1
|
Zarandi PK, Ghiasi M, Heiat M. The role and function of lncRNA in ageing-associated liver diseases. RNA Biol 2025; 22:1-8. [PMID: 39697114 PMCID: PMC11660375 DOI: 10.1080/15476286.2024.2440678] [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] [Revised: 10/09/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Liver diseases are a significant global health issue, characterized by elevated levels of disorder and death. The substantial impact of ageing on liver diseases and their prognosis is evident. Multiple processes are involved in the ageing process, which ultimately leads to functional deterioration of this organ. The process of liver ageing not only renders the liver more susceptible to diseases but also compromises the integrity of other organs due to the liver's critical function in metabolism regulation. A growing body of research suggests that long non-coding RNAs (lncRNAs) play a significant role in the majority of pathophysiological pathways. They regulate gene expression through a variety of interactions with microRNAs (miRNAs), messenger RNAs (mRNAs), DNA, or proteins. LncRNAs exert a major influence on the progression of age-related liver diseases through the regulation of cell proliferation, necrosis, apoptosis, senescence, and metabolic reprogramming. A concise overview of the current understanding of lncRNAs and their potential impact on the development of age-related liver diseases will be provided in this mini-review.
Collapse
Affiliation(s)
- Peyman Kheirandish Zarandi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Cancer Biology Signaling Pathway Interest Group (CBSPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohsen Ghiasi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
García-Juan M, Villa M, Benito-Cuesta I, Ordóñez-Gutiérrez L, Wandosell F. Reassessing the AMPK-MTORC1 balance in autophagy in the central nervous system. Neural Regen Res 2025; 20:3209-3210. [PMID: 39715086 PMCID: PMC11881726 DOI: 10.4103/nrr.nrr-d-24-00733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/26/2024] [Accepted: 09/19/2024] [Indexed: 12/25/2024] Open
Affiliation(s)
- Marta García-Juan
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Nicolas Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - Mario Villa
- Fisiología Animal, Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | - Irene Benito-Cuesta
- Department of Clinical Neuroscience, CMM Karolinska Universitetssjukhuset Solna, Stockholm, Sweden
| | - Lara Ordóñez-Gutiérrez
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Nicolas Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
- Departamento de Bioquímica y Biología Molecular, Universidad Complutense de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Francisco Wandosell
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Nicolas Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
3
|
Parchwani D, Singh R, Patel D. Biological and translational attributes of mitochondrial DNA copy number: Laboratory perspective to clinical relevance. World J Methodol 2025; 15:102709. [DOI: 10.5662/wjm.v15.i3.102709] [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: 10/26/2024] [Revised: 01/21/2025] [Accepted: 02/08/2025] [Indexed: 03/06/2025] Open
Abstract
The mitochondrial DNA copy number (mtDNAcn) plays a vital role in cellular energy metabolism and mitochondrial health. As mitochondria are responsible for adenosine triphosphate production through oxidative phosphorylation, maintaining an appropriate mtDNAcn level is vital for the overall cellular function. Alterations in mtDNAcn have been linked to various diseases, including neurodegenerative disorders, metabolic conditions, and cancers, making it an important biomarker for understanding the disease pathogenesis. The accurate estimation of mtDNAcn is essential for clinical applications. Quantitative polymerase chain reaction and next-generation sequencing are commonly employed techniques with distinct advantages and limitations. Clinically, mtDNAcn serves as a valuable indicator for early diagnosis, disease progression, and treatment response. For instance, in oncology, elevated mtDNAcn levels in blood samples are associated with tumor aggressiveness and can aid in monitoring treatment efficacy. In neurodegenerative diseases such as Alzheimer’s and Parkinson’s, altered mtDNAcn patterns provide insights into disease mechanisms and progression. Understanding and estimating mtDNAcn are critical for advancing diagnostic and therapeutic strategies in various medical fields. As research continues to uncover the implications of mtDNAcn alterations, its potential as a clinical biomarker is likely to expand, thereby enhancing our ability to diagnose and manage complex diseases.
Collapse
Affiliation(s)
- Deepak Parchwani
- Department of Biochemistry, All India Institute of Medical Sciences, Rajkot 360001, India
| | - Ragini Singh
- Department of Biochemistry, All India Institute of Medical Sciences, Rajkot 360001, India
| | - Digisha Patel
- Department of Physiology, Shantabaa Medical College and General Hospital Amreli, Amreli 365601, Gujarāt, India
| |
Collapse
|
4
|
Zhao Y, Wang Y, Gong W, Duan C, Ren J, Zhang H, Nie X. Energy metabolism disturbance induced by atorvastatin exposure on yellowstripe goby (Mugilogobius chulae) larvae based on transcriptome and metabolome analysis. J Environ Sci (China) 2025; 155:475-487. [PMID: 40246482 DOI: 10.1016/j.jes.2024.09.001] [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: 03/04/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 04/19/2025]
Abstract
Atorvastatin (ATV), a commonly prescribed lipid-lowering drug, has been widely detected in various aquatic environments due to its large use and low degradation rate. Since the target gene inhibited by ATV is highly conserved in organisms, many studies have shown that ATV can interfere with lipid metabolism in aquatic non-target organisms. However, studies on mitochondria, energy metabolism, and developmental toxicity of ATV on non-target organisms are limited. In this study, Mugilogobius chulae embryos were exposed to ATV (0.5 and 50 µg/L) until 96 hour post fertilization (hpf). The results confirmed that the environmental concentrations of ATV caused toxic effects including developmental malformations, pathological damage, hepatotoxicity, and oxidative stress in M. chulae larvae. Both transcriptomic and metabolomic analyses showed that ATV exposure interfered the normal processes of oxidative phosphorylation and TCA cycle, resulting in energy metabolic disorder. In addition, ATV exposure also damaged the mitochondrial structure of M. chulae larvae. Thus, M. chulae could regulate PI3K/AMPK/FoxO proteins to promote mitochondrial regeneration, support autophagy, and even initiate apoptosis to maintain metabolism homeostasis. Taken together, our findings suggested that mitochondrial dysfunction and metabolic disorder were involved in ATV-induced toxicity which may cause developmental malformations and abnormalities, providing novel insight into the toxic mechanisms of ATV.
Collapse
Affiliation(s)
- Yufei Zhao
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Yimeng Wang
- Department of Ecology, Jinan University, Guangzhou 510632, China; Guangdong Laboratory Animals Monitoring Institute, Guangzhou 510663, China
| | - Weibo Gong
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Chunni Duan
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Jinzhi Ren
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Huiyu Zhang
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Xiangping Nie
- Department of Ecology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
5
|
Xu Q, Zhou Y, Wu M, Wu S, Yu J, Xu Y, Wei Z, Jin L. MTHFD2: A metabolic checkpoint altering trophoblast invasion and migration by remodeling folate-nucleotide metabolism in recurrent spontaneous abortion. Cell Signal 2025; 132:111808. [PMID: 40250694 DOI: 10.1016/j.cellsig.2025.111808] [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: 01/03/2025] [Revised: 03/30/2025] [Accepted: 04/11/2025] [Indexed: 04/20/2025]
Abstract
Recurrent spontaneous abortion (RSA) affects female reproduction worldwide, yet its pathological mechanisms are still unclear. It has been reported that cellular metabolism reprogramming is a critical step for trophoblasts during embryo implantation. Herein, MTHFD2 was recognized as a key metabolic checkpoint attributed to RSA occurrence. This work figured out that the expression level of MTHFD2 was significantly inhibited in villus tissues from RSA patients, suggesting the potential role of MTHFD2 in RSA occurrence. Moreover, MTHFD2 knockdown impaired cellular folate-nucleotide metabolism, induced the accumulation of AICAR, and thereby impairing the EMT process to inhibit the invasion and migration of trophoblasts Besides, the AICAR accumulation further activated the downstream AMPK which deactivated the JAK/STAT/Slug pathway and ultimately deactivated the EMT process. Using a mouse model, MTHFD2 inhibition was observed to induce embryo implantation failure in vivo. Our results highlighted MTHFD2 as a metabolic checkpoint that remodeled folate-nucleotide metabolism to regulate the EMT process and ultimately altered the migration and invasion of trophoblasts in RSA occurrence. Our findings suggested that MTHFD2 was a promising therapeutic target in recurrent spontaneous abortion treatment.
Collapse
Affiliation(s)
- Qingxin Xu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yicheng Zhou
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Meijuan Wu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Shengnan Wu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Jing Yu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Pathology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yao Xu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Zhiyun Wei
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China; Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, China.
| | - Liping Jin
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China; Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, China.
| |
Collapse
|
6
|
Dai C, Tong Y, Bai N, Xu N, Zhao X, Zhou L, Tang Z, Liu M, Xu B, Liu X, Chen Y, Lin Z, Li J, Bian S, Zheng W. Decoding the role of nucleic acid binding protein 2 in lipid dysregulation and hepatocellular carcinoma progression through LKB1-mediated mitochondrial dysfunction. Cell Signal 2025; 132:111820. [PMID: 40250697 DOI: 10.1016/j.cellsig.2025.111820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/22/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
Nucleic Acid Binding Protein 2 (NABP2), a crucial regulator in the single-stranded DNA-binding protein family, has been linked to the progression of hepatocellular carcinoma (HCC) and poor prognosis. However, the precise mechanisms by which NABP2 regulates HCC development, especially through metabolic pathways, remain unclear. In this study, we evaluated NABP2 expression in clinical HCC samples and analyzed its correlation with patient survival outcomes. Functional assays, including cell proliferation, migration, and lipid metabolism analyses, were performed in vitro and in vivo to investigate the role of NABP2 in tumorigenesis. Additionally, we examined the molecular interactions of NABP2 with the E3 ubiquitin ligase STUB1 and its impact on the LKB1/AMPK signaling pathway. Our results revealed that NABP2 was overexpressed in HCC tissues and associated with worse survival outcomes. NABP2 promoted tumor cell proliferation, migration, and disrupted lipid metabolism. Mechanistically, NABP2 regulated the proteostasis of liver kinase B1 (LKB1) by recruiting STUB1, leading to the inhibition of the LKB1/AMPK signaling axis and mitochondrial dysfunction. In conclusion, our findings suggest that NABP2 may serve as both a biomarker and a potential therapeutic target for HCC, offering novel insights into its role in metabolic reprogramming and tumor progression.
Collapse
Affiliation(s)
- Chengchen Dai
- Research Center of Clinical Medicine & Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China; Department of Oncology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Yun Tong
- Department of Oncology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Nan Bai
- Research Center of Clinical Medicine & Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China; Department of Oncology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Nuo Xu
- Research Center of Clinical Medicine & Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Xuying Zhao
- Research Center of Clinical Medicine & Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Lihua Zhou
- Department of Oncology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Zhangzhi Tang
- Department of Oncology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Mingyu Liu
- Department of Oncology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Banglong Xu
- Research Center of Clinical Medicine & Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Xiaoquan Liu
- Research Center of Clinical Medicine & Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Yinqi Chen
- Research Center of Clinical Medicine & Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Zhaoyi Lin
- Research Center of Clinical Medicine & Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Jinlong Li
- College of Pharmacology, Nantong University, Nantong, China.
| | - Saiyan Bian
- Research Center of Clinical Medicine & Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| | - Wenjie Zheng
- Research Center of Clinical Medicine & Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China; Department of Oncology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China; Department of Pathology, Medical School of Nantong University, Nantong 226001, China.
| |
Collapse
|
7
|
He Z, Xiong H, Cai Y, Chen W, Shi M, Liu L, Wu K, Deng X, Deng X, Chen T. Clostridium butyricum ameliorates post-gastrectomy insulin resistance by regulating the mTORC1 signaling pathway through the gut-liver axis. Microbiol Res 2025; 297:128154. [PMID: 40188705 DOI: 10.1016/j.micres.2025.128154] [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: 03/23/2025] [Accepted: 03/25/2025] [Indexed: 05/04/2025]
Abstract
Postoperative insulin resistance (IR) is a metabolic disorder characterized by decreased insulin sensitivity and elevated blood glucose levels following major surgery. Our previous clinical study identified a notable correlation between postoperative IR and gut microbiota, particularly butyrate-producing bacteria, yet the mechanisms remain unclear. In this study, we established gastric resection SD rat models to evaluate the impact of Clostridium butyricum NCU-27 (butyrate-producing bacteria) on postoperative IR. The results demonstrated significant reductions in fasting blood glucose (FBG), fasting insulin (FIns) levels, and HOMA-IR (6.64 ± 0.76 vs. 11.47 ± 1.32; 4.27 ± 0.59 vs. 7.40 ± 0.54) in the postoperative period compared to the control group (P < 0.05). Additionally, glucose tolerance and hepatic glycogen content were markedly improved (P < 0.001). Further exploration of butyrate demonstrated effects similar to C. butyricum NCU-27, potentially mediated through the gut-liver axis by inhibiting mTORC1 expression in liver cells, activating the IRS1/AKT pathway, enhancing glucose uptake and glycogen synthesis, suppressing gluconeogenesis, increasing insulin sensitivity, and improving IR. Finally, the use of mTORC1 agonists and inhibitors further confirmed the critical role of the mTORC1 pathway in mediating the beneficial effects of C. butyricum NCU-27 and butyrate on postoperative IR. In conclusion, this study elucidated that C. butyricum NCU-27 improves postoperative IR by regulating butyrate metabolism and inhibiting the mTORC1 pathway, offering new insights for preventing and treating post-gastrectomy IR.
Collapse
Affiliation(s)
- Zhipeng He
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, China; Jiangxi Province Key Laboratory of Bioengineering Drugs, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Huan Xiong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Yujie Cai
- Jiangxi Province Key Laboratory of Bioengineering Drugs, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Wenjing Chen
- Jiangxi Province Key Laboratory of Bioengineering Drugs, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Meng Shi
- Department of Gastrointestinal Surgery, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei 442008, China
| | - Lulin Liu
- Department of Vascular Surgery, Heyuan Hospital of Guangdong Provincial People's Hospital, Heyuan, Guangdong 51700, China
| | - Kai Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Xi Deng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Xiaorong Deng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, China.
| | - Tingtao Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, China; Jiangxi Province Key Laboratory of Bioengineering Drugs, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, China.
| |
Collapse
|
8
|
Reglero-Real N, Rolas L, Nourshargh S. Aging microvasculature: Effects on immune cell trafficking and inflammatory diseases. J Exp Med 2025; 222:e20242154. [PMID: 40455014 PMCID: PMC12128883 DOI: 10.1084/jem.20242154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 04/07/2025] [Accepted: 05/14/2025] [Indexed: 06/11/2025] Open
Abstract
Leukocyte recruitment to sites of inflammation is vital for orchestrating an effective immune response. Key to this process is the ability of leukocytes to migrate through venular walls, engaging in sequential interactions with endothelial cells, pericytes, and the venular basement membrane. The aging process exerts profound effects on the molecular and functional properties of the vasculature, thereby influencing the profile and dynamics of leukocyte trafficking during inflammation. In this review, by focusing mainly on neutrophils, we summarize key examples of how the aged microvasculature and perivascular stroma cells promote dysregulated leukocyte-venular wall interactions and present the associated molecular mechanisms. Additionally, we discuss the functional implications of such aberrant leukocyte behavior to age-related and chronic inflammatory pathologies.
