1
|
Qin X, Lu F, Wan J, Teng X, Jin S, Xiao L, Xue H, Guo Q, Tian D, Wu Y. Hydrogen sulfide preserves the function of senescent endothelium through SIRT2 mediated inflammatory inhibition. J Mol Cell Cardiol 2025; 203:10-21. [PMID: 40209983 DOI: 10.1016/j.yjmcc.2025.04.005] [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: 10/18/2024] [Revised: 03/30/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Endothelial aging is an independent risk factor of cardiovascular diseases, and this study aims to explore the mechanism of endothelial aging. We first applied two animal aging models and two cellular aging models to observe the characteristics of senescent endothelium at the morphological, functional, and molecular levels. It was confirmed that the aging of endothelial cells was accompanied by activation of Nod like receptor protein 3 (NLRP3) inflammasome pathway, reduced levels of hydrogen sulfide (H2S) and sirtuin2 (SIRT2) activity. Endothelial specific knockout of cystathionine-γ-lyase (CSE) led to premature aging of blood vessels, and excessive activation of the SIRT2/NLRP3 inflammasome. Finally, H2S supplementation improved vascular and endothelial cell function, normalized inflammatory cytokine levels, and thereby reversed endothelial aging through SIRT2/NLRP3 mediated pathway. In this study, we found that the decrease in SIRT2 activity in aging endothelial cells increased the level of NLRP3 inflammasome and H2S inhibited inflammation to improve endothelial aging through the SIRT2/NLRP3 pathway. This provided H2S could be a new target for improving endothelial aging, and offered new strategies for defending human aging.
Collapse
Affiliation(s)
- Xueyuan Qin
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China; School of Clinical Medicine, North China University of Science and Technology, Tangshan 063210, China
| | - Fan Lu
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Jie Wan
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Xu Teng
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Sheng Jin
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Lin Xiao
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Hongmei Xue
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Qi Guo
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Danyang Tian
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China.
| | - Yuming Wu
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
2
|
Huang CL, Qi-En S, Cen XF, Ye T, Qu HS, Chen SJ, Liu D, Xia HG, Xu CF, Zhu JS. TJ0113 attenuates fibrosis in metabolic dysfunction-associated steatohepatitis by inducing mitophagy. Int Immunopharmacol 2025; 156:114678. [PMID: 40252468 DOI: 10.1016/j.intimp.2025.114678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/01/2025] [Accepted: 04/13/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatohepatitis (MASH) fibrosis is a liver disease accompanied by inflammatory cell infiltration. There is growing evidence that insufficient mitophagy can exacerbate inflammation and liver fibrosis (LF). TJ0113 is a novel mitophagy inducer. The study aimed to explore the role of TJ0113 in ameliorating fibrosis in MASH and its mechanisms. METHODS A high-fat diet (HFD)-induced MASH mice model and a transforming growth factor (TGF)-β1-induced LX-2 cells model were used, and then they were treated with TJ0113. Changes in hepatocyte damage were observed using electron microscopy. Expression of key molecules related to mitophagy, mitochondrial damage and inflammation in liver was detected by immunofluorescence staining (IF), immunohistochemistry (IHC) and western blotting (WB). RESULT TJ0113 induces mitophagy through parkin/PINK1 and ATG5 signaling pathways and reduces lipid accumulation, inflammation and fibrosis in the liver of MASH mice. TJ0113 attenuated hepatic injury and lowered serum ALT, AST, TC and TG levels. TJ0113 reduced pro-inflammatory factors (IL-1β, IL-6, TNF-α), TGF-β1/Smad pathway activation and typical fibrosis-related molecules (α-SMA, Collagen-1) expression. In addition, NF-κB/NLRP3 signaling pathway activation after MASH was significantly attenuated by enhanced Mitophagy. We found that TJ0113 was able to effectively and safely induce mitophagy in vitro and reduce TGF-β1/Smad signaling and downstream pro-fibrotic responses in TGF-β1-treated LX-2 cells. CONCLUSION TJ0113 enhances mitophagy to inhibit lipid accumulation, inflammation and fibrosis formation in MASH, and is a candidate for MASH treatment.
Collapse
Affiliation(s)
- Chun-Lian Huang
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China
| | - Shen Qi-En
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Xu-Feng Cen
- Research Center of Clinical Pharmacy of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Ting Ye
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China
| | - Hang-Shuai Qu
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China.
| | - Si-Jia Chen
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China
| | - Dong Liu
- Hangzhou PhecdaMed Co., Ltd., Third Floor, Building 2, No. 2626. Yuhangtang Road, Yuhang District, Hangzhou 310003, China.
| | - Hong-Guang Xia
- Research Center of Clinical Pharmacy of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Cheng-Fu Xu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Jian-Sheng Zhu
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China.
| |
Collapse
|
3
|
Saadh MJ, Muhammad FA, Albadr RJ, Sanghvi G, Ballal S, Pathak PK, Bareja L, Aminov Z, Taher WM, Alwan M, Jawad MJ, Al-Nuaimi AMA. Exosomal non-coding RNAs: key regulators of inflammation-related cardiovascular disorders. Eur J Med Res 2025; 30:395. [PMID: 40390035 PMCID: PMC12087048 DOI: 10.1186/s40001-025-02649-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 04/30/2025] [Indexed: 05/21/2025] Open
Abstract
Inflammation is a complex, tightly regulated process involving biochemical and cellular reactions to harmful stimuli. Often termed "the internal fire", it is crucial for protecting the body and facilitating tissue healing. While inflammation is essential for survival, chronic inflammation can be detrimental, leading to tissue damage and reduced survival. The innate immune system triggers inflammation, closely linked to the development of heart diseases, with significant consequences for individuals. Inflammation in arterial walls or the body substantially contributes to atherosclerotic disease progression, affecting the cardiovascular system. Altered lipoproteins increase the risk of excessive blood clotting, a hallmark of atherosclerotic cardiovascular disease and its complications. Integrating inflammatory biomarkers with established risk assessment techniques can enhance our ability to identify at-risk individuals, assess their risk severity, and recommend appropriate CVD prevention strategies. Exosomes, a type of extracellular vesicle, are released by various cells and mediate cell communication locally and systemically. In the past decade, exosomes have been increasingly studied for their vital roles in health maintenance and disease processes. They can transport substances like non-coding RNAs, lipids, and proteins between cells, influencing immune responses and inflammation to elicit harmful or healing effects. This study focuses on the critical role of inflammation in heart disease progression and how non-coding RNAs in exosomes modulate the inflammatory process, either exacerbating or alleviating inflammation-related damage in the cardiovascular system.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | | | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat, 360003, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Piyus Kumar Pathak
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Lakshay Bareja
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | |
Collapse
|
4
|
Tang Y, Tong W, Peng Y, Sun S. Targeting cholesterol-driven pyroptosis: a promising strategy for the prevention and treatment of atherosclerosis. Mol Biol Rep 2025; 52:459. [PMID: 40372511 DOI: 10.1007/s11033-025-10554-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Accepted: 04/28/2025] [Indexed: 05/16/2025]
Abstract
Funding Pyroptosis is a type of programmed cell death (PCD) pathway distinguished by inflammation. It is activated by specific inflammasomes. Once activated, it causes the physical breakdown of the cell, along with the discharge of pro-inflammatory cytokines, such as interleukin-1β (IL-1β) and interleukin-18 (IL-18). Abundant evidence has demonstrated the existence of pyroptotic cell death within atherosclerotic plaques, which has significance for the development of atherosclerosis (AS). As a result, pyroptosis has become a new and important topic in cardiovascular disease (CVD) research. Cholesterol, it is recognized to have a connection with inflammation, exerts a crucial function in the development process of AS, and has been linked to the initiation of pyroptosis. This review aims to briefly summarize the fundamental aspects of pyroptosis and the influence of cholesterol-related inflammation in AS. Additionally, this review will explore potential therapeutic approaches based on pyroptosis that could be utilized for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Yuehong Tang
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wenjuan Tong
- Department of Gynecology and Obstetrics, First Affiliated Hospital, University of South China, Hengyang, Hunan, 421001, China
| | - Yujiao Peng
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shaowei Sun
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
5
|
Jiang Y, Chen J, Du Y, Fan M, Shen L. Immune modulation for the patterns of epithelial cell death in inflammatory bowel disease. Int Immunopharmacol 2025; 154:114462. [PMID: 40186907 DOI: 10.1016/j.intimp.2025.114462] [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: 01/17/2025] [Revised: 02/23/2025] [Accepted: 03/08/2025] [Indexed: 04/07/2025]
Abstract
Inflammatory bowel disease (IBD) is an inflammatory disease of the intestine whose primary pathological presentation is the destruction of the intestinal epithelium. The intestinal epithelium, located between the lumen and lamina propria, transmits luminal microbial signals to the immune cells in the lamina propria, which also modulate the intestinal epithelium. In IBD patients, intestinal epithelial cells (IECs) die dysfunction and the mucosal barrier is disrupted, leading to the recruitment of immune cells and the release of cytokines. In this review, we describe the structure and functions of the intestinal epithelium and mucosal barrier in the physiological state and under IBD conditions, as well as the patterns of epithelial cell death and how immune cells modulate the intestinal epithelium providing a reference for clinical research and drug development of IBD. In addition, according to the targeting of epithelial apoptosis and necroptotic pathways and the regulation of immune cells, we summarized some new methods for the treatment of IBD, such as necroptosis inhibitors, microbiome regulation, which provide potential ideas for the treatment of IBD. This review also describes the potential for integrating AI-driven approaches into innovation in IBD treatments.
Collapse
Affiliation(s)
- Yuting Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Yaoyao Du
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Minwei Fan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Lan Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
6
|
Zheng X, Jiang GJ, Fan TJ. Blue Light Irradiation Elicits Senescence of Corneal Endothelial Cells In Vitro by Provoking Energy Crisis, Inflammasome Assembly and DNA Damage. Curr Eye Res 2025:1-12. [PMID: 40336349 DOI: 10.1080/02713683.2025.2497330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 05/09/2025]
Abstract
PURPOSE The blue light from the digital screens endangers the visual system among which the corneas at the outmost of eyes are vulnerable to the irradiation. Therein, the human corneal endothelial (HCE) cells are crucial to maintain corneal transparency and their damage leads to HCE decompensation resulting in blindness ultimately. Thus, understanding the phototoxic effects of the blue light on the HCE cells and the underlying mechanisms is important for taking measures to protect the vision clarity from the blue-light hazard. METHODS We pulse-irradiated the HCE cell line cells at logarithmic phase for 3 passages using 440 nm blue light and examined the levels of reactive oxygen species (ROS), ATP, nicotinamide adenine dinucleotide (NAD+) and autophagy using cytochemistry assay to investigate the alterations of energy metabolism. Moreover, we examined the γH2AX+ cells using immunofluorescence and expression of poly(ADP-Ribose)polymerase1 (PARP1) using western blotting to investigate the degrees of DNA damage and repair. We also monitored the levels of inflammasome using western blotting and senescence associated secretory phenotypes (SASPs) of interleukin (IL)-8, IL-1β and IL-6 using qPCR and ELISA to investigate the inflammasome assembly and secretion of SASPs. We detected the senescent features with senescence-associated-β-galactosidase assay, p16 levels by western blotting, Lamin B1 localization by immunofluorescence observation, cell growth by EdU incorporation assay and confluence forming time and alterations of the cell morphology and relative areas by microscopy observation. RESULTS The HCE cells exhibited senescent features after blue-light-pulse-irradiation. The blue light provokes overproduction of ROS to decrease the levels of ATP, NAD+ and autophagy leading to energy crisis. Moreover, the excess ROS injure DNA and downregulate PARP1 resulting in stable cell-cycle arrest. The excess ROS also facilitate inflammasome assembly leading to hypersecretion of SASPs. CONCLUSION The blue light elicits HCE cell senescence via inducing energy crisis, stable cell-cycle arrest and SASP hypersecretion.
Collapse
Affiliation(s)
- Xin Zheng
- College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong Province, China
| | - Guo-Jian Jiang
- College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong Province, China
| | - Ting-Jun Fan
- College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong Province, China
| |
Collapse
|
7
|
Zhang K, Du Y, Yang S, Sun G. Irisin suppressed the progression of TBI via modulating AMPK/MerTK/autophagy and SYK/ROS/inflammatory signaling. Sci Rep 2025; 15:15583. [PMID: 40320408 PMCID: PMC12050266 DOI: 10.1038/s41598-025-00066-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
Irisin is a hormone-like peptide secreted by muscle tissues and generated by hydrolysis of type III fibronectin domain-containing protein 5 by proteolytic hydrolases. Whether Irisin has a potential protective role in traumatic brain injury (TBI). In this study, we will investigate the relevant research progress of Irisin's protective role in traumatic brain injury (TBI) in recent years in terms of attenuating oxidative stress, inhibiting pyroptosis, suppressing inflammatory response, and improving autophagy, with the aim of providing valuable references for the diagnosis and treatment of traumatic brain injury (TBI). Utilize bioinformatics analysis to study the interactions between genes in TBI (Traumatic Brain Injury). Construct a TBI mouse model to observe the effects of Irisin on TBI. The Morris water maze test is used to assess the learning and spatial memory abilities of mice, TUNEL fluorescence is used to detect cell apoptosis, Nissl staining is employed to observe the survival of hippocampal neurons in mice, and HE staining is used to observe the extent of brain injury in mice. Western blot is used to detect protein expression in both in vivo and in vitro experiments. Q-PCR is employed to detect the levels of proteins related to autophagy/pyroptosis/inflammation. Irisin promotes MerTK overexpression by enhancing AMPK activation. Irisin can increase the expression of LC3I and Beclin-1 proteins, indicating the promotion of autophagic response. Additionally, Irisin reduces ROS levels and decreases SYK expression, thereby inhibiting the inflammatory response. Irisin improves the learning and spatial memory abilities of TBI mice and reduces cell apoptosis, as well as decreases hippocampal neuron death. HE staining shows that the brain injury in mice treated with Irisin is significantly alleviated. Irisin can enhance the expression of phosphorylated AMPK and phosphorylated MerTK proteins, promote autophagic response, and inhibit pyroptosis/inflammatory response. Correction experiments confirmed that after stimulation with an AMPK agonist, the expression of phosphorylated MerTK protein is significantly increased, autophagic response is enhanced, and pyroptosis/inflammatory response is weakened. When treated with a MerTK inhibitor during AMPK agonist stimulation, the autophagic response is weakened while pyroptosis/inflammatory response is enhanced. Irisin can inhibit the progression of traumatic brain injury by regulating AMPK/MerTK/autophagy and SYK/ROS/inflammatory signaling.
Collapse
Affiliation(s)
- Kuo Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang City, 050000, Hebei Province, China
- Department of Neurosurgery, Hebei General Hospital, Shijiazhuang City, 050000, Hebei Province, China
| | - Yihui Du
- Department of Neurosurgery, Hebei General Hospital, Shijiazhuang City, 050000, Hebei Province, China
| | - Sihui Yang
- Department of Neurosurgery, Hebei General Hospital, Shijiazhuang City, 050000, Hebei Province, China
| | - Guozhu Sun
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang City, 050000, Hebei Province, China.
