repair processes, as well as in modulating inflammation and vascular responses in calculous cholecystitis. The significant positive correlations between the factors studied highlight the importance of their coordinated interaction and intricate crosstalk in the morphopathogenesis of calculous cholecystitis.
Collapse
Affiliation(s)
- Kaiva Zīle Zariņa
- Institute of Anatomy and Anthropology, Riga Stradins University, Kronvalda Boulevard 9, LV-1010 Riga, Latvia
| | - Māra Pilmane
- Institute of Anatomy and Anthropology, Riga Stradins University, Kronvalda Boulevard 9, LV-1010 Riga, Latvia
| | - Aigars Pētersons
- Department of Pediatric Surgery, Riga Stradins University, Dzirciema Street 16, LV-1007 Riga, Latvia
| |
Collapse
|
23
|
Fernandes DC, Silva-de-França F, Pohl PC, Eto SF, Sardinha LR, Lambris JD, Tambourgi DV. Cp40-mediated complement C3 inhibition dampens inflammasome activation and inflammatory mediators storm induced by Bitis arietans venom. Int Immunopharmacol 2025; 147:113701. [PMID: 39809101 DOI: 10.1016/j.intimp.2024.113701] [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: 10/01/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 01/16/2025]
Abstract
The complement system plays a crucial role in various pathophysiological conditions, including snake envenomation. In this study, we investigated the effects of Bitis arietans venom on the complement system using an ex vivo human whole blood model. Our findings demonstrate that B. arietans venom was able to activate the complement system, leading to a significant increase in the production of anaphylatoxins (C3a/C3a-desArg, C5a/C5a-desArg) and the soluble Terminal Complement Complex (sTCC). Inhibition of the C3 component by Cp40, a C3-C3b inhibitor, resulted in the reduction of C3a/C3a-desArg, C5a/C5a-desArg, and sTCC levels to baseline in venom-stimulated samples. Furthermore, treatment with Cp40 promoted a substantial decrease in the production of pro-inflammatory mediators, such as Prostaglandin E2 (PGE2), IL-8/CXCL8, MCP-1/CCL2, and MIG/CXCL9. To further elucidate the molecular mechanisms, we utilized the THP-1 cell line differentiated into M0 macrophages. Incubation of these macrophages with human plasma, from the human whole blood treated with B. arietans venom, resulted in the expression of the NLRP3 inflammasome and the production of IL-8 and IL-1β. Importantly, Cp40 was able to diminish the production of these cytokines, as well as the levels of ASC and caspase-1 proteins. In conclusion, our results indicate that the inhibition of the complement by Cp40 at C3/C3b level can modulate the inflammatory response and inflammasome activation induced by B. arietans venom. These findings suggest that complement inhibition may be a promising therapeutic approach for managing the inflammatory complications associated with this snake envenomation.
Collapse
Affiliation(s)
- Dayanne Carla Fernandes
- Immunochemistry Laboratory, Butantan Institute, São Paulo, SP, Brazil; Center of Toxins, Cell Signaling and Immune Response (CeTICS), CEPID, FAPESP, Brazil
| | - Felipe Silva-de-França
- Immunochemistry Laboratory, Butantan Institute, São Paulo, SP, Brazil; Center of Toxins, Cell Signaling and Immune Response (CeTICS), CEPID, FAPESP, Brazil
| | | | | | | | - John D Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Denise V Tambourgi
- Immunochemistry Laboratory, Butantan Institute, São Paulo, SP, Brazil; Center of Toxins, Cell Signaling and Immune Response (CeTICS), CEPID, FAPESP, Brazil.
| |
Collapse
|
24
|
Wang L, Zhu K, Tian Z, Wang H, Jia Y, Feng C, Qi L, Tang W, Hu Y. Discovery of novel biaryl urea derivatives against IL-1β release with low toxicity based on NEK7 inhibitor. Eur J Med Chem 2025; 283:117125. [PMID: 39647417 DOI: 10.1016/j.ejmech.2024.117125] [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: 10/25/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 12/10/2024]
Abstract
Aberrant activation of NLRP3 inflammasome is involved in various inflammatory diseases, making it a promising target for therapeutic intervention. NEK7, a member of the NIMA-related kinase (NEK) family, functions as a key NLRP3-binding protein and plays a crucial role in the regulation of NLRP3 inflammasome assembly and activation. Thus, disrupting NLRP3-NEK7 interactions by targeting NEK7 could be a promising strategy to inhibit the activation of NLRP3 inflammasome. In this work, a series of novel urea derivatives were designed and synthesized based on the reported NEK7 inhibitors. Among these, compound 23 exhibited potent activity against IL-1β release with low cytotoxicity. Moreover, compound 23 enhanced the thermal stability of NEK7 and disrupted the NLRP3-NEK7 interaction, thereby regulating NLRP3 inflammasome assembly.
Collapse
Affiliation(s)
- Leibo Wang
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, 1 Xiangshanzhi Road, Hangzhou, 310024, China
| | - Kehan Zhu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 ZuChongZhi Road, Shanghai, 201203, China
| | - Ziyang Tian
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, 1 Xiangshanzhi Road, Hangzhou, 310024, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 110039, China
| | - Haoyu Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 ZuChongZhi Road, Shanghai, 201203, China
| | - Yulei Jia
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, 1 Xiangshanzhi Road, Hangzhou, 310024, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 110039, China
| | - Chunlan Feng
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 ZuChongZhi Road, Shanghai, 201203, China
| | - Luyao Qi
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 ZuChongZhi Road, Shanghai, 201203, China
| | - Wei Tang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 ZuChongZhi Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 110039, China.
| | - Youhong Hu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, 1 Xiangshanzhi Road, Hangzhou, 310024, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 110039, China; State Key Laboratory of Drug Research, Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 ZuChongZhi Road, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, 198 East Binhai Road, Yantai, Shandong, 264117, China.
| |
Collapse
|
25
|
Gu HY, Liu N. Mechanism of effect and therapeutic potential of NLRP3 inflammasome in spinal cord injury. Exp Neurol 2025; 384:115059. [PMID: 39571746 DOI: 10.1016/j.expneurol.2024.115059] [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: 10/09/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024]
Abstract
Spinal cord injury (SCI) is a serious and disabling central nervous system injury that can trigger various neuropathological conditions, resulting in neuronal damage and release of various pro-inflammatory mediators, leading to neurological dysfunction. Currently, surgical decompression, drugs and rehabilitation are primarily used to relieve symptoms and improve endogenous repair mechanisms; however, they cannot directly promote nerve regeneration and functional recovery. SCI can be divided into primary and secondary injuries. Secondary injury is key to determining the severity of injury, whereas inflammation and cell death are important pathological mechanisms in the process of secondary SCI. The activation of the inflammasome complex is thought to be a necessary step in neuro-inflammation and a key trigger for neuronal death. The NLRP3 inflammasome is a cytoplasmic multiprotein complex that is considered an important factor in the development of SCI. Once the NLRP3 inflammasome is activated after SCI, NLRP3 nucleates the assembly of an inflammasome, leading to caspase 1-mediated proteolytic activation of the interleukin-1β (IL-1β) family of cytokines, and induces an inflammatory, pyroptotic cell death. Inhibition of inflammasomes can effectively inhibit inflammation and cell death in the body and promote the recovery of nerve function after SCI. Therefore, inhibition of NLRP3 inflammasome activation may be a promising approach for the treatment of SCI. In this review, we describe the current understanding of NLRP3 inflammasome activation in SCI pathogenesis and its subsequent impact on SCI and summarize drugs and other potential inhibitors based on NLRP3 inflammasome regulation. The objective of this study was to emphasize the role of the NLRP3 inflammasome in SCI, and provide a new therapeutic strategy and theoretical basis for targeting the NLRP3 inflammasome as a therapy for SCI.
Collapse
Affiliation(s)
- Hou-Yun Gu
- Department of Spine Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China; Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital), Southern Medical University, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China.
| | - Ning Liu
- Department of Spine Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China; Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital), Southern Medical University, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China.
| |
Collapse
|
26
|
de Sousa VM, Almeida ÁMAN, Ferreira RS, dos Santos BL, da Silva VDA, David JM, dos Santos CC, Costa SL. The Flavonoid Agathisflavone Attenuates Glia Activation After Mechanical Injury of Cortical Tissue and Negatively Regulates Both NRLP3 and IL-1β Expression. Int J Mol Sci 2025; 26:1275. [PMID: 39941042 PMCID: PMC11818122 DOI: 10.3390/ijms26031275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Traumatic brain injury (TBI) has a complex and multifactorial pathology and is a major cause of death and disability for humans. Immediately after TBI, astrocytes and microglia react with complex morphological and functional changes known as reactive gliosis to form a glial scar in the area immediately adjacent to the lesion, which is the major barrier to neuronal regeneration. The flavonoid agathisflavone (bis-apigenin), present in Poincianella pyramidalis leaves, has been shown to have neuroprotective, neurogenic, and anti-inflammatory effects, demonstrated in vitro models of glutamate-induced toxicity, neuroinflammation, and demyelination. In this study, we evaluated the effect and mechanisms of agathisflavone in neuronal integrity and in the modulation of gliosis in an ex vivo model of TBI. For this, microdissections from the encephalon of Wistar rats (P6-8) were prepared and subjected to mechanical injury (MI) and treated or not with daily agathisflavone (5 μM) for 3 days. Astrocyte reactivity was investigated by measuring mRNA and expression of GFAP protein in the lesioned area by immunofluorescence and Western blot. The proportion of microglia was determined by immunofluorescence for Iba-1; mRNA expression for inflammasome NRPL3 and interleukin-1 beta (IL-1β) was determined by RT-qPCR. It was observed that lesions in the cortical tissue induced astrocytes overexpressing GFAP in the typical glial scar formed and that agathisflavone modulated GFAP expression at the transcriptional and post-transcriptional levels, which was associated with a reduction of the glial scar. MI induced an increase in the proportion of microglia (Iba-1+), which was not observed in agathisflavone-treated cultures. Moreover, the flavonoid modulated negatively both the NRLP3 and IL-1β mRNA expression that was increased in the lesioned area of the tissue. These findings support the regulatory properties of agathisflavone in the control of the inflammatory response in glial cells, which can impact neuroprotection and should be considered for future studies for TB and other pathological conditions of the central nervous system.