Collapse
Affiliation(s)
- Natalia Reglero-Real
- Departamento de Biología Molecular, Instituto Universitario de Biología Molecular (IUBM) and Centro de Biología Molecular Severo Ochoa (CBM), Universidad Autónoma de Madrid, UAM-CSIC, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | - Loïc Rolas
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sussan Nourshargh
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
9
|
Yin W, Xue H, Zhang Y, Li R, Liu M, Yue H, Ge D, Liu N. Steroid constituents of Solidago canadensis alleviate LPS-induced inflammation via AMPK regulated mitophagy/NLRP3 and NF-κB pathway. Eur J Pharmacol 2025; 998:177512. [PMID: 40113066 DOI: 10.1016/j.ejphar.2025.177512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 03/04/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
Inflammation is a major risk factor for a variety of human diseases, such as sepsis, Inflammatory Bowel Disease (IBD) and also major cardiovascular disease including atherosclerosis. Solidago canadensis is used as a traditional medicine to treat inflammation-related diseases. However, the component with anti-inflammatory activity of Solidago canadensis is not clear. In this study, we aimed to search for new bioactive steroids from Solidago canadensis and investigate their anti-inflammatory activity both in vitro and in vivo. Lipopolysaccharides (LPS)-stimulated RAW264.7 cells, mouse bone marrow-derived macrophages (BMDMs) and peripheral blood mononuclear cells (PBMCs) were used to induce an inflammation response. Compound 10 outperformed other compounds for superior anti-inflammatory activity and significant inhibition of NLR family, pyrin domain containing 3 (NLRP3) inflammasome activation. Mechanistically, compound 10 induced mitophagy by activating AMP-activated protein kinas (AMPK) to suppress NLRP3 inflammasome activation. Inhibiting AMPK by inhibitor BML-275 significantly attenuated compound 10 induced mitophagy and subsequent the NLRP3 inflammasome. Besides, the NF-κB activation, key step in NLRP3 inflammasome priming, was also suppressed by compound 10 via activation of AMPK. In addition, the in vivo experiments showed that compound 10 could alleviate LPS-induced inflammatory and dextran sulfate sodium salt -induced colitis in C57BL/6 mice. Collectively, the present study, for the first time, shows that the steroids compound 10 exhibited anti-inflammatory effect via AMPK/mitophagy/NLRP3 as well as AMPK/NF-κB/NLRP3 signaling pathway, which strongly suggests the therapeutic potential of compound 10 in various inflammatory diseases.
Collapse
Affiliation(s)
- Wenying Yin
- School of Biological Science and Technology, University of Jinan, Jinan, 250024, China.
| | - Han Xue
- School of Biological Science and Technology, University of Jinan, Jinan, 250024, China.
| | - Yongqi Zhang
- School of Biological Science and Technology, University of Jinan, Jinan, 250024, China.
| | - Rongxian Li
- School of Biological Science and Technology, University of Jinan, Jinan, 250024, China.
| | - Mengjia Liu
- School of Biological Science and Technology, University of Jinan, Jinan, 250024, China.
| | - Hongwei Yue
- Department of Emergency Medicine, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, China.
| | - Di Ge
- School of Biological Science and Technology, University of Jinan, Jinan, 250024, China.
| | - Na Liu
- School of Biological Science and Technology, University of Jinan, Jinan, 250024, China.
| |
Collapse
|
10
|
Asghari Alashti F, Goliaei B. Rethinking fat Browning: Uncovering new molecular insights into the synergistic roles of fasting, exercise, and cold exposure. Eur J Pharmacol 2025; 998:177651. [PMID: 40274179 DOI: 10.1016/j.ejphar.2025.177651] [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: 01/28/2025] [Revised: 04/13/2025] [Accepted: 04/17/2025] [Indexed: 04/26/2025]
Abstract
The global obesity epidemic highlights the need to understand the molecular mechanisms that regulate energy metabolism. Among emerging research areas, fat browning-the transformation of white adipose tissue into beige fat-has gained significant attention. This review explores the molecular pathways involved in fat browning triggered by fasting, physical exercise, and cold exposure, emphasizing both shared and distinct regulatory mechanisms. These stimuli consistently induce physiological responses such as lipolysis, mitochondrial biogenesis, and improved insulin sensitivity. Notably, PGC-1α and SIRT3 are upregulated across all three conditions, underscoring their central roles in mitochondrial function and energy metabolism and identifying them as promising therapeutic targets. In contrast, UCP1 and PRDM16 exhibit condition-specific regulation, suggesting they may not be universally essential for fat browning. In addition, the review discusses species-specific differences in brown adipose tissue (BAT) activation, particularly between rodents and humans, highlighting the challenges of translating animal model findings to human therapies. Future research should aim to develop selective pharmacological activators of PGC-1α and SIRT3 to enhance therapeutic outcomes while minimizing adverse effects. This review also proposes that integrating fasting, exercise, and cold exposure could provide innovative strategies to promote metabolic health.
Collapse
Affiliation(s)
- Fariborz Asghari Alashti
- Institute of Biochemistry and Biophysics (IBB), Laboratory of Biophysics and Molecular Biology, University of Tehran, Tehran, Iran; Sunnybrook Research Institute, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, M4N 3M5, Canada.
| | - Bahram Goliaei
- Institute of Biochemistry and Biophysics (IBB), Laboratory of Biophysics and Molecular Biology, University of Tehran, Tehran, Iran.
| |
Collapse
|
11
|
Luo Y, Yao M, Wang R, Liao S, Yu J. Netrin-1 binding to UNC5b improves post-stroke neuronal ferroptosis via AMPK-BACH1 pathway. Eur J Pharmacol 2025; 998:177507. [PMID: 40086580 DOI: 10.1016/j.ejphar.2025.177507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 03/08/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
Ferroptosis contributes to neuronal destruction after ischemic stroke which may be improved by inhibiting BTB domain and CNC homolog 1 (BACH1), a recently recognized ferroptosis facilitator. Axon guidance molecule netrin-1 (Ntn1) functions in neuroprotection against ischemic insult by engaging into its receptor of uncoordinated-5 homolog B (UNC5b) via adenosine 5'-monophosphate-activated protein kinase (AMPK), which potentially binds to BACH1. Whether Ntn1/UNC5b regulates post-stroke ferroptosis through AMPK-BACH1 pathway remains unclear. Ntn1 supplementation and UNC5b knockdown by siRNA were performed in photo-thrombosis stroke mice and oxygen-glucose deprivation-treated HT22 neurons. AMPK inhibitor BAY3827 and BACH1 activator Leptomycin B (LMB) were administrated. Ferroptosis was determined by ferroptosis-associated proteins (FSP1, GPX4 and ACSL4), Fe2+, malondialdehyde and mitochondrial morphology. BACH1 and p-AMPK/AMPK as well as the interaction between them were examined by Western blot and co-immunoprecipitation. Neuronal ferroptosis and the protein levels of BACH1 and p-AMPK were increased after photo-thrombosis and oxygen-glucose deprivation. Ntn1 supplementation or UNC5b knockdown relieved neuronal ferroptosis and neurological impairment with downregulated BACH1 and upregulated p-AMPK, nonetheless, UNC5b knockdown prevented the beneficial role of Ntn1. Both BAY3827 and LMB could reverse the change of ferroptosis caused by Ntn1 where BAY3827 inhibited the effects of Ntn1 to p-AMPK and BACH1 while LMB only inhibited the effect of Ntn1 to BACH1 without p-AMPK, suggesting BACH1 was regulated by AMPK. Co-immunoprecipitation verified that AMPK could physically bind to BACH1. Our results demonstrate UNC5b-evoked neuronal ferroptosis post stroke, and favor that Ntn1 improves post-stroke ferroptosis by its interaction with UNC5b via the AMPK-BACH1 pathway.
Collapse
Affiliation(s)
- Ying Luo
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China; Department of Neurology, The Affiliated Hospital of Southwest Medical University, Laboratory of Neurological Diseases and Brain Function, Luzhou, 646000, China
| | - Meiling Yao
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Rui Wang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Songjie Liao
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China.
| | - Jian Yu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China.
| |
Collapse
|
12
|
Mosalam EM, AboShabaan HS, Mahfouz MM, Sallam AS, Elhosary E, Allam A, Metwally EM, Shaldam MA, Ghoneim MES. Protective effect of empagliflozin against paracetamol-induced acute kidney injury through modulation of AMPK/SIRT1/PGC-1α pathway in experimental mice. Toxicol Appl Pharmacol 2025; 500:117382. [PMID: 40349789 DOI: 10.1016/j.taap.2025.117382] [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: 11/27/2024] [Revised: 05/05/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Emerging evidences about paracetamol-induced kidney injury in clinical settings are concerning, especially when administered at high doses. Empagliflozin, an oral SGLT2 inhibitor, employed in the management of diabetes mellitus, exhibits antioxidant, anti-inflammatory, and anti-apoptotic attributes. Thus, the objective of this study is to investigate whether empagliflozin may alleviate paracetamol-triggered nephrotoxicity and unravel the mechanistic insights responsible for its protective impact. In this regard, male mice were assigned to four groups: normal, paracetamol, empagliflozin 10, and empagliflozin 20. Kidney function tests, histopathological examination, immunohistochemistry, oxidative stress biomarkers, inflammatory cytokines, and other molecular targets were detected. Our results showed that paracetamol administration impaired kidney functions along with causing aberrations in renal histoarchitecture. Additionally, paracetamol triggered oxidative stress, inflammation, and apoptosis via hindering the AMPK/SIRT1/PGC-1α cascade and Nrf2/HO-1 while activating the NF-κB hub. Nevertheless, pretreatment with empagliflozin markedly enhanced the kidney function tests and mitigated histopathological alterations caused by paracetamol. Additionally, empagliflozin suppressed the oxidative stress as confirmed by an upregulation of Nrf2, which subsequently increased HO-1, SOD, and GSH, while reducing the MDA level. Moreover, it inhibited the NF-κB-mediated inflammatory process by dampening NF-κB, IL-1β, and TNF-α expressions as well as lowering Bax expression-induced apoptosis. The observed safeguards effects were facilitated via boosting AMPK/SIRT1/PGC-1α signaling trajectory. Collectively, our study verified the enduring reno-protective potential of empagliflozin, particularly at high dose, in the context of paracetamol-induced renal injury by instigating the AMPK/SIRT1/PGC-1α hinge.
Collapse
Affiliation(s)
- Esraa M Mosalam
- Biochemistry Department, Faculty of Pharmacy, Menoufia University, 32511 Shebin EL-Kom, Menoufia, Egypt; Department of Pharm D, Faculty of Pharmacy, Jadara University, Irbid 21110, Jordan.
| | - Hind S AboShabaan
- Clinical Pathology Department, National Liver Institute Hospital, Menoufia University, Shebin El-Kom, Menoufia, Egypt.
| | - Marwa M Mahfouz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Menoufia University, 32511 Shebin El-Kom, Menoufia, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Menoufia National University, Birket El-Sab, Menoufia, Egypt
| | - Amany Said Sallam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Menoufia University, 32511 Shebin El-Kom, Menoufia, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Menoufia National University, Birket El-Sab, Menoufia, Egypt.
| | - Enas Elhosary
- Department of Pathology, Faculty of Medicine, Helwan University, Cairo, Egypt.
| | - Albatoul Allam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), AL-Azhar University, Cairo, Egypt
| | - Ebtehal M Metwally
- Medical Physiology Department, Faculty of Medicine, Menoufia University, 32511 Shebin El-Kom, Menofia, Egypt.
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt; Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy.
| | - Mai El-Sayed Ghoneim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Sadat City (USC), 32897 Sadat City, Egypt.
| |
Collapse
|
13
|
Cao S, Wang X, Liu Q, Xu M, Wang Z, Jiang Y, Wang Y, Zhou Z. BDE-47 induces apoptosis and ferroptosis in Pseudosciaena crocea kidney (PCK) cells by disrupting mitochondrial energy metabolism. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2025; 284:107387. [PMID: 40347739 DOI: 10.1016/j.aquatox.2025.107387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 04/17/2025] [Accepted: 04/26/2025] [Indexed: 05/14/2025]
Abstract
2,2',4,4'-Tetrabromodiphenyl ether (BDE-47), an emerging contaminant (EC), is widely used in the production of brominated flame retardants and is biotoxic to marine organisms. However, our understanding of the mechanism of polybrominated diphenyl ethers (PBDEs)-induced toxicity remains incomplete. In this study, BDE-47 cytotoxicity after short-term exposure was investigated in PCK cells. BDE-47 significantly decreased cell viability, and morphological alterations were observed. Moreover, BDE-47 exposure induced apoptosis and ferroptosis, a newly described form of iron-mediated cell death, as demonstrated by transcriptomic analysis and physiological/biochemical tests. The observed cell death was associated with mitochondrial damage and a decrease in ATP production. Pharmacological intervention of cytotoxicity via 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), an activator of the adenosine monophosphate-activated protein kinase (AMPK) protein, a regulator of energy, strongly confirmed the causal relationship between cell death and energy metabolism dysfunction. Furthermore, lipidomic analysis revealed lipid metabolism disorders resulting from the accumulation of triglycerides (TGs) and glycerophospholipids (GPs) and the suppression of β-oxidation, ultimately inhibiting ATP synthesis. Molecular docking analysis revealed the binding potential of BDE-47 with energy metabolism checkpoints AMPK and Carnitine Palmitoyltransferase 1 (CPT1). Thus, our study broadens the understanding of the toxicity of BDE-47 and provides a new potential cellular and molecular mechanism.