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping West Road, Xinhua District, Shijiazhuang City, 050000, Hebei Province, China.
| |
Collapse
|
8
|
Bai X, Guo YR, Zhao ZM, Li XY, Dai DQ, Zhang JK, Li YS, Zhang CD. Macrophage polarization in cancer and beyond: from inflammatory signaling pathways to potential therapeutic strategies. Cancer Lett 2025; 625:217772. [PMID: 40324582 DOI: 10.1016/j.canlet.2025.217772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
Macrophages are innate immune cells distributed throughout the body that play vital roles in organ development, tissue homeostasis, and immune surveillance. Macrophages acquire a binary M1/M2 polarized phenotype through signaling cascades upon sensing different signaling molecules in the environment, thereby playing a core role in a series of immune tasks, rendering precise regulation essential. M1/M2 macrophage phenotypes regulate inflammatory responses, while controlled activation of inflammatory signaling pathways is involved in regulating macrophage polarization. Among the relevant signaling pathways, we focus on the six well-characterized NF-κB, MAPK, JAK-STAT, PI3K/AKT, inflammasome, and cGAS-STING inflammatory pathways, and elucidate their roles and crosstalk in macrophage polarization. Furthermore, the effects of many environmental signals that influence macrophage polarization are investigated by modulating these pathways in vivo and in vitro. We thus detail the physiological and pathophysiological status of these six inflammatory signaling pathways and involvement in regulating macrophage polarization in cancer and beyond, as well as describe potential therapeutic approaches targeting these signaling pathways. In this review, the latest research advances in inflammatory signaling pathways regulating macrophage polarization are reviewed, as targeting these inflammatory signaling pathways provides suitable strategies to intervene in macrophage polarization and various tumor and non-tumor diseases.
Collapse
Affiliation(s)
- Xiao Bai
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Yun-Ran Guo
- Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Zhe-Ming Zhao
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Xin-Yun Li
- Clinical Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Dong-Qiu Dai
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Cancer Center, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Jia-Kui Zhang
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Yong-Shuang Li
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Chun-Dong Zhang
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
9
|
Qiu D, Zhao N, Chen Q, Wang M. FOXC1 Aggravates the Ischemia-Reperfusion Induced Injury in Renal Tubular Epithelial Cells by Activating NF-κB/NLRP3 Signaling. J Biochem Mol Toxicol 2025; 39:e70301. [PMID: 40371539 DOI: 10.1002/jbt.70301] [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: 03/18/2025] [Revised: 03/19/2025] [Accepted: 04/25/2025] [Indexed: 05/16/2025]
Abstract
Renal ischemia-reperfusion injury (RIRI) is a condition characterized by inflammation and cell damage in the kidneys following a period of ischemia and subsequent reperfusion, which lacks effective treating method in the clinic. Exploring molecular mechanisms holds profound significance in guiding the clinical prevention and treatment of RIRI. Herein, the potential function of Forkhead box C1 (FOXC1), a protein belongs to FOX family, in I/R-induced injury in renal tubular epithelial cells (RTECs) was studied to explore potential targets for RIRI. FOXC1 was upregulated in RIRI rats, expressions of which were elevated as time prolonged. FOXC1-overexpressed or knockdown HK-2 cells were constructed, followed by I/R stimulation. FOXC1 was found markedly upregulated in I/R-stimulated HK-2 cells. Notably repressed cell viability, enhanced apoptosis, increased release of inflammatory cytokines, boosted reactive oxygen species (ROS) and malondialdehyde (MDA) levels, and inactivated superoxide dismutase (SOD) enzyme were observed in I/R-stimulated HK-2 cells, which were sharply reversed by silencing FOXC1 and aggravated by overexpression FOXC1. Furthermore, largely increased levels of NLRP3, caspase-1, GSDMD-N, IL-18, IL-1β, and p-p65/p65 were observed in I/R-stimulated HK-2 cells, which were notably suppressed by silencing FOXC1 and further elevated by overexpression FOXC1. Additionally, FOXC1-overexpressed HK-2 cells were stimulated by I/R with or without 10 μM MCC950, an inhibitor of NLRP3. The enhanced apoptosis, triggered inflammation, and facilitated ROS by FOXC1 overexpression in I/R-stimulated HK-2 cells were remarkably abolished by the coculture of MCC950, accompanied by an inhibition on the NF-κB/NLRP3 signaling. Collectively, FOXC1 aggravated the I/R induced injury in RTECs by activating NF-κB/NLRP3 signaling.
Collapse
Affiliation(s)
- Donghao Qiu
- Department of Nehprology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Ning Zhao
- Department of Nehprology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Qi Chen
- Department of Nehprology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Ming Wang
- Department of Nehprology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| |
Collapse
|
10
|
Yu Y, Jiang X, Li R, Xiang G, Zhang Y. Study on the Anti-Inflammatory Effect of Gentiana scabra Bunge Extract and Its Mechanism Using Zebrafish and RAW264.7 Cell Models. Biomed Chromatogr 2025; 39:e70050. [PMID: 40091705 DOI: 10.1002/bmc.70050] [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: 12/12/2024] [Revised: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 03/19/2025]
Abstract
Gentiana scabra Bunge (Gentian) is a traditional medicinal plant valued for its anti-inflammatory and analgesic effects, with historical use in treating atopic dermatitis. Despite its therapeutic reputation, a comprehensive scientific analysis of its constituents is lacking. This study systematically evaluates the anti-inflammatory effects of Gentian extract and explores its molecular mechanisms. We characterized the chemical profile of Gentian extracts using HPLC and assessed their anti-inflammatory activity in zebrafish and cellular models. Gentian extract significantly reduced inflammation, as shown by decreased neutrophil migration in response to sodium lauryl sulfate (SLS), reduced tail wagging in zebrafish embryos, and alleviated lipopolysaccharide (LPS)-induced edema. It also lowered reactive oxygen species (ROS) and malondialdehyde (MDA) levels, indicating antioxidant properties, and downregulated pro-inflammatory cytokines and genes. In LPS-stimulated RAW264.7 cells, the extract upregulated IκBα and reduced p65 and STAT3 phosphorylation, inhibiting NF-κB and JAK-STAT pathways. This study is the first to systematically evaluate the anti-inflammatory mechanisms of Gentian extract in zebrafish and RAW264.7 cell models, enhancing its understanding and providing a scientific basis for its application in anti-inflammatory products.
Collapse
Affiliation(s)
- Yang Yu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xufeng Jiang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, China
- Ugel Cosmetics Pte. Ltd., Singapore
| | - Ruirui Li
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | | | - Yang Zhang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
11
|
Xu Q, Zhu Q, Ling G, Huang T, Su T, Chen Y, Xie Y, Zhong Y. Activation of spinal PGC-1α regulates microglial polarization through a feedback loop between ROS-mediated mitochondrial dysfunction and the NLRP3 inflammasome in neuropathic pain. Brain Res Bull 2025:111365. [PMID: 40316183 DOI: 10.1016/j.brainresbull.2025.111365] [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: 12/27/2024] [Revised: 03/19/2025] [Accepted: 04/26/2025] [Indexed: 05/04/2025]
Abstract
BACKGROUND An imbalance in microglial polarization plays an important role in the pathogenesis of neuropathic pain. PPARγ coactivator-1α (PGC-1α), a master coregulator of gene expression in mitochondrial biogenesis, is related to microglial polarization. However, the underlying mechanism involved is poorly understood.The aim of the present study was to explore the role of PGC-1α in regulating microglial polarization through a feedback loop between reactive oxygen species (ROS)-mediated mitochondrial dysfunction and the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome in a rat model of chronic constriction injury (CCI). METHODS we quantified pain behaviour after CCI; analysed the localization of PGC-1α and the changes in the expression of CD68 (an M1 microglial marker)/IBA1 and ARG1 (an M2 microglial marker)/IBA1 in the dorsal horn (DH) via immunofluorescence. Western blotting and immunofluorescence were used to examine the expression of target proteins. Quantitative real-time PCR (qPCR) was used to investigate the mitochondrial DNA copy number (mtDNA). ROS production was measured via dihydroethidium (DHE). SOD activity and the MDA content were measured via SOD and MDA assay kits, respectively. In addition, tumour necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6 and IL-10 levels were measured via enzyme-linked immunosorbent assay (ELISA). RESULTS The results revealed ROS-mediated mitochondrial dysfunction and NLRP3 inflammasome activation, microglia phenotype from the M2 to the M1 phenotype in the CCI rats.Interesting, ROS-mediated mitochondrial dysfunction is one of the critical mediators of NLRP3 inflammasome activation.NLRP3 inflammasome in turn cause ROS production and mitochondrial dysfunction, suggesting for the first time a feedback loop between ROS-mediated mitochondrial dysfunction and NLRP3 inflammasome in the neuropathic pain.The activation of PGC-1α shifts the microglial phenotype via the modulation of a feedback loop between ROS-mediated mitochondrial dysfunction and the NLRP3 inflammasome. CONCLUSIONS These findings indicate that activation of PGC-1α could be a potential therapeutic approach to ameliorate neuropathic pain.
Collapse
Affiliation(s)
- Qingling Xu
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, PR China
| | - Qiulin Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, PR China
| | - Guoxu Ling
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, PR China
| | - Tonghong Huang
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, PR China
| | - Tingting Su
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, PR China
| | - Yanhua Chen
- Department of Anesthesiology, Cardiovascular Institute,Nanning, Guangxi 530021, PR China
| | - Yubo Xie
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, PR China.
| | - Yu Zhong
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, PR China.
| |
Collapse
|
12
|
Li H, Wu Y, Xiang L, Zhao Q, Liu L, Zhu Z, Lin W, Li Z, Yang Y, Ze Y, Zhang L, Fu P, Guo Y, Zhang P, Shao B. A20 attenuates oxidized self-DNA-mediated inflammation in acute kidney injury. Signal Transduct Target Ther 2025; 10:154. [PMID: 40280946 PMCID: PMC12032302 DOI: 10.1038/s41392-025-02194-y] [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: 05/23/2024] [Revised: 02/03/2025] [Accepted: 02/20/2025] [Indexed: 04/29/2025] Open
Abstract
The ubiquitin-editing enzyme A20 is known to regulate inflammation and maintain homeostasis, but its role in self-DNA-mediated inflammation in acute kidney injury (AKI) is not well understood. Here, our study demonstrated that oxidized self-DNA accumulates in the serum of AKI mice and patients. This oxidized self-DNA exacerbates the progression of AKI by activating the cGAS-STING pathway and NLRP3 inflammasome. While inhibition of the STING pathway only slightly attenuates AKI progression, suppression of NLRP3 inflammasome-mediated pyroptosis significantly alleviates AKI progression and improves the survival of AKI mice. Subsequently, we found that Tnfaip3 (encoding A20) is significantly upregulated following oxidized self-DNA treatment. A20 significantly alleviates AKI development by dampening STING signaling pathway and NLRP3-mediated pyroptosis. Moreover, A20-derived peptide (P-II) also significantly alleviates ox-dsDNA-induced pyroptosis and improves the survival and renal injury of AKI mice. Mechanistically, A20 competitively binds with NEK7 and thus inhibiting NLRP3 inflammasome. A20 and P-II interfere with the interaction between NEK7 and NLRP3 through Lys140 of NEK7. Mutation of Lys140 effects on the interaction of NEK7 with A20 and/or NLRP3 complex. Conditional knockout of NEK7 in macrophages or pharmacological inhibition of NEK7 both significantly rescue AKI mouse models. This study reveals a new mechanism by which A20 attenuates oxidized self-DNA-mediated inflammation and provides a new therapeutic strategy for AKI.
Collapse
Affiliation(s)
- Hanwen Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, PR China
| | - Yongyao Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, PR China
| | - Lisha Xiang
- Division of Thoracic Tumor Multimodality Treatment and Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Qing Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, PR China
| | - Lu Liu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Zhixiong Zhu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Weimin Lin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, PR China
| | - Zhan Li
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, PR China
| | - Yang Yang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yiting Ze
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, PR China
| | - Lulu Zhang
- College of Foreign Languages and Cultures, Sichuan University. Sichuan University, Chengdu, Sichuan, PR China
| | - Ping Fu
- Kidney Research Institute, National Clinical Research Center for Geriatrics and Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, PR China
| | - Yingqiang Guo
- Department of Cardiovascular Surgery and Cardiovascular Surgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China.
| | - Ping Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, PR China.
| | - Bin Shao
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China.
- Department of Cardiovascular Surgery and Cardiovascular Surgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China.
| |
Collapse
|
13
|
Li M, Sun X, Zeng L, Sun A, Ge J. Metabolic Homeostasis of Immune Cells Modulates Cardiovascular Diseases. RESEARCH (WASHINGTON, D.C.) 2025; 8:0679. [PMID: 40270694 PMCID: PMC12015101 DOI: 10.34133/research.0679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/20/2025] [Accepted: 03/28/2025] [Indexed: 04/25/2025]
Abstract
Recent investigations into the mechanisms underlying inflammation have highlighted the pivotal role of immune cells in regulating cardiac pathophysiology. Notably, these immune cells modulate cardiac processes through alternations in intracellular metabolism, including glycolysis and oxidative phosphorylation, whereas the extracellular metabolic environment is changed during cardiovascular disease, influencing function of immune cells. This dynamic interaction between immune cells and their metabolic environment has given rise to the novel concept of "immune metabolism". Consequently, both the extracellular and intracellular metabolic environment modulate the equilibrium between anti- and pro-inflammatory responses. This regulatory mechanism subsequently influences the processes of myocardial ischemia, cardiac fibrosis, and cardiac remodeling, ultimately leading to a series of cardiovascular events. This review examines how local microenvironmental and systemic environmental changes induce metabolic reprogramming in immune cells and explores the subsequent effects of aberrant activation or polarization of immune cells in the progression of cardiovascular disease. Finally, we discuss potential therapeutic strategies targeting metabolism to counteract abnormal immune activation.
Collapse
Affiliation(s)
- Mohan Li
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Xiaolei Sun
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Linqi Zeng
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
- Institutes of Biomedical Sciences,
Fudan University, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
- Institutes of Biomedical Sciences,
Fudan University, Shanghai 200032, China
| |
Collapse
|
14
|
Gong M, Luo J, Liang Q, Liu Y, Zheng Y, Yang XD. Chromatin-associated cullin-RING E3 ubiquitin ligases: keeping transcriptionally active NF-κB in check. Front Immunol 2025; 16:1584999. [PMID: 40308609 PMCID: PMC12040619 DOI: 10.3389/fimmu.2025.1584999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Nuclear factor-κB (NF-κB) constitutes a family of transcription factors that serve as a critical regulatory hub, dynamically orchestrating inflammatory and immune responses to maintain homeostasis and protect against pathogenic threats. Persistent activation of NF-κB has been implicated in the pathogenesis of various inflammatory diseases and cancer. A critical mechanism to prevent excessive inflammation and its harmful effects is the timely termination of NF-κB's transcriptional activity on target genes. This termination can be facilitated through the ubiquitination and subsequent proteasomal degradation of chromatin-bound RelA, the most active subunit of NF-κB. Several multi-subunit cullin-RING E3 ubiquitin ligases, composed of elongin B/C, cullin2/5, and SOCS-box proteins, have been identified to target RelA for degradation. These E3s, known as ECS complexes, use SOCS-box proteins as substrate-recognizing subunits to engage RelA. SOCS1 is the first identified SOCS-box member that functions in ECSSOCS1 to target chromatin-bound RelA for ubiquitination. Specifically, SOCS1 collaborates with accessory proteins COMMD1 and GCN5 to preferentially recognize Ser468-phosphorylated RelA. Our recent work demonstrates that WSB1 and WSB2 (WSB1/2), two additional SOCS-box proteins with structurally similar WD40 repeat domains, function as substrate-recognizing subunits of ECSWSB1/2 to specifically mediate the ubiquitination and degradation of chromatin-associated RelA methylated at Lys314/315. In this review, we summarize the discovery and functional importance of ECSSOCS1 and ECSWSB1/2 in terminating NF-κB activity, highlight the distinct molecular mechanisms by which they ubiquitinate chromatin-associated RelA in a modification- and gene-specific manner, and discuss their potential as therapeutic targets for inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Mengyao Gong
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Junqi Luo
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiankun Liang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi Liu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuejuan Zheng
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Dong Yang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
15
|
Ma X, Li B, Liu Y, Guo X. An inverted U-shaped association between high-sensitivity C-reactive protein and the albumin ratio and hepatic steatosis and liver fibrosis: a population-based study. Front Nutr 2025; 12:1534200. [PMID: 40303878 PMCID: PMC12037389 DOI: 10.3389/fnut.2025.1534200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/24/2025] [Indexed: 05/02/2025] Open
Abstract
Background The high-sensitivity C-reactive protein to albumin (CAR) ratio is a comprehensive measure of inflammation in vivo. Hepatic steatosis and fibrosis are significantly correlated with inflammation. The present study aimed to explore the possible associations between CAR and hepatic steatosis and fibrosis in the American population. Methods The study population involved the National Health and Nutrition Examination Survey (NHANES) participants from 2017 to 2020. The natural logarithm of CAR, calculated as Ln(CAR) with base "e," was used for further analyses. The relationships between Ln(CAR) and the controlled attenuation parameter (CAP) and between Ln(CAR) and liver stiffness measurement (LSM) were investigated through multivariate linear regression analysis. Interaction and subgroup analysis identified factors affecting these variables. Nonlinear relationships were elucidated by smoothing curves and threshold effect analysis. Receiver operating characteristic (ROC) curve analysis was performed to evaluate the predictive performance of the CAR for non-alcoholic fatty liver disease (NAFLD). The results were adjusted for U.S. population estimates. Results The study included a total of 7,404 individuals. Ln(CAR) was positively correlated with CAP in the fully adjusted model, with an effect value of β = 1.827 (95% CI, 0.611, 3.042). A more pronounced positive association was observed among participants with a BMI ≥ 25 kg/m2 in the subgroup analysis. An inverted U-shaped association was shown between Ln(CAR) and CAP through smooth curve fitting and a two-segment linear regression model, with an inflection point of (-9.594). ROC curve analysis showed that CAR had a moderate predictive value for NAFLD (AUC = 0.6895), with a sensitivity of 0.7276 and a specificity of 0.6092. No significant association was detected between Ln(CAR) and the LSM. Conclusion We demonstrate an inverted U-shaped relationship between Ln(CAR) and CAP risk within the U.S. demographic. Our results suggest that CAR may serve as a valuable diagnostic tool for NAFLD. Further prospective research is necessary to validate this conclusion.