Collapse
Affiliation(s)
- Verônica Moreira de Sousa
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (V.M.d.S.); (Á.M.A.N.A.); (R.S.F.); (B.L.d.S.); (V.D.A.d.S.)
| | - Áurea Maria Alves Nunes Almeida
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (V.M.d.S.); (Á.M.A.N.A.); (R.S.F.); (B.L.d.S.); (V.D.A.d.S.)
| | - Rafael Short Ferreira
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (V.M.d.S.); (Á.M.A.N.A.); (R.S.F.); (B.L.d.S.); (V.D.A.d.S.)
| | - Balbino Lino dos Santos
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (V.M.d.S.); (Á.M.A.N.A.); (R.S.F.); (B.L.d.S.); (V.D.A.d.S.)
- College of Nursing, Federal University of Vale do São Francisco, Petrolina 56304-917, PB, Brazil
| | - Victor Diogenes Amara da Silva
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (V.M.d.S.); (Á.M.A.N.A.); (R.S.F.); (B.L.d.S.); (V.D.A.d.S.)
| | - Jorge Mauricio David
- Department of General and Inorganic Chemistry, Institute of Chemistry, Federal University of Bahia, Salvador 40170-110, BA, Brazil;
| | - Cleonice Creusa dos Santos
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (V.M.d.S.); (Á.M.A.N.A.); (R.S.F.); (B.L.d.S.); (V.D.A.d.S.)
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (V.M.d.S.); (Á.M.A.N.A.); (R.S.F.); (B.L.d.S.); (V.D.A.d.S.)
| |
Collapse
|
27
|
Almutary AG, Begum MY, Kyada AK, Gupta S, Jyothi SR, Chaudhary K, Sharma S, Sinha A, Abomughaid MM, Imran M, Lakhanpal S, Babalghith AO, Abu-Seer EA, Avinash D, Alzahrani HA, Alhindi AA, Iqbal D, Kumar S, Jha NK, Alghamdi S. Inflammatory signaling pathways in Alzheimer's disease: Mechanistic insights and possible therapeutic interventions. Ageing Res Rev 2025; 104:102548. [PMID: 39419399 DOI: 10.1016/j.arr.2024.102548] [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: 01/26/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
The complex pathophysiology of Alzheimer's disease (AD) poses challenges for the development of therapies. Recently, neuroinflammation has been identified as a key pathogenic mechanism underlying AD, while inflammation has emerged as a possible target for the management and prevention of AD. Several prior studies have demonstrated that medications modulating neuroinflammation might lessen AD symptoms, mostly by controlling neuroinflammatory signaling pathways such as the NF-κB, MAPK, NLRP3, etc, and their respective signaling cascade. Moreover, targeting these inflammatory modalities with inhibitors, natural products, and metabolites has been the subject of intensive research because of their anti-inflammatory characteristics, with many studies demonstrating noteworthy pharmacological capabilities and potential clinical applications. Therefore, targeting inflammation is considered a promising strategy for treating AD. This review comprehensively elucidates the neuroinflammatory mechanisms underlying AD progression and the beneficial effects of inhibitors, natural products, and metabolites in AD treatment.
Collapse
Affiliation(s)
- Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, P.O. Box 59911, Abu Dhabi, United Arab Emirates
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Ashish Kumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, Gujarat 360003, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Swati Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab 140307, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Sorabh Lakhanpal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Ahmad O Babalghith
- Medical Genetics Department, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Eman Adnan Abu-Seer
- Department of Epidemiology and Medical Statistic, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Makkah, Saudi Arabia
| | - D Avinash
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Hassan A Alzahrani
- Department of Respiratory Care, Medical Cities at the Minister of Interior, MCMOl, Riyadh, Saudi Arabia
| | | | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Sandeep Kumar
- School of Pharmacy, Sharda University, Greater Noida, India; DST-FIST Laboratory, Sharda University, Greater Noida, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Biosciences and Technology (SBT), Galgotias University, Greater Noida, India; Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India.
| | - Saad Alghamdi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
28
|
Abrams ED, Basu A, Zavorka Thomas ME, Henrickson SE, Abraham RS. Expanding the diagnostic toolbox for complex genetic immune disorders. J Allergy Clin Immunol 2025; 155:255-274. [PMID: 39581295 DOI: 10.1016/j.jaci.2024.11.022] [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: 08/30/2024] [Revised: 10/29/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024]
Abstract
Laboratory-based immunology evaluation is essential to the diagnostic workup of patients with complex immune disorders, and is as essential, if not more so, depending on the context, as genetic testing, because it enables identification of aberrant pathways amenable to therapeutic intervention and clarifies variants of uncertain significance. There have been considerable advances in techniques and instrumentation in the clinical laboratory in the past 2 decades, although there are still "miles to go." One of the goals of the clinical laboratory is to ensure advanced diagnostic testing is widely accessible to physicians and thus patients, through reference laboratories, particularly in the context of academic medical centers. This ensures a greater likelihood of translating research discoveries into the diagnostic laboratory, on the basis of patient care needs rather than a sole emphasis on commercial utility. However, these advances are under threat from burdensome regulatory oversight that can compromise, at best, and curtail, at worst, the ability to rapidly diagnose rare immune disorders and ensure delivery of precision medicine. This review discusses the clinical utility of diagnostic immunology tools, beyond cellular immunophenotyping of lymphocyte subsets, which can be used in conjunction with clinical and other laboratory data for diagnosis as well as monitoring of therapeutic response in patients with genetic immunologic diseases.
Collapse
Affiliation(s)
- Eric D Abrams
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Amrita Basu
- Diagnostic Immunology Laboratory, Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio
| | - Megan E Zavorka Thomas
- Diagnostic Immunology Laboratory, Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio
| | - Sarah E Henrickson
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa; Institute for Immunology and Immune Health, University of Pennsylvania, Philadelphia, Pa; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Roshini S Abraham
- Diagnostic Immunology Laboratory, Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio.
| |
Collapse
|
29
|
Zhong X, Sun Y, Lin Y, Deng S, Wang H, Zhou X, Lu J, Zheng Y, Luo R, Huang M, Song J. Ginsenoside Rd protects against acute liver injury by regulating the autophagy NLRP3 inflammasome pathway. Sci Rep 2025; 15:3569. [PMID: 39875579 PMCID: PMC11775168 DOI: 10.1038/s41598-025-87991-9] [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] [Received: 09/29/2024] [Accepted: 01/23/2025] [Indexed: 01/30/2025] Open
Abstract
Ginsenoside Rd (Rd) is a bioactive compound predominantly found in Panax ginseng C.A. Meyer and Panax notoginseng (Burkill) F.H. Chen ex C.H. Chow, both species belonging to genus Panax in the Araliaceae family. However, its hepatic protective effect against acute liver injury and related mechanistic action remain unexplored. To investigate the protective effect of Rd against thioacetamide (TAA)-induced acute liver injury and assess its underlying regulatory mechanisms related to autophagy and inflammation. Forty-eight 8 weeks old C57BL/6 mice were treated with saline (control or model group), Rd (12.5 mg/kg, 25 mg/kg or 50 mg/kg), and diammonium glycyrrhizinate (DG, 30 mg/kg) for three days. Then the mice were stimulated with TAA to establish acute liver injury model, excluding the control group. HSC-T6 cells were treated with Rd at concentrations of 2.5, 5, or 10 µM, for 12 h with or without Lipopolysaccharide (LPS) stimulation at 100 ng/mL. Immunofluorescence staining, qPCR and Western blot were employed to analyze the expressions of genes and proteins associated with inflammation and autophagy. To validate the role of Rd in regulating autophagy and inflammation, the autophagy inducers, rapamycin and GSK621, were utilised in reverse validation experiments in cells. Rd exhibited significant hepatic protective effects in mice by reducing the serum levels of Aspartate aminotransferase (AST), Alanine aminotransferase (ALT), Glutathione S-transferase (GST) and Lactate dehydrogenase (LDH) with acute liver injury. It exhibited strong anti-inflammatory effect by reducing inflammation associated protein, such as cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), nod-like receptor protein 3 (NLRP3), associated speck-like protein containing a CARD (ASC), interleukin-18 (IL-18) and interleukin-1β(IL-1β) proteins and the mRNA expression levels of COX-2, Tumor Necrosis Factor α (TNF α), interleukin-6 (IL-6) and iNOS were decreased in liver tissue. And Rd inhibited LPS-induced inflammation by reducing the expression of COX-2 and NLRP3 in HSC-T6 cells. Moreover, not only in vivo but also in vitro, Rd downregulated the expression of LC3II, Beclin1, phosphorylation-AMP-activated protein kinase (p-AMPK), phosphorylation-ULK1 (p-ULK1) and upregulated the expression of p62 and phosphorylation-mechanistic target of rapamycin (p-mTOR) to suppress autophagy via the AMPK/mTOR/ULK1 pathway. Finally, the inhibitory effects of Rd on autophagy and inflammation in HSC-T6 cells were partially blocked by rapamycin and GSK621. Rd is a promising therapeutic agent to protect liver against TAA-induced acute liver injury by regulating the autophagy-NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Xiaomei Zhong
- The Affiliated People's Hospital, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, China
| | - Yibin Sun
- Kaifeng Hospital of Traditional Chinese Medicine, Kaifeng, 475000, China
| | - Yanxiang Lin
- The Affiliated People's Hospital, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, China
| | - Shan Deng
- The Affiliated People's Hospital, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, China
| | - Huan Wang
- The Affiliated People's Hospital, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, China
| | - Xian Zhou
- NICM Health Research Institute, Western Sydney University, Westmead, NSW, 2006, Australia
| | - Jinjian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao, China
| | - Yanfang Zheng
- The Affiliated People's Hospital, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, China.
