1
|
Ji L, Chen Y, Chen X. Circular RNA Circ_0002762 promotes cell migration and invasion in cervical squamous cell carcinoma via activating RelA/nuclear factor kappa B (Nf-kB) signalling pathway. RNA Biol 2025; 22:1-13. [PMID: 40083243 PMCID: PMC11934174 DOI: 10.1080/15476286.2025.2478539] [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: 08/25/2024] [Revised: 02/18/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
Cervical cancer is a leading cause of cancer-related deaths, with cervical squamous cell carcinoma (CSCC) accounting for a majority of cases. Circular RNAs (circRNAs) have been repeatedly suggested as crucial effectors in modulating the development of multiple malignancies. The expression of circ_0002762 was predicted to be high in CSCC tissues in GEO dataset, but the functional role and underlying regulatory mechanism of circ_0002762 in CSCC was unclear. By series of functional assays and mechanism assays, supported by bioinformatics analysis, reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR) analysis and western blot assays, we identified that circ_0002762 aberrantly up-regulated in CSCC, promoting CSCC cell migration and invasion. Mechanically, circ_0002762 was transcriptionally activated by Fork head box A1 (FOXA1). Moreover, the involvement of nuclear factor kappa B (NF-kB) signalling in circ_0002762 regulation mechanism in CSCC cells was ascertained. Additionally, circ_0002762, predominantly accumulated in cell cytoplasm, was proved to recruit Mov10 RISC complex RNA helicase (MOV10) to enhance RelA mRNA stability, thus affecting CSCC cell migration and invasion. In summary, FOXA1-mediated circ_0002762 up-regulation could enhance the migratory and invasive abilities of CSCC cells via the MOV10/RelA/NF-kB pathway.
Collapse
Affiliation(s)
- Lei Ji
- Department of Obstetrics and Gynecology, Yancheng First People’s Hospital of Jiangsu Province, Yancheng, Jiangsu, China
| | - Youguo Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaoping Chen
- Department of Obstetrics and Gynecology, Yancheng First People’s Hospital of Jiangsu Province, Yancheng, Jiangsu, China
| |
Collapse
|
2
|
Goncharov AP, Dicusari Elissaiou C, Ben Aharon Farzalla E, Akhvlediani G, Vashakidze N, Kharaishvili G. Signalling pathways in a nutshell: from pathogenesis to therapeutical implications in prostate cancer. Ann Med 2025; 57:2474175. [PMID: 40372974 PMCID: PMC12082737 DOI: 10.1080/07853890.2025.2474175] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 05/17/2025] Open
Abstract
From tumorigenesis to the establishment of local or metastatic high-grade tumours, an integral part of the cellular lifespan relies on various signalling pathways. Particular pathways that allow cells to proliferate by creating a network of new blood vessels have been documented, whereas other pathways are primarily involved with a migration to distant body parts, partially through the process of epithelial-mesenchymal transition (EMT). This review will discuss the different signalling pathways, such as TGF-β, Cripto-1, Wnt pathways, Hedgehog, Notch and NF-κB pathways, and how they promote tumour initiation and progression by influencing diverse cellular processes and EMT in general and in benign and malignant prostate tumours. This review will discuss only the critical pathways. Therefore, many other types of signalling pathways which are related to prostate cancer will not be discussed. Possibilities for further investigation will be mentioned, as many underlying mechanisms involved in these pathways have potential as targets in future tumour therapy. This review will also introduce some novel clinical trials relating to the inhibition of signalling pathways and their clinical outcomes.
Collapse
Affiliation(s)
- Aviv Philip Goncharov
- Department of Clinical and Molecular Pathology, Palacky University, Olomouc, Czech Republic
- Department of Oncology, University Hospital, Olomouc, Czech Republic
| | | | | | - Giorgi Akhvlediani
- Faculty of Medicine, Georgian-American University, Tbilisi, Georgia
- American Hospital in Tbilisi, Tbilisi, Georgia
- Faculty of Medicine, University of Georgia, Tbilisi, Georgia
| | - Nino Vashakidze
- Department of Clinical and Molecular Pathology, Palacky University, Olomouc, Czech Republic
| | - Gvantsa Kharaishvili
- Department of Human Morphology and Pathology, Medical Faculty, David Tvildiani Medical University, Tbilisi, Georgia
- Department of Clinical and Molecular Pathology, University Hospital, Olomouc, Czech Republic
| |
Collapse
|
3
|
Yu L, Tian D, Su Z, Zhang L, Jie L, Guo S, Zhu W, Zhang N, Wang P. Mechanical stress overload promotes NF-κB/NLRP3-mediated osteoarthritis synovitis and fibrosis through Piezo1. Cell Signal 2025; 132:111786. [PMID: 40221068 DOI: 10.1016/j.cellsig.2025.111786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/27/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
Mechanical stress is a pivotal factor in the development of knee osteoarthritis (KOA). Piezo1, an innovative mechanosensitive ion channel, plays a key role in detecting variations in mechanical stress and transforming them into electrical signals. This research focuses on examining how Piezo1 influences synovial inflammation and fibrosis induced by mechanical stress in KOA, as well as delving into the potential underlying mechanisms. In vivo, pathological changes and immunohistochemical staining were conducted on both normal and overexercise rat synovial tissues to analyze the expression of Piezo1 and the NF-κB/NLRP3 pathways. In vitro utilized a cell stretcher to replicate the mechanical conditions seen in KOA. Levels of pro-inflammatory cytokines and fibrosis-related markers were assessed to investigate the impact of Piezo1 on mechanical stress in fibroblast-like synoviocytes (FLS). Subsequently, following cell stretching interventions, the effects on synovial inflammation and fibrosis were observed with the use of the Piezo1 inhibitor GsMTx4 or the NLRP3 inhibitor MCC950. Mechanical stress significantly promoted the activation of Piezo1, increased the phosphorylation ratio of p65, and elevated the levels of NLRP3, caspase-1, ASC, GSDMD, IL-1β, IL-18, IL-6, and TNF-α. Both in vitro and in vivo, mechanical stress also promoted the occurrence and development of synovial fibrosis, with significant increases in the expression levels of fibrosis-related markers. Under mechanical stress overload, upregulation of Piezo1 can promote the secretion of pro-inflammatory cytokines and the fibrotic process in synovium through the NF-κB/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Likai Yu
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Di Tian
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Zishan Su
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Li Zhang
- Orthopedics of traditional Chinese Medicine, Zhongda Hospital Southeast University, Nanjing 210009, Jiangsu, China
| | - Lishi Jie
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Shaobo Guo
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Wenhui Zhu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Nongshan Zhang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China.
| | - Peimin Wang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China.
| |
Collapse
|
4
|
Tan WL, Yu X, Jia J, Chen RY, Xu XX, Liang LQ, Ruan YY, Wang FF, Chen YT, Peng YL, Peng J, Shi M, Tang L, Guo B, Wang YY. Alpha-lipoamide prevents acute kidney injury in mouse by inhibiting renal tubular epithelial cell pyroptosis. Biochem Pharmacol 2025; 237:116942. [PMID: 40228634 DOI: 10.1016/j.bcp.2025.116942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 03/28/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
Acute kidney injury (AKI) is a critical condition marked by a sudden decline in kidney function, frequently resulting in high morbidity and mortality. Renal ischemia-reperfusion injury (IRI) is a leading cause of AKI, characterized by reactive oxygen species (ROS) release, cell death, and inflammation. Alpha-lipoamide (ALM), a neutral derivative of lipoic acid, is recognized for its antioxidant and organ-protective properties. Prior research indicates that ALM mitigates diabetic nephropathy by decreasing ROS. This study examines ALM's protective role in a mouse model of IRI-induced AKI and its mechanisms using mouse renal tubular epithelial cells (mRTECs). Mice were subjected to IRI by renal artery occlusion for 30 min, followed by reperfusion, and treated with ALM (100 or 200 mg/kg) for three days before surgery. In vitro, mRTECs were exposed to hypoxia/reoxygenation injury, with ALM (200 μM) applied to assess oxidative stress. ALM significantly decreased serum creatinine levels, neutrophil gelatinase-associated lipocalin (NGAL), and kidney injury marker-1 (KIM-1), mitigated kidney injury, and reduced both ROS and Malondialdehyde(MDA) content. ALM increased glutathione (GSH) levels and upregulated SIRT1 expression. This resulted in the deacetylation of the NF-κB p65 subunit, facilitating its nuclear export, suppressing NF-κB signaling, and reducing the expression of the inflammatory marker NLRP3. ALM decreased the levels of pyroptosis-related proteins (Caspase-1, GSDMD, and IL-1β), which in turn suppressed IL-6 secretion and macrophage infiltration. These findings suggest that ALM reduces inflammation and pyroptosis-associated proteins by promoting the upregulation of SIRT1, ultimately preventing IRI-mediated renal tubular epithelial cell damage and inflammation.
Collapse
Affiliation(s)
- Wan-Lin Tan
- Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou 561113, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research On Common Chronic Diseases, Guizhou Medical University, Anshun, Guizhou 561113, China
| | - Xiong Yu
- Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou 561113, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research On Common Chronic Diseases, Guizhou Medical University, Anshun, Guizhou 561113, China
| | - Jing Jia
- Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou 561113, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research On Common Chronic Diseases, Guizhou Medical University, Anshun, Guizhou 561113, China
| | - Rong-Yu Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou 561113, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research On Common Chronic Diseases, Guizhou Medical University, Anshun, Guizhou 561113, China
| | - Xiao-Xiao Xu
- Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou 561113, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research On Common Chronic Diseases, Guizhou Medical University, Anshun, Guizhou 561113, China
| | - Lu-Qun Liang
- Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou 561113, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research On Common Chronic Diseases, Guizhou Medical University, Anshun, Guizhou 561113, China
| | - Yuan-Yuan Ruan
- Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou 561113, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research On Common Chronic Diseases, Guizhou Medical University, Anshun, Guizhou 561113, China
| | - Fang-Fang Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou 561113, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research On Common Chronic Diseases, Guizhou Medical University, Anshun, Guizhou 561113, China
| | - Yu-Ting Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou 561113, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research On Common Chronic Diseases, Guizhou Medical University, Anshun, Guizhou 561113, China
| | - Yu-Lin Peng
- Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou 561113, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research On Common Chronic Diseases, Guizhou Medical University, Anshun, Guizhou 561113, China
| | - Jin Peng
- Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou 561113, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research On Common Chronic Diseases, Guizhou Medical University, Anshun, Guizhou 561113, China
| | - Mingjun Shi
- Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou 561113, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research On Common Chronic Diseases, Guizhou Medical University, Anshun, Guizhou 561113, China
| | - Lei Tang
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research On Common Chronic Diseases, Guizhou Medical University, Anshun, Guizhou 561113, China; Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guizhou 550025 China.
| | - Bing Guo
- Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou 561113, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research On Common Chronic Diseases, Guizhou Medical University, Anshun, Guizhou 561113, China.
| | - Yuan-Yuan Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou 561113, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research On Common Chronic Diseases, Guizhou Medical University, Anshun, Guizhou 561113, China.
| |
Collapse
|
5
|
Liu X, Li Y, He M, Zhao Y, Li C, Wang Y, Zhou Q, Peng Y, Zhan L. Multi-bioluminescence based dynamic imaging of Pseudomonas aeruginosa-induced hepatic inflammation process. Microb Pathog 2025; 204:107521. [PMID: 40169074 DOI: 10.1016/j.micpath.2025.107521] [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: 12/10/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/03/2025]
Abstract
Bacterial infections are a major cause of death worldwide. However, it is difficult to track the in vivo dynamics of pathogenic bacteria and the expression of inflammatory factors in infected animals throughout the infection process. This work used Pseudomonas aeruginosa as an infection model and utilised genetically bioluminescence-labeled P. aeruginosa and hydrodynamic transfection technology to construct a liver-visual NF-κB, IL-6, TNF-α inflammation model, thereby enabling the tracking of the dynamic spread of P. aeruginosa in infected animals and the transient activation of the liver inflammation response. The results showed that P. aeruginosa introduced via the tail vein initially accumulates in the liver and gradually activates NF-κB, IL-6, and TNF-α. Subsequently, the P. aeruginosa infection gradually spreads to the lungs and small intestine, and final proliferation leads to septic death in mice. During the infection process, we observed a strictly negative correlation between platelet activation and bacterial proliferation; the higher the degree of platelet activation, the stronger the inhibitory effect on bacterial proliferation and liver inflammation. In conclusion, this bioluminescence-based in vivo imaging technique offers new opportunities to investigate the innate immune response in controlling pathogenic infections.
Collapse
Affiliation(s)
- Xingzhao Liu
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, People's Republic of China
| | - Yipu Li
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
| | - Minwei He
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, People's Republic of China
| | - Yan Zhao
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, People's Republic of China
| | - Chenyan Li
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, People's Republic of China
| | - Yi Wang
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, People's Republic of China
| | - Qianqian Zhou
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, People's Republic of China.
| | - Ying Peng
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China.
| | - Linsheng Zhan
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, People's Republic of China.
| |
Collapse
|
6
|
Zhang Q, Zhou Q, Li H. Action and mechanisms of neferine in inflammatory diseases (Review). Mol Med Rep 2025; 32:174. [PMID: 40242976 PMCID: PMC12046375 DOI: 10.3892/mmr.2025.13539] [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: 09/28/2024] [Accepted: 02/17/2025] [Indexed: 04/18/2025] Open
Abstract
Neferine is a bisbenzylisoquinoline alkaloid derived from the seed embryo of Nelumbo nucifera, a traditional Chinese medicine. It has been extensively studied for its therapeutic potential in various disease models. Extensive research has highlighted its diverse pharmacological activities, including antitumor, anti‑inflammatory, anti‑fibrosis, anti‑oxidative stress, anti‑platelet aggregation and anti‑arrhythmic effects. The present review, however, focuses on the anti‑inflammatory properties of neferine, emphasizing its fundamental mechanisms as demonstrated in both in vivo and in vitro studies. By critically evaluating its effect on inflammation and the underlying pathways, this review aims to provide a comprehensive understanding of the potential of neferine in the management of inflammatory diseases. Furthermore, it seeks to establish a foundational framework for the future development of neferine as a novel therapeutic agent for inflammatory conditions.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Nephropathy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400021, P.R. China
| | - Qiaoling Zhou
- Department of Nephropathy, Xiangya Hospital Central-South University, Changsha, Hunan 410028, P.R. China
| | - Huihui Li
- Department of Nephropathy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400021, P.R. China
| |
Collapse
|
7
|
Zhu Y, Wang Q, Zheng Y, Chen N, Kou L, Yao Q. Microenvironment responsive nanomedicine for acute pancreatitis treatment. Colloids Surf B Biointerfaces 2025; 251:114633. [PMID: 40112593 DOI: 10.1016/j.colsurfb.2025.114633] [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: 11/08/2024] [Revised: 02/26/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Acute pancreatitis (AP) is an acute inflammation of the pancreas, which is considered a prevalent gastrointestinal emergency characterized by rapid progression and significant mortality. Currently available medications primarily serve as adjunctive therapies, yielding suboptimal therapeutic outcomes. Consequently, there remains a dearth of specific and efficient treatment modalities for AP. In recent years, nanomedicine-based treatment strategies have exhibited significant potential as drug therapy approaches for pancreatitis. The distinctive features of the AP microenvironment encompass aberrant activation of pancreatic enzymes, oxidative stress induced by elevated reactive oxygen species levels, and excessive production of pro-inflammatory cytokines; these factors offer promising targeted sites for early diagnosis and treatment using nanomedicine. This article comprehensively delineates the pathological microenvironmental characteristics associated with AP while highlighting the application of microenvironment-responsive strategies in nanodrug delivery systems for its treatment, thereby providing insights into future prospects.