Collapse
Affiliation(s)
| | | | | | - Xiaoyan Guo
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
16
|
Tai GJ, Ma YJ, Feng JL, Li JP, Qiu S, Yu QQ, Liu RH, Wankumbu SC, Wang X, Li XX, Xu M. NLRP3 inflammasome-mediated premature immunosenescence drives diabetic vascular aging dependent on the induction of perivascular adipose tissue dysfunction. Cardiovasc Res 2025; 121:77-96. [PMID: 38643484 DOI: 10.1093/cvr/cvae079] [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: 05/16/2023] [Revised: 11/29/2023] [Accepted: 02/06/2024] [Indexed: 04/23/2024] Open
Abstract
AIMS The vascular aging process accelerated by type 2 diabetes mellitus (T2DM) is responsible for the elevated risk of associated cardiovascular diseases. Metabolic disorder-induced immune senescence has been implicated in multi-organ/tissue damage. Herein, we sought to determine the role of immunosenescence in diabetic vascular aging and to investigate the underlying mechanisms. METHODS AND RESULTS Aging hallmarks of the immune system appear prior to the vasculature in streptozotocin (STZ)/high-fat diet (HFD)-induced T2DM mice or db/db mice. Transplantation of aged splenocytes or diabetic splenocytes into young mice triggered vascular senescence and injury compared with normal control splenocyte transfer. RNA sequencing profile and validation in immune tissues revealed that the toll-like receptor 4-nuclear factor-kappa B-NLRP3 axis might be the mediator of diabetic premature immunosenescence. The absence of Nlrp3 attenuated immune senescence and vascular aging during T2DM. Importantly, senescent immune cells, particularly T cells, provoked perivascular adipose tissue (PVAT) dysfunction and alternations in its secretome, which in turn impair vascular biology. In addition, senescent immune cells may uniquely affect vasoconstriction via influencing PVAT. Lastly, rapamycin alleviated diabetic immune senescence and vascular aging, which may be partly due to NLRP3 signalling inhibition. CONCLUSION These results indicated that NLRP3 inflammasome-mediated immunosenescence precedes and drives diabetic vascular aging. The contribution of senescent immune cells to vascular aging is a combined effect of their direct effects and induction of PVAT dysfunction, the latter of which can uniquely affect vasoconstriction. We further demonstrated that infiltration of senescent T cells in PVAT was increased and associated with PVAT secretome alterations. Our findings suggest that blocking the NLRP3 pathway may prevent early immunosenescence and thus mitigate diabetic vascular aging and damage, and targeting senescent T cells or PVAT might also be the potential therapeutic approach.
Collapse
MESH Headings
- Animals
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/deficiency
- Inflammasomes/metabolism
- Inflammasomes/genetics
- Inflammasomes/immunology
- Signal Transduction
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/immunology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Experimental/pathology
- Male
- Adipose Tissue/metabolism
- Adipose Tissue/immunology
- Adipose Tissue/physiopathology
- Adipose Tissue/pathology
- Mice, Inbred C57BL
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/physiopathology
- Diabetes Mellitus, Type 2/pathology
- Immunosenescence
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/immunology
- Diabetic Angiopathies/physiopathology
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/pathology
- Diabetic Angiopathies/prevention & control
- Cellular Senescence
- Mice, Knockout
- Vasoconstriction
- T-Lymphocytes/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- T-Lymphocytes/pathology
- NF-kappa B/metabolism
- Mice
- Spleen/metabolism
- Spleen/transplantation
- Toll-Like Receptor 4
Collapse
Affiliation(s)
- Guang-Jie Tai
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Yan-Jie Ma
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Jun-Lin Feng
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Jia-Peng Li
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Shu Qiu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Qing-Qing Yu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Ren-Hua Liu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Silumbwe Ceaser Wankumbu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Xin Wang
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Xiao-Xue Li
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Dingjiaqiao, Nanjing 210009, China
| | - Ming Xu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| |
Collapse
|
17
|
Bhat KMR, Upadhya R, Adiga S, Kumar SEP, Manjula SD, Acharya N, Subramanian HH, Upadhya D. Regulation of chronic neuroinflammation through dietary herbal products. Front Nutr 2025; 12:1487786. [PMID: 40297338 PMCID: PMC12036043 DOI: 10.3389/fnut.2025.1487786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Chronic neuroinflammation is a consequence of disease pathogenesis underlying neurological disorders at large. While the immune response that triggers inflammatory signaling cascades is unresolved, its progression could cause functional damage to neurons and glial cells, including astrocytes, microglia, and oligodendrocytes. Controlling neuroinflammatory signaling at the early stage of disease pathogenesis is critical to prevent irreversible tissue necrosis. While the application of anti-inflammatory drugs is standard practice, their protracted use is known to cause gastrointestinal injuries, further enhancing the risk of cardiovascular, renal, liver, and lung diseases. Several medicinal herbs and herbal products with anti-inflammatory potential could be effective substitutes. This review aims to identify the preclinical data from important dietary herbal products that have demonstrated anti-neuroinflammatory efficacy in several animal models. The reviewed dietary herbal products are sourced from Bacopa monnieri, Centella asiatica, Emblica officinalis, Piper nigrum, Zingiber officinale, Punica granatum, Mucuna pruriens, Clitoria ternatea, Moringa oleifera, Phoenix dactylifera and Curcuma longa. This review is based on emphatic data from these products demonstrating the significant anti-neuro-inflammatory potential that could probably reduce neuroinflammatory signaling in a neurological disorder and promote brain health and well-being. Abundant scientific evidence shows that critical proinflammatory cytokines in the brain, such as tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), interleukin-six (IL-6), could be controlled through regular consumption of such dietary herbal products without debilitating side effects for their disease-modifying impacts.
Collapse
Affiliation(s)
- Kumar M. R. Bhat
- Department of Anatomy, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Raghavendra Upadhya
- Manipal Centre for Biotherapeutics Research, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Shripathi Adiga
- Department of Ayurveda, Centre for Integrative Medicine and Research, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - S. E. Praveen Kumar
- Department of Pharmacology, Manipal Tata Medical College, Jamshedpur, Manipal Academy of Higher Education, Manipal, India
| | - S. D. Manjula
- Department of Physiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Nanda Acharya
- Department of Physiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Hari H. Subramanian
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, United States
- Neuronano AB, Valencia, CA, United States
| | - Dinesh Upadhya
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
18
|
Li Y, Yang JY, Lin ML, Liu TZ, Lu WN, Yang Y, Liu ZC, Li JH, Zhang GQ, Guo JS. ACT001 improves OVX-induced osteoporosis by suppressing the NF-κB/NLRP3 signaling pathway. Mol Med 2025; 31:131. [PMID: 40197211 PMCID: PMC11977873 DOI: 10.1186/s10020-025-01189-3] [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: 09/18/2024] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
Osteoporosis (OP) is a common systemic metabolic bone disease characterized by the decrease in bone mass and hyperactivity of osteoclasts. ACT001 is approved as an orphan drug by FDA and has shown multiple protective effects against tissue injury. However, its role in prevention of osteoclast differentiation and the underlying mechanisms have not been elucidated. Herein, we show that ACT001 inhibited RANKL-induced osteoclast differentiation and F-actin ring formation through suppressing the expression of Nfatc1, TRAP, Ctsk, Dc-stamp without obvious cytotoxicity in vitro. ACT001 restrained the phosphorylation of NF-κB and the activation of NLRP3 inflammasome, thereby decreased the expression of pyroptosis-related protein. (GSDMD, caspase-1, IL-1β, IL-18). Consistent with ACT001, the NLRP3 inflammasome inhibitor MCC950 treatment also suppressed the osteoclastogenesis through inhibiting the transcriptional activation of Nfatc1. Furthermore, ACT001 protected ovariectomy-induced bone loss in mice, reduced the number of osteoclasts, downregulated the expression of NLRP3 and IL-1β. These data indicate that ACT001 can reduce RANKL-induced osteoclast differentiation through suppressing the NF-κB/NLRP3 pathway, and attenuate the bone loss induced by estrogen-deficiency, suggesting its therapeutic potential for bone homeostasis maintenance and osteoporosis treatment.
Collapse
Affiliation(s)
- Yuan Li
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China
| | - Jin-Yu Yang
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China
| | - Ma-Li Lin
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China
| | - Tian-Zhu Liu
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China
| | - Wen-Na Lu
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China
| | - Ying Yang
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China
| | - Zhong-Cheng Liu
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China
| | - Jian-Heng Li
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China.
| | - Guo-Qiang Zhang
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China.
| | - Jian-Shuang Guo
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China.
| |
Collapse
|
19
|
Wang J, Zhang C, Qin J, An N, Bai M, Du RH, Shen Y, Wu XD, Cheng JC, Wu XF, Xu Q. Direct inhibition of the TXNIP-NLRP3-GSDMD pathway reduces pyroptosis in colonocytes and alleviates ulcerative colitis in mice by the small compound PEITC. Acta Pharmacol Sin 2025:10.1038/s41401-025-01549-z. [PMID: 40195510 DOI: 10.1038/s41401-025-01549-z] [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: 10/25/2024] [Accepted: 03/18/2025] [Indexed: 04/09/2025]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease. The etiology of UC is multifaceted, and the underlying pathogenesis remains incompletely understood. Pyroptosis, programmed cell death mediated by the gasdermins, is a pivotal driver of UC pathology due to its dual role in epithelial barrier disruption and inflammatory amplification. We previously showed that phenethyl isothiocyanate (PEITC), an isothiocyanate derived from cruciferous vegetables, alleviated acute liver injury in mice by suppressing hepatocyte pyroptosis. In this study we evaluated the therapeutic potential of PEITC in the treatment of UC and the underlying mechanisms. UC mouse models were established by administration of 2.5% (w/v) dextran sulfate sodium (DSS) daily for 7 days. PEITC (5, 10, or 20 mg·kg-1·d-1, i.g.) was given 2 days before the start of modeling, and the dosing lasted for a total of 10 days. We showed that during the progression of DSS-induced UC, the pyroptosis pathway was activated accompanied by elevated expression levels of thioredoxin-interacting protein (TXNIP) and NOD-like receptor thermal protein domain associated protein 3 (NLRP3), as well as the activation of caspase-1, gasdermin D (GSDMD) and interleukin-1β (IL-1β). Treatment with PEITC dose-dependently reduced TXNIP and NLRP3 expression while inhibiting the cleavage of proteins associated with the pyroptosis pathway such as caspase-1, GSDMD, and IL-1β. We confirmed the inhibitory effect of PEITC on colonocyte pyroptosis in an in vitro model established in HT29 cells, where PEITC (0.2, 1, 5 µM) dose-dependently inhibited TXNIP and NLRP3 expression and the activation of pro-caspase-1, GSDMD and pro-IL-1β. We revealed that PEITC is directly bound to TXNIP and disrupted the interaction between TXNIP and NLRP3, leading to diminished cellular inflammation and oxidative stress levels. In conclusion, this study demonstrates that PEITC disrupts the interaction of TXNIP and NLRP3 by binding to TXNIP, inhibits NLRP3 activation and colonocyte pyroptosis, and thus effectively alleviates UC symptoms in mice. This study offers novel drug targets along with potential therapeutic candidates for the clinical prevention and treatment of UC.
Collapse
Affiliation(s)
- Jie Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing, 210000, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 210000, China
| | - Cui Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing, 210000, China
| | - Jia Qin
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing, 210000, China
| | - Ning An
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing, 210000, China
| | - Mei Bai
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing, 210000, China
| | - Rong-Hui Du
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing, 210000, China
| | - Yan Shen
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing, 210000, China
| | - Xu-Dong Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing, 210000, China
| | - Jing-Cai Cheng
- Drug R&D Institute, JC (Wuxi) Company, Inc., Wuxi, 214000, China
| | - Xue-Feng Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing, 210000, China.
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing, 210000, China.
| |
Collapse
|
20
|
Cai M, Zhang X, Gao X, Huo Q, Sun Y, Dai X. Chitooligosaccharide ameliorates cognitive deficits and neuroinflammation in APP/PS1 mice associated with the regulation of Nrf2/NF-κB axis. Int J Biol Macromol 2025; 303:140683. [PMID: 39914538 DOI: 10.1016/j.ijbiomac.2025.140683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 03/19/2025]
Abstract
Mounting evidence suggests that neuroinflammation is involved in the pathogenesis of neurodegenerative diseases such as Alzheimer's disease (AD). Amyloid β peptide (Aβ) could recruit and activate microglia, leading to the generation of pro-inflammatory factors, and ultimately neuroinflammation. Chitooligosaccharide (COS) is widely recognized as anti-inflammation bioactive substance, though whether it exerts beneficial effect on AD is unclear. In this study, we explored the effect of COS on AD prevention and treatment. We found that COS ameliorated cognitive deficiency, increased the expression of Nrf2 but decreased Aβ levels and the activation of NF-κB in APP/PS1 mice. In vitro, COS decreased the secretions of IL-6, IL-1β and TNF-α in Aβ25-35 + lipopolysaccharides (LPS) -exposed BV2 microglia. Meanwhile, COS down-regulated the expressions of iNOS, COX-2, NLRP3, caspase 1 and the nuclear translocation of NF-κB p65, while upregulated the expressions of Nrf2 and HO-1. Further, COS improved the viability of SK-N-SH cells that exposed to Aβ25-35 + LPS-stimulated microglial conditioned media, and the repressive effect of COS on NLRP3, iNOS, and phospho-NF-κB p65 expressions were markedly compromised upon Nrf2-siRNA transfection. Collectively, COS improved cognitive decline and suppressed neuroinflammation via the Nrf2/NF-κB signaling axis, suggesting COS might be a promising candidate in down-regulating inflammatory responses during AD progression.