| | - Ruoyin Luo
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Belfast, UK.
| | - Mingqing Huang
- The Affiliated People's Hospital, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, China.
| | - Jianyuan Song
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
30
|
Gong Z, Yang H, Gao L, Liu Y, Chu Q, Luo C, Kang L, Zhai H, Xu Q, Wu W, Li N, Li R. Mechanisms of wogonoside in the treatment of atherosclerosis based on network pharmacology, molecular docking, and experimental validation. BMC Complement Med Ther 2025; 25:28. [PMID: 39871254 PMCID: PMC11770944 DOI: 10.1186/s12906-025-04760-x] [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: 05/22/2024] [Accepted: 01/14/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Atherosclerosis serves as the fundamental pathology for a variety of cardiovascular disorders, with its pathogenesis being closely tied to the complex interplay among lipid metabolism, oxidative stress, and inflammation. Wogonoside is a natural flavonoid extracted from Scutellaria baicalensis with a variety of biological activities, including anti-inflammatory, hypolipidemic, and cardiac function improvement properties. Despite these known effects, the specific role of wogonoside in the context of atherosclerosis remains to be elucidated. PURPOSE To validate the efficacy of wogonoside in the treatment of atherosclerosis and to investigate its possible therapeutic mechanisms. METHODS Network pharmacology was used to obtain the core targets and signaling pathways that may be efficacious in the treatment of atherosclerosis with wogonoside, which were validated using molecular docking and molecular dynamics simulations. To further validate the core targets in the signaling pathway, we performed in vivo experiments using apolipoprotein E (ApoE)-/- mice. This included pathological morphology and lipid deposition analysis of mouse aorta, serum lipid level analysis, Elisa analysis, oxidative stress analysis, reactive oxygen species (ROS) fluorescence assay, immunohistochemical analysis and protein blot analysis. RESULTS Predictions were obtained that wogonoside treatment of atherosclerosis has 31 core targets, which are mainly focused on pathways such as Toll-like receptor (TLR) signaling pathway and NF-kappa B (NF-κB ) signaling pathway. Molecular docking and molecular dynamics simulations showed that wogonoside has good binding properties to the core targets. In vivo experimental results showed that wogonoside significantly inhibited aortic inflammatory response and lipid deposition, significantly reduced the release levels of total cholesterol (TC), triglycerides (TG), low-density-lipoprotein cholesterol (LDL-C), oxidized low density (ox-LDL) and free fatty acid (FFA), and significantly inhibited the release of inflammatory factors TNF-α, IL-1β, IL-6 and oxidative stress in ApoE-/- mice. Further molecular mechanism studies showed that wogonoside significantly inhibited the activation of TLR4/NF-κB signaling pathway in ApoE-/- mice. CONCLUSION Wogonoside may be an effective drug monomer for the treatment of atherosclerosis, and its mechanism of action is closely related to the inhibition of the activation of the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zhaohui Gong
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Haixin Yang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Li Gao
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Yi Liu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Qingmin Chu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Chuanjin Luo
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Liang Kang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Huiqi Zhai
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Qiang Xu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Wei Wu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Nan Li
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China.
| | - Rong Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
31
|
Lin L, Zheng J, Lin Q, Cai F, Li D. The Role of Key Molecules of Pyroptosis in Liver Damage of Rats With Exertional Heat Stroke. Gastroenterol Res Pract 2025; 2025:6864091. [PMID: 39872797 PMCID: PMC11772058 DOI: 10.1155/grp/6864091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 10/18/2024] [Accepted: 12/04/2024] [Indexed: 01/30/2025] Open
Abstract
Purpose: This study is aimed at investigating the role of key molecular elements involved in pyroptosis in liver injury caused by exertional heat stroke (EHS). Methods: We established a model of EHS-induced liver injury in Sprague-Dawley rats, with a control group (receiving no treatment) for comparison and 12 rats in each group. Alanine transaminase (ALT) and aspartate transaminase (AST) levels in the blood were detected. Interleukin-1 beta (IL-1β) and interleukin-18 (IL-18) levels were assessed using enzyme-linked immunosorbent assays (ELISA). Pathological changes in liver tissue were examined by hematoxylin and eosin (H&E) staining. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting were used to detect mRNA and protein expression levels of Caspase-1 and Gasdermin D. Results: Compared to the control group, the liver tissue of the EHS group showed congestion in hepatic sinusoids, hepatocyte edema, eosinophilic changes, necrosis, and infiltration of inflammatory cells. ALT and AST levels in the EHS group were significantly higher than those in the control group (p < 0.05). The mRNA expressions of Caspase-1, Gasdermin D, IL-1β, and IL-18 were significantly increased in the EHS group compared to the control group (p < 0.001). The protein expressions of Caspase-1, cleaved Caspase-1, Gasdermin D, and cleaved Gasdermin D were significantly increased in the EHS group. Conclusion: These findings indicated that hepatic pyroptosis plays an important role in EHS-induced liver injury.
Collapse
Affiliation(s)
- Lifang Lin
- Department of Hepatobiliary Disease, 900th Hospital of Joint Logistics Support Force, Fuzhou General Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
- Department of Gastroenterology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Jiaolong Zheng
- Department of Hepatobiliary Disease, 900th Hospital of Joint Logistics Support Force, Fuzhou General Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Qingqing Lin
- Department of Hepatobiliary Disease, 900th Hospital of Joint Logistics Support Force, Fuzhou General Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Fangze Cai
- Department of Hepatobiliary Disease, 900th Hospital of Joint Logistics Support Force, Fuzhou General Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Dongliang Li
- Department of Hepatobiliary Disease, 900th Hospital of Joint Logistics Support Force, Fuzhou General Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
32
|
Sun Y, Zhou Y, Peng T, Huang Y, Lu H, Ying X, Kang M, Jiang H, Wang J, Zheng J, Zeng C, Liu W, Zhang X, Ai L, Peng Q. Preventing NLRP3 inflammasome activation: Therapeutic atrategy and challenges in atopic dermatitis. Int Immunopharmacol 2025; 144:113696. [PMID: 39608174 DOI: 10.1016/j.intimp.2024.113696] [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: 09/09/2024] [Revised: 11/06/2024] [Accepted: 11/20/2024] [Indexed: 11/30/2024]
Abstract
Atopic dermatitis (AD) is a prevalent inflammatory skin disorder characterized by its chronic, persistent, and recurrent nature. The pathophysiology of this condition is complex, involving various factors including cell-mediated immune responses, compromised skin barrier function, and alterations in hypersensitivity reactions. These components synergistically contribute to the perpetuation of the bothersome "itch-scratch-itch" cycle. Recent research has highlighted the significant role of the NLRP3 inflammasome in the development of AD and other inflammatory conditions. Current research indicates that the NLRP3 inflammasome plays a pivotal role in both the acute and chronic phases of AD by modulating the Th2/Th1 immune deviation. Moreover, the pharmacological suppression of NLRP3 has shown promising results in mitigating the pathological aspects of AD. This review outlines potential drug development strategies that target the NLRP3 inflammasome as a therapeutic approach for AD and the challenges faced in this endeavor.
Collapse
Affiliation(s)
- Yiran Sun
- School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Yangang Zhou
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China
| | - Tong Peng
- Department of R&D, Keystonecare Technology (Chengdu) Co., Ltd, Chengdu 610094, China
| | - Yuhang Huang
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China
| | - Hao Lu
- School of Biosciences and Technology, Key Laboratory of Target Discovery and Protein Drug Development in Major Diseases at Chengdu Medical College of Sichuan Province, Chengdu Medical College, Chengdu 610500, China
| | - Xiran Ying
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China
| | - Mingsheng Kang
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China
| | - Hao Jiang
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China
| | - Jingying Wang
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China
| | - Jiayao Zheng
- School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Chenyu Zeng
- School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Wanting Liu
- School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Xiaoyu Zhang
- College of Life Sciences, Sichuan Normal University, Chengdu 610101, China
| | - Lin Ai
- Department of Dermatology and Venereology, Nanbu County People's Hospital, Nanchong 637399, China
| | - Quekun Peng
- School of Biosciences and Technology, Key Laboratory of Target Discovery and Protein Drug Development in Major Diseases at Chengdu Medical College of Sichuan Province, Chengdu Medical College, Chengdu 610500, China.