Collapse
Affiliation(s)
- Yixuan Zhu
- Wenzhou Municipal KeyLaboratory of Pediatric Pharmacy, Department of Pharmacy, The Second AffiliatedHospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Qian Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yaoyao Zheng
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Nuo Chen
- Wenzhou Municipal KeyLaboratory of Pediatric Pharmacy, Department of Pharmacy, The Second AffiliatedHospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325035, China
| | - Longfa Kou
- Wenzhou Municipal KeyLaboratory of Pediatric Pharmacy, Department of Pharmacy, The Second AffiliatedHospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| | - Qing Yao
- Wenzhou Municipal KeyLaboratory of Pediatric Pharmacy, Department of Pharmacy, The Second AffiliatedHospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
8
|
Liu H, Xue H, Guo Q, Xue X, Yang L, Zhao K, Liu Y. Ferroptosis meets inflammation: A new frontier in cancer therapy. Cancer Lett 2025; 620:217696. [PMID: 40189012 DOI: 10.1016/j.canlet.2025.217696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, has emerged as a critical player in cancer pathogenesis. Concurrently, inflammation, a key biological response to tissue injury or infection, significantly influences cancer development and progression. The interplay between ferroptosis and inflammation represents a promising yet underexplored area of research. This review synthesizes recent advances in understanding the molecular mechanisms governing their interaction, emphasizing how ferroptosis triggers inflammatory responses and how inflammatory mediators, such as TNF-α, regulate ferroptosis through iron metabolism and lipid peroxidation pathways. Key molecular targets within the ferroptosis-inflammation axis, including GPX4, ACSL4, and the NF-κB signaling pathway, offer therapeutic potential for cancer treatment. By modulating these targets, it may be possible to enhance ferroptosis and fine-tune inflammatory responses, thereby improving therapeutic outcomes. Additionally, this review explores the broader implications of targeting the ferroptosis-inflammation interplay in disease treatment, highlighting opportunities for developing innovative strategies to combat cancer. By bridging the gap in current knowledge, this review provides a comprehensive resource for researchers and clinicians, offering insights into the therapeutic potential of this intricate biological relationship.
Collapse
Affiliation(s)
- Hu Liu
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Hui Xue
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Qian Guo
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xutong Xue
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Lixue Yang
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China.
| | - Kaijun Zhao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Yu'e Liu
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA; Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
9
|
Xu Y, Zhang X, Zhang Y, Ma H, Zhou Z, Qin H, Liu H, Han X. Integrated multi-omics insight into the molecular networks of oxidative stress in triggering multiple sclerosis. Neurobiol Dis 2025; 210:106929. [PMID: 40280189 DOI: 10.1016/j.nbd.2025.106929] [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: 11/06/2024] [Revised: 04/22/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025] Open
Abstract
Oxidative stress (OS) is a key pathophysiological mechanism in multiple sclerosis (MS). However, the underlying mechanisms by which OS triggered MS remain unknown. To identify potential causal targets of 1216 OS-related genes for MS, a summary-data-based Mendelian randomization (SMR) method was applied. Given that genes can exert their biological functions through different omics levels, the multi-omics SMR integrating expression, methylation, and protein quantitative trait loci (eQTL, mQTL, and pQTL) of OS-related genes from blood and brain tissues was utilized. Bayesian colocalization test was conducted to examine potential regulatory mechanisms of QTL risk variation in MS. To verify the robustness of our results, we validated these findings in FinnGen cohort. Furthermore, the QTL evidence levels, colocalization findings, and replication cohort results were integrated and potential target genes were categorized into three levels. Consequently, three genes (BACH2, TRAF3, and MAPK3) were identified as potential contributors to MS in blood, and four genes (HMGCL, TSFM, TRAF3 and HLA-B) were identified as potential contributors to MS in brain tissue. Additionally, HMGCL and TSFM from brain tissue were supported by first-level evidence related to MS and were validated via in vitro experiments. This research not only contributed to fundamental research of OS in MS but also supported the identification of potential targets for clinical interventions in MS.
Collapse
Affiliation(s)
- Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xiaowei Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hongxuan Ma
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhaokai Zhou
- Department of Urology Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hongzhuo Qin
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Huimin Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China.
| |
Collapse
|
10
|
Mohamed AR, Georgey HH, Aidy EA, Al-Shafie TA, Elnagar MR, Ali MA, Elblehi SS, Alzahrani AYA, Mousa MHA. Unveiling the potential of xanthines, discovery of potential 7-benzyl-1,3-dimethyl-3,7-dihydro-1H-purine-2,6-dione derivatives with antifibrotic activity for liver injury. Bioorg Chem 2025; 160:108441. [PMID: 40199013 DOI: 10.1016/j.bioorg.2025.108441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/21/2025] [Accepted: 04/02/2025] [Indexed: 04/10/2025]
Abstract
A new series of xanthine-based derivatives were designed, synthesized, and investigated to achieve promising antifibrotic and antioxidant agents for management of liver injury. Structure-based optimizations of the methylxanthine-based KMUP-1 (IX) were performed for inhibiting NF-κB activation pathway. All the newly designed xanthine derivatives 3, 4, 5, 6a-d, 7a-d, and 9a-d were in vitro screened for the antioxidant activity using the DPPH method. Compounds 4 and 5 showed the highest antioxidant activity with an IC50 of 28.02 and 36.02 μM, respectively. Compounds 9c and 9d retained a promising interception of the NF-κB activation pathway in molecular docking simulations within I-κB kinase α (IKKα) crystal structure (PDB ID: 5EBZ). Subsequently, compounds 9c and 9d were evaluated for their in vivo antifibrotic and chemoprotective activity using CCl4-induced hepatic fibrosis rat model. Compounds 9c and 9d successfully ameliorated liver fibrosis, as evidenced by the improved liver histopathological examination and liver enzyme activity levels. Compounds 9c and 9d were evaluated for their effects on mRNA expression levels of key genes involved in liver fibrosis via real-time PCR assays. Compound 9c exhibited a greater inhibitory effect on the expression levels of NF-κB and HIF-1α and a more pronounced stimulation of Nrf2 than compound 9d. Moreover, all the new xanthine derivatives were screened for the cytotoxic activity against the NCI tumor cell lines. Compounds 9c and 9d revealed a non-significant cytotoxic activity against all the assayed tumor cell lines, which indicate their selectivity for the antifibrotic activity. While compounds 6a and 6c displayed promising selective activity against melanoma SK-MEL-5 cell line (GI = 125.6, 90.3 %, respectively), and breast T-47D cell line (GI =87.8, 80.6 %, respectively). The utilized design approach unveiled the versatility of xanthine scaffold to deliver potential antioxidant, liver antifibrotic and chemoprotective agents, along with anticancer candidates via structure modification and optimization.
Collapse
Affiliation(s)
- Abdalla R Mohamed
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo 11829, Egypt.
| | - Hanan H Georgey
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Esraa A Aidy
- Medical Biochemistry and Molecular Biology Unit, Cancer Biology Department, National Cancer Institute (NCI), Cairo University, Cairo 11796, Egypt
| | - Tamer A Al-Shafie
- Department of Oral Biology (Biochemistry), Faculty of Dentistry, Pharos University in Alexandria (PUA), Alexandria 21648, Egypt
| | - Mohamed R Elnagar
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Cairo 11823, Egypt; Department of Pharmacology, College of Pharmacy, The Islamic University, Najaf 54001, Iraq
| | - Mennatallah A Ali
- Pharmacology and Toxicology Department, PharmD Program, Egypt-Japan University of Science and Technology (E-JUST), Alexandria 21934, Egypt
| | - Samar S Elblehi
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Alexandria 22758, Egypt
| | - Abdullah Y A Alzahrani
- Department of Chemistry, Faculty of Science, King Khalid University, Mohail Assir 61421, Saudi Arabia
| | - Mai H A Mousa
- Pharmaceutical Chemistry Department, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo 19346, Egypt
| |
Collapse
|
11
|
Jin X, Zhang L, Ying C, Yu L, Guo X, Pan K, Zhu D, Chen H. S-adenosylmethionine inhibits non-small cell lung cancer and enhances chemosensitivity by targeting the P62/NF-κB axis and regulating autophagy and oxidative stress. Bioorg Chem 2025; 160:108509. [PMID: 40280015 DOI: 10.1016/j.bioorg.2025.108509] [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: 01/06/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
Non-small cell lung cancer (NSCLC) is a leading cause of cancer-related deaths worldwide. Despite advances in targeted therapies and immunotherapy, which have improved survival rates, drug resistance and immune-related side effects continue to necessitate the development of new treatments. S-adenosylmethionine (SAM), a key metabolite in the methionine cycle, has indicated potential for cancer therapy and enhancing chemotherapy sensitivity. However, its effects on NSCLC remain undetermined. In our study, SAM inhibits NSCLC growth and enhances chemosensitivity both in vitro and in vivo. Mechanistic investigations revealed that SAM plays a significant regulatory role in autophagy and oxidative stress within NSCLC. Furthermore, we identified P62 as a critical target of SAM by constructing biotin-labeled SAM for immunocoprecipitation-mass spectrometry. Both in vitro and in vivo studies confirmed that P62 mediates SAM regulatory effects on NSCLC. Furthermore, by constructing truncated P62 expression plasmids for immunocoprecipitation experiments, we discovered that SAM inhibits the NF-κB signaling pathway by directly targeting the ZZ and TB domains of the P62 protein, thereby blocking autophagy and activating oxidative stress. These findings highlight SAM as a novel inhibitor of the P62/NF-κB axis and suggest that SAM could be a potential therapeutic agent for NSCLC.
Collapse
Affiliation(s)
- Xuehang Jin
- Department of Respiratory and Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, 321000 Jinhua, Zhejiang Province, China
| | - Lvjun Zhang
- Department of Respiratory and Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, 321000 Jinhua, Zhejiang Province, China
| | - Chiqing Ying
- Department of Respiratory and Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, 321000 Jinhua, Zhejiang Province, China
| | - Long Yu
- Department of Respiratory and Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, 321000 Jinhua, Zhejiang Province, China
| | - Xuejing Guo
- Department of Respiratory and Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, 321000 Jinhua, Zhejiang Province, China
| | - Kailing Pan
- Department of Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Dan Zhu
- Department of Respiratory and Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, 321000 Jinhua, Zhejiang Province, China.
| | - Hui Chen
- Department of Respiratory and Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, 321000 Jinhua, Zhejiang Province, China.
| |
Collapse
|
12
|
Liu M, Zou G, Lu M, Fu J, Chen H, Pan C, Liu HM, Fu L. Mechanism of Rabdosia rubescens extract against gastric cancer microenvironment by SIRT1/NF-κB/p53 pathway and promoting tumor-associated macrophage polarization. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119935. [PMID: 40345273 DOI: 10.1016/j.jep.2025.119935] [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: 02/07/2025] [Revised: 04/23/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The traditional action of Rabdosia rubescens (Hemsl.) H. Hara is heat-clearing and detoxifying, relieve sore throat, dissipate binds and disperse swelling. DLC, as an extract prepared from Rabdosiae Rubescentis Herba, could regulate the polarization of tumor associated macrophages (TAMs). For TAMs play an important role in the tumor microenvironment. It is worthy to further explore the mechanism of DLC on the polarized function of macrophages. AIM OF THE STUDY The aim of this study is to investigate the activity and molecular mechanisms of DLC on dissipating binds and dispersing swelling by modulating the gastric cancer microenvironment and macrophage polarization. MATERIALS AND METHODS We conducted comprehensive qualitative and quantitative chromatographic analyses to characterize the main components of DLC. To evaluate its anti-tumor effects, immunofluorescence, MTT assay, plate cloning, transcriptomics analysis, western blotting, and siRNA knockdown experiments were performed to assess DLC's action on gastric cancer cell proliferation. Additionally, we utilized Trypan blue staining, a THP-1 and MGC-803 co-culture model, flow cytometry, enzyme-linked immunosorbent assay (ELISA), and a mouse xenograft model with five distinct dosage groups to systematically investigate DLC's effects on macrophage polarization. RESULTS Key compounds in DLC were identified. The vivo tests demonstrated the tumor inhibition rate of the 5 g/kg DLC group reached 66.99 %, surpassing that of the 5-fluorouracil group (59.94 %). Mechanistically, DLC upregulated SIRT1 expression and suppressed NF-κB pathway, thereby preventing p65 from translocating into nuclear and modulating downstream p53/MDM2/USP7 signaling. Moreover, DLC enhanced M1 macrophage factors such as TNF-α, IL-6 while inhibiting M2 marker TGF-β, effectively repolarizing M2 TAMs toward an M1 phenotype. This effect was associated with suppressed protein expression of HIF-1α, p-p65, and p-PI3K. CONCLUSION This study provides insights into DLC's mechanisms in regulating tumor microenvironment remodeling and promoting macrophage polarization toward an anti-tumor phenotype. These results provide a solid basis for DLC's potential clinical treament in gastric cancer, highlighting its promise as a natural therapeutic agent.
Collapse
Affiliation(s)
- Mengran Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Guona Zou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Mengyao Lu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Jiayue Fu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Han Chen
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Chengxue Pan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Hong-Min Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China.
| | - Ling Fu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
13
|
Wei R, Zeng Z, Chen S, Shi Y, Ding Q. Qingfei Litan decoction alleviated Klebsiella pneumoniae-induced pneumonia by targeting the TLR4/MyD88/NF-κB axis via miR-146a-5p. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119947. [PMID: 40378935 DOI: 10.1016/j.jep.2025.119947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/19/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Klebsiella pneumoniae (Kp) is a significant pathogen responsible for various clinical bacterial infections, including pneumonia, sepsis, and even death. However, effective treatment options remain limited due to the rising prevalence of antimicrobial resistance. Traditional Chinese medicine (TCM) has shown potential in the treatment of bacterial pneumonia. Qingfei Litan decoction (QFLT) has been reported to alleviate symptoms in patients with bacterial pneumonia, though its precise mechanisms in regulating pulmonary inflammation remain unclear. AIM OF THE STUDY This study aimed to investigate the therapeutic potential of QFLT in Kp-induced pneumonia and to elucidate its underlying molecular mechanisms. MATERIAL AND METHODS In vivo, a murine pneumonia model was established through intratracheal instillation of Kp, and QFLT was administered by oral gavage. miR-146a-5p expression was downregulated by tail vein injection of an antagomir. The therapeutic effects of QFLT on pulmonary pathology, inflammatory factors, and miR-146a-5p expression were evaluated using qRT-PCR, flow cytometry, and other methods. Bioinformatics tools were employed to predict miR-146a-5p targets and associated inflammatory pathways. In vitro, an alveolar macrophage inflammation model was established by stimulating MH-S cells with heat-inactivated Klebsiella pneumoniae (iKp), followed by QFLT treatment. Inhibition of miR-146a-5p was achieved through transfection with specific inhibitors. The effects of QFLT on inflammatory responses, miR-146a-5p expression, and TLR4/MyD88/NF-κB signaling were assessed using qRT-PCR, Western blotting (WB) and other methods. RESULTS Kp infection significantly exacerbated pulmonary inflammation and downregulated miR-146a-5p expression in both lung tissues and MH-S cells. QFLT treatment alleviated inflammatory responses and upregulated miR-146a-5p expression. Bioinformatics analysis demonstrated that miR-146a-5p targeted TRAF6, a key mediator of the TLR4/MyD88/NF-κB pathway. Western blot analysis further confirmed that QFLT reduced Kp-induced upregulation of the TLR4/MyD88/NF-κB pathway in MH-S cells. Moreover, inhibition of miR-146a-5p exacerbated inflammatory responses in both lung tissues and MH-S cells, whereas QFLT treatment effectively attenuated these inflammatory effects. Furthermore, miR-146a-5p suppression resulted in elevated expression of proteins in the TLR4/MyD88/NF-κB signaling pathway in MH-S cells, while QFLT administration significantly reduced the expression levels of these signaling components. CONCLUSIONS These findings demonstrated that QFLT ameliorated Kp-induced pneumonia by modulating the TLR4/MyD88/NF-κB axis via miR-146a-5p.
