1
|
Wang H, Qin Y, Niu J, Chen H, Lu X, Wang R, Han J. Evolving perspectives on evaluating obesity: from traditional methods to cutting-edge techniques. Ann Med 2025; 57:2472856. [PMID: 40077889 PMCID: PMC11912248 DOI: 10.1080/07853890.2025.2472856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Objective: This review examines the evolution of obesity evaluation methods, from traditional anthropometric indices to advanced imaging techniques, focusing on their clinical utility, limitations, and potential for personalized assessment of visceral adiposity and associated metabolic risks. Methods: A comprehensive analysis of existing literature was conducted, encompassing anthropometric indices (BMI, WC, WHR, WHtR, NC), lipid-related metrics (LAP, VAI, CVAI, mBMI), and imaging technologies (3D scanning, BIA, ultrasound, DXA, CT, MRI). The study highlights the biological roles of white, brown, and beige adipocytes, emphasizing visceral adipose tissue (VAT) as a critical mediator of metabolic diseases. Conclusion: Although BMI and other anthropometric measurements are still included in the guidelines, indicators that incorporate lipid metabolism information can more accurately reflect the relationship between metabolic diseases and visceral obesity. At the same time, the use of more modern medical equipment, such as ultrasound, X-rays, and CT scans, allows for a more intuitive assessment of the extent of visceral obesity.
Collapse
Affiliation(s)
- Heyue Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaxin Qin
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jinzhu Niu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Haowen Chen
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinda Lu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Rui Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jianli Han
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
2
|
Yang J, Zhang CR, Li ZX, Gao YH, Jiang L, Zhang J, Wang PY, Liu T. Spermine alleviates myocardial cell aging by inhibiting mitochondrial oxidative stress damage. Eur J Pharmacol 2025; 997:177477. [PMID: 40058754 DOI: 10.1016/j.ejphar.2025.177477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Myocardial aging, involving oxidative stress, mitochondrial dysfunction, and cellular senescence, is crucial to DOX - induced heart failure. DOX has dose - dependent cardiotoxicity. Sper a natural polyamine with antioxidant and anti - aging effects, remains unstudied in this context. AIM This study hypothesizes Sper can alleviate DOX - induced heart failure by curbing myocardial aging and oxidative stress. It aims to assess Sper's protective impacts on cardiac function, pathology, oxidative stress, mitochondrial damage, and aging in a rat model, using captopril as a control. METHODS 80 male Sprague Dawley rats were assigned to 8 groups: normal control, 150 mg/kg Sper, DOX, and DOX +10/50/100/150 mg/kg Sper, DOX +30 mg/kg captopril. DOX was given intraperitoneally at 15 mg/kg total dose, while Sper or captopril was administered daily via gavage for six weeks. Cardiac function was evaluated using echocardiography, and histopathological changes, oxidative stress markers, mitochondrial damage, and myocardial aging were assessed via H&E staining, immunofluorescence, Western blot, and electron microscopy. RESULTS Sper boosted cardiac function in DOX - treated rats, upping EF and SV, and lessening cardiac tissue damage. It cut oxidative stress by reducing MDA levels and boosting SOD activity. Sper also eased mitochondrial damage by enhancing mitochondrial membrane potential and cutting mitochondrial fission proteins (Drp1 and Fis1). Plus, Sper held back myocardial aging by trimming β - galactosidase activity and downregulating p - P53 and p21 expression. At 150 mg/kg/day, Sper worked much like 30 mg/kg/day captopril. CONCLUSION Sper effectively eased DOX - induced heart failure by targeting oxidative stress and aging, showing potential as an adjunct therapy for DOX - related cardiotoxicity. Future research should explore Sper's molecular mechanisms and clinical efficacy.
Collapse
Affiliation(s)
- Jing Yang
- Puyang Medical College, Puyang, 457000, China.
| | - Chun-Rui Zhang
- Cardiovascular Laboratory of Xinxiang, Xinxiang, 453003, China
| | - Zi-Xuan Li
- Xinxiang University Affiliated Middle School, Xinxiang, 453000, China
| | - Yi-He Gao
- Xinxiang University Affiliated Middle School, Xinxiang, 453000, China
| | - Li Jiang
- Cardiovascular Laboratory of Xinxiang, Xinxiang, 453003, China
| | - Jing Zhang
- Puyang Medical College, Puyang, 457000, China
| | | | - Tong Liu
- Puyang Medical College, Puyang, 457000, China
| |
Collapse
|
3
|
Yang W, Li Y, Feng R, Liang P, Tian K, Hu L, Wang K, Qiu T, Zhang J, Sun X, Yao X. PFOS causes lysosomes-regulated mitochondrial fission through TRPML1-VDAC1 and oligomerization of MCU/ATP5J2. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137685. [PMID: 39983639 DOI: 10.1016/j.jhazmat.2025.137685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/26/2025] [Accepted: 02/19/2025] [Indexed: 02/23/2025]
Abstract
Perfluorooctane sulfonate (PFOS), a listed persistent organic pollutant, poses risks to human health and is closely linked to chronic metabolic diseases. Although the role of mitochondrial fission in these diseases has garnered attention, whether and how PFOS induces mitochondrial fission remains obscure. Here, we found that PFOS induced mitochondrial fission, as demonstrated by the fragmentation of mitochondria and the upregulation of dynamin-related protein 1 (DRP1), phospho-DRP1 and mitochondrial fission protein 1 (FIS1) in human hepatocytes MIHA and mice liver. Blocking the calcium transfer from lysosomes to mitochondria that was executed by transient receptor potential mucolipin 1 (TRPML1) of lysosomes and voltage-dependent anion channel 1 (VDAC1) of mitochondria, did not affect PFOS-induced mitochondrial fission. In contrast, knockdown of TRPML1 or VDAC1 reversed this process. Knockdown of mitochondrial calcium uniporter (MCU), rather than inhibiting its activity, effectively alleviated PFOS-induced mitochondrial fission. Additionally, PFOS increased MCU oligomers without affecting MCU monomer. Inhibiting autophagy reversed the MCU oligomerization. Further investigation unveiled the interactions of MCU with VDAC1, TRPML1, mitochondrial Fo complex subunit F2 (ATP5J2) and DRP1 in PFOS-exposed mice liver and MIHA cells. We also discovered that knockdown of ATP5J2 alleviated PFOS-induced mitochondrial fission. Ulteriorly, PFOS upregulated ATP5J2 that underwent oligomerization. Knockdown of MCU reversed the increase in ATP5J2. Our study uncovers the presence and molecular basics of lysosomes-regulated mitochondrial fission under PFOS exposure, explains the regulatory pathways on MCU and ATP5J2 oligomerization and their pivotal roles in mitochondrial fission, highlighting the involvement of mitochondrial fission in PFOS-related health risks.
Collapse
Affiliation(s)
- Wei Yang
- Occupational and Environmental Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian 116044, China
| | - Yu Li
- Occupational and Environmental Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian 116044, China
| | - Ruzhen Feng
- Occupational and Environmental Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian 116044, China
| | - Peiyao Liang
- Occupational and Environmental Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian 116044, China
| | - Kefan Tian
- Occupational and Environmental Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian 116044, China
| | - Lingli Hu
- Occupational and Environmental Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian 116044, China
| | - Kejing Wang
- Occupational and Environmental Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian 116044, China
| | - Tianming Qiu
- Occupational and Environmental Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian 116044, China
| | - Jingyuan Zhang
- Occupational and Environmental Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian 116044, China
| | - Xiance Sun
- Occupational and Environmental Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian 116044, China
| | - Xiaofeng Yao
- Occupational and Environmental Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian 116044, China.
| |
Collapse
|
4
|
Luo H, Jin M, Hu H, Ying Q, Hu P, Sheng W, Huang Y, Xu K, Lu C, Zhang X. SIRT4 Protects Müller Glial Cells Against Apoptosis by Mediating Mitochondrial Dynamics and Oxidative Stress. Mol Neurobiol 2025; 62:6683-6702. [PMID: 39023793 DOI: 10.1007/s12035-024-04349-4] [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: 01/01/2024] [Accepted: 07/03/2024] [Indexed: 07/20/2024]
Abstract
SIRT4 is a member of the sirtuin family, which is related to mitochondrial function and possesses antioxidant and regulatory redox effects. Currently, the roles of SIRT4 in retinal Müller glial cells, oxidative stress, and mitochondrial function are still unclear. We confirmed, by immunofluorescence staining, that SIRT4 is located mainly in the mitochondria of retinal Müller glial cells. Using flow cytometry and Western blotting, we analyzed cell apoptosis, intracellular reactive oxygen species (ROS) levels, apoptotic and proapoptotic proteins, mitochondrial dynamics-related proteins, and mitochondrial morphology and number after the overexpression and downregulation of SIRT4 in rMC-1 cells. Neither the upregulation nor the downregulation of SIRT4 alone affected apoptosis. SIRT4 overexpression reduced intracellular ROS, reduced the BAX/BCL2 protein ratio, and increased the L-OPA/S-OPA1 ratio and the levels of the mitochondrial fusion protein MFN2 and the mitochondrial cleavage protein FIS1, increasing mitochondrial fusion. SIRT4 downregulation had the opposite effect. Mitochondria tend to divide after serum starvation for 24 h, and SIRT4 downregulation increases mitochondrial fragmentation and oxidative stress, leading to aggravated cell damage. The mitochondrial division inhibitor Mdivi-1 reduced oxidative stress levels and thus reduced cell damage caused by serum starvation. The overexpression of SIRT4 in rMC-1 cells reduced mitochondrial fragmentation caused by serum starvation, leading to mitochondrial fusion and reduced expression of cleaved caspase-3, thus alleviating the cellular damage caused by oxidative stress. Thus, we speculate that SIRT4 may protect retinal Müller glial cells against apoptosis by mediating mitochondrial dynamics and oxidative stress.
Collapse
Affiliation(s)
- Hongdou Luo
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Ming Jin
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Haijian Hu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Qian Ying
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Piaopiao Hu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Weiwei Sheng
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Yi Huang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Ke Xu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Chuming Lu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Xu Zhang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China.
| |
Collapse
|
5
|
Liu M, Qiao H. Role and mechanism of mitochondrial dysfunction‑related gene biomarkers in the progression of type 2 diabetes mellitus. Mol Med Rep 2025; 31:158. [PMID: 40211718 PMCID: PMC12004108 DOI: 10.3892/mmr.2025.13523] [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: 10/17/2024] [Accepted: 02/19/2025] [Indexed: 04/19/2025] Open
Abstract
The present study aimed to elucidate the roles and mechanisms of gene biomarkers associated with mitochondrial dysfunction in the progression of Type 2 diabetes mellitus (T2DM). It conducted an analysis of differentially expressed genes related to mitochondrial dysfunction in T2DM and employed bioinformatics approaches to predict potential target drugs for key biomarkers. Additionally, the present study used the EPIC algorithm to examine immune cell infiltration in T2DM. Furthermore, the single‑cell RNA sequencing dataset GSE221156 was analyzed to identify specific cell types involved in T2DM. The expression of biomarkers was investigated through cellular experiments to assess the effect of marker genes on macrophage polarization. A total of five biomarker genes associated with T2DM were identified, namely ERAP2, HLA‑DQB1, HLA‑DRB5, MAP1B and OAS3. The combined detection of these genes yielded a risk‑predictive area under the curve value of 0.833 for T2DM. These five marker genes may serve as potential targets for valproic acid (VPA). During the progression of T2DM, there is an increase in macrophage numbers, with these genes being highly expressed in macrophages. In a high glucose‑induced RAW264.7 macrophage model, the expressions of MAP1B and OAS3 were upregulated. Notably, the knockdown of OAS3 markedly reduced M1 macrophage polarization, indicating OAS3 facilitates M1 macrophage polarization in a high‑glucose environment. The downregulation of OAS3 expression attenuated M1 macrophage polarization by inhibiting mTORC activation. In conclusion, five candidate biomarkers for T2DM were identified that may serve as therapeutic targets for VPA and are associated with immune infiltration in T2DM. Among these, OAS3 enhances M1 macrophage polarization in a high‑glucose environment by regulating the mTORC1 pathway.
Collapse
Affiliation(s)
- Mengxue Liu
- Department of Endocrinology, The Fourth Hospital of Harbin, Harbin, Heilongjiang 150026, P.R. China
| | - Hong Qiao
- Department of Endocrinology and Metabolism, The Second Affliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
6
|
Shi H, Yang SA, Bai LY, Du JJ, Wu Z, He ZH, Liu H, Cui JY, Zhao M. Mechanism of myocardial damage induced by doxorubicin via calumenin-regulated mitochondrial dynamics and the calcium–Cx43 pathway. World J Cardiol 2025; 17:104839. [DOI: 10.4330/wjc.v17.i5.104839] [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: 01/10/2025] [Revised: 04/01/2025] [Accepted: 05/07/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND The clinical application of doxorubicin (DOX) is limited by its potential to cause cardiac cardiotoxicity.
AIM To investigate the correlation between calumenin (CALU) and mitochondrial kinetic-related proteins in rats with DOX cardiomyopathy.
METHODS A rat model of DOX-induced cardiomyopathy was used to evaluate the effects of DOX. We observed the effect of DOX on electrical conduction in cardiomyocytes using the electromapping technique. Masson staining was performed to evaluate myocardium fibrosis. Electron microscopy was used to observe the changes in pathological ultrastructure of the myocardium. Western blotting and ELISAs were performed to detect protein levels and intracellular free Ca2+ concentration.
RESULTS DOX slowed conduction and increased conduction dispersion in cardiomyocytes. The myocardial pathology in rats treated with DOX exhibited a significant deterioration, as demonstrated by an increase in mitochondrial Ca2+ concentration and a decrease in the expression of CALU, optic atrophy-1, and Bcl-2. Additionally, there was an increase in the expression of connexin 43 (Cx43) and the mitochondrial mitotic proteins dynamin-related protein 1, CHOP, Cytochrome C, and Bax in DOX rats. Decreased expression of CALU in cardiomyocytes triggered an increase in cytoplasmic free calcium concentration, which would normally be taken up by mitochondria, but decreased expression of mitochondrial outer membrane fusion proteins triggered a decrease in mitochondrial Ca2+ uptake, and the increase in cytoplasmic free calcium concentration triggered cell apoptosis.