Collapse
Affiliation(s)
- Sai Cao
- Department of Marine Ecology, College of Marine Life Science, Ocean University of China, Qingdao, 266003, China.
| | - Xiaocheng Wang
- National Marine Environmental Monitoring Center, Dalian, 116023, China.
| | - Qian Liu
- Qingdao Research academy of Environmental Sciences, Qingdao, 266003, China
| | - Mengxue Xu
- Marine Science Research Institute of Shandong Province, Qingdao 266100, China; Qingdao Key Laboratory of Coastal Ecological Restoration and Security, Qingdao 266100, China
| | - Zhaoning Wang
- Department of Marine Ecology, College of Marine Life Science, Ocean University of China, Qingdao, 266003, China
| | - Yongshun Jiang
- College of Marine Science and Technology, Qingdao Agricultural University, Qingdao 266237, China.
| | - You Wang
- Department of Marine Ecology, College of Marine Life Science, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China.
| | - Zhongyuan Zhou
- Department of Marine Ecology, College of Marine Life Science, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
14
|
Qiao Z, Feng X, Sun W, Wang F, Lu C. Independent and synergistic effects of extreme heat and NO 2 pollution on diabetic nephropathy in a type II diabetes mouse model. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 375:126321. [PMID: 40294690 DOI: 10.1016/j.envpol.2025.126321] [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: 11/10/2024] [Revised: 04/13/2025] [Accepted: 04/26/2025] [Indexed: 04/30/2025]
Abstract
Extreme heat and traffic-related air pollution (TRAP) have been linked to worsening chronic health disorders, however, their combined effects on diabetic nephropathy (DN) are little understood. Type II diabetic mice were exposed to heat (40 °C) and NO2 (5 ppm) separately for 4 h per day over 6 weeks to investigate the synergistic effects on the progression of DN. We found that exposure to high temperature and NO2 elevated blood glucose levels and exacerbated histopathological changes. Additionally, there were increased oxidation indicators (ROS, MDA, 8-OHdG) and decreased antioxidant indicators (CAT, SOD, GSH-PX), along with elevated inflammation markers (TNF-α, IL-1β, IL-6). The expressions of transient receptor potential (TRP) ion channels (TRPV1, TRPV4, TRPA1, TRPM2) were also upregulated. Our findings suggest that simultaneous exposure to high temperature and NO2 impairs metabolic and autophagy pathways. Exposure to both high temperature and NO2 produces a synergistic effect, leading to more severe damage than exposure to either factor individually. This resulted in increased expression of APOA1, P62, and p-mTOR/mTOR while decreasing the expression of p-AMPKα/AMPKα and LC3-II/I. This disruption promoted the progression of DN. In contrast, capsazepine (CZP) reduced TRP expression, inflammatory markers, oxidative stress, metabolic and autophagy disorders, thereby mitigating renal damage and alleviating the progression of diabetic nephropathy. Our study provides some potential strategies for early prevention and effective reduction of DN.
Collapse
Affiliation(s)
- Zipeng Qiao
- XiangYa School of Public Health, Central South University, Changsha, 410013, China
| | - Xiangling Feng
- XiangYa School of Public Health, Central South University, Changsha, 410013, China
| | - Wenying Sun
- XiangYa School of Public Health, Central South University, Changsha, 410013, China
| | - Faming Wang
- Centre for Molecular Biosciences and Non-communicable Diseases, Xi'an University of Science and Technology, Xi'an, 710054, China
| | - Chan Lu
- XiangYa School of Public Health, Central South University, Changsha, 410013, China; FuRong Laboratory, Changsha, 410078, Hunan, China; Hunan Provincial Key Laboratory of Low Carbon Healthy Building, Central South University, Changsha, 410083, China.
| |
Collapse
|
15
|
Wang X, Li H, Li J, Lei L, Xu J, Sun H, Li J, Jiang J, Li H, Tang M, Dong B, Gong Y, Jiang J, Peng Z. Berberine dissociates mitochondrial complex I by SIRT3-dependent deacetylation of NDUFS1 to improve hepatocellular glucose and lipid metabolism. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2834-8. [PMID: 40493314 DOI: 10.1007/s11427-024-2834-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 01/07/2025] [Indexed: 06/12/2025]
Abstract
Many metabolic diseases show mitochondrial abnormalities because of dysfunction of complex I (CI). Therefore, the discovery of drugs that target the CI is of great interest. Berberine (BBR) is a botanic agent and has been included in the latest ESC/EAS Guidelines for the management of dyslipidemias. Here, we showed that BBR enters hepatocyte mitochondria after oral administration and improves glucose and lipid metabolism by reducing oxidative phosphorylation in hepatocytes. BBR inhibits CI function rapidly, selectively, and reversibly, not by directly inhibiting CI enzyme activity but by reducing the abundance of CI in the mitochondria through dissociation of CI. BBR directly binds to and activates Sirtuin 3 (SIRT3), thereby reducing acetylation of the catalytic subunit NDUFS1 in the N-module of CI, leading to dissociation of mitochondrial CI. Conclusively, BBR, as a mitochondria-homing agent, selectively and reversibly dissociates mitochondrial CI through SIRT3-dependent NDUFS1 deacetylation to improve hepatocellular glucose and lipid metabolism, highlighting that CI may be a promising target for innovative natural products to treat metabolic diseases.
Collapse
Affiliation(s)
- Xuekai Wang
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Hu Li
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Key Laboratory of Biotechnology of Antibiotics, The National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jianrui Li
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Lei Lei
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jingchen Xu
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Han Sun
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jiayu Li
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jing Jiang
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Hongying Li
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Mei Tang
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Biao Dong
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Key Laboratory of Biotechnology of Antibiotics, The National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yue Gong
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jiandong Jiang
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
- Key Laboratory of Biotechnology of Antibiotics, The National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Zonggen Peng
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
- Key Laboratory of Biotechnology of Antibiotics, The National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
16
|
Shao Q, Ndzie Noah ML, Golubnitschaja O, Zhan X. Mitochondrial medicine: "from bench to bedside" 3PM-guided concept. EPMA J 2025; 16:239-264. [PMID: 40438494 PMCID: PMC12106218 DOI: 10.1007/s13167-025-00409-4] [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: 02/24/2025] [Accepted: 03/27/2025] [Indexed: 06/01/2025]
Abstract
Mitochondria are the primary sites for aerobic respiration and play a vital role in maintaining physiologic function at the cellular and organismal levels. Physiologic mitochondrial homeostasis, functions, health, and any kind of mitochondrial impairments are associated with systemic effects that are linked to the human health and pathologies. Contextually, mitochondria are acting as a natural vital biosensor in humans controlling status of physical and mental health in a holistic manner. So far, no any disorder is known as happening to humans independently from a compromised mitochondrial health as the cause (primary mitochondrial dysfunction) or a target of collateral damage (secondary mitochondrial injury). This certainty makes mitochondrial medicine be the superior instrument to reach highly ambitious objectives of predictive, preventive, and personalized medicine (PPPM/3PM). 3PM effectively implements the paradigm change from the economically ineffective reactive medical services to a predictive approach, targeted prevention and treatments tailored to individualized patient profiles in primary (protection against health-to-disease transition) and secondary (protection against disease progression) healthcare. Mitochondrial DNA (mtDNA) properties differ significantly from those of nuclear DNA (nDNA). For example, mtDNA as the cell-free DNA molecule is much more stable compared to nDNA, which makes mtDNA be an attractive diagnostic target circulating in human body fluids such as blood and tear fluid. Further, genetic variations in mtDNA contribute to substantial individual differences in disease susceptibility and treatment response. To this end, the current gene editing technologies, such as clustered regularly interspaced short palindromic repeats (CRISPR)/Cas, are still immature in mtDNA modification, and cannot be effectively applied in clinical practice posing a challenge for mtDNA-based therapies. In contrast, comprehensive multiomics technologies offer new insights into mitochondrial homeostasis, health, and functions, which enables to develop more effective multi-level diagnostics and targeted treatment strategies. This review article highlights health- and disease-relevant mitochondrial particularities and assesses involvement of mitochondrial medicine into implementing the 3PM objectives. By discussing the interrelationship between 3PM and mitochondrial medicine, we aim to provide a foundation for advancing early and predictive diagnostics, cost-effective targeted prevention in primary and secondary care, and exemplify personalized treatments creating proof-of-concept approaches for 3PM-guided clinical applications.
Collapse
Affiliation(s)
- Qianwen Shao
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Marie Louise Ndzie Noah
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Olga Golubnitschaja
- Predictive, Preventive and Personalised (3P) Medicine, University Hospital Bonn, Rheinische Friedrich-Wilhelms-University of Bonn, Venusberg Campus 1, 53127 Bonn, Germany
| | - Xianquan Zhan
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
- Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Jinan Key Laboratory of Cancer Multiomics, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| |
Collapse
|
17
|
Ma Z, Pan S, Yang Y, Ren H, Yin S, Chen Q, An Z, Zhao X, Xu Z. Lipid droplets: Emerging therapeutic targets for age-related metabolic diseases. Ageing Res Rev 2025; 108:102758. [PMID: 40300696 DOI: 10.1016/j.arr.2025.102758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/01/2025]
Abstract
Lipids metabolism is crucial in regulating aging and metabolic diseases. Lipid droplets (LDs) are dynamic, complex organelles responsible for the storage and release of neutral lipids, essential for maintaining lipid homeostasis and energy metabolism. Aging accelerates the accumulation of LDs, functional deterioration, and metabolic disorders, thereby inducing age-related metabolic diseases (ARMDs). This review examines published datasets on the association between LDs and ARMDs, focusing on the structure and function of LDs, their interactions with other organelles, and associated proteins. Furthermore, we explore the potential mechanisms by which LDs mediate the onset of ARMDs, including Alzheimer's disease (AD), sarcopenia, metabolic cardiomyopathy, non-alcoholic fatty liver disease (NAFLD), and cancer. Lastly, we discuss intervention strategies aimed at targeting LDs to improve outcomes in ARMDs, including exercise, dietary, and pharmacological interventions.
Collapse
Affiliation(s)
- Zheying Ma
- School of Physical Education and Health Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Shou Pan
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an 710119, China
| | - Yaming Yang
- School of Physical Education and Health Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Huiqian Ren
- School of Physical Education and Health Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Sikun Yin
- School of Physical Education and Health Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Qianyu Chen
- School of Physical Education and Health Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Zhenxian An
- School of Physical Education and Health Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Xiaoqin Zhao
- School of Physical Education and Health Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China.
| | - Zujie Xu
- School of Physical Education and Health Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China.
| |
Collapse
|
18
|
Zhang X, Sun J, Zhu X, Yang Z, Zhu Z, Zhou M, Li C, Yu H, Gan X. Low-magnitude high-frequency vibration ameliorates high glucose-induced endothelial injury by restoring mitochondrial function via AMPK/mTOR pathway. J Histotechnol 2025; 48:82-92. [PMID: 39564647 DOI: 10.1080/01478885.2024.2429855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 11/10/2024] [Indexed: 11/21/2024]
Abstract
High glucose-induced dysfunction of endothelial cells is a critical and initiating factor in the genesis of diabetic vascular complications. Low-magnitude high-frequency vibration (LMHFV) is a non-invasive biophysical intervention. It has been reported that it exhibits protective effects on high glucose-induced osteoblast dysfunction, but little was known on diabetic vascular complications. In this work, we aim to clarify the role of LMHFV on high glucose-induced endothelial dysfunction and hypothesized that the protective effects functioned through adenosine monophosphate-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway. We cultured primary murine aortic endothelial cells (MAECs) in normal or HG medium, respectively, before exposing to LMHFV. The tube formation, paracellular permeability assay, and aortic ring sprouting assay showed that the high glucose injured-function of MAECs was improved after LMHFV treatment. The intracellular ROS generation analysis, mitochondrial complex I activities measurement, ATP measurement and mitochondrial membrane potential (MMP), and mitochondrial ROS generation analysis of MAECs indicated that mitochondrial function was restored by LMHFV loading in a high glucose environment. Mechanically, western blot assays showed that AMPK phosphorylation was promoted and mTOR was inhibited in LMHFV-induced endothelial function restoration. After the administration of the AMPK inhibitor, Compound C, these protective effects resulting from LMHFV are reversed. These findings suggest that LMHFV plays a significant role in protecting endothelial cells' function and mitochondrial function in high glucose-induced injured MAECs via AMPK/mTOR signalling.
Collapse
Affiliation(s)
- Xidan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jiyu Sun
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xiting Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zhenghao Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zhuoli Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Min Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Chen Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Haiyang Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xueqi Gan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
19
|
Ou W, Li X, Tang K, Ding L, Sun T, Li Q, Li T. GLA deficiency causes cardiac hypertrophy via enhanced autophagy. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1689-1702. [PMID: 39969746 DOI: 10.1007/s11427-023-2731-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 09/15/2024] [Indexed: 02/20/2025]
Abstract
Fabry disease is a monogenic disease characterized by a deficiency or loss of α-galactosidase A (GLA). Cardiomyopathy is the leading cause of death in Fabry patients; however, a lack of understanding of the pathological mechanism impedes the development of effective therapies. Here, we used a Gla knockout (KO) mouse model and investigated its impact on cardiomyopathy. We found that globotriaosylceramide (Gb3) increased the uptake and accumulation of fatty acids in KO hearts by increasing the expression levels of CD36 and ACC2. The augmented fatty acid metabolism further increased autophagy activity, leading to age-related late-onset cardiac hypertrophy. Additionally, increased autophagy facilitates disturbances in fatty acid metabolism. The inhibition of autophagy by supplementation with 3-methyladenine (3-MA) or the overexpression of GLA by the cardiomyocyte-specific adeno-associated virus for 2 months could rebalance abnormal fatty acid metabolism and ameliorate cardiac hypertrophy and dysfunction in KO hearts, suggesting a central role of autophagy in GLA deficiency-related cardiomyopathy.
Collapse
Affiliation(s)
- Wei Ou
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China
- Laboratory of Mitochondrial Metabolism and Perioperative Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Xi Li
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China
- Laboratory of Mitochondrial Metabolism and Perioperative Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Kuo Tang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China
- Laboratory of Mitochondrial Metabolism and Perioperative Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Lin Ding
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China
- Laboratory of Mitochondrial Metabolism and Perioperative Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Tingting Sun
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China
- Laboratory of Mitochondrial Metabolism and Perioperative Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Qian Li
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China.
- Laboratory of Mitochondrial Metabolism and Perioperative Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China.
| | - Tao Li
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China.
