1
|
Mishra Y, Kumar A, Kaundal RK. Mitochondrial Dysfunction is a Crucial Immune Checkpoint for Neuroinflammation and Neurodegeneration: mtDAMPs in Focus. Mol Neurobiol 2025; 62:6715-6747. [PMID: 39115673 DOI: 10.1007/s12035-024-04412-0] [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: 07/14/2024] [Accepted: 07/30/2024] [Indexed: 01/03/2025]
Abstract
Neuroinflammation is a pivotal factor in the progression of both age-related and acute neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, and stroke. Mitochondria, essential for neuronal health due to their roles in energy production, calcium buffering, and oxidative stress regulation, become increasingly susceptible to dysfunction under conditions of metabolic stress, aging, or injury. Impaired mitophagy in aged or injured neurons leads to the accumulation of dysfunctional mitochondria, which release mitochondrial-derived damage-associated molecular patterns (mtDAMPs). These mtDAMPs act as immune checkpoints, activating pattern recognition receptors (PRRs) and triggering innate immune signaling pathways. This activation initiates inflammatory responses in neurons and brain-resident immune cells, releasing cytokines and chemokines that damage adjacent healthy neurons and recruit peripheral immune cells, further amplifying neuroinflammation and neurodegeneration. Long-term mitochondrial dysfunction perpetuates a chronic inflammatory state, exacerbating neuronal injury and contributing additional immunogenic components to the extracellular environment. Emerging evidence highlights the critical role of mtDAMPs in initiating and sustaining neuroinflammation, with circulating levels of these molecules potentially serving as biomarkers for disease progression. This review explores the mechanisms of mtDAMP release due to mitochondrial dysfunction, their interaction with PRRs, and the subsequent activation of inflammatory pathways. We also discuss the role of mtDAMP-triggered innate immune responses in exacerbating both acute and chronic neuroinflammation and neurodegeneration. Targeting dysfunctional mitochondria and mtDAMPs with pharmacological agents presents a promising strategy for mitigating the initiation and progression of neuropathological conditions.
Collapse
Affiliation(s)
- Yogesh Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - SAS Nagar, SAS Nagar, Punjab, India
| | - Ashutosh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - SAS Nagar, SAS Nagar, Punjab, India.
| | - Ravinder Kumar Kaundal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow, Uttar Pradesh, India.
| |
Collapse
|
2
|
Zhang H, Jiang N, Xu M, Jing D, Dong T, Liu Q, Lv Q, Huo R, Chen P, Li L, Wang X. M2 macrophage derived exosomal miR-20a-5p ameliorates trophoblast pyroptosis and placental injuries in obstetric antiphospholipid syndrome via the TXNIP/NLRP3 axis. Life Sci 2025; 370:123561. [PMID: 40127859 DOI: 10.1016/j.lfs.2025.123561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025]
Abstract
AIM Obstetric antiphospholipid syndrome (OAPS) is a pregnancy-related complication characterized by trophoblast pyroptosis and placental injury induced by antiphospholipid antibodies (aPLs). M2-polarized macrophage-derived exosomes (M2-exos) exert anti-inflammatory, immunomodulatory, and growth-promoting effects in various autoimmune diseases and tumors. However, their role in OAPS is not yet clear. Therefore, in this study, we isolated M2-exos from M2 macrophages and investigated their effects on trophoblast proliferation, death, migration, invasion, and pyroptosis following stimulation using aPLs. MAIN METHODS First, we established an animal model of OAPS and thereafter treated the OAPS mice with exogenous M2-exos via injection through the tail vein. Then to clarify the roles of miR-20a-5p and thioredoxin-interacting protein (TXNIP) in OAPS, we performed gain- or loss-of-function assays, and used GraphPad Prism software to analyze the collected data with statistical significance set at P < 0.05. KEY FINDINGS MicroRNAs (miRNAs) sequencing revealed the enrichment of miR-20a-5p in M2-exos, and these M2-exos significantly alleviated aPLs-induced trophoblast dysfunction. Our results also indicated that M2-exos delivered miR-20a-5p to trophoblast cells directly targeted thioredoxin-interacting protein (TXNIP), and thus suppressed the TXNIP/NLRP3 pathway, reduced pyroptosis and inflammation in trophoblast cells, and improved placental function and fetal development. SIGNIFICANCE M2-exos improve pregnancy outcomes in OAPS via the miR-20a-5p/TXNIP/NLRP3 axis, and thus represent as a novel therapeutic approach for aPLs-induced OAPS.
Collapse
Affiliation(s)
- Hongyuan Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China; The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan 250117, Shandong, China
| | - Ning Jiang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Mingyang Xu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Die Jing
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Tingting Dong
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Qian Liu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Feixian County People's Hospital, Linyi 273400, Shandong, China
| | - Qingfeng Lv
- The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, Shandong, China
| | - Ruiheng Huo
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Pengzheng Chen
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China.
| | - Lei Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China; The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan 250117, Shandong, China.
| | - Xietong Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China; The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan 250117, Shandong, China.
| |
Collapse
|
3
|
Zhang H, Liang H, Fan L, Zhu X, Ji P, Su Y, Li W, Li W. Ginsenoside Rg1 attenuates T2DM-induced renal damage and fibrosis by inhibiting TRPC6-ChREBP-TXNIP signaling. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119863. [PMID: 40311716 DOI: 10.1016/j.jep.2025.119863] [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: 02/25/2025] [Revised: 04/12/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As a traditional Chinese medicine, ginseng has many benefits, including regulating blood sugar, blood pressure and so on. Ginsenoside Rg1 is the main active component of ginseng and has been found to significantly improve renal pathological injury in type 2 diabetes mellitus (T2DM) mice. However, the effects and mechanisms of Rg1 in attenuating T2DM are not fully understood. AIM OF THE STUDY This study aims to investigate the role of Rg1 in the treatment of renal damage and fibrosis induced by T2DM and its molecular mechanism. MATERIALS AND METHODS T2DM models were constructed on mice and cells respectively and were administered with corresponding drugs. SA-β-Gal and Oil Red O were used to observe cell senescence and lipid droplet deposition; H&E and PAS were used to observe pathological changes in the kidney; masson and sirius red were used to evaluate the level of renal fibrosis. Immunohistochemistry, immunofluorescence and Western blotting were performed to analyze the relevant indexes which resulted in the detection of ROS levels in vitro and in vivo. Calcium imaging was used to test the level of [Ca2+]i. RESULTS Rg1 and Trpc6 knockout could significantly improve kidney dysfunction, attenuate renal injury and fibrosis and also decrease the expression levels of TRPC6, CaN, TXNIP, ChREBP, p-ASK1 and NLRP3 inflammasome. Meanwhile, Rg1 and Trpc6 knockout significantly inhibited mitochondrial damage and apoptosis protein release. Additionally, Rg1 treatment has been shown to markedly reduce lipid deposition and ROS accumulation in T2DM, while Trpc6 knockout exhibited no effect on these parameters. CONCLUSION Rg1 treatment can inhibit the TRPC6-ChREBP-TXNIP pathway, thereby improving chronic T2DM-induced renal injury and fibrosis.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Haoyu Liang
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Lei Fan
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Xing Zhu
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Pengmin Ji
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Yong Su
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Weiping Li
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Weizu Li
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
4
|
Yoo TT, Baek IH, Stoletniy L, Hilliard A, Sakr A, Doycheva D. Impact of sodium-glucose transport protein-2 (SGLT2) inhibitors on the inflammasome pathway in acute myocardial infarction in type 2 diabetes mellitus: a comprehensive review. Cardiovasc Diabetol 2025; 24:227. [PMID: 40420176 PMCID: PMC12105141 DOI: 10.1186/s12933-025-02777-7] [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: 03/06/2025] [Accepted: 05/06/2025] [Indexed: 05/28/2025] Open
Abstract
Sodium-glucose transport protein-2 (SGLT2) inhibitors, initially developed for glycemic control in type 2 diabetes mellitus (T2DM), have emerged as potential cardioprotective agents, reducing cardiovascular mortality and improving heart failure outcomes. Recent evidence suggests that SGLT2 inhibitors exert anti-inflammatory effects, particularly through modulating the inflammasome pathway. This review explores the role of the inflammasome in acute myocardial infarction (AMI) in T2DM and discusses the mechanisms by which SGLT2 inhibitors influence this pathway. We evaluate current studies on the impact of SGLT2 inhibitors on key inflammatory mediators, particularly the NLRP3 inflammasome, and discuss their potential therapeutic implications for reducing inflammation and myocardial injury in patients with T2DM experiencing AMI. In summary, the key novelties in this review lie in its focused mechanistic approach on the inflammasome pathway, its integration of diabetes and cardiovascular research, and its potential to influence future therapeutic strategies for AMI in T2DM patients. It offers a novel angle by tying together molecular mechanisms of inflammation with clinical implications in a specific patient population that faces high cardiovascular risk.
Collapse
Affiliation(s)
- Thomas T Yoo
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, CA, 92354, USA
| | - In Hae Baek
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, CA, 92354, USA
| | - Liset Stoletniy
- Division of Cardiology, School of Medicine, Loma Linda University, 11234 Anderson St, Loma Linda, CA, 92354, USA
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, CA, 92354, USA
| | - Anthony Hilliard
- Division of Cardiology, School of Medicine, Loma Linda University, 11234 Anderson St, Loma Linda, CA, 92354, USA
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, CA, 92354, USA
| | - Antoine Sakr
- Division of Cardiology, School of Medicine, Loma Linda University, 11234 Anderson St, Loma Linda, CA, 92354, USA
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, CA, 92354, USA
| | - Desislava Doycheva
- Division of Cardiology, School of Medicine, Loma Linda University, 11234 Anderson St, Loma Linda, CA, 92354, USA.
- Department of Physiology and Pharmacology, Loma Linda University, 11175 Campus St, Loma Linda, CA, 92354, USA.
| |
Collapse
|
5
|
Shen J, Ma X. Inhibition of the Foxo3/Txnip Axis Alleviates Ventilator-Induced Diaphragmatic Dysfunction by Downregulating MuRF1. Appl Biochem Biotechnol 2025:10.1007/s12010-025-05261-w. [PMID: 40377847 DOI: 10.1007/s12010-025-05261-w] [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] [Accepted: 05/02/2025] [Indexed: 05/18/2025]
Abstract
Ventilator-induced diaphragm dysfunction (VIDD) is one of the main causes of weaning from mechanical ventilation (MV). The forkhead box O3 (Foxo3) has been identified as being involved in regulating the contractile function of skeletal muscle. This study aimed to figure out the regulatory role and mechanism of Foxo3 on VIDD. The mouse myoblast C2C12 cells were stimulated using different intensities of stress to mimic the in-vitro VIDD model. 3- (4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and TdT-mediated dUTP nick end labeling (TUNEL) assays were applied to check cell viability and apoptosis, respectively. Cellular inflammation and oxidative stress levels were evaluated by measuring cellular inflammatory factors (IL-1β and TNF-α) and oxidative stress markers (SOD and MDA). The release of oxygen species (ROS) was assayed by cellular immunofluorescence. The expression of apoptosis-associated proteins (Bax and Bcl-2), Gpx4, Slc7a11, Ptgs2, Foxo3, Txnip, Murf1, Atrogin-1, Nlrp3, Asc, and Caspase1 was gauged using Western blot. The rats with or without MV therapy were treated with the Foxo3 inhibitor Carbenoxolone (CBX) to characterize the impact of Foxo3 on VIDD. Stress stimulation dampened myogenic cell viability, boosted apoptosis, inflammation, oxidative stress, and ROS release, and activated the expression of Foxo3 and Txnip pathways. Overexpression of Txnip or Murf1 lessened the protective effect of FOxO3 inhibition on myoblasts. Downregulation of Txnip or Murf1 mitigated myoblasts dysfunction that was induced by Foxo3 overexpression. In vivo, inhibition of Foxo3 mitigated MV-induced diaphragmatic atrophy and reduced contractility, inflammation, and oxidative stress in rats. Inhibition of Foxo3 eased VIDD by downregulating Txnip and Murf1.
Collapse
Affiliation(s)
- Jia Shen
- Department of Intensive Care Unit, General Hospital of Ningxia Medical University, No. 804, Shengli South Street, Xingqing District, Yinchuan, 750002, China.
| | - Xiaojun Ma
- Department of Orthopedics, People's Hospital of Ningxia Hui Autonomous Region, Jinfeng District, Yinchuan, 750004, China
| |
Collapse
|
6
|
Liang H, Yang S, Huang Y, Zhu Y, Wu Q, Wu Z, Li S, Shi Y, Chen Z, Jin H, Wang X. PTPN22 as a therapeutic target in intervertebral disc degeneration: Modulating mitophagy and pyroptosis through the PI3K/AKT/mTOR axis. J Adv Res 2025:S2090-1232(25)00311-X. [PMID: 40349959 DOI: 10.1016/j.jare.2025.05.017] [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: 01/25/2025] [Revised: 04/20/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025] Open
Abstract
INTRODUCTION Intervertebral disc degeneration (IDD) is a predominant risk factor for low back pain (LBP). However, the mechanisms underlying IDD progression remain unclear. OBJECTIVES The protein tyrosine phosphatase non-receptor type 22 (PTPN22) is associated with various chronic inflammatory and autoimmune conditions. However, its role in the progression of IDD remains obscure. This investigation delves into the function of PTPN22 within IDD and examines its molecular mechanisms. METHODS The expression levels of PTPN22 in human and rat degenerative nucleus pulposus (NP) cells were analyzed using Western blot and immunohistochemistry. Following PTPN22 knockdown via lentiviral transfection, pyroptosis, extracellular matrix (ECM) degradation, mitophagy, and mitochondrial function were assessed using Western blot, immunofluorescence, Calcein-AM/PI staining, qPCR, Seahorse, JC-1, and MitoSOX assays. The roles of autophagy and the PI3K/AKT/mTOR pathway were further investigated using the autophagy inhibitor 3-MA, Baf-A1, and the PI3K agonist 740Y-P. A puncture-induced rat model was established, and the effects of LV-shPTPN22 on IDD were evaluated through imaging and histological analyses. RESULTS We noted an upregulation of PTPN22 in degenerative NP cells. A deficiency in PTPN22 was found to enhance mitophagy, thereby alleviating hydrogen peroxide (H2O2)-induced mitochondrial dysfunction and consequently mitigating NP cell pyroptosis and ECM degradation. Inhibition of the PI3K/AKT/mTOR pathway appears to play a pivotal role in the protective effects of PTPN22 deficiency against IDD. Experiments conducted in vivo revealed that PTPN22 knockdown significantly curtails the progression of IDD. CONCLUSION In summary, PTPN22 knockdown alleviates IDD progression by reducing pyroptosis and ECM degradation through enhanced mitophagy. This highlights PTPN22 as a critical contributor to IDD and a promising therapeutic target.