Collapse
Affiliation(s)
- Mingyang Cai
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing 100023, China; Department of Food Science and Biomedicine, College of Biochemical Engineering, Beijing Union University, Beijing 100023, China
| | - Xiaoxia Zhang
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing 100023, China
| | - Xiaohan Gao
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing 100023, China
| | - Qing Huo
- Department of Food Science and Biomedicine, College of Biochemical Engineering, Beijing Union University, Beijing 100023, China
| | - Yaxuan Sun
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing 100023, China; Department of Food Science and Biomedicine, College of Biochemical Engineering, Beijing Union University, Beijing 100023, China
| | - Xueling Dai
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing 100023, China; Department of Food Science and Biomedicine, College of Biochemical Engineering, Beijing Union University, Beijing 100023, China.
| |
Collapse
|
21
|
Peng Y, Demidchik V, Li Y, Shen Z. Comparison of terpenoids in Nauclea officinalis and Paederia scandens and their anti-inflammatory effects on RAW264.7 macrophages. Fitoterapia 2025; 182:106411. [PMID: 39909359 DOI: 10.1016/j.fitote.2025.106411] [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: 10/18/2024] [Revised: 01/14/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Terpenoids are important components that exert pharmacological effects in Rubiaceae plants. In this study, two Rubiaceae plants, Nauclea officinalis and Paederia scandens, which are widely distributed in Hainan, China, were collected. The extracts of these two plants were obtained through boiling water extraction and analyzed using UHPLC-ESI-QE-Orbitrap-MS. By comparing with the mzCloud database, terpenoids with a matching rate of 85 % were identified. The results revealed that the aqueous extracts of N. officinalis mainly contain six pentacyclic triterpenoids, one diterpenoid, and one iridoid. The aqueous extracts of P. scandens contains one monoterpenoid, one diterpenoid, two pentacyclic triterpenes, and 4 iridoids. To verify the anti-inflammatory efficacy of the two extracts, in this study, they were added to the lipopolysaccharide (LPS)-induced RAW264.7 macrophages inflammation model. The results showed that the two extracts can reduce the secretion of proinflammatory cytokines IL-1β, IL-6, and TNF-α (p < 0.05) and the mortality rate of cell inflammation (p < 0.05) by inhibiting the activation of the NF-κB/NLRP3 pathway (p < 0.05). The study identified the terpenoids in N. officinalis and P. scandens and verified their anti-inflammatory effects in the RAW264.7 macrophages inflammation model.
Collapse
Affiliation(s)
- Yuxuan Peng
- Hainan College of Vocation and Technique, 95 Nanhai Ave., 570100 Haikou, China; Department of Plant Cell Biology and Bioengineering, Biology Faculty, Belarusian State University, 4 Independence Ave., 220030 Minsk, Belarus.
| | - Vadim Demidchik
- Institute of Experimental Botany, National Academy of Sciences of Belarus, 27 Botanichskaya St., 220072 Minsk, Belarus; International Research Centre for Environmental Membrane Biology and Department of Horticulture, Foshan University, Foshan, China
| | - Yan Li
- International Sakharov Environmental Institute, Belarusian State University, 15 Botanichskaya St., 220030 Minsk, Belarus
| | - Zhenguo Shen
- Hainan College of Vocation and Technique, 95 Nanhai Ave., 570100 Haikou, China.
| |
Collapse
|
22
|
Hume DA, Summers KM, O'Brien C, Pavli P. The Relationship Between CSF1R Signaling, Monocyte-Macrophage Differentiation, and Susceptibility to Inflammatory Bowel Disease. Cell Mol Gastroenterol Hepatol 2025; 19:101510. [PMID: 40154882 DOI: 10.1016/j.jcmgh.2025.101510] [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: 10/04/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025]
Abstract
More than 300 genomic loci have been associated with increased susceptibility to inflammatory bowel disease (IBD) through genome-wide association studies. A major challenge in the translation of genome-wide association studies to mechanistic insights lies in connecting noncoding variants to function. For example, single-nucleotide variants (SNVs) in the vicinity of the gene encoding the transcription factor ETS2 on human chromosome 21 are associated with the risk of developing IBD in Europeans. The peak of SNV association lies within a distal enhancer that may regulate ETS2 transcription. The interpretation of this and many other SNV associations with IBD depends on a model linking variation in transcriptional regulation to the likelihood of developing chronic intestinal inflammation. One model for the ETS2 locus is that overexpression in monocytes is causally associated with the risk allele, which in turn leads to a hyperinflammatory state. Here we summarize evidence for an alternative mechanism focused on negative regulators of monocyte-macrophage activation. We argue that IBD susceptibility arises from dysregulation of monocyte adaptation in the intestinal milieu to form resident intestinal macrophages that are anergic to inflammatory stimuli. This process depends on signals initiated by macrophage colony-stimulating factor (CSF1) binding to its receptor (CSF1R). Within this framework, ETS2 is a myeloid-specific transcription factor, expressed in pluripotent and committed progenitors and monocytes, and is down-regulated by CSF1, in common with many genes associated with IBD susceptibility, including NOD2. ETS2 is also both a downstream target and a mediator of the CSF1/CSF1R signaling pathway. Therapeutic targeting of ETS2 and its upstream regulators has the potential to prevent CSF1-dependent monocyte differentiation toward a prorepair resident macrophage phenotype and consequently exacerbate intestinal inflammation.
Collapse
Affiliation(s)
- David A Hume
- Mater Research Institute, University of Queensland, Woolloongabba, Brisbane, Australia.
| | - Kim M Summers
- Mater Research Institute, University of Queensland, Woolloongabba, Brisbane, Australia
| | - Claire O'Brien
- Centre for Research in Therapeutics Solutions, Faculty of Science and Technology, University of Canberra, Canberra, Australian Capital Territory, Australia
| | - Paul Pavli
- School of Medicine and Psychology, The Australian National University, Canberra, Australian Capital Territory, Australia; Gastroenterology and Hepatology Unit, Canberra Hospital, Canberra, Australian Capital Territory, Australia.
| |
Collapse
|
23
|
Feng X, Luo Y, Zheng M, Sun X, Shen X. Independent and Combined Associations between Metals Exposure and Inflammatory Markers among the General U.S. Adults. ENVIRONMENT & HEALTH (WASHINGTON, D.C.) 2025; 3:282-290. [PMID: 40144327 PMCID: PMC11934204 DOI: 10.1021/envhealth.4c00097] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/28/2024] [Accepted: 10/22/2024] [Indexed: 03/28/2025]
Abstract
Exposure to metals can trigger a series of diseases by dysregulating the human immune system, but there is still a lack of systematic studies assessing the independent and combined effects of exposure to metals on immune function in the general population, particularly concerning inflammation markers. This cross-sectional study was designed to mainly examine the associations between urinary metal mixtures and inflammatory markers, including white blood cell (WBC), platelet count (PLT), mean platelet volume (MPV), MPV/PLT ratio (MPR), platelet-to-lymphocyte ratio (PLR), and neutrophil-to-lymphocyte ratio (NLR). A total of 3451 participants aged ≥20 years were selected from the 2013-2016 National Health and Nutrition Examination Survey. Generalized linear models were used to investigate the relationships of exposure to single metals on inflammatory markers. Associations between coexposure to multiple metals and inflammatory markers were determined using weighted quantile sum regression and quantile g-computation. Barium, cadmium, lead, thallium, and cobalt showed significant associations with MPV, PLR, and NLR. Metal mixtures showed a negative association with MPV, while they had positive associations with PLR and NLR. Overall, our study highlights the significant effects of multiple metals exposure on inflammation markers, including MPV, PLR, and NLR, among U.S. adults. Thereinto, uranium, cadmium, and cobalt were identified as major contributors. Further prospective studies representative of other countries are warranted to either validate or refute our findings.
Collapse
Affiliation(s)
- Xinrui Feng
- State
Key Laboratory of Environment Health (Incubation), Key Laboratory
of Environment and Health, Ministry of Education, Key Laboratory of
Environment and Health (Wuhan), Ministry of Environmental Protection,
School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China
| | - Yaoyu Luo
- State
Key Laboratory of Environment Health (Incubation), Key Laboratory
of Environment and Health, Ministry of Education, Key Laboratory of
Environment and Health (Wuhan), Ministry of Environmental Protection,
School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China
| | - Min Zheng
- State
Key Laboratory of Environment Health (Incubation), Key Laboratory
of Environment and Health, Ministry of Education, Key Laboratory of
Environment and Health (Wuhan), Ministry of Environmental Protection,
School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China
| | - Xiaojie Sun
- Department
of Environmental Hygiene and Occupational Medicine, School of Public
Health, Wuhan University of Science and
Technology, Wuhan 430065, China
| | - Xiantao Shen
- State
Key Laboratory of Environment Health (Incubation), Key Laboratory
of Environment and Health, Ministry of Education, Key Laboratory of
Environment and Health (Wuhan), Ministry of Environmental Protection,
School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China
| |
Collapse
|
24
|
Shen S, Fang X, Zhang H, Lang T, Fu F, Du Y, Xu T, Jin H, Tong P, Wu C, Hu C, Ruan H. Systemic Lupus Erythematosus Stimulates Chondrocyte Pyroptosis to Aggravate Arthritis via Suppression of NRF-2/KEAP-1 and NF-κB Pathway. J Inflamm Res 2025; 18:4233-4250. [PMID: 40129871 PMCID: PMC11932136 DOI: 10.2147/jir.s502800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/14/2025] [Indexed: 03/26/2025] Open
Abstract
Purpose Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by diverse clinical manifestations, including joint symptoms. Arthritis represents one of the earliest manifestations of SLE, profoundly affecting the quality of life for affected individuals, yet the underlying mechanisms of SLE-associated arthritis remain insufficiently investigated. The study aimed to investigate the impact of SLE exacerbation on arthritis using the MRL/lpr mouse model, which closely mimics human SLE manifestations. Methods In the present study, we evaluated the impact of SLE onset on knee joint degeneration by comparing arthritic phenotype and complex molecular alterations between 6 female 14-week-old MRL/lpr mice, which manifest SLE, and MRL/MpJ mice, which remain unaffected. Results Our results demonstrated that MRL/lpr mice exhibited a more severe arthritic phenotype compared to MRL/MpJ mice, characterized by elevated Osteoarthritis Research Society International (OARSI) scores (P < 0.01), disrupted extracellular matrix metabolism, impaired chondrocyte proliferation and increased apoptosis. Notably, inflammatory cytokines proteins such as IL-1β and TNF-α (both P < 0.01), IL-18 and IL-6 (both P < 0.05), were significantly increased in articular cartilage of MRL/lpr mice, accompanied by increased expression of calcitonin gene-related peptide (CGRP) (P < 0.05), NETRIN-1, and NESTIN (both P < 0.01), indicating that SLE promotes inflammation response and sensory nerve ingrowth in the knee joint, contributing to the progression of arthritis. Mechanistic analysis revealed that SLE exacerbation intensified chondrocyte pyroptosis by upregulating pyroptotic-related proteins, including NLRP3, CASPASE-1, and gasdermin D (all P < 0.01), through the regulation of the nuclear factor erythroid 2-related factor (NRF-2)/KEAP-1 and nuclear factor kappa-B (NF-κB) pathway. Conclusion Collectively, our findings underscore the mechanistic connection between chondrocyte pyroptosis and arthritis exacerbation in SLE, suggesting potential therapeutic targets for mitigating arthritis progression in the context of SLE.
Collapse
Affiliation(s)
- Shuchao Shen
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Xuliang Fang
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Helou Zhang
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Tingting Lang
- School of Information and Electronic Engineering, Zhejiang University of Science and Technology, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Fangda Fu
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Yu Du
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Taotao Xu
- Department of Orthopaedics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Hongting Jin
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Peijian Tong
- Department of Orthopaedics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Chengliang Wu
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Changfeng Hu
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Hongfeng Ruan
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, 310053, People’s Republic of China
| |
Collapse
|
25
|
Cao R, Zhou J, Liu J, Wang Y, Dai Y, Jiang Y, Yamauchi A, Atlas D, Jin T, Zhou J, Wang C, Tan Q, Chen Y, Yodoi J, Tian H. TXM-CB13 Improves the Intestinal Mucosal Barrier and Alleviates Colitis by Inhibiting the ROS/TXNIP/TRX/NLRP3 and TLR4/MyD88/NF-κB/NLRP3 Pathways. Inflammation 2025:10.1007/s10753-025-02282-9. [PMID: 40085192 DOI: 10.1007/s10753-025-02282-9] [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: 05/31/2024] [Revised: 02/11/2025] [Accepted: 03/02/2025] [Indexed: 03/16/2025]
Abstract
The activation of inflammasomes (NLRP3 and NLRP1) is central to the pathogenesis of inflammatory bowel disease (IBD). Here we examined the protective effects of a thioredoxin-mimetic peptide CB13 (TXM-CB13), known for its antioxidative stress and anti-inflammatory properties. We examined the effects of TXM-CB13 on dextran sulfate sodium (DSS)-induced colitis and lipopolysaccharide (LPS)-induced NLRP3 inflammasome activation in RAW264.7 macrophages. TXM-CB13 appeared to alleviate symptoms of DSS-induced colitis and to significantly suppress the protein and mRNA levels of NLRP3, Mlck, and IL-1β in colonic tissues. Additionally, TXM-CB13 treatment increased the levels of the intestinal barrier proteins Occludin, ZO-1, and NLRP1, as shown through immunohistochemistry and Western blot analysis. In vitro, TXM-CB13 inhibited LPS-induced TLR4 signaling, reducing MyD88 levels and consequently attenuating the activation of the NF-κB pathways, including p-IκB-α/IκB-α and p-NF-κB-p65/NF-κB-p65. This inhibition further reduced the activation of the NLRP3 inflammasome components, NLRP3, ASC, Caspase-1, GSDMD, and IL-1β. In addition, TXM-CB13 prevented the ROS-mediated dissociation of TXNIP from TRX, inhibiting NLRP3 activation. These findings suggest that TXM-CB13 is a potential therapeutic candidate for IBD through its modulation of the TLR4/MyD88/NF-κB/NLRP3 and ROS/TXNIP/TRX/NLRP3 pathways.
Collapse
Affiliation(s)
- Ruijie Cao
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Jinhui Zhou
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Jiale Liu
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Yaxuan Wang
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Yandong Dai
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Yun Jiang
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Akira Yamauchi
- Department of Breast Surgery, Misugi-kai Sato Hospital Breast Center, HIrakata, Osaka, Japan
| | - Daphne Atlas
- Dept. Of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel
| | - Tiancheng Jin
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Jiedong Zhou
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Cuixue Wang
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Qihuan Tan
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Yifei Chen
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Junji Yodoi
- Laboratory of Infection and Prevention, Department of Biological Response, Institute for Virus Research, Kyoto University, Kyoto, Japan
| | - Hai Tian
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China.
- Jiaozhimei Biotechnology (Shaoxing) Co., Ltd., Shaoxing, China.
| |
Collapse
|
26
|
Li W, Chen Y, Li K, Chen Z, Zhang J, Zhao G, Sun F, Xiao P, Yang Y. Periplaneta americana extract improves recurrent oral ulcers through regulation of TLR4/NF-κB and Nrf2/HO-1 pathways. Sci Rep 2025; 15:8578. [PMID: 40075107 PMCID: PMC11903863 DOI: 10.1038/s41598-024-84703-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 12/26/2024] [Indexed: 03/14/2025] Open
Abstract
Recurrent oral ulcers (ROUs) of oral mucosa disease are difficult to cure and relapse easily, and immune imLbalance or dysfunction is considered an essential factor in their occurrence and recurrence. Periplaneta americana extract (PAD), a raw material used in Kangfuxin Liquid and Yunnan Baiyao toothpaste, contains a variety of growth factors such as polypeptides and sticky sugar amino acids that promote tissue repair; this can encourage the growth of the granulation tissue and reduce inflammation on the wound surface. This study aimed to investigate the interventional potential of PAD on recurrent oral ulcers in rats and to elucidate the underlying mechanism of action involving the TLR4/NF-κB and Nrf2/HO-1 signaling pathways. A rat model of recurrent oral ulcer (ROU) was established using an oral antigen emulsifier. Rats in the ROU group were administered PAD by gavage for 7 days. To observe the effect of PDA on ROU mice. HE staining revealed that PAD restored the structure of the oral mucosal tissue and reduced inflammatory infiltration. FCM revealed that PAD upregulated CD3 + and CD4 + levels and the CD4 + /CD8 + ratio in peripheral blood T lymphocytes. ELISA revealed that PAD increased the content of IgA, IgG, IgM, VEGF, IL-2, and IL-10, while decreasing IL-6 and TNF-α content. Microplate analysis revealed that PAD significantly increased CAT content in the serum of ROU rats and reduced GSH, NO, SOD, and MDA levels. IHC staining, RT-qPCR, and Western blotting revealed that PAD downregulated Keap1 and IκBα expression, inhibited the TLR4/NF-κB signaling pathway, upregulated Nrf2 and HO-1 expression, and activated the Nrf2/HO-1 signaling pathway. These fndings suggest that PAD improved immune imbalance and oxidative stress in ROU rats by activating the Nrf2/HO-1 pathway and inhibiting the TLR4/NF-κB signaling pathway, thereby promoting the healing of oral ulcer wounds.