- Laboratory of Mitochondrial Metabolism and Perioperative Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
20
|
Yao J, Song S, Liu T, Wang J, Li C, Liu J, Yuan Y, Zhao H. Isoguanosine-Induced ER Stress via AMPK Enhances Chemosensitivity in OSCC. J Dent Res 2025; 104:668-678. [PMID: 40071313 DOI: 10.1177/00220345241303168] [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: 03/17/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common malignancy of the head and neck; however, the efficacy of existing treatment is limited and new effective strategies need to be explored. Our previous work demonstrates that isoguanosine (isoG) is a promising nucleoside molecule with superior self-assembly capability and significant anti-OSCC potential. However, the antitumor mechanism of isoG remains unclear. In this study, we reveal that the antiproliferative effect of isoG is mediated by its cellular metabolite, isoguanosine 5'-monophosphate (isoGMP), which induces excessive endoplasmic reticulum (ER) stress and cell death through adenosine monophosphate-activated protein kinase (AMPK) activation. IsoG activates AMPK and induces ER stress at low concentrations, with minimal impact on cell viability at these concentrations. To further explore the therapeutic potential of isoG, we investigated its role in modulating chemosensitivity. Our findings show that AMPK activation enhances the sensitivity of OSCC cells to 5-fluorouracil (5-FU), and the combination of isoG and 5-FU exhibits a synergistic anticancer effect. Building on the self-assembly characteristics of isoG, we developed an innovative treatment platform by introducing dynamic borate ester bonds to form an isoguanosine-phenylenediboronic acid-isoguanosine (isoGPBisoG) structure. When combined with 5-FU, this platform achieved remarkable therapeutic efficacy in 2 OSCC cell-derived xenograft models, with tumor inhibition rates of 71.0% and 56.6%, respectively, compared with control. These findings establish isoG as a potent enhancer of chemotherapeutic efficacy in OSCC via AMPK activation. More importantly, the isoGPBisoG and 5-FU combination represents a significant paradigm of a synergistic therapy platform. This novel approach offers a promising direction for the development of more effective OSCC treatments.
Collapse
Affiliation(s)
- J Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - S Song
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - T Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - J Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - C Li
- Key Laboratory of Green Chemistry & Technology of Ministry of Education, College of Chemistry, Sichuan University, Chengdu, People's Republic of China
| | - J Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Y Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - H Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
21
|
Luo Z, Zheng S, Hu Z, Li P, Zeng J, Lu Y, Ali M, Chen Z, Wang Q, Qi F. Ultrasound-responsive taurine lipid nanoparticles attenuate oxidative stress and promote macrophage polarization for diabetic wound healing. Free Radic Biol Med 2025; 233:302-316. [PMID: 40187503 DOI: 10.1016/j.freeradbiomed.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/22/2025] [Accepted: 04/03/2025] [Indexed: 04/07/2025]
Abstract
Diabetic wound healing presents a significant clinical challenge due to disrupted neuro-immune interactions. This study identifies the α7 nicotinic acetylcholine receptor (α7nAChR) as a key regulator of wound repair by linking cholinergic signaling to macrophage reprogramming. GEO analysis of diabetic foot ulcer (DFU) microenvironments revealed neuronal loss, M1 macrophage dominance, and chronic inflammation, all driven by impaired acetylcholine (ACh) secretion and α7nAChR inactivation. Mechanistically, taurine (TA) restored PC12 cell function under high glucose conditions by activating AMPK, alleviating oxidative and endoplasmic reticulum stress, and promoting ACh production. ACh activated macrophage α7nAChR, modulating M1/M2 polarization through JAK2/STAT3 activation and NF-κB suppression. To enhance TA bioavailability, ultrasound-responsive Ccr2-targeted TA nanoparticles (Ccr2@TA@LNP) were developed for site-specific delivery via Ccl2/Ccr2 chemotaxis. In diabetic neuropathy (DPN) mice, Ccr2@TA@LNP accelerated wound healing by increasing ACh levels, enhancing α7nAChR/CD206 expression, and reducing Ccl2-mediated inflammation. By integrating neuroprotection, macrophage reprogramming, and targeted nanotherapy, this study highlights TA as a multi-target agent that restores neuro-immune balance through the AMPK/α7nAChR/JAK2-STAT3 axis, offering a novel therapeutic strategy for diabetic wound treatment.
Collapse
Affiliation(s)
- Zucheng Luo
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shaoluan Zheng
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Zhichao Hu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pengfei Li
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junhao Zeng
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yao Lu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mohyeddin Ali
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zijian Chen
- Department of Plastic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Qi Wang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fazhi Qi
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
22
|
Zhang Y, Shen M, Zhang B, Li X, Cheng H, Feng D, Han Y, Luo Z, Zhou Y. A Novel Role of Adipokine 'Intelectin-1': Ameliorating Renal Fibrosis Through Inhibition of Renal Tubular Epithelial Cell Senescence. FASEB J 2025; 39:e70653. [PMID: 40387543 DOI: 10.1096/fj.202403361r] [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: 12/24/2024] [Revised: 04/17/2025] [Accepted: 05/08/2025] [Indexed: 05/20/2025]
Abstract
Renal fibrosis is a common pathological process associated with chronic kidney disease (CKD) progression. Intelectin-1, a newly identified adipokine, has been demonstrated to protect renal function in mice with type 2 diabetic nephropathy. However, the role of intelectin-1 in renal fibrosis and the underlying mechanisms remain unclear. This study aimed to: (1) investigate the effects of intelectin-1 on renal fibrosis in mice, and (2) explore the potential involvement of intelectin-1 in regulating renal tubular epithelial cells (TECs) senescence and mitochondrial dysfunction. To our knowledge, these findings represent the first demonstration that intelectin-1 treatment significantly attenuates renal fibrosis in unilateral ureteral obstruction (UUO) in mice by effectively inhibiting TECs senescence. Furthermore, intelectin-1 treatment alleviated mitochondrial dysfunction in TECs, as evidenced by improved mitochondrial membrane potential and decreased mitochondrial reactive oxygen species (mtROS) production. Mechanistically, intelectin-1 treatment activated AMPK signaling that subsequently inhibited the mTOR and p38 pathways. In conclusion, our findings suggest that intelectin-1 attenuates renal fibrosis in mice by inhibiting TECs senescence and alleviating mitochondrial dysfunction via AMPK/mTOR/p38MAPK signaling. These results provide a potential therapeutic target for the treatment of renal fibrosis in CKD. Further studies are warranted to explore the clinical relevance and translational potential of adipokines, including intelectin-1, in human renal fibrosis.
Collapse
Affiliation(s)
- Yunna Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Mengxia Shen
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Bo Zhang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaohong Li
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Haipeng Cheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Dandan Feng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Yang Han
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Ziqiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Yan Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
23
|
Wu X, Guan X, Cheng C, Deng Z, Li Z, Ma Y, Xie Y, Zheng Q. Activation of the MEK1-CHK2 axis in macrophages by Staphylococcus aureus promotes mitophagy, resulting in a reduction in bactericidal efficacy. Mol Med 2025; 31:211. [PMID: 40437411 PMCID: PMC12121099 DOI: 10.1186/s10020-025-01274-7] [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: 04/02/2025] [Accepted: 05/20/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND Macrophages, which serve as the frontline defenders against microbial invasion, paradoxically become accomplices in Staphylococcus aureus (S. aureus)-driven osteomyelitis pathogenesis through poorly defined immunosuppressive mechanisms. METHODS In this study, we established an S. aureus implant-associated femoral infection model treated with MEK1 inhibitors and evaluated the degree of bone destruction and the bacterial load. We subsequently investigated changes in mitochondrial ROS (mtROS) levels, mitophagy activity, phagocytic-killing ability, and CHEK2 mitochondrial translocation in S. aureus-activated bone marrow-derived macrophages (BMDMs) following MEK1 inhibitor treatment. Finally, in vivo experiments involving different inhibitor combinations were conducted to assess mitophagy levels and the therapeutic potential for treating osteomyelitis. RESULTS Pharmacological inhibition of MEK1 significantly attenuated bone degradation and the pathogen burden in murine models of osteomyelitis, indicating its therapeutic potential. Investigations using BMDMs revealed that blockade of the MEK1-ERK1/2 axis increases mtROS levels by suppressing mitophagy, directly linking metabolic reprogramming to increased bactericidal activity. Mechanistically, inactivation of the MEK1-ERK1/2 pathway restores CHEK2 expression, facilitating its translocation from the nucleus to mitochondria to restore mtROS levels by inhibiting mitophagy. Importantly, in vivo studies confirmed that the MEK1-ERK1/2-CHEK2 axis is pivotal for controlling mitophagy-dependent bone pathology and bacterial persistence during S. aureus infection. CONCLUSIONS We identified a self-amplifying pathogenic loop in which S. aureus exploits macrophage MEK1 to hyperactivate ERK1/2, leading to the suppression of CHEK2 expression. This process results in excessive mitophagy and decreased mtROS levels, which impair the bactericidal function and enable uncontrolled osteolytic destruction. These findings redefine MEK1 as a metabolic-immune checkpoint and highlight its druggable vulnerability in osteomyelitis.
Collapse
Affiliation(s)
- Xiaohu Wu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Xin Guan
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chubin Cheng
- The Second People's Hospital of Shenzhen City (the First Affiliated Hospital of Shenzhen University), Shenzhen, 518000, China
| | - Zhantao Deng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Zeng Li
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yuanchen Ma
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yanjie Xie
- Department of Health Management Center, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| |
Collapse
|
24
|
Renziehausen T, Dirr A, Schmidt-Schippers R, Flashman E, Schippers J. Oxygen sensing and plant adaptation to flooding in a changing climate. Philos Trans R Soc Lond B Biol Sci 2025; 380:20240238. [PMID: 40439298 PMCID: PMC12121377 DOI: 10.1098/rstb.2024.0238] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 06/02/2025] Open
Abstract
In times of climate change, frequency and intensity of extreme weather events are rising and thus demand a higher resilience of crop plants to environmental influences, such as flooding events, to minimize yield losses. Flooding causes acute oxygen deprivation in plants, accompanied by an energy crisis, starvation, growth retardation and ultimately increased harvest losses. At a molecular level, fluctuating oxygen concentrations are sensed via plant cysteine oxidases (PCOs) that, as part of the N-degron pathway, oxygen-dependently oxidize substrates such as vernalization 2, little zipper 2 and, most prominently, transcription factors of the group VII ETHYLENE RESPONSE FACTORS (ERFVIIs) to render them for degradation. When stabilized under hypoxia owing to the lack of oxygen, ERFVIIs act as transcriptional activators of hypoxia-response genes to evoke appropriate acclimation. Crop engineering for improved submergence resilience is an important goal for future food security, and prolonging ERFVII stability is a promising strategy. Here we discuss the potential molecular consequences of this strategy in terms of stabilization of Cys-initiating proteins in commercially relevant crop species, as well as ways in which this may be achieved, including via PCO engineering.This article is part of the theme issue 'Crops under stress: can we mitigate the impacts of climate change on agriculture and launch the 'Resilience Revolution'?'.
Collapse
Affiliation(s)
| | - Anna Dirr
- University of Oxford, OxfordOX1 3TA, UK
| | | | | | - Jos Schippers
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Seeland, Sachsen-Anhalt06466, Germany
| |
Collapse
|
25
|
Gamal W, Goedhart NB, Simon-Molas H, Mediavilla-Varela M, Uriepero-Palma A, Peters FS, Maharaj K, Chavez JC, Powers J, Obermayer A, Shaw TI, Conejo-Garcia JR, Rodriguez PC, Sahakian E, Pinilla-Ibarz J, Kater AP. Mitigating T-cell mitochondrial dysfunction in CLL to augment CAR T-cell therapy: evaluation in an immunocompetent model. Blood Adv 2025; 9:2511-2529. [PMID: 39938006 DOI: 10.1182/bloodadvances.2024014822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/09/2025] [Accepted: 01/28/2025] [Indexed: 02/14/2025] Open
Abstract
ABSTRACT An unmet clinical need in chronic lymphocytic leukemia (CLL) is emerging due to the rapidly expanding group of patients with double refractory (Bruton's tyrosine kinase- and B-cell lymphoma 2-inhibitor) disease. So far, autologous T-cell-based therapies, including chimeric antigen receptor (CAR) T cells, have limited success in CLL, which has been attributed to an acquired CLL-mediated T-cell dysfunction and subset skewing toward effector cells at the expense of memory formation. T-cell responses rely on dynamic metabolic processes, particularly mitochondrial fitness. Although mitochondrial disruptions have been observed in solid tumor-infiltrating lymphocytes, their impact on T-cell immunity in lymphoproliferative disorders is unknown. Recent findings indicate that mitochondrial mass in CAR T cells correlates with CLL clinical outcomes. This prompted our investigation into the mitochondrial fitness in CLL T cells. Integrated metabolic and functional analyses revealed impaired, depolarized mitochondria across all T-cell subsets in untreated patients with CLL, leading to further ex vivo and in vivo mouse studies on the underlying signaling alterations. Multiomics profiling of transcriptome and epigenome revealed significant alterations in mitochondrial signaling, diminished adenosine monophosphate-activated protein kinase and autophagy activity, and upregulated glycolysis coupled with hyperactivation of Akt. Inhibition of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway during CLL T-cell culture induced metabolic reprogramming, enhancing mitochondrial activity, expression of peroxisome proliferator-activated receptor-gamma coactivator 1-alpha, and memory differentiation. Underscoring clinical relevance, supplementation with the PI3Kδ inhibitor idelalisib during CAR T-cell manufacturing improved persistence and long-term leukemia-free remissions in an immunocompetent murine model. Our study suggests that modulating the abnormal CLL T-cell metabolism can enhance the efficacy of autologous T-cell therapies.