Collapse
Affiliation(s)
- Haibo Liang
- Division of Spine Surgery, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shu Yang
- Division of Spine Surgery, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yeheng Huang
- Division of Spine Surgery, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yuxuan Zhu
- Division of Spine Surgery, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qihang Wu
- Division of Spine Surgery, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zhouwei Wu
- Division of Spine Surgery, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Sunlong Li
- Division of Spine Surgery, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yifeng Shi
- Division of Spine Surgery, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zhenya Chen
- School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Haiming Jin
- Division of Spine Surgery, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Xiangyang Wang
- Division of Spine Surgery, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
7
|
Trambas IA, Bowen L, Thallas-Bonke V, Snelson M, Sourris KC, Laskowski A, Tauc M, Rubera I, Zheng G, Harris DCH, Kantharidis P, Shimizu T, Cooper ME, Tan SM, Coughlan MT. Proximal tubular deletion of superoxide dismutase-2 reveals disparate effects on kidney function in diabetes. Redox Biol 2025; 82:103601. [PMID: 40127616 PMCID: PMC11979990 DOI: 10.1016/j.redox.2025.103601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/09/2025] [Accepted: 03/17/2025] [Indexed: 03/26/2025] Open
Abstract
There is a large body of evidence implicating mitochondrial reactive oxygen species (ROS) overproduction and oxidative stress in the development of diabetic kidney disease and the deficiency of mitochondrial antioxidant systems in the kidney, such as manganese superoxide dismutase (MnSOD/SOD2) have been identified. The proximal tubules of the kidney are densely packed with mitochondria thereby providing energy via oxidative phosphorylation in order to drive active transport for proximal tubular reabsorption of solutes from the glomerular filtrate. We hypothesized that maintenance of MnSOD function in the proximal tubules would be critical to maintain kidney health in diabetes. Here, we induced targeted deletion of SOD2 in the proximal tubules of the kidney in Ins2Akita diabetic mice (SODptKO mice) and show that 20 weeks of SOD2 deletion leads to no major impairment of kidney function and structure, despite these mice displaying enhanced albuminuria and kidney lipid peroxidation (8-isoprostanes). Plasma cystatin C, which is a surrogate marker of glomerular filtration was not altered in SODptKO diabetic mice and histological assessment of the kidney cortex revealed no change in kidney fibrosis. Thus, our findings suggest that deletion of SOD2 in the proximal tubular compartment of the kidney induces a more subtle phenotype than expected, shedding light on the involvement of SOD2 and the proximal tubular compartment in the pathogenesis of diabetic kidney disease.
Collapse
Affiliation(s)
- Inez A Trambas
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Lilliana Bowen
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Vicki Thallas-Bonke
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Matthew Snelson
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Karly C Sourris
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Adrienne Laskowski
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Michel Tauc
- Laboratoire de Physiomédecine Moléculaire, Université Côte D'Azur, CNRS, LP2M, 7370, Nice Cedex 2, France
| | - Isabelle Rubera
- Laboratoire de Physiomédecine Moléculaire, Université Côte D'Azur, CNRS, LP2M, 7370, Nice Cedex 2, France
| | - Guoping Zheng
- Centre for Transplantation and Renal Research, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, 2145, Australia
| | - David C H Harris
- Centre for Transplantation and Renal Research, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, 2145, Australia
| | - Phillip Kantharidis
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Takahiko Shimizu
- Department of Food and Reproductive Function Advanced Research, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Mark E Cooper
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Sih Min Tan
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Melinda T Coughlan
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia; Baker Heart and Diabetes Institute, Melbourne, 3004, Victoria, Australia; Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University Parkville Campus, 381 Royal Parade, Parkville, 3052, Victoria, Australia.
| |
Collapse
|
8
|
Sun DL, Guo ZY, Liu WY, Zhang L, Zhang ZY, Hu YL, Li SF, Zhang MY, Zhang G, Wang JJ, Fang JA. Astragaloside IV Alleviates Podocyte Injury in Diabetic Nephropathy through Regulating IRE-1α/NF-κ B/NLRP3 Pathway. Chin J Integr Med 2025; 31:422-433. [PMID: 39039342 DOI: 10.1007/s11655-024-3568-0] [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] [Accepted: 10/25/2023] [Indexed: 07/24/2024]
Abstract
OBJECTIVE To investigate the effects of astragaloside IV (AS-IV) on podocyte injury of diabetic nephropathy (DN) and reveal its potential mechanism. METHODS In in vitro experiment, podocytes were divided into 4 groups, normal, high glucose (HG), inositol-requiring enzyme 1 (IRE-1) α activator (HG+thapsigargin 1 µmol/L), and IRE-1α inhibitor (HG+STF-083010, 20 µmol/L) groups. Additionally, podocytes were divided into 4 groups, including normal, HG, AS-IV (HG+AS-IV 20 µmol/L), and IRE-1α inhibitor (HG+STF-083010, 20 µmol/L) groups, respectively. After 24 h treatment, the morphology of podocytes and endoplasmic reticulum (ER) was observed by electron microscopy. The expressions of glucose-regulated protein 78 (GRP78) and IRE-1α were detected by cellular immunofluorescence. In in vivo experiment, DN rat model was established via a consecutive 3-day intraperitoneal streptozotocin (STZ) injections. A total of 40 rats were assigned into the normal, DN, AS-IV [AS-IV 40 mg/(kg·d)], and IRE-1α inhibitor [STF-083010, 10 mg/(kg·d)] groups (n=10), respectively. The general condition, 24-h urine volume, random blood glucose, urinary protein excretion rate (UAER), urea nitrogen (BUN), and serum creatinine (SCr) levels of rats were measured after 8 weeks of intervention. Pathological changes in the renal tissue were observed by hematoxylin and eosin (HE) staining. Quantitative reverse transcription-polymerase chain reaction (RT-PCR) and Western blot were used to detect the expressions of GRP78, IRE-1α, nuclear factor kappa Bp65 (NF-κBp65), interleukin (IL)-1β, NLR family pyrin domain containing 3 (NLRP3), caspase-1, gasdermin D-N (GSDMD-N), and nephrin at the mRNA and protein levels in vivo and in vitro, respectively. RESULTS Cytoplasmic vacuolation and ER swelling were observed in the HG and IRE-1α activator groups. Podocyte morphology and ER expansion were improved in AS-IV and IRE-1α inhibitor groups compared with HG group. Cellular immunofluorescence showed that compared with the normal group, the fluorescence intensity of GRP78 and IRE-1α in the HG and IRE-1α activator groups were significantly increased whereas decreased in AS-IV and IRE-1α inhibitor groups (P<0.05). Compared with the normal group, the mRNA and protein expressions of GRP78, IRE-1α, NF-κ Bp65, IL-1β, NLRP3, caspase-1 and GSDMD-N in the HG group was increased (P<0.05). Compared with HG group, the expression of above indices was decreased in the AS-IV and IRE-1α inhibitor groups, and the expression in the IRE-1α activator group was increased (P<0.05). The expression of nephrin was decreased in the HG group, and increased in AS-IV and IRE-1α inhibitor groups (P<0.05). The in vivo experiment results revealed that compared to the normal group, the levels of blood glucose, triglyceride, total cholesterol, BUN, blood creatinine and urinary protein in the DN group were higher (P<0.05). Compared with DN group, the above indices in AS-IV and IRE-1α inhibitor groups were decreased (P<0.05). HE staining revealed glomerular hypertrophy, mesangial widening and mesangial cell proliferation in the renal tissue of the DN group. Compared with the DN group, the above pathological changes in renal tissue of AS-IV and IRE-1α inhibitor groups were alleviated. Quantitative RT-PCR and Western blot results of GRP78, IRE-1α, NF-κ Bp65, IL-1β, NLRP3, caspase-1 and GSDMD-N were consistent with immunofluorescence analysis. CONCLUSION AS-IV could reduce ERS and inflammation, improve podocyte pyroptosis, thus exerting a podocyte-protective effect in DN, through regulating IRE-1α/NF-κ B/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Da-Lin Sun
- The First College for Clinical Medicine, Shanxi Medical University, Taiyuan, 030001, China
| | - Zi-Yi Guo
- The First College for Clinical Medicine, Shanxi Medical University, Taiyuan, 030001, China
| | - Wen-Yuan Liu
- Department of Nephrology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Lin Zhang
- The First College for Clinical Medicine, Shanxi Medical University, Taiyuan, 030001, China
| | - Zi-Yuan Zhang
- Department of Nephrology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Ya-Ling Hu
- Department of Nephrology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Su-Fen Li
- Department of Nephrology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Ming-Yu Zhang
- The First College for Clinical Medicine, Shanxi Medical University, Taiyuan, 030001, China
| | - Guang Zhang
- Department of Nephrology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Jin-Jing Wang
- The First College for Clinical Medicine, Shanxi Medical University, Taiyuan, 030001, China
| | - Jing-Ai Fang
- Department of Nephrology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
9
|
Chen X, Zhang J, Lu F, Hu R, Du X, Xu C, Liang M, Chen L, Yao W, Ma Z, Zhong J, Wang M. Association between uric acid to high-density lipoprotein cholesterol ratio and chronic kidney disease in Chinese patients with type 2 diabetes mellitus: a cross-sectional study. Front Nutr 2025; 12:1582495. [PMID: 40297336 PMCID: PMC12034545 DOI: 10.3389/fnut.2025.1582495] [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: 02/24/2025] [Accepted: 03/28/2025] [Indexed: 04/30/2025] Open
Abstract
Objectives To examine the association between uric acid (UA) to high-density lipoprotein cholesterol (HDL-C) ratio (UHR) and chronic kidney disease (CKD) in type 2 diabetes mellitus (T2DM) patients in China. Methods The investigation stems from a survey conducted in the eastern Chinese province of Zhejiang, spanning from March to November 2018. A multivariable logistic regression model was employed to assess the relationship between UHR and CKD, while restricted cubic spline (RCS) analysis was used to evaluate the dose-response relationship. Receiver operating characteristic (ROC) curve analysis was performed to determine the optimal UHR cut-off value and assess its diagnostic performance for CKD. Model performance was further evaluated using net reclassification improvement (NRI) and integrated discrimination improvement (IDI) metrics. Sensitivity analyses, including propensity score matching (PSM) and k-means clustering, were conducted to enhance the robustness of the findings. Subgroup analyses were performed across various demographic and clinical categories to examine the consistency of the UHR-CKD association. Results This cross-sectional study included 1,756 Chinese patients with T2DM, among whom 485 (27.62%) were identified with CKD. Multivariable logistic regression analysis revealed a significant positive association between UHR and CKD. Per standard deviation (SD) increase in UHR was associated with a 40% higher odds of CKD (OR = 1.40, 95% CI: 1.23-1.60) after adjusting for potential covariates. When analyzed categorically, participants in the highest UHR tertile (T3) had 1.82-fold higher odds of CKD compared to the lowest tertile (T1) (95% CI: 1.32-2.50). RCS analysis demonstrated a consistent linear dose-response relationship between UHR and CKD across all models (all p for nonlinearity >0.05). ROC curve analysis identified an optimal UHR cut-off value of 12.28 for CKD prediction, with an area under the curve (AUC) of 0.710 (95% CI: 0.683-0.737) in the fully adjusted model. Subgroup analyses confirmed the robustness of the UHR-CKD association across most demographic and clinical variables, except for younger age groups (18-44 and 45-59 years) and smokers. Notably, BMI significantly modified the UHR-CKD relationship, with a nonlinear association observed in individuals with lower BMI (<24 kg/m2) and a linear association in those with higher BMI (≥24 kg/m2). Conclusion This study demonstrates a significant dose-response relationship between the UHR and CKD in Chinese patients with T2DM, highlighting UHR as a promising biomarker for CKD risk assessment. The identified UHR cut-off of 12.28 offers a practical threshold for early renal monitoring and targeted interventions. Future research should explore UHR-targeted therapies and its integration into personalized risk stratification models to improve CKD management in T2DM.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Meng Wang
- Department of Non-Communicable Disease Control and Prevention, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| |
Collapse
|
10
|
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
|
11
|
Srivastava SP, Kopasz-Gemmen O, Thurman A, Rajendran BK, Selvam MM, Kumar S, Srivastava R, Suresh MX, Kumari R, Goodwin JE, Inoki K. The molecular determinants regulating redox signaling in diabetic endothelial cells. Front Pharmacol 2025; 16:1563047. [PMID: 40290438 PMCID: PMC12023289 DOI: 10.3389/fphar.2025.1563047] [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: 01/18/2025] [Accepted: 03/14/2025] [Indexed: 04/30/2025] Open
Abstract
Oxidation and reduction are vital for keeping life through several prime mechanisms, including respiration, metabolism, and other energy supplies. Mitochondria are considered the cell's powerhouse and use nutrients to produce redox potential and generate ATP and H2O through the process of oxidative phosphorylation by operating electron transfer and proton pumping. Simultaneously, mitochondria also produce oxygen free radicals, called superoxide (O2 -), non-enzymatically, which interacts with other moieties and generate reactive oxygen species (ROS), such as hydrogen peroxide (H2O2), peroxynitrite (ONOO-), and hydroxyl radical (OH-). These reactive oxygen species modify nucleic acids, proteins, and carbohydrates and ultimately cause damage to organs. The nutrient-sensing kinases, such as AMPK and mTOR, function as a key regulator of cellular ROS levels, as loss of AMPK or aberrant activation of mTOR signaling causes ROS production and compromises the cell's oxidant status, resulting in various cellular injuries. The increased ROS not only directly damages DNA, proteins, and lipids but also alters cellular signaling pathways, such as the activation of MAPK or PI3K, the accumulation of HIF-1α in the nucleus, and NFkB-mediated transcription of pro-inflammatory cytokines. These factors cause mesenchymal activation in renal endothelial cells. Here, we discuss the biology of redox signaling that underlies the pathophysiology of diabetic renal endothelial cells.
Collapse
Affiliation(s)
- Swayam Prakash Srivastava
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
- Vascular Biology and Therapeutic Program, Yale University School of Medicine, New Haven, CT, United States
| | | | - Aaron Thurman
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
| | - Barani Kumar Rajendran
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - M. Masilamani Selvam
- Department of Pharmaceutical Technology, Paavai Engineering College, Namakkal, Tamil Nadu, India
| | - Sandeep Kumar
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| | - Rohit Srivastava
- Laboratory of Medical Transcriptomics, Department of Endocrinology, Nephrology Services, Hadassah Hebrew-University Medical Center, Jerusalem, Israel
| | - M. Xavier Suresh
- School of Advanced Sciences and Languages, VIT Bhopal University, Sehore, Madhya Pradesh, India
| | - Reena Kumari
- Department of Physiology, Augusta University, Augusta, GA, United States
| | - Julie E. Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
- Vascular Biology and Therapeutic Program, Yale University School of Medicine, New Haven, CT, United States
| | - Ken Inoki
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
12
|
Gao H, Xie T, Li Y, Xu Z, Song Z, Yu H, Zhou H, Li W, Yun C, Guan B, Luan S, Yin L. Role of gasdermins in chronic kidney disease. Front Immunol 2025; 16:1557707. [PMID: 40236694 PMCID: PMC11996640 DOI: 10.3389/fimmu.2025.1557707] [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: 01/09/2025] [Accepted: 03/14/2025] [Indexed: 04/17/2025] Open
Abstract
Gasdermins (GSDMs), functioning as membrane perforating proteins, can be activated by canonical inflammasomes, noncanonical inflammasomes, as well as non-inflammasomes, leading to cell pyroptosis and the subsequent release of inflammatory mediators. Increasing evidence has implicated that GSDMs are associated with chronic kidney disease (CKD), including diabetes nephropathy, lupus nephritis, obstructive nephropathy, and crystalline nephropathy. This review centers on the role of GSDMs-mediated pyroptosis in the pathogenesis of CKD, providing novel ideas for enhancing the prognosis and therapeutic strategies of CKD.