CONCLUSION Increased cytoplasmic free calcium ion concentration induces calcium overload in ventricular myocytes, leading to decreased Cx43 protein, slowed conduction in myocytes, and increased conduction dispersion, resulting in arrhythmias.
Collapse
Affiliation(s)
- He Shi
- Department of Cardiovascular Medicine, Affiliated Hospital of Beihua University, Jilin Province 132000, China
| | - Song-Ao Yang
- Department of Biological Sciences, Inner Mongolia Minzu University, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Ling-Yu Bai
- Department of Cardiovascular Medicine, Affiliated Hospital of Inner Mongolia Minzu University, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Jian-Jun Du
- Department of Cardiovascular Medicine, The First People's Hospital of Horqin District, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Zhe Wu
- Department of Cardiovascular Medicine, Tongliao Municipal Hospital, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Zhi-Hui He
- Department of Human Anatomy, Histology and Embryology, Inner Mongolia Minzu University, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Hao Liu
- Section of Anatomy, Inner Mongolia Minzu University, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Jia-Yue Cui
- College of Basic Medical Sciences, Jilin University, Changchun 130000, Jilin Province, China
| | - Ming Zhao
- Department of Cardiovascular Medicine, Affiliated Hospital of Inner Mongolia Minzu University, Tongliao 028000, Inner Mongolia Autonomous Region, China
| |
Collapse
|
7
|
Zhao J, Shu Z, Li X, Zhang W, Sun M, Song W, Cheng H, Shi S. Dehydrodiisoeugenol alleviates palmitate-induced mitochondrial dysfunction in human vascular smooth muscle cells through the activation of SIRT1-mediated Drp1 deacetylation. Lipids Health Dis 2025; 24:187. [PMID: 40413480 DOI: 10.1186/s12944-025-02611-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 05/14/2025] [Indexed: 05/27/2025] Open
Abstract
OBJECTIVE Dehydrodiisoeugenol (Deh) has demonstrated positive effects in the prevention and treatment of cardiovascular disease (CVD) caused by lipid overload, but its specific mechanism of action remains poorly understood. The aim of this study was to investigate the possible mechanisms by which Deh modulates the mitochondrial dysfunction induced by palmitate (PA) in vascular smooth muscle cells (VSMCs). METHODS A PA-induced high-fat model of VSMCs was established, and the effect of PA on the VSMCs on function was detected by evaluating the oxidative stress and apoptosis of cells, as well as mitochondrial function. The expression of dynamin-related protein 1 (Drp1) was detected by immunofluorescence and immunoprecipitation. The key targets of Deh for the treatment of mitochondria-related diseases were screened by bioinformatics analysis and molecular docking techniques. Finally, the role of Silent information regulator 1 (SIRT1) in the treatment of PA-induced mitochondrial dysfunction in VSMCs by Deh was explored by administrating Deh as well as SIRT1 activator (CAY10602, CAY) and SIRT1 inhibitor (JGB1741, JGB). RESULTS The results showed that PA concentration-dependently increased oxidative stress and apoptosis in VSMCs, while modulating the acetylation of Drp1, promoting its expression and mitochondrial ectopia, thereby inducing mitochondrial dysfunction. Bioinformatics analysis and molecular docking indicated that SIRT1 may be a key target of Deh for the treatment of mitochondria-related diseases. Follow-up experiments revealed that Deh significantly inhibited PA-induced mitochondrial dysfunction in VSMCs by suppressing acetylation and expression of Drp1 and reducing mitochondrial ectasia, an effect that was achieved by regulating SIRT1. CONCLUSION Deh was able to inhibit Drp1 expression and mitochondrial ectopia by reducing Drp1 acetylation through activation of SIRT1, thereby inhibiting PA-induced mitochondrial dysfunction effects in VSMCs, ameliorating pathological processes, such as cellular oxidative stress and apoptosis, and maintaining stable cellular functions.
Collapse
Affiliation(s)
- Jianjun Zhao
- Department of Respiratory Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130000, China
| | - Zhiyun Shu
- Department of Respiratory Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130000, China
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130000, China
| | - Xiangjun Li
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130000, China
| | - Wenqing Zhang
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130000, China
| | - Mengze Sun
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130000, China
| | - Wenxiao Song
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130000, China
| | - Hongyuan Cheng
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130000, China
| | - Shaomin Shi
- Department of Respiratory Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130000, China.
| |
Collapse
|
8
|
Araújo R, Bernardino RL, Monteiro MP, Gomes PS. Unveiling metabolic pathways in the hyperglycemic bone: bioenergetic and proteomic analysis of the bone tissue exposed to acute and chronic high glucose. Mol Med 2025; 31:194. [PMID: 40382540 DOI: 10.1186/s10020-025-01251-0] [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: 02/14/2025] [Accepted: 05/07/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Bone fragility due to poor glycemic control is a recognized complication of diabetes, but the mechanisms underlying diabetic bone disease remain poorly understood. Despite the importance of bioenergetics in tissue functionality, the impact of hyperglycemia on bone bioenergetics has not been previously investigated. OBJECTIVE To determine the effects of high glucose exposure on energy metabolism and structural integrity in bone tissue using an ex vivo organotypic culture model of embryonic chick femur. METHODS Femora from eleven-day-old Gallus gallus embryos were cultured for eleven days under physiological glucose conditions (5.5 mM, NG), chronic high glucose exposure (25 mM, HG-C), or acute high glucose exposure (25 mM, HG-A). Bioenergetic assessments (Seahorse assays), proteomic analysis (liquid chromatography-mass spectrometry), histomorphometric and microtomographic evaluations, and oxidative stress measurements (carbonyl content assay) were performed. Statistical analyses were conducted using IBM® SPSS® Statistics (v26.0). The Mann-Whitney nonparametric test was used for group comparisons in microtomographic analysis, ALP activity, and carbonyl content assays. For Seahorse assay results, ANOVA with Tukey's post-hoc test was applied after confirming data homoscedasticity with Levene's test. RESULTS Chronic high glucose exposure reduced bone mineral deposition, altered histomorphometric indices, and suppressed key osteochondral development regulators. Acute high glucose exposure enhanced glycolysis and oxidative phosphorylation, while chronic exposure caused oxygen consumption uncoupling, increased ROS generation, and downregulated mitochondrial proteins critical for bioenergetics. Elevated oxidative stress was confirmed in the chronic high glucose group. CONCLUSION Chronic high glucose exposure disrupted bone bioenergetics, induced mitochondrial dysfunction, and compromised bone structural integrity, emphasizing the metabolic impact of hyperglycemia in diabetic bone disease.
Collapse
Affiliation(s)
- Rita Araújo
- BoneLab, Faculdade de Medicina Dentária, Universidade do Porto, Rua Dr. Manuel Pereira da Silva, 4200-393, Porto, Portugal
- LAQV/REQUIMTE, Faculdade de Medicina Dentária, Universidade do Porto, Rua Dr. Manuel Pereira da Silva, 4200-393, Porto, Portugal
- Endocrine and Metabolic Research, UMIB-Unit for Multidisciplinary Research in Biomedicine, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
| | - Raquel L Bernardino
- Endocrine and Metabolic Research, UMIB-Unit for Multidisciplinary Research in Biomedicine, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Porto, Portugal
| | - Mariana P Monteiro
- Endocrine and Metabolic Research, UMIB-Unit for Multidisciplinary Research in Biomedicine, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Porto, Portugal
| | - Pedro S Gomes
- BoneLab, Faculdade de Medicina Dentária, Universidade do Porto, Rua Dr. Manuel Pereira da Silva, 4200-393, Porto, Portugal.
- LAQV/REQUIMTE, Faculdade de Medicina Dentária, Universidade do Porto, Rua Dr. Manuel Pereira da Silva, 4200-393, Porto, Portugal.
| |
Collapse
|
9
|
Fang X, Shao Z, Ding H, Xu H, Tu Z, Wang H, Li D, Huang C, Jiang C. Urolithin A enhances diabetic wound healing: Insights from parkin-mediated mitophagy in endothelial progenitor cells. Int Immunopharmacol 2025; 155:114572. [PMID: 40203794 DOI: 10.1016/j.intimp.2025.114572] [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: 10/11/2024] [Revised: 02/24/2025] [Accepted: 03/26/2025] [Indexed: 04/11/2025]
Abstract
Diabetes is often associated with delayed wound healing, where endothelial progenitor cells (EPCs) play a key role in maintaining vascular integrity and promoting angiogenesis. Urolithin A, a metabolite derived from pomegranates, strawberries, and nuts, has demonstrated therapeutic potential in reversing damage in various disease models, indicating its potential in facilitating diabetic wound healing. In this study, we investigated the effects of Urolithin A on mitochondrial dysfunction, apoptosis, and impaired function in EPCs treated with high glucose. Through sequencing and molecular docking analysis, we found that Urolithin A exerts its therapeutic action by upregulating Parkin and activating mitophagy. Furthermore, Urolithin A alleviated delayed wound healing in diabetic rat models. In conclusion, Urolithin A holds promise as a therapeutic agent for improving diabetes-related delayed wound healing by targeting mitochondrial dysfunction and enhancing EPC function.
Collapse
Affiliation(s)
- Xia Fang
- Department of Arthroplasty, The First People's Hospital of Wenling, The Affiliated Wenling Hospital of Wenzhou Medical University, Taizhou, Zhejiang 317500, China; Department of Plastic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310017, China
| | - Zhenxuan Shao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310017, China
| | - Hongfeng Ding
- Department of Arthroplasty, The First People's Hospital of Wenling, The Affiliated Wenling Hospital of Wenzhou Medical University, Taizhou, Zhejiang 317500, China
| | - Haoxiang Xu
- Department of Arthroplasty, The First People's Hospital of Wenling, The Affiliated Wenling Hospital of Wenzhou Medical University, Taizhou, Zhejiang 317500, China
| | - Zhuolong Tu
- Department of Burn, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Hui Wang
- Department of Plastic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310017, China
| | - Dawei Li
- Department of Plastic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310017, China.
| | - Cheng Huang
- Department of Plastic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310017, China.
| | - Chang Jiang
- Department of Arthroplasty, The First People's Hospital of Wenling, The Affiliated Wenling Hospital of Wenzhou Medical University, Taizhou, Zhejiang 317500, China.
| |
Collapse
|
10
|
Wu G, Wang X, Dong H, Yu J, Li T, Wang X. Coix Seed Oil Alleviates Hyperuricemia in Mice by Ameliorating Oxidative Stress and Intestinal Microbial Composition. Nutrients 2025; 17:1679. [PMID: 40431419 PMCID: PMC12114407 DOI: 10.3390/nu17101679] [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: 04/16/2025] [Revised: 05/09/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Coix seed oil (YRO), rich in unsaturated fatty acids, has emerged as a promising intervention for hyperuricemia (HUA) due to its potential to alleviate oxidative damage and support organ health. Methods: The fatty acid composition of YRO was determined by gas chromatography-mass spectrometry (GC-MS). A HUA mouse model was established, and serum markers and hepatic enzymes were evaluated. Renal mitochondrial function was assessed using immunohistochemistry and immunofluorescence, and urate transporter expression, along with key signaling proteins, was quantified by Western blot analysis. Additionally, gut microbiota composition was analyzed, and non-targeted metabolomics was performed to observe alterations in serum lipid metabolites. Results: YRO significantly reduced serum uric acid (UA) levels and normalized hepatic enzyme activities. Histological evaluation revealed less tissue damage in both the kidney and the intestine. In the kidney, YRO improved mitochondrial function and supported antioxidant defenses via regulation of Keap1/Nrf2 signaling. In the intestine, YRO enhanced barrier integrity by increasing ZO-1, Occludin, and Claudin-1 expression. Moreover, YRO modulated gut microbiota by increasing beneficial bacteria (Muribaculaceae, Prevotellaceae UCG-001, Lachnospiraceae_ NK4A136_group, Akkermansia) while suppressing harmful species (Bacteroides, Dubosiella). Lipid metabolomics indicated a restoration of phospholipid balance through modulation of the PI3K/AKT/mTOR pathway. Conclusions: YRO supported metabolic health by promoting UA homeostasis, enhancing mitochondrial function, reinforcing antioxidant capacity, and maintaining gut integrity. These findings suggest that coix seed oil could serve as a nutritional supplement in managing HUA and related metabolic disturbances.
Collapse
Affiliation(s)
- Guozhen Wu
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xinming Wang
- Institute of Chinese Materia Medica Chemistry, Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Hongjing Dong
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Jinqian Yu
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Tao Li
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Xiao Wang
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| |
Collapse
|
11
|
de Souza MC, Agneis MLG, das Neves KA, de Almeida MR, Feltran GDS, Souza Cruz EM, Schoffen JPF, Chuffa LGDA, Seiva FRF. Melatonin Improves Lipid Homeostasis, Mitochondrial Biogenesis, and Antioxidant Defenses in the Liver of Prediabetic Rats. Int J Mol Sci 2025; 26:4652. [PMID: 40429795 PMCID: PMC12111231 DOI: 10.3390/ijms26104652] [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: 03/14/2025] [Revised: 04/25/2025] [Accepted: 04/29/2025] [Indexed: 05/29/2025] Open
Abstract
Type 2 diabetes mellitus represents a major global health burden and is often preceded by a prediabetic state characterized by insulin resistance and metabolic dysfunction. Mitochondrial alterations, oxidative stress, and disturbances in lipid metabolism are central to the prediabetes pathophysiology. Melatonin, a pleiotropic indolamine, is known to regulate metabolic and mitochondrial processes; however, its therapeutic potential in prediabetes remains poorly understood. This study investigated the effects of melatonin on energy metabolism, oxidative stress, and mitochondrial function in a rat model of prediabetes induced by chronic sucrose intake and low-dose streptozotocin administration. Following prediabetes induction, animals were treated with melatonin (20 mg/kg) for four weeks. Biochemical analyses were conducted to evaluate glucose and lipid metabolism, and mitochondrial function was assessed via gene expression, enzymatic activity, and oxidative stress markers. Additionally, hepatic mitochondrial dynamics were examined by quantifying key regulators genes associated with biogenesis, fusion, and fission. Prediabetic animals exhibited dyslipidemia, hepatic lipid accumulation, increased fat depots, and impaired glucose metabolism. Melatonin significantly reduced serum glucose, triglycerides, and total cholesterol levels, while enhancing the hepatic high-density lipoprotein content. It also stimulated β-oxidation by upregulating hydroxyacyl-CoA dehydrogenase and citrate synthase activity. Mitochondrial dysfunction in prediabetic animals was evidenced by the reduced expression of peroxisome proliferator-activated receptor gamma coactivator-1 alpha and mitochondrial transcription factor A, both of which were markedly upregulated by melatonin. The indolamine also modulated mithocondrial dynamics by regulating fusion and fission markers, including mitosuin 1 and 2, optic atrophy protein, and dynamin-related protein. Additionally, melatonin mitigated oxidative stress by enhancing the activity of superoxide dismutase and catalase while reducing lipid peroxidation. These findings highlight melatonin's protective role in prediabetes by improving lipid and energy metabolism, alleviating oxidative stress, and restoring mitochondrial homeostasis. This study provides novel insights into the therapeutic potential of melatonin in addressing metabolic disorders, particularly in mitigating mitochondrial dysfunction associated with prediabetes.
