1
|
Wang H, Han J, Zhang XA. Interplay of m6A RNA methylation and gut microbiota in modulating gut injury. Gut Microbes 2025; 17:2467213. [PMID: 39960310 PMCID: PMC11834532 DOI: 10.1080/19490976.2025.2467213] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/12/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
The gut microbiota undergoes continuous variations among individuals and across their lifespan, shaped by diverse factors encompassing diet, age, lifestyle choices, medication intake, and disease states. These microbial inhabitants play a pivotal role in orchestrating physiological metabolic pathways through the production of metabolites like bile acids, choline, short-chain fatty acids, and neurotransmitters, thereby establishing a dynamic "gut-organ axis" with the host. The intricate interplay between the gut microbiota and the host is indispensable for gut health, and RNA N6-methyladenosine modification, a pivotal epigenetic mark on RNA, emerges as a key player in this process. M6A modification, the most prevalent internal modification of eukaryotic RNA, has garnered significant attention in the realm of RNA epigenetics. Recent findings underscore its potential to influence gut microbiota diversity and intestinal barrier function by modulating host gene expression patterns. Conversely, the gut microbiota, through its impact on the epigenetic landscape of host cells, may indirectly regulate the recruitment and activity of RNA m6A-modifying enzymes. This review endeavors to delve into the biological functions of m6A modification and its consequences on intestinal injury and disease pathogenesis, elucidating the partial possible mechanisms by which the gut microbiota and its metabolites maintain host intestinal health and homeostasis. Furthermore, it also explores the intricate crosstalk between them in intestinal injury, offering a novel perspective that deepens our understanding of the mechanisms underlying intestinal diseases.
Collapse
Affiliation(s)
- Haixia Wang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Juanjuan Han
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Xin-An Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
2
|
Liu D, Lin S, Hu Y, Xiong J, Wan H, Chen Y, Ding T, Zhao H, Jiang R, Huang Z, Yao D, Li M, Zhu X, Yi B. HNRNPC stabilizes m6A-modified AC145207.5 to accelerate tumorigenesis in colorectal cancer by impeding the Nrf2/GPX4 axis-mediated ferroptosis. Noncoding RNA Res 2025; 13:43-56. [PMID: 40290566 PMCID: PMC12032884 DOI: 10.1016/j.ncrna.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 03/26/2025] [Accepted: 04/06/2025] [Indexed: 04/30/2025] Open
Abstract
Ferroptosis is an apoptosis-independent cell death pathway characterized by heightened lipid peroxidation, which shows promise for tumor suppression. Despite extensive research on long non-coding RNAs (LncRNAs) in ferroptosis, their role in colorectal cancer (CRC) remains underexplored. We investigated the upregulation of AC145207.5 and HNRNPC expression in CRC tissues through public dataset analysis and in-house validation, identifying them as having significant diagnostic potential. In vitro experiments including MTS assay, transwell, and colony formation, alongside in vivo studies using xenograft models, elucidated the synergistic carcinogenic role of the HNRNPC/AC145207.5 axis in promoting the malignant characteristics of CRC. Mechanistically, the m6A reader HNRNPC stabilized m6A-modified AC145207.5, contributing to its stabilization and upregulation. Consequently, AC145207.5 activated the Nrf2/GPX4 axis, resulting in increased GPX4 expression, inhibition of GPX4-mediated ferroptosis, and facilitation of CRC progression. Our findings underscore the clinical relevance of the HNRNPC/AC145207.5 axis in CRC and illuminate its regulatory role in ferroptosis, suggesting implications for targeted precision medicine in CRC.
Collapse
Affiliation(s)
- Dan Liu
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China
| | - Shanshan Lin
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China
| | - Yueben Hu
- Department of Pharmacy, Nanchang People's Hospital, 330009, PR China
| | - Jianyong Xiong
- 2nd Abdominal Surgery Department, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330029, PR China
| | - Hongtao Wan
- 2nd Abdominal Surgery Department, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330029, PR China
| | - Yanglin Chen
- 2nd Abdominal Surgery Department, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330029, PR China
| | - Taohui Ding
- 2nd Abdominal Surgery Department, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330029, PR China
| | - Hu Zhao
- 2nd Abdominal Surgery Department, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330029, PR China
| | - Renjie Jiang
- 2nd Abdominal Surgery Department, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330029, PR China
| | - Zhijiang Huang
- 2nd Abdominal Surgery Department, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330029, PR China
| | - Dengke Yao
- 2nd Abdominal Surgery Department, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330029, PR China
| | - Ming Li
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330029, PR China
| | - Xiaojian Zhu
- 2nd Abdominal Surgery Department, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330029, PR China
| | - Bo Yi
- 2nd Abdominal Surgery Department, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330029, PR China
| |
Collapse
|
3
|
Chen H, Wang X, Xing J, Pu Y, Ye H, Ma Y, Zhang J. Role and mechanisms of cuproptosis in the pathogenesis of Wilson's disease (Review). Int J Mol Med 2025; 56:117. [PMID: 40476570 PMCID: PMC12140093 DOI: 10.3892/ijmm.2025.5558] [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: 04/08/2025] [Accepted: 05/21/2025] [Indexed: 06/11/2025] Open
Abstract
Copper, an indispensable trace element in living organisms, plays a pivotal role in human physiological processes. Wilson's disease (WD), an inherited disorder of copper metabolism, is caused by mutations in the ATP7B gene. This genetic malfunction disrupts the dynamics of copper transport and metabolism, thereby impairing ceruloplasmin synthesis and copper excretion. The resultant accumulation of copper in various tissues and organs precipitates a cascade of cellular demise and functional impairment. Notably, cuproptosis, a recently discovered copper‑dependent regulated cell death mechanism, distinctly deviates from conventional cell death paradigms. This novel mode of cell death involves the interaction of copper with lipoacylated proteins within the tricarboxylic acid cycle, leading to proteinotoxic stress and culminating in cell death. In the realm of pathophysiology, cuproptosis has emerged as a pivotal player in a spectrum of diseases, with WD standing as a paradigm closely intertwined with the dysregulation of copper metabolism. This study aimed to encapsulate the pivotal molecular underpinnings of cuproptosis and delve into its crucial involvement in the etiopathogenesis of WD. By elucidating these mechanisms, the present analysis contributes significantly to the nuanced understanding of the pathological underpinnings of WD, thereby providing fresh insights and evidence that may direct innovative therapeutic strategies for this condition.
Collapse
Affiliation(s)
| | | | - Jin Xing
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Yue Pu
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Hao Ye
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Ying Ma
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Juan Zhang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui 230031, P.R. China
| |
Collapse
|
4
|
Wang Y, Wu Q, Guo W, Chen Z, Tan L, Fu C, Ren X, Zhang J, Meng X, Gu B. Dual-upregulation of p53 for self-sensitized cuproptosis via microwave dynamic and NO gas therapy. J Colloid Interface Sci 2025; 691:137421. [PMID: 40154167 DOI: 10.1016/j.jcis.2025.137421] [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: 01/20/2025] [Revised: 03/21/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
Cuproptosis-a novel cell death mechanism-is an innovative strategy for tumor therapy. However, the insufficient efficacy of cuproptosis, primarily owing to the low sensitivity of tumor cells to Cu ions, remains a major challenge. In this study, we design TiCuMOF@PEG@l-Arg@TPP (TCPAT) nanoparticles to facilitate self-sensitized cuproptosis for anti-tumor therapy through the dual upregulation of p53. TiMOF serves as a microwave sensitizer by generating reactive oxygen species (ROS). Notably, the uniformly distributed Cu ions within the MOF serve as co-catalysts to provide reactive sites that enhance ROS generation. Additionally, the ROS generated are utilized to oxidize l-arginine, thus resulting in the release of nitric oxide (NO), which has a long half-life and diffusion distance, thereby enabling it to penetrate deep into the tumor regions that are typically inaccessible to ROS. Furthermore, TCPAT not only induces cuproptosis but also leverages the efficiently generated ROS and cascade-released NO for the dual upregulation of p53. This upregulation subsequently inhibits glycolysis, increases cellular sensitivity to Cu ions, and facilitates self-sensitized cuproptosis. Consequently, the self-sensitized cuproptosis strategy, dependent on the efficient generation of ROS, presents a promising avenue for tumor therapy based on cuproptosis mechanisms.
Collapse
Affiliation(s)
- Yuxin Wang
- State Key Laboratory of Cryogenic Science and Technology, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China; Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Qiong Wu
- State Key Laboratory of Cryogenic Science and Technology, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China; Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Wenna Guo
- State Key Laboratory of Cryogenic Science and Technology, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China; Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Zengzhen Chen
- State Key Laboratory of Cryogenic Science and Technology, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China; Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Longfei Tan
- State Key Laboratory of Cryogenic Science and Technology, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China; Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Changhui Fu
- State Key Laboratory of Cryogenic Science and Technology, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China; Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Xiangling Ren
- State Key Laboratory of Cryogenic Science and Technology, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China; Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Jiqing Zhang
- Department of Urology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China.
| | - Xianwei Meng
- State Key Laboratory of Cryogenic Science and Technology, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China; Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China.
| | - Bin Gu
- Department of Stomatology, The First Medical Center of PLA General Hospital, Beijing 100853, PR China.
| |
Collapse
|
5
|
Zhao L, Chang Y, Sun X, Chen H, Li N, Ma T, Jin S. LncRNA SNHG12 promotes EMT and metastasis of colorectal cancer via regulating TGF-β/Smad2/3 signaling pathway. Mol Immunol 2025; 183:225-235. [PMID: 40412277 DOI: 10.1016/j.molimm.2025.05.017] [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: 03/20/2025] [Revised: 04/21/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
OBJECTIVE In this study, we aimed to explore the molecular mechanism of SNHG12 promoting colorectal cancer (CRC) progression. METHODS Bioinformatics technology was utilized to identify SNHG12-targeted mRNA and the correlation with the prognosis of CRC patients. Transfected sequence of knockdown SNHG12 in HCT-116 cell line was established. CCK8 assay, colone formation assay, flow cytometry, cell migration and transwell assay were applied to detect the impact of SNHG12 on HCT-116 cells. Besides, qRT-PCR and western blot were employed to evaluate the apoptotic and EMT markers as well as the expression of TGF-β and p-Smad2/3. Additionally, the rescue test of overexpressing TGF-β and a nude mouse subcutaneous tumor model were established to validate the pivotal role of SNHG12 in driving the progression of CRC. RESULTS SNHG12 could predict the prognosis of CRC patients, and a target mRNA GOLT1B was obtained from bioinformatics. In vitro results indicated that SNHG12 facilitated the proliferation, migration, and invasion of HCT-116 cells. qRT-PCR and western blot showed SNHG12 was related to the expression of Caspase 3, EMT markers as well as TGF-β and p-Smad2/3. Meanwhile, the rescue experiment proved that overexpressed TGF-β had the ability to reverse the impact of SNHG12 knockout on cell function and phenotype. In vivo, SNHG12 knockdown significantly reduced tumor growth. CONCLUSION SNHG12 promotes EMT and metastasis of CRC by modulating the TGF-β/Smad2/3 signaling pathway and EMT process, which could function as a prognostic biomarker and a treatment target for CRC.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province 150086, China
| | - Yuan Chang
- Department of Rheumatology and Immunology,The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province 150086, China
| | - Xiaoli Sun
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province 150086, China
| | - Hongliang Chen
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province 150086, China
| | - Ning Li
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province 150086, China
| | - Tianyi Ma
- Department of Colorectal Surgery, Cancer Center,The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province 150086, China
| | - Shizhu Jin
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province 150086, China.
| |
Collapse
|
6
|
Akano OP, Amos VO, Fawole IO, Oyindamola AC, Ajayi AF. Epigenetics' Responsibility in Endometriosis: A comprehensive assessment. J Steroid Biochem Mol Biol 2025; 253:106814. [PMID: 40513963 DOI: 10.1016/j.jsbmb.2025.106814] [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: 04/17/2025] [Revised: 05/23/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025]
Abstract
Endometriosis is a chronic inflammatory disease characterized by the presence of endometrial-like tissue outside the uterus, affecting women of reproductive age. Despite extensive research, its pathophysiology remains unclear, with genetic, hormonal, and environmental factors playing interconnected roles. Epigenetic processes, including non-coding RNAs, histone modifications, and DNA methylation, have been implicated in the genesis and progression of endometriosis. These modifications impact physiological functions such as inflammation, cell division, apoptosis, and hormone sensitivity. Recent findings on epigenetic alterations in endometriosis highlight their role in the abnormal behavior of ectopic endometrial-like cells. Aberrant DNA methylation patterns in genes related to immunological control and oestrogen metabolism contribute to the invasiveness and durability of lesions. Histone modifications, such as methylation and acetylation, regulate gene expression by altering chromatin structure. Non-coding RNAs, particularly microRNAs, influence tissue remodeling and inflammation. Given the reversible nature of epigenetic modifications, they present promising therapeutic targets for innovative treatment strategies. Epigenetic-based therapies aim to reverse pathological gene expression patterns, offering hope for more personalized and effective management of endometriosis. Further research is needed to fully utilize epigenetic processes in treating this debilitating disease.
Collapse
Affiliation(s)
| | - Victor Oluwaloseyi Amos
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria.
| | | | | | - Ayodeji Folorunsho Ajayi
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria; Anchor Biomed Research Institute, Ogbomoso, Oyo State, Nigeria; Department of Physiology, Adeleke University, Ede, Osun State, Nigeria.
| |
Collapse
|
7
|
Hussain MS, Jakhmola V, Goyal K, Rekha A, Sultana A, Ali H, Gupta G. Targeting the NAD+/SIRT1 axis: A metabolic strategy to overcome oxaliplatin resistance in colorectal cancer. World J Gastroenterol 2025; 31:106530. [DOI: 10.3748/wjg.v31.i21.106530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/21/2025] [Accepted: 04/16/2025] [Indexed: 06/06/2025] Open
Abstract
Oxaliplatin resistance remains a significant clinical challenge in colorectal cancer (CRC), highlighting the urgent need to identify novel molecular targets for therapeutic intervention. Recent findings by Niu et al have elucidated the role of the NAD+/SIRT1 axis in mediating oxaliplatin resistance through metabolic reprogramming. Their study demonstrated that oxaliplatin-induced DNA damage activates PARP, resulting in NAD+ depletion and subsequent downregulation of SIRT1. This reduction in SIRT1 levels enhances glycolysis, as evidenced by increased expression of PKM2 and LDHA, thereby conferring a metabolic advantage to resistant CRC cells. Conversely, restoration of SIRT1 expression reverses resistance, while pharmacological inhibition of glycolysis effectively sensitizes cells to oxaliplatin. These findings underscore the therapeutic potential of targeting the NAD+/SIRT1 pathway as a metabolic vulnerability in CRC. Future studies should investigate the clinical feasibility of combining SIRT1 agonists and glycolysis inhibitors with oxaliplatin to overcome drug resistance and improve patient outcomes.