| |
Collapse
|
33
|
Min Q, Chen X, Yifei G, Baifeng S, Zichuan W, Xiaolong S, Huajiang C, Wen Y, Yang L. FOXO3a overexpression ameliorates intervertebral disc degeneration by decreasing NLRP3-mediated pyroptosis. Int Immunopharmacol 2025; 144:113596. [PMID: 39579536 DOI: 10.1016/j.intimp.2024.113596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/25/2024]
Affiliation(s)
- Qi Min
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, China
| | - Xu Chen
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, China
| | - Gu Yifei
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, China
| | - Sun Baifeng
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, China
| | - Wu Zichuan
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, China
| | - Shen Xiaolong
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, China
| | - Chen Huajiang
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, China
| | - Yuan Wen
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, China.
| | - Liu Yang
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, China.
| |
Collapse
|
34
|
Lu Y, Gao J, Hou Y, Yang H, Wang D, Zhang G, Qin Z, Du P, Wang Z, Wang Y, Chen Q, Sun Z, Li P, Zhang J, Tang J. Targeting the NLRP3 inflammasome abrogates cardiotoxicity of immune checkpoint blockers. J Immunother Cancer 2025; 13:e010127. [PMID: 39773567 PMCID: PMC11749606 DOI: 10.1136/jitc-2024-010127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of many malignant tumors. However, ICI-induced hyper-immune activation causes cardiotoxicity. Traditional treatments such as glucocorticoids and immunosuppressants have limited effectiveness and may even accelerate tumor growth. This study aimed to identify approaches that effectively reduce cardiotoxicity and simultaneously preserve or enhance the antitumor immunity of ICI therapy. METHODS ICI injection in melanoma-bearing C57BL/6J female mice was used to simulate cardiotoxicity in patients with tumor undergoing immune therapy. MCC950 was used to block nod-like receptor protein 3 (NLRP3) inflammasome activity. Echocardiography, immunofluorescence, flow cytometry, and reverse transcription quantitative polymerase chain reaction were used to assess cardiac function, immune cell populations, and inflammatory factor levels. Bulk and single-cell RNA sequencing was used to detect the changes in cardiac transcriptome and immunological network. RESULTS NLRP3 inhibition reduced inflammatory response and improved cardiac function. Notably, NLRP3 inhibition also resulted in a pronounced suppression of tumor growth. Single-cell RNA sequencing elucidated that MCC950 treatment reduced the cardiac infiltration of pathogenic macrophages, cytotoxic T cells, activated T cells, and their production of inflammatory cytokines, while enhancing the presence of reparative macrophages and naive T cells. In addition, MCC950 attenuated cardiotoxicity induced by dual programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) immunotherapy and promoted tumor regression, and showed efficacy in treating established cardiotoxicity. CONCLUSIONS Our findings provide a promising clinical approach for preventing and treating cardiotoxicity induced by ICIs, dissociating the antitumor efficacy of ICI-based therapies from their cardiotoxic side effects.
Collapse
Affiliation(s)
- Yang Lu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Jiamin Gao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Yachen Hou
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Han Yang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Dashuai Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Zhen Qin
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Pengchong Du
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Zhenwei Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Yunzhe Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Quanzhou Chen
- Department of Nutrition, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhaowei Sun
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Ping Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jinying Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Junnan Tang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| |
Collapse
|
35
|
Li JP, Qiu S, Tai GJ, Liu YM, Wei W, Fu MM, Fang PQ, Otieno JN, Battulga T, Li XX, Xu M. NLRP3 inflammasome-modulated angiogenic function of EPC via PI3K/ Akt/mTOR pathway in diabetic myocardial infarction. Cardiovasc Diabetol 2025; 24:6. [PMID: 39762890 PMCID: PMC11705910 DOI: 10.1186/s12933-024-02541-3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Inflammatory diseases impair the reparative properties of endothelial progenitor cells (EPC); however, the involvement of diabetes in EPC dysfunction associated with myocardial infarction (MI) remains unknown. METHODS A model was established combining high-fat diet (HFD)/streptozotocin (STZ)-induced diabetic mice with myocardial infarction. The therapeutic effects of transplanted wild-type EPC, Nlrp3 knockout EPC, and Nlrp3 overexpression EPC were evaluated. Chip and Luciferase assay revealed CEBPB regulated the transcriptional expression of Nlrp3 as a transcription factor in EPC stimulated by high glucose (HG) or advanced glycation end products (AGEs). CO-IP results suggested that USP14 selectively suppressed NLRP3 degradation. KEGG enrichment revealed PI3K/ Akt/mTOR signaling showed striking significance in the entire pathway. RESULTS In our study, wild-type, Nlrp3 knockout and Nlrp3 overexpressed EPC, intracardiac injections effectively improved cardiac function, increased angiogenesis, and reduced infarct size in mice with myocardial infarction. However, in the HFD/STZ-induced diabetic mice model combined with myocardial infarction, Nlrp3 knockout EPC significantly restored angiogenic capacity. Mechanically, CEBPB regulated the transcriptional level of Nlrp3 as a transcription factor in EPC. Meanwhile, we found that USP14 selectively suppressed NLRP3 protein degradation through the USP motif on the NACHT domain in mediating inflammasome activation. Cardiac functional outcomes in recipient mice after intramyocardial injection of shNlrp3 EPC overexpressing CEBPB or USP14 validated the modulation of EPC function by regulating Nlrp3 transcription or post-translational modification. Furthermore, KEGG enrichment and validation at the protein levels revealed PI3K/ Akt/mTOR cascade might be a downstream signal for NLRP3 inflammasome. CONCLUSION Our study provides a new understanding of how diabetes affected progenitor cell-mediated cardiac repair and identifies NLRP3 as a new therapeutic target for improving myocardial infarction repair in inflammatory diseases.
Collapse
Affiliation(s)
- Jia-Peng Li
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing, 210009, People's Republic of China
| | - Shu Qiu
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing, 210009, People's Republic of China
| | - Guang-Jie Tai
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing, 210009, People's Republic of China
| | - Yi-Ming Liu
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210009, People's Republic of China
| | - Wei Wei
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing, 210009, People's Republic of China
| | - Meng-Meng Fu
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing, 210009, People's Republic of China
| | - Pan-Qi Fang
- Department of Pharmacy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Joseph Nicolao Otieno
- Director Institute of Traditional Medicine, Muhimbili University of Health and Allied Sciencea, P.O.BOX 65001, Dar es Salaam, Tanzania
| | - Tungalag Battulga
- School of Pharmacy, Mongolian National University of Medical Sciences, 24210, Ulaanbaatar, Mongolia
| | - Xiao-Xue Li
- Department of Cardiology, School of Medicine, Zhongda Hospital, Southeast University, 87 Ding Jiaqiao, Nanjing, 210009, People's Republic of China
| | - Ming Xu
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
36
|
Florencio-Silva R, Sasso GRDS, Sasso-Cerri E, Cerri PS, Gil CD, de Jesus Simões M. Relationship between autophagy and NLRP3 inflammasome during articular cartilage degradation in oestrogen-deficient rats with streptozotocin-induced diabetes. Ann Anat 2025; 257:152318. [PMID: 39216675 DOI: 10.1016/j.aanat.2024.152318] [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: 05/14/2024] [Revised: 07/23/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Estrogen deficiency and Diabetes mellitus (DM) cause joint tissue deterioration, although the mechanisms are uncertain. This study evaluated the immunoexpression of autophagy and NLRP3-inflammasome markers, in rat articular cartilage with estrogen deficiency and DM. METHODS Twenty rats were sham-operated (SHAM) or ovariectomized (OVX) and equally allocated into four groups: SHAM and OVX groups administered with vehicle solution; SHAM and OVX groups treated with 60 mg/kg/body weight of streptozotocin, intraperitoneally, to induce DM (SHAM-DM and OVX-DM groups). After seven weeks, the rats were euthanized, and their joint knees were processed for paraffin embedding. Sections were stained with haematoxylin-eosin, toluidine blue, safranin-O/fast-green or subjected to picrosirius-red-polarisation method; immunohistochemistry to detect beclin-1 and microtubule-associated protein 1B-light chain 3 (autophagy markers), NLRP3 and interleukin-1β (IL-1β) (inflammasome activation markers), along with matrix metalloproteinase-9 (MMP-9), Nuclear factor-kappa B (NFκB), and Vascular endothelial growth factor A (VEGF-A) were performed. RESULTS Deterioration of articular cartilage and subchondral bone were greater in SHAM-DM and OVX-DM groups. Higher percentages of immunolabeled chondrocytes to NLRP3, IL-1β, MMP-9, NFκB, and VEGF-A, as well as lower percentages of chondrocytes immunolabeled to autophagy markers, were noticed in estrogen-deficient and diabetic groups. These differences were greater in the OVX-DM group. Percentages of immunolabeled chondrocytes showed negative correlation between autophagy markers v.s IL-1β, NLRP-3, MMP-9, NFκB, and VEGF-A, along with positive correlation between VEGF-A vs. MMP-9, NFκB, IL-1β, and NLRP3, and MMP-9 vs. NFκB. CONCLUSIONS In conclusion, autophagy reduction and NLRP3 inflammasome activation in chondrocytes may be implicated in articular cartilage degradation, under estrogen-deficient and DM conditions. Moreover, the combination of estrogen deficiency and DM may potentiate those effects.