Collapse
Affiliation(s)
- Milena Cremer de Souza
- Department of Parasitology, Immunology and General Pathology, State University of Londrina (UEL), Londrina 86057-970, Paraná, Brazil
- Department of Chemistry and Biochemistry, São Paulo State University (UNESP), Botucatu 18618-693, São Paulo, Brazil
| | - Maria Luisa Gonçalves Agneis
- Department of Chemistry and Biochemistry, São Paulo State University (UNESP), Botucatu 18618-693, São Paulo, Brazil
| | - Karoliny Alves das Neves
- Department of Chemistry and Biochemistry, São Paulo State University (UNESP), Botucatu 18618-693, São Paulo, Brazil
| | - Matheus Ribas de Almeida
- Department of Chemistry and Biochemistry, São Paulo State University (UNESP), Botucatu 18618-693, São Paulo, Brazil
| | - Geórgia da Silva Feltran
- Department of Chemistry and Biochemistry, São Paulo State University (UNESP), Botucatu 18618-693, São Paulo, Brazil
| | - Ellen Mayara Souza Cruz
- Department of Parasitology, Immunology and General Pathology, State University of Londrina (UEL), Londrina 86057-970, Paraná, Brazil
| | - João Paulo Ferreira Schoffen
- Center of Biological Sciences, State University of Northern Paraná (UENP), Bandeirantes 86360-000, Paraná, Brazil
| | | | | |
Collapse
|
12
|
Xie L, Cao J, Xu Y, Yang Q, Chang W, Song L, Sun Y. Mechanistic study of modulating mitochondrial fission and fusion to ameliorate neuropathic pain in mice. Sci Rep 2025; 15:15571. [PMID: 40320455 PMCID: PMC12050293 DOI: 10.1038/s41598-025-99300-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/18/2025] [Indexed: 05/08/2025] Open
Abstract
The emergence of neuropathic pain is significantly influenced by the impairment of mitochondrial processes. Ensuring the stability of mitochondrial activity requires a delicate equilibrium between the processes of mitochondrial fission and fusion. However, the specific alterations in mitochondrial activity across different models of neuropathic pain and the underlying mechanisms remain largely unclear. We developed a persistent compression injury (CCI) model targeting the sciatic nerve in mice. CCI induced pain like behaviors in mice, which were associated with increased levels of dynamin related protein 1 (Drp1) and decreased expression of the fusion protein OPA1 and an increase in the percentage of DRG nerve cell mitochondria in the fission form, and a decrease in percentage in the fusion form. Ultrastructural analysis showed that mitochondria in CCI mice were smaller in perimeter and area, adopting a more circular shape. Overexpression of OPA1 mediated by AAV attenuated pain hypersensitivity, lowered oxidative stress, and expanded mitochondrial circumference and area. Mdivi-1 treatment reduced pain, whereas blocking fusion with MYLS22 augmented pain and oxidative stress and further led to increased mitochondrial fragmentation. Our results illustrate that Mitochondria in DRG nerve cell are highly sensitive to neuropathic pain. Modulating mitochondrial fission and fusion through targeted gene overexpression and pharmacological inhibitors restores mitochondrial dynamics, reduces oxidative stress, and alleviates neuropathic pain in mice. These findings position mitochondrial dynamics as promising therapeutic targets for pain management.
Collapse
Affiliation(s)
- Liu Xie
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, China
| | - Jing Cao
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, China
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450015, China
| | - Qingqing Yang
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, China
- Xinyang Central Hospital, Xinyang, 464000, China
| | - Wanting Chang
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, China
| | - Linna Song
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, China
| | - Yanyan Sun
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou, 450001, China.
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
13
|
Fu X, Yu B, Lu L, Han Y, Liu Y, Zhang J, Chen T, Yu D. An injectable and photocurable methacrylate-silk fibroin/prussian blue nanozyme hydrogel with antioxidant and pyroptosis suppression properties for cartilage regeneration. Int J Biol Macromol 2025; 307:142154. [PMID: 40118414 DOI: 10.1016/j.ijbiomac.2025.142154] [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: 10/21/2024] [Revised: 12/15/2024] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Cartilage injury is one of the most prevalent, distressing, and disabling chronic conditions affecting degenerative joints worldwide; however, its underlying mechanisms remain elusive. Various biomaterials have been widely employed in the treatment of articular cartilage (AC) injuries. Despite these efforts, the key role of reactive oxygen species (ROS) as primary instigators of pyroptosis, combined with the lack of effective interventions, often results in suboptimal cartilage repair outcomes. Thus, there is an urgent need for the development of novel antioxidants and inflammasome-mediated pyroptosis modulators to create a favorable microenvironment for cartilage repair. In this study, we synthesized Prussian blue nanoparticles (PBNPs) capable of efficiently scavenging ROS. In vitro, these PBNPs protected against oxidative stress-induced cytotoxicity, preserved mitochondrial integrity, reduced the activation of nucleotide-binding domain and leucine-rich repeat family pyrin domain-containing 3 (NLRP3) inflammasomes and caspase-1, and subsequently downregulated gasdermin D (GSDMD) cleavage and inflammatory factor production, leading to the inhibition of chondrocyte pyroptosis. To extend these findings in vivo, we developed an injectable and photocurable methacrylate-silk fibroin (SilMA) hydrogel with homogeneously incorporated PBNPs, designed for releasing PBNPs. The resulting PBNPs@SilMA hydrogel decreased ROS production, reduced chondrocyte pyroptosis, and supported chondrocyte proliferation and matrix secretion, subsequently improved AC repair. Overall, our results indicate that the PBNPs@SilMA platform holds significant promise as a therapeutic strategy for AC injury.
Collapse
Affiliation(s)
- Xuefei Fu
- Department of Orthopedics, Anhui No.2 Provincial People's Hospital, Hefei 230041, China
| | - Bing Yu
- Department of Orthopedics, Anhui No.2 Provincial People's Hospital, Hefei 230041, China
| | - Lin Lu
- Department of Radiotherapy, Anhui No.2 Provincial People's Hospital, Hefei 230041, China
| | - Yafei Han
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Yancheng Liu
- Department of Bone Tumor and Soft Tissue Oncology, Tianjin Hospital, Tianjin University, Tianjin 300211, China
| | - Jingyu Zhang
- Department of Bone Tumor and Soft Tissue Oncology, Tianjin Hospital, Tianjin University, Tianjin 300211, China.
| | - Tao Chen
- Department of Orthopedics, Anhui No.2 Provincial People's Hospital, Hefei 230041, China.
| | - Defu Yu
- Department of Orthopedics, Anhui No.2 Provincial People's Hospital, Hefei 230041, China; School of Clinical Medicine, Anhui Medical College, China.
| |
Collapse
|
14
|
Li J, Lin Q, Ren C, Li X, Li X, Li H, Li S. The perspective of modern transplant science - transplant arteriosclerosis: inspiration derived from mitochondria associated endoplasmic reticulum membrane dysfunction in arterial diseases. Int J Surg 2025; 111:3430-3440. [PMID: 40146783 DOI: 10.1097/js9.0000000000002362] [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] [Accepted: 03/16/2025] [Indexed: 03/29/2025]
Abstract
The mitochondria-associated endoplasmic reticulum membrane (MAM) is a crucial structure connecting mitochondria and the endoplasmic reticulum (ER), regulating intracellular calcium homeostasis, lipid metabolism, and various signaling pathways essential for arterial health. Recent studies highlight MAM's significant role in modulating vascular endothelial cells (EC) and vascular smooth muscle cells (VSMC), establishing it as a key regulator of arterial health and a contributor to vascular disease pathogenesis. Organ transplantation is the preferred treatment for end-stage organ failure, but transplant arteriosclerosis (TA) can lead to chronic transplant dysfunction, significantly impacting patient survival. TA, like other vascular diseases, features endothelial dysfunction and abnormal proliferation and migration of VSMC. Previous research on TA has focused on immune factors; the pathological and physiological changes in grafts following immune system attacks have garnered insufficient attention. For example, the potential roles of MAM in TA have not been thoroughly investigated. Investigating the relationship between MAM and TA, as well as the mechanisms behind TA progression, is essential. This review aims to outline the fundamental structure and the primary functions of MAM, summarize its key molecular regulators of vascular health, and explore future prospects for MAM in the context of TA research, providing insights for both basic research and clinical management of TA.
Collapse
Affiliation(s)
- Jingyi Li
- Department of Urology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Qian Lin
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chao Ren
- Department of Urology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Xiaodong Li
- Department of Urology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Xiaowei Li
- Department of Urology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Haofeng Li
- Department of Urology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Shadan Li
- Department of Urology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| |
Collapse
|
15
|
Ma Y, Lei C, Ye T, Wan Q, Wang K, Zhu Y, Li L, Liu X, Niu L, Tay FR, Mu Z, Jiao K, Niu L. Silicon Enhances Functional Mitochondrial Transfer to Improve Neurovascularization in Diabetic Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415459. [PMID: 40125794 PMCID: PMC12097102 DOI: 10.1002/advs.202415459] [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] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/15/2025] [Indexed: 03/25/2025]
Abstract
Diabetes mellitus is a metabolic disorder associated with an increased risk of fractures and delayed fracture healing, leading to a higher prevalence of bone defects. Recent advancements in strategies aim at regulating immune responses and enhancing neurovascularization have not met expectations. This study demonstrates that a silicon-based strategy significantly enhances vascularization and innervation, thereby optimizing the repair of diabetic bone defects. Silicon improves mitochondrial function and modulates mitochondrial fission dynamics in macrophages via the Drp1-Mff signaling pathway. Subsequently, functional mitochondria are transferred from macrophages to endothelial and neuronal cells through microvesicles, providing a protective mechanism for blood vessels and peripheral nerves during early wound healing. On this basis, an optimized strategy combining a silicified collagen scaffold with a Drp1-Fis1 interaction inhibitor is used to further regulate mitochondrial fission in macrophages and enhance the trafficking of functional mitochondria into stressed receptor cells. In diabetic mice with critical-sized calvarial defects, the silicon-based treatment significantly promotes vessel formation, nerve growth, and mineralized tissue development. These findings provide therapeutic insights into the role of silicon in promoting diabetic bone regeneration and highlight the importance of intercellular communication in diabetic conditions.
Collapse
Affiliation(s)
- Yu‐Xuan Ma
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Chen Lei
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Tao Ye
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Qian‐Qian Wan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Kai‐Yan Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Yi‐Na Zhu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Ling Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Xu‐Fang Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Long‐Zhang Niu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Franklin R. Tay
- The Dental College of GeorgiaAugusta UniversityAugustaGA30912USA
| | - Zhao Mu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Kai Jiao
- Department of StomatologyTangdu hospitalState Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of Stomatology & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| | - Li‐Na Niu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| |
Collapse
|
16
|
Xi Y, Tao K, Wen X, Feng D, Mai Z, Ding H, Mao H, Wang M, Yang Q, Xiang J, Zhang J, Wu S. SIRT3-Mediated Deacetylation of DRP1 K711 Prevents Mitochondrial Dysfunction in Parkinson's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411235. [PMID: 39976201 PMCID: PMC12061286 DOI: 10.1002/advs.202411235] [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] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/08/2025] [Indexed: 02/21/2025]
Abstract
Dysregulation of mitochondrial dynamics is a key contributor to the pathogenesis of Parkinson's disease (PD). Aberrant mitochondrial fission induced by dynamin-related protein 1 (DRP1) causes mitochondrial dysfunction in dopaminergic (DA) neurons. However, the mechanism of DRP1 activation and its role in PD progression remain unclear. In this study, Mass spectrometry analysis is performed and identified a significant increased DRP1 acetylation at lysine residue 711 (K711) in the mitochondria under oxidative stress. Enhanced DRP1K711 acetylation facilitated DRP1 oligomerization, thereby exacerbating mitochondrial fragmentation and compromising the mitochondrial function. DRP1K711 acetylation also affects mitochondrial DRP1 recruitment and fission independent of canonical S616 phosphorylation. Further analysis reveals the critical role of sirtuin (SIRT)-3 in deacetylating DRP1K711, thereby regulating mitochondrial dynamics and function. SIRT3 agonists significantly inhibit DRP1K711 acetylation, rescue DA neuronal loss, and improve motor function in a PD mouse model. Conversely, selective knockout of SIRT3 in DA neurons exacerbates DRP1K711 acetylation, leading to increased DA neuronal damage, neuronal death, and worsened motor dysfunction. Notably, this study identifies a novel mechanism involving aberrant SIRT3-mediated DRP1 acetylation at K711 as a key driver of mitochondrial dysfunction and DA neuronal death in PD, revealing a potential target for PD treatment.