Collapse
Affiliation(s)
- Md Sadique Hussain
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehra Dun 248007, Uttarākhand, India
| | - Vikash Jakhmola
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehra Dun 248007, Uttarākhand, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Dehra Dun 248002, Uttarākhand, India
| | - Arcot Rekha
- Department of General Surgery, Dr. D.Y. Patil Medical College, Hospital and Research Centre, Pune 411018, Mahārāshtra, India
| | - Ayesha Sultana
- Department of Pharmaceutics, Sree Dattha Institute of Pharmacy, Hyderabad 501510, Telangāna, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 602105, Tamil Nādu, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman 13306, United Arab Emirates
| |
Collapse
|
8
|
Li N, Wei X, Dai J, Yang J, Xiong S. METTL3: a multifunctional regulator in diseases. Mol Cell Biochem 2025; 480:3429-3454. [PMID: 39853661 DOI: 10.1007/s11010-025-05208-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/04/2025] [Indexed: 01/26/2025]
Abstract
N6-methyladenosine (m6A) methylation is the most prevalent and abundant internal modification of mRNAs and is catalyzed by the methyltransferase complex. Methyltransferase-like 3 (METTL3), the best-known m6A methyltransferase, has been confirmed to function as a multifunctional regulator in the reversible epitranscriptome modulation of m6A modification according to follow-up studies. Accumulating evidence in recent years has shown that METTL3 can regulate a variety of functional genes, that aberrant expression of METTL3 is usually associated with many pathological conditions, and that its expression regulatory mechanism is related mainly to its methyltransferase activity or mRNA posttranslational modification. In this review, we discuss the regulatory functions of METTL3 in various diseases, including metabolic diseases, cardiovascular diseases, and cancer. We focus mainly on recent progress in identifying the downstream target genes of METTL3 and its underlying molecular mechanisms and regulators in the above systems. Studies have revealed that the use of METTL3 as a therapeutic target and a new diagnostic biomarker has broad prospects. We hope that this review can serve as a reference for further studies.
Collapse
Affiliation(s)
- Na Li
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiang Wei
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jian Dai
- Department of Critical Care Medicine, Wuhan Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Jinfeng Yang
- Department of Medical Affairs, Wuhan Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China.
| | - Sizheng Xiong
- Department of Vascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
9
|
Wang Z, Zhang HT, Li SY, Song XP, Shi CZ, Zhang YW, Han F. An integrative study on the effects of Lingguizhugan decoction in treating Alzheimer's disease rats through modulation of multiple pathways involving various components. Comput Biol Med 2025; 191:110149. [PMID: 40228446 DOI: 10.1016/j.compbiomed.2025.110149] [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: 10/29/2024] [Revised: 03/16/2025] [Accepted: 04/04/2025] [Indexed: 04/16/2025]
Abstract
OBJECTIVE To explore the active components and mechanisms of Lingguizhugan decoction (LGZGD) in the treatment of Alzheimer's disease (AD) through an integrated approach. METHODS The active components of LGZGD in rat serum were identified using HPLC-FTICR MS. Network pharmacology and molecular docking analyses were conducted, and their findings were validated using an Aβ1-42-induced AD rat model. RESULTS Twenty-four active components and 324 common targets were identified and used to construct the networks. KEGG pathway enrichment analysis linked key target genes with MAPK, Rap1, and NF-κB signaling pathways. Molecular docking results indicated that three key targets (IL-6, TNF, and EGFR) and 10 core components are closely associated with LGZGD in the treatment of AD. LGZGD improved the spatial learning and memory abilities of AD rats. LGZGD reduced neuronal damage and increased the number of neurons in the cortex and hippocampal CA1 region of AD rats. LGZGD decreased Aβ1-42 expression in the rat hippocampus, alleviated oxidative stress in AD rats, and decreased TNF-α, IL-6, IL-1β, and HMGB1 levels in the cerebral cortical tissue. LGZGD markedly decreased Iba-1 and iNOS expression and increased CD206 levels to inhibit M1 activation and promote M2 activation. LGZGD increased the expression of p-GSK-3β, ERK, and p-ERK, while decreasing the expression of p-Tau, IKKβ, p-IκBα, p-p65, p-p38, and p-JNK in the hippocampus of AD rats. CONCLUSION LGZGD treats AD by modulating targets like IL-6, TNF, MAPK3, and BCL2, thereby alleviating cognitive impairments in rats. Its neuroprotective effects in treating AD are mediated through the NF-κB/MAPK signaling pathways.
Collapse
Affiliation(s)
- Zhe Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, No.103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Hao-Tian Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, No.103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Si-Yue Li
- School of Pharmacy, Shenyang Pharmaceutical University, No.103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Xiu-Ping Song
- School of Pharmacy, Shenyang Pharmaceutical University, No.103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Chong-Zhen Shi
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Ye-Wen Zhang
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, People's Republic of China
| | - Fei Han
- School of Pharmacy, Shenyang Pharmaceutical University, No.103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| |
Collapse
|
10
|
Jafari Z, Honarmand S, Abbasirad Z, Sadeghi S, Bigham A. The Therapeutic Potential of Berberine in Lung Cancer. Arch Pharm (Weinheim) 2025; 358:e70013. [PMID: 40490958 DOI: 10.1002/ardp.70013] [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: 09/05/2024] [Revised: 04/17/2025] [Accepted: 04/30/2025] [Indexed: 06/11/2025]
Abstract
Lung cancer has the second-highest incidence rate after breast cancer and remains the leading cause of cancer-related mortality. The 1-year survival rate for lung cancer patients is below 50%, highlighting the urgent need for novel therapeutic strategies and drug development. Phytochemicals and their derivatives have been widely explored for their anticancer properties, serving as chemotherapeutic agents against various types of cancer. One of these herbal compounds, berberine (BBR), a quaternary isoquinoline alkaloid, has shown significant promise in preclinical studies and is currently undergoing clinical trials for cancer treatment. BBR exhibits diverse biological activities, contributing to its anticancer potential, including antioxidant, antidiarrheal, antidiabetic, antimicrobial, and so on. However, despite its multifunctional therapeutic potential, BBR faces several limitations, hindering its clinical application, like poor bioavailability, low tissue uptake, a short plasma half-life, and rapid metabolic elimination. To address these challenges, various targeted drug delivery approaches have been developed to improve its efficacy. This review aims to provide a comprehensive overview of drug delivery strategies designed to encapsulate BBR for enhanced lung cancer therapy, highlighting the most recent advancements in the field. Moreover, the molecular structure of BBR and the biological pathways it targets to inhibit lung cancer progression are discussed in detail. Finally, BBR-encapsulated nanocarriers specifically developed for lung cancer therapy are evaluated in terms of their benefits, limitations, and overall therapeutic potential.
Collapse
Affiliation(s)
- Zohreh Jafari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shokouh Honarmand
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zeinab Abbasirad
- Institute of Petroleum Engineering, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Sahar Sadeghi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ashkan Bigham
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy (IPCB-CNR), Naples, Italy
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Naples, Italy
| |
Collapse
|
11
|
Chen WC, Fang QQ, Qin Y, Li RS, Zhang QG, Liu W, Wang P, Zhu SX. Analysis of Chemical and Potential Anti-Heart Failure Components of Yan-Shi-Qiang-Xin Decoction by UHPLC-Q-Orbitrap HRMS Coupled With Molecular Networking. Biomed Chromatogr 2025; 39:e70100. [PMID: 40312847 DOI: 10.1002/bmc.70100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/03/2025]
Abstract
Yan-Shi-Qiang-Xin decoction (YSQXD), a traditional Chinese medicine formula, is clinically effective in treating chronic heart failure, yet its bioactive constituents remain unclear. To address this, a sensitive and reliable UHPLC-Q-Exactive Orbitrap HRMS method was established to separate and identify the chemical constituents in YSQXD and its absorbed constituents in rat serum, heart and liver following oral administration of YSQXD. With the optimized conditions, a total of 134 chemical components were tentatively identified, including 41 terpenoids, 34 flavonoids, 19 alkaloids, nine organic acids, eight amino acids, seven coumarins, six nucleotides, two sterols, and eight other compounds, based on retention times, MS/MS spectra, and literature references. Furthermore, 29, 10, 17 constituents were identified in the rat serum, heart and liver, respectively. Finally, the network pharmacology analysis based on absorbable components indicated that polyporenic acid, bavachalcone, albiflorin, biatractylolide, psoralen, angelicin, neobavaisoflavone, dictysine, isotalatizidine, and 26-hydroxyporicoic acid G exhibited high degree values, suggesting their potential as active ingredients for chronic heart failure treatment. These findings provide a comprehensive chemical profile of YSQXD and its absorbed components, offering valuable insights into its pharmacologically active substances.
Collapse
Affiliation(s)
- Wen-Chao Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qin-Qin Fang
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Qin
- Department of Internal Medicine of TCM, Shanghai Municipal Hospital of Traditional Chinese Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rong-Sheng Li
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qing-Guang Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Liu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Si-Xing Zhu
- Institute of Science, Technology and Humanities, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
12
|
Jia X, Qiang W, Chang L, Xiao K, Zhou R, Qiu Q, Jiang G, Li X, Chi C, Liu W, Zhang D. Integrative whole-genome methylation and transcriptome analysis reveals epigenetic modulation of glucose metabolism by dietary berberine in blunt snout bream (Megalobrama amblycephala). Comp Biochem Physiol B Biochem Mol Biol 2025; 278:111098. [PMID: 40250795 DOI: 10.1016/j.cbpb.2025.111098] [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: 01/16/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
The present research was designed to explore the epigenetic mechanism by which dietary berberine (BBR) affects glucose metabolism in fish. Blunt snout bream (Megalobrama amblycephala) is susceptible to disturbances in glucose metabolism when subjected to prolonged high-carbohydrate diets. This study aimed to elucidate whether BBR can enhance glucose regulation in M. amblycephala via modulating DNA methylation levels. Fish (average weight of 20.36 ± 1.44 g) were administered a normal-carbohydrate diet (NC, 30 % carbohydrate), a high-carbohydrate diet (HC, 43 % carbohydrate), or a high-carbohydrate diet supplemented with 50 mg/kg berberine (HB) for 10 weeks. Subsequently, global DNA methylation level, whole-genome bisulfite sequencing (WGBS), RNA-seq, bisulfite sequencing PCR, and real-time quantitative PCR were employed to analyze the DNA methylation patterns and transcription results of the liver genome. The findings indicated that high carbohydrate diets induced glucose metabolism disorders in M. amblycephala, whereas BBR mitigated these metabolic disturbances by reducing methylation levels. WGBS results revealed that CG-type cytosine methylation predominated, and that DNA methylation mainly occurred in promoter, intron, and exon regions. Furthermore, analyses demonstrated a negative correlation between DNA methylation around the transcriptional start site and gene expression levels for 47 genes. Functional enrichment analysis revealed that these genes were associated with 60 KEGG pathways, including 12 genes implicated in the amelioration of insulin resistance, reduction of gluconeogenesis, and maintenance of glucose homeostasis. Consequently, we generated a comprehensive catalog of liver DNA methylation in M. amblycephala, which provides a foundational framework for future investigations into the epigenetic regulation of glucose metabolism by BBR.
Collapse
Affiliation(s)
- Xiaoyan Jia
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wei Qiang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Le Chang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Kang Xiao
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Ronghua Zhou
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Qiyong Qiu
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Guangzhen Jiang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangfei Li
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Cheng Chi
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wenbin Liu
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Dingdong Zhang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
13
|
Zheng L, Lu Z, Ma Y, Cui P, Zhang X, Gan J, Li G. Hawthorn total flavonoids ameliorate hyperlipidemia through AMPK/SREBP1-c and PPARα/PGC-1α/CPT-1A pathway activation and gut microbiota modulation. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:4326-4337. [PMID: 40013442 DOI: 10.1002/jsfa.14188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/02/2025] [Accepted: 01/20/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND The increased prevalence of hyperlipidemia significantly affects human health worldwide. Although drug treatment is very effective, the harm to the human body cannot be ignored. Improvement of lipid metabolism by natural medicinal and food homologous products is an effective approach to ameliorate hyperlipidemia and it has gradually become a research focus. In this research, we adopted HepG2 cell models and high-fat-diet-fed C57BL/6j mouse models to explore the effect of hawthorn total flavonoids (HTF) on hyperlipidemia. Moreover, we utilized western blot and gut microbiota analysis to elucidate the specific mechanism of HTF's influence on hyperlipidemia. RESULTS We found that HTF significantly alleviated hyperlipidemia and its complications, as manifested by reduced body weight gain and fat accumulation, and improved the disorder of intestinal microorganisms. HTF protected the liver, reducing aspartate transaminase and lactate dehydrogenase levels, and ameliorating inflammatory infiltration. Fat droplet amounts and necrotic cell numbers in liver cells were also decreased. Mechanistically, HTF promoted AMP-activated protein kinase phosphorylation, inhibited sterol regulatory element binding protein 1c expression, downregulating the expression of lipid synthesis-related proteins (acetyl CoA carboxylase, fatty acid synthase, 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase), thus suppressing liver lipid synthesis. HTF also functioned as a natural peroxisome proliferator-activated receptor α (PPARα) agonist. Activated PPARα enhanced mitochondrial oxidation and lipid consumption via upregulating carnitine palmitoyltransferase 1A. Peroxisome proliferator-activated receptor-γ coactivator expression was also elevated, activating mitochondrial activity, increasing cholesterol 7α-hydroxylase activity and cholesterol consumption, and reducing blood lipids. Additionally, HTF regulated intestinal flora abundance, restored the ratio of Firmicutes to Bacteroidetes, balanced gut-liver axis crosstalk, and alleviated hyperlipidemia. CONCLUSION The results demonstrated that HTF alleviated the pathological symptoms caused by hyperlipidemia, and had a certain protective effect on the liver. HTF also stimulated the lipid metabolism pathway and accelerated lipid consumption. © 2025 Society of Chemical Industry.
Collapse
Affiliation(s)
- Liping Zheng
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries, Beijing, China
| | - Zhihao Lu
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries, Beijing, China
| | - Yurong Ma
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries, Beijing, China
| | - Penglei Cui
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries, Beijing, China
| | - Xinxue Zhang
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries, Beijing, China
| | - Jing Gan
- College of Life Science, Yantai University, Yantai, China
| | - Guoming Li
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries, Beijing, China
| |
Collapse
|
14
|
Zhou K, Cai H, Zhou Z, Yi D, Yao Y, Jin Z, Huang P. m6A methylation modification of RNA plays a significant role in the occurrence and development of colorectal cancer. Int J Biol Macromol 2025; 315:144666. [PMID: 40424908 DOI: 10.1016/j.ijbiomac.2025.144666] [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: 03/19/2025] [Revised: 05/23/2025] [Accepted: 05/24/2025] [Indexed: 05/29/2025]
Abstract
Colorectal cancer is the third most common malignant tumor worldwide and ranks second in terms of mortality. N6-methyladenosine (m6A) modification is the most prevalent internal covalent modification in eukaryotic mRNA and is involved in various stages of RNA processing, including splicing, degradation, and export, playing a crucial role in the onset and progression of many diseases. The m6A modification is co-regulated by methyltransferases, demethylases, and methyl-binding proteins, and it has become a hot topic in cancer research. Based on a systematic review of existing studies on the role of m6A modification in colorectal cancer, this article further expands the research horizon in this field and effectively overcomes the limitations of existing reviews that only focus on discussing a single or a class of methylation regulators.