Collapse
Affiliation(s)
- Rinaldo Florencio-Silva
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Ginecologia, São Paulo, SP, Brazil; Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, São Paulo, SP, Brazil.
| | - Gisela Rodrigues da Silva Sasso
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, São Paulo, SP, Brazil
| | - Estela Sasso-Cerri
- São Paulo State University (UNESP), School of Dentistry, Araraquara - Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry - Laboratory of Histology and Embryology, Araraquara, SP, Brazil
| | - Paulo Sérgio Cerri
- São Paulo State University (UNESP), School of Dentistry, Araraquara - Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry - Laboratory of Histology and Embryology, Araraquara, SP, Brazil
| | - Cristiane Damas Gil
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, São Paulo, SP, Brazil
| | - Manuel de Jesus Simões
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Ginecologia, São Paulo, SP, Brazil; Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, São Paulo, SP, Brazil
| |
Collapse
|
37
|
Xie Z, Chen Y, Xie J, Du S, Chen R, Zheng Y, You B, Feng M, Liao M, Dai M. Construction with recombinant epitope-expressing baculovirus enhances protective effects of inactivated H9N2 vaccine against heterologous virus. Vet Microbiol 2025; 300:110337. [PMID: 39671758 DOI: 10.1016/j.vetmic.2024.110337] [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: 10/17/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/15/2024]
Abstract
Although the use of inactivated vaccines has kept avian influenza (AI) outbreaks largely under control, they fail to prevent virus shedding. To enhance the efficacy of inactivated H9N2 AIV vaccines (InV), we constructed a multi-epitope recombinant baculovirus (BV-BNT) containing two B cell epitopes and nine T cell epitopes of H9N2 AIV for combined immunization with InV. The results showed that HI titer, IgG and IgM levels, and the percentage of B cells, CD4+ T cells, CD8+ T cells, and CD4+CD8+ T cells were significantly higher in the InV+BV-BNT immunization group than the InV immunization group. Besides, the expression levels of IL-1β, IFN-γ, IFN-α, IL-4, IL-13, and CXCLi1 were significantly higher in the InV+BV-BNT group than the InV group. Moreover, four conservative peptides (NP182-190, NP455-463, NS198-106, and NP380-393) significantly stimulated splenocytes to express IFN-γ in the InV+BV-BNT group instead of InV group. After heterologous virus challenging, the percentages of CD4+ T and CD8+ T cells were significantly upregulated in the InV+BV-BNT group compared to Inv group at 3 DPI. Viral loads in oropharyngeal of the InV+BV-BNT group was significantly lower than that in the InV group at 3 days post-infection (DPI). Furthermore, compared to the InV group, the virus positivity rate of oropharyngeal and cloacal swabs in the InV+BV-BNT group was lower at 5 DPI, with none positive at 7 DPI. Hence, this study indicated that the combined immunization of InV and BV-BNT could induce stronger humoral and cellular immune responses, shorten the detoxification period and reduce viral load compared to Inv alone, which suggests BV-BNT could act as a supplementary vaccine to potentially address the protection deficiency of the H9N2 inactivated vaccine.
Collapse
Affiliation(s)
- Zimin Xie
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Yingyi Chen
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Jun Xie
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Shanyao Du
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Rongmao Chen
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Yuqin Zheng
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Bowen You
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Min Feng
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Ming Liao
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; UK-China Centre of Excellence for Research on Avian Diseases, Guangzhou 510642, PR China.
| | - Manman Dai
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; UK-China Centre of Excellence for Research on Avian Diseases, Guangzhou 510642, PR China.
| |
Collapse
|
38
|
Li L, Xu T, Qi X. Balanced regulation of ROS production and inflammasome activation in preventing early development of colorectal cancer. Immunol Rev 2025; 329:e13417. [PMID: 39523732 DOI: 10.1111/imr.13417] [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] [Indexed: 11/16/2024]
Abstract
Reactive oxygen species (ROS) production and inflammasome activation are the key components of the innate immune response to microbial infection and sterile insults. ROS are at the intersection of inflammation and immunity during cancer development. Balanced regulation of ROS production and inflammasome activation serves as the central hub of innate immunity, determining whether a cell will survive or undergo cell death. However, the mechanisms underlying this balanced regulation remain unclear. Mitochondria and NADPH oxidases are the two major sources of ROS production. Recently, NCF4, a component of the NADPH oxidase complex that primarily contributes to ROS generation in phagocytes, was reported to balance ROS production and inflammasome activation in macrophages. The phosphorylation and puncta distribution of NCF4 shifts from the membrane-bound NADPH complex to the perinuclear region, promoting ASC speck formation and inflammasome activation, which triggers downstream IL-18-IFN-γ signaling to prevent the progression of colorectal cancer (CRC). Here, we review ROS signaling and inflammasome activation studies in colitis-associated CRC and propose that NCF4 acts as a ROS sensor that balances ROS production and inflammasome activation. In addition, NCF4 is a susceptibility gene for Crohn's disease (CD) and CRC. We discuss the evidence demonstrating NCF4's crucial role in facilitating cell-cell contact between immune cells and intestinal cells, and mediating the paracrine effects of inflammatory cytokines and ROS. This coordination of the signaling network helps create a robust immune microenvironment that effectively prevents epithelial cell mutagenesis and tumorigenesis during the early stage of colitis-associated CRC.
Collapse
Affiliation(s)
- Longjun Li
- Key Laboratory for Experimental Teratology of the Ministry of Education, Advanced Medical Research Institute, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Tao Xu
- Key Laboratory for Experimental Teratology of the Ministry of Education, Advanced Medical Research Institute, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiaopeng Qi
- Key Laboratory for Experimental Teratology of the Ministry of Education, Advanced Medical Research Institute, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
39
|
Yin W, Wang JH, Liang YM, Liu KH, Chen Y, Chen Y. Neferine Targeted the NLRC5/NLRP3 Pathway to Inhibit M1-type Polarization and Pyroptosis of Macrophages to Improve Hyperuricemic Nephropathy. Curr Mol Med 2025; 25:90-111. [PMID: 38549521 DOI: 10.2174/0115665240272051240122074511] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 02/19/2025]
Abstract
BACKGROUND Neferine (Nef) has a renal protective effect. This research intended to explore the impact of Nef on hyperuricemic nephropathy (HN). METHODS Adenine and potassium oxonate were administered to SD rats to induce the HN model. Bone marrow macrophages (BMDM) and NRK-52E were used to construct a transwell co-culture system. The polarization of BMDM and apoptosis levels were detected using immunofluorescence and flow cytometry. Renal pathological changes were detected using hematoxylin-eosin (HE) and Masson staining. Biochemical methods were adopted to detect serum in rats. CCK-8 and EDU staining were used to assess cell activity and proliferation. RT-qPCR and western blot were adopted to detect NLRC5, NLRP3, pyroptosis, proliferation, and apoptosis-related factor levels. RESULTS After Nef treatment, renal injury and fibrosis in HN rats were inhibited, and UA concentration, urinary protein, BUN, and CRE levels were decreased. After Nef intervention, M1 markers, pyroptosis-related factors, and NLRC5 levels in BMDM stimulated with uric acid (UA) treatment were decreased. Meanwhile, the proliferation level of NRK-52E cells co-cultured with UA-treated BMDM was increased, but the apoptosis level was decreased. After NLRC5 overexpression, Nef-induced regulation was reversed, accompanied by increased NLRP3 levels. After NLRP3 was knocked down, the levels of M1-type markers and pyroptosis-related factors were reduced in BMDM. CONCLUSION Nef improved HN by inhibiting macrophages polarized to M1-type and pyroptosis by targeting the NLRC5/NLRP3 pathway. This research provides a scientific theoretical basis for the treatment of HN.
Collapse
Affiliation(s)
- Wei Yin
- Department of Nephrology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410002, China
| | - Jin-Hua Wang
- Department of Nephrology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410002, China
| | - Yu-Mei Liang
- Department of Nephrology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410002, China
| | - Kang-Han Liu
- Department of Nephrology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410002, China
| | - Ying Chen
- Department of Nephrology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410002, China
| | - Yusa Chen
- Department of Nephrology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410002, China
| |
Collapse
|
40
|
Zhang L, Gao Y, Zhou H, Liang X, Jiang X, Gong W, Sun Y, Zhang D, Wang X, Nauwynck H, Bai J, Jiang P. PRRSV-2 nsp2 Ignites NLRP3 inflammasome through IKKβ-dependent dispersed trans-Golgi network translocation. PLoS Pathog 2025; 21:e1012915. [PMID: 39869629 PMCID: PMC11801707 DOI: 10.1371/journal.ppat.1012915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/06/2025] [Accepted: 01/17/2025] [Indexed: 01/29/2025] Open
Abstract
The NLRP3 inflammasome is a fundamental component of the innate immune system, yet its excessive activation is intricately associated with viral pathogenesis. Porcine reproductive and respiratory syndrome virus type 2 (PRRSV-2), belonging to the family Arteriviridae, triggers dysregulated cytokine release and interstitial pneumonia, which can quickly escalate to acute respiratory distress and death. However, a mechanistic understanding of PRRSV-2 progression remains unclear. Here, we screen that PRRSV-2 nsp2 activates the NLRP3 inflammasome, thereby instigating a state of hyperinflammation. Mechanistically, PRRSV-2 nsp2 interacts with the nucleotide-binding and oligomerization (NACHT) domain of NLRP3, augmenting IKKβ recruitment to driving NLRP3 translocation to the dispersed trans-Golgi network (dTGN) for oligomerization. This process facilitates ASC polymerization, culminating in the activation of the NLRP3 inflammasome. In addition, the IKKβ-dependent NLRP3 translocation to the dTGN is pivotal for pseudorabies virus (PRV) and encephalomyocarditis virus (EMCV)-induced inflammatory responses. Collectively, these results elucidate a novel mechanism of NLRP3 inflammasome activation during PRRSV-2 infection, providing valuable insights into PRRSV-2 pathogenesis.