Collapse
Affiliation(s)
- Hanchao Gao
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen, Guangdong, China
| | - Ting Xie
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yunyi Li
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zigan Xu
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen, Guangdong, China
| | - Zhuoheng Song
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen, Guangdong, China
| | - Huixia Yu
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Hongming Zhou
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen, Guangdong, China
| | - Weilong Li
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen, Guangdong, China
| | - Chen Yun
- Charité-Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Baozhang Guan
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Shaodong Luan
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen, Guangdong, China
| | - Lianghong Yin
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
13
|
Li JM, Song ZH, Li Y, Chen HW, Li H, Yuan L, Li J, Lv WY, Liu L, Wang N. NR4A1 silencing alleviates high-glucose-stimulated HK-2 cells pyroptosis and fibrosis via hindering NLRP3 activation and PI3K/AKT pathway. World J Diabetes 2025; 16:97544. [PMID: 40093286 PMCID: PMC11885978 DOI: 10.4239/wjd.v16.i3.97544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 10/15/2024] [Accepted: 12/16/2024] [Indexed: 01/21/2025] Open
Abstract
BACKGROUND The pathophysiology of diabetic kidney disease (DKD) is complex. Interfering with the processes of pyroptosis and fibrosis is an effective strategy for slowing DKD progression. Previous studies have revealed that nuclear receptor subfamily 4 group A member 1 (NR4A1) may serve as a novel pathogenic element in DKD; however, the specific mechanism by which it contributes to pyroptosis and fibrosis in DKD is unknown. AIM To investigate the role of NR4A1 in renal pyroptosis and fibrosis in DKD and possible molecular mechanisms. METHODS Streptozotocin 60 mg/kg was injected intraperitoneally to establish a rat model of DKD. Typically, 45 mmol/L glucose [high glucose (HG)] was used to activate HK-2 cells to mimic the DKD model in vitro. HK-2 cells were transfected with NR4A1 siRNA to silence NR4A1. RESULTS NR4A1 was elevated in renal tissues of DKD rats and HG-stimulated HK-2 cells. Concurrently, NOD-like receptor protein 3 (NLRP3) and phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathways were triggered, and pyroptosis and expression of fibrosis-linked elements was increased in vivo and in vitro. These alterations were significantly reversed via NR4A1 silencing. CONCLUSION Inhibition of NR4A1 mitigated pyroptosis and fibrosis via suppressing NLRP3 activation and the PI3K/AKT pathway in HG-activated HK-2 cells.
Collapse
Affiliation(s)
- Jin-Meng Li
- Department of Clinical Medicine, Jining Medical University, Jining 272013, Shandong Province, China
| | - Zi-Hua Song
- Department of General Medicine, Affiliated Hospital of Jining Medical University, Jining 272029, Shandong Province, China
| | - Yuan Li
- Department of General Medicine, Affiliated Hospital of Jining Medical University, Jining 272029, Shandong Province, China
| | - Han-Wen Chen
- Department of General Medicine, Affiliated Hospital of Jining Medical University, Jining 272029, Shandong Province, China
| | - Han Li
- Department of General Medicine, Affiliated Hospital of Jining Medical University, Jining 272029, Shandong Province, China
| | - Lu Yuan
- Department of Clinical Medicine, Jining Medical University, Jining 272013, Shandong Province, China
| | - Jing Li
- Department of Clinical Medicine, Jining Medical University, Jining 272013, Shandong Province, China
| | - Wen-Yue Lv
- Department of Clinical Medicine, Jining Medical University, Jining 272013, Shandong Province, China
| | - Lei Liu
- Department of General Medicine, Affiliated Hospital of Jining Medical University, Jining 272029, Shandong Province, China
| | - Na Wang
- Department of General Medicine, Affiliated Hospital of Jining Medical University, Jining 272029, Shandong Province, China
| |
Collapse
|
14
|
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
|
15
|
Liu X, Zhou D, Su Y, Liu H, Su Q, Shen T, Zhang M, Mi X, Zhang Y, Yue S, Zhang Z, Wang D, Tan X. PDIA4 targets IRE1α/sXBP1 to alleviate NLRP3 inflammasome activation and renal tubular injury in diabetic kidney disease. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167645. [PMID: 39743023 DOI: 10.1016/j.bbadis.2024.167645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
The role of ER stress in the pathogenesis of diabetic kidney diseases (DKD) remains unclear. We employed bioinformatics to identify the UPR pathway activation, inflammation, and programmed cell death patterns in diabetic tubules. Levels of IRE1α/sXBP1 signaling, NLRP3 inflammasome activity and pyroptosis in tubular cells under high glucose conditions were measured. IRE1α knockdown was used to determine its role in glucose-triggered activation of the NLRP3 inflammasome and pyroptosis. PDIA4 overexpression and silencing were used to assess its impact on the IRE1α/sXBP1 pathway. The dynamic interaction among PDIA4, GRP78, and IRE1α under high glucose were analyzed using immunoprecipitation and crosslinking assays. In STZ-induced and db/db mouse models of DKD, the regulatory role of PDIA4 on IRE1α/sXBP1 signaling and diabetic tubular inflammation and injury were evaluated. Our study showed that IRE1α/sXBP1, NLRP3 inflammasome, and pyroptosis are activated in the renal tubules of DKD patients. Induction of IRE1α pathway mediated the glucose-triggered activation of the NLRP3 inflammasome and pyroptosis. Moreover, overexpression of PDIA4 decreased the activation of IRE1α/sXBP1 under high glucose conditions. High glucose leads to the release of GRP78 from IRE1α and an increased interaction between IRE1α and PDIA4. In mouse models of DKD, overexpressing PDIA4 mitigated diabetic tubular injury and inflammation, marked by decreased IRE1α/sXBP1 and NLRP3 inflammasome. In conclusion, our findings demonstrate that high glucose triggers NLRP3 inflammasome and pyroptosis via the IRE1α/sXBP1 pathway in renal tubular cells. Overexpression of PDIA4 suppresses IRE1α signaling by binding to its oligomeric form, implying a promising therapeutic intervention for DKD.
Collapse
Affiliation(s)
- Xuan Liu
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Donghui Zhou
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Yu Su
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Hongling Liu
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Qiuyue Su
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Tianyu Shen
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Mianzhi Zhang
- Dongfang Hospital of Beijing University of Chinese medicine, Beijing, China
| | - Xue Mi
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Yuying Zhang
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Shijing Yue
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Zhujun Zhang
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Dekun Wang
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China.
| | - Xiaoyue Tan
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China.
| |
Collapse
|
16
|
Mo B, Ding Y, Ji Q. NLRP3 inflammasome in cardiovascular diseases: an update. Front Immunol 2025; 16:1550226. [PMID: 40079000 PMCID: PMC11896874 DOI: 10.3389/fimmu.2025.1550226] [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: 12/23/2024] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
Cardiovascular disease (CVD) continues to be the leading cause of mortality worldwide. The nucleotide oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasome is involved in numerous types of CVD. As part of innate immunity, the NLRP3 inflammasome plays a vital role, requiring priming and activation signals to trigger inflammation. The NLRP3 inflammasome leads both to the release of IL-1 family cytokines and to a distinct form of programmed cell death called pyroptosis. Inflammation related to CVD has been extensively investigated in relation to the NLRP3 inflammasome. In this review, we describe the pathways triggering NLRP3 priming and activation and discuss its pathogenic effects on CVD. This study also provides an overview of potential therapeutic approaches targeting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Binhai Mo
- People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yudi Ding
- First People’s Hospital of Nanning, Nanning, Guangxi, China
| | - Qingwei Ji
- People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
17
|
Xiang J, Lv M, Luo Y, Ke K, Zhang B, Wang M, Zhang K, Li H. Mechanistic studies of Ca 2+-induced classical pyroptosis pathway promoting renal adhesion on calcium oxalate kidney stone formation. Sci Rep 2025; 15:6669. [PMID: 39994305 PMCID: PMC11850917 DOI: 10.1038/s41598-025-91460-8] [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: 05/16/2024] [Accepted: 02/20/2025] [Indexed: 02/26/2025] Open
Abstract
This study aims to investigate the role of hypercalciuria and pyroptosis in the formation of calcium oxalate kidney stones. Bioinformatics analysis was performed to compare the correlation of pyroptosis scores and cell adhesion scores between Randall's plaques and normal tissues from kidney stone patients. For the in vitro experiments, we investigated the effects of high concentrations of Ca2+ on the pyroptosis and adhesion levels of renal tubular epithelial cells and examined the adhesion levels and crystal aggregation of the cells in high Ca2+ concentrations environment by knockdown and overexpression of the key pyroptosis gene, GSDMD, and we verified the effects of Ca2+ concentration on pyroptosis and adhesion levels, kidney injury, and crystal deposition by in vivo experiments. Bioinformatic results showed that the scores of pyroptosis and cell adhesion in Randall's plaques of patients with kidney stones were significantly higher than those in normal tissues, and pyroptosis was highly positively correlated with cell adhesion. In vitro and in vivo experiments showed that high concentrations of Ca2+ activated the NLRP3/Caspase-1/GSDMD pathway of pyroptosis through ROS and up-regulated the expression of adhesion-related proteins, and GSDMD could regulate the adhesion level of renal tubular epithelial cells by mediating the level of pyroptosis, thereby affecting the adhesion and deposition of calcium oxalate crystals. Our findings reveal that the Ca2+-induced classical pyroptosis pathway may be a potential mechanism to promote calcium oxalate kidney stone formation, which provides new insights into the etiology of kidney stones.
Collapse
Affiliation(s)
- Jinjie Xiang
- Department of Urology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Maoxin Lv
- Department of Urology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Yuhui Luo
- Department of Urology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Kunbin Ke
- Department of Urology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Baiyu Zhang
- Department of Urology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Mengyue Wang
- Department of Urology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Kun Zhang
- Department of Urology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Hao Li
- Department of Urology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| |
Collapse
|
18
|
Yang S, Guan Y, Zheng C, Xia X, Ma X, Jiang J. FOXO3-induced microRNA-128-3p promotes the progression of spinal cord injury in mice via regulating NLRP3 inflammasome-mediated pyroptosis. Front Immunol 2025; 16:1526721. [PMID: 40061945 PMCID: PMC11885150 DOI: 10.3389/fimmu.2025.1526721] [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: 11/12/2024] [Accepted: 02/03/2025] [Indexed: 05/13/2025] Open
Abstract
Background Spinal cord injury (SCI) remains a severe condition with an extremely high disability rate and complex pathophysiologic mechanisms. Pyroptosis, an inflammatory form of cell death triggered by certain inflammasomes, has a key role in a variety of inflammatory diseases, including SCI. However, it is unclear whether microRNAs (miRNAs), novel regulators in the SCI, are involved in SCI-induced pyroptosis. Methods Two GEO miRNA expression profiles (GSE158195 and GSE90452) were downloaded, and the differentially expressed miRNAs were analyzed by bioinformatics methods. An in vivo animal model and an in vitro cellular model of SCI were constructed in female C57BL/6 mice and BV-2 cells for studying the possible roles of FOXO3, miR-128-3p and NLRP3-mediated pyroptosis in SCI. Markers of ROS, cell pyroptosis and inflammation were measured by RT-qPCR, Western blotting, immunofluorescence, flow cytometry, and enzyme-linked immunosorbent assays. Histopathological changes in spinal cord tissue were detected using hematoxylin and eosin and immunohistochemical. The Basso-Beattie-Bresnahan (BBB) score was used to evaluate the motor function of mice in each group. Results Bioinformatics analysis of GSE158195 and GSE90452 datasets revealed a significant downregulation of miR-128-3p, a phenomenon that was consistently observed in the SCI mice model. Functionally, miR-128-3p upregulation improved functional behavioral recovery, relieved pathological injury, repressed oxidative stress, and alleviated pyroptosis and inflammation in the mouse SCI models. We also confirmed that Thioredoxin-interacting protein (TXNIP) was the target gene of miR-128-3p, and overexpression of TXNIP can effectively reverse the improvement of miR-128-3p in SCI cell model. Moreover, we found that transcription factor FOXO3 facilitated miR-128-3p expression, and its overexpression resulted in similar effects of miR-128-3p in the SCI cell model. Conclusion To the best of our knowledge, this is the first report demonstrating miR-128-3p improved secondary injury in SCI through the modulation of cell pyroptosis pathway. Our results suggest that FOXO3/miR-128-3p/TXNIP/NLRP3-mediated pyroptosis axis may be a potential therapeutic target for SCI.
Collapse
Affiliation(s)
| | | | - Chaojun Zheng
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinlei Xia
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | | | | |
Collapse
|
19
|
Letonja J, Nussdorfer P, Petrovič D. Single-Nucleotide Polymorphisms in the Thioredoxin Antioxidant System and Their Association with Diabetic Nephropathy in Slovenian Patients with Type 2 Diabetes-A Preliminary Study. Int J Mol Sci 2025; 26:1832. [PMID: 40076459 PMCID: PMC11899783 DOI: 10.3390/ijms26051832] [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: 12/30/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Diabetic nephropathy (DN) is a microvascular complication of type 2 diabetes mellitus (T2DM) that develops after years of T2DM and affects approximately one in four diabetic patients. Thioredoxin (TXN), thioredoxin reductase (TXNRD), and thioredoxin-interacting protein (TXNIP) are part of the thioredoxin antioxidant system, which is involved in DN. We included 897 Slovenian patients with T2DM lasting more than 10 years in our preliminary study. In total, 344 patients with DN were included in our case group, while 553 without DN comprised our control group. The genotypes of TXN2 rs8140110, TXNRD2 rs1548357, and TXNIP rs7212 were determined for all participants using real-time PCR. We found a statistically significant association between the T allele of the TXN2 rs8140110 polymorphism and DN (p < 0.001; OR: 0.52; 95% CI: 0.36-0.74). The TT and TC genotypes were also significantly less likely to develop DN in comparison to the CC genotype according to the dominant model of inheritance (p < 0.001; OR: 0.51; 95 CI: 0.34-0.75). We did not find a statistically significant association between rs1548357 or rs7212 and DN. To conclude, the rs8140110 polymorphism in the TXN2 gene is associated with DN in Slovenian patients with T2DM.
Collapse
Affiliation(s)
- Jernej Letonja
- Laboratory for Histology and Genetics of Atherosclerosis and Microvascular Diseases, Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia; (J.L.); (P.N.)
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Petra Nussdorfer
- Laboratory for Histology and Genetics of Atherosclerosis and Microvascular Diseases, Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia; (J.L.); (P.N.)
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Danijel Petrovič
- Laboratory for Histology and Genetics of Atherosclerosis and Microvascular Diseases, Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia; (J.L.); (P.N.)