Collapse
Affiliation(s)
- Ke Zhou
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, PR China
| | - Huazhong Cai
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, PR China
| | - Zhengrong Zhou
- School of Medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Dehao Yi
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, PR China
| | - Yuan Yao
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, PR China
| | - Zhesi Jin
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, PR China
| | - Pan Huang
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, PR China; School of Medicine, Jiangsu University, Zhenjiang 212013, PR China.
| |
Collapse
|
15
|
Thomas A, Thomas A. Managing Nonalcoholic Fatty Liver Disease Through Structured Lifestyle Modification Interventions. Am J Lifestyle Med 2025:15598276251346717. [PMID: 40438150 PMCID: PMC12106371 DOI: 10.1177/15598276251346717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 05/14/2025] [Accepted: 05/16/2025] [Indexed: 06/01/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a significant global health burden. It comprises a broad pathological spectrum ranging from simple liver steatosis to steatohepatitis with variable degrees of fibrosis, and liver failure. Patients with NAFLD have an increased risk of liver-related and overall mortality. While the trials to assess the efficacy of the medications are ongoing, lifestyle modification is the first line of therapy recommended. The primary aim of this review paper is to synthesize literature related to current evidence-based lifestyle interventions for preventing and managing NAFLD. The review and synthesis of the literature reveal that personalized nutritional, exercise, and behavior change interventions are effective in managing NAFLD. Evidence suggests that there are several gaps in managing NAFLD. The gaps discussed in this paper include a lack of awareness of the disease, ineffective patient-provider communication, shortage of specialists, under-recognition of the disease, and liver health disparities. This paper highlights the evidence-based opportunities to overcome those gaps, such as utilizing comprehensive models of care, clinical care pathways, and clinical practice guidelines. Primary care physicians and endocrinologists, who are the first point of contact must utilize these opportunities for diagnosing and managing patients with NAFLD.
Collapse
Affiliation(s)
- Andrew Thomas
- Internal Medicine, Southern Illinois Healthcare, Carbondale, IL, USA (AT)
| | - Annie Thomas
- Marcella Niehoff School of Nursing, Loyola University Chicago, Maywood, IL, USA (AT)
| |
Collapse
|
16
|
Grossini E, Ola Pour MM, Venkatesan S. The Role of Extracellular Vesicles in the Pathogenesis of Metabolic Dysfunction-Associated Steatotic Liver Disease and Other Liver Diseases. Int J Mol Sci 2025; 26:5033. [PMID: 40507843 PMCID: PMC12154092 DOI: 10.3390/ijms26115033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 05/09/2025] [Accepted: 05/19/2025] [Indexed: 06/16/2025] Open
Abstract
The increasing prevalence of liver diseases, such as metabolic dysfunction-associated steatotic liver disease (MASLD), presents considerable medical challenges, particularly given the absence of approved pharmacological treatments, which underscores the necessity to comprehend its underlying mechanisms. Extracellular vesicles (EVs), which are tiny particles released by cells, play a crucial role in facilitating communication and can transport harmful molecules that promote inflammation and tissue damage. These EVs are involved in the progression of various types of liver disorders since they aggravate inflammation and oxidative stress. Because of their critical role, it is believed that EVs are widely involved in the initiation and progression of MASLD, as well as in viral hepatitis, alcoholic liver disease, drug-induced liver injury, and hepatocellular carcinoma. This review emphasizes recent findings regarding the functions of EVs in the above liver pathologies and underscores their potential as new therapeutic targets, paving the way for innovative approaches to address those detrimental liver conditions.
Collapse
Affiliation(s)
- Elena Grossini
- Laboratory of Physiology, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (M.M.O.P.); (S.V.)
| | | | | |
Collapse
|
17
|
Műzes G, Sipos F. PANoptosis as a Two-Edged Sword in Colorectal Cancer: A Pathogenic Mechanism and Therapeutic Opportunity. Cells 2025; 14:730. [PMID: 40422233 DOI: 10.3390/cells14100730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2025] [Revised: 05/14/2025] [Accepted: 05/16/2025] [Indexed: 05/28/2025] Open
Abstract
The examination of PANoptosis in colorectal cancer is particularly important, as many tumor cells can evade apoptotic cell death while continuing to proliferate through inflammatory mediators and creating an immunosuppressive environment. The PANoptosome functions as a regulatory complex that unites proteins governing pyroptotic, apoptotic, and necroptotic pathways, rather than allowing distinct death pathways to compete. The expression and functional status of key molecules within the PANoptosome, such as ZBP1, RIPK1, RIPK3, CASP8, and ASC, may influence tumor viability and immune detection. The tumorigenic impact of PANoptosis is complex and predominantly manifests through chronic inflammation, immune response modulation, and changes in the tumor microenvironment. PANoptosis also aids in the defense against colon cancer by directly eradicating tumor cells and modifying the cellular environment. The expression profile of PANoptosis components may possess prognostic and predictive significance. The therapeutic ramifications of PANoptosis in colorectal cancer are now being investigated through many avenues. It provides an opportunity to develop targeted therapeutic techniques. In contrast, it may also be pertinent in conjunction with immunotherapy, as PANoptosis signifies an immunogenic type of cell death and may consequently enhance the anti-tumor immune response. A thorough comprehension of how these parameters influence PANoptosis is crucial for practical implementation.
Collapse
Affiliation(s)
- Györgyi Műzes
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| | - Ferenc Sipos
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| |
Collapse
|
18
|
Yang W, Wu X, Wang J, Ou W, Huang X. Integrated single-cell and transcriptome sequencing data reveal the value of IL1RAP in gastric cancer microenvironment and prognosis. Front Oncol 2025; 15:1584619. [PMID: 40444099 PMCID: PMC12119286 DOI: 10.3389/fonc.2025.1584619] [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: 02/27/2025] [Accepted: 04/23/2025] [Indexed: 06/02/2025] Open
Abstract
Background Investigating the pivotal role of IL1RAP in the tumor microenvironment of gastric cancer. Method Download and collate transcriptomic and single-cell data from gastric cancer patients. Three machine learning algorithms identified distinct sets of prognostic genes in gastric cancer patients. The CIBERSORT and ssGSEA algorithms elucidated immune infiltration patterns, while TIDE and TCGA predicted immune-related outcomes. Furthermore, single-cell sequencing data confirmed the interaction of IL1RAP within the tumor microenvironment. Finally, differential expression levels of IL1RAP protein and mRNA were validated. Result After machine learning screening and independent dataset validation, high IL1RAP expression was identified as a poor prognostic factor for gastric cancer patients. Immune infiltration analysis indicated that the low IL1RAP expression group was associated with higher infiltration of CD8+ T cells and M1-type macrophages, whereas the high IL1RAP expression group exhibited increased presence of M2-type macrophages. Immunotherapy prediction models suggested a more favorable response to PD-1 treatment in the low IL1RAP expression group. Prognostic models incorporating IL1RAP demonstrated superior predictive performance. Single-cell data analysis revealed that IL1RAP plays a critical role in regulating intercellular communication within the tumor microenvironment. Our findings were further validated by confirming elevated IL1RAP expression levels in gastric cancer tissues. Conclusion IL1RAP plays a critical role in the tumor microenvironment of gastric cancer and serves as a robust predictor of immunotherapy efficacy in gastric cancer.
Collapse
Affiliation(s)
| | | | | | | | - Xing Huang
- Gastrointestinal Surgery, Nanping First Hospital Affiliated to Fujian Medical
University, Nanping, Fujian, China
| |
Collapse
|
19
|
Bhattacharya I, Maity DK, Kumar A, Sarkar S, Bhattacharya T, Sahu A, Sreedhar R, Arumugam S. Beyond obesity: lean metabolic dysfunction-associated steatohepatitis from unveiling molecular pathogenesis to therapeutic advancement. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04257-x. [PMID: 40366398 DOI: 10.1007/s00210-025-04257-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD), now known by the name of metabolic dysfunction-associated fatty liver disease (MAFLD), with increased global incidence, has been recognized as a significant metabolic disorder. NAFLD includes a spectrum liver disease from hepatocellular fat accumulation (isolated steatosis) to an advanced form of liver injury known as nonalcoholic steatohepatitis (NASH), which refers to distinct histologic features, including hepatocellular steatosis and injury, necroinflammation, and eventually fibrosis. Nonobese or lean individuals associated with metabolic dysregulation usually demonstrated diverse risk factors compared to obese MAFLD. The presence of normal range body mass index (BMI) and excess visceral adiposity with increased cardiometabolic and renal comorbidities, along with sarcopenia, has been evidenced to be associated with lean MASH. Genetic predispositions accompanying lifestyle and environmental factors contribute to disease initiation and progression. The genetic influence in pathophysiology indicated the significant contributions of the following genes: PNPLA3, TM6SF2, APOB, LIPA, MBOAT7, and HSD17B13, and the impact of their disease-specific variants in the development of obesity-independent MASH. The epigenetic modifications exhibited differential DNA methylation patterns in the genes involved in lipid metabolism, particularly hypomethylation of PEMT. Diet-induced and genetic animal models of lean MASH, including Slc: Wistar/ST rats, PPAR-α, PTEN, and MAT1A knockout mice models, are indicated to be pivotal in the exploration of disease progression and observing the effect of therapeutic interventions. This comprehensive review comprises the molecular and genetic pathophysiology, molecular diagnostics, and therapeutic aspects of lean MASH to enunciate a diagnostic approach that combines detailed clinical phenotyping regarding genomic analysis.
Collapse
Affiliation(s)
- Indrajit Bhattacharya
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Deep Kumar Maity
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Amit Kumar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Sampriti Sarkar
- School of Biosciences & Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, 632014, India
| | - Teeshyo Bhattacharya
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Amrita Sahu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Remya Sreedhar
- School of Pharmacy, Sister Nivedita University, DG Block, Action Area I, 1/2, Newtown, Kolkata, 700156, West Bengal, India
| | - Somasundaram Arumugam
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India.
| |
Collapse
|
20
|
El-Shoukrofy MS, Ismail A, Elhamammy RH, Abdelhady SA, Nassra R, Makkar MS, Agami MA, Wahid A, Nematalla HA, Sai M, Merk D, El-Yazbi AF, Belal ASF, Eid AH, Elzahhar PA. Novel thiazolones for the simultaneous modulation of PPARγ, COX-2 and 15-LOX to address metabolic disease-associated portal inflammation. Eur J Med Chem 2025; 289:117415. [PMID: 40022874 DOI: 10.1016/j.ejmech.2025.117415] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 03/04/2025]
Abstract
A hybrid pharmacophore model, based on structural motifs previously identified by our team, was employed to generate ligands that simultaneously target COX-2, 15-LOX, and PPARγ in the context of metabolic dysfunction-associated fatty liver disease (MAFLD). Notable COX-2 inhibitory activities (IC50 = 0.065-0.24 μM) were observed relative to celecoxib (IC50 = 0.049 μM). The two most effective 15-LOX inhibitors, 2a and 2b, exhibited 69 % and 57 % of quercetin's action, respectively. Utilizing the rat hemi-diaphragm model to assess in vitro glucose uptake capacity, compounds 2a and 2b demonstrated significant glucose uptake potential in the absence of insulin, surpassing that of pioglitazone. Compound 2a activated PPARγ with an EC50 value of 3.4 μM in a Gal4-hybrid reporter gene assay, indicating partial agonistic action. Interesting binding interactions with targets of interest were identified by molecular docking studies. As well, the expression levels of 20-HETE, Il-1β and TNF-α were decreased in LPS-challenged RAW264.7 macrophages upon treatment with compound 2a. The pharmacokinetic analysis of 2a and assessment of its in vivo efficacy in addressing hepatic impairment in rat models of diabetes and pre-diabetes were carried out. Together, these findings may offer preliminary insights into the potential of these compounds for further refinement in the existing therapeutic arsenals for metabolic diseases.
Collapse
Affiliation(s)
- Mai S El-Shoukrofy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Azza Ismail
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Reem H Elhamammy
- Department of Biochemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Sherien A Abdelhady
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia Street, Beside Green Plaza Complex 21648, Alexandria, Egypt
| | - Rasha Nassra
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, 21131, Egypt
| | - Monica S Makkar
- Faculty of Pharmacy and the Research & Innovation Hub, Alamein International University, Alamein, 5060335, Egypt
| | - Mahmoud A Agami
- Faculty of Pharmacy and the Research & Innovation Hub, Alamein International University, Alamein, 5060335, Egypt
| | - Ahmed Wahid
- Department of Biochemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Hisham A Nematalla
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour, 22516, Egypt
| | - Minh Sai
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377, Munich, Germany
| | - Daniel Merk
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377, Munich, Germany; Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, 60438, Frankfurt, Germany
| | - Ahmed F El-Yazbi
- Faculty of Pharmacy and the Research & Innovation Hub, Alamein International University, Alamein, 5060335, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Ahmed S F Belal
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, 2713, Qatar.
| | - Perihan A Elzahhar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.
| |
Collapse
|
21
|
Alhosani F, Alhamidi RS, Ilce BY, Altaie AM, Ali N, Hamad AM, Künstner A, Khandanpour C, Busch H, Al-Ramadi B, Harati R, Sayed K, AlFazari A, Bendardaf R, Hamoudi R. Transcriptome-Wide Analysis and Experimental Validation from FFPE Tissue Identifies Stage-Specific Gene Expression Profiles Differentiating Adenoma, Carcinoma In-Situ and Adenocarcinoma in Colorectal Cancer Progression. Int J Mol Sci 2025; 26:4194. [PMID: 40362431 PMCID: PMC12071244 DOI: 10.3390/ijms26094194] [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: 03/21/2025] [Revised: 04/20/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Colorectal cancer (CRC) progression occurs through three stages: adenoma (pre-cancerous lesion), carcinoma in situ (CIS) and adenocarcinoma, with tumor stage playing a pivotal role in the prognosis and treatment outcomes. Despite therapeutic advancements, the lack of stage-specific biomarkers hinders the development of accurate diagnostic tools and effective therapeutic strategies. This study aims to identify stage-specific gene expression profiles and key molecular mechanisms in CRC providing insights into molecular alterations across disease progression. Our methodological approach integrates the use of absolute gene set enrichment analysis (absGSEA) on formalin-fixed paraffin-embedded (FFPE)-derived transcriptomic data, combined with large-scale clinical validation and experimental confirmation. A comparative whole transcriptomic analysis (RNA-seq) was performed on FFPE samples including adenoma (n = 10), carcinoma in situ (CIS) (n = 8) and adenocarcinoma (n = 11) samples. Using absGSEA, we identified significant cellular pathways and putative molecular biomarkers associated with each stage of CRC progression. Key findings were then validated in a large independent CRC patient cohort (n = 1926), with survival analysis conducted from 1336 patients to assess the prognostic relevance of the candidate biomarkers. The key differentially expressed genes were experimentally validated using real-time PCR (RT-qPCR). Pathway analysis revealed that in CIS, apoptotic processes and Wnt signaling pathways were more prominent than in adenoma samples, while in adenocarcinoma, transcriptional co-regulatory mechanisms and protein kinase activity, which are critical for tumor growth and metastasis, were significantly enriched compared to adenoma. Additionally, extracellular matrix organization pathways were significantly enriched in adenocarcinoma compared to CIS. Distinct gene signatures were identified across CRC stages that differentiate between adenoma, CIS and adenocarcinoma. In adenoma, ARRB1, CTBP1 and CTBP2 were overexpressed, suggesting their involvement in early tumorigenesis, whereas in CIS, RPS3A and COL4A5 were overexpressed, suggesting their involvement in the transition from benign to malignant stage. In adenocarcinoma, COL1A2, CEBPZ, MED10 and PAWR were overexpressed, suggesting their involvement in advanced disease progression. Functional analysis confirmed that ARRB1 and CTBP1/2 were associated with early tumor development, while COL1A2 and CEBPZ were involved in extracellular matrix remodeling and transcriptional regulation, respectively. Experimental validation with RT-qPCR confirmed the differential expression of the candidate biomarkers (ARRB1, RPS3A, COL4A5, COL1A2 and MED10) across the three CRC stages reinforcing their potential as stage-specific biomarkers in CRC progression. These findings provide a foundation to distinguish between the CRC stages and for the development of accurate stage-specific diagnostic and prognostic biomarkers, which helps in the development of more effective therapeutic strategies for CRC.