Collapse
Affiliation(s)
- Ruojun Wei
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, China.
| | - Ziyuan Zeng
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, China.
| | - Sheng Chen
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China.
| | - Yuanyuan Shi
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, China; Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China.
| | - Qi Ding
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, China.
| |
Collapse
|
14
|
Gu T, Raval R, Bashkin Z, Zhou C, Ko S, Kong N, Hong S, Bhaskara A, Shah S, Joshi A, Thellakal S, Rim K, Marimuthu A, Venkatesan S, Wang E, Li S, Jayabalan A, Tao A, Fang Y, Xia L, Chui A, Shu E, Zhang T, Chen Z, Njoo E. Synthesis, antiproliferative activity, and biological profiling of C-19 trityl and silyl ether andrographolide analogs in colon cancer and breast cancer cells. Bioorg Med Chem Lett 2025; 121:130163. [PMID: 40043819 DOI: 10.1016/j.bmcl.2025.130163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/08/2025] [Accepted: 02/28/2025] [Indexed: 03/10/2025]
Abstract
Andrographolide, a labdane diterpenoid isolated from Andrographis paniculata, putatively functions through covalent inhibition of NF-κB, a transcription factor that modulates tumor survival and metastasis. Previous studies have found that functionalization of the C-19 hydroxyl alters the primary mode of action from inhibition of NF-κB to the modulation of the Wnt1/β-catenin signaling pathway. Here, we synthesized a series of twelve C-19 trityl and silyl ether analogs, including three novel substituted trityl analogs and four novel substituted silyl analogs of andrographolide. MTT assays revealed cell line selectivity between colorectal and breast cancer cells, which is consistent with known mechanisms of β-catenin-driven cell proliferation in colorectal cancer cell lines. Most compounds exhibited cell line specific antiproliferative activity in HCT-116 and HT-29 colorectal cancer cell lines. Specifically, within 24 h, C-19 analogs of andrographolide exhibit far more limited antiproliferative activity in MCF-7 breast cancer cells compared to HCT-116, HT-29, and MDA-MB-231 cells. Through in vitro TNF-α-dependent NF-κB reporter and Wnt1-dependent luciferase reporter assays, we observed that several analogs generally exhibit greater inhibitory activity compared to andrographolide. Fluorescence imaging demonstrated that cells treated with andrographolide and its C-19 analogs retained similar distributions of active β-catenin, but notable differences in antiproliferative potency upon co-delivery with GSK-3β inhibitor CHIR99021 indicate that several lead compounds exhibit attenuated biological activity selectively in HT-29 cells. Collectively, this work indicates that modest structural modifications at C-19 of andrographolide can have profound implications for its biological activity in mechanisms connected to its anticancer activity.
Collapse
Affiliation(s)
- Tiffany Gu
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Rushika Raval
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Zachary Bashkin
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Carina Zhou
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Sanghyuk Ko
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Natalie Kong
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Seoyeon Hong
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Aditya Bhaskara
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Samarth Shah
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Aditi Joshi
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Samahith Thellakal
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Kaitlyn Rim
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Anushree Marimuthu
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Srishti Venkatesan
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Emma Wang
- Department of Computer Science & Engineering, Aspiring Scholars Directed Research Program, USA
| | - Sophia Li
- Department of Computer Science & Engineering, Aspiring Scholars Directed Research Program, USA
| | - Aditi Jayabalan
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Alice Tao
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Yilin Fang
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Lorelei Xia
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Aidan Chui
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Emily Shu
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Tracy Zhang
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Zhan Chen
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Edward Njoo
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA.
| |
Collapse
|
15
|
Saamarthy K, Daams R, Sime W, Persson C, Chygorin E, Ahlqvist K, Evans-Axelsson S, Strand D, Massoumi R. An optimised Bcl-3 inhibitor for melanoma treatment. Br J Pharmacol 2025; 182:2426-2446. [PMID: 39943627 DOI: 10.1111/bph.17467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND AND PURPOSE Malignant melanoma is the most lethal form of skin cancer, characterised by a poor survival rate. One of the key factors driving the aggressive growth of melanoma cells is the elevated expression of the proto-oncogene Bcl-3. This study aims to optimise, evaluate and characterise a second-generation Bcl-3 inhibitor, using melanoma as a model to demonstrate its potential therapeutic efficacy. EXPERIMENTAL APPROACH We synthesised and screened a series of structural analogues and selected A27, the most promising candidate for further investigation. We assessed whether A27 disrupted the interaction between Bcl-3 and its binding partner, p50, and examined the subsequent effects on cyclin D1 expression. Additionally, we evaluated the impact of A27 on melanoma cell proliferation and migration in vitro, as well as its therapeutic efficacy in various in vivo melanoma models. KEY RESULTS Nuclear magnetic resonance (NMR) confirmed that A27 directly binds to Bcl-3, effectively inhibiting its function. By disrupting the Bcl-3/p50 interaction, A27 led to a significant down-regulation of cyclin D1 expression. In cellular assays, A27 markedly reduced proliferation and migration of melanoma cells. In vivo, treatment with A27 resulted in a substantial reduction in melanoma tumour growth, with no observed toxicity in treated animals. CONCLUSIONS AND IMPLICATIONS At present, no other Bcl-3 inhibitors exist for clinical application in the field of oncology, and as a result, our novel findings provide a unique opportunity to develop a highly specific drug against malignant melanoma to meet an urgent clinical need.
Collapse
Affiliation(s)
- Karunakar Saamarthy
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| | - Renée Daams
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| | - Wondossen Sime
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| | - Cecilia Persson
- Swedish NMR Center, Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Eduard Chygorin
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Lund, Sweden
| | - Kristofer Ahlqvist
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| | - Susan Evans-Axelsson
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| | - Daniel Strand
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Lund, Sweden
| | - Ramin Massoumi
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| |
Collapse
|
16
|
Oliveira MDS, Dias IRSB, Costa RGA, Rodrigues ACBDC, Silva SLR, Soares MBP, Dias RB, Valverde LF, Gurgel Rocha CA, Batista AA, Correa RS, Silva VR, Granado Pina ET, Bezerra DP. Ru(II)-thymine complex suppresses acute myeloid leukemia stem cells by inhibiting NF-κB signaling. Biomed Pharmacother 2025; 187:118080. [PMID: 40288174 DOI: 10.1016/j.biopha.2025.118080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 04/02/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
Acute myeloid leukemia (AML) is a lethal hematologic malignancy caused by leukemic blasts that fail to mature normally. AML has a high relapse rate, primarily due to a small subset known as leukemic stem cells (LSCs). In this work, we investigated the ability of a Ru(II)-thymine complex (RTC) with the formula [Ru(PPh3)2(Thy)(bipy)]PF6 (where PPh3 = triphenylphosphine, Thy = thymine, and bipy = 2,2'-bipyridine) to suppress AML LSCs. RTC exhibited potent cytotoxicity toward both solid and hematologic malignancies and suppressed primary AML LSCs, as observed by the reduction in the CD34 +CD38- cell population. In the AML cell line KG-1a, which has an LSC-like population, RTC reduced the number of CD34 + and CD123 + cells. A reduction in leukemic blasts was detected in the bone marrow of RTC-treated NSG mice bearing KG-1a xenografts. Increased DNA fragmentation, YO-PRO-1 staining, active caspase-3 and cleaved PARP (Asp 214) levels, and mitochondrial superoxide levels were detected in RTC-treated KG-1a cells. The pancaspase inhibitor Z-VAD-(OMe)-FMK, but not the antioxidant N-acetylcysteine, partially prevented RTC-induced cell death in KG-1a cells, indicating that RTC induces caspase-mediated apoptosis in KG-1a cells via an oxidative stress-independent pathway. In molecular mechanism studies, transcripts of the NF-κB inhibitor NFKBIA were upregulated, and the level of NF-κB p65 phosphorylated at the Ser529 residue was reduced in RTC-treated KG-1a cells, indicating that RTC may inhibit NF-κB signaling. Overall, these results indicate the anti-AML potential of RTC in AML LSCs via the suppression of NF-κB signaling.
Collapse
Affiliation(s)
- Maiara de S Oliveira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil
| | - Ingrid R S B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil
| | - Rafaela G A Costa
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil
| | | | - Suellen L R Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil
| | - Milena B P Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil; SENAI Institute for Innovation in Advanced Health Systems, SENAI CIMATEC, Salvador, BA 41650-010, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil; Department of Biological Sciences, State University of Feira de Santana, Feira de Santana, BA 44036-900, Brazil
| | - Ludmila F Valverde
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil; Department of Dentistry, Federal University of Sergipe, Lagarto, SE 49400-000, Brazil
| | - Clarissa A Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil; Department of Propaedeutics, Faculty of Dentistry, Federal University of Bahia (UFBA), Salvador, BA 40301-155, Brazil
| | - Alzir A Batista
- Department of Chemistry, Federal University of São Carlos, São Carlos, SP 13561-901, Brazil
| | - Rodrigo S Correa
- Department of Chemistry, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - Valdenizia R Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil
| | - Eugênia T Granado Pina
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil; Research Center, National Cancer Institute (INCA), Rio de Janeiro, RJ 20230-130, Brazil
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, BA 40296-710, Brazil.
| |
Collapse
|
17
|
Zhu P, Ren Y, Wei C, Luo J, Wu D, Ye X, Donlao N, Tian J. Compounds from sea buckthorn and their application in food: A review. Food Chem 2025; 476:143428. [PMID: 39986072 DOI: 10.1016/j.foodchem.2025.143428] [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: 08/24/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/24/2025]
Abstract
Sea buckthorn is a fruit rich in many bioactive compounds and shows the benefits of antioxidant, anti-inflammatory, anti-obesity, hepatoprotective, anti-tumor, and immunomodulatory properties, etc. The main bioactive compounds extracted and characterized in sea buckthorn are polyphenols, carotenoids, and functional lipids, which could provide health benefits by scavenging free radicals, regulating enzyme activities, and modulating signaling pathways, etc. Although there are many studies focused on the values of sea buckthorn, a comprehensive review on its chemical composition, functional mechanism and food application are still lacking. Thus, this paper aims to review the bioactive compounds in sea buckthorn, their underlying mechanisms for health benefits, as well as the applications in health food development. Particularly, the potential value of sea buckthorn and the novel technologies applied in previous studies are also discussed to improve its use for human health.
Collapse
Affiliation(s)
- Peiyi Zhu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China; Zhejiang University Zhongyuan Institute, Zhengzhou 450000, China
| | - Yanming Ren
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China; Zhejiang University Zhongyuan Institute, Zhengzhou 450000, China
| | - Changqing Wei
- Food College, Shihezi University, Shihezi 832000, China
| | - Jingjing Luo
- Agricultural Science Research Institute of Baiyang City, Tacheng 834601, China
| | - Dan Wu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China
| | - Xingqian Ye
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China; Zhejiang University Zhongyuan Institute, Zhengzhou 450000, China
| | - Natthawuddhi Donlao
- School of Agro-Industry, Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Jinhu Tian
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China; Zhejiang University Zhongyuan Institute, Zhengzhou 450000, China; Zhejiang University-Wuxi Xishan Modern Agriculture Joint Research Center, Wuxi 214117, China.
| |
Collapse
|
18
|
Wu X, Chen Y, He W, Yao Y, Liu Y, Xia P, Zhang H, Li X, Guo Y, Chen X, Ma W, Yuan Y. UBE2Q2 promotes tumor progression and glycolysis of hepatocellular carcinoma through NF-κB/HIF1α signal pathway. Cell Oncol (Dordr) 2025; 48:637-654. [PMID: 39833608 PMCID: PMC12119716 DOI: 10.1007/s13402-025-01037-w] [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] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
PURPOSE Metabolic reprogramming, particularly the Warburg effect, plays a crucial role in the onset and progression of tumors. The ubiquitin-conjugating enzyme E2 Q2 (UBE2Q2) has been identified overexpressed in hepatocellular carcinoma (HCC). Our aim was to determine if UBE2Q2 plays a role in regulating glycolysis, contributing to the carcinogenesis of HCC. METHODS Bioinformatics analysis, western blot and qPCR were used to detect the expression of UBE2Q2. Functional experiments, proteomics analysis and subcutaneous tumors were constructed to find the biological function of UBE2Q2 in HCC. Co-immunoprecipitation, western blot and ubiquitination assays were used to identify the mechanisms involved. RESULTS We found a significant association between high UBE2Q2 expression and poor prognosis in HCC patients. Functionally, UBE2Q2 was shown to advance tumor progression in HCC through both in vitro assays and in vivo assessments. Proteomics analysis and glycolysis stress tests corroborated an increase in glycolytic activity due to UBE2Q2. Our findings reveal that UBE2Q2 augments glycolysis by boosting the transcription levels of hypoxia-inducible factor 1α (HIF1α), primarily through the activation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway. At the molecular level, UBE2Q2 interaction with baculoviral IAP repeat-containing 2 (cIAP1) orchestrates the K63-linked ubiquitination of receptor-interacting serine/threonine-protein kinase 1 (RIP1), which in turn, activates the NF-κB signaling pathway. CONCLUSIONS Our investigation reveals that UBE2Q2 regulates the glycolysis in HCC through modulation of the NF-κB/HIF1α signaling pathway, pinpointing UBE2Q2 as a promising therapeutic target for the disease.
Collapse
Affiliation(s)
- Xiaoling Wu
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei, 430071, PR China
- Department of Liver Surgery, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Yiran Chen
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei, 430071, PR China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
| | - Wenzhi He
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei, 430071, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, 430071, PR China
| | - Ye Yao
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei, 430071, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, 430071, PR China
| | - Yingyi Liu
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei, 430071, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, 430071, PR China
| | - Peng Xia
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei, 430071, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, 430071, PR China
| | - Hao Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei, 430071, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, 430071, PR China
| | - Xiaomian Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei, 430071, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, 430071, PR China
| | - Yonghua Guo
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei, 430071, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, 430071, PR China
| | - Xi Chen
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei, 430071, PR China.
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, 430071, PR China.
| | - Weijie Ma
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei, 430071, PR China.
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, 430071, PR China.
| | - Yufeng Yuan
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei, 430071, PR China.
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, 430071, PR China.
| |
Collapse
|
19
|
Mannan A, Mohan M, Singh TG. Revenge unraveling the fortress: Exploring anticancer drug resistance mechanisms in BC for enhanced therapeutic strategies. Crit Rev Oncol Hematol 2025; 210:104707. [PMID: 40122355 DOI: 10.1016/j.critrevonc.2025.104707] [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: 01/22/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025] Open
Abstract
Breast cancer (BC) is the most prevalent form of cancer in women worldwide and the main cause of cancer-related fatalities in females. BC can be classified into various types based on where cancer has begun to grow or spread, specific characteristics that influence how cancer behaves, and treatment choices. BC is multifaceted, and due to its diverse nature, the mechanisms involved are complex and have not yet been understood. Overexpression and expression of various factors involved in the functioning of mechanisms lead to abnormal changes, providing an environment supporting cancer cell growth. Understanding BC risk factors and early diagnosis through screening techniques like mammography and diagnostic techniques such as imaging and biopsies has advanced significantly. A wide range of treatment options, including surgery, radiation, chemotherapy, targeted treatments, and hormonal therapies, are now available. Daily advancements are being made in the clinical treatment of BC. Still, BC drug resistance cases remain highly prevalent and are currently one of the biggest problems faced by medical science. To increase response rates and possibly lengthen survival, there is a critical requirement for novel medicines with minimal sensitivity to overcome drug resistance. This review classifies different mechanisms that are involved in the development of BC and workable pharmacological targets and explains how they relate to the development of BC drug resistance. By concentrating on the mechanisms covered in this review, we can have a deep understanding of different mechanisms and learn innovative ways to develop novel therapeutics for the disease to combat medication resistance.