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| |
Collapse
|
20
|
Zhou Y, Fang X, Huang LJ, Wu PW. Transcriptome and single-cell profiling of the mechanism of diabetic kidney disease. World J Diabetes 2025; 16:101538. [PMID: 39959271 PMCID: PMC11718477 DOI: 10.4239/wjd.v16.i2.101538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/29/2024] [Accepted: 11/26/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND The NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome may play an important role in diabetic kidney disease (DKD). However, the exact link remains unclear. AIM To investigate the role of the NLRP3 inflammasome in DKD. METHODS Using datasets from the Gene Expression Omnibus database, 30 NLRP3 inflammasome-related genes were identified. Differentially expressed genes were selected using differential expression analysis, whereas intersecting genes were selected based on overlapping differentially expressed genes and NLRP3 inflammasome-related genes. Subsequently, three machine learning algorithms were used to screen genes, and biomarkers were identified by overlapping the genes from the three algorithms. Potential biomarkers were validated by western blotting in a db/db mouse model of diabetes. RESULTS Two biomarkers, sirtuin 2 (SIRT2) and caspase 1 (CASP1), involved in the Leishmania infection pathway were identified. Both biomarkers were expressed in endothelial cells. Pseudo-temporal analysis based on endothelial cells showed that DKD mostly occurs during the mid-differentiation stage. Western blotting results showed that CASP1 expression was higher in the DKD group than in the control group (P < 0.05), and SIRT2 content decreased (P < 0.05). CONCLUSION SIRT2 and CASP1 provide a potential theoretical basis for DKD treatment.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Endocrinology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
| | - Xiao Fang
- Department of Kidney Transplantation, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350001, Fujian Province, China
| | - Lin-Jing Huang
- Department of Endocrinology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
- Department of Endocrinology National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou 350212, Fujian Province, China
- Clinical Research Center for Metabolic Diseases of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolism, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
- Diabetes Research Institute of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
| | - Pei-Wen Wu
- Department of Endocrinology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
- Department of Endocrinology National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou 350212, Fujian Province, China
- Clinical Research Center for Metabolic Diseases of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolism, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
- Diabetes Research Institute of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
| |
Collapse
|
21
|
Zhu Y, Yu J, Yang Q, Xie Y, Li X, Chen Z, Xiong Y, Fu W, He H, Yin S, Lan D, Li J, Xiong X. Mitochondria-targeted antioxidant MitoQ improves the quality of low temperature-preserved yak semen via alleviating oxidative stress. Anim Reprod Sci 2025; 273:107680. [PMID: 39709684 DOI: 10.1016/j.anireprosci.2024.107680] [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: 07/08/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
Low-temperature preservation of yak semen during transportation and conservation is crucial to accelerate yak breeding. The effects of low-temperature cooling on yak semen quality, however, are poorly understood. This study aimed to determine the dose-dependent effect of mitochondria-targeted antioxidant "MitoQ" on the motility, oxidative status, and mitochondrial function of yak semen during low-temperature preservation. Semen samples were collected from six adult healthy Maiwa yaks and preserved at 4 ℃ in semen extender containing 0, 50, 100, 200, and 400 nM MitoQ, respectively. Firstly, the motility, membrane integrity, acrosome integrity, and abnormity index of yak spermatozoa were evaluated to determine the optimal MitoQ concentration. Next, the effect of MitoQ at the optimal concentration on spermatozoa antioxidant capacity, including reactive oxygen species (ROS) and malondialdehyde (MDA) contents, total antioxidant capacity (T-AOC), and superoxide dismutase content (SOD) levels, as well as mitochondrial membrane potential were analyzed. Up to 96 h of low-temperature storage, 200 nM MitoQ showed the most optimal effect on motility, membrane integrity, and acrosome integrity (P < 0.05) but not on sperm morphology (P > 0.05). Also, 200 nM MitoQ markedly reduced yak spermatozoa ROS and MDA contents for up to 48 h of low-temperature storage (P < 0.05). Finally, 200 nM MitoQ significantly improved T-AOC, SOD, and mitochondrial membrane potential for up to 24, 48, and 72 h of low-temperature storage, respectively (P < 0.05). In summary, semen extender supplementation with 200 nM MitoQ is beneficial for low-temperature yak semen preservation via improving the oxidative status.
Collapse
Affiliation(s)
- Yanjin Zhu
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu 610041, China
| | - Jun Yu
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu 610041, China
| | - Qinhui Yang
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu 610041, China
| | - Yumian Xie
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Southwest Minzu University, Chengdu 610041, China
| | - Xupeng Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Southwest Minzu University, Chengdu 610041, China
| | - Zhuo Chen
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Southwest Minzu University, Chengdu 610041, China
| | - Yan Xiong
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu 610041, China
| | - Wei Fu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Southwest Minzu University, Chengdu 610041, China
| | - Honghong He
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu 610041, China
| | - Shi Yin
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Southwest Minzu University, Chengdu 610041, China
| | - Daoliang Lan
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Southwest Minzu University, Chengdu 610041, China
| | - Jian Li
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu 610041, China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Southwest Minzu University, Chengdu 610041, China
| | - Xianrong Xiong
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu 610041, China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Southwest Minzu University, Chengdu 610041, China.
| |
Collapse
|
22
|
Chen Y, Mei E, Nan S, Chen X, Zhang P, Zhu Q, Lan D, Qi S, Wang Y. Fibrin aggravates periodontitis through inducing NETs formation from mitochondrial DNA. Oral Dis 2025; 31:577-588. [PMID: 39054859 DOI: 10.1111/odi.15073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/22/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVES This study investigated the role of fibrin on neutrophil extracellular traps (NETs) formation from neutrophils and to elucidate the involvement of mitochondria in NETs formation during periodontitis. MATERIALS AND METHODS Plasminogen-deficient (Plg-/-) mice were employed to evaluate the effects of fibrin deposition on inflammation, bone resorption, and neutrophil infiltration in periodontal tissues. In addition, in vitro tests evaluated fibrin's impact on neutrophil-driven inflammation. Mitochondrial reactive oxygen species (mtROS) levels within neutrophils were quantified utilizing flow cytometry and immunofluorescence in vitro. Furthermore, the anti-inflammatory properties of the mtROS scavenger, Mito-TEMPO, were confirmed to regulate the NET formation in vitro and in vivo. RESULTS Plasminogen deficiency resulted in increased fibrin deposition, neutrophil infiltration, inflammatory factors concentration, and alveolar bone resorption in periodontal tissues. After neutrophils were treated by fibrin in vitro, the expression of inflammatory factors, the formation of mtROS, and NETs enriched in mitochondrial DNA (mtDNA) were upregulated, which were reversed by Mito-TEMPO in vitro. Moreover, Mito-TEMPO alleviated inflammation in Plg-/- mice. CONCLUSIONS This study showed that fibrin deposition in gingiva induced the NET formation in Plg-/- mice, in which the DNA in NETs was from mitochondria depending on increasing mtROS.
Collapse
Affiliation(s)
- Yinan Chen
- Department of Preventive Dentistry, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
- Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| | - Enhua Mei
- Department of Preventive Dentistry, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
- Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| | - Shunxue Nan
- Department of Preventive Dentistry, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
- Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| | - Xueting Chen
- Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
- Department of Prothodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Pengye Zhang
- Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
- Department of Prothodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Qingyu Zhu
- Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
- Department of Prothodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Dongmei Lan
- Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
- Department of Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Shengcai Qi
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
- Department of Prothodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Yan Wang
- Department of Preventive Dentistry, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| |
Collapse
|
23
|
Li C, Pan Y, Wang Y, Li X, Tie Y, Li S, Wang R, Zhao X, Fan J, Yan X, Wang Y, Sun X. Single-cell RNA sequencing of the carotid artery and femoral artery of rats exposed to hindlimb unloading. Cell Mol Life Sci 2025; 82:50. [PMID: 39833543 PMCID: PMC11747068 DOI: 10.1007/s00018-024-05572-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/20/2024] [Accepted: 12/30/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Prolonged spaceflight is known to cause vascular deconditioning and remodeling. Tail suspension, a widely used spaceflight analog, is reported to result in vascular remodeling of rats. However, little is known about the cellular atlas of the heterogeneous cells of CA and FA from hindlimb-unloaded rats. METHODS Firstly, we leveraged scRNA-seq to perform clustering analysis to identify diverse cell populations and sub-clusters within CA and FA from rats subjected to 3 months of hindlimb unloading. The dysregulated genes specific for artery types and cell types in HU group compared to Con were unraveled. Then R package "Cellchat" was used to reveal ligand-receptor cellular communication. At last, the TF network analysis was performed using the SCENIC R package to predict the pivotal TFs in rat artery remodeling induced by hindlimb unloading. RESULTS Clustering analysis identified ECs, SMCs, fibroblasts, and a spectrum of immune cells, as well as neuronal and stem cells. Notably, an increased percentage of ECs in the CA and a diminished proportion of SMCs in both CA and FA were observed following tail suspension. Intersection of dysregulated genes specific for artery type and cell type after tail suspension revealed several gene sets involved in ECM remodeling, inflammation, vasoconstriction, etc. Fibroblasts, in particular, exhibited the most significant gene expression variability, highlighting their plasticity. Subclustering within ECs, SMCs and fibroblasts revealed specialized subsets engaged in processes such as EndoMT and cell cycle checkpoint regulation. Additionally, enhanced intercellular interactions among major cell types, especially between SMC and fibroblast, underscored the importance of cell communication in vascular remodeling. Several TFs were identified as potentially influential in the vascular remodeling process under simulated microgravity conditions. CONCLUSIONS This study presents the first cellular atlas of the conductive arteries in hindlimb-unloaded rats, revealing a spectrum of dysregulated gene profiles. The identification of the subclusters of ECs, SMCs and fibroblasts, cellular communication analysis and transcription factors prediction are also included in this work. The findings provide a reference for future research on vascular deconditioning following long-duration spaceflight.
Collapse
Affiliation(s)
- Chengfei Li
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, China
| | - Yikai Pan
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, China
| | - Yuan Wang
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, China
| | - Xi Li
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, China
| | - Yateng Tie
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, China
| | - Shuhan Li
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, China
| | - Ruonan Wang
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, China
| | - Xingcheng Zhao
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, China
| | - Jieyi Fan
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, China
| | - Xianchun Yan
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, China.
| | - Yongchun Wang
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, China.
| | - Xiqing Sun
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, China.
| |
Collapse
|
24
|
Stanigut AM, Tuta L, Pana C, Alexandrescu L, Suceveanu A, Blebea NM, Vacaroiu IA. Autophagy and Mitophagy in Diabetic Kidney Disease-A Literature Review. Int J Mol Sci 2025; 26:806. [PMID: 39859520 PMCID: PMC11766107 DOI: 10.3390/ijms26020806] [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: 11/20/2024] [Revised: 01/08/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Autophagy and mitophagy are critical cellular processes that maintain homeostasis by removing damaged organelles and promoting cellular survival under stress conditions. In the context of diabetic kidney disease, these mechanisms play essential roles in mitigating cellular damage. This review provides an in-depth analysis of the recent literature on the relationship between autophagy, mitophagy, and diabetic kidney disease, highlighting the current state of knowledge, existing research gaps, and potential areas for future investigations. Diabetic nephropathy (DN) is traditionally defined as a specific form of kidney disease caused by long-standing diabetes, characterized by the classic histological lesions in the kidney, including mesangial expansion, glomerular basement membrane thickening, nodular glomerulosclerosis (Kimmelstiel-Wilson nodules), and podocyte injury. Clinical markers for DN are albuminuria and the gradual decline in glomerular filtration rate (GFR). Diabetic kidney disease (DKD) is a broader and more inclusive term, for all forms of chronic kidney disease (CKD) in individuals with diabetes, regardless of the underlying pathology. This includes patients who may have diabetes-associated kidney damage without the typical histological findings of diabetic nephropathy. It also accounts for patients with other coexisting kidney diseases (e.g., hypertensive nephrosclerosis, ischemic nephropathy, tubulointerstitial nephropathies), even in the absence of albuminuria, such as a reduction in GFR.
Collapse
Affiliation(s)
- Alina Mihaela Stanigut
- Clinical Medical Disciplines Department, Faculty of Medicine, Ovidius University of Constanta, 900470 Constanta, Romania; (A.M.S.); (L.T.); (L.A.); (A.S.)
- Nephrology Department, County Emergency Clinical Hospital of Constanta, 145 Tomis Street, 900591 Constanta, Romania
| | - Liliana Tuta
- Clinical Medical Disciplines Department, Faculty of Medicine, Ovidius University of Constanta, 900470 Constanta, Romania; (A.M.S.); (L.T.); (L.A.); (A.S.)
- Nephrology Department, County Emergency Clinical Hospital of Constanta, 145 Tomis Street, 900591 Constanta, Romania
| | - Camelia Pana
- Clinical Medical Disciplines Department, Faculty of Medicine, Ovidius University of Constanta, 900470 Constanta, Romania; (A.M.S.); (L.T.); (L.A.); (A.S.)
- Nephrology Department, County Emergency Clinical Hospital of Constanta, 145 Tomis Street, 900591 Constanta, Romania
| | - Luana Alexandrescu
- Clinical Medical Disciplines Department, Faculty of Medicine, Ovidius University of Constanta, 900470 Constanta, Romania; (A.M.S.); (L.T.); (L.A.); (A.S.)
- Gastroenterology Department, County Emergency Clinical Hospital of Constanta, 145 Tomis Street, 900591 Constanta, Romania
| | - Adrian Suceveanu
- Clinical Medical Disciplines Department, Faculty of Medicine, Ovidius University of Constanta, 900470 Constanta, Romania; (A.M.S.); (L.T.); (L.A.); (A.S.)
- Gastroenterology Department, County Emergency Clinical Hospital of Constanta, 145 Tomis Street, 900591 Constanta, Romania
| | - Nicoleta-Mirela Blebea
- Department of Pharmacotherapy, Faculty of Pharmacy, Ovidius University of Constanta, Aleea Universitatii Nr. 1, 900470 Constanta, Romania
| | - Ileana Adela Vacaroiu
- Department of Nephrology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Nephrology, Sf. Ioan Clinical Emergency Hospital, 042122 Bucharest, Romania
| |
Collapse
|
25
|
Cao X, Liu K, Yuan J, Hua Q, Rong K, Zhou T, He W, Pang Y, Yang X, Yu Y, Zhang P, Ma P, Cao Y, Zhao J, Morahan G, Xu J, Qin A. Transcriptional regulation of Rankl by Txnip-Ecd in aging and diabetic related osteoporosis. J Adv Res 2025:S2090-1232(25)00046-3. [PMID: 39826612 DOI: 10.1016/j.jare.2025.01.027] [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: 12/25/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025] Open
Abstract
INTRODUCTION Bone homeostasis between osteoclast bone resorption and osteoblastic bone formation is tightly regulated by a series of factors such as the receptor activator of nuclear factor-κB ligand (RANKL). Denosumab that neutralizes RANKL is effective and widely applied in the treatment of postmenopausal osteoporosis. However, factors that participated in the RANKL-related bone remodeling process in primary and secondary osteoporosis are less known. OBJECTIVES Revealing the novel transcriptional regulatory mechanism of RANKL is of great significance for the treatment of osteoporosis. METHODS After differential expression genes (DEGs) intersection and screening, we generated Thioredoxin-interacting protein (Txnip) bone marrow-derived mesenchymal stromal cells (BMSCs) genetic knockout mice and performed bone histomorphometry and histological analysis. RNA-Sequencing, Western blotting and immunofluorescence staining verified Rankl downregulation. Co-immunoprecipitation and immunofluorescence staining were used for Rankl regulation mechanism exploration. A specific inhibitor was selected for treatment effect verification. RESULTS Txnip knockout in BMSCs impaired its osteogenic differentiation, suppressed Rankl expression and subsequent osteoclast formation and thus led to increased bone mass. The regulatory function of Txnip on Rankl expression was revealed for the first time through the novel transcription-related Ecdysoneless (Ecd)-P300 axis. Pharmacological inhibition of Txnip can effectively prevent bilateral ovariectomy (OVX)-induced osteoporosis. CONCLUSIONS Inhibition of Txnip is an alternative way to suppress Rankl-mediated osteoblast and osteoclast crosstalk. This interesting finding rendered Txnip an ideal therapeutic target for the treatment of both ovariectomy-induced and diabetes-induced osteoporosis.