Collapse
Affiliation(s)
- Faisal Alhosani
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany; (A.K.); (H.B.)
- Forensic Laboratory Department, Sharjah Police Headquarters, Sharjah P.O. Box 1965, United Arab Emirates
| | - Reem Sami Alhamidi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
| | - Burcu Yener Ilce
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
| | - Alaa Muayad Altaie
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
| | - Nival Ali
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
| | - Alaa Mohamed Hamad
- College of Health Sciences, Abu Dhabi University, Abu Dhabi P.O. Box 59911, United Arab Emirates;
| | - Axel Künstner
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany; (A.K.); (H.B.)
| | - Cyrus Khandanpour
- Department of Hematology and Oncology, University Cancer Center Schleswig-Holstein, University Hospital Schleswig-Holstein, University of Lübeck, 23562 Lübeck, Germany;
| | - Hauke Busch
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany; (A.K.); (H.B.)
| | - Basel Al-Ramadi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Rania Harati
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Kadria Sayed
- Department of Pathology and Laboratory Medicine, American Hospital Dubai, Dubai P.O. Box 3050, United Arab Emirates;
| | - Ali AlFazari
- Mediclinic Welcare Hospital, Dubai P.O. Box 31500, United Arab Emirates;
| | - Riyad Bendardaf
- Oncology Unit, University Hospital Sharjah, Sharjah P.O. Box 72772, United Arab Emirates;
| | - Rifat Hamoudi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Center of Excellence for Precision Medicine, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- BIMAI-Lab, Biomedically Informed Artificial Intelligence Laboratory, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London WC1E 6BT, UK
| |
Collapse
|
22
|
Zhong C, Yuan Y, Jiang Y, Qiao G, Deng Z, Liu Z, Yu L, Lin H, Mao J, Ma L, Zhang J. Predictive role of inflammatory markers for the efficacy of first-line immunotherapy plus chemotherapy in advanced gastric cancer. Discov Oncol 2025; 16:618. [PMID: 40285934 PMCID: PMC12033139 DOI: 10.1007/s12672-025-01857-0] [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: 12/02/2024] [Accepted: 01/28/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) plus chemotherapy have become a new first-line treatment option for patients with locally advanced or metastatic gastric cancer. However, it is still controversial whether to choose chemotherapy alone or ICIs plus chemotherapy as the first-line treatment option due to a lack of ideal predictive biomarkers for the efficacy. This study intended to explore the predictive value of inflammatory markers for the efficacy of first-line ICIs plus chemotherapy in this disease. METHODS This retrospective study included 131 patients with locally advanced or metastatic gastric cancer who received first-line treatment between July, 2020 and June, 2023. Among them, 76 received first-line ICIs plus chemotherapy and 55 received chemotherapy alone. Firstly, Kaplan-Meier and Cox regression analyses were used to explore the correlation between inflammatory markers and efficacy of first-line ICIs plus chemotherapy. Subsequently, the predictive value of combined baseline and dynamic changes in inflammatory markers was explored. Moreover, the predictive value of baseline inflammatory markers was further verified by comparing efficacy of ICIs plus chemotherapy with that of chemotherapy alone. RESULTS In patients receiving first-line ICIs plus chemotherapy, low baseline monocyte-to-lymphocyte ratio (MLR) in peripheral blood was significantly associated with better progression-free survival (PFS) and overall survival (OS), and was an independent prognostic factor for OS. In addition, dynamic early changes of MLR also played predictive role. Patients whose MLR was lower at baseline and after two cycles of treatment had better OS (P = 0.009). Furthermore, compared to chemotherapy alone, patients with a lower baseline MLR were more likely to benefit from first-line ICIs plus chemotherapy. CONCLUSION MLR could serve as a new biomarker to predict the efficacy of first-line ICIs plus chemotherapy in patients with locally advanced or metastatic gastric cancer. And it is helpful to select the candidates for first-line ICIs plus chemotherapy, which is worthy of further study.
Collapse
Affiliation(s)
- Chenming Zhong
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Yuan
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Jiang
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanglei Qiao
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhoufeng Deng
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zimei Liu
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liping Yu
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongjian Lin
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiuang Mao
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lijun Ma
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianjun Zhang
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
23
|
Miller C, Ealy A, Gregory A, Janarthanam C, Albers W, Richardson G, Jin H, Zenitsky G, Anantharam V, Kanthasamy A, Kanthasamy AG. Pathological α-synuclein dysregulates epitranscriptomic writer METTL3 to drive neuroinflammation in microglia. Cell Rep 2025; 44:115618. [PMID: 40279247 DOI: 10.1016/j.celrep.2025.115618] [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: 10/03/2024] [Revised: 02/11/2025] [Accepted: 04/04/2025] [Indexed: 04/27/2025] Open
Abstract
Recent reports suggest dysregulation of the N6-methyladenosine (m6A) RNA modification may contribute to the pathology of neurodegenerative diseases. Herein, we show the m6A methyltransferase complex including METTL3-the catalytic component of the nuclear-localized complex-is robustly upregulated in human microglia and astrocytes exposed to αSynf and Mn. Subcellular localization studies reveal METTL3 was predominantly cytoplasmic following Mn insult but remained nuclear following αSynf stimulation in activated microglia. Functional analysis revealed METTL3 and downstream m6A readers, including YTHDF2 and IGF2BP1-3, may regulate the proinflammatory secretome of activated microglia. Notably, methyltransferase activity and m6A abundance were significantly increased following Mn and αSynf treatment. METTL3 in Mn and αSynfin vivo models of neuroinflammation, along with human postmortem tissues from Alzheimer's disease (AD), Parkinson's disease (PD), and dementia with Lewy bodies (DLB) patients, was significantly upregulated. This was further confirmed by single-cell RNA sequencing (scRNA-seq) analysis. Overall, we demonstrate the m6A writer METTL3 may function as a major regulator of chronic neuroinflammation in synucleinopathies.
Collapse
Affiliation(s)
- Cameron Miller
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA; Department of Biochemistry and Molecular Biology, The University of Georgia, Athens, GA 30602, USA
| | - Alyssa Ealy
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA; Department of Physiology and Pharmacology, The University of Georgia, Athens, GA 30602, USA
| | - Amanda Gregory
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA
| | - Chelva Janarthanam
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA; Department of Physiology and Pharmacology, The University of Georgia, Athens, GA 30602, USA
| | - William Albers
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA; Department of Biology, The University of Georgia, Athens, GA 30602, USA
| | - Gabriel Richardson
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA
| | - Huajun Jin
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA; Department of Physiology and Pharmacology, The University of Georgia, Athens, GA 30602, USA
| | - Gary Zenitsky
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA
| | - Vellareddy Anantharam
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA; Department of Physiology and Pharmacology, The University of Georgia, Athens, GA 30602, USA
| | - Arthi Kanthasamy
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA; Department of Biochemistry and Molecular Biology, The University of Georgia, Athens, GA 30602, USA
| | - Anumantha G Kanthasamy
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA; Department of Biology, The University of Georgia, Athens, GA 30602, USA; Department of Biochemistry and Molecular Biology, The University of Georgia, Athens, GA 30602, USA; Department of Physiology and Pharmacology, The University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
24
|
Wu Z, Miao C, Zhang H. METTL3-mediated m6A modification in sepsis: current evidence and future perspectives. Epigenomics 2025:1-13. [PMID: 40251974 DOI: 10.1080/17501911.2025.2494983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 04/15/2025] [Indexed: 04/21/2025] Open
Abstract
Sepsis, a severe systemic inflammatory condition triggered by infection, is associated with high morbidity and mortality worldwide. While medical diagnosis and treatment have advanced in recent years, a specific therapy remains unavailable. Recently, significant progress has been made in studying the epigenetic RNA modification N6-methyladenosine (m6A) and its core methyltransferase METTL3. The role of m6A in sepsis has also been increasingly elucidated. This review aims to explore the pathological mechanisms of sepsis and its relationship with m6A, focusing on the role of the key m6A writer, METTL3, in sepsis.
Collapse
Affiliation(s)
- Zijun Wu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Wu B, Li D, Wang Y, Pan T, Xu J, Li L. The m6A methyltransferase METTL3 affects ferroptosis in non-small cell lung cancer by regulating the PTEN/PI3K/AKT pathway. Discov Oncol 2025; 16:559. [PMID: 40249573 PMCID: PMC12008084 DOI: 10.1007/s12672-025-02330-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) poses a major threat to human health, METTL3 has been reported to promote numerous tumor development by inhibiting ferroptosis. The aim of the present study was to explore the mechanism of action of METTL3 in NSCLC. METHODS The UALCAN online platform was applied to analyze METTL3 and PTEN expression in NSCLC and their relationship with tumor stages. NCI-H23 and NCI-H1975 cells were transfected with sh-METTL3, or oe-METTL3 respectively. Then EdU assay was employed to assess cell proliferation and the transwell assay was employed to assess the ability of cells to migrate and invade. Apoptosis was detected using flow cytometry. In addition, m6A methylation levels, oxidative stress indicators, and Fe2+ content were determined. Furthermore, GPX4 and PTEN expression, as well as PI3K and AKT phosphorylation were quantified. Finally, the cells with METTL3 knockdown were further transfected with sh-PTEN. RESULTS METTL3 expression was up-regulated in NSCLC and was closely related to the tumor stages. METTL3 overexpression significantly promoted the malignant phenotype of NSCLC cells, increased the methylation level of m6A mRNA, reduced oxidative stress, inhibited the occurrence of ferroptosis and apoptosis, and led to increased expression of GPX4 and activation of the PTEN/PI3K/AKT pathway. Conversely, METTL3 knockdown produced the opposite effect. Importantly, METTL3 knockdown-induced oxidative stress and ferroptosis in NCI-H23 cells were rescued by sh-PTEN or ferroptosis inhibitor Ferrostatin-1. CONCLUSION METTL3 may inhibit ferroptosis in NSCLC by activating the PTEN/PI3K/AKT pathway, suggesting that METTL3-mediated PTEN/PI3K/AKT pathway may be a promising therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Bin Wu
- Department of Respiratoty, Affiliated Xiaoshan Hospital, Hangzhou Normal Uiniversity, No. 728, Yucai North Road, Xiaoshan District, Hangzhou, 311200, Zhejiang, China.
| | - Danhong Li
- Department of Respiratoty, Affiliated Xiaoshan Hospital, Hangzhou Normal Uiniversity, No. 728, Yucai North Road, Xiaoshan District, Hangzhou, 311200, Zhejiang, China.
| | - Yu Wang
- Department of Respiratoty, Affiliated Xiaoshan Hospital, Hangzhou Normal Uiniversity, No. 728, Yucai North Road, Xiaoshan District, Hangzhou, 311200, Zhejiang, China
| | - Tingting Pan
- Department of Respiratoty, Affiliated Xiaoshan Hospital, Hangzhou Normal Uiniversity, No. 728, Yucai North Road, Xiaoshan District, Hangzhou, 311200, Zhejiang, China
| | - Jieru Xu
- Department of Respiratoty, Affiliated Xiaoshan Hospital, Hangzhou Normal Uiniversity, No. 728, Yucai North Road, Xiaoshan District, Hangzhou, 311200, Zhejiang, China
| | - Lihong Li
- Department of Respiratoty, Affiliated Xiaoshan Hospital, Hangzhou Normal Uiniversity, No. 728, Yucai North Road, Xiaoshan District, Hangzhou, 311200, Zhejiang, China
| |
Collapse
|
26
|
Wang JX, Liu XZ, Guo Z, Zhang HL, Qi L, Liu J, Liu P, Xie GX, Wang XN. Differences in Fatty Acid Metabolism between MCDD and HFD Induced Metabolic Dysfunction-associated Fatty Liver Disease Model Mice. Biol Proced Online 2025; 27:14. [PMID: 40229695 PMCID: PMC11998272 DOI: 10.1186/s12575-025-00276-3] [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: 02/04/2025] [Accepted: 03/28/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND The global incidence of metabolic dysfunction-associated fatty liver disease (MAFLD) is increasing annually, which has become a major public-health concern. MAFLD is typically associated with obesity, hyperlipemia, or metabolic syndrome. Dietary induction is one of the most common methods for preparing animal models of MAFLD. However, there are phenotypic differences between methionine-choline-deficient diet (MCDD) and high fat diet (HFD) models. METHODS To explore the differences in hepatic fatty acid metabolism between MCDD and HFD induced MAFLD, we analyzed serum and liver tissue from the two MAFLD models. RESULTS We found that liver fat accumulation and liver function damage were common pathological features in both MAFLD models. Furthermore, in the MCDD model, the expression of hepatic fatty acid transport proteins increased, while the expression of hepatic fatty acid efflux proteins and mRNA decreased, along with a decrease in blood lipid levels. In the HFD model, the expression of hepatic fatty acid uptake proteins, efflux proteins and efflux mRNA increased, along with an increase in blood lipid levels. CONCLUSION Impaired fatty acid oxidation and increased hepatic fatty acid uptake play key roles in the pathogenesis of the two MAFLD models. The inverse changes in de novo lipogenesis and fatty acid efflux may represent an important pathological mechanism that leads to the phenotypic differences between the MCDD and HFD models.
Collapse
Affiliation(s)
- Jia-Xuan Wang
- Institute of Interdisciplinary Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Hepatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Xin-Zhu Liu
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Zhen Guo
- Institute of Interdisciplinary Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Sichuan, 646000, China
| | - Hui-Lin Zhang
- Institute of Interdisciplinary Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li Qi
- Institute of Interdisciplinary Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jia Liu
- Institute of Interdisciplinary Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ping Liu
- Institute of Interdisciplinary Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Guo-Xiang Xie
- Human Metabolomics Institute, Inc., Shenzhen, 518109, Guangdong, China.