Collapse
Affiliation(s)
- Ye Xi
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'anShaanxi710032China
| | - Kai Tao
- Department of Experimental SurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Xiaomin Wen
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'anShaanxi710032China
| | - Dayun Feng
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Zifan Mai
- Department of BiophysicsInstitute of NeuroscienceNHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang University School of MedicineHangzhou310058China
| | - Hui Ding
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'anShaanxi710032China
| | - Honghui Mao
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'anShaanxi710032China
| | - Mingming Wang
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'anShaanxi710032China
| | - Qian Yang
- Department of Experimental SurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Jie Xiang
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'anShaanxi710032China
| | - Jie Zhang
- Institute of NeuroscienceCollege of MedicineXiamen University XiamenFujian361105China
| | - Shengxi Wu
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'anShaanxi710032China
| |
Collapse
|
17
|
Liu G, Jia G, Ren Y, Yin C, Xiao X, Wu H, Liu J, Chen M. Mechanism of lncRNA gadd7 regulating mitofusin 1 expression by recruiting LSD1 to down-regulate H3K9me3 level, and mediating mitophagy in alveolar type II epithelial cell apoptosis in hyperoxia-induced acute lung injury. Cell Biol Toxicol 2025; 41:77. [PMID: 40301157 PMCID: PMC12041145 DOI: 10.1007/s10565-025-10021-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 04/13/2025] [Indexed: 05/01/2025]
Abstract
OBJECTIVE Hyperoxic exposure induces acute lung injury (ALI). We analyzed the mechanism of long non-coding RNA (lncRNA) growth-arrested DNA damage-inducible gene 7 (gadd7) regulating mitofusin 1 (MFN1) in Hyperoxia-induced ALI (HALI) type II alveolar epithelial cell (AEC II) apoptosis. METHODS The HALI rat model was generated using hyperoxic induction and treated with shRNA-gadd7 and rapamycin (Rapa), with ALI, apoptotic level, total protein concentration and total cell, neutrophil and macrophage counts assessed. The HALI cell model was developed on hyperoxia-induced RLE-6TN cells and processed with oe-MFN1, si-gadd7 and Rapa. Cell viability, apoptosis, TOM20/LC3BII co-localization, mitochondrial membrane potential (MMP), superoxide dismutase activity, malonaldehyde, reactive oxygen species (ROS), tumor necrosis factor-α, interleukin (IL)-10, IL-6, IL-1β, gadd7, MFN1, Cleaved caspase-3, Cleaved poly (ADP-ribose) polymerase, B-cell lymphoma-2 (Bcl-2), Bcl-2-associated X, LC3BI/II, lysine-specific demethylase 1 (LSD1), p62, and H3K9me3 protein levels were measured. gadd7-LSD1 interaction was predicted and verified by RPISeq database, RIP, and RNA pull-down assay. RESULTS In HALI rats, gadd7 was up-regulated in lung tissues, and gadd7 silencing alleviated oxidative stress, ALI and apoptosis. gadd7 knockdown inhibited oxidative stress and apoptosis though MFN1, and mediated mitophagy (evidenced by diminished LC3BII/LC3BI ratio, TOM20/LC3BII co-localization and ROS level, and elevated p62 level and MMP), which were reversed by mitophagy activation. By recruiting LSD1 to down-regulate H3K9me3 level and promote MFN1 expression, gadd7-mediated mitophagy affected ALI and apoptosis in HALI rats. CONCLUSION LncRNA gadd7 regulated MFN1 expression by recruiting LSD1 to down-regulate H3K9me3 level and mediate mitophagy, thereby promoting AEC II apoptosis in HALI.
Collapse
Affiliation(s)
- Guoyue Liu
- Intensive Care Unit, The Second Affiliated Hospital of Zunyi Medical University, Intersection of Xinlong Avenue and Xinpu Avenue, Xinpu New District, Zunyi, 563000, Guizhou, China
- Graduate School, Zunyi Medical University, No. 6, Xuefu West Road, Xinpu New District, Zunyi, 563000, Guizhou, China
| | - Guiyang Jia
- Intensive Care Unit, The Second Affiliated Hospital of Zunyi Medical University, Intersection of Xinlong Avenue and Xinpu Avenue, Xinpu New District, Zunyi, 563000, Guizhou, China
- Graduate School, Zunyi Medical University, No. 6, Xuefu West Road, Xinpu New District, Zunyi, 563000, Guizhou, China
| | - Yingcong Ren
- Graduate School, Zunyi Medical University, No. 6, Xuefu West Road, Xinpu New District, Zunyi, 563000, Guizhou, China
| | - Cunzhi Yin
- Intensive Care Unit, The Second Affiliated Hospital of Zunyi Medical University, Intersection of Xinlong Avenue and Xinpu Avenue, Xinpu New District, Zunyi, 563000, Guizhou, China
| | - Xuan Xiao
- Intensive Care Unit, The Second Affiliated Hospital of Zunyi Medical University, Intersection of Xinlong Avenue and Xinpu Avenue, Xinpu New District, Zunyi, 563000, Guizhou, China
| | - Hang Wu
- Intensive Care Unit, The Second Affiliated Hospital of Zunyi Medical University, Intersection of Xinlong Avenue and Xinpu Avenue, Xinpu New District, Zunyi, 563000, Guizhou, China
| | - Jun Liu
- Department of Preventive Medicine, School of Public Health, Zunyi Medical University, No. 6, Xuefu West Road, Xinpu New District, Zunyi, 563000, Guizhou, China.
| | - Miao Chen
- Intensive Care Unit, Affiliated Hospital of Zunyi Medical University, No.149, Dalian Road, Huichuan District, Zunyi, Guizhou, China.
| |
Collapse
|
18
|
Vandersmissen J, Dewachter I, Cuypers K, Hansen D. The Impact of Exercise Training on the Brain and Cognition in Type 2 Diabetes, and its Physiological Mediators: A Systematic Review. SPORTS MEDICINE - OPEN 2025; 11:42. [PMID: 40274715 PMCID: PMC12022206 DOI: 10.1186/s40798-025-00836-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/16/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Type 2 diabetes (T2DM) affects brain structure and function, and is associated with an increased risk of dementia and mild cognitive impairment. It is known that exercise training has a beneficial effect on cognition and brain structure and function, at least in healthy people, but the impact of exercise training on these aspects remains to be fully elucidated in patients with T2DM. OBJECTIVE To determine the impact of exercise training on cognition and brain structure and function in T2DM, and identify the involved physiological mediators. METHODS This paper systematically reviews studies that evaluate the effect of exercise training on cognition in T2DM, and aims to indicate the most beneficial exercise modality for improving or preserving cognition in this patient group. In addition, the possible physiological mediators and targets involved in these improvements are narratively described in the second part of this review. Papers published up until the 14th of January 2025 were searched by means of the electronic databases PubMed, Embase, and Web of Science. Studies directly investigating the effect of any kind of exercise training on the brain or cognition in patients with T2DM, or animal models thereof, were included, with the exception of human studies assessing cognition only at one time point, and studies combining exercise training with other interventions (e.g. dietary changes, cognitive training, etc.). Study quality was assessed by means of the TESTEX tool for human studies, and the CAMARADES tool for animal studies. RESULTS For the systematic part of the review, 22 papers were found to be eligible. 18 out of 22 papers (81.8%) showed a significant positive effect of exercise training on cognition in T2DM, of which two studies only showed significant improvements in the minority of the cognitive tests. Four papers (18.2%) could not find a significant effect of exercise on cognition in T2DM. Resistance and endurance exercise were found to be equally effective for achieving cognitive improvement. Machine-based power training is seemingly more effective than resistance training with body weight and elastic bands to reach cognitive improvement. In addition, BDNF, lactate, leptin, adiponectin, GSK3β, GLP-1, the AMPK/SIRT1 pathway, and the PI3K/Akt pathway were identified as plausible mediators directly from studies investigating the effect of exercise training on brain structure and function in T2DM. Via these mediators, exercise training induces multiple beneficial brain changes, such as increased neuroplasticity, increased insulin sensitivity, and decreased inflammation. CONCLUSION Overall, exercise training beneficially affects cognition and brain structure and function in T2DM, with resistance and endurance exercise having similar effects. However, there is a need for additional studies, and more methodological consistency between different studies in order to define an exercise program optimal for improving cognition in T2DM. Furthermore, we were able to define several mediators involved in the effect of exercise training on cognition in T2DM, but further research is necessary to unravel the entire process.
Collapse
Affiliation(s)
- Jitske Vandersmissen
- Faculty of Rehabilitation Sciences, REVAL - Rehabilitation Research Center, Hasselt University, Wetenschapspark 7, 3590, Diepenbeek, Belgium.
| | - Ilse Dewachter
- Biomedical Research Institute, BIOMED, Hasselt University, 3590, Diepenbeek, Belgium
| | - Koen Cuypers
- Faculty of Rehabilitation Sciences, REVAL - Rehabilitation Research Center, Hasselt University, Wetenschapspark 7, 3590, Diepenbeek, Belgium
- Movement Control and Neuroplasticity Research Group, Department of Movement Sciences, Group Biomedical Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Dominique Hansen
- Faculty of Rehabilitation Sciences, REVAL - Rehabilitation Research Center, Hasselt University, Wetenschapspark 7, 3590, Diepenbeek, Belgium
- Heart Centre Hasselt, Jessa Hospital, 3500, Hasselt, Belgium
| |
Collapse
|
19
|
Radovic M, Gartzke LP, Wink SE, van der Kleij JA, Politiek FA, Krenning G. Targeting the Electron Transport System for Enhanced Longevity. Biomolecules 2025; 15:614. [PMID: 40427507 PMCID: PMC12109555 DOI: 10.3390/biom15050614] [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: 03/20/2025] [Revised: 04/15/2025] [Accepted: 04/20/2025] [Indexed: 05/29/2025] Open
Abstract
Damage to mitochondrial DNA (mtDNA) results in defective electron transport system (ETS) complexes, initiating a cycle of impaired oxidative phosphorylation (OXPHOS), increased reactive oxygen species (ROS) production, and chronic low-grade inflammation (inflammaging). This culminates in energy failure, cellular senescence, and progressive tissue degeneration. Rapamycin and metformin are the most extensively studied longevity drugs. Rapamycin inhibits mTORC1, promoting mitophagy, enhancing mitochondrial biogenesis, and reducing inflammation. Metformin partially inhibits Complex I, lowering reverse electron transfer (RET)-induced ROS formation and activating AMPK to stimulate autophagy and mitochondrial turnover. Both compounds mimic caloric restriction, shift metabolism toward a catabolic state, and confer preclinical-and, in the case of metformin, clinical-longevity benefits. More recently, small molecules directly targeting mitochondrial membranes and ETS components have emerged. Compounds such as Elamipretide, Sonlicromanol, SUL-138, and others modulate metabolism and mitochondrial function while exhibiting similarities to metformin and rapamycin, highlighting their potential in promoting longevity. The key question moving forward is whether these interventions should be applied chronically to sustain mitochondrial health or intermittently during episodes of stress. A pragmatic strategy may combine chronic metformin use with targeted mitochondrial therapies during acute physiological stress.
Collapse
Affiliation(s)
| | | | | | | | | | - Guido Krenning
- Department of Clinical Pharmacy and Pharmacology, Section of Experimental Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (AP50), 9713 GZ Groningen, The Netherlands; (M.R.); (J.A.v.d.K.); (F.A.P.)
| |
Collapse
|
20
|
Hu T, Fang Z. Explore potential immune-related targets of leeches in the treatment of type 2 diabetes based on network pharmacology and machine learning. Front Genet 2025; 16:1554622. [PMID: 40296871 PMCID: PMC12036332 DOI: 10.3389/fgene.2025.1554622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder that poses a significant global health burden due to its profound effects on systemic physiological homeostasis. Without timely intervention, the disease can progress insidiously, leading to multisystem complications such as cardiovascular, renal, and neuropathic pathologies. Consequently, pharmacological intervention becomes crucial in managing the condition. Leeches have been traditionally used in Chinese medicine for their potential to inhibit the progression of T2DM and its associated complications; however, the specific mechanisms underlying their action and target pathways remain poorly understood. The objective of this study was to predict potential therapeutic targets of leeches in the treatment of T2DM. Methods We collected active components and targets associated with leeches from four online databases, while disease-related targets were sourced from the GeneCards and OMIM databases. Following this, we performed Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Gene expression data were obtained from the GSE184050 dataset. Important immune cell types were identified through immunoinfiltration analysis in conjunction with single sample enrichment analysis (ssGSEA). Additionally, weighted co-expression network analysis (WGCNA) was utilized to identify significantly associated genes. Finally, we employed LASSO regression, SVM-RFE, XGBoost, and random forest algorithms to further predict potential targets, followed by validation through molecular docking. Results Leeches may influence cellular immunity by modulating immune receptor activity, particularly through the activation of RGS10, CAPS2, and OPA1, thereby impacting the pathology of Type 2 Diabetes Mellitus (T2DM). Discussion However, it is important to note that our results lack experimental validation; therefore, further research is warranted to substantiate these findings.
Collapse
Affiliation(s)
- Tairan Hu
- Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Zhaohui Fang
- Department of Cardioiogy, First Hospital Affiliated to Anhui University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Institution of Cardiovascular Disease, Anhui Academy of Chinese Medicine Sciences, Hefei, China
| |
Collapse
|
21
|
Kharitonova A, Patel RS, Osborne B, Krause-Hauch M, Lui A, Vidyarthi G, Li S, Cai J, Patel NA. NPC86 Increases LncRNA Gas5 In Vivo to Improve Insulin Sensitivity and Metabolic Function in Diet-Induced Obese Diabetic Mouse Model. Int J Mol Sci 2025; 26:3695. [PMID: 40332318 PMCID: PMC12027414 DOI: 10.3390/ijms26083695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/04/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
In the United States, an estimated 38 million individuals (10% of the population) have type 2 diabetes mellitus (T2D), while approximately 97.6 million adults (38%) have prediabetes. Long noncoding RNAs (lncRNAs) are critical regulators of gene expression and metabolism. We were the first to demonstrate that lncRNA Growth Arrest-Specific Transcript 5 (GAS5 (human)/gas5 (mouse)) is decreased in the serum of T2D patients and established GAS5 as a biomarker for T2D diagnosis and onset prediction, now validated by other groups. We further demonstrated that GAS5 depletion impaired glucose uptake, decreased insulin receptor levels, and inhibited insulin signaling in human adipocytes, highlighting its potential as a therapeutic target in T2D. To address this, we developed NPC86, a small-molecule compound that stabilizes GAS5 by disrupting its interaction with UPF-1, an RNA helicase involved in nonsense-mediated decay (NMD) that regulates RNA stability. NPC86 increased GAS5 and insulin receptor (IR) levels, enhanced insulin signaling, and improved glucose uptake in vitro. In this study, we tested the efficacy of NPC86 in vivo in a diet-induced obese diabetic (DIOD) mouse model, and NPC86 treatment elevated gas5 levels, improved glucose tolerance, and enhanced insulin sensitivity, with no observed toxicity or weight changes. A transcriptomics analysis of adipose tissue revealed the upregulation of insulin signaling and metabolic pathways, including oxidative phosphorylation and glycolysis, while inflammatory pathways were downregulated. These findings highlight NPC86's therapeutic potential in T2D.