Collapse
Affiliation(s)
- Xiankun Cao
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Kexin Liu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jinbo Yuan
- School of Biomedical Sciences, the University of Western Australia, Perth, Western Australia 6009, Australia
| | - Qi Hua
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Kewei Rong
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Tangjun Zhou
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Wenxin He
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; Centre National de la Recherche Scientifique-Laboratoire International Associé (CNRS-LIA) Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yichuan Pang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200011, China
| | - Xiao Yang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yating Yu
- Department of Orthopedic and Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Shanghai 200011, People's Republic of China
| | - Pu Zhang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Peixiang Ma
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yu Cao
- Department of Orthopedic and Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Shanghai 200011, People's Republic of China
| | - Jie Zhao
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Grant Morahan
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, 6 Verdun Street, Nedlands, Perth, WA 6009, Australia; Australian Centre for Advancing Diabetes Innovations, The University of Melbourne, Parkville, Victoria 3050, Australia.
| | - Jiake Xu
- School of Biomedical Sciences, the University of Western Australia, Perth, Western Australia 6009, Australia; Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - An Qin
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
26
|
Zhang R, Lu M, Ran C, Niu L, Qi Q, Wang H. Ginsenoside Rg1 improves hypoxia-induced pulmonary vascular endothelial dysfunction through TXNIP/NLRP3 pathway-modulated mitophagy. J Ginseng Res 2025; 49:80-91. [PMID: 39872289 PMCID: PMC11764820 DOI: 10.1016/j.jgr.2024.10.002] [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: 06/12/2024] [Revised: 10/15/2024] [Accepted: 10/23/2024] [Indexed: 01/30/2025] Open
Abstract
Background Vascular endothelial dysfunction (VED) is one of the main pathogenic events in pulmonary arterial hypertension (PAH). Previous studies have demonstrated that the ginsenoside Rg1 (Rg1) can ameliorate PAH, but the mechanism by which Rg1 affects pulmonary VED in hypoxia-induced PAH remains unclear. Methods Network pharmacology, molecular docking and other experiments were used to explore the mechanisms by which Rg1 affects PAH. A PAH mouse model was established via hypoxia combined with the vascular endothelial growth factor (VEGFR) inhibitor su5416 (SuHx), and a cell model was established via hypoxia. The functions of Rg1 in VED, oxidative stress, inflammation, mitophagy, and TXNIP and NLRP3 expression were examined. Results In hypoxia-induced VED, progressive exacerbation of oxidative stress, inflammation, and mitophagy were observed, and were associated with elevated TXNIP and NLRP3 expression in vivo and in vitro. Rg1 improved hypoxia-induced impaired endothelium-dependent vasodilation and increased nitric oxide (NO) and endothelial NO synthase (eNOS) expression. Rg1, SRI37330 (a TXNIP inhibitor), MCC950 (an NLRP3 inhibitor), and Liensinine (a mitophagy inhibitor) attenuated oxidative stress, inflammation, and mitophagy by reducing the expression of TXNIP and NLRP3 in mice and cells. Furthermore, the combination of SB203580 (a mitophagy agonist) with Rg1 disrupted the protective effect of Rg1 on hypoxia-induced pulmonary artery and human pulmonary artery endothelial cells (HPAECs). Conclusion Rg1 improves hypoxia-induced pulmonary vascular endothelial dysfunction through TXNIP/NLRP3 pathway-modulated oxidative stress, inflammation and mitophagy.
Collapse
Affiliation(s)
- Ru Zhang
- The Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Meili Lu
- The Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Chenyang Ran
- The Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Linchao Niu
- The Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Qi Qi
- The Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Hongxin Wang
- The Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
27
|
Fu Y, Tang M, Duan Y, Pan Y, Liang M, Yuan J, Wang M, Laher I, Li S. MOTS-c regulates the ROS/TXNIP/NLRP3 pathway to alleviate diabetic cardiomyopathy. Biochem Biophys Res Commun 2024; 741:151072. [PMID: 39616938 DOI: 10.1016/j.bbrc.2024.151072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/06/2024] [Accepted: 11/25/2024] [Indexed: 12/11/2024]
Abstract
Chronic low-grade inflammation is a characteristic of diabetes, which often culminates in cardiovascular events including myocardial damage, thereby increasing the risk of debilitating cardiac complications. The mitochondria-derived peptide MOTS-c regulates glucose and lipid metabolism while improving insulin resistance, making it a potential candidate for the treatment of diabetes and cardiovascular diseases. We investigated the impact of MOTS-c on cardiac structure and inflammation in diabetic rats induced by a high-sugar-fat diet combined with low-dose streptozotocin (30 mg/kg, i.p.). Our results confirm that high glucose levels activate the nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome and increase reactive oxygen species (ROS), ultimately leading to myocardial injury. Furthermore, treatment with MOTS-c (0.5 mg/kg/day, i.p.) for 8 weeks reduced the expression of ROS/TXNIP/NLRP3 pathway proteins to inhibit the diabetic myocardial inflammatory response. These findings suggested that MOTS-c alleviates myocardial damage by inhibiting the ROS/TXNIP/NLRP3 pathway.
Collapse
Affiliation(s)
- Yu Fu
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Mi Tang
- School of Physical Education, Xihua University, Chengdu, China
| | - Yimei Duan
- School of Physical Education, Sichuan Normal University, Chengdu, China
| | - Yanrong Pan
- School of Physical Education, Sichuan Minzu College, Kangding, China
| | - Min Liang
- College of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinghan Yuan
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Manda Wang
- School of Sport Science, Beijing Sport University, Beijing, China
| | - Ismail Laher
- Department of Pharmacology and Therapeutics, Medicine, University of British Columbia, Vancouver, Canada
| | - Shunchang Li
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China.
| |
Collapse
|
28
|
Ouyang J, Wang H, Gan Y, Huang J. Uric acid mediates kidney tubular inflammation through the LDHA/ROS/NLRP3 pathway. Clin Exp Hypertens 2024; 46:2424834. [PMID: 39488824 DOI: 10.1080/10641963.2024.2424834] [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: 07/01/2024] [Revised: 09/26/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
PURPOSE Hyperuricemia (HUA) is an important factor leading to chronic kidney disease (CKD). The kidney tubular inflammatory response is activated in HUA. This study aimed to investigate whether lactate dehydrogenase A (LDHA) is involved in mediating uric acid-induced kidney tubular inflammatory response. METHODS In vivo, an HUA mouse model was established by continuous intraperitoneal injection of potassium oxonate (PO) for one week. A total of 18 C57BL/6J male adult mice were divided into three groups: control group, HUA group, and HUA+oxamate group, with six mice in each group. Oxamate was intraperitoneally injected into the mice one hour after PO injection. In vitro, an HUA model was simulated by stimulating HK-2 cells with uric acid. Oxamate and tempol inhibited LDHA and reactive oxygen species (ROS) in HK-2 cells. RESULTS In HUA mice, blood uric acid levels were significantly elevated. LDHA in kidney tubular cells was significantly increased in both in vivo and in vitro HUA models, accompanied by an increase in kidney tubular inflammation and ROS. Mechanistically, LDHA mediates uric acid-induced inflammation to kidney tubular cells through the ROS/NLRP3 pathway. Pharmacologic inhibition of LDHA or ROS in kidney tubular cells can significantly ameliorate inflammation response caused by uric acid. CONCLUSIONS LDHA in kidney tubular cells significantly was increased in HUA models. LDHA mediates kidney inflammation response induced by uric acid through the ROS/NLRP3 pathway. This study may provide a new intervention target for preventing kidney tubular inflammation caused by uric acid.
Collapse
Affiliation(s)
- Jun Ouyang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hui Wang
- School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Yumei Gan
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiangnan Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
29
|
Li J, Zhang D, Li H, Zhang Z, Shen Z, An R, Qiu H, Guo X, Zhang C, Chen L, Liu Z. Isodeoxyelephantopin ameliorates LPS-induced acute peritonitis by inhibiting NLRP3 inflammasome in vitro and in vivo. Int Immunopharmacol 2024; 143:113144. [PMID: 39536489 DOI: 10.1016/j.intimp.2024.113144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/02/2024] [Accepted: 09/08/2024] [Indexed: 11/16/2024]
Abstract
Isodeoxyelephantopin (IDET), a sesquiterpene lactone compound isolated from the Asteraceae plant Inula helenium, has been demonstrated to possess excellent antimicrobial, antidiabetic, hepatoprotective, wound healing, anti-inflammatory, and anticancer activities. However, the underlying mechanisms of IDET's anti-inflammatory properties remain unclear. In this study, we investigated the anti-inflammatory effects and mechanisms of IDET using both in vitro and in vivo models. Our findings revealed that IDET dose-dependently inhibited the upregulation of inflammatory cytokines (IL-1β, IL-6, TNF-α) and pro-inflammatory chemokines (MCP-1, CCL20) induced by Lipopolysaccharide (LPS). IDET suppressed the activation of cyclooxygenase-2 (COX-2) and the NLRP3 inflammasome. Subsequent mechanistic investigations demonstrated that IDET inhibited the mRNA and protein levels of Txnip, FOXO1, and EFhd2 in THP-1 cells in a time- and concentration-dependent manner. Furthermore, in an LPS-induced acute peritonitis mouse model, IDET effectively ameliorated symptoms, preserved ileal tissue integrity, attenuated immune cell infiltration, and reduced the expression of inflammatory cytokines in mouse serum. Notably, IDET exhibited an improvement in acute peritonitis by inhibiting the activation of NLRP3 inflammasome. Overall, our study highlights the significant anti-inflammatory activity of IDET, providing valuable insights into its therapeutic potential for acute peritonitis.
Collapse
Affiliation(s)
- Junhao Li
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Dongli Zhang
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Hengzhen Li
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Zhimeng Zhang
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Zehui Shen
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Ran An
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Honghong Qiu
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xindong Guo
- Guangzhou Quality Supervision and Testing Institute, Guangzhou City Research Center of Risk Dynamic Detection and Early Warning for Food Safety, Guangzhou City, Key Laboratory of Detection Technology for Food Safety, Guangzhou 511447, China
| | - Chen Zhang
- Rui Fu Biomedical (Shenzhen) Co., Ltd., Shenzhen 518055, China
| | - Lifeng Chen
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| | - Zhong Liu
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
30
|
Hu Z, Zhou Y, Gao C, Liu J, Pan C, Guo J. Astragaloside IV attenuates podocyte apoptosis via regulating TXNIP/NLRP3/GSDMD signaling pathway in diabetic nephropathy. Diabetol Metab Syndr 2024; 16:296. [PMID: 39696607 DOI: 10.1186/s13098-024-01546-y] [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: 09/28/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
OBJECTIVES Among all the diabetes complications brought on by persistent inflammation is diabetic kidney disease (DKD). One essential method of the inflammatory response's programmed cell death is anthrax. One of the main causes of diabetic renal disease progression in a high-glycemic environment is the lysis of renal resident cells. METHOD This investigation sought to determine whether Astragaloside IV (AS-IV)'s anti-pyroptosis action provides a protective function for the kidneys. For 12 weeks, db/db mice received 40 mg/kg of AS-IV by transgastric gavage. To validate the possible in vitro mechanism, mouse podocytes were cultivated for additional experiments. RESULTS In vitro, AS-IV led to a significant reduction in blood urea nitrogen (BUN), urine albumen-to-creatinine ratio (UACR), serum creatinine (CREA), and hyperglycemia in db/db mice and lessen the pathological alterations in the kidney. Moreover, pyrin structural domain of the NLR family pyrin domain containing 3 (NLRP3), cleaved-caspase-1, gasdermin D (GSDMD), IL-18, and IL-1β were down-expressed and podocyte markers podocin and nphs1 were up-regulated following AS-IV intervention. By silencing GSDMD, we demonstrated in vitro that HG-stimulated podocytes undergo pyroptosis. We also discovered that AS-IV can mitigate this pyroptosis. To confirm that AS-IV prevented the NLRP3 inflammasome from activating, the NLRP3 inhibitor CY-09 was employed. It was also discovered that AS-IV prevents the expression of TXNIP and NLRP3 as well as their interaction. GSDMD expression was significantly downregulated following TXNIP-siRNA treatment, whereas GSDMD expression was upregulated in TXNIP overexpression cells; this upregulation could be undone with AS-IV. CONCLUSIONS The anti-pyroptosis effect of AS-IV via the TXNIP-NLRP3-GSDMD axis improves the renal function and podocyte damage of db/db mice and delays the onset of DKD, according to in vivo and in vitro experimental data.
Collapse
Affiliation(s)
- Zhibo Hu
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Yu Zhou
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Cailing Gao
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Junfen Liu
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Congqing Pan
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China.
| | - Jun Guo
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China.
| |
Collapse
|
31
|
Chen T, Bai D, Gong C, Cao Y, Yan X, Peng R. Hydrogen sulfide mitigates mitochondrial dysfunction and cellular senescence in diabetic patients: Potential therapeutic applications. Biochem Pharmacol 2024; 230:116556. [PMID: 39332692 DOI: 10.1016/j.bcp.2024.116556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/08/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
Diabetes induces a pro-aging state characterized by an increased abundance of senescent cells in various tissues, heightened chronic inflammation, reduced substance and energy metabolism, and a significant increase in intracellular reactive oxygen species (ROS) levels. This condition leads to mitochondrial dysfunction, including elevated oxidative stress, the accumulation of mitochondrial DNA (mtDNA) damage, mitophagy defects, dysregulation of mitochondrial dynamics, and abnormal energy metabolism. These dysfunctions result in intracellular calcium ion (Ca2+) homeostasis disorders, telomere shortening, immune cell damage, and exacerbated inflammation, accelerating the aging of diabetic cells or tissues. Hydrogen sulfide (H2S), a novel gaseous signaling molecule, plays a crucial role in maintaining mitochondrial function and mitigating the aging process in diabetic cells. This article systematically explores the specific mechanisms by which H2S regulates diabetes-induced mitochondrial dysfunction to delay cellular senescence, offering a promising new strategy for improving diabetes and its complications.
Collapse
Affiliation(s)
- Ting Chen
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Dacheng Bai
- Guangdong Institute of Mitochondrial Biomedicine, Room 501, Coolpad Building, No.2 Mengxi Road, High-tech Industrial Park, Nanshan District, Shenzhen, Guangdong Province 518000, China
| | - Changyong Gong
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Yu Cao
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Xiaoqing Yan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Renyi Peng
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
32
|
Wang Y, Wang Q, Wang M, Wang X, Liu Q, Lv S, Nie H, Liu G. Epigallocatechin-3-Gallate Ameliorates Diabetic Kidney Disease by Inhibiting the TXNIP/NLRP3/IL-1β Signaling Pathway. Food Sci Nutr 2024; 12:10800-10815. [PMID: 39723074 PMCID: PMC11666909 DOI: 10.1002/fsn3.4617] [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: 08/17/2024] [Revised: 10/14/2024] [Accepted: 11/06/2024] [Indexed: 12/28/2024] Open
Abstract
Recent research indicates that the activation of the NLRP3 inflammasome is crucial in the development of diabetic kidney disease (DKD). Epigallocatechin-3-gallate (EGCG), the predominant catechin in green tea, has been noted for its anti-inflammatory properties in DKD. However, the specific mechanisms are not yet fully understood. In this study, our objective was to explore the effects of EGCG on podocytes and in diabetic kidney disease (DKD) mice and investigate how EGCG modulates the TXNIP/NLRP3/IL-1β signaling pathway in DKD, both in podocytes and animal models. In vitro, we co-cultured podocytes with EGCG and detected the viability, apoptosis, inflammation and the TXNIP/NLRP3/IL-1β signaling pathway. In vivo, DKD mice were given EGCG via oral gavage, followed by evaluations of renal function, inflammation, and the aforementioned signaling pathway. Our findings revealed that oxidative stress, inflammatory cytokines, and the TXNIP/NLRP3/IL-1β pathway were upregulated in podocytes exposed to high glucose (HG) and in the kidneys of DKD mice. However, EGCG treatment reduced the expression of the NLRP3 inflammasome and its associated proteins, including TXNIP, ASC, caspase-1, and IL-1β, as well as the levels of ROS and inflammatory factors such as TNF-α, IL-6, and IL-18. Furthermore, in vivo, EGCG improved kidney function, reduced albuminuria and body weight, and alleviated renal pathological damage. In summary, our study suggests that EGCG mitigates inflammation in podocytes and DKD through the TXNIP/NLRP3/IL-1β signaling pathway, indicating potential benefits of EGCG or green tea in managing DKD.