Collapse
Affiliation(s)
- Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Maneesh Mohan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
20
|
Zhu YS, Shah SAA, Yang BY, Fan SS, He L, Sun YR, Shang WB, Qian Y, Zhang X. Gen-17, a beta-methyl derivative of Genipin, attenuates LPS-induced ALI by regulating Keap1-Nrf2/HO-1 and suppressing NF-κB and MAPK-dependent signaling pathways. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167770. [PMID: 40037266 DOI: 10.1016/j.bbadis.2025.167770] [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: 09/04/2024] [Revised: 12/03/2024] [Accepted: 02/27/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND AND OBJECTIVE Acute lung injury (ALI) represents a complicated and debilitating pulmonary disorder for which therapeutic options are currently limited. Genipin is an aglycone derived from the geniposide, the most abundant iridoid glucoside constituent of Gardenia jasminoides Ellis, and has demonstrated beneficial effects in ALI. The objective of this study was to investigate the protective effect of Gen-17, a beta-methyl derivative of genipin, against ALI in vitro and in vivo, and explore its mechanism of action. METHODS In this study, we prepared a beta-methyl derivative of genipin, Gen-17, and assessed the antioxidative and anti-inflammatory effects of Gen-17 in LPS-induced murine macrophages and ALI in mice, and explored the mechanism of action of Gen-17. In an in vivo model, the impact of Gen-17 on lipopolysaccharide (LPS)-induced ALI in mice was investigated by assessing pro-inflammatory cytokine levels, lung histology, edema, and vascular and alveolar barrier integrity, and in an in vitro model, murine macrophages-Raw 264.7 cells were used to establish a cell model of inflammation and oxidative stress by incubating with LPS. Keap1-Nrf2/HO-1, NF-κB and MAPK signaling pathways related factors were tested in vitro and in vivo to explore the possible mechanism of Gen-17. RESULTS The study showed that administration of Gen-17 conferred protection against LPS-induced ALI in mice, characterized by the mitigation of histological lung tissue alterations, reduction in lung edema, diminished protein content in bronchoalveolar lavage fluid, attenuation of inflammatory cell infiltration, and a decrease in cytokine secretion. Furthermore, Gen-17 exhibited the capacity to inhibit the nuclear factor-kappa B (NF-κB) and extracellular signal-regulated kinase (ERK) in the context of LPS-induced lung injury. In vitro, research findings revealed that Gen-17 demonstrated notable efficacy in reducing oxidative stress and inflammation in RAW 264.7 cells induced by LPS. Its central mechanism of action revolved around enhancing the antioxidant defense pathway, mediated through nuclear factor erythroid 2-related factor 2 (Nrf2). Consequently, this intervention repressed the release of pro-inflammatory mediators initiated by LPS, along with the modulation of the mitogen-activated protein kinase (MAPK) signaling pathway. CONCLUSION Gen-17 demonstrates the ability to mitigate oxidative stress and inflammation in the context of LPS-induced ALI via modulation of the MAPK, NF-κBp65, and Keap1/Nrf2/heme oxygenase-1 (HO-1) pathways. As such, it emerges as a promising and novel therapeutic candidate for treating ALI.
Collapse
Affiliation(s)
- Yu-Shan Zhu
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products/Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, China
| | - Syed Alfakhar Ali Shah
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products/Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, China
| | - Bi-Ying Yang
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products/Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, China
| | - Su-Su Fan
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products/Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, China
| | - Lei He
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products/Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, China
| | - Ya-Ru Sun
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products/Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, China
| | - Wen-Bin Shang
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products/Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, China.
| | - Yiyi Qian
- Department of Pharmacy, Fuwai Yunnan Cardiovascular Hospital, Kunming, China.
| | - Xuan Zhang
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products/Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, China.
| |
Collapse
|
21
|
Askarizadeh F, Butler AE, Kesharwani P, Sahebkar A. Regulatory effect of curcumin on CD40:CD40L interaction and therapeutic implications. Food Chem Toxicol 2025; 200:115369. [PMID: 40043936 DOI: 10.1016/j.fct.2025.115369] [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: 11/14/2024] [Revised: 01/24/2025] [Accepted: 03/02/2025] [Indexed: 04/21/2025]
Abstract
Natural compounds have garnered significant attention as potential therapeutic agents due to their inherent properties. Their notable qualities, including safety, efficacy, favorable pharmacokinetic properties, and heightened effectiveness against certain diseases, particularly inflammatory conditions, make them particularly appealing. Among these compounds, curcumin has attracted considerable interest for its unique therapeutic properties and has therefore been extensively studied as a potential therapeutic agent for treating various diseases. Curcumin exhibits diverse anti-inflammatory, antioxidant, and antimicrobial effects. Curcumin's immune system regulatory ability has made it a promising compound for treatment of various inflammatory diseases, such as psoriasis, atherosclerosis, asthma, colitis, IBD, and arthritis. Among the signaling pathways implicated in these conditions, the CD40 receptor together with its ligand, CD40L, are recognized as central players. Studies have demonstrated that the interaction between CD40 and CD40L interaction acts as the primary mediator of the immune response in inflammatory diseases. Numerous studies have explored the impact of curcumin on the CD40:CD40L pathway, highlighting its regulatory effects on this inflammatory pathway and its potential therapeutic use in related inflammatory conditions. In this review, we will consider the evidence concerning curcumin's modulatory effects in inflammatory disease and its potential therapeutic role in regulating the CD40:CD40L pathway.
Collapse
Affiliation(s)
- Fatemeh Askarizadeh
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
22
|
Xu S, Sun X, Gu Y, Liu T, Liu S, Weng Y, Zhang W, Wang L, Zhou M, Lu G, Tang M, Wang H, Li J. ECM characterization and 3D bioprinted models of NSCLC for investigating stiffness-dependent tumor behavior and drug response. Mater Today Bio 2025; 32:101823. [PMID: 40416783 PMCID: PMC12098162 DOI: 10.1016/j.mtbio.2025.101823] [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: 12/29/2024] [Revised: 04/02/2025] [Accepted: 04/30/2025] [Indexed: 05/27/2025] Open
Abstract
The heterogeneity and complex extracellular matrix (ECM) characteristics of non-small cell lung cancer (NSCLC) present significant challenges for understanding its pathological mechanisms and advancing precise treatment strategies. This study characterized the physicochemical properties of native NSCLC ECM to inform the biomimetic design of 3D models utilizing biomaterials and light-based 3D bioprinting technologies. A tunable 3D model was constructed that replicates the interfacial structures and matrix stiffness of both lung cancer tissue and adjacent normal tissue. This model elucidates the impact of matrix stiffness on cellular behaviors, including proliferation, invasion, and drug sensitivity, and delineates the role of the CCN1 gene under different mechanical conditions. Specifically, it demonstrates that a reduction in CCN1 expression within soft matrices can attenuate the migratory and proliferative capabilities of tumor cells. Furthermore, primary NSCLC patient-derived bioprinted tissues validated the model fidelity to clinical samples and its predictive potential for responses to combined chemotherapy and immunotherapy. This study establishes a versatile platform for NSCLC modeling and research, advancing biomaterial and bioprinting strategies to replicate the tumor microenvironment and optimize therapeutic approaches.
Collapse
Affiliation(s)
- Shiwei Xu
- Department of Thoracic Surgery, Afflicated Hospital of Jiangnan University, Wuxi, 214122, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Xin Sun
- Jiangsu Hansoh Pharmaceutical, Shanghai, 200120, China
| | - Yexin Gu
- Cyberiad Biotechnology, Shanghai, 201112, China
| | - Tong Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Shiyin Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Yuan Weng
- Department of Thoracic Surgery, Afflicated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Weimin Zhang
- Department of Thoracic Surgery, Afflicated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Leisheng Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Mengzhen Zhou
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Guye Lu
- Department of Pulmonary Medicine, Wuxi People's Hospital, Wuxi, 214023, China
| | - Min Tang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Haifeng Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Shanghai, 200433, China
| | - Jinyou Li
- Department of Thoracic Surgery, Afflicated Hospital of Jiangnan University, Wuxi, 214122, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| |
Collapse
|
23
|
Zhu L, Liu C, Wang Y, Zhu X, Wu L, Chen L, Zhou J, Wang F. METTL3/IGF2BP2/IκBα axis participates in neuroinflammation in Alzheimer's disease by regulating M1/M2 polarization of microglia. Neurochem Int 2025; 186:105964. [PMID: 40107503 DOI: 10.1016/j.neuint.2025.105964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Microglia-mediated neuroinflammation is closely related to the development of Alzheimer's disease (AD). This study further elucidated the regulatory mechanism of microglia polarization in AD. METHOD Microglia polarization was assessed using RT-qPCR, ELISA, and immunofluorescence (IF). Western blot (WB) analyzed inflammation-related, p-tau, and apoptosis-related proteins. Neuronal damage was evaluated by immunofluorescence, and neuronal apoptosis by flow cytometry and TUNEL assay. METTL3 and IκBα expression were detected using RT-qPCR and WB. N6-methyladenosine (m6A) levels were quantified with a colorimetric assay. RNA pull-down assay examined METTL3, IGF2BP2, and IκBα mRNA binding. IGF2BP expression was assessed by RT-qPCR. Learning and memory abilities were evaluated using morris water maze (MWM) test and novel object recognition (NOR) test. Inflammation-related proteins were detected using IF. RESULTS Stimulation with Aβ1-42 led to microglia M1 polarization, upregulation of inflammation-related proteins, and exacerbation of neuronal injury and apoptosis, along with increased p-tau expression in neurons. METTL3/IGF2BP2 modulated IκBα m6A modification through binding to IκBα mRNA, enhancing its expression. Enhanced METTL3 or IGF2BP2 expression suppressed M1 polarization, inflammation, and neuronal apoptosis in microglia, reversed by knockdown of IκBα. AD model mice exhibited cognitive impairments, neuroinflammation, and elevated M1 polarization. METTL3 or IGF2BP2 overexpression improved cognitive function, reduced neuroinflammation, and inhibited M1 polarization, and this effect was similarly reversed by knockdown of IκBα. CONCLUSION Our study demonstrates that the METTL3/IGF2BP2/IκBα axis is involved in neuroinflammation in AD by modulating microglia M1/M2 polarization, which sheds light on the treatment of AD.
Collapse
Affiliation(s)
- Ling Zhu
- Department of neurology, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China
| | - Congyan Liu
- Department of pharmacy, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China
| | - Yang Wang
- Department of radiology, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China
| | - Xuanang Zhu
- Department of neurology, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China
| | - Lei Wu
- Department of neurology, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China
| | - Lvan Chen
- Department of neurosurgery, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China
| | - Jing Zhou
- College of Medical, Jingchu University of Technology, Jingmen, 448000, China.
| | - Fan Wang
- Department of neurosurgery, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China; College of Medical, Jingchu University of Technology, Jingmen, 448000, China.
| |
Collapse
|
24
|
Wang J, Wu L, Tian Z, Chen J. Effect of deubiquitinases in head and neck squamous cell carcinoma (Review). Oncol Lett 2025; 29:307. [PMID: 40337608 PMCID: PMC12056481 DOI: 10.3892/ol.2025.15053] [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: 10/08/2024] [Accepted: 02/04/2025] [Indexed: 05/09/2025] Open
Abstract
HNSCC includes nasopharyngeal, laryngeal and oral cancers, and its pathogenesis is influenced by various factors. As an essential part of the ubiquitin (Ub)-proteasome system (UPS), deubiquitinating enzymes (DUBs) maintain the homeostasis of Ub molecules and influence the physiological functions of cells and disease processes by removing ubiquitinated proteins. Accumulating evidence has confirmed that the aberrant expression of DUBs is involved in cell proliferation, metastasis, and apoptosis during the development of HNSCC, with some acting as oncogenes and others as tumor-suppressor genes. In this review, the DUBs implicated in HNSCC were summarized and the mechanisms underlying abnormal DUBs expression in signaling pathways were discussed. In addition, given the important role of DUBs in tumorigenesis, recent studies were reviewed and agonists and inhibitors of DUBs were summarized to identify more effective therapeutic strategies.
Collapse
Affiliation(s)
- Jiahui Wang
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Liangpei Wu
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Zhifeng Tian
- Cancer Center, Lishui Municipal Central Hospital, Lishui, Zhejiang 323000, P.R. China
| | - Jun Chen
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
25
|
Huang Y, Li G, Li D, Liu C, Chen M, Cai L, Sun M, Xu Q. Ethyl caffeate alleviates inflammatory response and promotes recovery in septic-acute lung injury via the TNF-α/NF-κB/MMP9 Axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156700. [PMID: 40220405 DOI: 10.1016/j.phymed.2025.156700] [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: 12/12/2024] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Septic acute lung injury (Septic-ALI, SA) is a severe complication of sepsis with limited clinical treatment options. Ethyl Caffeate (EC) is a phenolic compound isolated from Ilex latifolia Thunb (I. latifolia) of the Aquifoliaceae family. PURPOSE This study aimed to investigate the potential mechanisms of EC in treating SA by integrating network pharmacology and transcriptomics. METHODS We used network pharmacology to predict the potential pathways and targets of EC and validated these predictions using the GEO database, molecular docking and MDS. Subsequently, LPS-induced inflammation models in RAW cells and a mouse model of SA were established to evaluate the therapeutic effects of EC. Cell transcriptomic sequencing, along with ELISA, qRT-PCR, and Western blot analyses, were performed on both cellular and animal models to validate the key pathways and targets. RESULTS EC targeted TNF-α and MMP9, significantly alleviating LPS-induced SA through the TNF-α/NF-κB/MMP9 axis. Specifically, network pharmacology and molecular docking suggested that EC may target TNF, MMP9, EGFR, PRKACA, and MAPK3. Transcriptomic analyses, MDS and in vitro and in vivo experiments showed that EC primarily reduced the expression of p-p65 and p-IκBα in the TNF pathway by inhibiting TNF-α, thereby downregulating the expression of downstream effector molecules MMP9 and MMP14, and improving lung tissue damage, cell apoptosis, and inflammation levels in mice. CONCLUSION This study was the first to integrate network pharmacology and transcriptomic results, revealing the mechanism by which EC ameliorated SA through the TNF-α/NF-κB/MMP9 axis. Furthermore, experimental validation identified TNF-α and MMP9 as two core targets of EC, providing a valuable reference for the clinical treatment of SA.
Collapse
Affiliation(s)
- Yuanlan Huang
- Department of Blood Transfusion, Naval Specialty Medical Center, Naval Medical University, Shanghai 200050, PR China
| | - Gang Li
- Department of Blood Transfusion, Naval Specialty Medical Center, Naval Medical University, Shanghai 200050, PR China.
| | - Dan Li
- Special Food Equipment Research Laboratory, Naval Specialty Medical Center, Naval Medical University, Shanghai 200050, PR China
| | - Chang Liu
- Department of Blood Transfusion, Naval Specialty Medical Center, Naval Medical University, Shanghai 200050, PR China
| | - Mengying Chen
- Department of Blood Transfusion, Naval Specialty Medical Center, Naval Medical University, Shanghai 200050, PR China
| | - Linli Cai
- Department of Blood Transfusion, Naval Specialty Medical Center, Naval Medical University, Shanghai 200050, PR China
| | - Mingxue Sun
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, PR China.
| | - Qingqiang Xu
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, PR China; Basic Medical Center for Pulmonary Disease, Naval Medical University, 800, Xiangyin Road, Shanghai 200433, PR China.
| |
Collapse
|
26
|
Men X, Chiou WC, Li X, Li Q, Chen X, Zhang K, Jiang X, Chen S. Klotho senses mechanical stimuli and modulates tension-induced osteogenesis. Bone 2025; 195:117464. [PMID: 40118264 DOI: 10.1016/j.bone.2025.117464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
Delicate external mechanosensing, efficient intracellular mechanotransduction and effective alveolar bone remodeling lay the foundation of orthodontic tooth movement (OTM). Periodontal ligament stem cells (PDLSCs) are thought to be the primary cells that withstand mechanical stimuli and respond to biomechanical signals during orthodontic treatment. Nevertheless, the cellular and molecular mechanisms of orthodontic force-induced mechanosignaling and osteogenesis in PDLSCs still remain unclear. In the present study, we hypothesize that the ageing suppressor, Klotho, is correlated with orthodontic force-triggered mechanical signaling cascades, further contributing to alveolar bone remodeling. This study reveals that Klotho expression is notably upregulated via cytoskeletal-nuclei-mediated epigenetic modifications, consistent with osteogenic differentiation on the tension side during OTM. Additionally, Klotho deficiency undermines tensile force-induced new bone formation in NFκB- and PI3K/Akt-dependent manners. Notably, RNA sequencing (RNA-seq) results and targeted force application experiments unveil that Klotho not only functions as a downstream effector of external stress but also acts as an upstream regulator in mechanical signaling for the first time. In summary, we identify the indispensable role of Klotho in mechanotransduction and alveolar bone formation, which provide a latent target of linking cell senescence to mechanical force in future studies and offer novel insights into orthodontic force-induced tooth movement and bone remodeling.