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China.
| | - Xiao-Ning Wang
- Institute of Interdisciplinary Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
27
|
Chen J, Li W, Yu L, Zhang B, Li Z, Zou P, Ding B, Dai X, Wang Q. Combined Effects of Ketogenic Diet and Aerobic Exercise on Skeletal Muscle Fiber Remodeling and Metabolic Adaptation in Simulated Microgravity Mice. Metabolites 2025; 15:270. [PMID: 40278399 PMCID: PMC12029359 DOI: 10.3390/metabo15040270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/11/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
Objective: Prolonged microgravity environments impair skeletal muscle homeostasis by triggering fiber-type transitions and metabolic dysregulation. Although exercise and nutritional interventions may alleviate disuse atrophy, their synergistic effects under microgravity conditions remain poorly characterized. This study investigated the effects of an 8-week ketogenic diet combined with aerobic exercise in hindlimb-unloaded mice on muscle fiber remodeling and metabolic adaptation. Methods: Seven-week-old male C57BL/6J mice were randomly divided into six groups: normal diet control (NC), normal diet with hindlimb unloading (NH), normal diet with hindlimb unloading and exercise (NHE), ketogenic diet control (KC), ketogenic diet with hindlimb unloading (KH), and ketogenic diet with hindlimb unloading and exercise (KHE). During the last two weeks of intervention, hindlimb unloading was applied to simulate microgravity. Aerobic exercise groups performed moderate-intensity treadmill running (12 m/min, 60 min/day, and 6 days/week) for 8 weeks. Body weight, blood ketone, and glucose levels were measured weekly. Post-intervention assessments included the respiratory exchange ratio (RER), exhaustive exercise performance tests, and biochemical analyses of blood metabolic parameters. The skeletal muscle fiber-type composition was evaluated via immunofluorescence staining, lipid deposition was assessed using Oil Red O staining, glycogen content was analyzed by Periodic Acid-Schiff (PAS) staining, and gene expression was quantified using quantitative real-time PCR (RT-qPCR). Results: Hindlimb unloading significantly decreased body weight, induced muscle atrophy, and reduced exercise endurance in mice. However, the combination of KD and aerobic exercise significantly attenuated these adverse effects, as evidenced by increased proportions of oxidative muscle fibers (MyHC-I) and decreased proportions of glycolytic fibers (MyHC-IIb). Additionally, this combined intervention upregulated the expression of lipid metabolism-associated genes, including CPT-1b, HADH, PGC-1α, and FGF21, enhancing lipid metabolism and ketone utilization. These metabolic adaptations corresponded with improved exercise performance, demonstrated by the increased time to exhaustion in the KHE group compared to other hindlimb unloading groups. Conclusions: The combination of a ketogenic diet and aerobic exercise effectively ameliorates simulated microgravity-induced skeletal muscle atrophy and endurance impairment, primarily by promoting a fiber-type transition from MyHC-IIb to MyHC-I and enhancing lipid metabolism gene expression (CPT-1b, HADH, and PGC-1α). These findings underscore the potential therapeutic value of combined dietary and exercise interventions for mitigating muscle atrophy under simulated microgravity conditions.
Collapse
Affiliation(s)
- Jun Chen
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China;
- Sports Nutrition Center, National Institute of Sports Medicine, Beijing 100029, China
| | - Wenjiong Li
- National Key Laboratory of Space Medicine, Beijing 100094, China; (W.L.); (Z.L.); (P.Z.); (B.D.)
| | - Liang Yu
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China; (L.Y.); (B.Z.)
| | - Bowei Zhang
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China; (L.Y.); (B.Z.)
| | - Zhili Li
- National Key Laboratory of Space Medicine, Beijing 100094, China; (W.L.); (Z.L.); (P.Z.); (B.D.)
| | - Peng Zou
- National Key Laboratory of Space Medicine, Beijing 100094, China; (W.L.); (Z.L.); (P.Z.); (B.D.)
| | - Bai Ding
- National Key Laboratory of Space Medicine, Beijing 100094, China; (W.L.); (Z.L.); (P.Z.); (B.D.)
| | - Xiaoqian Dai
- National Key Laboratory of Space Medicine, Beijing 100094, China; (W.L.); (Z.L.); (P.Z.); (B.D.)
| | - Qirong Wang
- Sports Nutrition Center, National Institute of Sports Medicine, Beijing 100029, China
| |
Collapse
|
28
|
Sabouri M, Etemadifar M, Dehghani Firoozabadi F, Sindarreh S, Akhavan-Sigari A. Primary central nervous system tumors in patients with multiple sclerosis. BMC Neurol 2025; 25:147. [PMID: 40205341 PMCID: PMC11983722 DOI: 10.1186/s12883-025-04095-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 02/19/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic neuroinflammatory disorder that can present with clinical and radiological features indistinguishable from a central nervous system (CNS) tumor. Previous studies suggest that whilepatients with MS have a reduced overall risk of cancer, they may have an increased risk of developing CNS malignancies. METHODS In this cross-sectional observational study, we investigated the prevalence of CNS tumors in patients with MS using data from the Isfahan MS clinic registry between 2020 and 2023 who had been diagnosed with primary CNS tumors following their diagnosis of MS. RESULTS Among the 2,280 registered patients, 36 individuals were diagnosed with CNS tumors, yielding a prevalence of 1.58%. The distribution of primary CNS tumors among these patients was as follows: 41.7% had pituitary adenomas, 30.6% had meningiomas, 13.9% had primary CNS lymphoma, 5.6% had acoustic neuroma, and the remaining cases included epidermoid cysts (2.8%), neurofibromas (2.8%), and glioblastoma multiforme (2.8%). The mean age at tumor diagnosis was approximately 45 years, while the mean age at MS diagnosis among those who subsequently developed a CNS tumor was 31.5 years. CONCLUSION The overall prevalence of primary CNS tumors in our MS population was 1.58%. Meningiomas and pituitary adenomas were the most common types of CNS tumors observed in these patients. Given potential symptom overlap, new or unusual symptoms not typical of MS should be closely monitored or assessed for possible CNS malignancies.
Collapse
Affiliation(s)
- Masih Sabouri
- Department of Neurosurgery, Alzahra University Hospital, Isfahan, Iran
| | - Masoud Etemadifar
- Department of Neurosurgery, Alzahra University Hospital, Isfahan, Iran
| | | | - Setayesh Sindarreh
- Cancer Prevention Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amirhossein Akhavan-Sigari
- Department of Neurosurgery, Alzahra University Hospital, Isfahan, Iran.
- Cancer Prevention Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
29
|
Fan B, Chen G, Huang S, Li Y, Nabil ZUH, Yang Z. Summary of the mechanism of ferroptosis regulated by m6A modification in cancer progression. Front Cell Dev Biol 2025; 13:1507171. [PMID: 40271153 PMCID: PMC12014555 DOI: 10.3389/fcell.2025.1507171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/25/2025] [Indexed: 04/25/2025] Open
Abstract
The most common form of internal RNA modification in eukaryotes is called n6-methyladenosine (m6A) methylation. It has become more and more well-known as a research issue in recent years since it alters RNA metabolism and is involved in numerous biological processes. Currently, m6A alteration offers new opportunities in clinical applications and is intimately linked to carcinogenesis. Ferroptosis-a form of iron-dependent, lipid peroxidation-induced regulated cell death-was discovered. In the development of cancer, it has become an important factor. According to newly available data, ferroptosis regulates tumor growth, and cancer exhibits aberrant m6A levels in crucial ferroptosis regulatory components. On the other hand, m6A has multiple roles in the development of tumors, and the relationship between m6A-modified ferroptosis and malignancies is quite intricate. In this review, we first give a thorough review of the regulatory and functional roles of m6A methylation, focusing on the molecular processes of m6A through the regulation of ferroptosis in human cancer progression and metastasis, which are strongly associated to cancer initiation, progression, and drug resistance. Therefore, it is crucial to clarify the relationship between m6A-mediated regulation of ferroptosis in cancer progression, providing a new strategy for cancer treatment with substantial clinical implications.
Collapse
Affiliation(s)
| | | | | | | | | | - Zuozhang Yang
- Bone and Soft Tissue Tumors Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, Yunnan, China
| |
Collapse
|
30
|
Sun Y, Qu Y, Yang Z, Lv B, Wang G, Fan K, Wang Y, Pan J, Du Z, Yu Y. Suppressed intestinal secondary bile acids in moxifloxacin-induced hyperglycemia: studies in normal and diabetic GK rats. Front Pharmacol 2025; 16:1569856. [PMID: 40255568 PMCID: PMC12006139 DOI: 10.3389/fphar.2025.1569856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 03/19/2025] [Indexed: 04/22/2025] Open
Abstract
Objective Moxifloxacin (MFLX) frequently induces dysglycemia when used in the treatment of infectious diseases, particularly in patients with diabetes. However, the mechanism through which MFLX affects host glucose metabolism remains unclear. This study aimed to investigate the possible mechanism underlying MFLX-induced hyperglycemia. Methods In this study, we investigated the short-term (3 days) and long-term (14 days) effects of MFLX on glucose metabolism in normal and type 2 diabetic GK rats. After oral administration of 40 mg/kg of MFLX, blood glucose, insulin, GLP-1, and fibroblast growth factor 15 (FGF15) levels in the blood of rats, as well as bile acids in both blood and feces, and gut microbiota, were examined. Liver and ileum tissues were promptly harvested for detecting the expression of hepatic 7α-hydroxylase (CYP7A1) and intestinal Takeda G-protein-coupled receptor 5 (TGR5) and farnesoid X receptor (FXR). In addition, we explored the effect of secondary bile acids (SBAs) on GLP-1 secretion in NCI-H716 cells, and observed the direct effect of MFLX on the expression of CYP7A1 in HepG2 cells and TGR5, FXR in NCI-H716 cells. Results It was demonstrated that MFLX induced hyperglycemia in diabetic rats, with a more pronounced reduction in serum insulin, GLP-1, and FGF15 levels than observed in normal rats. Gut microbiota associated with SBAs metabolism were significantly reduced, leading to decreased intestinal deoxycholic acid (DCA) and lithocholic acid (LCA). In vitro studies revealed that DCA and LCA (25 μM, 50 μM, and 100 μM) promoted GLP-1 secretion in a concentration-dependent manner in NCI-H716 cells. Meanwhile, we observed that the expression of intestinal TGR5 and FXR significantly downregulated, whereas CYP7A1 expression in liver was increased in GK rats after MFLX treatment. MFLX itself (0.1 μM, 1 μM, and 10 μM) did not directly altered TGR5 or FXR expressions in NCI-H716 cells, nor did it alter CYP7A1 expression in HepG2 cells, which indicated that the impact of MFLX on glucose metabolism was primarily induced by changes in bile acids metabolism resulting from alterations in the gut microbiota. Conclusion Our studies showed MFLX more likely to cause hyperglycemia when used in diabetic states and highlighted the critical role of gut microbiota-SBAs-TGR5/FXR pathway in MFLX-induced hyperglycemia.
Collapse
Affiliation(s)
- Yewen Sun
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, China
- College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Yuchen Qu
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Zhuan Yang
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, China
- College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Bo Lv
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Guanjun Wang
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, China
- College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Kai Fan
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuyuan Wang
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jie Pan
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ziyan Du
- Department of Respiration Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yunli Yu
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
31
|
Abdelmoneim D, Eldomany EB, El-Adl M, Farghali A, El-Sayed G, El-Sherbini ES. Possible protective effect of natural flavanone naringenin-reduced graphene oxide nanosheets on nonalcoholic fatty liver disease. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4071-4086. [PMID: 39414698 PMCID: PMC11978702 DOI: 10.1007/s00210-024-03495-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/25/2024] [Indexed: 10/18/2024]
Abstract
Utilizing naringenin as a safe, natural compound for reducing graphene oxide and to determine whether Nar-RGO more effectively mitigates the harmful effects of HFFD-induced NAFLD compared to crude naringenin. Using a straightforward experimental setup, we utilize the bioactive flavonoid naringenin (NAR) as the reducing agent to synthesize naringenin-reduced graphene oxide nanosheets (Nar-RGO). Naringenin loading on graphene oxide was validated using electroscopic methods (SEM and TEM) and zeta potential measurements. Utilization of reduced graphene oxide for naringenin encapsulation resulted in a significant improvement in hepatic steatosis, insulin resistance, oxidative stress, and signs of inflammation in HFFD-induced NAFLD compared to crude naringenin. This study demonstrates that Nar-RGO exhibits significantly greater efficacy compared to free naringenin. Therefore, it can be used as a promising medicine in counteracting high-fat-fructose diet (HFFD)-induced NAFLD.
Collapse
Affiliation(s)
- Doaa Abdelmoneim
- Biochemistry and Chemistry of Nutrition Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt.
| | - Ehab B Eldomany
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Mohamed El-Adl
- Biochemistry and Chemistry of Nutrition Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Ahmed Farghali
- Material Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Gehad El-Sayed
- Biochemistry and Chemistry of Nutrition Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - El Said El-Sherbini
- Biochemistry and Chemistry of Nutrition Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
32
|
Luo X, He C, Yang B, Yin S, Li K. WTAP Promotes Atherosclerosis by Inducing Macrophage Pyroptosis and M1 Polarization through Upregulating NLRP3. Appl Biochem Biotechnol 2025; 197:2397-2416. [PMID: 39747738 DOI: 10.1007/s12010-024-05106-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2024] [Indexed: 01/04/2025]
Abstract
The study was designed to investigate the impact of N6-methyladenosine (m6A) writer Wilms tumor 1-associated protein (WTAP) on the progression of atherosclerosis (AS) and to further elucidate its possible regulatory mechanism. The m6A levels and WTAP expressions were initially assessed through RIP, qRT-PCR, and western blotting. An in vitro model of AS was constructed by ox-LDL treatment in RAW264.7 cells. Next, the impact of WTAP on macrophage pyroptosis and M1 polarization was evaluated. The relationship between WTAP and NLRP3 was then investigated using m6A modification quantification and RIP-qPCR assay. To investigate the effect of WTAP on AS development in vivo, we created an ApoE-/-mouse model of AS by feeding high-fat diet (HFD). Furthermore, the influence of WTAP on macrophage pyroptosis and M1 polarization through NLRP3 was explored by NLRP3 overexpression AAV injection. Here, we found that WTAP was significantly upregulated in peripheral blood mononuclear cells (PBMCs) from AS patients, accompanied by increased total m6A methylation levels. The silencing of WTAP suppressed macrophage pyroptosis and M1 polarization induced by ox-LDL and also ameliorated aortic root lesion damage in AS mice. Mechanistically, m6A modification mediated by WTAP enhanced NLRP3 mRNA stabilization, thereby upregulating NLRP3 expression. Overexpression of NLRP3 was found to enhance macrophage pyroptosis and M1 polarization, contributing to the progression of AS. In conclusion, our findings suggest that WTAP knockdown mitigated AS progression by modulating NLRP3 in an m6A-dependent manner. Our study proposes that targeting WTAP could be a potential preventive and therapeutic strategy for AS patients.