Collapse
Affiliation(s)
- Yinghui Wang
- Department of Nephrology, Multidisciplinary Innovation Center for NephrologyThe Second Hospital of Shandong UniversityJinanShandongChina
- Nephrology Research Institute of Shandong UniversityJinanShandongChina
| | - Qimeng Wang
- Department of Nephrology, Multidisciplinary Innovation Center for NephrologyThe Second Hospital of Shandong UniversityJinanShandongChina
- Nephrology Research Institute of Shandong UniversityJinanShandongChina
| | - Mingming Wang
- Department of Nephrology, Multidisciplinary Innovation Center for NephrologyThe Second Hospital of Shandong UniversityJinanShandongChina
- Nephrology Research Institute of Shandong UniversityJinanShandongChina
| | - Xueling Wang
- Department of Nephrology, Multidisciplinary Innovation Center for NephrologyThe Second Hospital of Shandong UniversityJinanShandongChina
- Nephrology Research Institute of Shandong UniversityJinanShandongChina
| | - Qingzhen Liu
- Department of Nephrology, Multidisciplinary Innovation Center for NephrologyThe Second Hospital of Shandong UniversityJinanShandongChina
- Nephrology Research Institute of Shandong UniversityJinanShandongChina
| | - Shasha Lv
- Department of Nephrology, Multidisciplinary Innovation Center for NephrologyThe Second Hospital of Shandong UniversityJinanShandongChina
- Nephrology Research Institute of Shandong UniversityJinanShandongChina
| | - Huibin Nie
- Department of Nephrology, Chengdu First People's HospitalIntegrated TCM and Western Medicine Hospital Affiliated to Chengdu University of TCMChengduSichuanChina
| | - Gang Liu
- Department of Nephrology, Multidisciplinary Innovation Center for NephrologyThe Second Hospital of Shandong UniversityJinanShandongChina
- Nephrology Research Institute of Shandong UniversityJinanShandongChina
- Key Laboratory of Reproductive Endocrinology of Ministry of EducationShandong UniversityJinanShandongChina
| |
Collapse
|
33
|
Shou L, He H, Wei Y, Xu X, Wang W, Zheng J. Identification of TXN and F5 as novel diagnostic gene biomarkers of the severe asthma based on bioinformatics and machine learning analysis. Autoimmunity 2024; 57:2427085. [PMID: 39531229 DOI: 10.1080/08916934.2024.2427085] [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: 09/12/2024] [Revised: 10/22/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
Asthma poses a major threat to human health. The aim of this study was to identify genetic markers of severe asthma and analyze the relationship between key genes and immune infiltration. Differentially expressed genes (DEGs) were first screened by downloading the training set GSE69683 and validation set GSE137268 from the GEO dataset. SVM-RFE analysis and the LASSO regression model were used to screen key genes, and CIBERSORT was used to assess immune infiltration in the samples. A total of 20 DEGs were identified in this study, mainly enriched for lymph node-like receptors, b-cell receptors, and neutrophil extracellular trap pathway. Comparative validation set GSE137268 identified thioredoxin (TXN) and coagulation factor V (F5) were identified as diagnostic markers of severe asthma. CIBERSORT analysis revealed that TXN and F5 are associated with multiple immune cell infiltrates. In addition, we identified miRNA and TF at the transcriptional level that may regulate F5 and TXN, and found that several commonly used drugs may exert therapeutic effects by targeting F5 and TXN. Taken together, TXN and F5 may be key genes in the development of severe asthma and are associated with immune infiltration. Our study can help to better understand the pathogenesis of asthma and provide new ideas for clinical treatment.
Collapse
Affiliation(s)
- Lu Shou
- Tongde Hospital of Zhejiang Province, Pulmonary and Critical Care Medicine, Hangzhou, Zhejiang, China
| | - Haidong He
- Tongde Hospital of Zhejiang Province, Pulmonary and Critical Care Medicine, Hangzhou, Zhejiang, China
| | - Yi Wei
- Tongde Hospital of Zhejiang Province, Pulmonary and Critical Care Medicine, Hangzhou, Zhejiang, China
| | - Xianrong Xu
- Tongde Hospital of Zhejiang Province, Pulmonary and Critical Care Medicine, Hangzhou, Zhejiang, China
| | - Wenmin Wang
- The Yangtze River Delta Biological Medicine Research and Development Center of Zhejiang Province, Yangtze Delta Region Institution of Tsinghua University, Hangzhou, Zhejiang, China
| | - Jisheng Zheng
- Tongde Hospital of Zhejiang Province, Pulmonary and Critical Care Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
34
|
Zhang L, Zhang D, Liu C, Tang B, Cui Y, Guo D, Duan M, Tu Y, Zheng H, Ning X, Liu Y, Chen H, Huang M, Niu Z, Zhao Y, Liu X, Xie J. Outer Membrane Vesicles Derived From Fusobacterium nucleatum Trigger Periodontitis Through Host Overimmunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400882. [PMID: 39475060 DOI: 10.1002/advs.202400882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 10/11/2024] [Indexed: 12/19/2024]
Abstract
The virulent bacteria-induced host immune response dominates the occurrence and progression of periodontal diseases because of the roles of individual virulence factors from these pathogens in the initiation and spread of inflammation. Outer membrane vesicles (OMVs) as a pathogenic entity have recently attracted great attention as messenger bridges between bacteria and host tissues. Herein, the novel role of OMVs derived from Fusobacterium nucleatum in the occurrence of periodontitis is dissected. In a rat periodontitis model, it is found that OMVs derived from F. nucleatum caused deterioration of periodontitis by enhancing inflammation of the periodontium and absorption of alveolar bone, which is almost equivalent to the effect of F. nucleatum itself. Furthermore, that OMVs can independently induce periodontitis is shown. The pathogenicity of OMVs is attributed to multiple pathogenic components identified by omics. After entering human periodontal ligament stem cells (hPDLSCs) by endocytosis, OMVs activated NLRP3 inflammasomes and impaired the mineralization of hPDLSCs through NF-κB (p65) signaling, leading to the final injury of the periodontium and damage of alveolar bone in periodontitis. These results provide a new understanding of OMVs derived from pathogens and cues for the prevention of periodontitis.
Collapse
Affiliation(s)
- Li 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, 610041, China
| | - Demao Zhang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Chengcheng Liu
- 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, 610041, China
| | - Boyu Tang
- 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, 610041, China
| | - Yujia Cui
- 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, 610041, China
| | - Daimo Guo
- 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, 610041, China
| | - Mengmeng Duan
- 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, 610041, China
| | - Ying Tu
- 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, 610041, China
| | - Huiling Zheng
- 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, 610041, China
| | - Xinjie Ning
- 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, 610041, China
| | - Yang Liu
- 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, 610041, China
| | - Haoran Chen
- 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, 610041, China
| | - Minglei Huang
- 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, 610041, China
| | - Zhixing Niu
- 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, 610041, China
| | - Yanfang Zhao
- Department of Pediatric Dentistry, School of Dentistry, University of Alabama Birmingham, Birmingham, 35233, USA
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Jing Xie
- 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, 610041, China
| |
Collapse
|
35
|
Ye S, Zhang M, Zheng X, Li S, Fan Y, Wang Y, Peng H, Chen S, Yang J, Tan L, Zhang M, Xie P, Li X, Luo N, Wang Z, Jin L, Wu X, Pan Y, Fan J, Zhou Y, Tang SCW, Li B, Chen W. YAP1 preserves tubular mitochondrial quality control to mitigate diabetic kidney disease. Redox Biol 2024; 78:103435. [PMID: 39608245 PMCID: PMC11629574 DOI: 10.1016/j.redox.2024.103435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/23/2024] [Accepted: 11/16/2024] [Indexed: 11/30/2024] Open
Abstract
Renal tubule cells act as a primary site of injury in diabetic kidney disease (DKD), with dysfunctional mitochondrial quality control (MQC) closely associated with progressive kidney dysfunction in this context. Our investigation delves into the observed inactivation of yes-associated protein 1 (YAP1) and consequential dysregulation of MQC within renal tubule cells among DKD subjects through bioinformatic analysis of transcriptomics data from the Gene Expression Omnibus (GEO) dataset. Receiver operating characteristic curve analysis unequivocally underscores the robust diagnostic accuracy of YAP1 and MQC-related genes for DKD. Furthermore, we observed YAP1 inactivation, accompanied by perturbed MQC, within cultured tubule cells exposed to high glucose (HG) and palmitic acid (PA). This pattern was also evident in the tubulointerstitial compartment of kidney sections from biopsy-approved DKD patients. Additionally, renal tubule cell-specific Yap1 deletion exacerbated kidney injury in diabetic mice. Mechanistically, Yap1 deletion disrupted MQC, leading to mitochondrial aberrations in mitobiogenesis and mitophagy within tubule cells, ultimately culminating in histologic tubular injury. Notably, Yap1 deletion-induced renal tubule injury promoted the secretion of C-X-C motif chemokine ligand 1 (CXCL1), potentially augmenting M1 macrophage infiltration within the renal microenvironment. These multifaceted events were significantly ameliorated by administrating the YAP1 activator XMU-MP-1 in DKD mice. Consistently, bioinformatic analysis of transcriptomics data from the GEO dataset revealed a noteworthy upregulation of tubule cells-derived chemokine CXCL1 associated with macrophage infiltration among DKD patients. Crucially, overexpression of YAP1 via adenovirus transfection sustained mitochondrial membrane potential, mtDNA copy number, oxygen consumption rate, and activity of mitochondrial respiratory chain complex, but attenuated mitochondrial ROS production, thereby maintaining MQC and subsequently suppressing CXCL1 generation within cultured tubule cells exposed to HG and PA. Collectively, our study establishes a pivotal role of tubule YAP1 inactivation-mediated MQC dysfunction in driving DKD progression, at least in part, facilitated by promoting M1 macrophage polarization through a paracrine-dependent mechanism.
Collapse
Affiliation(s)
- Siyang Ye
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Meng Zhang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Xunhua Zheng
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Suchun Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Yuting Fan
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Yiqin Wang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Huajing Peng
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Sixiu Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Jiayi Yang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Li Tan
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Manhuai Zhang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Peichen Xie
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Xiaoyan Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Ning Luo
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Zhipeng Wang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Leigang Jin
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xiaoping Wu
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yong Pan
- Department of Pathophysiology, School of Basic Medical Science, Shenzhen University Medical School, Shenzhen, 518000, China
| | - Jinjin Fan
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Yi Zhou
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Sydney C W Tang
- Division of Nephrology, Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Bin Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China.
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China.
| |
Collapse
|
36
|
Li M, Xiao J, Chen B, Pan Z, Wang F, Chen W, He Q, Li J, Li S, Wang T, Zhang G, Wang H, Chen J. Loganin inhibits the ROS-NLRP3-IL-1β axis by activating the NRF2/HO-1 pathway against osteoarthritis. Chin J Nat Med 2024; 22:977-990. [PMID: 39510640 DOI: 10.1016/s1875-5364(24)60555-8] [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: 01/06/2024] [Indexed: 11/15/2024]
Abstract
Loganin (LOG), a bioactive compound derived from Cornus officinalis Siebold & Zucc, has been understudied in the context of osteoarthritis (OA) treatment. In this study, we induced an inflammatory response in chondrocytes using lipopolysaccharide (LPS) and subsequently treated these cells with LOG. We employed fluorescence analysis to quantify reactive oxygen species (ROS) levels and measured the expression of NLRP3 and nuclear factor erythropoietin-2-related factor 2 (NRF2) using real-time quantitative polymerase chain reaction (qRT-PCR), Western blotting, and immunofluorescence (IF) techniques. Additionally, we developed an OA mouse model by performing medial meniscus destabilization (DMM) surgery and monitored disease progression through micro-computed tomography (micro-CT), hematoxylin and eosin (H&E) staining, safranin O and fast green (S&F) staining, and immunohistochemical (IHC) analysis. Our results indicate that LOG significantly reduced LPS-induced ROS levels in chondrocytes, inhibited the activation of the NLRP3 inflammasome, and enhanced NRF2/heme oxygenase 1 (HO-1) signaling. In vivo, LOG treatment mitigated cartilage degradation and osteophyte formation triggered by DMM surgery, decreased NLRP3 expression, and increased NRF2 expression. These findings suggest that LOG has a protective effect against OA, potentially delaying disease progression by inhibiting the ROS-NLRP3-IL-1β axis and activating the NRF2/HO-1 pathway.
Collapse
Affiliation(s)
- Miao Li
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jiacong Xiao
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Baihao Chen
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zhaofeng Pan
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Fanchen Wang
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Weijian Chen
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qi He
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jianliang Li
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shaocong Li
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Ting Wang
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Gangyu Zhang
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Biomedicine, University of Basel, Basel, Switzerland.
| | - Haibin Wang
- Department of Orthopaedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Jianfa Chen
- Department of Orthopaedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| |
Collapse
|
37
|
Kang M, Kwon H, Song J, Jang Y, Yang SH, Cha SM, Moon JH, Kim YC, Kim HJ. Spatial Transcriptomic Signatures of Early Acute T Cell-mediated Rejection in Kidney Transplants. Transplant Direct 2024; 10:e1705. [PMID: 39399058 PMCID: PMC11469874 DOI: 10.1097/txd.0000000000001705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 10/15/2024] Open
Abstract
Background Kidney transplantation significantly improves the quality of life for those with end-stage renal failure, yet allograft rejection resulting from immune cell interactions remains a persistent challenge. Although T cell-directed immunosuppressive drugs effectively contain graft rejection in most patients, a notable proportion still experiences acute T cell-mediated rejection (TCMR). Despite an emphasis on suppressing T cell-mediated immune responses, successful control over TCMR is not always achieved, suggesting the potential involvement of factors beyond T cells. Methods Biopsy samples from suspicious (borderline) for acute TCMR (borderline TCMR) and non-TCMR patients were obtained 9 d postsurgery, and spatial transcriptomics profiling was conducted using the GeoMx Digital Spatial Profiler platform. Regions of interest in the glomerulus and interstitium were selected on the basis of immunohistochemistry staining anti-CD3 to identify areas with T-lymphocyte infiltration. Differential gene expression analysis was performed using unpaired t tests. Results Unbiased clustering of transcriptional profiles across all regions of interest showed distinct transcriptional profiles between glomeruli and interstitium in non-TCMR samples, whereas borderline TCMR samples displayed no distinct transcriptional profiles between these regions. Contrary to the prevailing T cell-centric view, we observed pathways and genes associated with innate immunity-related inflammatory conditions expressed in glomerular regions of borderline TCMR biopsies. Immunofluorescence staining for CD68 confirmed the presence of macrophages in the glomeruli of the post-TCMR sample in a validation cohort, indicating macrophage involvement in the glomerular response after TCMR. Conclusions Activation of the innate immune response in borderline TCMR appears to impact not only the interstitium but also the glomerulus. Glomerulus-specific immune signatures suggest the role of the innate immune system in rejection. This nuanced understanding proposes the necessity for tailored therapeutic interventions targeting both innate and adaptive immune pathways to enhance transplant outcomes.