Collapse
Affiliation(s)
- Xinrui Men
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Wei-Cho Chiou
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xingjian Li
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Qiming Li
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xinyi Chen
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Kaiwen Zhang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoge Jiang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Song Chen
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
27
|
Qiu H, Liu F, Qiu M, Yang J, Peng X. Monotropein attenuates renal cell carcinoma cell progression and M2 macrophage polarization by weakening NF-κB. Int Urol Nephrol 2025; 57:1785-1795. [PMID: 39776402 DOI: 10.1007/s11255-024-04358-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE The study aimed to investigate the effect and mechanism of monotropein on renal cell carcinoma (RCC). METHODS After monotropein and NF-κB receptor activator (RANKL) treatment, cell proliferation, invasion, and apoptosis were evaluated using CCK-8, Transwell, and flow cytometry. Primary macrophages co-cultured with monotropein-treated RCC cells were analyzed to evaluate macrophage polarization using qRT-PCR, western blot, and ELISA assays by detecting the expression of M2 markers (CD206, CD168) and cytokines (IL-10, TGF-β). Additionally, the therapeutic efficacy of monotropein was examined using an RCC mouse xenograft model. RESULTS Monotropein could inhibit the proliferation, invasion, and M2 macrophage polarization and accelerate the apoptosis of RCC cells. Mechanistically, monotropein suppressed NF-κB pathway activation in RCC cells and reduced the expression of NF-κB downstream targets, including Bcl-2, c-Myc, and MMP9. RANKL could eliminate the effect of monotropein on RCC progression. In primary macrophages co-cultured with monotropein-treated RCC cells, monotropein downregulated M2 polarization markers and cytokines, further supporting its role in modulating the tumor microenvironment. In mouse models, monotropein reduced RCC tumor growth, induced apoptosis, and blocked NF-κB pathway. CONCLUSIONS Monotropein prevents RCC malignant progression and reduces M2 macrophage polarization by suppressing the NF-κB pathway, suggesting that monotropein may serve as a potential therapeutic agent for RCC by targeting both tumor cells and the tumor microenvironment.
Collapse
Affiliation(s)
- Heping Qiu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, East Lake District, Nanchang, 330006, Jiangxi, China
| | - Fei Liu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, East Lake District, Nanchang, 330006, Jiangxi, China
| | - Mei Qiu
- Department of Urology, Shanghai Changzheng Hospital, Shanghai, 341099, China
| | - Juliang Yang
- Department of Urology, The Affiliated Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou, 341099, China
| | - Xiang Peng
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, East Lake District, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
28
|
Chathuranga K, Rathnapala P, Weerawardhana A, Kim TH, Seong Y, Gayan Chathuranga WA, Subasinghe A, Haluwana DK, Gamage N, Choi YJ, Jung JU, Lee JS. The E3 ubiquitin ligase MARCH2 controls TNF-α mediated inflammation by autoubiquitination. Cell Commun Signal 2025; 23:257. [PMID: 40450320 DOI: 10.1186/s12964-025-02260-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Accepted: 05/20/2025] [Indexed: 06/03/2025] Open
Abstract
BACKGROUND Regulation of the nuclear factor-kappa B (NF-kB) signaling pathway is a major host homeostatic mechanism for controlling hyper-inflammation or chronic inflammation. Despite extensive research, the regulatory factors of NF-kB signaling required to preserve homeostasis and control inflammatory disorders are not fully understood. Moreover, the role of MARCH2 in chronic inflammation models and the regulation of MARCH2 activation remain to be elucidated. METHODS We monitored disease severity and mortality in MARCH2-/- or MARCH2+/+ mice induced experimental colitis. Susceptibility to DSS-induced experimental colitis was determined by various methods, including Swiss roll assay and fluorescein isothiocyanate (FITC)-dextran treatment, respectively. RNA-sequencing was conducted to recognize the inflammatory response-related genes in the distal colon of colitis-induced mice. Enzyme-linked immunosorbent assay (ELISA) was used to measure the cytokines and chemokines with in vitro and in vivo samples. Affinity purification and LC-MS/MS analysis were used to identify the MARCH2 interacting proteins and posttranslational modifications. The underlying mechanism was elucidated using immunoblotting, co-immunoprecipitation, ubiquitination assay, and confocal microscopy. RESULT Here, we report that MARCH2-/- mice were more susceptible to experimental inflammatory bowel disease (IBD) due to the massive production of cytokines. Stimulation by inflammatory cytokines such as TNF induces dimerization of MARCH2 at a later stage and dimerized MARCH2 undergoes K63-linked autoubiquitination at lysine 127 and 238, which promotes NEMO recognition, ubiquitination and proteasomal degradation. We also show an interaction between MARCH2 and MARCH8 in resting cells that inhibits MARCH2 activation. Taken together, these findings provide new insights into the molecular mechanism of MARCH2 and suggest a crucial role of MARCH2 in the modulation of inflammation and cellular homeostasis. CONCLUSION Our results indicate that MARCH2 plays a critical role in regulating NEMO/IKKγ under the inflammatory and resting conditions, thereby suppressing excessive or unexpected inflammatory responses. Our findings here not only demonstrate a biological role of MARCH2 in inflammatory signaling pathways but also provide a novel insight in the underlying mechanism.
Collapse
Affiliation(s)
- Kiramage Chathuranga
- Laboratory of Microbiology, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Pramodya Rathnapala
- Laboratory of Microbiology, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Asela Weerawardhana
- Laboratory of Microbiology, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Tae-Hwan Kim
- Laboratory of Microbiology, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Yebin Seong
- Laboratory of Microbiology, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - W A Gayan Chathuranga
- Laboratory of Microbiology, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Ashan Subasinghe
- Laboratory of Microbiology, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - D K Haluwana
- Laboratory of Microbiology, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Nuwan Gamage
- Laboratory of Microbiology, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Youn Jung Choi
- Kao Autoimmunity Institute and Division of Rheumatology, Department of Medicine, Ce dars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jae U Jung
- Department of Cancer Biology, Infection Biology Program, and Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jong-Soo Lee
- Laboratory of Microbiology, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
29
|
Zuo S, Yuan H, Li X, Chen M, Peng R, Chen S, Zou X, Yang Y, Long H, Liu Z, Wang T, Guo B, Liu L. SMYD2 Promotes Renal Tubular Cell Apoptosis and Chronic Kidney Disease Following Cisplatin Nephrotoxicity. FASEB J 2025; 39:e70651. [PMID: 40391402 PMCID: PMC12090038 DOI: 10.1096/fj.202402703r] [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: 11/02/2024] [Revised: 03/25/2025] [Accepted: 05/07/2025] [Indexed: 05/21/2025]
Abstract
The protein lysine methyltransferase 2 (SMYD2) can affect cell proliferation, differentiation, and survival through methylation of its histone and non-histone substrates. SMYD2 has been shown to act as an oncogene to promote disease progression in a variety of cancer diseases, but its role in chronic kidney diseases (CKD) pathogenesis has not been fully elucidated. This study aims to investigate the effect of SMYD2 on cisplatin-induced CKD and its underlying mechanisms. In this study, we found that cisplatin caused severe renal injury in mice, which was accompanied by the up-regulation of SMYD2 expression. AZ505 treatment significantly down-regulated cisplatin-induced renal injury and fibrosis. It also alleviated renal apoptosis and inhibited the phosphorylation level of NF-κB p65. Conditional knockdown of Smyd2 achieved similar effects as AZ505. In renal tubular epithelial cells, inhibition or silencing of SMYD2 down-regulated cisplatin-induced apoptotic response, while overexpression of SMYD2 induced apoptotic response and activated NF-κB in response to the up-regulation of SMYD2 expression. Up-regulation of SMYD2 induced interaction and phosphorylation of SMYD2 and NF-κB p65, and inhibition of NF-κB activation further suppressed cisplatin-induced NF-κB activation and apoptosis. The present study suggests that up-regulation of SMYD2 expression in cisplatin-induced CKD may promote apoptosis of renal tubular epithelial cells and accelerate the process of renal injury through NF-κB activation. SMYD2 may serve as a potential target for effective CKD treatment.
Collapse
Affiliation(s)
- Siyang Zuo
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Huixiong Yuan
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Xia Li
- Key Laboratory of Kidney Disease Pathogenesis Research and Transformation ApplicationGuizhou UniversityGuiyangChina
| | - Ming Chen
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Rui Peng
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Siyu Chen
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Xue Zou
- Key Laboratory of Kidney Disease Pathogenesis Research and Transformation ApplicationGuizhou UniversityGuiyangChina
| | - Yuan Yang
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Hehua Long
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Zeying Liu
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Teng Wang
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Bing Guo
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic DiseasesGuizhou Medical UniversityGui'an New DistrictChina
| | - Lirong Liu
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| |
Collapse
|
30
|
Chen Y, Gao Y, Zhang Z, Jiang Y, Wang R, Zhang H, Su Y. POSTN Silencing Ameliorates LL37-Induced Rosacea and Inhibits the JAK2/STAT3 and NF-κB Pathways. FASEB J 2025; 39:e70643. [PMID: 40387439 DOI: 10.1096/fj.202403202r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/08/2025] [Accepted: 05/07/2025] [Indexed: 05/20/2025]
Abstract
Rosacea is a chronic inflammatory skin disease and its pathogenesis remains unclear. Key genes were screened in the GSE155141 and GSE65914 datasets through a bioinformatics approach. To establish a rosacea-like mouse model with periostin (POSTN) knockdown, mice were subcutaneously injected with lentivirus-packaged Lv-shPOSTN, followed by LL37 treatment on the dorsal skin. Skin tissues were collected for the assessment of skin lesion area, skin thickness, redness score, as well as for hematoxylin and eosin (HE) staining, toluidine blue staining, and immunofluorescence staining. The inflammatory factors and chemokine levels were determined by enzyme-linked immunosorbent assay. Wound healing and Transwell assays were performed to assess cell migration and invasion. Phosphorylation levels of JAK2, STAT3, IKKβ, and p65 were evaluated via western blotting. Hub genes, including COL1A2, POSTN, LOX, BGN, COL3A1, DCN, and COL1A1 were screened. POSTN was highly expressed in rosacea and POSTN silencing ameliorated pathological changes and suppressed inflammation, immune infiltration, and angiogenesis. The levels of inflammatory factors (TNF-α, IL-1β, and IL-6) and chemokines (CCL2, CXCL10, and CXCL2), as well as the KLK5, CAMP, TLR2, and VEGF expression levels were reduced after POSTN knockdown. POSTN silencing inhibited migration and invasion of LL37-induced human umbilical vein endothelial cells (HUVEC). Importantly, POSTN silencing suppressed the JAK2/STAT3 and NF-κB pathways both in vivo and in vitro. POSTN knockdown suppresses inflammation and angiogenesis in rosacea possibly by obstructing the JAK2/STAT3 and NF-κB pathways, which offers a potential therapeutic strategy for rosacea.
Collapse
Affiliation(s)
- Yan Chen
- Department of Dermatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yang Gao
- Department of General Practice, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhang Zhang
- Department of Dermatology, Baoan Central Hospital of Shenzhen, Shenzhen, China
| | - Yang Jiang
- Shenzhen Center for Chronic Disease Control, Shenzhen Institute of Dermatology, Shenzhen, China
| | - Ruiqi Wang
- Department of Dermatology, Sichuan Tianfu New Area People's Hospital, Chengdu, China
| | - Hao Zhang
- Shenzhen Center for Chronic Disease Control, Shenzhen Institute of Dermatology, Shenzhen, China
| | - Yang Su
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
31
|
Jeon Y, Bae H, Woo SW, Kim J, Yu D. Identifying functional roles and pathways of shared mutations in canine solid tumors by whole-genome sequencing. PLoS One 2025; 20:e0307792. [PMID: 40446009 PMCID: PMC12124556 DOI: 10.1371/journal.pone.0307792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 02/04/2025] [Indexed: 06/02/2025] Open
Abstract
Identifying genetic mutations contributing to solid tumors by altering the biological pathways related to tumor formation and development is essential for the development of targeted therapies. This study aimed to identify commonly mutated genes and altered pathways in canine solid tumors. Four dogs with different types of naturally occurring neoplasias (urothelial carcinoma, adenocarcinoma, rhabdomyosarcoma, and chondrosarcoma) were randomly selected and classified into carcinoma and sarcoma groups based on histopathological findings. Tumor tissues were analyzed using whole-genome sequencing, and significant variants shared within each tumor group were identified. Gene set enrichment analyses were conducted to compare the biological and functional pathways altered by the mutations in each carcinoma and sarcoma group. Forty-three and fifty-eight genes were identified in the carcinoma and sarcoma groups, respectively. Distinctions between the two tumor groups were noted for mutations related to tumor metastatic function. Mutations were identified in genes encoding cell adhesion molecules in the carcinoma group, whereas significant variations in extracellular matrix-related molecules were evident in the sarcoma group. This study revealed mutations and modified pathways associated with immune and tumor metastatic functions in canine carcinoma and sarcoma, indicating their significant relevance to the development and progression of each tumor group. Additionally, the distinctions indicated that different therapeutic approaches were required for each tumor group.
Collapse
Affiliation(s)
- YeSeul Jeon
- College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Hyeona Bae
- College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Seung-Wan Woo
- Division of Applied Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Jaemin Kim
- Division of Applied Life Science, Gyeongsang National University, Jinju, Republic of Korea
- Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - DoHyeon Yu
- College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
32
|
Meng Y, Jia Y, He Z, Li J, Yuan L. Aryl Hydrocarbon Receptor-Dependent miRNA-382-5p Mediates the Classical Pyroptosis Induced by Foodborne Benzo(a)pyrene through Targeting IκB in Liver. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:12978-12992. [PMID: 40368843 DOI: 10.1021/acs.jafc.5c00511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Benzo(a)pyrene (BaP), a carcinogen prevalent in high-temperature processed foods, activates the aryl hydrocarbon receptor (AhR) and induces liver pyroptotic injury. MicroRNAs regulate mRNA expression and are involved in BaP toxicity. In this study, we investigated the essential role of microRNA in BaP-induced pyroptosis. In vivo, BaP induces liver pyroptotic injury by activating AhR, which may be attributed to AhR's influence on microRNA expression. The miRNA-382-5p/Akt and miRNA-382-5p/IκB pairs were predicted to post-transcriptionally regulate pyroptosis. In vitro, miRNA-382-5p activates the classical NF-κB/NLRP3/Caspase-1 pyroptosis signaling pathway by targeting and inhibiting the IκB gene. Furthermore, AhR activation by BaP could promote the high expression of miRNA-382-5p, thereby upregulating the NF-κB/NLRP3/Caspase-1 pyroptosis signaling pathway. In summary, we established that the AhR-mediated miR-382-5p/NF-κB/NLRP3/Caspase-1 axis is a key driver of BaP-induced pyroptosis in hepatocytes and elucidated the underlying mechanisms. These findings provide a valuable theoretical basis for considering miRNA-382-5p as a potential target for preventing BaP toxicity.
Collapse
Affiliation(s)
- Yao Meng
- Engineering Research Center of High Value Utilization of Western China Fruit Resources, Key Laboratory of Food Processing Byproducts for Advanced Development and High Value Utilization, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi' an 710119, People' s Republic of China
| | - Yingyu Jia
- Engineering Research Center of High Value Utilization of Western China Fruit Resources, Key Laboratory of Food Processing Byproducts for Advanced Development and High Value Utilization, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi' an 710119, People' s Republic of China
| | - Ziyan He
- Engineering Research Center of High Value Utilization of Western China Fruit Resources, Key Laboratory of Food Processing Byproducts for Advanced Development and High Value Utilization, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi' an 710119, People' s Republic of China
| | - Jianke Li
- Engineering Research Center of High Value Utilization of Western China Fruit Resources, Key Laboratory of Food Processing Byproducts for Advanced Development and High Value Utilization, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi' an 710119, People' s Republic of China
| | - Li Yuan
- Engineering Research Center of High Value Utilization of Western China Fruit Resources, Key Laboratory of Food Processing Byproducts for Advanced Development and High Value Utilization, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi' an 710119, People' s Republic of China
| |
Collapse
|
33
|
Gao Y, Xu H, Zhao Q, Cai D, Zhou X, Chen X, Yuan S, Li D, Ning T, Dong X, Li M, Li G, Nan A. The key regulator circPDE3B promotes arsenic-induced bladder carcinogenesis by affecting STAT3 and NF-κB stability. Cell Biol Toxicol 2025; 41:91. [PMID: 40437145 PMCID: PMC12119782 DOI: 10.1007/s10565-025-10038-2] [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: 11/28/2024] [Accepted: 05/09/2025] [Indexed: 06/01/2025]
Abstract
Long-term exposure to arsenic (As), which is a ubiquitous environmental contaminant, significantly enhances the risk of multiple cancers, including bladder and lung cancers. In recent years, the important roles of circular RNAs (circRNAs) in tumorigenesis and development have attracted widespread attention. However, the specific molecular mechanisms by which circRNAs promote bladder cancer development following exposure to arsenic remain incompletely understood. This study is the first to demonstrate that circPDE3B is significantly upregulated in a cell model of transformation triggered by arsenic and that it promotes this transformation process. Our study elucidated the biological function of circPDE3B in vitro, in SV-HUC-1 cells, showing that it accelerates the malignant transformation from arsenic via increasing cell proliferation and inhibiting apoptosis. Furthermore, we delineated a novel molecular mechanism whereby circPDE3B directly binds to NF-κB and STAT3, inhibiting their ubiquitination and increasing their stability. This, in turn, affects downstream HIF-1α expression, promoting the malignant transformation of SV-HUC-1 cells and eventually resulting in bladder carcinogenesis. Our research reveals the critical regulatory role of circPDE3B in the arsenic-triggered malignant transformation within SV-HUC-1 cells. This study offers broader perspectives on the molecular mechanisms driving bladder cancer progression, while also identifying potential targets for early diagnosis and treatment of bladder tumour.