Collapse
MESH Headings
- Animals
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Mice
- Insulin Resistance/genetics
- Humans
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/drug therapy
- Disease Models, Animal
- Male
- Mice, Obese
- Obesity/metabolism
- Mice, Inbred C57BL
- Diet, High-Fat/adverse effects
- Signal Transduction/drug effects
- Insulin/metabolism
- Glucose/metabolism
- Receptor, Insulin/metabolism
Collapse
Affiliation(s)
- Anna Kharitonova
- James A. Haley Veteran’s Hospital, Research Service, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (A.K.); (R.S.P.); (B.O.); (M.K.-H.); (G.V.)
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA; (S.L.); (J.C.)
| | - Rekha S. Patel
- James A. Haley Veteran’s Hospital, Research Service, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (A.K.); (R.S.P.); (B.O.); (M.K.-H.); (G.V.)
| | - Brenna Osborne
- James A. Haley Veteran’s Hospital, Research Service, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (A.K.); (R.S.P.); (B.O.); (M.K.-H.); (G.V.)
| | - Meredith Krause-Hauch
- James A. Haley Veteran’s Hospital, Research Service, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (A.K.); (R.S.P.); (B.O.); (M.K.-H.); (G.V.)
| | - Ashley Lui
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Gitanjali Vidyarthi
- James A. Haley Veteran’s Hospital, Research Service, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (A.K.); (R.S.P.); (B.O.); (M.K.-H.); (G.V.)
| | - Sihao Li
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA; (S.L.); (J.C.)
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA; (S.L.); (J.C.)
| | - Niketa A. Patel
- James A. Haley Veteran’s Hospital, Research Service, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (A.K.); (R.S.P.); (B.O.); (M.K.-H.); (G.V.)
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
22
|
Ren B, Fang Z, Zhang Y, Yang H, Gou L, Yuan M, Wang Y, Gao D. BDH1 reduces apoptosis and alleviates mitochondrial damage of cardiomyocytes under high glucose condition as a downstream target of miR-125b. Biochem Biophys Res Commun 2025; 757:151561. [PMID: 40090116 DOI: 10.1016/j.bbrc.2025.151561] [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: 10/15/2024] [Revised: 02/27/2025] [Accepted: 02/27/2025] [Indexed: 03/18/2025]
Abstract
Diabetes is a chronic metabolic disease, characterized prominently by a persistent elevation of blood glucose level beyond the normal range. Prolonged hyperglycemia exerts deleterious effects on systems and organs of the body, leading to complications like diabetic cardiomyopathy (DCM). Our study commenced by screening the gene 3-hydroxybutyrate dehydrogenase 1 (BDH1) with low expression in DCM via Gene Expression Omnibus (GEO) analysis (GSE123975). Subsequently, we cultivated AC16 human cardiomyocytes in high glucose (HG) conditions and observed a reduction in BDH1 expression. To further investigate, we constructed plasmids for BDH1 knockdown (sh-BDH1) and overexpression (OE-BDH1). When BDH1 was overexpressed in HG-treated AC16 cells, apoptosis decreased, with reduced Bax/Bcl2 and Cleaved Caspase3/Caspase3 ratios. Additionally, mitochondrial ROS decreased, while expression of mitochondrial fusion protein mitofusin 2 (MFN2) and mitochondrial repair protein folliculin interacting protein 1 (FNIP1) increased. Notably, microRNA-125 b was upregulated in AC16 cells with hyperglycemia, and dual-luciferase reporter assays confirmed its targeting and inhibition of BDH1 mRNA. Inhibition of miR-125 b in HG-treated AC16 cells reversed apoptosis and mitochondrial ROS increase, yet simultaneous inhibition of both miR-125 b and BDH1 abolished this effect. In addition, we overexpressed BDH1 in diabetic mice by tail vein injection, and proved that overexpression of BDH1 could reduce cardiomyocyte apoptosis in vivo. In conclusion, our findings suggested that the miR-125-BDH1 axis could inhibit the production of mitochondrial ROS, promote mitochondrial fusion and repair, and reduce the apoptosis and mitochondrial damage of cardiomyocytes in HG condition.
Collapse
Affiliation(s)
- Bincheng Ren
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Cardiovascular Medicine, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhiyi Fang
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yimin Zhang
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Huan Yang
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lingjuan Gou
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Miao Yuan
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yu Wang
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Dengfeng Gao
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
23
|
Li D, Dai D, Wang J, Wang Y, Tian Y, Zhang C. Vine-inspired zinc-ion modified black phosphorus coating accelerates bone tissue infiltration of 3D printed scaffolds. Theranostics 2025; 15:5073-5086. [PMID: 40303335 PMCID: PMC12036863 DOI: 10.7150/thno.113623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Rationale: In the reconstruction of diabetic bone defects, 3D-printed scaffolds often encounter the challenge of limited and delayed tissue ingrowth in their central regions, which is critical for successful osseointegration and prognostic outcomes. Hyperglycemia induces endothelial apoptosis and impedes angiogenesis, thus inhibiting osteogenic differentiation of bone marrow stem cells (BMSCs). Methods: Drawing inspiration from the growth pattern of vines, we developed a Zn@BP/Si coating on the 3D-printed titanium scaffold to promote the coupling of angiogenesis and osteogenesis. This coating was achieved by Zn2+-modified black phosphorus (BP), which not only enhances the stability and photothermal properties of BP, but also prevents endothelial apoptosis. The effectiveness of Zn@BP/Si in the reconstruction of diabetic bone defects was investigated in rat model of diabetic femoral defect. Its effect on osteogenesis-angiogenesis coupling has also been explored in BMSCs and HUVECs. Results: Zn@BP/Si regulated mitochondrial dynamics and provided motivation for cell adhesion and migration, just like the climbing of vines. Notably, the regulation of enzymatic activity plays a crucial role in its inhibition of excessive mitochondrial fission. The results demonstrate that the Zn@BP/Si promotes the growth of "vascular vines" and ameliorates the angiogenic and osteogenic inhibition in diabetes. Conclusions: The study reveals the potential of bio-inspired Zn@BP/Si coating in angiogenesis-osteogenesis coupling and the treatment of diabetic bone defects.
Collapse
Affiliation(s)
| | | | | | | | | | - Chao Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
24
|
Jiang S, Nong T, Yu T, Qin Z, Huang J, Yin Z, Luo S, Lai Y, Jin J. Long term exposure to multiple environmental stressors induces mitochondrial dynamics imbalance in testis: Insights from metabolomics and transcriptomics. ENVIRONMENT INTERNATIONAL 2025; 198:109390. [PMID: 40139032 DOI: 10.1016/j.envint.2025.109390] [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: 11/28/2024] [Revised: 02/05/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025]
Abstract
Long-term exposure to adverse environment stressors (e.g. noise pollution, temperature, and crowding) impaired human health. However, research on the toxic effects of adverse environmental stressors on the male reproductive system is limited. This study employed integrated phenomics, metabolomics, and transcriptomics to investigate physiological disturbances in the testis of mice exposed to multiple adverse environmental stressors for two months. Phenotypic studies indicated that long-term environmental stimuli resulted in significant damage to the blood-testis barrier (BTB) and testes, evidenced by reduced testicular index, disrupted testicular tissue structure, abnormal tight junction protein expression, and spermatozoa abnormalities. Comprehensive multi-omics analysis revealed that long-term exposure to environmental stressors disrupted the BTB and testes, which was associated with mitochondrial metabolism disorders, including oxidative phosphorylation and fatty acid beta-oxidation, as well as glutathione and lipid metabolism alterations. Among these dysregulated pathways, significant alterations were observed in the critical regulators of mitochondrial fusion (MFN2) and fission (DRP1) within the BTB. Specifically, corticosterone treatment decreased tight junction protein expression, increased reactive oxygen species (ROS) levels, and impaired mitochondrial morphology and function, as evidenced by reduced mitochondrial membrane potential, elevated calcium ion concentration, and shortened mitochondrial length and network in vitro. Moreover, inhibiting DRP1 with Mdivi-1 or overexpressing MFN2 mitigated the corticosterone-induced reduction of tight junctions and mitochondrial dysregulation in TM4 cells. Collectively, maintaining mitochondrial homeostasis emerges as a promising strategy to alleviate the BTB and testicular injury induced by long-term exposure to multiple environmental stressors.
Collapse
Affiliation(s)
- Shiqin Jiang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Tianli Nong
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Ting Yu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhiyan Qin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Junyuan Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhaokun Yin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shiqi Luo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yating Lai
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jing Jin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
25
|
Lin L, Wei J, Xue J, Fan G, Zhu W, Zhu Y, Wu R. Drp1 Promotes Macrophage M1 Polarization and Inflammatory Response in Autoimmune Myocarditis by Driving Mitochondrial Fission. J Cardiovasc Transl Res 2025; 18:237-246. [PMID: 39388091 DOI: 10.1007/s12265-024-10570-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
Autoimmune myocarditis (AM) is characterized by an intricate inflammatory response within the myocardium. Dynamin-related protein 1 (Drp1), a pivotal modulator of mitochondrial fission, plays a role in the pathogenesis of various diseases. A myosin-induced experimental autoimmune myocarditis (EAM) mouse model was successfully established. Flow cytometry was employed to detect M1/M2-like macrophages. Mitochondrial fragmentation was assessed using Mito-Tracker Red CMXRos. Drp1 was upregulated and activated in EAM mice. Depletion of Drp1 was observed to mitigate inflammation, macrophage infiltration and M1 polarization within the cardiac tissue of EAM mice. In M1-like macrophages derived from the hearts of EAM mice, Drp1 was found to promote mitochondrial fission and diminish mitochondrial fusion. Furthermore, the depletion of Drp1 reduced the NF-κB-related pro-inflammatory response in EAM-associated M1-like macrophages. Drp1 drives mitochondrial fission in macrophages, driving their M1 polarization and the subsequent inflammatory response. Drp1 may represent an effective target for the prevention and treatment of AM.
Collapse
Affiliation(s)
- Lin Lin
- Department of Cardiovascular Medicine, Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Daminggong Campus, No. 5 Jianqiang Road, Xi'an, 710016, Weiyang District, China.
| | - Jin Wei
- Department of Cardiovascular Medicine, Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Daminggong Campus, No. 5 Jianqiang Road, Xi'an, 710016, Weiyang District, China
| | - Jiahong Xue
- Department of Cardiovascular Medicine, Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Daminggong Campus, No. 5 Jianqiang Road, Xi'an, 710016, Weiyang District, China
| | - Gang Fan
- Second Department of Cardiology, Xianyang First People's Hospital, Shaanxi University of Chinese Medicine, Xianyang, 712000, China
| | - Wenjing Zhu
- Department of Cardiovascular Medicine, Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Daminggong Campus, No. 5 Jianqiang Road, Xi'an, 710016, Weiyang District, China
| | - Yanhe Zhu
- Institute of Endiquidiopathies, School of Public Health, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ruiyun Wu
- Department of Internal Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| |
Collapse
|
26
|
Feng C, Hu Z, Zhao M, Leng C, Li G, Yang F, Fan X. Region-specific mitophagy in nucleus pulposus, annulus fibrosus, and cartilage endplate of intervertebral disc degeneration: mechanisms and therapeutic strategies. Front Pharmacol 2025; 16:1579507. [PMID: 40248091 PMCID: PMC12003974 DOI: 10.3389/fphar.2025.1579507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/24/2025] [Indexed: 04/19/2025] Open
Abstract
Intervertebral disc degeneration (IVDD) is a prevalent condition contributing to various spinal disorders, posing a significant global health burden. Mitophagy plays a crucial role in maintaining mitochondrial quantity and quality and is closely associated with the onset and progression of IVDD. Well-documented region-specific mitophagy mechanisms in IVDD are guiding the development of therapeutic strategies. In the nucleus pulposus (NP), impaired mitochondria lead to apoptosis, oxidative stress, senescence, extracellular matrix degradation and synthesis, excessive autophagy, inflammation, mitochondrial instability, and pyroptosis, with key regulatory targets including AMPK, PGC-1α, SIRT1, SIRT3, Progerin, p65, Mfn2, FOXO3, NDUFA4L2, SLC39A7, ITGα5/β1, Nrf2, and NLRP3 inflammasome. In the annulus fibrosus (AF), mitochondrial damage induces apoptosis and oxidative stress mediated by PGC-1α, while in the cartilage endplate (CEP), mitochondrial dysfunction similarly triggers apoptosis and oxidative stress. These mechanistic insights highlight therapeutic strategies such as activating Parkin-dependent and Ub-independent mitophagy pathways for NP, enhancing Parkin-dependent mitophagy for AF, and targeting Parkin-mediated mitophagy for CEP. These strategies include the use of natural ingredients, hormonal modulation, gene editing technologies, targeted compounds, and manipulation of related proteins. This review summarizes the mechanisms of mitophagy in different regions of the intervertebral disc and highlights therapeutic approaches using mitophagy modulators to ameliorate IVDD. It discusses the complex mechanisms of mitophagy and underscores its potential as a therapeutic target. The objective is to provide valuable insights and a scientific basis for the development of mitochondrial-targeted drugs for anti-IVDD.
Collapse
Affiliation(s)
- Chaoqun Feng
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ziang Hu
- Department of Orthopedics, The TCM Hospital of Longquanyi District, Chengdu, China
| | - Min Zhao
- International Ward (Gynecology), Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuan Leng
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guangye Li
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Yang
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaohong Fan
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
27
|
Hu H, Ding G, Liang W. Dietary therapy to halt the progression of diabetes to diabetic kidney disease. Food Funct 2025; 16:2622-2636. [PMID: 40047381 DOI: 10.1039/d4fo06011c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
Diabetic Kidney Disease (DKD) is a common and serious complication of diabetes, particularly Type 2 Diabetes Mellitus (T2DM), which significantly contributes to patient morbidity and mortality. The limitations of traditional treatments like ACE inhibitors and ARBs in managing DKD progression highlight the need for innovative therapeutic strategies. This review examines the impact of various dietary patterns, such as the Mediterranean diet, ketogenic diet, intermittent fasting, DASH diet, and vegetarian diet, on the management of DKD. Evidence suggests these diets can halt the progression of DKD, although further research is needed to confirm their long-term effectiveness and safety. Personalized dietary approaches tailored to individual needs may enhance outcomes for DKD patients.