Collapse
Affiliation(s)
- Weijun Li
- Engineering Research Center for Development of the Periplaneta Americana Industry of Yunnan Provincial Department of Education, Dali University, Dali, 671000, Yunnan, China
| | - Yi Chen
- Engineering Research Center for Development of the Periplaneta Americana Industry of Yunnan Provincial Department of Education, Dali University, Dali, 671000, Yunnan, China
| | - Kailing Li
- College of Pharmacy, Dali University, Dali, 671000, Yunnan, China
| | - Zhongze Chen
- Engineering Research Center for Development of the Periplaneta Americana Industry of Yunnan Provincial Department of Education, Dali University, Dali, 671000, Yunnan, China
| | - Jingyu Zhang
- College of Pharmacy, Dali University, Dali, 671000, Yunnan, China
| | - Guanhua Zhao
- Engineering Research Center for Development of the Periplaneta Americana Industry of Yunnan Provincial Department of Education, Dali University, Dali, 671000, Yunnan, China
| | - Fanfan Sun
- Engineering Research Center for Development of the Periplaneta Americana Industry of Yunnan Provincial Department of Education, Dali University, Dali, 671000, Yunnan, China
| | - Peiyun Xiao
- Engineering Research Center for Development of the Periplaneta Americana Industry of Yunnan Provincial Department of Education, Dali University, Dali, 671000, Yunnan, China.
| | - Yongshou Yang
- College of Pharmacy, Dali University, Dali, 671000, Yunnan, China.
| |
Collapse
|
27
|
He F, Lang Z, Huang Y, Qiu Y, Xiong P, Li N, Zhao G, Peng Y. Exogenous L-Serine Alleviates Pasteurella multocida-Induced Inflammation by Reprogramming the Transcription and Metabolism of Macrophages. Vet Sci 2025; 12:254. [PMID: 40267013 PMCID: PMC11945856 DOI: 10.3390/vetsci12030254] [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: 01/20/2025] [Revised: 02/05/2025] [Accepted: 03/04/2025] [Indexed: 04/25/2025] Open
Abstract
P. multocida is notorious for inducing excessive inflammation with high lethality in multiple animals, such as cattle, pigs, and chickens. Our previous study revealed that L-serine was decreased in the lungs of mice infected with P. multocida capsular type A strain CQ2 (PmCQ2), and 2 mg/kg of L-serine could alleviate PmCQ2-induced lung inflammation in vivo, which may largely depend on macrophages. However, the underlying intrinsic alterations remain unknown. Here, we demonstrated that 10 mM of L-serine significantly inhibited the release of inflammatory cytokines (e.g., IL-1β and TNF-α) by blocking inflammasome activation (including NALP1, NLRP3, NLRC4, AIM2, and Caspase-1) in PmCQ2-infected macrophages. Furthermore, the results of RNA-seq and metabonomics revealed that exogenous L-serine supplementation substantially reprogrammed macrophage transcription and metabolism. Mechanically, L-serine reduced inflammatory responses via the inhibition of glycolysis in macrophages based on a seahorse assay. Together, these findings characterize the intrinsic molecular alterations in activated macrophages and provide new targets for modulating P. multocida infection-induced macrophage inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Guangfu Zhao
- College of Veterinary Medicine, Southwest University, Chongqing 400715, China; (F.H.); (Z.L.); (Y.H.); (Y.Q.); (P.X.); (N.L.)
| | - Yuanyi Peng
- College of Veterinary Medicine, Southwest University, Chongqing 400715, China; (F.H.); (Z.L.); (Y.H.); (Y.Q.); (P.X.); (N.L.)
| |
Collapse
|
28
|
Wu F, Chen Y, Chen X, Tong D, Zhou J, Du Z, Yao C, Yang Y, Du A, Ma G. Nematode serine protease inhibitor SPI-I8 negatively regulates host NF-κB signalling by hijacking MKRN1-mediated polyubiquitination of RACK1. Commun Biol 2025; 8:356. [PMID: 40032982 PMCID: PMC11876351 DOI: 10.1038/s42003-025-07803-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 02/24/2025] [Indexed: 03/05/2025] Open
Abstract
Parasitic roundworms are remarkable for their ability to manipulate host immune systems and ameliorate inflammatory diseases. Although much is known about the nature of nematode effectors in immune modulation, little is known about the action mode of these molecules. Here, we report that a serine protease inhibitor SPI-I8 in the extracellular vesicles of blood-feeding nematodes like Ancylostoma ceylanicum, Haemonchus contortus and Nippostrongylus brasiliensis, effectively halts excessive inflammatory responses in vitro and in vivo. We demonstrate that H. contortus SPI-I8 promotes the role of a negative regulator of RACK1 and enhances the effects of RACK1 on tumor necrosis factor (TNF)-α-IκB kinases (IKKs)-nuclear factor kappa beta (NF-κB) axis in mammalian cells, by hijacking E3 ubiquitin protein ligase MKRN1-mediated polyubiquitination of RACK1. Administration of recombinant N. brasiliensis SPI-I8 effectively protects mice from dextran sulfate sodium (DSS)-induced colitis and lipopolysaccharide (LPS)-induced sepsis. Considering the structural and functional conservation of SPI-I8s among Strongylida nematodes and the conservation of interactive mediators (i.e., MKRN1 and RACK1) among mammals, our findings provide insights into the host-parasite interface where parasitic roundworms secret molecules to suppress host inflammatory responses. Harnessing these findings should underpin the exploitation of nematode's immunomodulators to relief excessive inflammation associated diseases in animals and humans.
Collapse
Affiliation(s)
- Fei Wu
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- College of Veterinary Medicine, Anhui Agricultural University, Hefei, China
| | - Yanqiong Chen
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xueqiu Chen
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Danni Tong
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingru Zhou
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Frontier Science Center for Brain and Brain-machine integration, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhendong Du
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chaoqun Yao
- Ross University School of Veterinary Medicine and One Health Center for Zoonoses and Tropical Veterinary Medicine, Ross University School of Veterinary Medicine, Basseterre, St. Kitts, Trinidad and Tobago
| | - Yi Yang
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Aifang Du
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Guangxu Ma
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
- ZJU-Xinchang Joint Innovation Centre (TianMu Laboratory), Gaochuang Hi-Tech Park, Xinchang, China.
| |
Collapse
|
29
|
Okasha AH, Hegab II, Seleem MA, Azzam AR, Ibrahim S, Ghalwash AA, El-Gohary RM. Effects of Fisetin and Nicorandil on adjuvant-induced rheumatoid arthritis in rats: Emerging role of TLR4/NF-κB-induced Pyroptosis, Nrf-2/HO-1, and OPG/RANKL pathways. Cytokine 2025; 187:156876. [PMID: 39884184 DOI: 10.1016/j.cyto.2025.156876] [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: 10/18/2024] [Revised: 01/04/2025] [Accepted: 01/24/2025] [Indexed: 02/01/2025]
Abstract
AIM AND BACKGROUND Our study explored the novel mechanisms implicated in the anti-rheumatic potential of fisetin and/or nicorandil (NIC) intervention. METHODS AND MATERIALS Fifty male rats were categorized into; control, rheumatoid arthritis (RA), fisetin-treated RA, NIC-treated RA, and co-treated RA groups. We assessed paw thickness, arthritis indices, serum CRP, RF, OPG, RANKL, and gene expressions of synovial TLR4, NLRP3, caspase-1, GSDMD, Nrf-2, and HO, along with synovial histopathology and NF-κB immunoreactivity. RESULTS The combined therapy demonstrated significantly better anti-rheumatic potential, suppressing oxidative stress and NF-κB, downregulating synovial TLR4, NLRP3, caspase-1, GSDMD, and increasing serum OPG while decreasing RANKL, confirmed by histopathological findings. CONCLUSION Our investigation uncovered the TLR4/NF-κB pyroptotic signaling, Nrf-2/HO-1, and OPG/RANKL pathways as novel mechanistic insights into the anti-rheumatoid potential of fisetin and/or NIC, with superiority of combination approach, providing a beacon of hope for RA patients in terms of optimizing treatment protocol effectiveness and patient outcomes.
Collapse
Affiliation(s)
- Asmaa H Okasha
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt.
| | - Islam Ibrahim Hegab
- Physiology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt,; Department of Bio-Physiology, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia.
| | - Monira A Seleem
- Medical Pharmacology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt.
| | - Asmaa R Azzam
- Human Anatomy and Embryology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt.
| | - Sarah Ibrahim
- Human Anatomy and Embryology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt.
| | - Asmaa A Ghalwash
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt.
| | - Rehab M El-Gohary
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt.
| |
Collapse
|
30
|
Cassavaugh J, Longhi MS, Robson SC. Impact of Estrogen on Purinergic Signaling in Microvascular Disease. Int J Mol Sci 2025; 26:2105. [PMID: 40076726 PMCID: PMC11900469 DOI: 10.3390/ijms26052105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Microvascular ischemia, especially in the heart and kidneys, is associated with inflammation and metabolic perturbation, resulting in cellular dysfunction and end-organ failure. Heightened production of adenosine from extracellular nucleotides released in response to inflammation results in protective effects, inclusive of adaptations to hypoxia, endothelial cell nitric oxide release with the regulation of vascular tone, and inhibition of platelet aggregation. Purinergic signaling is modulated by ectonucleoside triphosphate diphosphohydrolase-1 (NTPDase1)/CD39, which is the dominant factor dictating vascular metabolism of extracellular ATP to adenosine throughout the cardiovascular tissues. Excess levels of extracellular purine metabolites, however, have been associated with metabolic and cardiovascular diseases. Physiological estrogen signaling is anti-inflammatory with vascular protective effects, but pharmacological replacement use in transgender and postmenopausal individuals is associated with thrombosis and other side effects. Crucially, the loss of this important sex hormone following menopause or with gender reassignment is associated with worsened pro-inflammatory states linked to increased oxidative stress, myocardial fibrosis, and, ultimately, diastolic dysfunction, also known as Yentl syndrome. While there is a growing body of knowledge on distinctive purinergic or estrogen signaling and endothelial health, much less is known about the relationships between the two signaling pathways. Continued studies of the interactions between these pathways will allow further insight into future therapeutic targets to improve the cardiovascular health of aging women without imparting deleterious side effects.
Collapse
Affiliation(s)
- Jessica Cassavaugh
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (M.S.L.); (S.C.R.)
| | | | | |
Collapse
|
31
|
Xiao M, Pan Y, Tang S, Guo T, Yang L, Chen G. Carbon quantum dots amplify beneficial effects of EGCG against neural injuries by NLRP3 inflammasome after intracerebral hemorrhage. Int J Pharm 2025; 671:125281. [PMID: 39875034 DOI: 10.1016/j.ijpharm.2025.125281] [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: 11/28/2024] [Revised: 01/10/2025] [Accepted: 01/24/2025] [Indexed: 01/30/2025]
Abstract
Neuroinflammation plays an indispensable role in neural damages after ICH, responsible for the induced high mortality and poor prognosis. NLRP3 inflammasome, which is known mediated by ROS, has been widely documented to aggravate brain injuries. Therefore, suppressing neural injuries by ROS/NLRP3 pathway may be beneficial in treating ICH. As the major catechin found in green tea, epigallocatechin-3-gallate (EGCG) shows excellent anti-oxidative and anti-inflammatory effects. In this study, EGCG-carbon quantum dots (EGCG-CQDs) were successfully fabricated based on EGCG by hydrothermal synthesis method. EGCG-CQDs exhibited an excellent aqueous solubility, and emerged more abundant phenolic oxygens as well as oxygen-rich functional groups. Importantly, EGCG-CQDs showed superior free radical scavenging activity by DPPH and ABTS assays in vitro than EGCG. In vivo, a significant antioxidative activity was presented by EGCG-CQDs rather than EGCG. Furthermore, the upregulated NLRP3 and the induced inflammatory cascades (NF-κB, Caspase-1 and GSDMD) in ICH were attenuated by EGCG-CQDs. Inflammatory factor productions were also decreased by EGCG-CQDs, such as IL-1β, IL-18, IL-6 and TNF-α. Finally, the disturbed neural viability, disordered cytomorphology, and neurological deficits were significantly improved by EGCG-CQDs rather than EGCG. Therefore, CQDs might be an effective form to amplify the efficacy and bioavailability of EGCG, exerting considerable effects on treating ICH by suppressing ROS/NLRP3.
Collapse
Affiliation(s)
- Min Xiao
- School of Food Science and Engineering, Chongqing Technology and Business University, Chongqing 400067, China
| | - Yun Pan
- Department of Gastroenterology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Shijin Tang
- College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Tingwang Guo
- School of Food Science and Engineering, Chongqing Technology and Business University, Chongqing 400067, China.
| | - Lin Yang
- School of Food Science and Engineering, Chongqing Technology and Business University, Chongqing 400067, China
| | - Gang Chen
- School of Food Science and Engineering, Chongqing Technology and Business University, Chongqing 400067, China
| |
Collapse
|
32
|
Börmel L, Geisler AR, Hupfer Y, Liao S, Schubert T, Kluge S, Lorkowski S, Wallert M. The Vitamin E Derivative Garcinoic Acid Suppresses NLRP3 Inflammasome Activation and Pyroptosis in Murine Macrophages. Inflammation 2025:10.1007/s10753-025-02269-6. [PMID: 39982672 DOI: 10.1007/s10753-025-02269-6] [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: 10/24/2024] [Revised: 01/30/2025] [Accepted: 02/11/2025] [Indexed: 02/22/2025]
Abstract
Excessive inflammation in cells are a common cause of inflammation-related diseases such as cardiometabolic diseases. The cellular multiprotein complex nucleotide-binding domain and leucine-rich repeat pyrin domain 3 (NLRP3) inflammasome is a cellular key modulator of inflammatory processes. In addition to classic medications, phytochemicals are known for their anti-inflammatory potential. In African folk medicine the seeds of Garcinia kola are used to support the treatment of inflammatory diseases. Of particular interest is the phytochemical garcinoic acid (GA, trans-13'-carboxy-δ-tocotrienol), which is isolated from the Garcinia kola seeds. This derivative and potential metabolite of the vitamin E congener δ-tocotrienol (T3) shows anti-inflammatory properties in vitro. However, the underlying mechanisms are largely unknown. To get better insights into the molecular mode of action, murine J774A.1 macrophages were stimulated with lipopolysaccharides (LPS) only or in combination with adenosine triphosphate (ATP), which led to canonical activation of the NLRP3 inflammasome and subsequent pyroptosis. A combined treatment with GA resulted in significantly reduced stimulation of the transcription factor nuclear factor 'ĸ-light-chain-enhancer' of activated B-cells (NF-ĸB), decreased expression of inflammasome-related genes and marked downregulation of autoproteolytic cleavage of caspase-1 (Casp-1). Consequently, GA had an inhibitory effect on pyroptosis. The results have been validated using the well-known NLRP3 inflammasome inhibitor MCC950. In conclusion, GA was shown to have relevant effects on the regulation of the NLRP3 inflammasome and pyroptosis in vitro. Our study provides new mechanistic insights into the anti-inflammatory mode of action of GA and highlights its relevance as a potential phytochemical drug for the treatment of inflammation.