Collapse
Affiliation(s)
- Lujie Zhang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yanni Gao
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Haoran Zhou
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiao Liang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiaolin Jiang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Wenqin Gong
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yangyang Sun
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Desheng Zhang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xianwei Wang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hans Nauwynck
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan Merelbeke, Belgium
| | - Juan Bai
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, PR China
| | - Ping Jiang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, PR China
| |
Collapse
|
41
|
Peñin-Franch A, García-Vidal JA, Gómez AI, Escolar-Reina P, Medina-Mirapeix F, Pelegrín P. The total electric charge and time of application of galvanic currents to macrophages can optimize the release of IL-1β with low cell death. Sci Rep 2024; 14:30871. [PMID: 39730677 DOI: 10.1038/s41598-024-81848-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 11/29/2024] [Indexed: 12/29/2024] Open
Abstract
Galvanic current has been emerging as a novel therapy to regenerate chronic tissue lesions, including musculoskeletal and dermatological lesions. Recently, the NLRP3 inflammasome and IL-1β release have been identified as a signaling pathway triggered upon galvanic current application. However, the parameters for the clinical application of galvanic current remain subjective to the experience of the facultative in charge. In this study we used an in vitro model of macrophage culture and application of different combinations of the parameters of galvanic current to study IL-1β production and cell death. Increasing electric charge of galvanic current induces the release of IL-1β, but electric charges equal or higher to 144 mC also increase cell death. The release of IL-1β have a substantial variation within different electric charge of galvanic currents, being increased by decreasing the current and increasing the time of current application. Within the range of current intensities studied, the most optimal protocol for maximizing IL-1β release without inducing cell death was identified at electric charges equal to or near 144 mC, applied over a total duration of approximately 25 s. Our findings lay the groundwork for future in vivo studies assessing different electric charge of galvanic current, with the aim of yielding clinically relevant outcomes.
Collapse
Affiliation(s)
| | - José Antonio García-Vidal
- Biomedical Research Institute of Murcia IMIB-Pascual Parrilla, Murcia, Spain
- Department of Physical Therapy, University of Murcia, Murcia, Spain
| | - Ana Isabel Gómez
- Biomedical Research Institute of Murcia IMIB-Pascual Parrilla, Murcia, Spain
| | - Pilar Escolar-Reina
- Biomedical Research Institute of Murcia IMIB-Pascual Parrilla, Murcia, Spain
- Department of Physical Therapy, University of Murcia, Murcia, Spain
| | - Francesc Medina-Mirapeix
- Biomedical Research Institute of Murcia IMIB-Pascual Parrilla, Murcia, Spain.
- Department of Physical Therapy, University of Murcia, Murcia, Spain.
| | - Pablo Pelegrín
- Biomedical Research Institute of Murcia IMIB-Pascual Parrilla, Murcia, Spain.
- Department of Biochemistry and Molecular Biology B and Immunology, University of Murcia, Murcia, Spain.
| |
Collapse
|
42
|
Li C, Ma Z, Wei X, Wang Y, Wu J, Li X, Sun X, Ding Z, Yang C, Zou Y. Bufalin Ameliorates Myocardial Ischemia/Reperfusion Injury by Suppressing Macrophage Pyroptosis via P62 Pathway. J Cardiovasc Transl Res 2024:10.1007/s12265-024-10577-9. [PMID: 39733202 DOI: 10.1007/s12265-024-10577-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 11/29/2024] [Indexed: 12/30/2024]
Abstract
Bufalin, which is isolated from toad venom, exerts positive effects on hearts under pathological circumstance. We aimed to investigate the effects and mechanisms of bufalin on myocardial I/R injury. In vivo, bufalin ameliorated myocardial I/R injury, which characteristics with better ejection function, decreased infarct size and less apoptosis. The levels of pyroptotic proteins were increased in I/R-treated macrophages and inflammatory cytokines expressed more in I/R-induced mouse, which could be attenuated by bufalin. Bufalin also reduced H/R-treated macrophage pyroptosis in vitro. Autophagic flux blockage and ROS accumulation were reduced by bufalin in impaired macrophages. Overexpression of p62 abrogated the anti-proptosis and anti-oxidative effects of bufalin. The levels of apoptosis related proteins were changed and TUNEL-positive ratio was raised in cardiomyocytes that received conditioned medium treatment with H/R-treated macrophages, while bufalin pretreatment could reduce apoptosis. These findings indicate that bufalin may attenuate myocardial I/R injury by suppressing macrophage pyroptosis via P62 pathway.
Collapse
Affiliation(s)
- Chang Li
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Zhen Ma
- Institutes of Biomedical Sciences, Fudan University, 131 Dong'an Road, Shanghai, 200032, China
| | - Xiang Wei
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai, 200032, China
| | - Ying Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Jian Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Xuan Li
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Xiaolei Sun
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Zhiwen Ding
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
- Institutes of Biomedical Sciences, Fudan University, 131 Dong'an Road, Shanghai, 200032, China.
| | - Cheng Yang
- Department of Cardiac Surgery, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
- Institutes of Biomedical Sciences, Fudan University, 131 Dong'an Road, Shanghai, 200032, China.
- State Key Laboratory of Genetic Engineering, Fudan University, 138 Yixueyuan Road, Shanghai, 200438, China.
| |
Collapse
|
43
|
Li J, Song L, Li H, Gao Y, Chen T, Zhang Z, Hou H, Ye Z, Zhang G. Aerosol Inhalation of Luteolin-7-O-Glucuronide Exerts Anti-Inflammatory Effects by Inhibiting NLRP3 Inflammasome Activation. Pharmaceuticals (Basel) 2024; 17:1731. [PMID: 39770573 PMCID: PMC11677241 DOI: 10.3390/ph17121731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/14/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Luteolin-7-O-glucuronide (L7Gn) is a flavonoid isolated from numerous traditional Chinese herbal medicines that exerts anti-inflammatory effects. Previous research has revealed that aerosol inhalation is the most straightforward way of administration for the delivery of respiratory agents. Thus far, the impact of aerosol inhalation of L7Gn on lung inflammation and the underlying mechanisms remain unknown. Methods: The real-time particle size for L7Gn aerosol inhalation was detected by the Spraytec spray droplet size measurement system, including transmission and size diameters. The acute lung injury (ALI) rat model was induced by aerosol inhalation of LPS to evaluate the protective effect of L7Gn. The inhibitory effect of NLRP3 inflammasome activation assays was conducted in LPS-induced MH-S cells. Elisa, Western blotting, and RT-PCR were utilized to investigate the expression of NLRP3 inflammasome-relevant proteins and genes. Results: In this study, we found that inhalation of L7Gn aerosol significantly reduced pulmonary injury by inhibiting inflammatory infiltration and enhancing lung function. Meanwhile, the NLR family pyrin domain containing 3 (NLRP3) inflammasome was activated dramatically, accompanied by upregulated expression of IL-1β and IL-18, both in the ALI rat model and in LPS-induced MH-S cells. Moreover, L7Gn was found to significantly downregulate the expression of NLRP3, ASC, caspase-1, and cleaved caspase-1, which are critical components of the NLRP3 inflammasome, as well as the expression of IL-1β and IL-18. Conclusions: Based on our findings, L7Gn could exert anti-inflammatory effects by inhibiting NLRP3 inflammasome activation, which may emerge as potential therapeutic agents for the treatment of ALI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zuguang Ye
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiao Street, Dongzhimen, Dongcheng District, Beijing 100700, China; (J.L.); (L.S.); (H.L.); (Y.G.); (T.C.); (Z.Z.); (H.H.)
| | - Guangping Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiao Street, Dongzhimen, Dongcheng District, Beijing 100700, China; (J.L.); (L.S.); (H.L.); (Y.G.); (T.C.); (Z.Z.); (H.H.)
| |
Collapse
|
44
|
Han X, Fu X, Guo W, Liu Y, Sun J, Wang T, Yang W. Ghrelin Inhibits Inflammasomes Activation in Astrocytes, Alleviates Pyroptosis, and Prevents Lipopolysaccharide-induced Depression-like Behavior in Mice. Inflammation 2024:10.1007/s10753-024-02190-4. [PMID: 39702621 DOI: 10.1007/s10753-024-02190-4] [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: 09/05/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 12/21/2024]
Abstract
Depression is the leading cause of disability worldwide and places a significant burden on society. Neuroinflammation is closely associated with the pathophysiology of depression. Increasing evidence suggests that astrocytes, as the most abundant glial cells in the brain, are involved in the occurrence and development of depression due to morphological abnormalities and dysfunction. Astrocytes express the NOD-like receptor protein 2 (NLRP2) and NLRP3 inflammasomes, and the activation of inflammasomes induces pyroptosis. Ghrelin, a gastrointestinal peptide, plays vital role in regulating inflammation and alleviating stress. Therefore, we proposed a hypothesis that ghrelin inhibits the activation of inflammasomes on astrocytes, reduces pyroptosis, and consequently prevents depression. We used lipopolysaccharide (LPS)-induced mouse depression model and cultured primary astrocytes in vitro to explore the mechanism of the antidepressant effect of ghrelin. Our results showed that ghrelin effectively inhibited acute inflammatory responses and damage in the hippocampus and prefrontal cortex. The activation of NLRP2 and NLRP3 in astrocytes induced by LPS was significantly inhibited by ghrelin. Pretreatment with ghrelin effectively suppressed LPS-induced upregulation of pyroptosis-related proteins and mRNA. Ghrelin alleviated cell membrane pore formation and cell swelling, ultimately improved LPS-induced depression-like behavior. In vitro, ghrelin prevented the LPS-induced upregulation of pyroptosis-related proteins and mRNA expression in astrocytes, and inhibited the initiation and assembly of NLRP2 and NLRP3. Ghrelin exhibits antidepressant effects, inhibits inflammasomes activation in astrocytes, and prevents pyroptosis, suggesting a novel strategy for treating depression. This groundbreaking study reveals new avenues for targeting potential therapeutic interventions to alleviate depression.