Collapse
Affiliation(s)
- Minji Kang
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea
| | - Haeyoon Kwon
- Department of Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Jeongin Song
- Department of Internal Medicine, Dongguk University Ilsan Hospital, Goyang, South Korea
| | - Yunyoung Jang
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
- Transplantation Center, Seoul National University Hospital, Seoul, South Korea
| | - Seung Hee Yang
- Kidney Research Institute, Seoul National University Medical Research Center, Seoul, South South Korea
| | - Seung-Min Cha
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea
| | - Ji Hwan Moon
- Samsung Genome Institute, Samsung Medical Center, Seoul, South Korea
| | - Yong Chul Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
- Transplantation Center, Seoul National University Hospital, Seoul, South Korea
| | - Hyun Je Kim
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea
- Department of Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University
| |
Collapse
|
38
|
Gui R, Ren Y, Wang Z, Li Y, Wu C, Li X, Li M, Li Y, Qian L, Xiong Y. Deciphering interleukin-18 in diabetes and its complications: Biological features, mechanisms, and therapeutic perspectives. Obes Rev 2024; 25:e13818. [PMID: 39191434 DOI: 10.1111/obr.13818] [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: 02/28/2024] [Revised: 07/16/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024]
Abstract
Interleukin-18 (IL-18), a potent and multifunctional pro-inflammatory cytokine, plays a critical role in regulating β-cell failure, β-cell death, insulin resistance, and various complications of diabetes mellitus (DM). It exerts its effects by triggering various signaling pathways, enhancing the production of pro-inflammatory cytokines and nitric oxide (NO), as well as promoting immune cells infiltration and β-cells death. Abnormal alterations in IL-18 levels have been revealed to be strongly associated with the onset and development of DM and its complications. Targeting IL-18 may present a novel and promising approach for DM therapy. An increasing number of IL-18 inhibitors, including chemical and natural inhibitors, have been developed and have been shown to protect against DM and diabetic complications. This review provides a comprehensive understanding of the production, biological functions, action mode, and activated signaling pathways of IL-18. Next, we shed light on how IL-18 contributes to the pathogenesis of DM and its associated complications with links to its roles in the modulation of β-cell failure and death, insulin resistance in various tissues, and pancreatitis. Furthermore, the therapeutic potential of targeting IL-18 for the diagnosis and treatment of DM is also highlighted. We hope that this review will help us better understand the functions of IL-18 in the pathogenesis of DM and its complications, providing novel strategies for DM diagnosis and treatment.
Collapse
Affiliation(s)
- Runlin Gui
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Yuanyuan Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Zhen Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Yang Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Chengsong Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Xiaofang Li
- Department of Gastroenterology, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Man Li
- Department of Endocrinology, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Yujia Li
- Department of Traditional Chinese Medicine, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Lu Qian
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
- Scientific Research Center, Xi'an Mental Health Center, Xi'an, Shaanxi, China
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| |
Collapse
|
39
|
Liu CC, Ji JL, Wang Z, Zhang XJ, Ding L, Zhang Y, Zhou Y, Zhang DJ, Tang ZL, Cao JY, Zhang AQ, Liu BC, Li ZL, Ma RX. TRPC6-Calpain-1 Axis Promotes Tubulointerstitial Inflammation by Inhibiting Mitophagy in Diabetic Kidney Disease. Kidney Int Rep 2024; 9:3301-3317. [PMID: 39534194 PMCID: PMC11551102 DOI: 10.1016/j.ekir.2024.08.019] [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: 04/20/2024] [Revised: 07/23/2024] [Accepted: 08/16/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Renal tubulointerstitial inflammation represents an effective indicator for predicting the progression of diabetic kidney disease (DKD). Mitophagy abnormality is 1 of the most important factors involved in tubule injury. However, the exact molecular mechanism underlying mitophagy abnormality-mediated tubulointerstitial inflammation in DKD remains poorly understood. Methods In this study, a streptozotocin-induced DKD mouse model was established and HK-2 cells treated with high glucose (HG) served as an in vitro model. Tubular mitophagy was regulated through pharmacological urolithin A (UA) administration. The functional effect of the transient receptor potential cation channel, subfamily C, member 6 (TRPC6) was explored using genetic interventions in vivo and in vitro. Results We found that renal tubulointerstitial inflammation in DKD was closely associated with mitophagy inhibition, which was mediated by disturbance of PINK1/Parkin pathway. Mitophagy activation significantly attenuated tubular injury and tubulointerstitial inflammation. Further, it was found that TRPC6 was markedly increased in DKD and played an essential role in mitophagy inhibition by activating calpain-1. Knockdown of Trpc6 partially reversed mitophagy abnormality and consequently attenuated tubular injury and tubulointerstitial inflammation in vivo and in vitro. Finally, we found that tubular TRPC6-mediated mitophagy inhibition was blocked with BAPTA (a specific Ca2+ chelator) or calpeptin (a specific calpain-1 inhibitor). Conclusion Our study reveals that TRPC6-calpain-1 axis promotes tubulointerstitial inflammation in DKD by inhibiting mitophagy.
Collapse
Affiliation(s)
- Cong-Cong Liu
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jia-Ling Ji
- Department of Pediatrics, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ze Wang
- Department of Nephrology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xing-Jian Zhang
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Lin Ding
- Department of Nephrology, Minda Hospital Affiliated to Hubei Minzu University, Enshi, Hubei, China
| | - Yao Zhang
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yan Zhou
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Dong-Jie Zhang
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhen-Lin Tang
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jing-Yuan Cao
- Department of Nephrology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ai-Qing Zhang
- Department of Pediatrics, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Zuo-Lin Li
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Rui-Xia Ma
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
40
|
Hong J, Li X, Hao Y, Xu H, Yu L, Meng Z, Zhang J, Zhu M. The PRMT6/STAT1/ACSL1 axis promotes ferroptosis in diabetic nephropathy. Cell Death Differ 2024; 31:1561-1575. [PMID: 39134684 PMCID: PMC11519485 DOI: 10.1038/s41418-024-01357-8] [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: 03/12/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
Hyperglycaemia-induced ferroptosis is a significant contributor to kidney dysfunction in diabetic nephropathy (DN) patients. In addition, targeting ferroptosis has clinical implications for the treatment of DN. However, effective therapeutic targets for ferroptosis have not been identified. In this study, we aimed to explore the precise role of protein arginine methyltransferase 6 (PRMT6) in regulating ferroptosis in DN. In the present study, we utilized a mouse DN model consisting of both wild-type and PRMT6-knockout (PRMT6-/-) mice. Transcriptomic and lipidomic analyses, along with various molecular biological methodologies, were used to determine the potential mechanism by which PRMT6 regulates ferroptosis in DN. Our results indicate that PRMT6 downregulation participates in kidney dysfunction and renal cell death via the modulation of ferroptosis in DN. Moreover, PRMT6 reduction induced lipid peroxidation by upregulating acyl-CoA synthetase long-chain family member 1 (ACSL1) expression, ultimately contributing to ferroptosis. Furthermore, we investigated the molecular mechanism by which PRMT6 interacts with signal transducer and activator of transcription 1 (STAT1) to jointly regulate ACSL1 transcription. Additionally, treatment with the STAT1-specific inhibitor fludarabine delayed DN progression. Furthermore, we observed that PRMT6 and STAT1 synergistically regulate ACSL1 transcription to mediate ferroptosis in hyperglycaemic cells. Our study demonstrated that PRMT6 and STAT1 comodulate ACSL1 transcription to induce the production of phospholipid-polyunsaturated fatty acids (PL-PUFAs), thus participating in ferroptosis in DN. These findings suggest that the PRMT6/STAT1/ACSL1 axis is a new therapeutic target for the prevention and treatment of DN.
Collapse
Affiliation(s)
- Jia Hong
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue Li
- Department of Anesthesiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yingxiang Hao
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongjiao Xu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lang Yu
- Department of Anesthesiology, Huzhou Central Hospital, Affiliated Central Hospital of HuZhou University, No.1558 Sanhuan North Road, Huzhou, Zhejiang, China
| | - Zhipeng Meng
- Department of Anesthesiology, Huzhou Central Hospital, Affiliated Central Hospital of HuZhou University, No.1558 Sanhuan North Road, Huzhou, Zhejiang, China.
| | - Jianhai Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Minmin Zhu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
41
|
Zhang W, Yu W, Zhu Y, Gu J, Gu X. Alda-1 Ameliorates Oxidative Stress-Induced Cardiomyocyte Damage by Inhibiting the Mitochondrial ROS/TXNIP/NLRP3 Pathway. J Biochem Mol Toxicol 2024; 38:e70032. [PMID: 39467157 DOI: 10.1002/jbt.70032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/23/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
Alda-1 functions as an agonist of aldehyde dehydrogenase (ALDH2) within the mitochondria, contributing to the preservation of mitochondrial structure and function. This study aimed to determine whether Alda-1 inhibited the accumulation of mitochondrial reactive oxygen species (mtROS) and improved cardiomyocyte damage under oxidative stress. Cardiomyocytes derived from human induced pluripotent embryonic stem cells (iPSC-CMs) and human AC16 cardiomyocytes were chosen for the experiments. Oxidative stress was induced in both cardiomyocyte types using hydrogen peroxide (H2O2), a commonly employed agent. The mtROS accumulation and mitochondrial functional status were assessed by measuring the ROS content, mitochondrial membrane potential, and mitochondrial respiratory chain function. Co-IP experiments were used to analyze whether mtROS promoted protein interactions between TXNIP and NLRP3 and induced NLRP3 inflammasome activation. The results showed that Alda-1 mitigated damage to mitochondrial structure and function under oxidative stress, concurrently reducing the accumulation of mtROS. Co-IP experiments revealed that elevated mtROS levels attenuated the protein interaction between TXNIP and TRX while intensifying the interaction between TXNIP and NLRP3. Consequently, this triggers activation of the NLRP3 inflammasome, leading to cardiomyocyte damage. In contrast, TXNIP knockdown inhibited H2O2-induced myocardial injury and enhanced the therapeutic effects of Alda-1. Collectively, the results show that, in an H2O2 environment, Alda-1 increased the production of ALDH2 activity in cardiomyocytes, hindered the production of mtROS, disrupted the interaction between TXNIP and NLRP3, and alleviated myocardial damage during oxidative stress.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Cardiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
- Medicine College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Wei Yu
- Department of Cardiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Ye Zhu
- Medicine College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Jianjun Gu
- Department of Cardiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiang Gu
- Department of Cardiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
- Medicine College of Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
42
|
Li H, Wang R, Chen Y, Zhao M, Lan S, Zhao C, Li X, Li W. Integrated network pharmacology and pharmacological investigations to discover the active compounds of Toona sinensis pericarps against diabetic nephropathy. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118441. [PMID: 38851471 DOI: 10.1016/j.jep.2024.118441] [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: 04/14/2024] [Revised: 05/22/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Toona sinensis (A. Juss.) Roem. Is a deciduous woody plant native to Eastern and Southeastern Asia. Different parts of this plant have a long history of being applied as traditional medicines to treat various diseases. The fruits have been used for antidiabetic, antidiabetic nephropathy (anti-DN), antioxidant, anti-inflammatory, and other activities. AIM OF THE STUDY The purpose of this study was to investigate the effects of EtOAc (PEAE) and n-BuOH extracts (PNBE) from T. sinensis pericarps (TSP) on kidney injury in high-fat and high-glucose diet (HFD)/streptozotocin (STZ)-induced DN mice by network pharmacology and pharmacological investigations, as well as to further discover active compounds that could ameliorate oxidative stress and inflammation, thereby delaying DN progression by regulating the Nrf2/NF-κB pathway in high glucose (HG)-induced glomerular mesangial cells (GMCs). MATERIALS AND METHODS The targets of TSP 1-16 with DN were analyzed by network pharmacology. HFD/STZ-induced DN mouse models were established to evaluate the effects of PEAE and PNBE. Six groups were divided into normal, model, PEAE100, PEAE400, PNBE100, and PNBE400 groups. Fasting blood glucose (FBG) levels, organ indices, plasma MDA, SOD, TNF-α, and IL-6 levels, as well as renal tissue Nrf2, HO-1, NF-κB, TNF-α, and TGF-β1 levels were determined, along with hematoxylin-eosin (H&E) and immunohistochemical (IHC) analysis of kidney sections. Furthermore, GMC activity screening combined with molecular docking was utilized to discover active compounds targeting HO-1, TNF-α, and IL-6. Moreover, western blotting assays were performed to validate the mechanism of Nrf2 and NF-κB in HG-induced GMCs. RESULTS Network pharmacology predicted that the main targets of PEAE and PNBE in the treatment of DN include IL-6, INS, TNF, ALB, GAPDH, IL-1β, TP53, EGFR, and CASP3. Additionally, major pathways include AGE-RAGE and IL-17. In vivo experiments, treatment with PEAE and PNBE effectively reduced FBG levels and organ indices, while plasma MDA, SOD, TNF-α, and IL-6 levels, renal tissue Nrf2, HO-1, NF-κB, TNF-α, and TGF-β1 levels, and renal function were significantly improved. PEAE and PNBE significantly improved glomerular and tubule injury, and inhibited the development of DN by regulating the levels of oxidative stress and inflammation-related factors. In vitro experiments, compound 11 strongly activated HO-1 and inhibited TNF-α and IL-6. The molecular docking results revealed that compound 11 exhibited a high binding affinity towards the targets HO-1, TNF-α, and IL-6 (<-6 kcal/mol). Western blotting results showed compound 11 effectively regulated Nrf2 and NF-κB p65 protein levels, and significantly improved oxidative stress damage and inflammatory responses in HG-induced GMCs. CONCLUSION PEAE, PNBE, and their compounds, especially compound 11, may have the potential to prevent and treat DN, and are promising natural nephroprotective agents.
Collapse
Affiliation(s)
- Huiting Li
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China.
| | - Rongshen Wang
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China; Key Laboratory of Molecular Pharmacology and Translational Research, Shandong Second Medical University, Weifang, 261053, China.
| | - Ying Chen
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China.
| | - Mengyao Zhao
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China.
| | - Shuying Lan
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China.
| | - Chunzhen Zhao
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China; Key Laboratory of Molecular Pharmacology and Translational Research, Shandong Second Medical University, Weifang, 261053, China.
| | - Xu Li
- Affiliated Hospital of Shandong Second Medical University, Weifang, 261041, China.
| | - Wanzhong Li
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China; Key Laboratory of Molecular Pharmacology and Translational Research, Shandong Second Medical University, Weifang, 261053, China.
| |
Collapse
|
43
|
Huang B, Zhang J, Tian H, Ren S, Chen K, Feng J, Fan S, Tuo Y, Wang X, Yu L, Ma C, Peng Q, Chen X, He R, Li G. Metformin modulates the TXNIP-NLRP3-GSDMD pathway to improve diabetic bladder dysfunction. Sci Rep 2024; 14:23868. [PMID: 39396086 PMCID: PMC11470931 DOI: 10.1038/s41598-024-72129-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 09/04/2024] [Indexed: 10/14/2024] Open
Abstract
To validate the therapeutic efficacy of metformin on diabetic bladder dysfunction (DBD) and further elucidate whether the TXNIP-NLRP3-GSDMD axis serves as a target for metformin in ameliorating DBD. C57BL/6J mice were induced with diet-induced obesity by being fed a high-fat diet (HFD) for 16 weeks. After establishing the model, the mice were treated with metformin for 4 weeks, and their glucose metabolism-related parameters were assessed. Urine spot assays and urodynamic measurements were conducted to reflect the bladder function and urinary behavior in mice, while histological examination was performed to observe morphological changes. Western blot analysis was employed to measure the expression levels of pyroptotic factors such as TXNIP, NLRP3, GSDMD, and tight junction proteins. Metformin treatment significantly improved glucose tolerance and insulin sensitivity in mice. Moreover, it showed promise in decreasing urinary spot occurrence, reducing urination frequency, alleviating non-voiding contractions, and stabilizing peak urinary pressure. Following metformin therapy, mice displayed restored epithelial fold structure, increased thickness of the muscular layer, substantial decrease in muscle fiber content, notably reduced levels of TXNIP and GSDMD proteins in the metformin-treated group compared to the DBD group, and restored expression of tight junction proteins Zo-1, Claudin-1, and Occludin. Metformin ameliorates urothelial cells damage in DBD mice by inhibiting TXNIP generation and reducing NLRP3 and GSDMD production.