Collapse
Affiliation(s)
- Lisa Börmel
- Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany
| | - Anja R Geisler
- Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany
| | - Yvonne Hupfer
- Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany
| | - Sijia Liao
- Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany
| | - Tina Schubert
- Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany
| | - Stefan Kluge
- Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany
| | - Stefan Lorkowski
- Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany
| | - Maria Wallert
- Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany.
| |
Collapse
|
33
|
Kaszycki J, Kim M. Epigenetic regulation of transcription factors involved in NLRP3 inflammasome and NF-kB signaling pathways. Front Immunol 2025; 16:1529756. [PMID: 40046056 PMCID: PMC11879833 DOI: 10.3389/fimmu.2025.1529756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 02/03/2025] [Indexed: 05/13/2025] Open
Abstract
The NLRP3 inflammasome and NF-κB signaling pathways play crucial roles in orchestrating inflammation and immune defense. This review explores the intricate relationship between these pathways and epigenetic regulation, a field of growing importance in understanding immune responses. Epigenetic modifications, including DNA methylation, histone modifications, and non-coding RNAs (ncRNAs), significantly influence the activity of genes involved in these pathways, thereby modulating inflammatory responses. The review provides a comprehensive overview of current research on how epigenetic mechanisms interact with and regulate the NLRP3 inflammasome and NF-κB signaling pathways. It delves into advanced epigenetic concepts such as RNA modifications and 3D genome organization, and their impact on immune regulation. Furthermore, the implications of these findings for developing novel therapeutic strategies targeting epigenetic regulators in inflammatory diseases are discussed. By synthesizing recent advancements in this rapidly evolving field, this review underscores the critical role of epigenetic regulation in immune signaling and highlights the potential for epigenetic-based therapies in treating a wide range of inflammatory conditions, including autoimmune disorders and cancer.
Collapse
Affiliation(s)
- John Kaszycki
- Department of Biological Sciences, University of Connecticut, Storrs, CT, United States
| | - Minji Kim
- School of Pharmacy, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
34
|
Li H, Zhang P, Lin M, Li K, Zhang C, He X, Gao K. Pyroptosis: candidate key targets for mesenchymal stem cell-derived exosomes for the treatment of bone-related diseases. Stem Cell Res Ther 2025; 16:68. [PMID: 39940049 PMCID: PMC11816542 DOI: 10.1186/s13287-025-04167-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/21/2025] [Indexed: 02/14/2025] Open
Abstract
Bone-related diseases impact a large portion of the global population and, due to their high disability rates and limited treatment options, pose significant medical and economic challenges. Mesenchymal stem cells (MSCs) can differentiate into multiple cell types and offer strong regenerative potential, making them promising for treating various diseases. However, issues with the immune response and cell survival limit the effectiveness of cell transplantation. This has led to increased interest in cell-free stem cell therapy, particularly the use of exosomes, which is the most studied form of this approach. Exosomes are extracellular vesicles that contain proteins, lipids, and nucleic acids and play a key role in cell communication and material exchange. Pyroptosis, a form of cell death involved in innate immunity, is also associated with many diseases. Studies have shown that MSC-derived exosomes have therapeutic potential for treating a range of conditions by regulating inflammation and pyroptosis. This study explored the role of MSC-derived exosomes in modulating pyroptosis to improve the treatment of bone-related diseases.
Collapse
Affiliation(s)
- Haiming Li
- Shandong University of Traditional Chinese Medicine, Jinan, CN, China
| | - Peng Zhang
- Department of Orthopaedics, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China
| | - Minghui Lin
- Shandong University of Traditional Chinese Medicine, Jinan, CN, China
| | - Kang Li
- Department of Spine Surgery, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China
| | - Cunxin Zhang
- Department of Spine Surgery, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China.
| | - Xiao He
- Department of Orthopaedics, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China.
| | - Kai Gao
- Shandong University of Traditional Chinese Medicine, Jinan, CN, China.
- Department of Orthopaedics, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China.
| |
Collapse
|
35
|
Xu C, Yi M, Xiao Z, Xiang F, Wu M, Zhang Z, Zheng Y, Gong Y, Li Y, Su L, Liao Y, Zhang P, Xia B, Liao D, Lin L. New idea of Fuke Qianjin capsule in treating sequelae of pelvic inflammatory disease: Anti-inflammatory in the early stage and reparative in the later stage. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119066. [PMID: 39528116 DOI: 10.1016/j.jep.2024.119066] [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: 09/01/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sequelae of pelvic inflammatory disease (SPID) occurs in female internal genitalia and surrounding connective tissue. Recent clinical studies have shown that the traditional Chinese medicine Fuke Qianjin capsule (FKQ) can shorten the course of this disease, but its pharmacological effects and potential mechanism have not been fully elucidated. AIM OF THE STUDY This study aimed to investigate the efficacy and underlying mechanisms of FKQ in the treatment of SPID. METHODS In this study, we first established a mixed infection model to explore the protective effect of FKQ on common pathogens of SPID. Afterwards, mixed bacterial infection and mechanical injury were used in a SPID rat model to explore the protective mechanism of FKQ on SPID rats. Inflammation, repair and immune cells were tested. RESULTS FKQ has a protective effect against infections caused by SPID pathogenic bacterial and may reduce mortality from mixed infections. In the SPID model, FKQ improved pathological damage to the uterus, reduced the area of uterine fibrosis, and inhibited the levels of cytokines (TNF-α, IL-6, IL-1β, IL-18, TGF-β1 and VEGF) caused by pathogenic bacteria. Moreover, FKQ treatment reduced the accumulation of NLRP3, Caspase-1, GSDMD Vimentin, and Cytokeratin 18 in the uterus and suppressed the expression of TGF-β1 and VEGF in the fallopian tubes, thereby reducing inflammation and promoting mucosal repair. In addition, FKQ can restore the immune function balance of SPID rats by increasing the proportion of Treg cells in the spleen and thymus in a rat model of SPID, reducing the proportion of Th17 lymphocytes, and promoting an immunological balance of Treg/Th17 cells, thereby regulating the immune system of the body. CONCLUSION In summary, FKQ treatment for SPID is the result of a fourfold combination of antibacterial, anti-inflammatory, reparative and immune-enhancing activities.
Collapse
Affiliation(s)
- Chunfang Xu
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Meijin Yi
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Zhikui Xiao
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Feng Xiang
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Mengyao Wu
- Zhuzhou Qianjin Pharmaceutical Co., Ltd., Zhuzhou, 412003, China.
| | - Zhimin Zhang
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Yuanqing Zheng
- Zhuzhou Qianjin Pharmaceutical Co., Ltd., Zhuzhou, 412003, China.
| | - Yun Gong
- Zhuzhou Qianjin Pharmaceutical Co., Ltd., Zhuzhou, 412003, China.
| | - Yamei Li
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Liang Su
- Zhuzhou Qianjin Pharmaceutical Co., Ltd., Zhuzhou, 412003, China.
| | - Yingyan Liao
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Peng Zhang
- Zhuzhou Qianjin Pharmaceutical Co., Ltd., Zhuzhou, 412003, China.
| | - Bohou Xia
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Duanfang Liao
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Limei Lin
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
36
|
Yücer R, Schröder A, Topçu G, Efferth T. Identification of anti-inflammatory and anti-cancer compounds targeting the NF-κB-NLRP3 inflammasome pathway from a phytochemical library of the Sideritis genus. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119074. [PMID: 39522840 DOI: 10.1016/j.jep.2024.119074] [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: 03/06/2024] [Revised: 10/09/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
ETHNOBOTANICAL RELEVANCE For centuries, the aerial parts of Sideritis species have been known for their medicinal properties as herbal teas. Although the antioxidant and anti-inflammatory properties of the genus have been widely documented, the underlying mechanisms are yet to be sufficiently clarified. AIM OF THE STUDY We investigated the anti-inflammatory and anticancer activities of phytochemicals of the Sideritis genus. MATERIAL AND METHODS Through literature mining, a chemical library containing 657 components of the Sideritis genus was formed. We studied these compounds for binding to NLRP3 and NF-κB proteins in silico by virtual drug screening and molecular docking, and in vitro by microscale thermophoresis (MST). Liquid chromatography-high-resolution mass spectrometry analysis (LC-HRMS) was performed in the Sideritis extracts. One of the identified compounds, verbascoside, was investigated for its cytotoxic activity by mining a panel of 49 tumor cell lines in the data repository of the National Cancer Institute (NCI, USA). RESULTS Virtual screening and molecular docking results highlighted two compounds targeting both proteins of interest, i.e., verbascoside (acteoside) and apigenin 7,4'-bis(trans-p-coumarate), as both had lowest binding energies of less than -10 kcal/mol. Using MST, we then verified that both compounds bound to the target proteins. Verbascoside bound to NLRP3 and NF-κB with Kd values of 0.67 ± 0.18 μM and 0.01 ± 0.08 μM, while apigenin 7,4'-bis(trans-p-coumarate) had Kd values of 4.60 ± 1.66 μM and 0.27 ± 0.75 μM, respectively. Verbascoside was abundant in the Sideritis extracts, according to LC-HRMS analysis. Since inflammation is strongly related to carcinogenesis, we investigated the anticancer activity of verbascoside in the second part of this study. We investigated the activity of verbascoside in 49 tumor cell lines of the NCI. Comparing its activity with 81 standard anticancer drugs revealed numerous interactions with DNA-damaging agents (alkylators, topoisomerase I/II inhibitors, antimetabolites), indicating that verbascoside may also affect the DNA of tumor cells. We further investigated the involvement of verbascoside in several main drug resistance mechanisms, i.e., ABC transporters, oncogenes, tumor suppressors, cellular proliferation rates, and other parameters. Except for the correlation to the mutational status of NRAS, no other significant relationships were found, indicating that verbascoside is not involved in most of the common drug resistance mechanisms. Two-dimensional cluster analysis-based heatmap generation of a proteomic profile from 40 out of 3171 proteins revealed a significant correlation between the expression of these proteins in 49 tumor cell lines, and the cellular response to verbascoside. This indicates that the presence of these proteins is a determinant for sensitivity or resistance to this natural product. CONCLUSION The database established here represents a valuable resource for the screening of bioactivites of the Sideritis genus. The experimental validation of the anti-inflammatory and cytotoxic activities of selected compounds proved that virtual drug screening and molecular docking are suitable tools for the identification of putative drug candidates. Verbascoside was among the top 10 compounds binding to two key anti-inflammatory proteins, NLRP3 and NF-kB. Additionally, data from the NCI indicate that verbascoside is not linked to main drug resistance mechanisms.
Collapse
Affiliation(s)
- Rümeysa Yücer
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128, Mainz, Germany.
| | - Angela Schröder
- Theophrastus Paracelsus Foundation, 64367, Mühltal, Germany.
| | - Gülaçtı Topçu
- Department of Pharmacognosy and Phytochemistry, Faculty of Pharmacy, Bezmialem Vakif University, 34093, Fatih, Istanbul, Turkiye; Drug Application & Research Center (DARC), Bezmialem Vakif University, 34093, Fatih, Istanbul, Turkiye.
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128, Mainz, Germany.
| |
Collapse
|
37
|
Jiang Y, Wang Z, Wang W, Liu Y, Meng Y, Wang Y, Fan M, Cai C. Ganoderma lucidum polysaccharide alleviates cognitive dysfunction by inhibiting neuroinflammation via NLRP3/NF-κB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119065. [PMID: 39522844 DOI: 10.1016/j.jep.2024.119065] [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: 08/11/2024] [Revised: 10/21/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ganoderma lucidum (G. lucidum), a traditional Chinese medicinal herb, is commonly recommended for its potential to promote mental relaxation and alleviate memory impairment. Recently, there have been reports suggesting that it exhibits anti-neuroinflammatory activity through the gut-brain axis. Cognitive dysfunction is among the most prevalent neurodegenerative diseases. AIM OF THE STUDY This study aimed to investigate the efficacy of polysaccharides extracted from G. lucidum in alleviating cognitive dysfunction. METHODS AND MATERIALS A polysaccharide was extracted through the process of alkali extraction followed by alcohol precipitation. Comprehensive analysis was conducted to characterize the total sugar content, amino acid composition, and sugar chain structure. The levels of inflammatory related factors were assessed using griess reagent, qPCR and western blotting assay in vitro. The efficacy of alleviating cognitive dysfunction was evaluated through a series of behavioral studies in mice model induced by the high-fat high-sugar diet combined with chronic unpredictable mild stress (HFFD/CUMS) in vivo. The mechanism was investigated by 16S rRNA sequence, immunohistochemistry, flow cytometry and short-chain fatty acid detection. RESULTS Ganoderma lucidum polysaccharide (GLP) is a polysaccharide identified as β-glucan. Bioactivity experiments have demonstrated that GLP possesses the potential to ameliorate cognitive dysfunction. The mechanism study revealed that GLP can modulate the composition of gut microbiota and suppress the activation of inflammasomes via NLRP3/NF-κB signaling pathway, thereby attenuating neuroinflammatory. Furthermore, GLP may enhance the peripheral immunity response of the body, leading to a comprehensive regulatory effect. CONCLUSION A polysaccharide alleviates cognitive dysfunction via inhibiting neuroinflammation.
Collapse
Affiliation(s)
- Yudi Jiang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Shandong Key Laboratory of Glycoscience and Glycotherapeutic, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Zhe Wang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Shandong Key Laboratory of Glycoscience and Glycotherapeutic, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Wanshuai Wang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Shandong Key Laboratory of Glycoscience and Glycotherapeutic, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Yang Liu
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Shandong Key Laboratory of Glycoscience and Glycotherapeutic, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Yang Meng
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, 266001, Shandong, China
| | - Yaozhong Wang
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, 266001, Shandong, China
| | - Minghao Fan
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Key Laboratory of Biological Fermentation Engineering of Beer, Tsingtao Brewery Co., Ltd, Qingdao, 266100, Shandong, China.
| | - Chao Cai
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Shandong Key Laboratory of Glycoscience and Glycotherapeutic, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| |
Collapse
|
38
|
Xing Y, Zheng Y, Zhang J, Chen L, Xu Y, Jin X, Hong L, Yan S, Shi B. Purification, Characterization, and Potential Immune-Regulation Mechanism of Polysaccharides from Artemisia odosica Krasch. Molecules 2025; 30:675. [PMID: 39942779 PMCID: PMC11820876 DOI: 10.3390/molecules30030675] [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: 01/01/2025] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
Artemisia ordosica Krasch. represents a medicinal species traditionally and extensively employed in traditional medicine for treating ailments such as rheumatic arthritis, sore throat, and inflammation. This study initially focuses on the extraction, purification, and characterization of Artemisia ordosica Krasch. polysaccharides (AOP). The purified AOP exhibits a molecular mass corresponding to 9.00 kDa and consists of multiple monosaccharide units, with glucose (54.08%) as the predominant component, followed by arabinose (13.75%), mannose (13.43%), galactose (12.79%), xylose (3.15%), glucuronic acid (0.93%), galacturonic acid (0.67%), ribose (0.63%), and fucose (0.56%), respectively. Furthermore, to explore the immune-regulatory mechanisms of AOP, peripheral blood lymphocytes (PBLs) were cultured and exposed to inhibitors targeting receptors and signaling molecules. The results indicated that TLR4 serves as a potential target through which AOP exerts its immunomodulatory functions. AOP mitigates immune stress in PBLs triggered by LPS by disrupting the interaction between LPS and TLR and downregulating the over-activation of the nuclear factor kappa B (NF-κB) signaling pathway. In summary, AOP shows promise as a feed additive to protect animals from immune stress.