Collapse
Affiliation(s)
- Xiaoou Han
- The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China.
| | - Xiying Fu
- The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Wanxu Guo
- The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Yaqi Liu
- The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Jiangjin Sun
- The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Tian Wang
- The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Wei Yang
- The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China.
| |
Collapse
|
45
|
Tan Y, Chen S, Gao T, Wang S, Zhou X, Liu M. Exploring the role of NLRP3 infalmmasome in diabetes: a literature review and bibliometric analysis. Front Endocrinol (Lausanne) 2024; 15:1443798. [PMID: 39717099 PMCID: PMC11663631 DOI: 10.3389/fendo.2024.1443798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 11/21/2024] [Indexed: 12/25/2024] Open
Abstract
Background Diabetes has emerged as the foremost public health challenge of the 21st century, with a notable shift towards managing it through an inflammatory lens. This study seeks to investigate the role of NLRP3 infalmmasome in diabetes over the past ten years, leveraging bibliometric analysis to pinpoint prevailing trends, underscore critical focal points, and establish a roadmap for subsequent research endeavors. Method A literature search was conducted based on the SCI-E database, and all recorded results were downloaded in plain text format for subsequent analysis. The analysis was carried out using Vosviewer1.6.18, citespace6.3R1, and Microsoft Excel 2021 software, focusing on the following terms: country, institution, author, journal, references, and keywords. Results From January 1, 2014, to December 31, 2023, a total of 1373 articles were retrieved, with China, the United States, and Italy contributing the majority of records. Harbin Medical University, Nanjing Medical University, and Central South University stand as the top three most productive institutions. "International Journal of Molecular Sciences" leads the way with the highest number of publications, closely followed by "Frontiers in Immunology" and "Frontiers in Pharmacology." Authors Wang Wei boast the most publications, closely followed by Li Xiang and Wang Yan. Within the superimposed keyword network, four primary clusters emerge: (1) exploring the link between NLRP3 infalmmasome and inflammatory diseases like diabetes; (2) investigating the cellular-level pathogenesis of diabetes-related conditions; (3) examining diabetes characteristics and associated suppression techniques; (4) studying cell morphology alterations, including pyroptosis. Over the past five years, key topics in this field have revolved around the "heart", "damage", "caspase 1 activation", "NLRP3", and "diabetic kidney disease". Conclusion This paper has identified the hot spots and trends concerning the role of NLRP3 infalmmasome in diabetes, thereby providing a valuable reference for future research. Furthermore, it is anticipated that pyroptosis and diabetes-related diseases will become frontier research topics that may garner significant attention in the coming years.
Collapse
Affiliation(s)
- Yi Tan
- Departments of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Shaotao Chen
- Departments of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Tianjiao Gao
- Office of Scientific Research, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Sixian Wang
- Departments of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xinfeng Zhou
- Departments of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Mingjun Liu
- Departments of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
46
|
Wei X, Wu J, Pi X, Zhang Q, Tian J, Qi Z. Characterization of NLRP3 inflammasome components in the endangered Chinese giant salamander (Andrias davidianus). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 161:105263. [PMID: 39265857 DOI: 10.1016/j.dci.2024.105263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Chinese giant salamander (Andrias davidianus) is the largest extant urodela species and has unique evolutionary position. Studying the immune system of Chinese giant salamander contributes to understanding the evolution of immune systems of vertebrates. The NLR-related protein 3 (NLRP3) inflammasome comprised of NLRP3, ASC and caspase-1 play important roles in the host innate immunity. However, little is know about the NLRP3 inflammasome components in Chinese giant salamander. In this study, the NLRP3, apoptosis-associated speck-like protein (ASC) and caspase-1 (adaNLRP3, adaASC and adaCaspase-1) were characterized from Chinese giant salamander. The proteins of these three genes shared similar motifs and structures with their mammalian counterparts, with a PYD motif, a nucleotide-binding domain (NACHT) motif, and four leucine-rich repeat domain (LRR) motifs identified in adaNLRP3, a pyrin domain (PYD) motif and a caspase recruitment domain (CARD) motif in adaASC, and a CARD motif and a CASc motif in adaCaspase-1. These three genes were constitutively expressed in the skin, heart, lung, kidney, muscle, brain, spleen, and liver of Chinese giant salamander. Following Aeromonas hydrophia infection, all the three genes were up-regulated in various tissues. Molecular docking analysis revealed that the key residues involved in forming the adaNLRP3/adaASC complex were located in the PYD motifs, and that involved in forming the adaASC/adaCaspase-1 complex were located in the CARD motifs. Further analysis revealed that the hydrogen bonds and salt bridges had crucial roles in the formation of adaNLRP3/acaASC and adaASC/adaCaspase-1 complexes. To the best of our knowledge, this is the first report on the NLRP3 inflammasome components in Chinese giant salamander which will be helpful in further understanding the function of the NLRP3 inflammasome and in elucidating its role in the immune response to microbes.
Collapse
Affiliation(s)
- Xuan Wei
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, Yancheng Institute of Technology, Yancheng, Jiangsu Province, 224051, China
| | - Jianxiong Wu
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, Yancheng Institute of Technology, Yancheng, Jiangsu Province, 224051, China
| | - Xiangyu Pi
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, Yancheng Institute of Technology, Yancheng, Jiangsu Province, 224051, China
| | - Qihuan Zhang
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, Yancheng Institute of Technology, Yancheng, Jiangsu Province, 224051, China
| | - Jingyu Tian
- Marine Science Research Institute of Shandong Province, Qingdao, 266104, China
| | - Zhitao Qi
- Jiangsu Key Laboratory of Biochemistry and Biotechnology of Marine Wetland, Yancheng Institute of Technology, Yancheng, Jiangsu Province, 224051, China.
| |
Collapse
|
47
|
Zeng F, Lai Y, Huang Y, Zhu F, Gao J, Chen Z, Zeng L, Feng M, Qiu P, Yuan S, Deng G. Shikonin from lithospermum erythrorhizon induces pyroptosis in trophoblast cells by activating the CTSB-NLRP3 inflammasome. Ann Med 2024; 56:2394584. [PMID: 39183455 PMCID: PMC11348813 DOI: 10.1080/07853890.2024.2394584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND With the decline of global fertility, drug therapeutic of ectopic pregnancy is of great significance. Lithospermum erythrorhizon is using for embryo killing as herbal medicine. Shikonin is the critical nucleus of Lithospermum erythrorhizon; however, the mechanism is still unclear. The study aimed to explore the mechanism of shikonin against ectopic pregnancy. MATERIAL AND METHODS In this study, we examined the viability and LDH release of HTR-8/SVneo cells by assays, observed pore formation in cell membranes by microscopy imaging and PI staining, and IL-1β release by WB and ELISA assay kit. Then, we used network pharmacology to analyse the potential interaction between shikonin, ectopic pregnancy and pyroptosis and used molecular docking techniques to verify interactions between shikonin and core common targets. Finally, western blotting and immunofluorescence assay were used to explore the mechanism of shikonin-inducing pyroptosis of HTR-8/SVneo cells. RESULTS Shikonin could cause a significant inhibition of HTR-8/SVneo cell viability in a concentration- and time-dependent manner. In HTR-8/SVneo cells, shikonin-induced cell swelling, bubble formation, an increase in the release of lactate dehydrogenase (LDH) and up-regulation of several pyroptosis-associated factors. And network pharmacology showed that The main targets of shikonin-ectopic pregnancy-pyroptosis were IL-1β and caspase-1, and molecular docking results showed that shikonin can closely bind to IL-1β, caspase-1 and GSDMD. Additionally, the necroptosis inhibitor GSK'872 could not suppress the expression of mature-IL-1β and prevent the pyroptosis phenotype from developing. However, the nucleotide oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inhibitor MCC-950 could downregulate the expression of pyroptosis-associated factors and prevent the pyroptosis phenotype from developing. Shikonin led to an elevation in the expression of cathepsin B (CTSB), and the CTSB inhibitor CA-074 abolished pyroptosis induced by shikonin; however, the NLRP3 inhibitor MCC-950 could not inhibit the expression of CTSB. CONCLUSIONS Our results suggest that shikonin activates CTSB to induce NLRP3-dependent pyroptosis in HTR-8/SVneo cells. This study has important clinical implications for the treatment of ectopic pregnancy.