Collapse
Affiliation(s)
- Bincheng Huang
- Urology Department of General Hospital, Ningxia Medical University, Yinchuan, Ningxia, China
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Jin Zhang
- Urology Department of General Hospital, Ningxia Medical University, Yinchuan, Ningxia, China
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Haifu Tian
- Urology Department of General Hospital, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Shuai Ren
- Urology Department of General Hospital, Ningxia Medical University, Yinchuan, Ningxia, China
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Keming Chen
- Urology Department of General Hospital, Ningxia Medical University, Yinchuan, Ningxia, China
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Jiajin Feng
- Urology Department of General Hospital, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Shuzhe Fan
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Yunshang Tuo
- Urology Department of General Hospital, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xuehao Wang
- Urology Department of General Hospital, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Leyi Yu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Cunling Ma
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Qingjie Peng
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Xiaojiang Chen
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Rui He
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, China.
| | - Guangyong Li
- Urology Department of General Hospital, Ningxia Medical University, Yinchuan, Ningxia, China.
| |
Collapse
|
44
|
Ji Y, Hua H, Jia Z, Zhang A, Ding G. Therapy Targeted to the NLRP3 Inflammasome in Chronic Kidney Disease. KIDNEY DISEASES (BASEL, SWITZERLAND) 2024; 10:369-383. [PMID: 39430292 PMCID: PMC11488838 DOI: 10.1159/000539496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 05/07/2024] [Indexed: 10/22/2024]
Abstract
Background The NLRP3 inflammasome is a cytoplasmic polymeric protein complex composed of the cytoplasmic sensor NLRP3, the apoptosis-related spot-like protein ASC, and the inflammatory protease caspase-1. NLRP3 activates and releases IL-1β through classical pathways, and IL-18 mediates inflammation and activates gasdermin-D protein to induce cellular pyroptosis. Numerous studies have also emphasized the non-classical pathway activated by the NLRP3 inflammasome in chronic kidney disease (CKD) and the inflammasome-independent function of NLRP3. Summary The NLRP3-targeting inflammasome and its associated pathways have thus been widely studied in models of CKD treatment, but no drug that targets NLRP3 has thus far been approved for the treatment of CKD. Key Messages We herein reviewed the current interventional methods for targeting the NLRP3 inflammasome in various CKD models, analyzed their underlying mechanisms of action, classified and compared them, and discussed the advantages and follow-up directions of various interventional methods. This review therefore provides novel ideas and a reference for the development of targeted NLRP3-inflammasome therapy in CKD.
Collapse
Affiliation(s)
- Yong Ji
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Hu Hua
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Guixia Ding
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
45
|
Wang XL, Li L, Meng X. Interplay between the Redox System and Renal Tubular Transport. Antioxidants (Basel) 2024; 13:1156. [PMID: 39456410 PMCID: PMC11505102 DOI: 10.3390/antiox13101156] [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: 07/20/2024] [Revised: 09/03/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
The kidney plays a critical role in maintaining the homeostasis of body fluid by filtration of metabolic wastes and reabsorption of nutrients. Due to the overload, a vast of energy is required through aerobic metabolism, which inevitably leads to the generation of reactive oxygen species (ROS) in the kidney. Under unstressed conditions, ROS are counteracted by antioxidant systems and maintained at low levels, which are involved in signal transduction and physiological processes. Accumulating evidence indicates that the reduction-oxidation (redox) system interacts with renal tubular transport. Redox imbalance or dysfunction of tubular transport leads to renal disease. Here, we discuss the ROS and antioxidant systems in the kidney and outline the metabolic dysfunction that is a common feature of renal disease. Importantly, we describe the key molecules involved in renal tubular transport and their relationship to the redox system and, finally, summarize the impact of their dysregulation on the pathogenesis and progression of acute and chronic kidney disease.
Collapse
Affiliation(s)
- Xiao-Lan Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Lianjian Li
- Department of Vascular Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Hubei Academy of Chinese Medicine, Wuhan 430061, China;
| | - Xianfang Meng
- Department of Neurobiology, Institute of Brain Research, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
46
|
Qiu X, Lan X, Li L, Chen H, Zhang N, Zheng X, Xie X. The role of perirenal adipose tissue deposition in chronic kidney disease progression: Mechanisms and therapeutic implications. Life Sci 2024; 352:122866. [PMID: 38936605 DOI: 10.1016/j.lfs.2024.122866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024]
Abstract
Chronic kidney disease (CKD) represents a significant and escalating global health challenge, with morbidity and mortality rates rising steadily. Evidence increasingly implicates perirenal adipose tissue (PRAT) deposition as a contributing factor in the pathogenesis of CKD. This review explores how PRAT deposition may exert deleterious effects on renal structure and function. The anatomical proximity of PRAT to the kidneys not only potentially causes mechanical compression but also leads to the dysregulated secretion of adipokines and inflammatory mediators, such as adiponectin, leptin, visfatin, tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and exosomes. Additionally, PRAT deposition may contribute to renal lipotoxicity through elevated levels of free fatty acids (FFA), triglycerides (TAG), diacylglycerol (DAG), and ceramides (Cer). PRAT deposition is also linked to the hyperactivation of the renin-angiotensin-aldosterone system (RAAS), which further exacerbates CKD progression. Recognizing PRAT deposition as an independent risk factor for CKD underscores the potential of targeting PRAT as a novel strategy for the prevention and management of CKD. This review further discusses interventions that could include measuring PRAT thickness to establish a baseline, managing metabolic risk factors that promote its deposition, and inhibiting key PRAT-induced signaling pathways.
Collapse
Affiliation(s)
- Xiang Qiu
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Xin Lan
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Langhui Li
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Huan Chen
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China; Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Ningjuan Zhang
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xiaoli Zheng
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China.
| | - Xiang Xie
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China.
| |
Collapse
|
47
|
Shi W, Gao Y, Yang H, Li H, Liu T, Zhao J, Wei Z, Lin L, Huang Y, Guo Y, Xu A, Bai Z, Xiao X. Bavachinin, a main compound of Psoraleae Fructus, facilitates GSDMD-mediated pyroptosis and causes hepatotoxicity in mice. Chem Biol Interact 2024; 400:111133. [PMID: 38969277 DOI: 10.1016/j.cbi.2024.111133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024]
Abstract
Psoraleae Fructus (PF, Psoralea corylifolia L.), a traditional medicine with a long history of application, is widely used clinically for the treatment of various diseases. However, the reports of PF-related adverse reactions, such as hepatotoxicity, phototoxic dermatitis, and allergy, are increasing year by year, with liver injury being the mostly common. Our previous studies have demonstrated that PF and its preparations can cause liver injury in lipopolysaccharide (LPS)-mediated susceptibility mouse model, but the mechanism of PF-related liver injury is unclear. In this study, we showed that PF and bavachinin, a major component of PF, can directly induce the expression of caspase-1 and interleukin-1β (IL-1β), indicating that PF and bavachinin can directly triggered the activation of inflammasome. Furthermore, pretreatment with NLR family pyrin domain-containing 3 (NLRP3), NLR family CARD domain containing 4 (NLRC4) or absent in melanoma 2 (AIM2) inflammasome inhibitors, containing MCC950, ODN TTAGGG (ODN) and carnosol, all significantly reversed bavachinin-induced inflammasome activation. Mechanistically, bavachinin dose-dependently promote Gasdermin D (GSDMD) post-shear activation and then induce mitochondrial reactive oxygen species (mtROS) production and this effect is markedly inhibited by pretreatment with N-Acetylcysteine amide (NAC). In addition, combination treatment of LPS and bavachinin significantly induced liver injury in mice, but not LPS or bavachinin alone, and transcriptome analysis further validated these results. Thus, PF and bavachinin can induce the activation of inflammasome by promoting GSDMD cleavage and cause hepatotoxicity in mice. Therefore, PF, bavachinin, and PF-related preparations should be avoided in patients with inflammasome activation-associated diseases.
Collapse
Affiliation(s)
- Wei Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China; Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yuan Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Huijie Yang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China; Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hui Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China; Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Tingting Liu
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jia Zhao
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ziying Wei
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Li Lin
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China; Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yujiao Huang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yuming Guo
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Anlong Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.
| | - Zhaofang Bai
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; National Key Laboratory of Kidney Diseases, China.
| | - Xiaohe Xiao
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; National Key Laboratory of Kidney Diseases, China.
| |
Collapse
|
48
|
Li J, Zhang J, Zhao X, Tian L. MSU crystallization promotes fibroblast proliferation and renal fibrosis in diabetic nephropathy via the ROS/SHP2/TGFβ pathway. Sci Rep 2024; 14:20251. [PMID: 39215017 PMCID: PMC11364842 DOI: 10.1038/s41598-024-67324-y] [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: 02/06/2024] [Accepted: 07/10/2024] [Indexed: 09/04/2024] Open
Abstract
Monosodium urate (MSU) crystallisation deposited in local tissues and organs induce inflammatory reactions, resulting in diseases such as gout. MSU has been recognized as a common and prevalent pathology in various clinical conditions. In this study, we investigated the role of MSU in the pathogenesis of diabetic kidney disease (DKD). We induced renal injury in diabetic kidney disease mice using streptozotocin (STZ) and assessed renal histopathological damage using Masson's trichrome staining and Collagen III immunofluorescence staining. We measured the levels of malondialdehyde (MDA), superoxide dismutase (SOD), and uric acid (UA) using ELISA. Protein expression levels of NLRP3, p-NF-κB, SHP2, p-STAT3, and p-ERK1/2 were analyzed by Western blot. To further investigate the role of MSU in diabetic kidney disease, we conducted in vitro experiments. In our in vivo experiments, we found that compared to the Model group, there was a significant increase in interstitial fibrosis in the kidneys of mice after treatment with MSU, accompanied by elevated levels of MDA, SOD, and UA. Furthermore, the protein expression of NLRP3, p-NF-NB, SHP2, p-STAT3, and p-ERK1/2 was upregulated. In our subsequent studies on mouse fibroblasts (L929 cells), we discovered that high glucose, MSU, and TGF-β could promote the expression of P22, GP91, NLRP3, NF-κB, p-NF-κB, p-SHP2, p-EGFR, p-STAT3, and Collagen-III proteins. Additionally, we found that SHP2 could counteract the upregulation trend induced by MSU on the expression of p-SHP2, p-EGFR, p-STAT3, and Collagen-III proteins, and inhibitors YQ128, NAC, and Cetuximab exhibited similar effects. Furthermore, immunofluorescence results indicated that SHP2 could inhibit the expression of the fibrosis marker α-SMA in L929 cells. These findings suggest that MSU can promote renal fibroblast SHP2 expression, induce oxidative stress, activate the NLRP3/NF-κB pathway, and enhance diabetic kidney disease fibroblast proliferation through the TGFβ/STAT3/ERK1/2 signaling pathway, leading to renal fibrosis.
Collapse
Affiliation(s)
- Jing Li
- Department of Nephrology, Affiliated Hospital of Hebei University, 212 Yuhua East Road, Baoding, China
| | - Jiwei Zhang
- Department of Cardiovascular Medicine, Affiliated Hospital of Hebei University, Baoding, China
| | - Xuying Zhao
- Department of Endocrinology, Affiliated Hospital of Hebei University, 212 Yuhua East Road, Baoding, China.
| | - Ling Tian
- Department of Nephrology, Affiliated Hospital of Hebei University, 212 Yuhua East Road, Baoding, China.
| |
Collapse
|
49
|
Dupont N, Terzi F. Lipophagy and Mitophagy in Renal Pathophysiology. Nephron Clin Pract 2024; 149:36-47. [PMID: 39182483 DOI: 10.1159/000540688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/31/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND The lysosomal autophagic pathway plays a fundamental role in cellular and tissue homeostasis, and its deregulation is linked to human pathologies including kidney diseases. Autophagy can randomly degrade cytoplasmic components in a nonselective manner commonly referred to as bulk autophagy. In contrast, selective forms of autophagy specifically target cytoplasmic structures such as organelles and protein aggregates, thereby being important for cellular quality control and organelle homeostasis. SUMMARY Research during the past decades has begun to elucidate the role of selective autophagy in kidney physiology and kidney diseases. KEY MESSAGES In this review, we will summarize the knowledge on lipophagy and mitophagy, two forms of selective autophagy important in renal epithelium homeostasis, and discuss how their deregulations contribute to renal disease progression.
Collapse
Affiliation(s)
- Nicolas Dupont
- NSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades, Université Paris Cité, Paris, France
| | - Fabiola Terzi
- NSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades, Université Paris Cité, Paris, France
| |
Collapse
|
50
|
Zhou W, Yang Y, Feng Z, Zhang Y, Chen Y, Yu T, Wang H. Inhibition of Caspase-1-dependent pyroptosis alleviates myocardial ischemia/reperfusion injury during cardiopulmonary bypass (CPB) in type 2 diabetic rats. Sci Rep 2024; 14:19420. [PMID: 39169211 PMCID: PMC11339408 DOI: 10.1038/s41598-024-70477-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024] Open
Abstract
Cardiovascular complications pose a significant burden in type 2 diabetes mellitus (T2DM), driven by the intricate interplay of chronic hyperglycemia, insulin resistance, and lipid metabolism disturbances. Myocardial ischemia/reperfusion (MI/R) injury during cardiopulmonary bypass (CPB) exacerbates cardiac vulnerability. This study aims to probe the role of Caspase-1-dependent pyroptosis in global ischemia/reperfusion injury among T2DM rats undergoing CPB, elucidating the mechanisms underlying heightened myocardial injury in T2DM. This study established a rat model of T2DM and compared Mean arterial pressure (MAP), heart rate (HR), and hematocrit (Hct) between T2DM and normal rats. Myocardial cell morphology, infarction area, mitochondrial ROS and caspase-1 levels, NLRP3, pro-caspase-1, caspase-1 p10, GSDMD expressions, plasma CK-MB, cTnI, IL-1β, and IL-18 levels were assessed after reperfusion in both T2DM and normal rats. The role of Caspase-1-dependent pyroptosis in myocardial ischemia/reperfusion injury during CPB in T2DM rats was examined using the caspase-1 inhibitor VX-765 and the ROS scavenger NAC. T2DM rats demonstrated impaired glucose tolerance but stable hemodynamics during CPB, while showing heightened vulnerability to MI/R injury. This was marked by substantial lipid deposition, disrupted myocardial fibers, and intensified cellular apoptosis. The activation of caspase-1-mediated pyroptosis and increased reactive oxygen species (ROS) production further contributed to tissue damage and the ensuing inflammatory response. Notably, myocardial injury was mitigated by inhibiting caspase-1 through VX-765, which also attenuated the inflammatory cascade. Likewise, NAC treatment reduced oxidative stress and partially suppressed ROS-mediated caspase-1 activation, resulting in diminished myocardial injury. This study proved that Caspase-1-dependent pyroptosis significantly contributes to the inflammation and injury stemming from global MI/R in T2DM rats under CPB, which correlate with the surplus ROS generated by oxidative stress during reperfusion.
Collapse
Affiliation(s)
- Wenjing Zhou
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, 563000, Guizhou, People's Republic of China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, People's Republic of China
| | - Yingya Yang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, 563000, Guizhou, People's Republic of China
| | - Zhouheng Feng
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, People's Republic of China
| | - Yu Zhang
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, People's Republic of China
| | - Yiman Chen
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, 563000, Guizhou, People's Republic of China
| | - Tian Yu
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, People's Republic of China
| | - Haiying Wang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, 563000, Guizhou, People's Republic of China.
| |
Collapse
|