Collapse
MESH Headings
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Mitochondria/metabolism
- Mitochondria/pathology
- Humans
- Animals
- Mice
- Immunotherapy, Adoptive/methods
- T-Lymphocytes/metabolism
- T-Lymphocytes/immunology
- Disease Models, Animal
- Receptors, Chimeric Antigen/metabolism
Collapse
Affiliation(s)
- Wael Gamal
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Nienke B Goedhart
- Departments of Hematology and Experimental Immunology and Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, The Netherlands
| | - Helga Simon-Molas
- Departments of Hematology and Experimental Immunology and Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, The Netherlands
| | | | | | - Fleur S Peters
- Departments of Hematology and Experimental Immunology and Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, The Netherlands
| | - Kamira Maharaj
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Julio C Chavez
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - John Powers
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Alyssa Obermayer
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Timothy I Shaw
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | | | - Paulo C Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Eva Sahakian
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Javier Pinilla-Ibarz
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Arnon P Kater
- Departments of Hematology and Experimental Immunology and Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, The Netherlands
| |
Collapse
|
26
|
Zhang Y, Ma P, Wang S, Chen S, Deng H. Restoring calcium crosstalk between ER and mitochondria promotes intestinal stem cell rejuvenation through autophagy in aged Drosophila. Nat Commun 2025; 16:4909. [PMID: 40425608 PMCID: PMC12116733 DOI: 10.1038/s41467-025-60196-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
Breakdown of calcium network is closely associated with cellular aging. Previously, we found that cytosolic calcium (CytoCa2+) levels were elevated while mitochondrial calcium (MitoCa2+) levels were decreased and associated with metabolic shift in aged intestinal stem cells (ISCs) of Drosophila. How MitoCa2+ was decoupled from the intracellular calcium network and whether the reduction of MitoCa2+ drives ISC aging, however, remains unresolved. Here, we show that genetically restoring MitoCa2+ can reverse ISC functional decline and promote intestinal homeostasis by activating autophagy in aged flies. Further studies indicate that MitoCa2+ and Mitochondria-ER contacts (MERCs) form a positive feedback loop via IP3R to regulate autophagy independent of AMPK. Breakdown of this loop is responsible for MitoCa2+ reduction and ISC dysfunction in aged flies. Our results identify a regulatory module for autophagy initiation involving calcium crosstalk between the ER and mitochondria, providing a strategy to treat aging and age-related diseases.
Collapse
Affiliation(s)
- Yao Zhang
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 20092, China
| | - Peng Ma
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 20092, China
| | - Saifei Wang
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 20092, China
| | - Shuxin Chen
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 20092, China
| | - Hansong Deng
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 20092, China.
| |
Collapse
|
27
|
de Oliveira MR. Pre-clinical evidence for mitochondria as a therapeutic target for luteolin: A mechanistic view. Chem Biol Interact 2025; 413:111492. [PMID: 40154935 DOI: 10.1016/j.cbi.2025.111492] [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: 12/20/2024] [Revised: 03/05/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Pre-clinical evidence indicates that mitochondria may be a therapeutic target for luteolin (3',4',5,7-tetrahydroxyflavone; LUT) in different conditions. LUT modulates mitochondrial physiology in in vitro, ex vivo, and in vivo experimental models. This flavone exerted mitochondria-related antioxidant and anti-apoptotic effects, stimulated mitochondrial fusion and fission, induced mitophagy, and promoted mitochondrial biogenesis in human and animal cells and tissues. Moreover, LUT modulated the activity of components of the oxidative phosphorylation (OXPHOS) system, improving the ability of mitochondria to produce adenosine triphosphate (ATP) in certain circumstances. The mechanism of action by which LUT promoted mitochondrial benefits and protection are not completely clear yet. Nonetheless, LUT is a potential candidate to be utilized in mitochondrial therapy in the future. In this work, it is explored the mechanisms of action by which LUT modulates mitochondrial physiology in different pre-clinical experimental models.
Collapse
Affiliation(s)
- Marcos Roberto de Oliveira
- Grupo de Estudos em Neuroquímica e Neurobiologia de Moléculas Bioativas, Departamento de Química, Universidade Federal de Mato Grosso (UFMT), CEP 78060-900, Cuiaba, Mato Grosso, Brazil; Grupo de Estudos em Terapia Mitocondrial, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), CEP 90035-003, Porto Alegre, Rio Grande do Sul, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), CEP 90035-003, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
28
|
Xiong G, Yun F, Jiang L, Yi Z, Yi X, Yang L, Zhang X, Li X, Yang Z, Zhang Q, Sai B, Kuang Y, Zhu Y. NDUFS3 promotes proliferation via glucose metabolism reprogramming inducing AMPK phosphorylating PRPS1 to increase the purine nucleotide synthesis in melanoma. Cell Death Differ 2025:10.1038/s41418-025-01525-4. [PMID: 40404919 DOI: 10.1038/s41418-025-01525-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 04/02/2025] [Accepted: 05/06/2025] [Indexed: 05/24/2025] Open
Abstract
NADH dehydrogenase [ubiquinone] iron-sulfur protein 3 (NDUFS3) is the core subunit of the respiratory chain complex I (CI). We found NDUFS3 were abnormally elevated in human melanoma and promoted melanoma proliferation. Furthermore, NDUFS3 could promote the oxidative phosphorylation (OXPHOS) and the pentose phosphate pathway (PPP), as well as attenuated glycolysis. As NDUFS3-mediated the metabolic changes of OXPHOS and glucose metabolism, melanoma cells produced more ATP, resulting in the inhibition of AMP kinase (AMPK). AMPK induced phosphoribosyl pyrophosphate synthetase1 (PRPS1) phosphorylation, which resulted in suppressed PRPS1 activity. Briefly, the NDUFS3-AMPK-PRPS1 signaling axis coupled OXPHOS, glucose metabolism, and purine nucleotide biosynthesis to regulate melanoma proliferation. Our study highlighted an unrecognized role for NDUFS3 in melanoma, which might be used as a potential therapeutic target for the treatment of this type of cancer. NDUFS3 regulating PRPS1 activity through AMPK to affect melanoma proliferation.
Collapse
Affiliation(s)
- Guohang Xiong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, 650500, China
- Research Center for Clinical Medicine, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, 650032, China
| | - Fang Yun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, 650500, China
| | - Lu Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, 650500, China
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, 210000, China
| | - Zihan Yi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, 650500, China
- Department of Medical Oncology, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, 650118, China
| | - Xiaojia Yi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, 650500, China
- Department of Pathology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 434000, China
| | - Lijuan Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, 650500, China
| | - Xuedan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, 650500, China
| | - Xiaoyu Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, 650500, China
| | - Zhe Yang
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Qiao Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, 650500, China
| | - Buqing Sai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, 650500, China
| | - Yingmin Kuang
- Department of Organ Transplantation, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| | - Yuechun Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, 650500, China.
| |
Collapse
|
29
|
Liu S, Ding R, Huang L, Lv J, Sun Z, Wang X, Duan J. Maternal exposure to urban particulate matter induces cardiac developmental toxicity in zebrafish offspring by disrupting mitochondrial homeostasis. J Adv Res 2025:S2090-1232(25)00359-5. [PMID: 40409463 DOI: 10.1016/j.jare.2025.05.041] [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: 01/11/2025] [Revised: 04/20/2025] [Accepted: 05/18/2025] [Indexed: 05/25/2025] Open
Abstract
INTRODUCTION Urban particulate matter (UPM) is a major air pollutant affecting public health, with maternal exposure potentially leading to cardiac developmental disorders in offspring. However, the exact mechanisms underlying the intergenerational effects of UPM remain unclear. OBJECTIVE This study aimed to investigate the molecular mechanisms involved in cardiac developmental defects caused by maternal UPM exposure in offspring zebrafish. METHODS AND RESULTS Female zebrafish were exposed to UPM for 21 days to examine intergenerational effects. The results indicated that maternal zebrafish in the exposed group exhibited ovarian damage and a reduced number of embryos and fertilization rates. Zebrafish offspring exhibited abnormal cardiac development, including pericardial edema and pathological heart injury. Mechanistically, transcriptomic analysis of the offspring indicated that UPM exposure induced significant modifications in the mitochondrial biogenesis pathway, with altered expression of mitochondrial function-related genes. Maternal UPM exposure impaired respiration in zebrafish embryos and increased angiopoietin-like 4 (ANGPTL4) expression in offspring hearts. In vitro, Angptl4 knockdown alleviated UPM-induced mitochondrial membrane potential reduction and mitochondrial reactive oxygen species overproduction in cardiomyocytes, whereas Angptl4 overexpression exacerbated UPM-induced mitochondrial toxicity. CONCLUSION These findings show that maternal UPM exposure disrupts mitochondrial homeostasis by upregulating ANGPTL4 expression, leading to abnormal cardiac development in zebrafish offspring.
Collapse
Affiliation(s)
- Shiqian Liu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing 100069, PR China
| | - Ruiyang Ding
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing 100069, PR China
| | - Linyuan Huang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing 100069, PR China
| | - Jianong Lv
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing 100069, PR China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing 100069, PR China
| | - Xiaoxiao Wang
- College of Life Sciences, Institute of Life Sciences and Green Development, Hebei University, Baoding 071002, PR China.
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
30
|
Ang B, Yang T, Jiang H, Cheng Y, Chen Y, Qie X, Yin L, Wang T, Chen Q, Wang Z, Zeng M, Adhikari B, He Z, Chen J. Enzymatic Synthesis and Evaluation of Eight Methylated Quercetin Products: In Vitro Chemical Properties and Adipogenesis Regulation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40393977 DOI: 10.1021/acs.jafc.5c00426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
The methoxylated modification of flavonoids has been reported to enhance stability and permeability; however, its effect on the improvement of activity is not clear. In this study, Citrus depressa flavonoid O-methyltransferase 5 and Sorghum vulgare 7-O-methyltransferase were recombinantly expressed and successfully converted quercetin (QUE) into eight methoxylated products, which were isolated and identified with a purity exceeding 95%. All products except rhamnetin (RHA) showed improved stability, while only 5,7,3',4'-EMQ, 7,3',4'-TMQ, and 3,7,3',4'-EMQ had higher uptake ratios. Compared to QUE, 5,7,3',4'-EMQ and RHA significantly reduced the intracellular triglyceride level, while 3,5,7,3',4'-PMQ, 3,3',4'-TMQ and 3,7,3',4'-EMQ increased it. 5,7,3',4'-EMQ and RHA also significantly downregulated both the mRNA and protein levels of peroxisome proliferator-activated receptor γ, while 3,5,7,3',4'-PMQ and 3,7,3',4'-EMQ upregulated PPARγ at the transcriptional level to about ten times higher than that of QUE. The structure-activity relationship analysis highlighted the importance of C3-OH retention and dual methoxylation of the A-ring. In summary, this study efficiently produced eight structurally well-defined QUE methoxylation products via biotransformation, established an in vitro initial structure-activity relationship for regulating adipogenesis, and provided a potential structure for PPARγ regulation, a central target of lipid metabolism.
Collapse
Affiliation(s)
- Beijun Ang
- State Key Laboratory of Food Science and Resource, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Tian Yang
- Analytical and Testing Center, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hongtao Jiang
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yong Cheng
- State Key Laboratory of Food Science and Resource, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yang Chen
- State Key Laboratory of Food Science and Resource, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xuejiao Qie
- MOE Key Laboratory of Population Health across Life Cycle/School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Liduan Yin
- Yantai New Era Health Industry Co., Ltd., Yantai, Shandong 264000, China
| | - Tong Wang
- Yantai New Era Health Industry Co., Ltd., Yantai, Shandong 264000, China
| | - Qiuming Chen
- State Key Laboratory of Food Science and Resource, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Zhaojun Wang
- State Key Laboratory of Food Science and Resource, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Maomao Zeng
- State Key Laboratory of Food Science and Resource, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Benu Adhikari
- School of Science, RMIT University, Melbourne, Victoria 3083, Australia
| | - Zhiyong He
- State Key Laboratory of Food Science and Resource, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jie Chen
- State Key Laboratory of Food Science and Resource, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
31
|
Zhang Y, Chen H, Feng Y, Liu M, Lu Z, Hu B, Chen L, Zhang Y, Liu J, Cai F, Zhao Y, Pan W, Liao X, Pan S, Bestard-Lorigados I, Wu Y, Song W. Activation of AMPK by GLP-1R agonists mitigates Alzheimer-related phenotypes in transgenic mice. NATURE AGING 2025:10.1038/s43587-025-00869-3. [PMID: 40394225 DOI: 10.1038/s43587-025-00869-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 04/07/2025] [Indexed: 05/22/2025]
Abstract
Individuals with type 2 diabetes mellitus have an increased risk of developing Alzheimer's disease (AD). GLP-1 receptor agonists (GLP-1RAs) are used for glycemic control in diabetes and show potential neuroprotective properties, but their effects on AD and the underlying mechanisms are not well understood. Here we demonstrate that GLP-1RAs can alleviate AD-related phenotypes by activating 5' AMP-activated protein kinase (AMPK) signaling. We found that plasma GLP-1 levels were decreased in AD model mice and negatively correlated with amyloid-beta (Aβ) load in patients with AD. Enhancing GLP-1 signaling through GLP-1RAs increased CaMKK2-AMPK signaling, which subsequently reduced BACE1-mediated cleavage of amyloid precursor protein (APP) and Aβ generation. GLP-1RAs also increased AMPK activity in microglia, inhibiting neuroinflammation and promoting Aβ phagocytosis. Consequently, GLP-1RAs inhibited plaque formation and improved memory deficits in AD model mice. Our findings indicate that AMPK activation mediates the effects of GLP-1RAs on AD, highlighting the therapeutic potential of GLP-1RAs for the treatment of AD.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Huaqiu Chen
- National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yijia Feng
- The Second Affiliated Hospital and Yuying Children's Hospital, Zhejiang Key Laboratory of Alzheimer's Disease, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou Medical University, Wenzhou, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Center for Geriatric Medicine and Institute of Aging, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Mingjing Liu
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhi Lu
- Department of Nuclear Medicine, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Bolang Hu
- The Second Affiliated Hospital and Yuying Children's Hospital, Zhejiang Key Laboratory of Alzheimer's Disease, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou Medical University, Wenzhou, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Center for Geriatric Medicine and Institute of Aging, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Lifen Chen
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Center for Geriatric Medicine and Institute of Aging, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yang Zhang
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jiawen Liu
- National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Fang Cai
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Center for Geriatric Medicine and Institute of Aging, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yifan Zhao
- The Second Affiliated Hospital and Yuying Children's Hospital, Zhejiang Key Laboratory of Alzheimer's Disease, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou Medical University, Wenzhou, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Center for Geriatric Medicine and Institute of Aging, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Wenhao Pan
- The Second Affiliated Hospital and Yuying Children's Hospital, Zhejiang Key Laboratory of Alzheimer's Disease, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou Medical University, Wenzhou, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Center for Geriatric Medicine and Institute of Aging, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xinxin Liao
- Department of Geriatric Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Sipei Pan
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Center for Geriatric Medicine and Institute of Aging, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Isabel Bestard-Lorigados
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yili Wu
- The Second Affiliated Hospital and Yuying Children's Hospital, Zhejiang Key Laboratory of Alzheimer's Disease, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou Medical University, Wenzhou, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Center for Geriatric Medicine and Institute of Aging, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Weihong Song
- The Second Affiliated Hospital and Yuying Children's Hospital, Zhejiang Key Laboratory of Alzheimer's Disease, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou Medical University, Wenzhou, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Center for Geriatric Medicine and Institute of Aging, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China.