Collapse
Affiliation(s)
- Yihong Gao
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Haotian Xu
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Qingyun Zhao
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Dunyu Cai
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Xiaodong Zhou
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Xingcai Chen
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Shengyi Yuan
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Deqing Li
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Taoran Ning
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Xiangyu Dong
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Mengyao Li
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Gang Li
- School of Public Health, Guangxi Medical University, Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China.
| | - Aruo Nan
- School of Public Health, Guangxi Medical University, Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
34
|
Ye Y, Yao Y, He S, Zhao X, Wang W, Lin M, Su Y, Wu G, Zhou F, Wang L, Zhong C, Huang M, Wu H, Lu Y. Biyuantong decoction improves chronic rhinosinusitis by inhibiting inflammatory cell adhesion via NF-кB pathway modulation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119907. [PMID: 40316153 DOI: 10.1016/j.jep.2025.119907] [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: 02/08/2025] [Revised: 04/14/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Biyuantong decoction (BYT), a traditional Chinese medicinal formulation, has been used for years to treat chronic rhinosinusitis (CRS) with good clinical results. However, the underlying mechanisms of its treatment for CRS remain to be fully elucidated. STUDY AIM This research investigates the molecular mechanism by which BYT ameliorates CRS and provide new perspectives for CRS treatment research. MATERIALS AND METHODS Clinical research is conducted on CRS patients who underwent surgery, and post-operative treatments and observations were performed. The pathological alterations of CRS were inspected by H&E staining and nasal endoscopy. Flow cytometry and ELISA were employed to measure the levels of inflammatory cells and cytokines in the peripheral blood of CRS patients. The cytotoxic impacts of BYT were assessed by cell viability, cell cycle and apoptosis assays. The effects of BYT on the adhesion and invasion of inflammatory cells to endothelial cells were evaluated by hetero-adhesion and transwell assay. Flow cytometry was employed to analyze the expression of cell adhesion molecules (CAMs) on HUVECs. The effects of BYT on NF-κB signaling pathway was analyzed by Western blot and immunofluorescence staining. The chemical components of BYT was determined by UPLC-HRMS, and network pharmacology analysis was adopted to predict potential targets in the NF-κB pathway. RESULTS Clinical samples demonstrated that BYT treatment could effectively alleviate sinus mucosal edema and significantly decreased the recurrence rate after surgery. H&E staining disclosed obvious inflammatory cell infiltration in the sinus mucosa of CRS patients. Flow cytometry and ELISA results indicated that BYT treatment reduced the levels of eosinophils (median decrease 16.21 %) and cytokines in peripheral blood. Cell adhesion and transwell assays manifested that BYT inhibited the adhesion and invasion of U937 cells to TNF-α-induced HUVECs. Moreover, BYT counteracted the TNF-α-induced upregulation of CAMs on endothelial cells. Western blot and immunofluorescence analyses confirmed that BYT reduced the expression of NF-κB-related proteins and hindered the nuclear translocation of NF-κB. Network pharmacology analysis and component identification of BYT further supported the function of its compounds in synergistically modulating NF-κB signaling. CONCLUSION BYT enhances the clinical efficacy of CRS by suppressing inflammatory cell adhesion and infiltration into the nasal mucosa via NF-кB pathway regulation. These findings provide a robust foundation for the clinical application of BYT in CRS treatment and suggest interrupting inflammatory cell adhesion as a potential new approach.
Collapse
Affiliation(s)
- Yuying Ye
- Department of Otorhinolaryngology, Affiliated People's Hospital (Fujian Provincial People's Hospital), Fujian University of Traditional Chinese Medicine, Fuzhou, 350004, China
| | - Yinyin Yao
- College of Pharmacy, Fujian Key Laboratory of Chinese Materia Medica, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Fuzhou Institute of Oceanography, Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China
| | - Shanshan He
- College of Pharmacy, Fujian Key Laboratory of Chinese Materia Medica, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Fuzhou Institute of Oceanography, Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China
| | - Xinyi Zhao
- College of Pharmacy, Fujian Key Laboratory of Chinese Materia Medica, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Fuzhou Institute of Oceanography, Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China
| | - Weiyu Wang
- College of Pharmacy, Fujian Key Laboratory of Chinese Materia Medica, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Fuzhou Institute of Oceanography, Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China
| | - Mengting Lin
- College of Pharmacy, Fujian Key Laboratory of Chinese Materia Medica, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Fuzhou Institute of Oceanography, Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China
| | - Yafei Su
- Department of Otorhinolaryngology, Fuzhou City Second Genera Hospital, Fuzhou, 350007, China
| | - Guoqing Wu
- Department of Otorhinolaryngology, Affiliated People's Hospital (Fujian Provincial People's Hospital), Fujian University of Traditional Chinese Medicine, Fuzhou, 350004, China
| | - Feng Zhou
- Department of Otorhinolaryngology, Affiliated People's Hospital (Fujian Provincial People's Hospital), Fujian University of Traditional Chinese Medicine, Fuzhou, 350004, China
| | - Li Wang
- Department of Pathology, Affiliated People's Hospital (Fujian Provincial People's Hospital), Fujian University of Traditional Chinese Medicine, Fuzhou, 350004, China
| | - Chunlian Zhong
- Fuzhou Institute of Oceanography, Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China.
| | - Mingqing Huang
- College of Pharmacy, Fujian Key Laboratory of Chinese Materia Medica, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Hui Wu
- Department of Otorhinolaryngology, Affiliated People's Hospital (Fujian Provincial People's Hospital), Fujian University of Traditional Chinese Medicine, Fuzhou, 350004, China.
| | - Yusheng Lu
- Fuzhou Institute of Oceanography, Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China.
| |
Collapse
|
35
|
Li X, Ding L, Li Z, Cao Z, Li M, Yin K, Song S, Cao L, Xia Q, Wang Z, Zhao D, Tong X, Li X, Wang Z. Yangke powder alleviates OVA-induced allergic asthma by inhibiting the PI3K/AKT/NF-κB signaling pathway. Chin Med 2025; 20:69. [PMID: 40420184 PMCID: PMC12105270 DOI: 10.1186/s13020-025-01125-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 05/06/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Asthma is a chronic inflammatory airway disease that remains inadequately controlled by existing conventional treatments. A traditional Chinese medicine (TCM) formula of Yangke powder (yǎng ké sǎn-YKS) has demonstrated potential in alleviating asthma symptoms and reducing its acute exacerbation. Despite clinical evidence supporting its benefit, there is still insufficient understanding of the active compounds in YKS and their underlying mechanisms, which limits its broader clinical application. OBJECTIVE This study aims to identify the key active ingredients in YKS and explore their mechanisms, particularly through the PI3K/AKT/NF-κB pathways, to provide a scientific basis for its application in asthma treatment. METHODS We employed UPLC-Q-Exactive Orbitrap-MS to analyze YKS constituents, identified key ingredients, and explored asthma treatment mechanisms through bioinformatics, network pharmacology, Mendelian randomization, and molecular docking. The asthma model was evaluated using ovalbumin (OVA) and pulmonary function tests, while pathological examination was conducted using hematoxylin and eosin (HE), periodic acid-Schiff (PAS), and Masson trichrome stains. Concentrations of IgE, IL-4, and IL-5 were measured by ELISA, and protein and mRNA expressions were confirmed via qPCR, immunohistochemistry, and Western blot analysis. RESULTS A total of 174 compounds were identified in YKS by UPLC-MS, with 49 detected in the bloodstream, indicating their role as active ingredients. Bioinformatics analysis revealed 353 asthma-related targets and 972 potential targets for YKS. Key targets such as AKT1, TNF, and IL1B were validated by molecular docking. Our studies indicated that YKS modulates asthma primarily through the PI3K/Akt and NF-κB pathways, improving airway resistance, reducing inflammation, mucus production, and airway remodeling, and decreasing Th2 cytokines and IgE levels. CONCLUSION This investigation identifies Kaempferol, Norephedrine, Cynaroside, Genistein, and Rutin as critical active ingredients in YKS, impacting key biomarkers such as AKT1, TNF, and IL1B. These substances effectively modulate the PI3K/AKT/NF-κB pathway, enhancing the management of allergic asthma.
Collapse
Affiliation(s)
- Xueyan Li
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Lu Ding
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Zirui Li
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Zhenghua Cao
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Min Li
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Kai Yin
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Siyu Song
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Liyuan Cao
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Qinjing Xia
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Zihan Wang
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Daqing Zhao
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Xiaolin Tong
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China.
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Xiangyan Li
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China.
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130021, China.
| | - Zeyu Wang
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China.
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130021, China.
| |
Collapse
|
36
|
Bakrim S, Fessikh ME, Elhrech H, Omari NE, Amanullah M, Ming LC, Moshawih S, Bouyahya A. Targeting inflammation in cancer therapy: from mechanistic insights to emerging therapeutic approaches. J Transl Med 2025; 23:588. [PMID: 40420174 DOI: 10.1186/s12967-025-06583-3] [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: 03/08/2025] [Accepted: 05/07/2025] [Indexed: 05/28/2025] Open
Abstract
Inflammation is a complex and finely tuned component of the host defense mechanism, responding sensitively to a range of physical, chemical, and biological stressors. Current research is advancing our grasp of both cellular and molecular mechanisms that initiate and regulate interactions within inflammatory pathways. Substantial evidence now indicates a profound link between inflammation, innate immunity, and cancer. Dysregulation of inflammatory pathways is known to be a pivotal factor in the induction, growth, and metastasis of tumors through multiple mechanistic pathways. Basically, the tumor microenvironment (TME), characterized by dynamic interplay between cancerous cells and surrounding inflammatory and stromal cells, plays a central role in these processes. Increasingly, controlled acute inflammation is being explored as a promising therapeutic tool in certain types of cancer. However, inflammatory cells in the TME exhibit remarkable plasticity, with shifting phenotypic and functional roles that facilitate cancer cell survival, proliferation, and migration, especially under chronic inflammatory conditions. Additionally, signaling molecules associated with the innate immune system, like chemokines, are co-opted by malignant cells to support invasion, migration, and metastasis. These findings underscore the need for deeper insights into the mechanisms connecting inflammation to cancer pathology, which could pave the way for innovative diagnostic approaches and targeted anti-inflammatory therapies to counter tumor development. The current review underlines the critical involvement of inflammation in cancer development, examining the connection between the immune system, key inflammatory mediators, biomarkers, and their associated pathways in cancer. We also discuss the impact of inflammation-targeted therapies on anticancer signaling pathways. Furthermore, we review major anti-inflammatory drugs with potential applications in oncology, assessing how inflammation is modulated in cancer management. Lastly, we outline an overview of ongoing discoveries in the field, highlighting both the challenges and the therapeutic promise of targeting inflammation in cancer therapy.
Collapse
Affiliation(s)
- Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir, 80000, Morocco
| | - Meriem El Fessikh
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| | - Hamza Elhrech
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| | - Nasreddine El Omari
- High Institute of Nursing Professions and Health Techniques of Tetouan, Tetouan, Morocco
| | - Mohammed Amanullah
- Department of clinical Biochemistry, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Long Chiau Ming
- Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education and Research (deemed to be University), Sawangi (M), Wardha, India
- Faculty of Medical and Life Sciences, Sunway University, Sunway City, Malaysia
| | - Said Moshawih
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco.
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan.
| |
Collapse
|
37
|
Fan D, Shang Y, Cong Y, Jiao Y, Li N, Zhao H. Reciprocal regulation between m6 A modifications and non-coding RNAs: emerging roles in cancer therapeutic resistance. Discov Oncol 2025; 16:920. [PMID: 40413672 DOI: 10.1007/s12672-025-02641-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 05/09/2025] [Indexed: 05/27/2025] Open
Abstract
In recent years, the interplay between N6-methyladenosine (m6A) modifications and non-coding RNAs (ncRNAs) has emerged as a pivotal research area, owing to their crucial involvement in the pathophysiological mechanisms underlying various diseases. A significant hurdle in cancer therapy is therapeutic resistance, which frequently contributes to adverse patient outcomes. Recent investigations have underscored the vital role that interactions between m6A modifications and ncRNAs play in mediating cancer therapeutic resistance via the MAPK, PI3K/Akt/mTOR, Wnt/β-catenin, HIPPO, and NF-κB pathways. This review elucidates how these interactions drive tumor therapeutic resistance by modulating these pathways. By dissecting the regulatory dynamics between m6A and ncRNAs in the context of cancer therapeutic resistance, this review aims to deepen the understanding of m6A-ncRNA interaction in cancer therapeutic resistance and identify potential therapeutic targets to improve cancer treatment efficacy.
Collapse
Affiliation(s)
- Dan Fan
- Nanshan Class, The First Clinical Institute, Zunyi Medical University, Zunyi, 563000, China
| | - Yan Shang
- Department of Pathophysiology, Zunyi Medical University, Zunyi, 563000, China
| | - Yating Cong
- Department of Pathophysiology, Zunyi Medical University, Zunyi, 563000, China
| | - Yanlin Jiao
- Department of Pathophysiology, Zunyi Medical University, Zunyi, 563000, China
| | - Na Li
- The First Clinical Institute, Zunyi Medical University, Zunyi, 563000, China
| | - Hailong Zhao
- Department of Pathophysiology, Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
38
|
Ou Y, Huang Y, Yang X, Li L, Mei R, Yu Z, So KF, Chen J, Ma JH, Tang S. Lycium barbarum glycopeptide alleviates retinal inflammation by suppressing microglial M1 polarization via NF-κB/MAPK pathways. Exp Eye Res 2025; 257:110452. [PMID: 40412789 DOI: 10.1016/j.exer.2025.110452] [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: 01/15/2025] [Revised: 03/22/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
Lycium barbarum glycopeptide (LBGP) is a known glycoconjugate with various pharmacological benefits, notably anti-inflammatory properties, though its impact on retinal inflammatory conditions is not fully understood. This research evaluated the impact of LBGP on retinal inflammation using a diabetic retinopathy (DR) mouse model induced by streptozotocin (STZ), along with LPS/IFN-γ (L/I)-stimulated BV2 microglia and primary retinal microglia. In vivo, administration of LBGP effectively enhances retinal thickness, structure, and function in diabetic mice. Additionally, it prevents microglial activation and inflammation. In vitro, LBGP pretreatment significantly reversed L/I-induced morphological alterations in microglial area, perimeter, Feret's diameter, and roundness. LBGP significantly alleviated L/I-induced microglial activation in primary and BV2 microglia. LBGP shifted M1 pro-inflammatory phenotype to M2 anti-inflammatory phenotype by downregulating M1 markers (IL-6, IL-1β, iNOS, COX2, CD86, and CD16) and upregulating M2 markers (CD206 and arginase 1). Additionally, LBGP reduced the upregulation of NF-κB and MAPK pathways in L/I-stimulated BV2 microglial cells. Our study suggests that LBGP protects against microglial overactivation and diminishes the secretion of inflammatory molecules from microglia in vivo and vitro, potentially through attenuation of the NF-κB and MAPK signaling pathways.