Collapse
Affiliation(s)
- Hongtu Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China.
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
| | - Guohua Ding
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China.
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China.
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
| |
Collapse
|
28
|
Niu W, Liu X, Deng B, Hong T, Wang C, Yan Y, Liu J, Jiang Y, Li J. Piezo1 deletion mitigates diabetic cardiomyopathy by maintaining mitochondrial dynamics via ERK/Drp1 pathway. Cardiovasc Diabetol 2025; 24:127. [PMID: 40114130 PMCID: PMC11927149 DOI: 10.1186/s12933-025-02625-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/03/2025] [Indexed: 03/22/2025] Open
Abstract
OBJECTIVE Increasing evidence highlights the critical role of Piezo1 in cardiovascular diseases, with its expression upregulated in diabetic heart. However, the involvement of Piezo1 in the pathogenesis of diabetic cardiomyopathy (DCM) remains unclear. This study aims to elucidate the regulatory role of Piezo1 in mitochondrial dynamics within the context of DCM and to investigate the underlying mechanisms. METHODS We constructed cardiac-specific knockout of Piezo1 (Piezo1∆Myh6) mice. Type 1 diabetes was induced using streptozotocin (STZ) injection while type 2 diabetes was established through a high-fat diet combined with STZ. Echocardiography assessed left ventricular function, histological evaluations used HE and Masson staining to examine cardiac pathology in Piezo1fl/fl controls, Piezo1∆Myh6 controls, Piezo1fl/fl diabetic and Piezo1∆Myh6 diabetic mice. Mitochondrial function including oxygen species level, mitochondrial morphology, and respiration rate were also assessed. RESULTS Our findings revealed that Piezo1 expression was upregulated in the myocardium of diabetic mice and in high-glucose-treated cells. Cardiac-specific knockout of Piezo1 improved cardiac dysfunction and ameliorated cardiac fibrosis in diabetic mice. Moreover, Piezo1 deficiency also attenuated mitochondrial impairment. Piezo1fl/fl diabetic mice exhibited increased calpain activity and excessive mitochondrial fission mediated by Drp1 and obvious reduced fusion; however, Piezo1 deficiency restored calpain levels and mitochondrial dysfunction. These observations were also corroborated in H9C2 cells and neonatal mouse cardiomyocytes. Cardiac-specific knockout of Piezo1 increased phosphorylation of Drp1 and ERK1/2 in vivo and in vitro. Piezo1 knockout or treatment with inhibitor improved mitochondrial function. CONCLUSIONS This study provides the first evidence that Piezo1 is elevated in DCM through the modulation of mitochondrial dynamics, which is reversed by Piezo1 deficiency. Thus, Piezo1 inhibition may provide a promising therapeutic strategy for the treatment of DCM.
Collapse
MESH Headings
- Animals
- Diabetic Cardiomyopathies/genetics
- Diabetic Cardiomyopathies/enzymology
- Diabetic Cardiomyopathies/pathology
- Diabetic Cardiomyopathies/prevention & control
- Diabetic Cardiomyopathies/physiopathology
- Diabetic Cardiomyopathies/etiology
- Mitochondrial Dynamics
- Dynamins/metabolism
- Mice, Knockout
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/pathology
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/chemically induced
- Ion Channels/genetics
- Ion Channels/deficiency
- Ion Channels/metabolism
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Male
- Fibrosis
- Mice, Inbred C57BL
- Phosphorylation
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/enzymology
- Diabetes Mellitus, Type 1/complications
- Ventricular Function, Left
- Extracellular Signal-Regulated MAP Kinases/metabolism
- MAP Kinase Signaling System
- Rats
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/enzymology
- Diabetes Mellitus, Type 2/complications
- Mice
Collapse
Affiliation(s)
- Weipin Niu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
- Central Laboratory, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, 250014, China
| | - Xin Liu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
- Shandong Institute of Commerce and Technology, Jinan, 250103, China
| | - Bo Deng
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China
| | - Tianying Hong
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Cuifen Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yameng Yan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Jiali Liu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yuehua Jiang
- Central Laboratory, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, 250014, China.
| | - Jing Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
29
|
Mihalikova D, Stamm P, Kvandova M, Pednekar C, Strohm L, Ubbens H, Oelze M, Kuntic M, Witzler C, Bayo Jimenez MT, Rajlic S, Frenis K, Tang Q, Ruan Y, Karbach S, Kleinert H, Hahad O, von Kriegsheim A, Xia N, Grune T, Li H, Kröller-Schön S, Gericke A, Ruf W, Wild PS, Lurz P, Münzel T, Daiber A, Jansen T. Exposure to aircraft noise exacerbates cardiovascular and oxidative damage in three mouse models of diabetes. Eur J Prev Cardiol 2025; 32:301-314. [PMID: 39351780 DOI: 10.1093/eurjpc/zwae320] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/12/2024] [Accepted: 09/27/2024] [Indexed: 03/06/2025]
Abstract
AIMS Epidemiology links noise to increased risk of metabolic diseases like diabetes and obesity. Translational studies in humans and experimental animals showed that noise causes reactive oxygen species (ROS)-mediated cardiovascular damage. The interaction between noise and diabetes, specifically potential additive adverse effects, remains to be determined. METHODS AND RESULTS C57BL/6 mice were treated with streptozotocin (i.p. injections, 50 mg/kg/day for 5 days) to induce type 1 diabetes mellitus, with S961 (subcutaneous osmotic mini-pumps, 0.57 mg/kg/day for 7 days) or fed a high-fat diet (HFD, 20 weeks) to induce type 2 diabetes mellitus. Control and diabetic mice were exposed to aircraft noise to an average sound pressure level of 72 dB(A) for 4 days. While body weight was unaffected, noise reduced insulin production in all diabetes models. The oral glucose tolerance test showed only an additive aggravation by noise in the HFD model. Noise increased blood pressure and aggravated diabetes-induced aortic, mesenteric, and cerebral arterioles' endothelial dysfunction. ROS formation in cerebral arterioles, the aorta, the heart, and isolated mitochondria was consistently increased by noise in all models of diabetes. Mitochondrial respiration was impaired by diabetes and noise, however without additive effects. Noise increased ROS and caused inflammation in adipose tissue in the HFD model. RNA-sequencing data and alteration of gene pathway clusters also supported additive damage by noise in the setting of diabetes. CONCLUSION In all three models of diabetes, aircraft noise exacerbates oxidative stress, inflammation, and endothelial dysfunction in mice with pre-existing diabetes. Thus, noise may potentiate the already increased cardiovascular risk in diabetic patients.
Collapse
MESH Headings
- Animals
- Oxidative Stress
- Mice, Inbred C57BL
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/genetics
- Male
- Reactive Oxygen Species/metabolism
- Noise, Transportation/adverse effects
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/physiopathology
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/genetics
- Aircraft
- Diet, High-Fat
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/physiopathology
- Diabetes Mellitus, Type 1/chemically induced
- Diabetes Mellitus, Type 1/genetics
- Blood Glucose/metabolism
- Mice
- Disease Models, Animal
- Insulin/blood
Collapse
Affiliation(s)
- Dominika Mihalikova
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Paul Stamm
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Miroslava Kvandova
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy ofSciences, Bratislava, Slovakia
| | | | - Lea Strohm
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Henning Ubbens
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Matthias Oelze
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Marin Kuntic
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Claudius Witzler
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Maria Teresa Bayo Jimenez
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- Laboratory of RNA Archaeology, Instituto de Parasitología y Biomedicina 'López-Neyra' (CSIC), Granada, Spain
| | - Sanela Rajlic
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Katie Frenis
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- Hematology/Oncology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | - Qi Tang
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Yue Ruan
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Susanne Karbach
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Hartmut Kleinert
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Omar Hahad
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
| | | | - Ning Xia
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Huige Li
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Swenja Kröller-Schön
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Adrian Gericke
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Wolfram Ruf
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Philipp S Wild
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
- Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Institute of Molecular Biology (IMB), Johannes Gutenberg University, Mainz, Germany
| | - Philipp Lurz
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Thomas Münzel
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Andreas Daiber
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Thomas Jansen
- Department for Cardiology, Cardiology 1, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
- Department of Cardiology, KVB Hospital, Königstein, Germany
| |
Collapse
|
30
|
Han L, Ho CT, Lu M. Regulatory Role of Bioactive Compounds from Natural Spices on Mitochondrial Function. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:5711-5723. [PMID: 40019340 DOI: 10.1021/acs.jafc.4c12341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Natural spices have gained much attention for their aromatic and pungent flavors as well as their multiple beneficial health effects. As complex organelles that play a central role in energy production, stress response control, cell signal transduction, and metabolism regulation, mitochondria could be regulated by many bioactive components in spices. In this review, the role of mitochondria in maintaining cellular and metabolism homeostasis is summarized. The regulatory effects of mitochondrial function by major bioactive compounds from natural spices are evaluated, including capsaicin, 6-gingerol, 6-shogaol, allicin, quercetin, curcumin, tetrahydrocurcumin, and cinnamaldehyde. The underlying molecular mechanisms are also discussed. This work could enhance our understanding toward health-promoting properties of spice compounds as well as provide new insights into the prevention and treatment of disorders associated with mitochondrial dysfunctions by those nutraceuticals.
Collapse
Affiliation(s)
- Liguang Han
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, New Jersey 08901, United States
| | - Muwen Lu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
31
|
Huang B, Yang Y, Liu J, Zhang B, Lin N. Ubiquitination regulation of mitochondrial homeostasis: a new sight for the treatment of gastrointestinal tumors. Front Immunol 2025; 16:1533007. [PMID: 40134432 PMCID: PMC11933043 DOI: 10.3389/fimmu.2025.1533007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
Mitochondrial homeostasis (MH) refers to the dynamic balance of mitochondrial number, function, and quality within cells. Maintaining MH is significant in the occurrence, development, and clinical treatment of Gastrointestinal (GI) tumors. Ubiquitination, as an important post-translational modification mechanism of proteins, plays a central role in the regulation of MH. Over the past decade, research on the regulation of MH by ubiquitination has focused on mitochondrial biogenesis, mitochondrial dynamics, Mitophagy, and mitochondrial metabolism during these processes. This review summarizes the mechanism and potential therapeutic targets of ubiquitin (Ub)-regulated MH intervention in GI tumors.
Collapse
Affiliation(s)
- Bingqian Huang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Yulin Yang
- School of Clinical Chinese Medicine, Gansu University of Chinese Medicine, Gansu, China
| | - Jinming Liu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Biao Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Nengming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou, China
| |
Collapse
|
32
|
Rashad S, Marahleh A. Metabolism Meets Translation: Dietary and Metabolic Influences on tRNA Modifications and Codon Biased Translation. WILEY INTERDISCIPLINARY REVIEWS. RNA 2025; 16:e70011. [PMID: 40119534 PMCID: PMC11928779 DOI: 10.1002/wrna.70011] [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] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/08/2025] [Accepted: 03/11/2025] [Indexed: 03/24/2025]
Abstract
Transfer RNA (tRNA) is not merely a passive carrier of amino acids, but an active regulator of mRNA translation controlling codon bias and optimality. The synthesis of various tRNA modifications is regulated by many "writer" enzymes, which utilize substrates from metabolic pathways or dietary sources. Metabolic and bioenergetic pathways, such as one-carbon (1C) metabolism and the tricarboxylic acid (TCA) cycle produce essential substrates for tRNA modifications synthesis, such as S-Adenosyl methionine (SAM), sulfur species, and α-ketoglutarate (α-KG). The activity of these metabolic pathways can directly impact codon decoding and translation via regulating tRNA modifications levels. In this review, we discuss the complex interactions between diet, metabolism, tRNA modifications, and mRNA translation. We discuss how nutrient availability, bioenergetics, and intermediates of metabolic pathways, modulate the tRNA modification landscape to fine-tune protein synthesis. Moreover, we highlight how dysregulation of these metabolic-tRNA interactions contributes to disease pathogenesis, including cancer, metabolic disorders, and neurodegenerative diseases. We also discuss the new emerging field of GlycoRNA biology drawing parallels from glycobiology and metabolic diseases to guide future directions in this area. Throughout our discussion, we highlight the links between specific modifications, their metabolic/dietary precursors, and various diseases, emphasizing the importance of a metabolism-centric tRNA view in understanding many pathologies. Future research should focus on uncovering the interplay between metabolism and tRNA in specific cellular and disease contexts. Addressing these gaps will guide new research into novel disease interventions.
Collapse
Affiliation(s)
- Sherif Rashad
- Department of Neurosurgical Engineering and Translational NeuroscienceTohoku University Graduate School of MedicineSendaiJapan
- Department of Neurosurgical Engineering and Translational Neuroscience, Graduate School of Biomedical EngineeringTohoku UniversitySendaiJapan
| | - Aseel Marahleh
- Frontier Research Institute for Interdisciplinary SciencesTohoku UniversitySendaiJapan
- Graduate School of DentistryTohoku UniversitySendaiJapan
| |
Collapse
|
33
|
DeStephanis D, Long MR, Williams AG, Santiago M, Tonkin J, Stevens CM, Davis MA, Ruggiero AD, Henstridge DC, Premilovac D, Kavanagh K. Metabolically unhealthy adipose tissue is characterized by reductions in mitochondrial size and function. Obesity (Silver Spring) 2025; 33:537-547. [PMID: 39948771 PMCID: PMC11897863 DOI: 10.1002/oby.24221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/23/2024] [Accepted: 11/11/2024] [Indexed: 02/23/2025]
Abstract
OBJECTIVE Adipose function, not mass, underpins metabolic health. Lean and obese nonhuman primates (NHPs) naturally develop metabolic syndrome. Mitochondria-related measures in subcutaneous adipose tissue (SQ AT) and peripheral blood mononuclear cells may elucidate differences that transcend adiposity measures. METHODS Obesity statuses ranged from very lean to severely obese (<9%->50%, n = 44), which were equivalent in healthy or unhealthy NHPs (metabolic syndrome score difference, p < 0.001). We evaluated SQ AT histology, electron microscopy, tissue proteins, and bioenergetics. RESULTS Unhealthy adipocytes had mitochondria one-half the size of healthy adipocytes (p < 0.01), whereas adipocyte cell sizes were comparable. Consistent with small mitochondria, we saw deficiencies in mitochondrial fusion and quality-control proteins in SQ AT from unhealthy NHPs (all p < 0.05). Smaller mitochondria in unhealthy adipocytes were consistent with low SQ AT tissue respiration (p < 0.05). Mitochondrial size was specifically reduced with unhealthiness, as mitochondrial abundance, size, and related metrics were unrelated to adiposity. Isolated stromal vascular cells showed comparable respirometry profiles, substantiating specificity of adipocyte-related mitochondrial defects. Peripheral blood mononuclear cell bioenergetic indices were increased in unhealthy NHPs, indicative of immune cell activation, and correlated to SQ AT inflammatory cytokines. CONCLUSIONS We conclude that targeting mitochondrial fusion processes would be a rational strategy to improve metabolic health, independent of total fat mass.