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
32
|
Zhao Z, Sun Y, Jia M, Jiang M, Ruan X. Prophylactic administration of Kanli granule maintains fatty acid oxidation in the myocardium to prevent heart failure via activating AMPK/PPARα/CPT1A pathway: A network pharmacology-based study. Fitoterapia 2025; 184:106633. [PMID: 40398514 DOI: 10.1016/j.fitote.2025.106633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/18/2025] [Accepted: 05/17/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Abnormal energy metabolism plays a crucial role in the pathogenesis of heart failure (HF). Kanli granule (KLG), as an effective herbal medicine for treating HF, has been used in clinical practice for nearly 30 years. However, its underlying mechanisms have not been fully elucidated. METHODS This study combined network pharmacology, molecular docking, and in vivo experiments to explore KLG's effect on HF. Wistar rats with AAC-induced HF were orally administered KLG (0.675/1.35/2.7 g/kg) for 32 weeks. Assessments included heart weight index, echocardiography, histopathology, fatty acid metabolism (FAM) targets, and myocardial energy metrics. We focused on fatty acid oxidation (FAO) pathway, measuring AMPK, PPARα, and CPT1A at protein and gene levels. RESULTS KLG maintained cardiac function in AAC rats. Network pharmacology identified PPAR and AMPK pathways as key in FAM. Molecular docking showed strong affinity of KLG components to FAO targets PPARα and CPT1A. KLG significantly enhanced myocardial energy metabolism, reduced myocardial FFA levels, and increased ATP/ADP ratios. It activated AMPK and upregulated FAO-related genes, including PPARα and CPT1A. CONCLUSION KLG improves FAO in AAC-induced HF rats by activating the AMPK/PPARα/CPT1A pathway, reducing myocardial FFA levels, and improving myocardial microstructure and cardiac function.
Collapse
Affiliation(s)
- Zhejun Zhao
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuanlong Sun
- Cardiovascular Department, Shuguang Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Meijun Jia
- Cardiovascular Department, Shuguang Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Meixian Jiang
- Cardiovascular Department, Shuguang Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xiaofen Ruan
- Cardiovascular Department, Shuguang Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
33
|
Ma T, He J, Long Q, Wang Y, Chen F, Chen S, Xu K, Cao Y. Orientin attenuates UVB-induced skin photodamage by inhibiting ROS generation via the AMPK/Nrf2 axis. Int Immunopharmacol 2025; 155:114655. [PMID: 40239333 DOI: 10.1016/j.intimp.2025.114655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
The accumulation of reactive oxygen species (ROS) in the skin following UVB exposure is a key contributor to ultraviolet-induced skin photodamage. Orientin, a bioactive flavonoid, has demonstrated antioxidant properties in previous studies. However, its efficacy in treating skin photodamage remains inadequately understood. This study investigates the effects of orientin in preventing UVB-induced immortalized human keratinocytes (HaCaT cells) and BALB/c mouse skin photodamage by activating the AMPK/Nrf2 axis. Results show that orientin protects HaCaT cell viability after UVB exposure, reduces ROS levels, and upregulates antioxidant enzymes, including SOD1, HO-1, and NQO-1, while concurrently suppressing the expression of inflammatory mediators such as COX-2, IL-6, and IL-8. Additionally, orientin promotes AMPK phosphorylation, which facilitates Nrf2 nuclear translocation, thereby enhancing the antioxidant defense of cells. This effect is diminished upon inhibition of AMPK or Nrf2. In the BALB/c mouse model of photodamage, topical application of orientin alleviates symptoms like skin roughness, scaling, and erythema induced by UVB irradiation, while also elevating antioxidant enzyme expression in skin tissues. These findings suggest that orientin mitigates ultraviolet-induced skin photodamage both in vitro and in vivo, boosts cellular antioxidant capacity, and diminishes inflammatory responses, suggesting its potential for further exploration in skin photodamage management.
Collapse
Affiliation(s)
- Ting Ma
- School of Clinical Medicine, Guizhou Medical University, Guiyang 550004, Guizhou Province, China; Department of Dermatology, Guizhou Provincial People's Hospital, Guiyang 550000, Guizhou Province, China; Molecular Biology Laboratory, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Jing He
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550004, China
| | - Qiu Long
- School of Clinical Medicine, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Ye Wang
- School of Clinical Medicine, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Feng Chen
- School of Clinical Medicine, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Shaojie Chen
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Kexin Xu
- School of Clinical Medicine, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Yu Cao
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China.
| |
Collapse
|
34
|
Lin HH, Yu PR, Tseng CY, Lee MS, Chen JH. Protective Effects of Lotus Seedpod Extract on Hepatic Lipid and Glucose Metabolism via AMPK-Associated Mechanisms in a Mouse Model of Metabolic Syndrome and Oleic Acid-Induced HepG2 Cells. Antioxidants (Basel) 2025; 14:595. [PMID: 40427477 PMCID: PMC12108490 DOI: 10.3390/antiox14050595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 05/07/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Metabolic syndrome (MetS) poses considerable toxicological risks due to its association with an increased likelihood of metabolic dysfunction-associated steatotic liver disease (MASLD), and is characterized by hypertension, hyperglycemia, dyslipidemia, and obesity. This study aimed to investigate the therapeutic potential of flavonoid-rich lotus seedpod extract (LSE) in alleviating MetS and MASLD-related hepatic disturbances. In vivo, mice subjected to a high-fat diet (HFD) and streptozotocin (STZ) injection were supplemented with LSE or simvastatin for 6 weeks. Obesity indicators included body weight and epididymal fat, while insulin resistance was measured by fasting serum glucose, serum insulin, homeostasis model assessment-insulin resistance index (HOMA-IR), and oral glucose tolerance (OGTT). Also, the levels of serum lipid profiles and blood pressure were evaluated. Adipokines, proinflammatory cytokines, liver fat droplets, and peri-portal fibrosis were analyzed to clarify the mechanism of MetS. LSE significantly reduced the HFD/STZ-induced MetS markers better than simvastatin, as demonstrated by hypoglycemic, hypolipidemic, antioxidant, and anti-inflammatory effects. In vitro, LSE improved oleic acid (OA)-triggered phenotypes of MASLD in hepatocyte HepG2 cells by reducing lipid accumulation and enhancing cell viability. This effect might be mediated through proteins involved in lipogenesis that are downregulated by adenosine monophosphate-activated protein kinase (AMPK). In addition, LSE reduced reactive oxygen species (ROS) generation and glycogen levels, as demonstrated by enhancing insulin signaling involving reducing insulin receptor substrate-1 (IRS-1) Ser307 phosphorylation and increasing glycogen synthase kinase 3 beta (GSK3β) and protein kinase B (PKB) expression. These benefits were dependent on AMPK activation, as confirmed by the AMPK inhibitor compound C. These results indicate that LSE exhibits protective effects against MetS-caused toxicological disturbances in hepatic carbohydrate and lipid metabolism, potentially contributing to its efficacy in preventing MASLD or MetS.
Collapse
Affiliation(s)
- Hui-Hsuan Lin
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City 40201, Taiwan;
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City 40201, Taiwan
| | - Pei-Rong Yu
- Department of Nutrition, Chung Shan Medical University, Taichung City 40201, Taiwan; (P.-R.Y.); (C.-Y.T.)
| | - Chiao-Yun Tseng
- Department of Nutrition, Chung Shan Medical University, Taichung City 40201, Taiwan; (P.-R.Y.); (C.-Y.T.)
| | - Ming-Shih Lee
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City 40201, Taiwan;
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City 40201, Taiwan
| | - Jing-Hsien Chen
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City 40201, Taiwan
- Department of Nutrition, Chung Shan Medical University, Taichung City 40201, Taiwan; (P.-R.Y.); (C.-Y.T.)
| |
Collapse
|
35
|
Wolska W, Gutowska I, Wszołek A, Żwierełło W. The Role of Intermittent Fasting in the Activation of Autophagy Processes in the Context of Cancer Diseases. Int J Mol Sci 2025; 26:4742. [PMID: 40429883 PMCID: PMC12112746 DOI: 10.3390/ijms26104742] [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: 04/27/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Intermittent fasting (IF) is a dietary approach that influences key metabolic pathways, including autophagy-a crucial mechanism in maintaining cellular homeostasis. Autophagy plays a dual role in oncogenesis, acting both as a tumor suppressor and a survival mechanism under metabolic stress. IF has shown potential for reducing cancer risk and enhancing therapeutic efficacy by sensitizing tumor cells to chemotherapy and radiotherapy. However, its effects depend heavily on the type and stage of cancer. Potential risks, such as excessive weight loss and malnutrition, require careful evaluation. Further clinical studies are needed to optimize IF protocols as adjuncts to cancer therapy. This review discusses autophagy mechanisms induced by IF, their therapeutic implications in oncology, and the limitations of this dietary strategy.
Collapse
Affiliation(s)
- Waleria Wolska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (W.W.); (W.Ż.)
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Erling Skjalgssons gate 1, 7030 Trondheim, Norway
| | - Izabela Gutowska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (W.W.); (W.Ż.)
| | - Agata Wszołek
- Institute of Biology, University of Szczecin, Wąska 13, 71-415 Szczecin, Poland;
| | - Wojciech Żwierełło
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (W.W.); (W.Ż.)
| |
Collapse
|
36
|
Wang X, Gui H, Liu C, Huo F, Lan W, Zhu X, Wang W, Ma A, Lan J. ENTR1 regulates periodontitis by modulating macrophage M1 polarization via AMPK activation. Life Sci 2025; 369:123525. [PMID: 40054733 DOI: 10.1016/j.lfs.2025.123525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 02/26/2025] [Accepted: 03/01/2025] [Indexed: 03/30/2025]
Abstract
AIMS Periodontitis is a chronic inflammatory disorder arising from an imbalance between oral microbiota and the host's immune response, with macrophages as pivotal targets for prevention and treatment. Endosome-associated Trafficking Regulator 1 (ENTR1) is indispensable for protein trafficking and implant osseointegration. However, its specific role in periodontitis has yet to be clarified. This research seeks to explore the effects of ENTR1 on macrophage polarization, elucidate its mechanisms, and evaluate its regulatory functions in the regeneration of periodontal tissues. MATERIALS AND METHODS A ligature-induced periodontitis mouse model was established to investigate the correlation between macrophage polarization markers and ENTR1 expression. Techniques including qRT-PCR, Western blot, ELISA, flow cytometry, and immunofluorescence staining were utilized to evaluate the impact of ENTR1 on macrophage polarization under inflammatory stimuli. Micro-CT and histological staining were applied to assess periodontal bone resorption. The interaction between ENTR1 and AMP-activated protein kinase (AMPK) was explored through Western blot and co-immunoprecipitation, further validated by applying the AMPK inhibitor Compound C (CpC). KEY FINDINGS ENTR1 expression was down-regulated in the mice with periodontitis relative to healthy controls. Overexpressing ENTR1 suppressed macrophage M1 polarization and mitigated bone loss in periodontitis, while knocking down ENTR1 exacerbated these effects. ENTR1 directly interacted with AMPK, enhancing its phosphorylation. Furthermore, the inhibitory impact of ENTR1 on macrophage M1 polarization and inflammation-induced alveolar bone resorption were partially attenuated by CpC treatment. SIGNIFICANCE ENTR1 regulates periodontitis by suppressing macrophage M1 polarization through enhancing AMPK phosphorylation, presenting a promising therapeutic target for its prevention and management.
Collapse
Affiliation(s)
- Xi Wang
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Houda Gui
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Chenghang Liu
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Fenglei Huo
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Weipeng Lan
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Xingyan Zhu
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Wenhao Wang
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Anquan Ma
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Jing Lan
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, 250012 Jinan, Shandong, China.
| |
Collapse
|
37
|
Sureka N, Zaheer S. Regulatory T Cells in Tumor Microenvironment: Therapeutic Approaches and Clinical Implications. Cell Biol Int 2025. [PMID: 40365758 DOI: 10.1002/cbin.70031] [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: 03/10/2025] [Revised: 04/19/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025]
Abstract
Regulatory T cells (Tregs), previously referred to as suppressor T cells, represent a distinct subset of CD4+ T cells that are uniquely specialized for immune suppression. They are characterized by the constitutive expression of the transcription factor FoxP3 in their nuclei, along with CD25 (the IL-2 receptor α-chain) and CTLA-4 on their cell surface. Tregs not only restrict natural killer cell-mediated cytotoxicity but also inhibit the proliferation of CD4+ and CD8+ T-cells and suppress interferon-γ secretion by immune cells, ultimately impairing an effective antitumor immune response. Treg cells are widely recognized as a significant barrier to the effectiveness of tumor immunotherapy in clinical settings. Extensive research has consistently shown that Treg cells play a pivotal role in facilitating tumor initiation and progression. Conversely, the depletion of Treg cells has been linked to a marked delay in tumor growth and development.
Collapse
Affiliation(s)
- Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
38
|
Yang X, Liang N, Liu D, Yan J, Yang X, Lv J, Xiao S, Wei X, Chen X, Yang Z, Gui S, Jin L, Yu S, Lyu J, Ren X. Antioxidant capacity of the iron-sulfur cluster assembly protein IscU2 is mediated by aspartate metabolism to promote tumor survival. J Biol Chem 2025:110234. [PMID: 40378953 DOI: 10.1016/j.jbc.2025.110234] [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: 01/11/2025] [Revised: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025] Open
Abstract
Environmental nutrient levels affect cancer cell metabolism, activating adaptive mechanisms in cancer cells to deal with nutrient stress. However, it remains unclear how tumor cells sustain survival under nutrient-stress circumstances through metabolic reprogramming. Our study focused on nutrient deficiency-induced oxidative damage, revealing that increased expression of the iron-sulfur (Fe-S) cluster assembly protein, IscU2, is essential for the survival of pancreatic ductal adenocarcinoma (PDAC) cells in glucose-deficient conditions. Glucose deficiency induces IscU2 expression via the activation of the AMPK pathway, allowing IscU2 to exhibit antioxidant properties that are absent under glucose-sufficient conditions. Upregulated IscU2 stimulates aspartate synthesis by bolstering mitochondrial metabolism, including respiration and the tricarboxylic acid cycle, in a Fe-S cluster-dependent manner. Notably, oxidative stress and apoptosis induced by IscU2 depletion in glucose-deficient PDAC cells can be restored by aspartate-mediated NADPH production. These findings highlight the importance of IscU2 in PDAC cell metabolism and its essential function in supporting cell survival under nutrient-deficient conditions.