Collapse
Affiliation(s)
- Yiwen Ou
- Aier Academy of Ophthalmology, Central South University, Changsha, 410000, China; Aier Eye Institute, Changsha, 410000, China
| | - Yinhua Huang
- Shanghai Aier Eye Hospital, Shanghai, 200000, China; Shanghai Aier Eye Institute, Shanghai, 200000, China
| | - Xu Yang
- Aier Eye Institute, Changsha, 410000, China
| | - Lian Li
- Aier Eye Hospital, Jinan University, Guangzhou, 510000, China
| | - Rangsheng Mei
- Aier Academy of Ophthalmology, Central South University, Changsha, 410000, China; Aier Eye Institute, Changsha, 410000, China
| | - Zhexiong Yu
- Tianren Goji Biotechnology Co., Ltd, Zhongning, 755100, China
| | - Kwok-Fai So
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, 510000, China; Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510000, China; Aier Academician Station, Changsha, 410000, China
| | - Jiansu Chen
- Aier Academy of Ophthalmology, Central South University, Changsha, 410000, China; Aier Eye Institute, Changsha, 410000, China
| | - Jacey Hongjie Ma
- Aier Academy of Ophthalmology, Central South University, Changsha, 410000, China; Aier Eye Institute, Changsha, 410000, China; Aier Eye Hospital, Jinan University, Guangzhou, 510000, China.
| | - Shibo Tang
- Aier Academy of Ophthalmology, Central South University, Changsha, 410000, China; Aier Eye Institute, Changsha, 410000, China; Aier Eye Hospital, Jinan University, Guangzhou, 510000, China.
| |
Collapse
|
39
|
Li Z, Fan H, Xiong J, Tian M, Ye C, Liu S, Li G, Segbo JAG, Wu K, Zhu C. Vesicular Stomatitis Virus Induces NF-κB-Dependent Senescence to Mediate Persistent Inflammation and Injury. Viral Immunol 2025. [PMID: 40401441 DOI: 10.1089/vim.2025.0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025] Open
Abstract
Cell senescence, induced by various internal and external stresses, plays a significant role in the development of various diseases such as cancer, neurodegeneration, and infections. Viral infections can also induce cellular senescence, known as virus-induced senescence (VIS), which occurs in close correlation with the severity of the viral infections. However, due to the unclear mechanisms underlying VIS, the effective inhibition of VIS during viral infections is challenging, leading to rapid disease progression. This study utilized the widely used vesicular stomatitis virus (VSV) model virus to simulate RNA virus infections for exploring the mechanisms by which RNA viruses induce cellular senescence. The results indicated that VSV infection, both in vitro and in vivo, could significantly induce the upregulation of senescence-associated markers and the secretion of the senescence-associated secretory phenotype (SASP), promoting the senescence process. Further research found that the activation of the NF-κB pathway played a crucial role in VSV-induced cellular senescence. Targeted inhibition of the NF-κB pathway could reduce the level of organ senescence induced by viral infections, decrease the expression of SASP inflammatory factors, and ameliorate tissue damage in mice. Overall, our findings reveal the mechanisms underlying RNA virus-associated VIS and provide potential targets for inhibiting the occurrence of VIS and preventing disease progression.
Collapse
Affiliation(s)
- Zhiqiang Li
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Hong Fan
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Jiali Xiong
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Mingfu Tian
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, PR China
| | - Chenglin Ye
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Siyu Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, PR China
| | - Guangli Li
- Postgraduate Training Base at Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, PR China
| | | | - Kailang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, PR China
| | - Chengliang Zhu
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, PR China
| |
Collapse
|
40
|
Yurchenko AA, Sharkhith H, Rajabi F, Bogiatzi S, Wang J, Huber M, Hohl D, Nikolaev S. Integrative Molecular Analysis of Skin Tumors from CYLD Cutaneous Syndrome Patients. J Invest Dermatol 2025:S0022-202X(25)00496-8. [PMID: 40412469 DOI: 10.1016/j.jid.2025.05.006] [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: 03/18/2025] [Revised: 05/01/2025] [Accepted: 05/04/2025] [Indexed: 05/27/2025]
Abstract
CYLD cutaneous syndrome (CCS) is a rare genetic disorder caused by germline CYLD mutations, leading to multiple benign skin tumors, including cylindromas, spiradenomas, and trichoepitheliomas. While these tumors are well-characterized histologically, their molecular landscape remains unclear, and no targeted treatments exist. To comprehensively analyze the molecular features of CCS tumors, we integrated newly generated and publicly available data using multiomic approaches. We profiled 24 CCS tumors through whole-exome/genome sequencing, RNA-seq, immunohistochemistry, and methylation arrays. To enhance statistical power, we incorporated existing CCS tumor datasets, forming a cohort of 50 tumors. CCS tumors exhibited a low mutational burden and scarce UV damage. The driver landscape was defined by mutations in CYLD, DNMT3A, and BCOR. The tumor microenvironment of cylindromas was rich in CD8+ T cells. Methylation profiling identified two groups: cylindromas, which appeared to originate from apocrine sweat glands, and trichoepitheliomas, which clustered with basal cell carcinoma. RNA sequencing revealed specific activation of the non-canonical NF-κB pathway in cylindromas, a finding confirmed by immunohistochemistry. This pathway may drive tumor formation, highlighting a potential therapeutic target for CCS. Our study provides insights into CCS tumor biology and suggests that targeting the non-canonical NF-κB pathway could be explored as a treatment strategy.
Collapse
Affiliation(s)
- Andrey A Yurchenko
- INSERM U981, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France
| | - Hiba Sharkhith
- Department of Dermatology, CHUV-FBM UNIL, Hôpital de Beaumont, Lausanne, Switzerland
| | - Fatemeh Rajabi
- INSERM U981, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France
| | - Sofia Bogiatzi
- Department of Dermatology, CHUV-FBM UNIL, Hôpital de Beaumont, Lausanne, Switzerland; Department of Clinical Pathology and Dermatology, HUG, Geneva, Switzerland
| | - Jinxin Wang
- INSERM U981, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France
| | - Marcel Huber
- Department of Dermatology, CHUV-FBM UNIL, Hôpital de Beaumont, Lausanne, Switzerland
| | - Daniel Hohl
- Department of Dermatology, CHUV-FBM UNIL, Hôpital de Beaumont, Lausanne, Switzerland.
| | - Sergey Nikolaev
- INSERM U981, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France.
| |
Collapse
|
41
|
Jiang H, Yang H, Li H, Wang Y, Shen L, Adu X, Zhang J, Zhou H, Pu L, Zhang S, Guo J, Tong A, Yan H. Anti-Herpes simplex virus type 1 activity of the Rohdea chinensis (Baker) N. Tanaka aqueous extracts. JOURNAL OF ETHNOPHARMACOLOGY 2025:120024. [PMID: 40412773 DOI: 10.1016/j.jep.2025.120024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 05/21/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Herpes simplex virus type 1 (HSV-1) is a prevalent neurotropic pathogen that establishes lifelong latency in the peripheral nervous system, posing substantial clinical challenges due to its recurrent infections and emerging drug resistance. Rohdea chinensis (Baker) N. Tanaka, a traditional Chinese herbal medicine, has been historically used for heat-clearing, detoxifying, wind-dampness dispelling, blood-stasis removing, and pain-relieving properties. Despite its common folk use in Leshan, Sichuan Province, for treating herpes simplex infections, the chemical composition and antiviral activity of Rohdea chinensis remain largely uninvestigated. AIM OF THE STUDY The aim of this study was to clarify the chemical composition and antiviral activity of the Rohdea chinensis aqueous extract (RCAE), and preliminarily speculate the possible mechanism of its antiviral effect. MATERIALS AND METHODS The chemical profile of RCAE was characterized using UPLC-Q-TOF-MS/MS, identifying 9 major compounds. The antiviral effects of RCAE were assessed through in vitro and in vivo models, including direct virucidal assays, HSV-1 adhesion inhibition, and a mouse model of flank scratch HSV-1 infection. Immunomodulatory effects were evaluated by analyzing dendritic cell (DC) maturation, CD8+ T cell differentiation, and cytokine expression. Additionally, HSV-1 latent reactivation models were established to assess RCAE's impact on viral persistence in the dorsal root ganglion (DRG). Molecular docking studies were conducted to determine potential interactions between RCAE bioactive compounds and HSV-1 glycoprotein D (gD). RESULTS The overall anti-HSV-1 effect of RCAE on Vero cells was determined and the EC50 value was 7.994 μg/mL. RCAE directly killed HSV-1 and inhibited HSV-1 adhesion in vitro (P<0.001, 30 μg/mL RCAE treated groups vs HSV-1 group), and alleviated viral infection and inflammation of the skin involving the NF-κB pathway (P<0.001, different doses of RCAE treated groups vs HSV-1 group), as well as anti-viral infection of the brain and DRG in HSV-1-induced mice (P<0.05, 0.6 g/mL RCAE treated groups vs HSV-1 group). Interestingly, RCAE played an antiviral role by increasing the maturation of DC in mice, enhancing the antigen presentation ability of DC, promoting the differentiation of CD8+T cells and the secretion of CXCL1 and IFN-γ, and inhibiting the immune escape of HSV-1 virus (P<0.05, 0.6 g/mL RCAE treated groups vs HSV-1 group). Besides, RCAE prevented the reactivation of HSV-1 in vitro (P<0.001, 0.6 g/mL RCAE treated groups vs HSV-1 group). HSV gD is a critical viral glycoprotein that plays a central role in viral attachment to host cells, intercellular spread, and entry into host cells. To clarify the anti-HSV-1 mechanism of RCAE, molecular docking was employed to preliminarily confirm stable interactions between RCAE compounds (notably compound 9, with a binding energy of -10.5 kcal/mol) and HSV-1 gD. In vitro and in vivo experiments showed that RCAE reduced the expression of gD gene (P<0.001, different doses of RCAE treated groups vs HSV-1 group) and protein in skin tissues of HSV-1-infected mice, as well as the gD mRNA levels (P<0.001, different doses of RCAE treated groups vs HSV-1 group) in HSV-1-treated HaCaT cells. These findings suggest that the anti-HSV-1 effect of RCAE may be associated with the inhibition of gD function. CONCLUSIONS Our results indicate that RCAE has multi-target antiviral effects, including direct virus-killing activity, inhibition of viral replication, immune regulation, and inhibition of latent reactivation of HSV-1 in preclinical models. Integrating traditional Chinese medicine principles (heat-clearing, detoxifying, and blood-stasis-removing properties) with modern pharmacological activities (antiviral, anti-inflammatory, and immunomodulatory effects), RCAE demonstrates potential as a natural product candidate for addressing HSV-1 infection. Given RCAE's complex and undefined composition, future studies are needed to identify key antiviral components and their in vivo efficacy and mechanisms.
Collapse
Affiliation(s)
- Huiling Jiang
- Immunotherapy Laboratory, College of Pharmacy and Food, Southwest Minzu University, Chengdu, Sichuan, China
| | - Huantao Yang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Hui Li
- Immunotherapy Laboratory, College of Pharmacy and Food, Southwest Minzu University, Chengdu, Sichuan, China
| | - Yujuan Wang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Lidu Shen
- Immunotherapy Laboratory, College of Pharmacy and Food, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xiaobi Adu
- Immunotherapy Laboratory, College of Pharmacy and Food, Southwest Minzu University, Chengdu, Sichuan, China
| | - Junxiong Zhang
- Immunotherapy Laboratory, College of Pharmacy and Food, Southwest Minzu University, Chengdu, Sichuan, China
| | - Hang Zhou
- Immunotherapy Laboratory, College of Pharmacy and Food, Southwest Minzu University, Chengdu, Sichuan, China
| | - Likai Pu
- Immunotherapy Laboratory, College of Pharmacy and Food, Southwest Minzu University, Chengdu, Sichuan, China
| | - Shiying Zhang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Jiacheng Guo
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, 610041, Chengdu, China.
| | - Hengxiu Yan
- Immunotherapy Laboratory, College of Pharmacy and Food, Southwest Minzu University, Chengdu, Sichuan, China.
| |
Collapse
|
42
|
Ren Q, Zhu X, Wang N, Yu K, Lv W, Wang L, Zang Y, Ma D, Zhou X, Yao J, Shen M, Yu L, Li T. MiR-133a-5p Facilitates Cuproptosis in Hepatocellular Carcinoma Through Targeting of ATP7B. J Inflamm Res 2025; 18:6607-6622. [PMID: 40433054 PMCID: PMC12106915 DOI: 10.2147/jir.s515647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Purpose We explored the effects of miR-133a-5p and ATP7B on cuproptosis in hepatocellular carcinoma. Methods Initially, we assessed the impact of miR-133a-5p on hepatocellular carcinoma (HCC) using CCK-8 assays, cell scratch assays, and flow cytometry. Subsequently, we utilized elesclomol in combination with copper ions to induce cuproptosis in the HCC cell lines PLC/PRF/5 and Huh-7. We evaluated the influence of miR-133a-5p on cuproptosis using CCK-8 assays, cell scratch assays, flow cytometry, and Western blotting. To elucidate the underlying mechanisms, we employed bioinformatics to identify potential downstream target genes of miR-133a-5p and conducted dual-luciferase reporter assays to confirm the binding sites. Finally, we validated the regulatory effect of miR-133a-5p on ATP7B by modulating miR-133a-5p expression through cell transfection experiments. Results The results from the CCK-8 assay, cell scratch assay, and flow cytometry demonstrated that miR-133a-5p significantly inhibits the proliferation and migration of HCC cells while promoting their apoptosis. Furthermore, Elesclomol in combination with copper ions induces cuproptosis in HCC cells. Compared to the cuproptosis observed in HCC as a control, miR-133a-5p further suppresses the proliferation and migration of HCC cells, enhances their death, and increases the expression of cuproptosis-related proteins more prominently. Bioinformatics analysis suggested that ATP7B might be a downstream target gene of miR-133a-5p. This was confirmed by dual luciferase assays, which identified a binding site between miR-133a-5p and ATP7B. Additionally, the expression levels of ATP7B were found to decrease or increase in response to the regulation by miR-133a-5p. Conclusion MiR-133a-5p facilitates cuproptosis in hepatocellular carcinoma through targeting of ATP7B.
Collapse
Affiliation(s)
- Qiaohui Ren
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Xinyue Zhu
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Nannan Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Kang Yu
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Wei Lv
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Lianzi Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Yan Zang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Dongyue Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Xinyi Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Junxiao Yao
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Mengjiao Shen
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Li Yu
- Anhui Province Key Laboratory of Zoonoses, Anhui Medical University, Hefei, People’s Republic of China
| | - Tao Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| |
Collapse
|
43
|
Lin Z, Sun M. Phytochemical regulation of CaMKII in Alzheimer's disease: A review of molecular mechanisms and therapeutic potential. Pharmacol Res 2025; 216:107790. [PMID: 40409522 DOI: 10.1016/j.phrs.2025.107790] [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: 01/29/2025] [Revised: 04/08/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disorder that leads to cognitive decline. CaMKII is a calcium-regulated kinase that is crucial for synaptic plasticity and memory. Phytochemicals with diverse origins, safety, and biological activity have attracted considerable attention in AD research. This systematic analysis of phytochemicals targeting CaMKII reveals their neuroprotective mechanisms against AD pathogenesis, highlighting CaMKII as a promising therapeutic target that warrants further preclinical investigation and drug development. We conducted a comprehensive review of the literature of phytochemicals that target CaMKII as a protective mechanism against AD. The search was conducted across multiple databases, including PubMed, Web of Science, China National Knowledge Internet, and Google Scholar, and covered the period from January 2000 to October 2024. A total of 301 articles were retrieved, of which 22 articles were included. The results showed that flavonoid, glycoside, terpene, and polyphenol analogs positively regulated CaMKII expression, whereas alkaloid analogs negatively regulated CaMKII expression. Different components of traditional Chinese medicine played different roles in CaMKII expression. Flavonoid compounds upregulated the expression of SYN, PSD-95, MAP2, and GluR1 to exert neuroprotective effects. Alkaloid and glycoside analogs inhibited Aβ deposition and tau hyperphosphorylation. Terpene analogs upregulated the SYN, PSD-95, NMDAR, BDNF, and PI3K/Akt signaling pathways to exert neuroprotection. Polyphenol analogs upregulated PSD-95, Munc18-1, SNAP25, SYN, and BDNF to exert neuroprotective effects. Emerging evidence demonstrates that select phytochemicals and traditional Chinese medicine compounds exert neuroprotective effects in AD by modulating CaMKII activity, thereby reducing Aβ accumulation, attenuating tau hyperphosphorylation, and enhancing synaptic plasticity, suggesting promising therapeutic potential.