Collapse
Affiliation(s)
- Fuling Zeng
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Gynecology, Guangzhou Baiyun District Maternal and Child Health Hospital, Guangzhou, China
| | - Yuling Lai
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, China
| | - Yanxi Huang
- Department of Gynecology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fangfang Zhu
- Department of Gynecology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Jie Gao
- Department of Gynecology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhenyue Chen
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lihua Zeng
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min Feng
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pin Qiu
- Department of Gynecology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuo Yuan
- Department of Gynecology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Gaopi Deng
- Department of Gynecology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
48
|
Soares-Bezerra RJ, da Silva Ferreira NC, de Almeida Alves TM, Zani CL, Rosa LH, Calheiros AS, de Souza CZ, Miranda JAA, Lima-Quaresma KRF, Alves LA, da Silva Frutuoso V. The analgesic and gastroprotective activities of the three fungal extracts and their possible correlation with the inhibition of the P2X7 receptor. Biomed Pharmacother 2024; 181:117657. [PMID: 39515112 DOI: 10.1016/j.biopha.2024.117657] [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: 06/17/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
P2X7 is a purinergic receptor physiologically activated by extracellular ATP. Its activation induces proinflammatory responses, including cytokine release, reactive oxygen species formation, and cell death. Previous in vivo experimental models demonstrated that P2X7 blockade has anti-inflammatory effects; however, there are no drugs used in clinical therapy that act on the P2X7 receptor. In the context of inflammatory diseases, nonsteroidal anti-inflammatory drugs (NSAIDs) are widely used as the first-line treatment; however, their major side effects include stomach ulcer formation, which increases patient morbidity and mortality. Here, we analyzed for the first time the analgesic and gastroprotective activities of three fungal extracts that showed antagonistic effects on P2X7 in vitro. The Antarctic fungal extracts obtained from Vishniacozyma victoriae, Metschnikowia australis, and Ascomycota sp. were tested in animal models of acute pain and ethanol-induced ulceration. These three extracts reduced paw licking by approximately 50 %, which is related to pain behavior, and reduced the number of stomach ulcers 3-7 times compared with the control (70 % ethanol), making them more efficient than the lansoprazole, an NSAID drug, and Brilliant Blue G (BBG), a known P2X7 antagonist, which only halves the number of ulcers. Furthermore, the extracts also protected the gastric mucosa and significantly reduced the levels of liver and renal enzymes compared with those in the ethanol group. Taken together, the fungal extracts presented both analgesic and possibly anti-inflammatory activities and had a protective effect on the gastric epithelium.
Collapse
Affiliation(s)
- Rômulo José Soares-Bezerra
- Laboratory of Cellular Communication, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ 21040-360, Brazil.
| | | | - Tânia Maria de Almeida Alves
- Laboratory of Chemistry of Bioactive Natural Products, René Rachou Research Center, Oswaldo Cruz Foundation, Belo Horizonte, MG 30190-009, Brazil
| | - Carlos Leomar Zani
- Laboratory of Chemistry of Bioactive Natural Products, René Rachou Research Center, Oswaldo Cruz Foundation, Belo Horizonte, MG 30190-009, Brazil
| | - Luiz Henrique Rosa
- Laboratory of Polar Microbiology and Tropical Connections, Federal University of Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Andrea Surrage Calheiros
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ 21040-360, Brazil
| | - Cristiane Zanon de Souza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ 21040-360, Brazil
| | | | | | - Luiz Anastacio Alves
- Laboratory of Cellular Communication, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ 21040-360, Brazil
| | - Válber da Silva Frutuoso
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ 21040-360, Brazil
| |
Collapse
|
49
|
Zhao P, Ning J, Huang J, Huang X. Mechanism of Resveratrol on LPS/ATP-induced pyroptosis and inflammatory response in HT29 cells. Autoimmunity 2024; 57:2427094. [PMID: 39534992 DOI: 10.1080/08916934.2024.2427094] [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: 06/17/2024] [Revised: 10/25/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
Pyroptosis plays an important role in maintenance of intestinal homeostasis, the abnormal activation of NOD-like receptor thermal protein domain-associated protein 3 (NLRP3) inflammasome can promote the event and development of ulcerative colitis (UC). Its protective effects such as inhibiting pyroptosis in various inflammation-related diseases have been demonstrated, but whether resveratrol (RES) can also alleviate the progression of the disease by inhibiting pyroptosis in UC and the mechanism have rarely been studied. In this study, lipopolysaccharide (LPS) combined with adenosine triphosphate (ATP) was used to induce HT29 human colon cancer cells to construct an intestinal epithelial cell pyroptosis and inflammation model in vitro to investigate the anti-inflammatory effect of RES, reveal the regulatory mechanism of RES on pyroptosis, and provide a new theoretical basis for the treatment of UC. In vitro experiences, HT29 cells were dividing into control group, LPS/ATP group, RES low-dose group, RES high-dose group, NF-κB inhibitor pyrrolidine dithiocarbamate group (PDTC group), and LPS/ATP+PDTC group. The mRNA expressions of pyroptosis-related indicators such as NLRP3, apoptosis-associated speck-like protein containing CARD (ASC), Caspase-1(CASP1), IL-18, IL-1β, and inflammatory factors such as TNF-α and IL-6 were detected by qRT-PCR. The protein expressions of pyroptosis-related indicators NLRP3, ASC, CASP1, IL-18, IL-1β, NF-κB-p65 in the nucleus, and IκBα and p-IκBα in the cytoplasm were detected by Western blot. Immunofluorescence saw the distribution and expression of NLRP3, ASC and NF-κB-p65 protein in each group. The morphology and degree of pyroptosis in each group were observed by transmission electron microscope. The results showed that compared with the control group, the pyroptosis-related proteins including NLRP3, ASC, CASP1, IL-18, IL-1β, and inflammatory factors including TNF-α and IL-6 in the LPS/ATP group were increased, and LPS/ATP activated the activity of NF-κB signaling pathway. Compared with the LPS/ATP group, RES downregulated the expression of pyroptosis-related proteins and inflammatory factors in HT29 cells, and inhibited the activation of the NF-κB signaling pathway in HT29 cells pyroptosis. RES down-regulates the pyroptosis of HT29 cells induced by LPS/ATP and the expression of pyroptosis-related indicators NLRP3, ASC, CASP1, IL-18, IL-1β and inflammatory factors TNF-α and IL-6 in the inflammatory response and inhibits the occurrence of pyroptosis. The mechanism is related to the inhibition of NF-κB pathway activity.
Collapse
Affiliation(s)
- Peizhuang Zhao
- Department of Geriatrics and Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jiajia Ning
- Department of Geriatrics and Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jun Huang
- Department of Geriatrics and Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xue Huang
- Department of Geriatrics and Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
50
|
Mazhar F, Faucon AL, Fu EL, Szummer KE, Mathisen J, Gerward S, Reuter SB, Marx N, Mehran R, Carrero JJ. Systemic inflammation and health outcomes in patients receiving treatment for atherosclerotic cardiovascular disease. Eur Heart J 2024; 45:4719-4730. [PMID: 39211962 PMCID: PMC11578643 DOI: 10.1093/eurheartj/ehae557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/11/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND AND AIMS The burden and outcomes of inflammation in patients with atherosclerotic cardiovascular disease (ASCVD) are not well defined beyond the controlled settings of trials and research cohorts. METHODS This was an observational study of ASCVD adults undergoing C-reactive protein testing in Stockholm's healthcare (2007-21). After excluding C-reactive protein tests associated with acute illness or medications/conditions that bias C-reactive protein interpretation, systemic inflammation was evaluated over a 3-month ascertainment window. Determinants of C-reactive protein ≥ 2 mg/L were explored with logistic regression. C-reactive protein categories were compared via negative-binomial/Cox regression for subsequent healthcare resource utilization and occurrence of major adverse cardiovascular events, heart failure hospitalization, and death. RESULTS A total of 84 399 ASCVD adults were included (46% female, mean age 71 years, 59% with C-reactive protein ≥ 2 mg/L). Female sex, older age, lower kidney function, albuminuria, diabetes, hypertension, and recent anaemia were associated with higher odds of C-reactive protein ≥ 2 mg/L. The use of renin-angiotensin system inhibitors, antiplatelets, and lipid-lowering therapy was associated with lower odds. Over a median of 6.4 years, compared with C-reactive protein < 2 mg/L, patients with C-reactive protein ≥ 2 mg/L had higher rates of hospitalizations, days spent in hospital, outpatient consultations, and dispensed medications (P < .05 for all). They also had a higher rate of major adverse cardiovascular events [hazard ratio (HR) 1.30; 95% confidence interval (CI) 1.27-1.33], heart failure (HR 1.24; 95% CI 1.20-1.30), and death (HR 1.35; 95% CI 1.31-1.39). Results were consistent across subgroups and granular C-reactive protein categories and robust to the exclusion of extreme C-reactive protein values or early events. CONCLUSIONS Three in five adults with ASCVD have systemic inflammation, which is associated with excess healthcare resource utilization and increased rates of cardiovascular events and death.
Collapse
Affiliation(s)
- Faizan Mazhar
- Department of Medical Epidemiology and Biostatistics, Campus Solna, Karolinska Institutet, Nobels väg 12A, 171 65 Stockholm, Sweden
| | - Anne-Laure Faucon
- Department of Medical Epidemiology and Biostatistics, Campus Solna, Karolinska Institutet, Nobels väg 12A, 171 65 Stockholm, Sweden
| | - Edouard L Fu
- Department of Medical Epidemiology and Biostatistics, Campus Solna, Karolinska Institutet, Nobels väg 12A, 171 65 Stockholm, Sweden
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Karolina E Szummer
- Department of Cardiology, Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | | | | | | | - Nikolaus Marx
- Department of Internal Medicine I, RWTH Aachen University, Aachen, Germany
| | - Roxana Mehran
- Mount Sinai School of Medicine, Mount Sinai Health System, New York City, NY, USA
| | - Juan-Jesus Carrero
- Department of Medical Epidemiology and Biostatistics, Campus Solna, Karolinska Institutet, Nobels väg 12A, 171 65 Stockholm, Sweden
- Division of Nephrology, Department of Clinical Sciences, Danderyd Hospital, Danderyd, Sweden
| |
Collapse
|