Collapse
Affiliation(s)
- Xunjun Yang
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Department of Laboratory Medicine, Wenzhou, Zhejiang, China; Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Na Liang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dandan Liu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jimei Yan
- Department of Laboratory Medicine, Linyi Peoples' Hospital, Linyi, Shandong, China
| | - Xiali Yang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jinya Lv
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Saijun Xiao
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiujuan Wei
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xuyang Chen
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhengquan Yang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Shanying Gui
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Liqin Jin
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Shihui Yu
- School of Basic Medical Sciences, the Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Jianxin Lyu
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Xiaojun Ren
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China; Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
39
|
Li Z, Liu J, Ju J, Peng X, Zhao W, Ren J, Jia X, Wang J, Tu Y, Gao F. Contributions of synaptic energetic dysfunction by microtubule dynamics and microtubule-based mitochondrial transport disorder to morphine tolerance. Br J Pharmacol 2025. [PMID: 40361281 DOI: 10.1111/bph.70048] [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: 11/12/2024] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND AND PURPOSE Morphine is among the most powerful analgesic, but its long-term use can cause tolerance. Synaptic ATP supply is critical for maintaining synaptic transmission. Microtubule-based mitochondrial transport ensures synaptic energy supply. How synaptic energy changes with morphine and the role of microtubule tracks in synaptic mitochondrial energy supply remain elusive. Chronic morphine treatment can destroy microtubule cytoskeletons. We investigated the effect of the microtubule cytoskeleton on synaptic mitochondrial energy supply and the mechanism of microtubule dynamics after morphine exposure. EXPERIMENTAL APPROACH Rats were treated with long-term morphine and the effect on thermal pain thresholds was evaluated by the tail-flick latency test. Various antagonists and agonists were used elucidated the role and mechanism of synaptic mitochondrial energy supply and microtubules in morphine tolerance in vivo and in SH-SY5Y cells. KEY RESULTS Chronic morphine treatment reduced synaptic mitochondrial ATP production. Improving mitochondrial oxidative phosphorylation (OXPHOS) alleviated the downregulation of synaptic ATP levels. Microtubule-stabilizing agents prevented microtubule disruption and ameliorated synaptic energy deficit via microtubule-based microtubule transport. In SH-SY5Y cells, morphine exposure reduced microtubule expression. And re-opening the synaptic Ca2+ channel by agonist alleviated microtubule decrease by calcium/calmodulin-dependent protein kinase 2 (CAMKK2)/AMP-activated protein kinase (AMPK) pathway. CONCLUSION AND IMPLICATIONS This study demonstrates that the microtubule cytoskeleton regulated by the Ca2+-CAMKK2-AMPK axis is critical for synaptic mitochondrial transport and ATP production, explaining an interplay between chronic morphine-induced abnormal neuroadaptation and synaptic energetic dysfunction. These findings implicated a potential clinical strategy for prolonging the opioid antinociceptive effect during long-term pain control.
Collapse
Affiliation(s)
- Zheng Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Anesthesiology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Jie Liu
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Ju
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoling Peng
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Zhao
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jihao Ren
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqian Jia
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jihong Wang
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ye Tu
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Gao
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
40
|
Zhang Y, Bai C, Sha J, Huo X, Qu D, Chen J. Ginseng Soluble Dietary Fiber Reverses Obesity via the PPAR/AMPK Signaling Pathway and Improves Intestinal Flora in Mice. Foods 2025; 14:1716. [PMID: 40428495 PMCID: PMC12111629 DOI: 10.3390/foods14101716] [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: 04/07/2025] [Revised: 05/04/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Ginseng soluble dietary fiber (GSDF) has been shown to have good physicochemical properties; however, its in vivo benefits in obesity are yet to be fully elucidated. METHODS To explore this, C57BL/6J obese mice were given metformin hydrochloride and different doses of GSDF for 60 days. The levels of blood lipids and inflammatory factors were detected by ELISA, and the pathological alterations were detected through the application of HE staining. The level of adipose tissue protein in epididymis was detected by Western blotting and through the effects of 16S rRNA sequencing on gut microbiota. RESULTS The results showed that GSDF significantly improved basal physiological indices, lipid levels, and serum cytokine levels in the obese mice. GSDF increased the expression levels of PPAR-γ, AMPK, and P-AMPK proteins, and lowered the expression of IL-1β, TNF-α, and other proteins in the adipose tissues of the epididymis, in turn inhibiting adipogenesis and ameliorating lipid metabolism disorders. By lowering the Firmicutes/Bacteroidetes ratio in the gut and altering the abundance of thick-walled bacteria and mycobacterium, the abundance of species such as Lactobacillus, Alloprevotella, and Faecalibaculum was altered to improve cecum health. CONCLUSIONS These results suggest that GSDF may have a positive effect on growth, obesity, and cecal health in obese mice.
Collapse
Affiliation(s)
| | | | | | | | | | - Jianbo Chen
- Institute of Special Animals and Plants, Chinese Academy of Agricultural Sciences, Changchun 130112, China; (Y.Z.); (C.B.); (J.S.); (X.H.); (D.Q.)
| |
Collapse
|
41
|
Cheng M, Hou Y, Chen Q, Ge S, Chen C, Zheng X, Zhang C. Exploring the wound-healing mechanism of Cayratia japonica extract: A combined experimental and network pharmacology study. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119810. [PMID: 40239878 DOI: 10.1016/j.jep.2025.119810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/10/2025] [Accepted: 04/13/2025] [Indexed: 04/18/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Wound healing is a complex biological process and remains a significant challenge due to the lack of effective therapeutic drugs. Cayratia japonica (CJG), a traditional folk medicine, has been widely used for its anti-inflammatory and efficacy in treating traumatic injuries. AIM OF THE STUDY This study aimed to investigate the wound-healing effects of CJG and elucidate its underlying mechanism. METHODS First, the phytochemical composition of CJG was identified using high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS), and its potential wound-healing mechanisms were predicated via network pharmacology. Next, in vivo experiments were conducted by dividing subjects into control, CJG (1.5-6 mg/cm2), and bFGF (150 IU/cm2) groups to assess its therapeutic efficacy. Finally, the mechanism of CJG and its key bioactive component, luteolin-7-O-glucoside (LUT-7G), were explained through polymerase chain reaction (PCR), Western blotting, histopathology, immunofluorescence, plasmid transfection, colony formation unit assays, and cellular thermal shift assay (CETSA). RESULTS LC-MS/MS identified 15 major constituents of CJG and 102 potential wound healing-related targets. Network pharmacology analysis revealed key enriched pathways, including AMPK, TNF, and metabolic pathways. In vivo, CJG significantly accelerated wound-healing by inhibiting inflammatory responses, promoting angiogenesis, and modulating collagen deposition. In vitro, LUT-7G treatment markedly enhanced the proliferation and migration of HaCaT and HSF cells. Mechanistically, LUT-7G exerted its wound-healing effects by activating the AMPK/CTHRC1/TGF-β1 signaling pathway in HaCaT cells. In conclusion, CJG significantly promotes wound healing by regulating AMPK signaling pathways, indicating its promising clinical application prospects.
Collapse
Affiliation(s)
- Mengqin Cheng
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, 211198, China
| | - Yi Hou
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 21000, China
| | - Qi Chen
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, 211198, China
| | - Shanchun Ge
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, 211198, China
| | - Ce Chen
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, 211198, China
| | - Xueping Zheng
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 21000, China.
| | - Chaofeng Zhang
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
42
|
Han J, Song J, Jung ES, Choi JW, Ji HY, Mook-Jung I. SGLT2 Inhibition by Enavogliflozin Significantly Reduces Aβ Pathology and Restores Cognitive Function via Upregulation of Microglial AMPK Signaling in 5XFAD Mouse Model of Alzheimer's Disease. Aging Cell 2025:e70101. [PMID: 40346951 DOI: 10.1111/acel.70101] [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: 03/28/2025] [Accepted: 04/22/2025] [Indexed: 05/12/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline. Metabolic dysfunctions, particularly type 2 diabetes mellitus (T2DM), have been implicated in AD pathogenesis, highlighting the potential for novel therapeutic approaches targeting shared underlying mechanisms. Here, we investigate sodium-glucose cotransporter 2 (SGLT2) inhibition as a therapeutic strategy for AD using Enavogliflozin, a potent SGLT2 inhibitor, in the 5XFAD mouse model. Five-month-old 5XFAD mice were treated with Enavogliflozin (0.1 or 1 mg/kg) or vehicle for 8 weeks. The higher dose significantly improved cognitive performance in Y-maze and Morris Water Maze tests, which correlated with enhanced synaptic plasticity and increased acetylcholine levels. Moreover, Enavogliflozin treatment reduced Aβ pathology and plaque burden, particularly affecting larger plaques. Mechanistically, SGLT2 inhibition attenuated neuroinflammation by suppressing NF-κB signaling and proinflammatory cytokine production while promoting microglial recruitment to plaques. In vitro and ex vivo analyses further revealed that Enavogliflozin enhances microglial phagocytic capacity via AMPK-mediated mitochondrial biogenesis and function. These findings highlight the multifaceted neuroprotective effects of SGLT2 inhibition in AD, demonstrating its potential to mitigate pathology and improve cognitive function. By uncovering its impact on neuroinflammation and microglial function, this study establishes SGLT2 inhibition as a promising therapeutic avenue for AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Jihui Han
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jaehoon Song
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Eun Sun Jung
- Convergence Dementia Research Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Ji Won Choi
- Life Science Institute, Daewoong Pharmaceutical, Yongin, Republic of Korea
| | - Hye Young Ji
- Life Science Institute, Daewoong Pharmaceutical, Yongin, Republic of Korea
| | - Inhee Mook-Jung
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Convergence Dementia Research Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
43
|
Yang J, Yang Y, Tan X, Du H, Zhou Z, Chen L, Tian X, Zheng G, Hu J, Zhang C, Qiu Z. Unlocking the potential of the ACE2/Ang-(1-7)/Mas Axis in liver diseases: From molecular mechanisms to translational applications. Diabetes Obes Metab 2025. [PMID: 40344459 DOI: 10.1111/dom.16435] [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: 01/25/2025] [Revised: 04/11/2025] [Accepted: 04/21/2025] [Indexed: 05/11/2025]
Abstract
Over the past two decades, the identification of new functions within the renin-angiotensin system (RAS) has extended beyond its traditional roles, with the emergence of the angiotensin-converting enzyme 2 (ACE2)/Ang-(1-7)/Mas axis being particularly significant. This axis is hypothesized to balance or modulate the effects of the traditional ACE/Ang II/AT1 axis in various physiological and pathological contexts. ACE2, a membrane-bound carboxypeptidase and an ancient homologue of ACE converts Angiotensin II (Ang II) into Angiotensin 1-7 (Ang-(1-7)). The Mas receptor is a G-protein-coupled receptor that specifically binds Ang-(1-7). Recent research has increasingly focused on the local expression of RAS in different tissues. Ang-(1-7) produces a variety of biological effects by binding to the Mas receptor, including anti-inflammatory, antioxidant, anti-apoptotic and anti-fibrotic actions, thereby influencing a range of mechanisms in the heart, kidneys, brain and other tissues. Preclinical animal model studies indicate that manipulating the protective RAS can significantly alter the progression of multiple liver diseases. Hepatic overexpression of ACE2 or administration of Ang-(1-7) and its analogues has been shown to be therapeutically effective against drug-induced liver injury, metabolic-associated fatty liver disease, liver fibrosis and hepatocellular carcinoma progression. These effects are achieved through various pathways, including the regulation of lipid metabolism, inhibition of epithelial-mesenchymal transition (EMT) and extracellular matrix (ECM) production, as well as suppression of aerobic glycolysis. In current clinical trials, while recombinant human ACE2 (Rh-ACE2) has demonstrated safety and good tolerance in most studies, research on the relevance of activating the ACE2/Ang-(1-7) axis in the mechanisms and evolution of human diseases remains in its early stages. Therefore, further elucidation of the complex interactions between the classical and counter-regulatory RAS axes in clinical settings is crucial. This review will summarize the roles of selective activation of the ACE2/Ang-(1-7)/Mas axis, with a focus on its mechanisms in the treatment of liver diseases. Additionally, we will discuss the safety concerns regarding selective activation of the ACE2/Ang-(1-7)/Mas axis in clinical applications and the challenges of tissue-specific activation of this axis, providing effective therapeutic strategies for targeted activation of the hepatic ACE2/Ang-(1-7)/Mas axis in clinical practice.
Collapse
Affiliation(s)
- Jun Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Yuan Yang
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiangyun Tan
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Hongzhi Du
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Zhongshi Zhou
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Liang Chen
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Xianxiang Tian
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Guohua Zheng
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Junjie Hu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Cong Zhang
- College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Zhenpeng Qiu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
- Center of Traditional Chinese Medicine Modernization for Liver Diseases, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
- Hubei Shizhen Laboratory, Wuhan, People's Republic of China
| |
Collapse
|
44
|
Yu YS, Kim IS, Baek SH. Decoding the dual role of autophagy in cancer through transcriptional and epigenetic regulation. FEBS Lett 2025. [PMID: 40346781 DOI: 10.1002/1873-3468.70060] [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: 03/31/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 05/12/2025]
Abstract
Autophagy is a conserved catabolic process that is essential for maintaining cellular homeostasis by degrading and recycling damaged organelles and misfolded proteins. In cancer, autophagy exhibits a context-dependent dual role: In early stages, autophagy acts as a tumor suppressor by preserving genomic integrity and limiting oxidative stress. In advanced stages, autophagy supports tumor progression by facilitating metabolic adaptation, therapy resistance, immune evasion, and metastasis. This review highlights the molecular mechanisms underlying this dual function and focuses on the transcriptional and epigenetic regulation of autophagy in cancer cells. Key transcription factors, including the MiT/TFE family, FOXO family, and p53, as well as additional regulators, are discussed in the context of stress-responsive pathways mediated by mTORC1 and AMPK. A deeper understanding of the transcriptional and epigenetic regulation of autophagy in cancer is crucial for developing context-specific therapeutic strategies to either promote or inhibit autophagy depending on the cancer stage, thereby improving clinical outcomes in cancer treatment.