Collapse
Affiliation(s)
- Yuanyuan Xing
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.X.); (Y.Z.); (Y.X.); (X.J.); (L.H.); (S.Y.)
| | - Yankai Zheng
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.X.); (Y.Z.); (Y.X.); (X.J.); (L.H.); (S.Y.)
| | - Jing Zhang
- Intellectual Property Protection Center of Inner Mongolia Autonomous Region, Hohhot 010000, China;
| | - Lu Chen
- Animal Husbandry and Veterinary Department, Shanxi Animal Husbandry and Veterinary School, Taiyuan 030024, China;
| | - Yuanqing Xu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.X.); (Y.Z.); (Y.X.); (X.J.); (L.H.); (S.Y.)
| | - Xiao Jin
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.X.); (Y.Z.); (Y.X.); (X.J.); (L.H.); (S.Y.)
| | - Lei Hong
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.X.); (Y.Z.); (Y.X.); (X.J.); (L.H.); (S.Y.)
| | - Sumei Yan
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.X.); (Y.Z.); (Y.X.); (X.J.); (L.H.); (S.Y.)
| | - Binlin Shi
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.X.); (Y.Z.); (Y.X.); (X.J.); (L.H.); (S.Y.)
| |
Collapse
|
39
|
Tork MAB, Fotouhi S, Roozi P, Negah SS. Targeting NLRP3 Inflammasomes: A Trojan Horse Strategy for Intervention in Neurological Disorders. Mol Neurobiol 2025; 62:1840-1881. [PMID: 39042218 DOI: 10.1007/s12035-024-04359-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 07/09/2024] [Indexed: 07/24/2024]
Abstract
Recently, a growing focus has been on identifying critical mechanisms in neurological diseases that trigger a cascade of events, making it easier to target them effectively. One such mechanism is the inflammasome, an essential component of the immune response system that plays a crucial role in disease progression. The NLRP3 (nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 3) inflammasome is a subcellular multiprotein complex that is widely expressed in the central nervous system (CNS) and can be activated by a variety of external and internal stimuli. When activated, the NLRP3 inflammasome triggers the production of proinflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18) and facilitates rapid cell death by assembling the inflammasome. These cytokines initiate inflammatory responses through various downstream signaling pathways, leading to damage to neurons. Therefore, the NLRP3 inflammasome is considered a significant contributor to the development of neuroinflammation. To counter the damage caused by NLRP3 inflammasome activation, researchers have investigated various interventions such as small molecules, antibodies, and cellular and gene therapy to regulate inflammasome activity. For instance, recent studies indicate that substances like micro-RNAs (e.g., miR-29c and mR-190) and drugs such as melatonin can reduce neuronal damage and suppress neuroinflammation through NLRP3. Furthermore, the transplantation of bone marrow mesenchymal stem cells resulted in a significant reduction in the levels of pyroptosis-related proteins NLRP3, caspase-1, IL-1β, and IL-18. However, it would benefit future research to have an in-depth review of the pharmacological and biological interventions targeting inflammasome activity. Therefore, our review of current evidence demonstrates that targeting NLRP3 inflammasomes could be a pivotal approach for intervention in neurological disorders.
Collapse
Affiliation(s)
- Mohammad Amin Bayat Tork
- Clinical Research Development Unit, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soroush Fotouhi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Roozi
- Department of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sajad Sahab Negah
- Clinical Research Development Unit, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran.
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Pardis Campus, Azadi Square, Kalantari Blvd., Mashhad, Iran.
| |
Collapse
|
40
|
Zhao Q, Wang Q, Yao Q, Yang Z, Li W, Cheng X, Wen Y, Chen R, Xu J, Wang X, Qin D, Zhu S, He L, Li N, Wu Y, Yu Y, Cao X, Wang P. Nonenzymatic lysine D-lactylation induced by glyoxalase II substrate SLG dampens inflammatory immune responses. Cell Res 2025; 35:97-116. [PMID: 39757301 PMCID: PMC11770101 DOI: 10.1038/s41422-024-01060-w] [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: 05/17/2024] [Accepted: 11/22/2024] [Indexed: 01/07/2025] Open
Abstract
Immunometabolism is critical in the regulation of immunity and inflammation; however, the mechanism of preventing aberrant activation-induced immunopathology remains largely unclear. Here, we report that glyoxalase II (GLO2) in the glycolysis branching pathway is specifically downregulated by NF-κB signaling during innate immune activation via tristetraprolin (TTP)-mediated mRNA decay. As a result, its substrate S-D-lactoylglutathione (SLG) accumulates in the cytosol and directly induces D-lactyllysine modification of proteins. This nonenzymatic lactylation by SLG is greatly facilitated by a nearby cysteine residue, as it initially reacts with SLG to form a reversible S-lactylated thiol intermediate, followed by SN-transfer of the lactyl moiety to a proximal lysine. Lactylome profiling identifies 2255 lactylation sites mostly in cytosolic proteins of activated macrophages, and global protein structure analysis suggests that proximity to a cysteine residue determines the susceptibility of lysine to SLG-mediated D-lactylation. Furthermore, lactylation is preferentially enriched in proteins involved in immune activation and inflammatory pathways, and D-lactylation at lysine 310 (K310) of RelA attenuates inflammatory signaling and NF-κB transcriptional activity to restore immune homeostasis. Accordingly, TTP-binding site mutation or overexpression of GLO2 in vivo blocks this feedback lactylation in innate immune cells and promotes inflammation, whereas genetic deficiency or pharmacological inhibition of GLO2 restricts immune activation and attenuates inflammatory immunopathology both in vitro and in vivo. Importantly, dysregulation of the GLO2/SLG/D-lactylation regulatory axis is closely associated with human inflammatory phenotypes. Overall, our findings uncover an immunometabolic feedback loop of SLG-induced nonenzymatic D-lactylation and implicate GLO2 as a promising target for combating clinical inflammatory disorders.
Collapse
Affiliation(s)
- Qihang Zhao
- National Key Laboratory of Immunity & Inflammation, Second Military Medical University, Shanghai, China
| | - Qiang Wang
- Department of Urology, People's Hospital, Peking University, Beijing, China
| | - Qinghua Yao
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Xinhua Hospital of Zhejiang Province, Zhejiang, China
| | - Zhengdong Yang
- National Key Laboratory of Immunity & Inflammation, Second Military Medical University, Shanghai, China
| | - Wenfang Li
- Department of Emergency and Intensive Care Unit, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xiaojie Cheng
- National Key Laboratory of Immunity & Inflammation, Second Military Medical University, Shanghai, China
| | - Yingling Wen
- National Key Laboratory of Immunity & Inflammation, Second Military Medical University, Shanghai, China
| | - Rong Chen
- Department of Urology, People's Hospital, Peking University, Beijing, China
| | - Junfang Xu
- Clinical and Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xuanying Wang
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Xinhua Hospital of Zhejiang Province, Zhejiang, China
| | - Dexiang Qin
- National Key Laboratory of Immunity & Inflammation, Second Military Medical University, Shanghai, China
| | - Shuyang Zhu
- National Key Laboratory of Immunity & Inflammation, Second Military Medical University, Shanghai, China
| | - Liujie He
- National Key Laboratory of Immunity & Inflammation, Second Military Medical University, Shanghai, China
| | - Nan Li
- National Key Laboratory of Immunity & Inflammation, Second Military Medical University, Shanghai, China
| | - Yanfeng Wu
- National Key Laboratory of Immunity & Inflammation, Second Military Medical University, Shanghai, China
| | - Yizhi Yu
- National Key Laboratory of Immunity & Inflammation, Second Military Medical University, Shanghai, China.
| | - Xuetao Cao
- National Key Laboratory of Immunity & Inflammation, Second Military Medical University, Shanghai, China.
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China.
- Frontier Research Center for Cell Response, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China.
| | - Pin Wang
- National Key Laboratory of Immunity & Inflammation, Second Military Medical University, Shanghai, China.
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, China.
| |
Collapse
|
41
|
Pan Y, Nie L, Chen W, Guan D, Li Y, Yang C, Duan L, Wan T, Zhuang L, Lai J, Li W, Zhang Y, Wang Q. Buyang Huanwu Decoction prevents hemorrhagic transformation after delayed t-PA infusion via inhibiting NLRP3 inflammasome/pyroptosis associated with microglial PGC-1α. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119275. [PMID: 39710159 DOI: 10.1016/j.jep.2024.119275] [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: 09/23/2024] [Revised: 11/30/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Delayed tissue-type plasminogen activator (t-PA) thrombolysis, which has a restrictive therapeutic time window within 4.5 h following ischemic stroke (IS), increases the risk of hemorrhagic transformation (HT) and subsequent neurotoxicity. Studies have shown that the NLRP3 inflammasome activation reversely regulated by the PGC-1α leads to microglial polarization and pyroptosis to cause damage to nerve cells and the blood-brain barrier. The effect of Buyang Huanwu Decoction (BHD), a traditional Chinese medicine prescription widely used in the recovery of IS, on HT injury after delayed t-PA treatment had been found with clinical studies, while the underlying mechanisms are reminded to be further clarified. AIM OF THE STUDY This study sought to investigate the therapeutic effect and the underlying mechanisms of BHD in ischemic rat brains with delayed t-PA treatment. MATERIALS AND METHODS The components of BHD extracts were identified by High Performance Liquid Chromatography (HPLC) and the effective components in the rat brains from BHD were analyzed by liquid chromatography-mass spectrometry (LC-MS). In vivo experiment was carried out by 5 h of middle cerebral artery occlusion (MCAO) following by t-PA infusion for 0.5 h plus reperfusion 19 h, while the in vitro BV2 cells were stimulated by lipopolysaccharide (LPS)-adenosine triphosphate (ATP) to activate microglia pyroptosis, of which the MCC950 (NLRP3 inhibitor) and NSA (GSDMD inhibitor) were adopted as reverse validation. PGC-1α siRNA was utilized to study the mechanisms of BHD against microglial polarization and pyroptosis in BV2 cells. RESULTS HPLC analysis demonstrated the fingerprint of BHD with six reference standards (Hydroxysafflor yellow A, Calycosin-7-glucoside, Paeoniflorin, Formononetin, Ferulic acid and Amygdalin), the last two of which can be found in rat brains by LC-MS analysis. In the following experiments, we found the major discoveries as follow: (1) BHD improved the neurological outcomes, the structural integrity of the blood-brain barrier and the neuronal structure in HT rats with MCAO following by delayed t-PA infusion; (2) the presence of t-PA promoted the suppression of PGC-1α and the activation of microglial NLRP3 inflammasome and pyroptosis in the HT rats; (3) BHD promoted the transformation of microglia from M1 to M2 type for inhibiting inflammatory response; (4) BHD restrained NLRP3 inflammasome/pyroptosis activation in microglia, prevented the translocations of NF-κB into the nucleus, as well as enhanced microglia-specific PGC-1α in ischemic rats following t-PA delayed thrombolysis; (5) BHD suppressed NLRP3 inflammasome assembly and increased PGC-1α expression in the LPS-ATP-induced BV2 cells; (6) PGC-1α silencing withdrew the protective role of BHD against NLRP3 inflammasome/pyroptosis. CONCLUSION Mechanistically, BHD existed the protective effect against HT injury after delayed t-PA treatment through up-regulating microglial PGC-1α to inhibit NLRP3 inflammasome and pyroptosis, and serves as a potential adjuvant therapy for HT injury.
Collapse
Affiliation(s)
- Yaru Pan
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Linlin Nie
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Weitao Chen
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Danni Guan
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Yongyi Li
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Cong Yang
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Lining Duan
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China; The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Ting Wan
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China; The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Lixing Zhuang
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China; The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Jianbo Lai
- Shenzhen Integrated Traditional Chinese and Western Medicine Hospital, Shenzhen, Guangdong, 518100, China
| | - Weirong Li
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Yifan Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China; The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, 510405, Guangdong, China.
| | - Qi Wang
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
42
|
Chen W, Peng J, Tang X, Ouyang S. MSC-derived exosome ameliorates pulmonary fibrosis by modulating NOD 1/NLRP3-mediated epithelial-mesenchymal transition and inflammation. Heliyon 2025; 11:e41436. [PMID: 39872463 PMCID: PMC11761938 DOI: 10.1016/j.heliyon.2024.e41436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 12/12/2024] [Accepted: 12/20/2024] [Indexed: 01/30/2025] Open
Abstract
Background Pulmonary fibrosis (PF) is an irreversible and usually fatal lung disease. In recent years, the therapeutic role of exosomes derived from mesenchymal stem cells (MSC-exos) in anti-fibrotic treatment has received much attention. In this study, we aimed to determine the anti-fibrotic properties and related molecular mechanisms of MSC-exos in Bleomycin(BLM)-induced PF. Methods We used BLM-induced mice model of PF and in vitro model. MSC-exos were isolated from BMSCs cells using Exo Quick-TC kit and identified using conventional methods. Using cell counting kit-8 (CCK-8) to detect cell viability. Classic molecular biology approaches such as RT-qPCR, Western blot, immunofluorescence, and ELISA were used to examine molecular pathways. Histopathological examination was performed using HE and Masson staining. Results MSC-exos alleviated inflammation, inhibited epithelial-mesenchymal transition (EMT), and ameliorated PF. Further studies showed that MSC-exos regulated NOD1/NF-kB signaling pathway to suppress the activation of NLRP3 inflammasomes both in vivo and in vitro. Additionally, overexpression of NLRP3 significantly reversed the anti-fibrotic effects of MSC-exos in BLM-induced lung epithelial cells. Conclusion MSC-derived exosome ameliorates pulmonary fibrosis by modulating NOD 1/NLRP3-mediated epithelial-mesenchymal transition and inflammation.
Collapse
Affiliation(s)
- Wei Chen
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Jie Peng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xiangyi Tang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Shao Ouyang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of University of South China, Key Laboratory of Heart Failure Prevention & Treatment of Hengyang, Clinical Medicine Research Center of Arteriosclerotic Disease of Hunan Province, Hengyang, Hunan, China
| |
Collapse
|
43
|
Liao Y, Kong Y, Chen H, Xia J, Zhao J, Zhou Y. Unraveling the priming phase of NLRP3 inflammasome activation: Molecular insights and clinical relevance. Int Immunopharmacol 2025; 146:113821. [PMID: 39674000 DOI: 10.1016/j.intimp.2024.113821] [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: 09/13/2024] [Revised: 11/10/2024] [Accepted: 12/05/2024] [Indexed: 12/16/2024]
Abstract
The NLRP3 inflammasome plays a pivotal role in the innate immune response. Its activation involves a two-step mechanism that consists of priming and activation. The priming of the NLRP3 inflammasome is a vital initial phase necessary for its activation and subsequent involvement in the immune response, though its understanding varies across studies. Recent research has identified key proteins that influence the priming process, revealing a sophisticated regulatory network. This review provides a comprehensive review of the priming phase of NLRP3 inflammasome activation, with a particular focus on the underlying molecular mechanisms, including transcriptional regulation, orchestration of the phosphorylation status, deubiquitination and the relationships with the inflammation-associated diseases. Understanding the intricacies of NLRP3 inflammasome priming not only elucidates fundamental aspects of immune regulation, but also provides potential avenues for therapeutic intervention in inflammatory diseases.