Collapse
Affiliation(s)
- Xing Luo
- Department of Neurology, The Third Hospital of Changsha, Changsha, Hunan, China
| | - Chaogui He
- Department of Vascular Surgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University (The First Hospital of Changsha), Changsha, Hunan, China
| | - Bo Yang
- Department of Vascular Surgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University (The First Hospital of Changsha), Changsha, Hunan, China
| | - Shuheng Yin
- University of South China, Hengyang, Hunan, China
| | - Ke Li
- University of South China, Hengyang, Hunan, China.
| |
Collapse
|
33
|
Wang J, Wang X, Jiang M, Lang T, Wan L, Dai J. 5-aminosalicylic acid alleviates colitis and protects intestinal barrier function by modulating gut microbiota in mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3681-3695. [PMID: 39352537 DOI: 10.1007/s00210-024-03485-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/23/2024] [Indexed: 04/10/2025]
Abstract
5-aminosalicylic acid (5-ASA) is widely used in the treatment of ulcerative colitis (UC), but its anti-inflammatory mechanism is complex and has not been fully understood. DSS model was used to test the effect of 5-ASA. Tight junction and Ki-67 were detected by western blot, immunofluorescence, and immunohistochemistry or qPCR. 16S rRNA gene sequencing of gut microbiota and subsequent bioinformatics and statistical analysis were performed to identify the specific bacteria which were associated with the treatment effect of 5-ASA. GC-MS was performed to test short-chain fatty acids (SCFAs). Antibiotic-treated mice were used to demonstrate the key role of endogenous gut microbiota. Here, we found that 5-ASA alleviated dextran sulfate sodium (DSS)-induced colitis in mice. Moreover, 5-ASA significantly repaired the intestinal barrier. At the molecular level, 5-ASA markedly raised the expression of tight junction proteins including JAM-A and occludin and cell proliferation marker Ki-67 in mice. In addition, bacterial 16S rRNA gene sequencing and bioinformatics analysis showed that 5-ASA significantly modulated the DSS-induced gut bacterial dysbiosis. In detail, it stimulated the growth of protective bacteria belonging to Faecalibaculum and Dubosiella, which were negatively correlated with colitis parameters, and blocked the expansion of pro-inflammatory bacteria such as Escherichia-Shigella and Oscillibacter, which were positively correlated with colitis in mice. Meanwhile, 5-ASA increased the cecal acetate level. Most notably, 5-ASA was no longer able to treat colitis and reverse gut barrier dysfunction in antibiotic-treated mice that lacked endogenous gut microbiota. Our data suggested that the anti-inflammatory activity of 5-ASA required the inherent intestinal flora, and the gut microbiota was a potential and effective target for the treatment of ulcerative colitis.
Collapse
Affiliation(s)
- Jingjing Wang
- Shanghai Key Laboratory of Pancreatic Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxin Wang
- Shanghai Key Laboratory of Pancreatic Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingjie Jiang
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, P.R. China
| | - Tao Lang
- Shanghai Key Laboratory of Pancreatic Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Leilei Wan
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Juanjuan Dai
- Shanghai Key Laboratory of Pancreatic Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Intensive Care Unit, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, P.R. China.
| |
Collapse
|
34
|
Wang X, Ling W, Zhu Y, Ji C, An X, Qi Y, Li S, Zhang C, Tong R, Jiang D, Kang B. Spermidine alleviates copper-induced oxidative stress, inflammation and cuproptosis in the liver. FASEB J 2025; 39:e70453. [PMID: 40079199 DOI: 10.1096/fj.202403002r] [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: 11/27/2024] [Revised: 01/13/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025]
Abstract
Copper exposure poses potential detrimental effects on both public and ecosystem health. Spermidine, an antioxidant, has shown promise in reducing oxidative stress and inflammation within the liver. However, its specific role in mitigating copper-induced hepatic cuproptosis and disturbances in copper metabolism remains unexplored. Consequently, this research aims to investigate to examine the impact of spermidine on hepatic cuproptosis and the related disturbances in copper metabolism. In the study, we established a model of copper-induced liver toxicity by feeding C57BL/6 mice a high-copper diet for three months. Histopathological and biochemical analyses revealed that copper exposure induced hepatic inflammatory cell infiltration, hepatocyte degeneration, elevated levels of MDA, ROS, and Cu2+ accumulation in the liver, and increased ALT and AST activities in serum (p < .05). Regarding inflammation, copper exposure significantly increased serum levels of IL-1β, IL-6, and TNF-α (p < .05), upregulated TNF-α and IFN-γ expression, and downregulated IL-10 expression in the liver (p < .05). Meanwhile, copper exposure inhibited the expression of copper metabolism and Fe-S cluster-related proteins (p < .05). Exogenous spermidine administration effectively reduced ROS, MDA, and Cu2+ accumulation in the liver, while also decreasing ALT and AST activites, IL-1β, IL-6, and TNF-α levels in the serum (p < .05), and downregulated TNF-α and IFN-γ expression (p < .001). Additionally, spermidine combined with CuSO4 treatment significantly promotes the expression of copper metabolism and Fe-S cluster-related proteins, compared to the CuSO4 group (p < .05). In summary, spermidine reduces Cu2+ accumulation in the liver, alleviates hepatic cuproptosis, oxidative damage, and inflammation, and exerts a protective effect on the liver.
Collapse
Affiliation(s)
- Xin Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - Weikang Ling
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - Yang Zhu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - Chengweng Ji
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - Xiaoguang An
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - Yuxin Qi
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - Shuo Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - Chengye Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - Ruixue Tong
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - Dongmei Jiang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - Bo Kang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| |
Collapse
|
35
|
Mindru DE, Bozomitu LI, Păduraru DTA, Țarcă E, Curici A, Elkan EM, Butnariu LI, Moraru DC, Diaconescu C, Luca AC. Biomarkers and Mechanisms in the Early Assessment of Childhood Obesity from a Multidisciplinary Perspective-A Narrative Review. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:607. [PMID: 40282897 PMCID: PMC12028513 DOI: 10.3390/medicina61040607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025]
Abstract
Obesity has been the subject of research focused on preventive policies among the young population due to epidemiological studies which have shown devastating figures in recent years in terms of the incidence and prevalence of this condition. A number of previously known biomarkers have proven useful in the early diagnosis of complications associated with obesity, while others remain in the study stage. The intestinal microbiota are also relevant in the secondary prevention of obesity complications, another area that has turned into a hot topic of current research. The primary goal of this review is to highlight markers and mechanisms that can enhance specialists' understanding of obesity assessment and its systemic complications. Salivary markers have been proven useful in the evaluation of obesity, with the advantage of being low-cost and easy to sample. Another interesting topic is the role of the renin-angiotensin and the kallikrein-kinin systems in obesity-related systemic complications. One well-known fact is the connection between obesity and high blood pressure, which is closely related to these systems. This paper also explores the link between gut microbiota and adiposity, particularly the potential of the Firmicutes/Bacteroidetes ratio as a useful biomarker.
Collapse
Affiliation(s)
- Dana Elena Mindru
- Department of Mother and Child Medicine, Faculty of Medicine, University of Medicine and Pharmacy “Gr.T.Popa”, 700115 Iasi, Romania; (D.E.M.); (L.I.B.); (D.T.A.P.); (A.C.L.)
- Emergency Clinical Hospital for Children “Sfanta Maria” Iasi, 700309 Iasi, Romania
| | - Laura Iulia Bozomitu
- Department of Mother and Child Medicine, Faculty of Medicine, University of Medicine and Pharmacy “Gr.T.Popa”, 700115 Iasi, Romania; (D.E.M.); (L.I.B.); (D.T.A.P.); (A.C.L.)
- Emergency Clinical Hospital for Children “Sfanta Maria” Iasi, 700309 Iasi, Romania
| | - Dana Teodora Anton Păduraru
- Department of Mother and Child Medicine, Faculty of Medicine, University of Medicine and Pharmacy “Gr.T.Popa”, 700115 Iasi, Romania; (D.E.M.); (L.I.B.); (D.T.A.P.); (A.C.L.)
- Emergency Clinical Hospital for Children “Sfanta Maria” Iasi, 700309 Iasi, Romania
| | - Elena Țarcă
- Department of Surgery II—Pediatric and Orthopedic Surgery, Faculty of Medicine, University of Medicine and Pharmacy “Gr.T.Popa”, 700115 Iasi, Romania
| | - Antoanela Curici
- Department of Cellular and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Eva Maria Elkan
- Department of Morfofunctional Sciences, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University of Galați, 47 Domnească Street, 800008 Galați, Romania;
| | - Lăcrămioara Ionela Butnariu
- Department of Medical Genetics, Faculty of Medicine, University of Medicine and Pharmacy “Gr.T.Popa”, 700115 Iasi, Romania;
| | - Dan Cristian Moraru
- Department of Plastic Surgery and Reconstructive Microsurgery, “Sf. Spiridon” Emergency County Hospital, 700111 Iasi, Romania;
| | - Cosmin Diaconescu
- Faculty of Medicine, Doctoral School of Medical Sciences, University of Medicine and Pharmacy “Gr.T.Popa”, 700115 Iasi, Romania;
| | - Alina Costina Luca
- Department of Mother and Child Medicine, Faculty of Medicine, University of Medicine and Pharmacy “Gr.T.Popa”, 700115 Iasi, Romania; (D.E.M.); (L.I.B.); (D.T.A.P.); (A.C.L.)
- Emergency Clinical Hospital for Children “Sfanta Maria” Iasi, 700309 Iasi, Romania
| |
Collapse
|
36
|
de Oliveira THC, Gonçalves GKN. Liver ischemia reperfusion injury: Mechanisms, cellular pathways, and therapeutic approaches. Int Immunopharmacol 2025; 150:114299. [PMID: 39961215 DOI: 10.1016/j.intimp.2025.114299] [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: 12/16/2024] [Revised: 01/28/2025] [Accepted: 02/11/2025] [Indexed: 03/03/2025]
Abstract
Liver ischemia-reperfusion injury (LIRI) is a critical challenge in liver transplantation, resection, and trauma surgeries, leading to significant hepatic damage due to oxidative stress, inflammation, and mitochondrial dysfunction. This review explores the cellular and molecular mechanisms underlying LIRI, focusing on ATP depletion, mitochondrial dysfunction, and the involvement of reactive oxygen species (ROS). Inflammatory pathways, including the activation of nuclear factor-kappa B (NF-κB) and the NLRP3 inflammasome, as well as pro-inflammatory cytokines such as TNF-α and IL-1β, play a crucial role in exacerbating tissue damage. Various types of cell death, including necrosis, apoptosis, necroptosis, pyroptosis, ferroptosis and cuproptosis are also discussed. Therapeutic interventions targeting these mechanisms, such as antioxidants, anti-inflammatories, mitochondrial protectors, and signaling modulators, have shown promise in pre-clinical studies. However, translating these findings into clinical practice faces challenges due to the limitations of animal models and the complexity of human responses. Emerging therapies, such as RNA-based treatments, genetic editing, and stem cell therapies, offer potential breakthroughs in LIRI management. This review highlights the need for further research and the development of innovative therapeutic approaches to improve clinical outcomes.
Collapse
|
37
|
Chae HS, Ahmed NH, Dale OR, Avula B, Khan IA, Khan SI. Damiana ( Turnera diffusa) Reduces Adipocyte Cell Differentiation and Ameliorates Myocyte Glucose Uptake. J Diet Suppl 2025; 22:401-416. [PMID: 40125913 DOI: 10.1080/19390211.2025.2480582] [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] [Indexed: 03/25/2025]
Abstract
Turnera diffusa Willd. ex Schult. leaf has been used in traditional medicine as an aphrodisiac, tonic, and in the management of diabetes. Based on the traditional use and recent evidence of antidiabetic activity, we investigated the effects of a 95% ethanolic extract of T. diffusa leaf (TDE) on a series of ligand-activated transcription factors, namely PPARα, PPARγ, LXR and NRF2, which are involved in the regulation of metabolic pathways associated with obesity, diabetes and inflammation. Further, the effects of TDE on α-glucosidase enzyme, lipid accumulation in adipocytes (adipogenesis) and glucose uptake in myocytes were also evaluated. Phytochemical analysis of TDE was performed by mass spectrometry. TDE (50 μg/mL) demonstrated a strong agonistic effect on LXR (2.7-fold) and NRF2 (21.6-fold), while the activation of PPARα, and PPARγ was in the range of 1.4-1.8-folds under similar experimental conditions. At a concentration of 100 μg/mL TDE decreased lipid accumulation in adipocytes by 55.3% and increased glucose uptake in muscle cells by 91.3%. The adipogenic effect induced by a full PPARγ agonist (rosiglitazone) was antagonized by TDE showing a decrease of 57.6% in lipid accumulation. This is the first report to reveal the agonistic action of TDE on multiple nuclear receptors along with its glucose uptake enhancing and antiadipogenic effects. The results indicate the potential utility of TDE in alleviating the symptoms of metabolic syndrome and in preventing the undesired adipogenic effects of antidiabetic drugs of the glitazone class. Phytochemical analysis of TDE indicated the presence of flavonoids as major constituents. Further studies in animal models of type II diabetes and obesity are warranted to explore its utility as an anti-diabetic and anti-obesity supplement.
Collapse
Affiliation(s)
- Hee-Sung Chae
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, University, MS, USA
| | - Nessma H Ahmed
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, University, MS, USA
| | - Olivia R Dale
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, University, MS, USA
| | - Bharathi Avula
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, University, MS, USA
| | - Ikhlas A Khan
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, University, MS, USA
- Department of Biomolecular Sciences, School of Pharmacy, The University of Mississippi, University, MS, USA
| | - Shabana I Khan
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, University, MS, USA
- Department of Biomolecular Sciences, School of Pharmacy, The University of Mississippi, University, MS, USA
| |
Collapse
|
38
|
He Q, Li X, Li H, Tan A, Chi Y, Fang D, Li X, Liu Z, Shang Q, Zhu Y, Cielecka-Piontek J, Chen J. TGR5 Activation by Dietary Bioactives and Related Improvement in Mitochondrial Function for Alleviating Diabetes and Associated Complications. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6293-6314. [PMID: 40045496 DOI: 10.1021/acs.jafc.4c10395] [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: 03/20/2025]
Abstract
Takeda G protein-coupled receptor 5 (TGR5), also known as G protein-coupled bile acid receptor 1 (GPBAR1), is a cell surface receptor involved in key physiological processes, including glucose homeostasis and energy metabolism. Recent research has focused on the role of TGR5 activation in preventing or treating diabetes while also highlighting its potential impact on the progression of diabetic complications. Functional foods and edible plants have emerged as valuable sources of natural compounds that can activate TGR5, offering potential therapeutic benefits for diabetes management. Despite growing interest, studies on the activation of TGR5 by dietary bioactive compounds remain scattered. This Review aims to provide a comprehensive analysis of how dietary bioactives act as potential agents for TGR5 activation in managing diabetes and its complications. It explores the mechanisms of TGR5 activation through both direct agonistic effects and indirect pathways via modulation of the gut microbiota and bile acid metabolism.