Collapse
Affiliation(s)
- Young Suk Yu
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Ik Soo Kim
- Department of Microbiology, Gachon University College of Medicine, Incheon, South Korea
| | - Sung Hee Baek
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
45
|
Li Y, Zheng L, Chen M, Li R, Yu Y, Qiao L, Liu J, Zhang X, Zhang Y, Zhang Y, Zheng W. Nootkatone Alleviates Type 2 Diabetes in db/db Mice Through AMPK Activation and ERK Inhibition: An Integrated In Vitro and In Vivo Study. Molecules 2025; 30:2111. [PMID: 40430283 PMCID: PMC12114572 DOI: 10.3390/molecules30102111] [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: 03/19/2025] [Revised: 05/03/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a common chronic metabolic disorder that imposes a substantial healthcare burden globally. Recent advances highlight the potential of natural products in ameliorating T2DM. In this study, we investigated the therapeutic efficacy of nootkatone (Nok), a natural sesquiterpene ketone, in T2DM and elucidated its underlying mechanisms. In vivo experiments demonstrated that Nok administration markedly improved dysregulated glucose metabolism and ameliorated serum biochemical abnormalities in db/db mice. Leveraging a network pharmacology-based approach, we identified putative molecular targets of Nok. Subsequent in vitro analyses revealed that Nok significantly enhanced glucose consumption in cultured cells. Mechanistically, Nok robustly activated AMP-activated protein kinase (AMPK) while suppressing mitogen-activated protein kinase (MAPK) signaling. Western blot validation further indicated that Nok downregulated the phosphorylation of MAPK1/3 (ERK2/1), attenuating MAPK pathway activation and thereby alleviating metabolic dysfunction-associated fatty liver disease (MAFLD) progression in the diabetic model. Collectively, our findings suggest that Nok exerts anti-diabetic effects via dual modulation of AMPK activation and MAPK inhibition, effectively restoring metabolic homeostasis and mitigating inflammation in T2DM. This study positions Nok as a promising natural compound for therapeutic intervention in T2DM and associated metabolic disorders.
Collapse
Affiliation(s)
- Yingjie Li
- School of Pharmacy, Harbin University of Commerce, Harbin 150028, China;
| | - Linlin Zheng
- Department of Pharmacology, College of Basic Medicine and Life Sciences, Hainan Medical College, Haikou 571199, China
| | - Mimi Chen
- Hainan Academy of Medical Sciences, Haikou 571199, China
| | - Ruodi Li
- Department of Pharmacology, College of Basic Medicine and Life Sciences, Hainan Medical College, Haikou 571199, China
| | - Yansu Yu
- Department of Pharmacology, College of Basic Medicine and Life Sciences, Hainan Medical College, Haikou 571199, China
| | - Lu Qiao
- Department of Pharmacology, College of Basic Medicine and Life Sciences, Hainan Medical College, Haikou 571199, China
| | - Jialu Liu
- School of Pharmacy, Hainan Medical College, Haikou 571199, China
| | - Xiaopo Zhang
- School of Pharmacy, Hainan Medical College, Haikou 571199, China
| | - Yong Zhang
- Department of Pharmacology, College of Basic Medicine and Life Sciences, Hainan Medical College, Haikou 571199, China
- Hainan Academy of Medical Sciences, Haikou 571199, China
| | - Yuxin Zhang
- Hainan Academy of Medical Sciences, Haikou 571199, China
| | - Wei Zheng
- School of Pharmacy, Harbin University of Commerce, Harbin 150028, China;
| |
Collapse
|
46
|
Nie Y, Yu H, Wan X, Zheng L, Cao S, Yang D, Xiao D. Identification of autophagy-related biomarker and analysis of immune infiltrates in diabetic nephropathy: PTGER1 protein macromolecular structure and function. Int J Biol Macromol 2025; 311:144063. [PMID: 40348252 DOI: 10.1016/j.ijbiomac.2025.144063] [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: 03/17/2025] [Revised: 04/24/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Diabetic nephropathy (DKD) is a common complication of diabetic patients, which seriously affects their quality of life and longevity. In recent years, autophagy has been found to play an important role in the pathogenesis of DKD. The aim of this study is to identify markers of autophagy associated with diabetic nephropathy, with special attention to the structural and biological role of PTGER1 protein and its role in immune infiltration. The DKD differential genes were identified by means of various sexually expressed genes (DEGs) analysis, and functional enrichment (GO) and pathway enrichment (KEGG) analysis were performed for the significant differential genes combined with weighted gene co-expression network analysis (WGCNA). Meanwhile, gene collection enrichment analysis (GSEA) was used to further explore the function of autophagy related genes. By constructing protein-protein interaction network (PPI) and GeneMania analysis, PTGER1 was identified as a key hub gene. The diagnostic value of biomarkers was evaluated by ROC curve, and the infiltration of different immune cells was analyzed. The results showed that PTGER1 was significantly up-regulated in the renal tissues of diabetic nephropathy patients. WGCNA and functional enrichment analysis revealed a close correlation between PTGER1-related autophagy pathways and immune responses. The constructed PPI network indicated that PTGER1 had significant interactions with multiple autophagy and inflammation-related proteins, and ROC curve analysis indicated the potential application value of PTGER1 in the early screening of DKD.
Collapse
Affiliation(s)
- Yanfang Nie
- Department of Nephrology, Taizhou Central Hospital, The First Affiliated Hospital of Taizhou University, Taizhou 318000, Zhejiang Province, China
| | - Haifeng Yu
- Department of Nephrology, Taizhou Central Hospital, The First Affiliated Hospital of Taizhou University, Taizhou 318000, Zhejiang Province, China
| | - Xiaoqing Wan
- Department of Nephrology, Taizhou Central Hospital, The First Affiliated Hospital of Taizhou University, Taizhou 318000, Zhejiang Province, China
| | - Lu Zheng
- Department of Nephrology, Taizhou Central Hospital, The First Affiliated Hospital of Taizhou University, Taizhou 318000, Zhejiang Province, China
| | - Susu Cao
- Department of Nephrology, Taizhou Central Hospital, The First Affiliated Hospital of Taizhou University, Taizhou 318000, Zhejiang Province, China
| | - Dongdong Yang
- Department of Nephrology, Taizhou Central Hospital, The First Affiliated Hospital of Taizhou University, Taizhou 318000, Zhejiang Province, China
| | - Daping Xiao
- Department of Nephrology, The First People's Hospital of Taizhou City, Huangyan Hospital Affiliated to Wenzhou Medical University, Taizhou 318000, Zhejiang Province, China.
| |
Collapse
|
47
|
André DCA, Oliveira PF, Alves MG, Martins AD. Caloric Restriction and Sirtuins as New Players to Reshape Male Fertility. Metabolites 2025; 15:303. [PMID: 40422880 DOI: 10.3390/metabo15050303] [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: 03/11/2025] [Revised: 04/25/2025] [Accepted: 04/30/2025] [Indexed: 05/28/2025] Open
Abstract
Over the years, caloric intake has remained a subject of profound scrutiny. Within the scientific community, there has been rigorous debate to ascertain which path is most ideal for enhancing quality of life and extending the human lifespan. Caloric restriction has been shown to be a promising contributor towards longevity and delaying the onset of age-related diseases. This diet consists of a reduction in caloric intake while maintaining essential energy and nutritional requirements to achieve optimal health while avoiding malnutrition. However, the effects of this nutritional regimen on male reproductive health have not yet been comprehensively studied. Nevertheless, such a complex process will certainly be regulated by a variety of metabolic sensors, likely sirtuins. Evidence has been gathered regarding this group of enzymes, and their ability to regulate processes such as chromatin condensation, the cell cycle, insulin signaling, and glucose and lipid metabolism, among many others. Concerning testicular function and male fertility, sirtuins can modulate certain metabolic processes through their interaction with the hypothalamic-pituitary-gonadal axis and mitochondrial dynamics, among many others, which remain largely unexplored. This review explores the impact of caloric restriction on male fertility, highlighting the emerging role of sirtuins as key regulators of male reproductive health through their influence on cellular metabolism.
Collapse
Affiliation(s)
- Diana C A André
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Pedro F Oliveira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Marco G Alves
- Institute of Biomedicine, Department of Medical Sciences (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ana D Martins
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
48
|
Machado IF, Palmeira CM, Rolo AP. Sestrin2 is a central regulator of mitochondrial stress responses in disease and aging. Ageing Res Rev 2025; 109:102762. [PMID: 40320152 DOI: 10.1016/j.arr.2025.102762] [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: 01/08/2025] [Revised: 04/09/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025]
Abstract
Mitochondria supply most of the energy for cellular functions and coordinate numerous cellular pathways. Their dynamic nature allows them to adjust to stress and cellular metabolic demands, thus ensuring the preservation of cellular homeostasis. Loss of normal mitochondrial function compromises cell survival and has been implicated in the development of many diseases and in aging. Although exposure to continuous or severe stress has adverse effects on cells, mild mitochondrial stress enhances mitochondrial function and potentially extends health span through mitochondrial adaptive responses. Over the past few decades, sestrin2 (SESN2) has emerged as a pivotal regulator of stress responses. For instance, SESN2 responds to genotoxic, oxidative, and metabolic stress, promoting cellular defense against stress-associated damage. Here, we focus on recent findings that establish SESN2 as an orchestrator of mitochondrial stress adaptation, which is supported by its involvement in the integrated stress response, mitochondrial biogenesis, and mitophagy. Additionally, we discuss the integral role of SESN2 in mediating the health benefits of exercise as well as its impact on skeletal muscle, liver and heart injury, and aging.
Collapse
Affiliation(s)
- Ivo F Machado
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CiBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Institute of Interdisciplinary Research, Doctoral Program in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, Coimbra, Portugal
| | - Carlos M Palmeira
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CiBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Anabela P Rolo
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CiBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
49
|
Ma J, Yang Y, Zang C, Chen Q, Jiang Y, Dong Y, Wang J, Zhou N, Yang X, Li F, Bao X, Zhang D. Inhibiting mitochondrial excessive fission alleviates the neuronal damage in Parkinson's disease via regulating PGC-1α mediated mitochondrial biogenesis. Exp Neurol 2025; 391:115288. [PMID: 40320116 DOI: 10.1016/j.expneurol.2025.115288] [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: 12/06/2024] [Revised: 04/24/2025] [Accepted: 04/30/2025] [Indexed: 05/17/2025]
Abstract
Mitochondrial excessive fission is one of representative pathological features and a principal element triggering the neuronal damage in Parkinson's disease (PD). Inhibiting mitochondrial excessive fission benefits the pathology of PD through promoting mitochondrial biogenesis, but the detailed mechanism has not been clarified. In our study, we revealed that inhibiting mitochondrial excessive fission by Mdivi-1, the dynamin related protein 1 (DRP1) inhibitor, increased the expression and nuclear translocation of peroxisome proliferator-activated receptor γ (PPARγ) coactivator 1α (PGC-1α), as well as its downstream transcriptional factors, nuclear respiratory factor 1/2 (NRF1/2) and mitochondrial transcription factor A (TFAM), and therefore promoted mitochondrial biogenesis. Suppression of mitochondrial excessive fission alleviated dopaminergic synaptic injury, neuronal apoptosis and motor dysfunction, while inhibiting PGC-1α attenuated these ameliorative effects in both in-vitro and in-vivo PD models. Mechanistic study showed that inhibiting mitochondrial excessive fission facilitated the expression of PGC-1α, NRF1 and TFAM by activation of Ca2+/calmodulin-dependent serine/threonine kinase II (CaMKII)/cAMP-response element binding protein (CREB) pathway. Inhibiting mitochondrial excessive fission also activated AMP-activated serine/threonine kinase (AMPK)/Sirtuin1 (Sirt1) pathway, and then phosphorylated and deacetylated PGC-1α by post-translational modifications. In conclusion, inhibiting mitochondrial excessive fission could promote mitochondrial biogenesis through activation of PGC-1α and therefore rescue the impaired dopaminergic neurons, which provided evidence for targeting mitochondrial excessive fission for the treatment of PD and new drug developments.
Collapse
Affiliation(s)
- Jingwei Ma
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Yang Yang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Caixia Zang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Qiuzhu Chen
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Yueqi Jiang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Yirong Dong
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Jinrong Wang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Ning Zhou
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Xing Yang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Fangfang Li
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Xiuqi Bao
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Dan Zhang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China.
| |
Collapse
|
50
|
Huang Q, Wang Y, Zhang Z, Wu M, Liu J, Chen J, Li J, Yao Y, Guo C, Zhao D, Qi W, Li X. Organ dysfunction induced by hemorrhagic shock: From mechanisms to therapeutic medicines. Pharmacol Res 2025; 216:107755. [PMID: 40315969 DOI: 10.1016/j.phrs.2025.107755] [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: 02/10/2025] [Revised: 04/03/2025] [Accepted: 04/27/2025] [Indexed: 05/04/2025]
Abstract
Hemorrhagic shock (HS) leads to organ dysfunction, which increases the incidence of unfavorable outcomes in patients. However, adjuvant drug therapy for HS has not been widely accepted, and the benefits of vasopressors are generally considered to have insufficient evidence. Energy homeostasis disruption and excessive immune system activation are the main mechanisms underlying HS-induced organ dysfunction. Recent reports on HS animal models and clinical trials have revealed potential drugs that target the immune response, oxidative damage, and energy homeostasis in HS, providing new insights for the treatment of HS-induced organ dysfunction. In this review, we first discuss the pathophysiology of organ dysfunction involved in HS injury and then systematically review potential drugs that regulate immunity, the inflammatory response, oxidative damage, energy homeostasis, and cell death. We also review the available drugs with clinical evidence of HS-induced organ dysfunction efficacy. Treatment strategies combined with an improved understanding of the organ injury mechanisms of HS may help identify and develop targeted therapeutic modalities that mitigate severe organ dysfunction and reduce mortality caused by HS injury.
Collapse
Affiliation(s)
- Qingxia Huang
- Research Center of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China; Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Yisa Wang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Zepeng Zhang
- Research Center of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China; Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Mingxia Wu
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Jiaqi Liu
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Jinjin Chen
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Jing Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Yao Yao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Chen Guo
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Daqing Zhao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Wenxiu Qi
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China.
| | - Xiangyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China.
| |
Collapse
|