Collapse
Affiliation(s)
- Darla DeStephanis
- Department of PathologyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Masha R. Long
- Department of PathologyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Abigail G. Williams
- Department of PathologyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - McKinley Santiago
- Department of PathologyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Jack Tonkin
- Department of PathologyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Christina M. Stevens
- Department of PathologyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Matthew A. Davis
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Alistaire D. Ruggiero
- Department of PathologyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | | | - Dino Premilovac
- College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Kylie Kavanagh
- Department of PathologyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
- College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| |
Collapse
|
34
|
Wang HN, Wang Y, Zhang SY, Bai L. Emerging roles of the acid sphingomyelinase/ceramide pathway in metabolic and cardiovascular diseases: Mechanistic insights and therapeutic implications. World J Cardiol 2025; 17:102308. [DOI: 10.4330/wjc.v17.i2.102308] [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: 10/14/2024] [Revised: 12/10/2024] [Accepted: 02/08/2025] [Indexed: 02/25/2025] Open
Abstract
Metabolic diseases have emerged as a leading cause of mortality from non-communicable diseases, posing a significant global public health challenge. Although the association between ceramides (Cers) and metabolic diseases is well-established, the role of the acid sphingomyelinase (ASMase)/Cer pathway in these diseases remains underexplored. This review synthesizes recent research on the biological functions, regulatory mechanisms, and targeted therapies related to the ASMase/Cer pathway in metabolic conditions, including obesity, diabetes, non-alcoholic fatty liver disease, and cardiovascular disease. The effects of the ASMase/Cer pathway on metabolic disease-related indicators, such as glycolipid metabolism, insulin resistance, inflammation, and mitochondrial homeostasis are elucidated. Moreover, this article discusses the therapeutic strategies using ASMase/Cer inhibitors for inverse prevention and treatment of these metabolic diseases in light of the possible efficacy of blockade of the ASMase/Cer pathway in arresting the progression of metabolic diseases. These insights offered herein should provide insight into the contribution of the ASMase/Cer pathway to metabolic diseases and offer tools to develop therapeutic interventions for such pathologies and their severe complications.
Collapse
Affiliation(s)
- Hong-Ni Wang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Ye Wang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Si-Yao Zhang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Lan Bai
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| |
Collapse
|
35
|
Wen X, Ji Y, Tang H, Jin Z, Su W, Zhou L, Xia ZY, Li L, Lei S. Caveolin-3: therapeutic target for diabetic myocardial ischemia/reperfusion injury. Mol Med 2025; 31:80. [PMID: 40012041 PMCID: PMC11866611 DOI: 10.1186/s10020-025-01117-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/05/2025] [Indexed: 02/28/2025] Open
Abstract
Myocardial ischemia/reperfusion (I/R) injury is a major global health problem with high rates of mortality and disability, which is more severe in patients with diabetes. Substantial researches have documented that diabetic myocardium are more susceptible to I/R injury, but many current intervention strategies against myocardial I/R injury have limited effectiveness in diabetic hearts. Caveolin-3 (Cav-3) is the signature protein of caveolae and serves as a signal integration and transduction platform in the plasma membrane of cardiomyocytes, which plays a vital role in myocardial functions, metabolism and protection of multiple conditioning strategies against I/R injury. Nevertheless, numerous studies have revealed that the expression of Cav-3 is impaired in diabetic hearts, which contributes to increased vulnerability of myocardium to I/R injury and resistance to protective conditioning strategies. In this review, we outline the basic structure and function of Cav-3, emphatically present the unique role of Cav-3 as a signal integration and transduction element in diabetic myocardial I/R injury and discuss its therapeutic perspective in strategies against myocardial I/R injury in diabetes.
Collapse
Affiliation(s)
- Xinyu Wen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan City, China
| | - Yanwei Ji
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan City, China
| | - Hepeng Tang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan City, China
| | - Zhenshuai Jin
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan City, China
| | - Wating Su
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan City, China
| | - Lu Zhou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan City, China
| | - Zhong-Yuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan City, China
| | - Lin Li
- Department of Anesthesiology, Affiliated RenHe Hospital of China, Second Clinical Medical College, Three Gorges University, Yichang, Hubei Province, China.
| | - Shaoqing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan City, China.
| |
Collapse
|
36
|
Wei L, Fang C, Jiang Y, Zhang H, Gao P, Zhou X, Zhu S, Du Y, Su R, Guo L, He M, Wang S, Feng L, Yu J. The Role of Placental MFF-Mediated Mitochondrial Fission in Gestational Diabetes Mellitus. Diabetes Metab Syndr Obes 2025; 18:541-554. [PMID: 39995822 PMCID: PMC11849531 DOI: 10.2147/dmso.s484002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Introduction Gestational diabetes mellitus (GDM) refers to hyperglycemia first recognized during pregnancy, characterized by decreased insulin sensitivity and impaired glucose metabolism. Dynamic fusion and fission processes within mitochondria play critical roles in maintaining glucose metabolism homeostasis. Given the fundamental role of mitochondrial fission factor (MFF) in mitochondrial fission, the intention of this study was to investigate mitochondrial dynamics in the placentae of GDM patients and explore the role of MFF in the etiopathogenesis and progression of GDM through the modulation of glucose metabolism and insulin resistance. Methods 40 Placental tissues were obtained from pregnant women undergoing cesarean section with GDM (n=20) and those with normoglycemia (n=20). To mimic the intrauterine high glucose environment, immortalized human-derived first-trimester extravillous trophoblast cells HTR8/SVneo were used and treated in a high glucose environment. Immunofluorescence was utilized to analyze MFF expression in placental tissues and mitochondrial length in HTR8/SVneo cells. The expression levels of glucose transporters (GLUTs) and other pivotal proteins involved in mitochondrial dynamics and the insulin signaling pathway, were assessed by Western blotting. Additionally, cellular glucose uptake capacity was determined using a glucose assay kit. Results MFF expression was greater in the GDM group than in the normoglycemic group. In a high-glucose environment, the expression of fusion-related proteins OPA1, MFN1 and MFN2 decreased while the expression of DRP1 and MFF increased, indicating that the mitochondrial dynamics of trophoblast cells shift toward fission. Elevated mitochondrial fission hinders the insulin signaling pathway, resulting in a reduction in glucose uptake by HTR8/SVneo cells and a concurrent decrease in GLUT4 expression. Discussion Our study demonstrates that MFF-mediated mitochondrial fission inhibits insulin sensitivity and upregulates glucose transport in GDM, which is related to offspring exposure to a hyperglycemic intrauterine environment. These results provide a novel therapeutic target for addressing GDM that may mitigate unfavorable pregnancy outcomes.
Collapse
Affiliation(s)
- Lijie Wei
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Chenyun Fang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Yi Jiang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Huiting Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Peng Gao
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Xuan Zhou
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Shenglan Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Yuanyuan Du
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Rui Su
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Lili Guo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Mengzhou He
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Shaoshuai Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Ling Feng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Jun Yu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| |
Collapse
|
37
|
Chen S, Zhang X, Mo H, Peng Y, An Z, Wu J, Wei X, Zhang S, Xiong Y, Jiang W, Peng X, Zhuo L, Lei Z, Wang Z, Hu Z. Structure-activity relationship study of novel evodiamine amino acid conjugates with potent anti-colorectal cancer efficacy. Eur J Med Chem 2025; 283:117132. [PMID: 39647421 DOI: 10.1016/j.ejmech.2024.117132] [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: 06/15/2024] [Revised: 11/29/2024] [Accepted: 12/01/2024] [Indexed: 12/10/2024]
Abstract
Evodiamine has been a promising lead structure with broad-spectrum antitumor activity. Druggability optimization is the most challenging part of evodiamine-based lead-to-candidate campaign. Amino acids as building blocks for conjugates are widely incorporated into approved drug and drug candidates, demonstrating highly attractive druggability. Herein, a series of evodiamine amino acid conjugates were designed and synthesized based on the evodiamine lead compound (±)-8b discovered in our previous work. The structure-activity relationship (SAR) studies culminated in the identification of a promising conjugate (-)-15h featuring a N-Boc-l-glutamine group and a chiral carbon atom (sinister), which exhibited nanomolar antiproliferative activity against LoVo and RKO colorectal cancer cells. Moreover, (-)-15h could inhibit topoisomerases I, arrest the cell cycle in the G2/M phase, and induce apoptosis. Importantly, (-)-15h (tumor growth inhibition rate was 82.53 % in 40 mpk) showed better efficacy and tolerability to that of parent compound (-)-8b (tumor growth inhibition rate was 51.22 % in 40 mpk) in LoVo xenograft model. Further, (-)-15h (tumor growth inhibition rate was 70.09 % in 40 mpk) showed comparable efficacy and better tolerability to that of topotecan (tumor growth inhibition rate was 70.67 % in 0.5 mpk) in HT-29 xenograft model. Collectively, this study further provided a strong scientific basis for amino acid-based structural modifications and a drug lead for anti-colorectal cancer applications.
Collapse
Affiliation(s)
- Shuting Chen
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xi Zhang
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Hanxuan Mo
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Ying Peng
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zhigang An
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Junbo Wu
- Department of Colorectal Surgery, Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, Hunan, 421001, China
| | - Xiuzhen Wei
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Siyi Zhang
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yongxia Xiong
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Weifan Jiang
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xue Peng
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Linsheng Zhuo
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Zhengwen Lei
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Zhen Wang
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, 810008, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410008, China; MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, China.
| | - Zecheng Hu
- The First Affiliated Hospital, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
38
|
Gu H, Li Q, Liu Z, Li Y, Liu K, Kong X, Zhang Y, Meng Q, Song K, Xie Q, Gao Y, Cheng L. SPP1-ITGα5/β1 Accelerates Calcification of Nucleus Pulposus Cells by Inhibiting Mitophagy via Ubiquitin-Dependent PINK1/PARKIN Pathway Blockade. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411162. [PMID: 39721032 PMCID: PMC11831503 DOI: 10.1002/advs.202411162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/02/2024] [Indexed: 12/28/2024]
Abstract
Low back pain (LBP) caused by nucleus pulposus degeneration and calcification leads to great economic and social burden worldwide. Unexpectedly, no previous studies have demonstrated the association and the underlying mechanism between nucleus pulposus tissue degeneration and calcification formation. Secreted Phosphoprotein 1 (SPP1) exerts crucial functions in bone matrix mineralization and calcium deposition. Here, a novel function of SPP1 is reported, namely that it can aggravate nucleus pulposus cells (NPs) degeneration by negatively regulating extracellular matrix homeostasis. The degenerated NPs have a higher mineralization potential, which is achieved by SPP1. Mechanistically, SPP1 can accelerate the degeneration of nucleus pulposus cells by activating integrin α5β1 (ITGα5/β1), aggravating mitochondrial damage and inhibiting mitophagy. SPP1-ITGα5/β1 axis inhibits mitophagy by PINK1/PARKIN pathway blockade. In conclusion, SPP1 activates ITGα5/β1 to inhibit mitophagy, accelerates NPs degeneration, and induces calcification, thereby leading to intervertebral disc degeneration (IVDD) and calcification, identifying the potentially unknown mechanism and relationship between IVDD and calcification. Important insights are provided into the role of SPP1 in nucleus pulposus calcification in IVDD by inducing nucleus pulposus cell senescence through inhibition of mitophagy and may help develop potential new strategies for IVDD treatment.
Collapse
Affiliation(s)
- Hanwen Gu
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Qi Li
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Zhenchuan Liu
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Yanlin Li
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Kaiwen Liu
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Xiangzhen Kong
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Yuanqiang Zhang
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Qunbo Meng
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Kangle Song
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Qing Xie
- Department of PharmacyQilu HospitalCheeloo College of MedicineShandong UniversityJinan250012China
| | - Yuan Gao
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Lei Cheng
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| |
Collapse
|
39
|
Old V, Davies M, Papamargaritis D, Choudhary P, Watson E. The Effects of Glucagon-Like Peptide-1 Receptor Agonists on Mitochondrial Function Within Skeletal Muscle: A Systematic Review. J Cachexia Sarcopenia Muscle 2025; 16:e13677. [PMID: 39815782 PMCID: PMC11735953 DOI: 10.1002/jcsm.13677] [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: 07/02/2024] [Revised: 11/05/2024] [Accepted: 11/16/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Obesity is a chronic disease associated with increased risk of multiple metabolic and mental health-related comorbidities. Recent advances in obesity pharmacotherapy, particularly with glucagon-like peptide-1 (GLP-1) receptor agonists (RAs), have the potential to transform obesity and type 2 diabetes mellitus (T2DM) care by promoting marked weight loss, improving glycaemic control and addressing multiple obesity-related comorbidities, with added cardio-renal benefits. Dual agonists combining GLP-1 with other enteropancreatic hormones such as glucose-dependent insulinotropic polypeptide (GIP) have also been developed in recent years, leading to greater weight loss than using GLP-1 RAs alone. However, up to 40% of the weight lost with GLP-1 RAs comes from lean body mass, raising concerns about potential adverse effects on skeletal muscle function. Mitochondrial dysfunction, characterized by reduced mitochondrial size and activity, is prevalent in individuals with obesity and T2DM and is a known contributor to muscle wasting in ageing and some chronic diseases. This systematic review investigates the impact of GLP-1-based therapies on skeletal muscle mitochondrial function in individuals with obesity and T2DM or in related animal and cell models. METHODS A comprehensive search of MEDLINE, Scopus, CINAHL and clinicaltrials.gov was conducted. Inclusion criteria included randomized controlled trials, randomized crossover trials, cluster randomized control trials and basic science studies involving any GLP-1 RA or GLP-1/GIP dual agonist. Outcomes of interest were skeletal muscle respiratory function either in the form of measurements of mass, number, content, oxidative capacity/respiratory function, mitochondrial dynamics, mitochondrial biogenesis and mitophagy. RESULTS Eight studies were eligible for analysis; no human studies were identified. All of the included studies used GLP-1 RAs (single agonists) as intervention. The emerging evidence suggests that GLP-1 RAs increase mitochondrial area, number and morphology (i.e., reduces swelling). Data are conflicting on the effect of GLP-1 RAs upon mitochondrial mass, respiration and the expression of uncoupling proteins and PGC-1α. Data also demonstrate muscle specific (i.e., soleus vs. extensor digitorum longus) responses to GLP-1 RAs. CONCLUSION GLP-1 RAs appear to have a positive effect upon mitochondria area, number and morphology, but effects upon other aspects of mitochondrial health remain inconclusive. Data are very limited and solely presented in animal and in vitro models. Future studies should be conducted in human populations in order to begin to understand the effect of GLP-1 RAs and GLP-1-based therapies on human skeletal muscle mitochondria.