Collapse
Affiliation(s)
- Quanrun He
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Shenzhen-Hong Kong International Science and Technology Park, No. 3 Binglang Road, Futian Free Trade Zone, Futian District, Shenzhen, Guangdong 518045, P.R. China
| | - Xinhang Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Shenzhen-Hong Kong International Science and Technology Park, No. 3 Binglang Road, Futian Free Trade Zone, Futian District, Shenzhen, Guangdong 518045, P.R. China
| | - Haimeng Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Shenzhen-Hong Kong International Science and Technology Park, No. 3 Binglang Road, Futian Free Trade Zone, Futian District, Shenzhen, Guangdong 518045, P.R. China
| | - Aditya Tan
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
| | - Yunlin Chi
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
| | - Daozheng Fang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Shenzhen-Hong Kong International Science and Technology Park, No. 3 Binglang Road, Futian Free Trade Zone, Futian District, Shenzhen, Guangdong 518045, P.R. China
| | - Xinyue Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Shenzhen-Hong Kong International Science and Technology Park, No. 3 Binglang Road, Futian Free Trade Zone, Futian District, Shenzhen, Guangdong 518045, P.R. China
| | - Zhihao Liu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Shenzhen-Hong Kong International Science and Technology Park, No. 3 Binglang Road, Futian Free Trade Zone, Futian District, Shenzhen, Guangdong 518045, P.R. China
| | - Qixiang Shang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Shenzhen-Hong Kong International Science and Technology Park, No. 3 Binglang Road, Futian Free Trade Zone, Futian District, Shenzhen, Guangdong 518045, P.R. China
| | - Yong Zhu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Shenzhen-Hong Kong International Science and Technology Park, No. 3 Binglang Road, Futian Free Trade Zone, Futian District, Shenzhen, Guangdong 518045, P.R. China
| | - Judyta Cielecka-Piontek
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3 Str., 60-806 Poznan, Poland
| | - Jihang Chen
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Shenzhen-Hong Kong International Science and Technology Park, No. 3 Binglang Road, Futian Free Trade Zone, Futian District, Shenzhen, Guangdong 518045, P.R. China
| |
Collapse
|
39
|
Sharma J, Dey P. Differential modulation of the hepatocellular metabolome, cytoprotective and inflammatory responses due to endotoxemia and lipotoxicity. Mol Omics 2025; 21:152-163. [PMID: 39744997 DOI: 10.1039/d4mo00140k] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2025]
Abstract
The present work aimed to examine the primary mechanisms of liver damage, namely the impact of gut-derived endotoxins along the gut-liver axis and adipose-derived free fatty acids along the adipose-liver axis. These processes are known to play a significant role in the development of hepatic inflammation and steatosis. Although possible overlapping in the pathogenesis was expected, these processes have unique pathophysiological consequences. Therefore, we used HepG2 cells as a model system to investigate the impact of lipopolysaccharides (LPS) and free fatty acid (FFA; albumin conjugated palmitic acid) on the intracellular metabolome. Although both LPS and FFA triggered the expression of nuclear factor κB (NFκB)-dependent inflammation, only LPS treatment was able to trigger a Toll-like receptor 4 (TLR4) dependent response. The intracellular cytoprotective enzymatic levels (catalase, peroxidase, glutathione) were increased due to FFA but lowered due to LPS. The free-radical neutralizing efficacies of cell-free metabolites of FFA-treated cells were better than those of the LPS-treated ones. The use of untargeted metabolomics allowed for the identification of distinct metabolic pathway enrichments, providing further insights into the differential effects of LPS and FFA on the metabolism of hepatocytes. Collectively, the current study highlights the distinct impacts of endotoxemia and lipotoxicity on the metabolome of hepatocytes, hence offering valuable insights into hepatocellular function.
Collapse
Affiliation(s)
- Jyoti Sharma
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India.
| | - Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India.
| |
Collapse
|
40
|
Nie T, Nepovimova E, Wu Q. Circadian rhythm, hypoxia, and cellular senescence: From molecular mechanisms to targeted strategies. Eur J Pharmacol 2025; 990:177290. [PMID: 39863143 DOI: 10.1016/j.ejphar.2025.177290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/03/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
Cellular senescence precipitates a decline in physiological activities and metabolic functions, often accompanied by heightened inflammatory responses, diminished immune function, and impaired tissue and organ performance. Despite extensive research, the mechanisms underpinning cellular senescence remain incompletely elucidated. Emerging evidence implicates circadian rhythm and hypoxia as pivotal factors in cellular senescence. Circadian proteins are central to the molecular mechanism governing circadian rhythm, which regulates homeostasis throughout the body. These proteins mediate responses to hypoxic stress and influence the progression of cellular senescence, with protein Brain and muscle arnt-like 1 (BMAL1 or Arntl) playing a prominent role. Hypoxia-inducible factor-1α (HIF-1α), a key regulator of oxygen homeostasis within the cellular microenvironment, orchestrates the transcription of genes involved in various physiological processes. HIF-1α not only impacts normal circadian rhythm functions but also can induce or inhibit cellular senescence. Notably, HIF-1α may aberrantly interact with BMAL1, forming the HIF-1α-BMAL1 heterodimer, which can instigate multiple physiological dysfunctions. This heterodimer is hypothesized to modulate cellular senescence by affecting the molecular mechanism of circadian rhythm and hypoxia signaling pathways. In this review, we elucidate the intricate relationships among circadian rhythm, hypoxia, and cellular senescence. We synthesize diverse evidence to discuss their underlying mechanisms and identify novel therapeutic targets to address cellular senescence. Additionally, we discuss current challenges and suggest potential directions for future research. This work aims to deepen our understanding of the interplay between circadian rhythm, hypoxia, and cellular senescence, ultimately facilitating the development of therapeutic strategies for aging and related diseases.
Collapse
Affiliation(s)
- Tong Nie
- College of Life Science, Yangtze University, Jingzhou, 434025, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, 434025, China.
| |
Collapse
|
41
|
Xin M, Bi F, Wang C, Huang Y, Xu Y, Liang S, Cai T, Xu X, Dong L, Li T, Wang X, Fang Y, Xu Z, Wang M, Song X, Zheng Y, Sun W, Li L. The circadian rhythm: A new target of natural products that can protect against diseases of the metabolic system, cardiovascular system, and nervous system. J Adv Res 2025; 69:495-514. [PMID: 38631431 PMCID: PMC11954810 DOI: 10.1016/j.jare.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/17/2024] [Accepted: 04/07/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND The treatment of metabolic system, cardiovascular system, and nervous system diseases remains to be explored. In the internal environment of organisms, the metabolism of substances such as carbohydrates, lipids and proteins (including biohormones and enzymes) exhibit a certain circadian rhythm to maintain the energy supply and material cycle needed for the normal activities of organisms. As a key factor for the health of organisms, the circadian rhythm can be disrupted by pathological conditions, and this disruption accelerates the progression of diseases and results in a vicious cycle. The current treatments targeting the circadian rhythm for the treatment of metabolic system, cardiovascular system, and nervous system diseases have certain limitations, and the identification of safer and more effective circadian rhythm regulators is needed. AIM OF THE REVIEW To systematically assess the possibility of using the biological clock as a natural product target for disease intervention, this work reviews a range of evidence on the potential effectiveness of natural products targeting the circadian rhythm to protect against diseases of the metabolic system, cardiovascular system, and nervous system. This manuscript focuses on how natural products restore normal function by affecting the amplitude of the expression of circadian factors, sleep/wake cycles and the structure of the gut microbiota. KEY SCIENTIFIC CONCEPTS OF THE REVIEW This work proposes that the circadian rhythm, which is regulated by the amplitude of the expression of circadian rhythm-related factors and the sleep/wake cycle, is crucial for diseases of the metabolic system, cardiovascular system and nervous system and is a new target for slowing the progression of diseases through the use of natural products. This manuscript provides a reference for the molecular modeling of natural products that target the circadian rhythm and provides a new perspective for the time-targeted action of drugs.
Collapse
Affiliation(s)
- Meiling Xin
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100000, China
| | - Fangjie Bi
- Heart Center, Zibo Central Hospital, Zibo, Shandong 255000, China
| | - Chao Wang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Yuhong Huang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Yujia Xu
- Department of Echocardiography, Zibo Central Hospital, Zibo, Shandong 255000, China
| | - Shufei Liang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Tianqi Cai
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Xiaoxue Xu
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Ling Dong
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Tianxing Li
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100000, China; Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xueke Wang
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100000, China; The Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yini Fang
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100000, China; Basic Medical College, Zhejiang Chinese Medical University, Hangzhou 310053 China
| | - Zhengbao Xu
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Meng Wang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Xinhua Song
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China.
| | - Yanfei Zheng
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100000, China.
| | - Wenlong Sun
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China.
| | - Lingru Li
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100000, China.
| |
Collapse
|
42
|
Zheng T, Zhao J, Sun C, Zhu C, Li C, Zhou J, Yang J, Zhang Y. Phenylthiourea synergistically enhances the hepatotoxicity of bavachalcone by interfering with metabolism in zebrafish: A factor to consider when evaluating toxicity of environmental pollutants using zebrafish models. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:118016. [PMID: 40056746 DOI: 10.1016/j.ecoenv.2025.118016] [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: 11/05/2024] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 03/10/2025]
Abstract
Phenylthiourea (PTU) is a well-known inhibitor of melanin synthesis that has been extensively utilized in ecotoxicological studies involving zebrafish. Although there are reports suggesting that PTU may influence the toxicity of various compounds, the underlying mechanisms of its action remain unclear. Bavachalcone (BavaC) has a wide range of applications in agriculture and medicine, and it can enter groundwater through a variety of pathways that may pose a risk to aquatic ecosystems. We found that PTU enhanced the hepatotoxicity of BavaC in zebrafish, but the mechanism was unclear. In this study, the interactive effects of 200 μM PTU and 2.5 μM BavaC on the toxicity of zebrafish larvae were evaluated after 72 h of exposure. PTU significantly increased BavaC-induced hepatotoxicity, which was characterized by liver hypoplasia, hepatocyte vacuolation, and lipid accumulation. Matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI) revealed that the contents of fatty acids, phosphatidylcholine and glutathione significantly increased. The results of RNA sequencing (RNA-seq) and Real-time PCR (RT-qPCR) analyses indicated that several metabolic pathways, including xenobiotic biodegradation and metabolism, amino acid metabolism, lipid metabolism and carbohydrate metabolism, were differentially regulated in the PTU+BavaC group compared to the BavaC group. Our findings indicate that PTU-induced metabolic disorders exacerbate BavaC hepatotoxicity and highlight the need to reconsider the use of PTU in zebrafish-based toxicity assessments of environmental pollutants.
Collapse
Affiliation(s)
- Te Zheng
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan, Shandong Province, China
| | - Jingcheng Zhao
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan, Shandong Province, China
| | - Chenglong Sun
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China; Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Chengyue Zhu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan, Shandong Province, China
| | - Chenqinyao Li
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan, Shandong Province, China
| | - Jiashuo Zhou
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan, Shandong Province, China
| | - Jing Yang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan, Shandong Province, China
| | - Yun Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan, Shandong Province, China.
| |
Collapse
|
43
|
Bian Y, Wu H, Jiang W, Kong X, Xiong Y, Zeng L, Zhang F, Song J, Wang C, Yang Y, Zhang X, Zhang Y, Pang P, Duo T, Wang Z, Pan T, Yang B. Anti-b diminishes hyperlipidaemia and hepatic steatosis in hamsters and mice by suppressing the mTOR/PPARγ and mTOR/SREBP1 signalling pathways. Br J Pharmacol 2025; 182:1254-1272. [PMID: 39614407 DOI: 10.1111/bph.17397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/02/2024] [Accepted: 10/15/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND AND PURPOSE As a chronic metabolic syndrome, hyperlipidaemia is manifested as aberrantly elevated cholesterol and triglyceride (TG) levels, primarily attributed to disorders in lipid metabolism. Despite the promising outlook for hyperlipidaemia treatment, the need persists for the development of lipid-lowering agents with heightened efficiency and minimal toxicity. This investigation aims to elucidate the lipid-lowering effects and potential pharmacodynamic mechanisms of Anti-b, a novel low MW compound. EXPERIMENTAL APPROACH We employed high-fat diet (HFD) in hamsters and mice or oleic acid (OA) in cultures of HepG2 cells and LO2 cells to induce hyperlipidaemia models. We administered Anti-b to assess its therapeutic effects on dyslipidaemia and hepatic steatosis. We used western blotting, RNA sequencing, GO and KEGG analysis, oil red O staining, along with molecular docking and molecular dynamics simulation to elucidate the mechanisms underlying the effects of Anti-b. KEY RESULTS Anti-b exhibited a substantial reduction in HFD-induced elevation of blood lipids, liver weight to body weight ratio, liver diameter and hepatic fat accumulation. Moreover, Anti-b demonstrated therapeutic effects in alleviating total cholesterol (TC), TG levels, and lipid accumulation derived from OA in HepG2 cells and LO2 cells. Mechanistically, Anti-b selectively bound to the mTOR kinase protein and increased mTOR thermal stability, resulting in downregulation of phosphorylation level. Notably, Anti-b exerted anti-hyperlipidaemia effects by modulating PPARγ and SREBP1 signalling pathways and reducing the expression level of mSREBP1 and PPARγ proteins. CONCLUSION AND IMPLICATIONS In conclusion, our study has provided initial data of a novel low MW compound, Anti-b, designed and synthesised to target mTOR protein directly. Our results indicate that Anti-b may represent a novel class of drugs for the treatment of hyperlipidemia and hepatic steatosis.