Collapse
Affiliation(s)
- Yonghong Liao
- College of Veterinary Medicine, Southwest University, 402460 Chongqing, China; National Center of Technology Innovation for Pigs, 402460, Rongchang, Chongqing, China
| | - Yueyao Kong
- College of Veterinary Medicine, Southwest University, 402460 Chongqing, China
| | - Hongyu Chen
- College of Veterinary Medicine, Southwest University, 402460 Chongqing, China
| | - Jing Xia
- College of Veterinary Medicine, Southwest University, 402460 Chongqing, China
| | - Jianjun Zhao
- College of Animal Science and Technology, Southwest University, 402460 Chongqing, China
| | - Yang Zhou
- College of Veterinary Medicine, Southwest University, 402460 Chongqing, China; National Center of Technology Innovation for Pigs, 402460, Rongchang, Chongqing, China.
| |
Collapse
|
44
|
Dou Y, Zhang Y, Liu Y, Sun X, Liu X, Li B, Yang Q. Role of macrophage in intervertebral disc degeneration. Bone Res 2025; 13:15. [PMID: 39848963 PMCID: PMC11758090 DOI: 10.1038/s41413-024-00397-7] [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: 07/01/2024] [Revised: 12/04/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
Intervertebral disc degeneration is a degenerative disease where inflammation and immune responses play significant roles. Macrophages, as key immune cells, critically regulate inflammation through polarization into different phenotypes. In recent years, the role of macrophages in inflammation-related degenerative diseases, such as intervertebral disc degeneration, has been increasingly recognized. Macrophages construct the inflammatory microenvironment of the intervertebral disc and are involved in regulating intervertebral disc cell activities, extracellular matrix metabolism, intervertebral disc vascularization, and innervation, profoundly influencing the progression of disc degeneration. To gain a deeper understanding of the inflammatory microenvironment of intervertebral disc degeneration, this review will summarize the role of macrophages in the pathological process of intervertebral disc degeneration, analyze the regulatory mechanisms involving macrophages, and review therapeutic strategies targeting macrophage modulation for the treatment of intervertebral disc degeneration. These insights will be valuable for the treatment and research directions of intervertebral disc degeneration.
Collapse
Affiliation(s)
- Yiming Dou
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Yiming Zhang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, 300070, China
| | - Yang Liu
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Xun Sun
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Xinyu Liu
- Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China.
| | - Bin Li
- Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215007, China.
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China.
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
45
|
Moura FA, Siqueira AIDAN. Gut-liver axis in sepsis-associated liver injury: Epidemiology, challenges and clinical practice. World J Gastroenterol 2025; 31:99987. [PMID: 39777244 PMCID: PMC11684188 DOI: 10.3748/wjg.v31.i1.99987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/15/2024] [Accepted: 11/13/2024] [Indexed: 12/09/2024] Open
Abstract
Although the liver has a remarkable regenerative capacity, sepsis-associated liver injury (SLI) is a complication often seen in intensive care units. Due to its role in immune and inflammatory regulation, the liver is particularly vulnerable during severe infections. Understanding the global prevalence, causes, and management of SLI is essential to improve outcomes and reduce healthcare costs. This paper aims to explore these factors, with an emphasis on identifying effective strategies for clinical management. Zhang et al's bibliometric analysis of 787 publications (745 original articles and 42 reviews, mostly in animal models) from 2000 to 2023 highlights the growing interest in SLI, focusing on oxidative stress, gut microbiota, and inflammatory processes. Key components such as nuclear factor-kappa B and the NOD-like receptor thermal protein domain associated protein 3 inflammasome pathway, along with their links to gut microbiota imbalance and oxidative stress, are crucial for understanding SLI pathogenesis. The gut-liver axis, particularly the role of intestinal permeability and bacterial translocation in liver inflammation, is emphasized. In this context, bacterial translocation is especially relevant for critically ill patients, as it can exacerbate liver inflammation. The findings underscore the need for integrated care in intensive care units, prioritizing gut health and careful antibiotic use to prevent dysbiosis. Despite extensive research, there remains a lack of clinical trials to validate therapeutic approaches. The abundance of experimental studies highlights potential therapeutic targets, stressing the need for high-quality randomized clinical trials to translate these findings into clinical practice.
Collapse
Affiliation(s)
- Fabiana Andréa Moura
- College of Nutrition, Federal University of Alagoas, Maceio 57072-970, Alagoas, Brazil
- College of Medicine, Federal University of Alagoas, Maceio 57072-970, Alagoas, Brazil
| | | |
Collapse
|
46
|
Chen J, Jia S, Xue X, Guo C, Dong K. Gut microbiota: a novel target for exercise-mediated regulation of NLRP3 inflammasome activation. Front Microbiol 2025; 15:1476908. [PMID: 39834360 PMCID: PMC11743191 DOI: 10.3389/fmicb.2024.1476908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
The NOD-like receptor family pyrin domain-containing 3 (NLRP3) is a key pattern recognition receptor in the innate immune system. Its overactivation leads to the production of pro-inflammatory cytokines, such as IL-1β and IL-18, which contribute to the development and progression of various diseases. In recent years, evidence has shown that gut microbiota plays an important role in regulating the activation of NLRP3 inflammasome. Variations in the function and composition of gut microbiota can directly or indirectly influence NLRP3 inflammasome activation by influencing bacterial components and gut microbiota metabolites. Additionally, exercise has been shown to effectively reduce NLRP3 inflammasome overactivation while promoting beneficial changes in gut microbiota. This suggests that gut microbiota may play a key role in mediating the effects of exercise on NLRP3 inflammasome regulation. This review explores the impact of exercise on gut microbiota and NLRP3 inflammasome activation, and examines the mechanisms through which gut microbiota mediates the anti-inflammatory effects of exercise, providing new avenues for research.
Collapse
Affiliation(s)
- Jun Chen
- School of Graduate of Wuhan Sports University, Wuhan, China
| | - Shaohui Jia
- School of Sports Medicine of Wuhan Sports University, Wuhan, China
| | - Xinxuan Xue
- School of Sports Training of Wuhan Sports University, Wuhan, China
| | - Chenggeng Guo
- School of Sports Training of Wuhan Sports University, Wuhan, China
| | - Kunwei Dong
- School of Art of Wuhan Sports University, Wuhan, China
| |
Collapse
|
47
|
Wang W, Asiru, Luo G, Chen Y, Cui Y, Ping S, Chen Y. A Novel Effect of Id2 in Microglia TNFα Regulation. Mol Neurobiol 2025; 62:304-321. [PMID: 38850351 DOI: 10.1007/s12035-024-04278-2] [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: 03/07/2024] [Accepted: 06/01/2024] [Indexed: 06/10/2024]
Abstract
Microglia are the most important immune cells in the central nervous system (CNS), which can defend against external pathogens and stimuli. Dysregulation of microglia releases excessive proinflammatory cytokines and leads to neuroinflammation, which is fundamental to the pathophysiology of multiple neurological diseases. However, the molecular mechanisms underlying the regulation of proinflammatory cytokines in microglia are still not well-understood. Here, we identified that inhibitor of DNA binding protein 2 (Id2) was a negative regulator of tumor necrosis factor-α (TNFα) in cultured microglia. Knockdown of Id2 significantly increased the expression of TNFα in microglia, while overexpression of Id2 inhibited TNFα expression. Furthermore, by interacting with the p65 subunit of nuclear factor kappa-B (NF-κB), Id2 suppressed the transcription activation of NF-κB and inhibited TNFα expression. Interestingly, in lipopolysaccharides (LPS)-treated microglia, Id2 increased and underwent a cytoplasmic relocation. Immunoprecipitation and immunostaining results showed that by binding to the LIM domain of Id2, a scaffold protein PDZ and LIM 5 (PDLIM5) involved in the Id2 cytoplasmic relocation, which inactivated Id2 and resulted in higher TNFα expression in LPS-treated microglia. Collectively, our data delineate a novel effect of Id2 on TNFα regulation in microglia, which may shed a light on the proinflammatory cytokines regulating in microglia associated neuroimmune disorders.
Collapse
Affiliation(s)
- Wenhui Wang
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Asiru
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Guoya Luo
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Yanmei Chen
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Yu Cui
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Suning Ping
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China.
- Department of Histology and Embryology, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China.
| | - Yuan Chen
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China.
| |
Collapse
|
48
|
Zhang L, Zhu W, Zhang ZP, Huang Y. Alleviating Acute Lung Injury Induced by Lipopolysaccharide: Dayuan Yin Suppresses Inflammation and Oxidative Stress in Elderly Male Rats. Comb Chem High Throughput Screen 2025; 28:840-852. [PMID: 38920065 DOI: 10.2174/0113862073294620240527102409] [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: 12/22/2023] [Revised: 04/07/2024] [Accepted: 04/17/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND The occurrence of acute lung injury (ALI) caused by lipopolysaccharide (LPS) is prevalent and perilous among older individuals. Inflammation and oxidative stress are vital factors in the progression of ALI in this population. Dayuan Yin (DYY) is a classic Chinese herbal formula used for treating pulmonary diseases. Therefore.this situation can be well simulated by selecting suitable aged rats and induced by LPS, which is helpful to evaluate the role of DYY. OBJECTIVE The objective of this study is to assess the therapeutic efficacy of DYY in reducing pulmonary inflammation and oxidative stress injury in aged rats induced by LPS. METHODS In elderly male Sprague Dawley (SD) rats, the ALI model was induced by injecting LPS into the peritoneal cavity. The therapeutic effect of the DYY group was evaluated after 3 days of oral administration. Lung tissue damage was assessed using hematoxylin-eosin staining and the lung wet/dry (W/D) ratio. Inflammatory reaction in lung tissue was analyzed by counting inflammatory agents, measuring total protein (TP), and examining the concentration of inflammatory components in bronchoalveolar lavage fluid (BALF). Lung oxidative stress was assessed by measuring malondialdehyde (MDA), inducible nitric oxide synthase (iNOS), and superoxide dismutase (SOD) levels in BALF. The impact of DYY on the phosphorylation of PI3K, AKT, and NF-κBp65 protein was analyzed using Western Blot (WB). RESULTS The administration of DYY exhibited a dose-dependent reduction in the severity of lung injury caused by LPS, leading to a reversal of the LPS-induced lung W/D ratio. Furthermore, DYY treatment resulted in decreased levels of leukocytes, eosinophils, neutrophils, macrophages, lymphocytes, and total protein in BALF. Additionally, DYY significantly inhibited the upregulation of Interleukin -6, Interleukin -10, and Interleukin -1β (IL-6, IL-10, IL-1β) as well as Tumor necrosis factor-α(TNF-α) induced by LPS (P<0.01). The lungs experienced oxidative stress due to LPS, leading to the production of MDA and iNOS, as well as a decrease in SOD activity. DYY reduced oxidative stress in the lungs and inhibited the activation of p-PI3K, p-Akt, and p-NF-κBp65, with a greater effect at higher doses. CONCLUSION In a dose-dependent manner, DYY suppresses the inflammatory response and oxidative stress in the lung tissue of elderly rats, thereby reducing ALI caused by LPS. This effect may be attributed to the inhibition of the PI3K/AKT/NF-κB pathway activation.
Collapse
Affiliation(s)
- Lei Zhang
- Kunshan Hospital of Traditional Chinese Medicine, Kunshan, 215300, China
| | - Wei Zhu
- Kunshan Hospital of Traditional Chinese Medicine, Kunshan, 215300, China
| | - Ze-Peng Zhang
- Kunshan Hospital of Traditional Chinese Medicine, Kunshan, 215300, China
| | - Yu Huang
- Kunshan Hospital of Traditional Chinese Medicine, Kunshan, 215300, China
| |
Collapse
|
49
|
Zhao T, Zhou ZR, Wan HQ, Feng T, Hu XH, Li XQ, Zhao SM, Li HL, Hou JW, Li W, Lu DY, Qian MY, Shen X. Otilonium bromide ameliorates pulmonary fibrosis in mice through activating phosphatase PPM1A. Acta Pharmacol Sin 2025; 46:107-121. [PMID: 39160244 PMCID: PMC11695943 DOI: 10.1038/s41401-024-01368-8] [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: 03/19/2024] [Accepted: 07/24/2024] [Indexed: 08/21/2024]
Abstract
Pulmonary fibrosis (PF) is a chronic, progressive and irreversible interstitial lung disease characterized by unremitting pulmonary myofibroblasts activation, extracellular matrix (ECM) deposition and inflammatory recruitment. PF has no curable medication yet. In this study we investigated the molecular pathogenesis and potential therapeutic targets of PF and discovered drug lead compounds for PF therapy. A murine PF model was established in mice by intratracheal instillation of bleomycin (BLM, 5 mg/kg). We showed that the protein level of pulmonary protein phosphatase magnesium-dependent 1A (PPM1A, also known as PP2Cα) was significantly downregulated in PF patients and BLM-induced PF mice. We demonstrated that TRIM47 promoted ubiquitination and decreased PPM1A protein in PF progression. By screening the lab in-house compound library, we discovered otilonium bromide (OB, clinically used for treating irritable bowel syndrome) as a PPM1A enzymatic activator with an EC50 value of 4.23 μM. Treatment with OB (2.5, 5 mg·kg-1·d-1, i.p., for 20 days) significantly ameliorated PF-like pathology in mice. We constructed PF mice with PPM1A-specific knockdown in the lung tissues, and determined that by targeting PPM1A, OB treatment suppressed ECM deposition through TGF-β/SMAD3 pathway in fibroblasts, repressed inflammatory responses through NF-κB/NLRP3 pathway in alveolar epithelial cells, and blunted the crosstalk between inflammation in alveolar epithelial cells and ECM deposition in fibroblasts. Together, our results demonstrate that pulmonary PPM1A activation is a promising therapeutic strategy for PF and highlighted the potential of OB in the treatment of the disease.
Collapse
Affiliation(s)
- Tong Zhao
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhi-Ruo Zhou
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Hui-Qi Wan
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tian Feng
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xu-Hui Hu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiao-Qian Li
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shi-Mei Zhao
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hong-Lin Li
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ji-Wei Hou
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, 210093, China
| | - Wei Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Da-Yun Lu
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Min-Yi Qian
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xu Shen
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
50
|
Luetragoon T, Daowtak K, Thongsri Y, Potup P, Calder PC, Usuwanthim K. Anti-Inflammatory Potential of 3-Hydroxy-β-Ionone from Moringa oleifera: Decreased Transendothelial Migration of Monocytes Through an Inflamed Human Endothelial Cell Monolayer by Inhibiting the IκB-α/NF-κB Signaling Pathway. Molecules 2024; 29:5873. [PMID: 39769962 PMCID: PMC11678794 DOI: 10.3390/molecules29245873] [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: 10/25/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Moringa leaves provide numerous health benefits due to their anti-inflammatory properties. This study presents the first evidence that endothelial cell inflammation can potentially be ameliorated by moringa leaf extract. Here, we established an experimental human blood vessel cell model of inflammation using EA.hy926 cells. TNF-α was added after pre-treating the cells with crude leaf extract from Moringa oleifera Lam., a constituent fraction of the extract, and the bioactive component 3-hydroxy-β-ionone. The extract and the active ingredient significantly decreased the levels of pro-inflammatory mediators such as IL-6, IL-8, and MCP-1; decreased IκB-α and NF-κB p65 phosphorylation; and decreased the expression of VCAM-1, PECAM-1, and ICAM-1, three significant adhesion molecules. Furthermore, they attenuated THP-1 monocyte adhesion to the EA.hy926 monolayer and decreased monocyte transmigration across the monolayer. These findings suggest that 3-hydroxy-β-ionone and moringa leaf extract have anti-inflammatory properties and can be used as therapeutic agents to reduce the progression of diseases involving the inflamed endothelium by decreasing the production of inflammatory cytokines, chemokines, and adhesion molecules. This is promising for conditions such as atherosclerosis and neuroinflammation.
Collapse
Affiliation(s)
- Thitiya Luetragoon
- Department of Medical Technology, Faculty of Allied Health Sciences, Nakhon Ratchasima College, Nakhon Ratchasima 30000, Thailand;
- Cellular and Molecular Immunology Research Unit, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand; (K.D.); (Y.T.); (P.P.)
| | - Krai Daowtak
- Cellular and Molecular Immunology Research Unit, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand; (K.D.); (Y.T.); (P.P.)
| | - Yordhathai Thongsri
- Cellular and Molecular Immunology Research Unit, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand; (K.D.); (Y.T.); (P.P.)
| | - Pachuen Potup
- Cellular and Molecular Immunology Research Unit, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand; (K.D.); (Y.T.); (P.P.)
| | - Philip C. Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK;
| | - Kanchana Usuwanthim
- Cellular and Molecular Immunology Research Unit, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand; (K.D.); (Y.T.); (P.P.)
| |
Collapse
|