Collapse
Affiliation(s)
- Zhongying Lin
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| | - Miao Sun
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
44
|
Liu X, Liu X, Zhao L, Wu J, Wang X, Hu Y. RIP2/NF-κB/PD-L1 signaling pathway is involved in temozolomide resistance by inducing autophagy in glioblastoma cells. Transl Oncol 2025; 58:102424. [PMID: 40403475 DOI: 10.1016/j.tranon.2025.102424] [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: 12/15/2024] [Revised: 05/09/2025] [Accepted: 05/16/2025] [Indexed: 05/24/2025] Open
Abstract
Autophagy is an important factor in temozolomide (TMZ) resistance in glioblastoma (GBM). Receptor-interacting protein 2 (RIP2) is associated with autophagy, but its role and mechanism in regulating autophagy in GBM cells remain unclear. To analyze RIP2 expression in GBM in The Cancer Genome Atlas (TCGA) dataset. GBM cells were stimulated using recombinant human RIP2 protein (rRIP2) or RIP2 plasmid. Cell proliferation and apoptosis were assessed using CCK-8 assay and flow cytometry. Western blotting and immunofluorescence (IF) assays were performed to detect protein expression in cells and tumor tissues. Moreover, the relationship between RIP2-induced autophagy and TMZ resistance was verified in a GBM xenograft model. We determined that RIP2 expression was upregulated in GBM. rRIP2 and RIP2 overexpression induced TMZ resistance in the GBM cell lines. RIP2 overexpressing xenograft tumors have reduced sensitivity to TMZ. In addition, we showed that PD-L1 protein expression was upregulated in GBM tissues with RIP2 overexpression. rRIP2 and RIP2 overexpression induced autophagy in GBM cells through AMPK. Notably, RIP2 upregulated PD-L1 expression through the NF-κB signaling pathway, which induced autophagy and TMZ resistance in GBM cells. Moreover, NF-κB or autophagy inhibition reversed TMZ resistance in RIP2 overexpressing GBM cells in a xenograft model. In conclusion, RIP2 induces TMZ resistance in GBM cells by promoting autophagy through the NF-κB/PD-L1 signaling pathway, indicating that the RIP2/NF-κB/PD-L1 pathway may be therapeutic target for TMZ resistance.
Collapse
Affiliation(s)
- Xiaomeng Liu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Xiaosong Liu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Lei Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Jianliang Wu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Xiaoliang Wang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Yuhua Hu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China.
| |
Collapse
|
45
|
Chen D, Guo Z, Yao L, Sun Y, Dian Y, Zhao D, Ke Y, Zeng F, Zhang C, Deng G, Li L. Targeting oxidative stress-mediated regulated cell death as a vulnerability in cancer. Redox Biol 2025; 84:103686. [PMID: 40424719 DOI: 10.1016/j.redox.2025.103686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
Reactive oxygen species (ROS), regulators of cellular behaviors ranging from signaling to cell death, have complex production and control mechanisms to maintain a dynamic redox balance under physiological conditions. Redox imbalance is frequently observed in tumor cells, where ROS within tolerable limits promote oncogenic transformation, while excessive ROS induce a range of regulated cell death (RCD). As such, targeting ROS-mediated regulated cell death as a vulnerability in cancer. However, the precise regulatory networks governing ROS-mediated cancer cell death and their therapeutic applications remain inadequately characterized. In this Review, we first provide a comprehensive overview of the mechanisms underlying ROS production and control within cells, highlighting their dynamic balance. Next, we discuss the paradoxical nature of the redox system in tumor cells, where ROS can promote tumor growth or suppress it, depending on the context. We also systematically explored the role of ROS in tumor signaling pathways and revealed the complex ROS-mediated cross-linking networks in cancer cells. Following this, we focus on the intricate regulation of ROS in RCD and its current applications in cancer therapy. We further summarize the potential of ROS-induced RCD-based therapies, particularly those mediated by drugs targeting specific redox balance mechanisms. Finally, we address the measurement of ROS and oxidative damage in research, discussing existing challenges and future prospects of targeting ROS-mediated RCD in cancer therapy. We hope this review will offer promise for the clinical application of targeting oxidative stress-mediated regulated cell death in cancer therapy.
Collapse
Affiliation(s)
- Danyao Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China; Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziyu Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Lei Yao
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Deze Zhao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yizhe Ke
- The First Affliated Hospital of Shihezi University, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China.
| | - Linfeng Li
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
46
|
Zhang Y, Guan Y, Dai M, Yang Y, Yang F. Microcystin-LR induces lung injury in mice through the NF-κB/NLRP3 pathway. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:385-394. [PMID: 39773316 DOI: 10.1080/15287394.2024.2443525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Microcystin-LR (MC-LR) a cyclic toxin produced by cyanobacterial species is known to exert detrimental effects on various organs, including lung. Several investigators demonstrated that MC-LR exerts pulmonary toxicity, but the underlying mechanisms remain unclear. This study aimed to investigate whether exposure to MC-LR-induced lung inflammation and examine the underlying mechanisms. Thirty specific pathogen-free (SPF) male mice were allocated into control and MC-LR treatment groups. Mice were intraperitoneally injected with physiological saline or MC-LR (20 μg/kg) daily for a total of 21 days. Our findings indicated that exposure to MC-LR-produced histopathological changes in lung tissue, including thickening of alveolar walls and inflammatory infiltration. MC-LR was found to upregulate mRNA expression levels of pro-inflammatory cytokines TNFα, IL-6, IL-1β, and IL-18. Further, MC-LR significantly elevated the expression levels of proteins associated with the NF-κB/NLRP3 pathway p-NF-κB, NLRP3, Caspase-1, ASC. The activation of NF-κB/NLRP3 pathway further promoted the release of inflammatory cytokine IL-1β and cleavage of pyroptosis-associated GSDMD protein. These findings indicate that MC-LR may induce lung inflammation by promoting cell pyroptosis via the activation of the NF-κB/NLRP3 pathway.
Collapse
Affiliation(s)
- Yin Zhang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ying Guan
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Manni Dai
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yue Yang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Department of Public Health, The Central Hospital of Shaoyang, Shaoyang, China
| | - Fei Yang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| |
Collapse
|
47
|
Das D, Wu Y, Hong J. Signaling Pathways and Promising Small-Molecule Therapeutic Agents for Ischemic Stroke. ChemMedChem 2025; 20:e202400975. [PMID: 40025810 DOI: 10.1002/cmdc.202400975] [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: 12/04/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/04/2025]
Abstract
Stroke is the second highest cause of death and leading cause of disability with high economic burden worldwide. The incidence of stroke is increasing faster and more prevalent for the global population over age 65. Ischemic stroke (IS) has a higher incidence than hemorrhagic stroke, accounting over 80 % of the total incidence of stroke. The rate of ischemic stroke is increasing in all age groups and both sexes. In present era, hypertension, high blood pressure and modern lifestyle are considered as the causes of the disease. The treatment options for stroke is still limited, mainly thrombolytic and thrombectomy therapy are available options. In the past decade, a number of therapeutic agents have been studied for the acute ischemic stroke to protect the brain from ischemic injury. Several study methods focus to improve neurons functions around the ischemic core and protect from the shock. Many signalling pathways including NF-kB, NrF, Nrf2-Keap1, PI3K/AKT, JAK/STAT signalling pathways are strongly associated for the indication. Controlling the signalling pathways by small molecules potentially improve the neuronal functions. In this article, we review the recent advancement of the drug discovery, controlling the signalling pathways by small molecules, and kinase inhibitors in ischemic stroke.
Collapse
Affiliation(s)
- Debasis Das
- Arromax Pharmatech Co. Ltd., Sangtiandao Innovation Park, No. 1 Huayun Road, SIP, Suzhou, 215123, P. R. China
| | - Yimeng Wu
- Arromax Pharmatech Co. Ltd., Sangtiandao Innovation Park, No. 1 Huayun Road, SIP, Suzhou, 215123, P. R. China
| | - Jian Hong
- Arromax Pharmatech Co. Ltd., Sangtiandao Innovation Park, No. 1 Huayun Road, SIP, Suzhou, 215123, P. R. China
| |
Collapse
|
48
|
Choo MZY, Lim ET, Wong WSF, Chai CLL. Discovery of an NF-κB1 p105 Degrader for Anti-Inflammatory Therapy via Structural Optimization of the Coumarin Natural Product Minutuminolate. J Med Chem 2025. [PMID: 40378174 DOI: 10.1021/acs.jmedchem.5c00055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
In this study, the coumarin natural product minutuminolate (MNT) was used as a starting point for the development of anti-inflammatory agents. Through structure-activity relationship studies, a lead compound MD-1 was designed and synthesized, exhibiting significantly improved anti-inflammatory activities. Mechanistic studies revealed that MD-1 is a degrader of the p105 subunit of NF-κB. Gene knockdown experiments further showed that the Cullin-ring ligase (CRL) SCFβTrCP is involved in MD-1-induced p105 degradation. This leads to suppressed NF-κB transcriptional activity, which is consistent with its potent anti-inflammatory effects. Taken together, our work challenges the longstanding notion that NF-κB is undruggable, as we demonstrate that the p105 subunit of NF-κB is indeed tractable with small molecules. More importantly, our study highlights that natural products are valuable starting points for the discovery and development of anti-inflammatory agents with novel mechanisms of action.
Collapse
Affiliation(s)
- Malcolm Z Y Choo
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, 117543 Singapore
| | - En Tong Lim
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, 117543 Singapore
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, 16 Medical Drive, 117600 Singapore
- Drug Discovery and Optimization Platform, Yong Loo Lin School of Medicine, National University Health System, 117600 Singapore
| | - Christina L L Chai
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, 117543 Singapore
| |
Collapse
|
49
|
Li Y, Gong S, Yan K, Shi Z, Bao Y, Ning K. Artery tertiary lymphoid organs in atherosclerosis: A review. Life Sci 2025; 369:123549. [PMID: 40058576 DOI: 10.1016/j.lfs.2025.123549] [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: 11/06/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
Atherosclerosis (AS) is the common pathological basis for many cardiovascular diseases. Initial investigations into AS predominantly centered on endothelial immune responses associated with plaque formation. However, recent studies increasingly underscore the salutary immune modulation occurring on the aorta adventitia as the atheromatous plaque progresses. The immune responses extend from the intima of the vessel to the adventitia, and the artery tertiary lymphoid organ (ATLO) assumes a major immune role in advanced stages of AS, according to available studies conducted on ApoE-/- mice. In this review, we collate the history of studies on the participation of ATLOs in immunity to AS, detailing its structure, classification, cellular composition, and formation mechanisms. We elucidate the distinct roles of ATLO components in immune regulation, emphasizing unique features such as territorial organization, T cell-driven autoimmunity, and the T follicular helper-germinal center B cell axis, which distinguish ATLOs from conventional lymphoid responses. Furthermore, based on the latest research, we propose that ATLOs cooperate with the nervous system to regulate the progression of AS. Moreover, we highlight that aging has a great impact on the deterioration of AS and this impact is related to ATLOs. We conclude by suggesting that a focus on ATLOs is important for the clinical management of AS, and we offer a perspective for further research on ATLO and suggest whether it will be beneficial or detrimental to ATLOs.
Collapse
Affiliation(s)
- Yanni Li
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Sihe Gong
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Kaijie Yan
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Zhonghong Shi
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Yimin Bao
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China.
| | - Ke Ning
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Preston Research Building, Room 359, Nashville, TN 37232, United States.
| |
Collapse
|
50
|
Chen J, Fei S, Chan LWC, Gan X, Shao B, Jiang H, Li S, Kuang P, Liu X, Yang S. Inflammatory signaling pathways in pancreatic β-cell: New insights into type 2 diabetes pathogenesis. Pharmacol Res 2025; 216:107776. [PMID: 40378943 DOI: 10.1016/j.phrs.2025.107776] [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: 03/03/2025] [Revised: 04/27/2025] [Accepted: 05/10/2025] [Indexed: 05/19/2025]
Abstract
Type 2 diabetes (T2D) is a complex metabolic disorder with a growing global prevalence, and there is a linking between inflammation in pancreatic β-cell and impaired glucose homeostasis which has emerged as a key player in the pathogenesis of T2D. Recent advances in research have provided new insights into various inflammatory signaling cascades in β-cell among which we focus on Toll-like Receptor 4 (TLR4), Nuclear Factor kappa B (NF-κB), Janus Kinase-Signal Transducer and Activator of Transcription (JAK/STAT), Platelet-Derived Growth Factor Receptor α (PDGFR-α), Stimulator of Interferon Genes (STING), and the death receptor TMEM219. TLR4 activation by pathogen- or damage-associated molecular patterns initiates NF-κB and mitogen-activated protein kinase (MAPK) cascades, promoting pro-inflammatory cytokine release and β-cell apoptosis. NF-κB acts as a central hub, integrating metabolic stress signals (e.g., glucolipotoxicity, ER stress) and amplifying inflammatory responses through crosstalk with JAK/STAT and STING pathways. Meanwhile, JAK/STAT signaling exhibits dual roles in β-cell survival and inflammation, influenced by cytokine milieu and feedback regulation. PDGFR-α, traditionally linked to β-cell proliferation, paradoxically contributes to pathological hyperplasia in obesity, while STING activation by cytosolic DNA triggers β-cell senescence and ferroptosis via IRF3/NF-κB. In this review, we synthesize recent advancements of these inflammatory signaling pathways in β-cells, and current therapeutic strategies targeting TLR4/NF-κB inhibitors, JAK/STAT modulators, STING antagonists, and the death receptor TMEM219 are discussed, alongside challenges in pathway specificity and clinical translation. Understanding these inflammatory signaling pathways and their interactions in pancreatic β-cell is essential for the development of novel therapeutic strategies to prevent or treat T2D.
Collapse
Affiliation(s)
- Jie Chen
- Department of Respiratory Medicine, Huangshi Maternity and Children's Health Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi Key Laboratory of Birth Defects Prevention, Huangshi, Hubei 435000, PR China
| | - Shinuan Fei
- Pediatrics Department, Huangshi Maternal and Child Health Care Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi Key Laboratory of Birth Defects Prevention, Huangshi, Hubei 435003, PR China
| | - Lawrence W C Chan
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, 99077, Hong Kong
| | - Xueting Gan
- Department of Pathology, Huangshi maternal and Child Health Care Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi Key Laboratory of Birth Defects Prevention, Huangshi, Hubei 435003, PR China
| | - Bibo Shao
- Department of Intensive Care Unit, Huangshi Maternity and Children's Health Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi Key Laboratory of Birth Defects Prevention, Huangshi, Hubei 435000, PR China
| | - Hong Jiang
- Department of Laboratory Medicine, Huangshi Maternity and Children's Health Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi Key Laboratory of Birth Defects Prevention, Huangshi, Hubei 435000, PR China
| | - Sheng Li
- Department of Laboratory Medicine, Huangshi Maternity and Children's Health Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi Key Laboratory of Birth Defects Prevention, Huangshi, Hubei 435000, PR China
| | - Peng Kuang
- Huangshi Maternal and Child Health Hospital Huangshi Maternity and Children's Health Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi Key Laboratory of Birth Defects Prevention, Huangshi, Hubei 435000, PR China
| | - Xin Liu
- Department of Ultrasound Medicine Huangshi Maternity and Children's Health Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi Key Laboratory of Birth Defects Prevention, Huangshi, Hubei 435000, PR China
| | - Sijun Yang
- Department of Laboratory Medicine, Huangshi Maternity and Children's Health Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi Key Laboratory of Birth Defects Prevention, Huangshi, Hubei 435000, PR China.
| |
Collapse
|