Collapse
Affiliation(s)
- Yu Bian
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Han Wu
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Weitao Jiang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xue Kong
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yuting Xiong
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Linghua Zeng
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Feng Zhang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jinglun Song
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Chunlei Wang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yang Yang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xinyue Zhang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yuning Zhang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Ping Pang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Tianqi Duo
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Zhuo Wang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Tengfei Pan
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Baofeng Yang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| |
Collapse
|
44
|
Bayoude A, Zhang J, Shen Y, Tilyek A, Chai C. Ribes diacanthum Pall modulates bile acid homeostasis and oxidative stress in cholestatic mice by activating the SIRT1/FXR and Keap1/Nrf2 signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119400. [PMID: 39864603 DOI: 10.1016/j.jep.2025.119400] [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: 12/22/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 01/28/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cholestatic liver injury (CLI) is a pathophysiological syndrome characterized by the accumulation of bile acids (BAs), which leads to significant hepatic dysfunction. This condition is frequently associated with disturbances in BAs homeostasis and the induction of oxidative stress. Ribes diacanthum Pall (RDP), a conventional folk medicinal plant, has been employed in Mongolia, the Inner Mongolia region of China, and other areas for the remediation of hepatic disorders. However, the specific mechanism and chemical composition by which RDP exerts its effects remain unknown. AIM OF THE STUDY The aim of this research was to assess the protective impact of RDP on CLI and probe into the underlying mechanism and pinpoint the active constituents of RDP. MATERIALS AND METHODS For this study, a CLI mouse model induced via bile duct ligation (BDL) was used to investigate the hepatoprotective effect of RDP. Mice were administered low, medium, or high doses of RDP for 6 consecutive days, beginning 3 days prior to BDL induction. Subsequently, serum biochemical parameters, hepatic histopathology, and cholestatic markers were analyzed. An HPLC-QTOF-MS/MS analysis was also conducted to identify the prototype constituents in RDP. Furthermore, component-directed network pharmacology was utilized to identify the active constituents, central targets, and signaling cascades of RDP. Eventually, quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting were adopted to confirm the associated antioxidant enzymes, BAs transporters, and metabolic enzymes. Molecular docking was applied to forecast the binding affinity between the components and core targets. RESULTS RDP effectively ameliorated the pathological liver damage and cholestasis in BDL-induced CLI mice. Moreover, 43 components within RDP were identified through HPLC-QTOF-MS/MS analysis. Altogether 106 potential targets were detected, and the high-affinity targets, namely Keap1 and SIRT1, were located through the PPI network. The results of GO and KEGG analysis indicated that the reaction to oxidative stress and BAs homeostasis are significantly associated with the RDP treatment of CLI. In the in vivo experimental study, the findings revealed that RDP alleviated the BDL-induced oxidative damage. Simultaneously, RDP augmented the expressions of BAs efflux transporters and the metabolic enzymes in liver tissues, thus promoting BAs excretion and metabolism in cholestatic rodents. Mechanically, RDP attenuated hepatic oxidative stress and the accumulation of BAs, protecting the liver from BDL-induced cholestasis via the Keap1/Nrf2 and SIRT1/FXR signaling axis. The molecular docking result indicated that bolusanthol C and 3,6,3',4'-tetrahydroxyflavone possess a superior binding affinity to the two core targets (Keap1, SIRT1). CONCLUSION These results suggest that RDP ameliorate CLI by regulating BAs homeostasis and alleviating oxidative stress through the SIRT1/FXR and Keap1/Nrf2 signaling pathways, presenting a novel therapeutic strategy for cholestasis. Additionally, bolusanthol C and 3,6,3',4'-tetrahydroxyflavone may function as key pharmacological agents in RDP, responsible for its protective effects against CLI.
Collapse
Affiliation(s)
- Alamusi Bayoude
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing, 211198, China
| | - Jiaxin Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing, 211198, China
| | - Yuanjiang Shen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing, 211198, China
| | - Akhtolkhyn Tilyek
- Department of Pharmaceutical Chemistry and Pharmacognosy, Mongolian University of Pharmaceutical Sciences, Ulaanbaatar, 18130, Mongolia
| | - Chengzhi Chai
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
45
|
Shi M, Ma G, Yang X. Artesunate: A Review of Its Potential Therapeutic Effects and Mechanisms in Digestive Diseases. Pharmaceutics 2025; 17:299. [PMID: 40142963 PMCID: PMC11945051 DOI: 10.3390/pharmaceutics17030299] [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: 01/13/2025] [Revised: 01/31/2025] [Accepted: 02/10/2025] [Indexed: 03/28/2025] Open
Abstract
Artesunate (ART), an artemisinin-derived semi-synthetic sesquiterpene lactone distinguished by its unique endoperoxide group, has become a cornerstone of clinical antimalarial therapy. Recent research has demonstrated its broad pharmacological profile, including its potent antimalarial, anti-inflammatory, anti-tumor, antidiabetic, immunomodulatory, and anti-fibrotic properties. These discoveries have significantly broadened the therapeutic scope of ART and offer new perspectives for its potential use in treating gastrointestinal disorders. Mechanistically, ART exerts significant therapeutic effects against diverse gastrointestinal pathologies-such as gastric ulcers, ulcerative colitis (UC), hepatic fibrosis (HF), gastric cancer, hepatocellular carcinoma, and colorectal cancer-via multimodal mechanisms, including cell cycle modulation, apoptosis induction, the suppression of tumor cell invasion and migration, proliferation inhibition, ferroptosis activation, and immune regulation. This review evaluates existing evidence on ART's therapeutic applications and molecular mechanisms in digestive diseases, intending to elucidate its clinical translation potential. ART emerges as a promising multi-target agent with significant prospects for improving the management of gastrointestinal disorders.
Collapse
Affiliation(s)
| | | | - Xiulan Yang
- Department of Pharmacology, The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (M.S.)
| |
Collapse
|
46
|
Markouli M, Skouras P, Piperi C. Impact of cuproptosis in gliomas pathogenesis with targeting options. Chem Biol Interact 2025; 408:111394. [PMID: 39848557 DOI: 10.1016/j.cbi.2025.111394] [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: 11/05/2024] [Revised: 01/10/2025] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
Gliomas constitute the most prevalent primary central nervous system tumors, often characterized by complex metabolic profile, genomic instability, and aggressiveness, leading to frequent relapse and high mortality rates. Traditional treatments are commonly ineffective because of gliomas increased heterogeneity, invasive characteristics and resistance to chemotherapy. Among several pathways affecting cellular homeostasis, cuproptosis has recently emerged as a novel type of programmed cell death, triggered by accumulation of copper ions. Although the precise molecular mechanisms of cuproptosis are not fully elucidated, there is evidence that copper ions can target mitochondrial lipoylated proteins, disrupting the tricarboxylic acid cycle and electron transport chain, thus leading to deregulated mitochondrial metabolism, protein aggregation and cell death. Of importance, altered expression of copper transporters and abnormally high intracellular copper levels have been observed in several cancer types, including gliomas, contributing to tumor growth and metastasis. Furthermore, a range of prognostic models incorporating cuproptosis-related genes and lncRNAs have been proposed and are currently under clinical validation. Drugs modulating cuproptosis or interfering with copper-binding proteins are under development, causing metabolic failure and cell death, thus offering potential new avenues for glioma diagnosis and therapy. In this article, we explore the role of copper metabolism in gliomas and the potential synergistic effects of cuproptosis-based treatments with current therapies, in effective targeting of tumor progression and chemoresistance.
Collapse
Affiliation(s)
- Mariam Markouli
- Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA, 02118, USA.
| | - Panagiotis Skouras
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece.
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece.
| |
Collapse
|
47
|
Li D, Lan X, Xu L, Zhou S, Luo H, Zhang X, Yu W, Yang Y, Fang X. Influence of gut microbial metabolites on tumor immunotherapy: mechanisms and potential natural products. Front Immunol 2025; 16:1552010. [PMID: 40066456 PMCID: PMC11891355 DOI: 10.3389/fimmu.2025.1552010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 02/06/2025] [Indexed: 05/13/2025] Open
Abstract
In recent years, tumor immunotherapy has made significant breakthroughs in the treatment of malignant tumors. However, individual differences in efficacy have been observed in clinical practice. There is increasing evidence that gut microbial metabolites influence the efficacy of distal tumor immunotherapy via the gut-liver axis, the gut-brain axis and the gut-breast axis, a process that may involve modulating the expression of immune cells and cytokines in the tumor microenvironment (TME). In this review, we systematically explore the relationship between gut microbial metabolites and tumor immunotherapy, and examine the corresponding natural products and their mechanisms of action. The in-depth exploration of this research area will provide new ideas and strategies to enhance the efficacy of tumor immunotherapy and mitigate adverse effects.
Collapse
Affiliation(s)
- Dongyang Li
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Xintian Lan
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Linyi Xu
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Shuo Zhou
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Haoming Luo
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Xiaoying Zhang
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, China
| | - Wenbo Yu
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Yonggang Yang
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Xiaoxue Fang
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
48
|
Gao C, Yang H, Cheng J, He S, Yang Y, Xu L, Ma Q, Guo X, Zhong X. STM2457 impairs the proliferation of esophageal squamous cell carcinoma by activating DNA damage response through ATM-Chk2 axis. Med Oncol 2025; 42:82. [PMID: 39985567 DOI: 10.1007/s12032-025-02634-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 02/13/2025] [Indexed: 02/24/2025]
Abstract
METTL3 has been proven to play an important role in the proliferation of Esophageal squamous cell carcinoma (ESCC). In this study, we focused on investigating the therapeutic role and molecular mechanism of STM2457 in ESCC, which is a novel small-molecule inhibitor of METTL3. The effect of STM2457 on ESCC was evaluated using ESCC cell lines by the cell viability measurement, cloning formation assay, scratching assay, transwell assay, and flow cytometry techniques. Furthermore, the molecular mechanism study was employed to evaluate by RT-qPCR, Western blotting, proteomics analysis, comet assay, etc. Additionally, the anticancer effect of STM2457 was carried out by nude mice tumor xenograft in vivo. This study showed STM2457 could significantly inhibit the proliferation and migration of Eca109 and KYSE150 cells, which promoted G0/G1 phase arrest and apoptosis in a dose-dependent manners in vitro. Moreover, proteomics analysis suggested the important role of ATM in action mechanism of STM2457. Further studies showed that STM2457 may activate DNA damage response and the expression of ATM, p-ATM, p-Chk2, and γ-H2AX protein in ATM-Chk2 pathway. Intriguingly, ATM inhibitor CGK-733 and knocking down ATM significantly reduced the expression of ATM in Eca109 and KYSE150 cells treated with STM2457. Importantly, STM2457 significantly upregulated the expression of ATM and γ-H2AX protein and inhibited the growth of ESCC in vivo. Finally, STM2457 combined with PTX could also significantly inhibit the proliferation and migration ability of Eca109 and KYSE150 cells by targeting the ATM-mediated DDR pathway. In tumor-bearing nude mice model, STM2457 combined with Paclitaxel can inhibit the growth of ESCC and increased the expression of ATM and γ-H2AX protein. These findings revealed ATM-Chk2 pathway is a promising therapeutic target for STM2457 to effectively inhibit the proliferation of ESCC.
Collapse
Affiliation(s)
- Chuanli Gao
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Hui Yang
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Jiao Cheng
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Shuang He
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Yong Yang
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Lei Xu
- Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Qiang Ma
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.
| | - Xiaolan Guo
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.
| | - Xiaowu Zhong
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.
| |
Collapse
|
49
|
He Q, Liu X, Ding G, Wang Y, Luo X, Cao W, Xing W. The relationship between serum uric acid level and non-alcoholic fatty liver disease in northern China: a retrospective cohort study. BMC Public Health 2025; 25:718. [PMID: 39984884 PMCID: PMC11843771 DOI: 10.1186/s12889-025-21943-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 02/13/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) has become the most prevalent chronic liver disease among adults. High uric acid (UA) increases the incidence of NAFLD in the general population. However, further exploration is warranted to determine the relationship between UA levels and NAFLD in various populations. We conducted a historical cohort study to investigate the causality between UA and NAFLD across different weight categories. METHODS A historical cohort was established from the Jidong community cohort. All participants were enrolled and followed up from July 1st, 2013 to August 1st, 2018. The study participants were retrospectively assigned to four groups according to their UA levels (Q1, 69-210 μmol/L; Q2, 211-255 μmol/L; Q3, 256-310 μmol/L; Q4, 311-593 μmol/L). The NAFLD incidence was investigated in each group. We used the UA level determined by an automatic analyzer. NAFLD was diagnosed with abdominal ultrasonography examination. Demographic information, lifestyle history, clinical anthropometric data, and blood samples of participants were collected. Univariate analysis and multivariable Cox regression were applied to analyze the relationship between UA and NAFLD by stratification of participants' body mass index (BMI) categories (underweight, normal weight, overweight, and obese). RESULTS Two thousand nine hundred eighty four participants were enrolled. 740 (24.8%) were assigned to UA Q1 group, 755 (25.3%) to UA Q2, 743 (24.9%) to UA Q3, and 746 (25.0%) to UA Q4. The global incidence of NAFLD was 26.0% (777/2984). The risk of NAFLD significantly increased with elevated UA levels in underweight and normal-weight participants (HR = 3.498, 95% CI: 2.413-5.072, P < 0.05). In multivariable analysis, UA showed a positive association with NAFLD, independent of other risk factors in underweight and normal-weight participants (UA Q2: 1.152 (0.761-1.743), UA Q3: 2.168 (1.489-3.157), UA Q4: 3.075 (2.103-4.196), P < 0.05). In the absence of other risk factors, high UA levels independently explained 17% of NAFLD risk in underweight and normal-weight participants. CONCLUSIONS High UA levels serve as an independent risk factor for NAFLD in underweight and normal-weight individuals, highlighting the necessity of early NAFLD screening through monitoring liver function and UA levels, and personalized treatment plans for NAFLD patients with higher UA levels, which may include uric acid-lowering therapy and lifestyle modifications. However, the relationship between UA levels and NAFLD in overweight and obese individuals remains inconclusive.
Collapse
Affiliation(s)
- Qian He
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xinyue Liu
- Tai'an City Center for Disease Control and Prevention, Taian, China
| | - Guoyong Ding
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yiying Wang
- Department of Medical, Rizhao Mental Health Center, Rizhao, China
| | - Xiaoting Luo
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wenyuan Cao
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Weijia Xing
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Shandong First Medical University & Shandong Academy of Medical Sciences, The Second Affiliated Hospital, Taian, 271000, China.
| |
Collapse
|
50
|
Li Q, Xiang J. METTL3 promotes the progression of non-alcoholic fatty liver disease by mediating m6A methylation of FAS. Sci Rep 2025; 15:6162. [PMID: 39979577 PMCID: PMC11842791 DOI: 10.1038/s41598-025-90419-z] [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: 11/07/2024] [Accepted: 02/12/2025] [Indexed: 02/22/2025] Open
Abstract
N6-methyladenosine (m6A) is involved in the development of non-alcoholic fatty liver disease (NAFLD). Here, we aimed to investigate the effect of m6A methyltransferase METTL3 on liver damage in high-fat diet (HFD)-induced mouse model and hepatocyte damage treated with free fatty acid (FFA). Plasma lipid, lipogenesis, viability, and apoptosis were measured to assess injury. m6A methylation was evaluated using m6A dot blot, methylated RNA immunoprecipitation, dual-luciferase reporter assay, and RNA decay assay. The results indicated that METTL3 was highly expressed in the liver of HFD mice, which knockdown improved plasma lipid and reduced liver lipids. Additionally, silencing of METTL3 promoted cell viability, inhibited apoptosis, reduced lipid concentrations, and downregulated lipogenesis-related marker levels. Moreover, METTL3 promoted the m6A methylation of FAS and enhanced its stability. In conclusion, silencing of METTL3 attenuates the progression of NAFLD by FAS m6A methylation, suggesting that METTL3 may be a promising target for treating NAFLD.
Collapse
Affiliation(s)
- Qunhua Li
- Department of Gastroenterology, Affiliated Hospital of Chengdu University, 2nd N Section of 2nd Ring Rd, Chengdu, 610036, Sichuan, China
| | - Junying Xiang
- Department of Gastroenterology, Affiliated Hospital of Chengdu University, 2nd N Section of 2nd Ring Rd, Chengdu, 610036, Sichuan, China.
| |
Collapse
|