Collapse
Affiliation(s)
- Victoria J. Old
- Department of Cardiovascular Sciences, College of Life SciencesUniversity of LeicesterLeicesterUK
| | - Melanie J. Davies
- Diabetes Research Centre, College of Life SciencesUniversity of LeicesterLeicesterUK
| | | | - Pratik Choudhary
- Diabetes Research Centre, College of Life SciencesUniversity of LeicesterLeicesterUK
| | - Emma L. Watson
- Department of Cardiovascular Sciences, College of Life SciencesUniversity of LeicesterLeicesterUK
| |
Collapse
|
40
|
He S, Yan C, Wang Z, Mao Y, Liu K, Sun J, Zang Y, Wang M, Li G, Yang Y. Icariside II relieves radiation enteritis by regulating PINK/Parkin-mediated mitophagy. Int Immunopharmacol 2025; 146:113861. [PMID: 39733645 DOI: 10.1016/j.intimp.2024.113861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/10/2024] [Accepted: 12/11/2024] [Indexed: 12/31/2024]
Abstract
Radiation enteritis (RE) is one of the major side effects of radiotherapy. So far, there are no effective drugs for preventing the disease process. Icariside II (ICS II) is a highly efficient monomer compound extracted and purified from the classic Chinese medicinal herb Epimedium. It has anti-inflammatory, antioxidant, and immunomodulatory effects. However, the role and mechanism of ICS II on radiation enteritis are not clear. Here, we reveal the role of ICS II in radiation enteritis by using an irradiation-induced rat model and a human colorectal cancer cell (CaCo2). After intragastric administration, HE staining and Tunel staining to observe the histopathological changes in the colon, and TEM to observe the ultrastructure of mitochondria; The antioxidant indexes and mitochondrial function-related markers of colon tissues were determined; DCFH-DA fluorescent probe were used to detect the cellular ROS level, JC-1 staining was used to detect the changes in mitochondrial membrane potential, and Western Blot was used to detect related protein expression. The results showed that ICS II could reduce intestinal injury and attenuate the radiation-induced oxidative stress and inflammatory response. In addition, ICS II could effectively attenuate mitochondrial damage and activate mitochondrial autophagy in rats. Mechanistically, ICS II activates mitochondrial autophagy-related protein expression to rescue radiation-induced damage to mitophagy. We found that by inhibiting mitophagy, the therapeutic effect of ICS II can be eliminated and our data suggest that ICS II may be a new and effective drug candidate for the treatment of radiation enteritis.
Collapse
Affiliation(s)
- Shuangyan He
- Changchun University of Chinese Medicine, Changchun, China
| | - Chengqiu Yan
- Department of Anorectal, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Zhuo Wang
- Changchun University of Chinese Medicine, Changchun, China
| | - Yinhui Mao
- Changchun University of Chinese Medicine, Changchun, China
| | - Kunjian Liu
- Changchun University of Chinese Medicine, Changchun, China
| | - Juntao Sun
- Changchun University of Chinese Medicine, Changchun, China
| | - Yueyue Zang
- Changchun University of Chinese Medicine, Changchun, China
| | - Mingxing Wang
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China.
| | - Guofeng Li
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China; Anorectal Department, Shenzhen Bao'an Authentic TCM Therapy Hospital, Shenzhen, China.
| | - Yong Yang
- Changchun University of Chinese Medicine, Changchun, China; Department of Urology, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China.
| |
Collapse
|
41
|
Młynarska E, Czarnik W, Dzieża N, Jędraszak W, Majchrowicz G, Prusinowski F, Stabrawa M, Rysz J, Franczyk B. Type 2 Diabetes Mellitus: New Pathogenetic Mechanisms, Treatment and the Most Important Complications. Int J Mol Sci 2025; 26:1094. [PMID: 39940862 PMCID: PMC11817707 DOI: 10.3390/ijms26031094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM), a prevalent chronic disease affecting over 400 million people globally, is driven by genetic and environmental factors. The pathogenesis involves insulin resistance and β-cell dysfunction, mediated by mechanisms such as the dedifferentiation of β-cells, mitochondrial dysfunction, and oxidative stress. Treatment should be based on non-pharmacological therapy. Strategies such as increased physical activity, dietary modifications, cognitive-behavioral therapy are important in maintaining normal glycemia. Advanced therapies, including SGLT2 inhibitors and GLP-1 receptor agonists, complement these treatments and offer solid glycemic control, weight control, and reduced cardiovascular risk. Complications of T2DM, such as diabetic kidney disease, retinopathy, and neuropathy, underscore the need for early diagnosis and comprehensive management to improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Witold Czarnik
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Natasza Dzieża
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Weronika Jędraszak
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Gabriela Majchrowicz
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Filip Prusinowski
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Magdalena Stabrawa
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
42
|
Shi J, Liu M, Zhu H, Jiang C. SIRT3 mitigates high glucose-induced damage in retinal microvascular endothelial cells via OPA1-mediated mitochondrial dynamics. Exp Cell Res 2025; 444:114320. [PMID: 39491778 DOI: 10.1016/j.yexcr.2024.114320] [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: 09/21/2024] [Revised: 10/29/2024] [Accepted: 11/02/2024] [Indexed: 11/05/2024]
Abstract
Oxidative stress in endothelial cells is pivotal in diabetic retinopathy (DR), with mitochondrial homeostasis being crucial to mitigate this stress. This study explored the roles of mitochondrial sirtuins (SIRTs) in high glucose (HG)-induced oxidative stress, related endothelial impairment, and mitochondrial homeostasis damage in rat retinal microvascular endothelial cells (RMECs). RMECs were cultured under HG or equivalent osmotic conditions. Cell viability was assessed using the Cell Counting Kit-8 assay, whereas cell death and survival were determined via calcein-AM/propidium iodide double staining. Reactive oxygen species (ROS) levels were measured using 2',7'-dichlorofluorescein fluorescence. Expression of mitochondrial SIRTs3-5 and key mitochondrial homeostasis molecules was quantified by the quantitative real-time polymerase chain reaction and confirmed by western blotting. Mitochondrial morphology was evaluated using electron microscopy and the MitoTracker fluorescent probe. A SIRT3-overexpressing RMEC line was constructed to assess the role of SIRT3 in oxidative stress and mitochondrial dynamics. After 48 h of HG exposure, cell viability was significantly reduced, with a concomitant increase in cell death and ROS levels, alongside a marked decrease in SIRT3 expression and molecules associated with mitochondrial dynamics. SIRT3 overexpression reversed these effects, particularly increasing the mitochondrial fusion-related molecule, optic atrophy 1 (OPA1). However, the OPA1 inhibitor, MYLS22, blocked the protective effect of SIRT3, leading to more dead cells, a higher ROS level, and intensified mitochondrial fragmentation. These results suggest that SIRT3 is involved in HG-induced imbalanced mitochondrial dynamics of endothelial cells in DR, potentially through the OPA1 pathway.
Collapse
Affiliation(s)
- Jiemei Shi
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Fudan University, Shanghai, China; Key Laboratory of Myopia of State Health Ministry, and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai, China
| | - Min Liu
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Fudan University, Shanghai, China; Key Laboratory of Myopia of State Health Ministry, and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai, China
| | - Haohao Zhu
- Department of Ophthalmology, People's Hospital of Shanghai No. 5, Shanghai, 200240, China.
| | - Chunhui Jiang
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Fudan University, Shanghai, China; Key Laboratory of Myopia of State Health Ministry, and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai, China.
| |
Collapse
|
43
|
Gad ES, Aldossary SA, El-Ansary MR, Abd El-Galil MM, Abd-El-Hamid AH, El-Ansary AR, Hassan NF. Cilostazol counteracts mitochondrial dysfunction in hepatic encephalopathy rat model: Insights into the role of cAMP/AMPK/SIRT1/ PINK-1/parkin hub and p-CREB /BDNF/ TrkB neuroprotective trajectory. Eur J Pharmacol 2025; 987:177194. [PMID: 39667427 DOI: 10.1016/j.ejphar.2024.177194] [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: 06/26/2024] [Revised: 11/17/2024] [Accepted: 12/09/2024] [Indexed: 12/14/2024]
Abstract
A devasting stage of chronic hepatic dysfunction is strictly correlated with neurological impairment, signifying hepatic encephalopathy (HE). HE is a multifactorial condition; therefore, hyperammonemia, oxidative stress, neuroinflammation, and mitochondrial dysfunction interplay in HE's progressive development. Cilostazol (Cilo) has shown promising neuroprotective and hepatoprotective effectiveness in different neuronal and hepatic disorders; however, its efficiency against HE hasn't yet been explored. This study aimed to investigate the protective role of Cilo against thioacetamide (TAA)-induced HE in rats targeting mitochondrial dysfunction via modulation of Adenosine monophosphate-activated protein kinase (AMPK)/Silent information regulator 1 (SIRT1) dependent pathways. Rats were allocated into three groups: the normal control group, the TAA group received (100 mg/kg, three times per week, for six weeks) to induce HE, and the Cilo group received (Cilo 100 mg/kg/day for six weeks, oral gavage) concurrently with TAA. Cilo counteracted HE indicated in the enhancement of cognitive impairment and the motor performance of rats (P < 0.0001), modulation AMPK/SIRT1signaling pathway causing reduction of NF-kB p65 (P < 0.0001) evoked inflammation along with histopathological alterations and glial fibrillary acidic protein (GFAP) immunoreactivity (P < 0.0001), restoration nuclear factor E2-related factor 2 (Nrf2) (P < 0.0001) antioxidant effects, reduction of Bax and elevation of Bcl2 immunoreactivity (P < 0.0001) in addition to boosting mitochondrial biogenesis by upregulation of PTEN-induced kinase-1 (PINK-1)/Parkin (P < 0.0001)and restoration of Brain-derived neurotrophic factor (BDNF) (P = 0.0002)/tropomyosin-related kinase B (TrkB) (P < 0.0001)/cAMP response element-binding (CREB) (P < 0.0001) neuroprotective axis. Collectively, Cilo activates the SIRT1 trajectory to abridge mitochondrial dysfunction invigorated in the HE rat model via restoration of mitochondrial hemostasis.
Collapse
Affiliation(s)
- Enas S Gad
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, AL Ahsa, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University, Kantara Branch, Ismailia, Egypt
| | - Sara A Aldossary
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, AL Ahsa, Saudi Arabia
| | - Mona R El-Ansary
- Department of Biochemistry, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Mona M Abd El-Galil
- Department of Histology and Cell Biology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Asmaa Hassan Abd-El-Hamid
- Department of Histology and Cell Biology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Amira R El-Ansary
- Department of Internal Medicine, Faculty of Medicine, Misr University for Science and Technology, Cairo, Egypt
| | - Noha F Hassan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt.
| |
Collapse
|
44
|
Zeng W, Wang L, Wang C, Xiong X, Huang Q, Chen S, Liu C, Liu W, Wang Y, Huang Q. SENP1 prevents high fat diet-induced non-alcoholic fatty liver diseases by regulating mitochondrial dynamics. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167527. [PMID: 39332783 DOI: 10.1016/j.bbadis.2024.167527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 09/12/2024] [Accepted: 09/22/2024] [Indexed: 09/29/2024]
Abstract
Mitochondrial dynamics plays a crucial role in the occurrence and development of non-alcoholic fatty liver diseases (NAFLD). SENP1, a SUMO-specific protease, catalyzes protein de-SUMOylation and involves in various physiological and pathological processes. However, the exact role of SENP1 in NAFLD remains unclear. Therefore, we investigated the regulatory role of SENP1 in mitochondrial dynamics during the progression of NAFLD. In the study, the NAFLD in vivo model induced by high fat diet (HFD) and in vitro model induced by free fatty acids (FFA) were established to investigate the role and underlying mechanism of SENP1 through detecting mitochondrial morphology and dynamics. Our results showed that the down-regulation of SENP1 expression and the mitochondrial dynamics dysregulation occurred in the NAFLD, evidenced as mitochondrial fragmentation, up-regulation of p-Drp1 ser616 and down-regulation of MFN2, OPA1. However, over-expression of SENP1 significantly alleviated the NAFLD, rectified the mitochondrial dynamics disorder, reduced Cyt-c release and ROS levels induced by FFA or HFD; moreover, the over-expression of SENP1 also reduced the SUMOylation levels of Drp1 and prevented the Drp1 translocation to mitochondria. Our findings suggest that the possible mechanisms of SENP1 were through rectifying the mitochondrial dynamics disorder, reducing Cyt-c release and ROS-mediated oxidative stress. The findings would provide a novel target for the prevention and treatment of NALFD.
Collapse
Affiliation(s)
- Wenjing Zeng
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Li Wang
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Chaowen Wang
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Xiaowei Xiong
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Qianqian Huang
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Sheng Chen
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Chen Liu
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Wentao Liu
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Yuan Wang
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Qiren Huang
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China.
| |
Collapse
|
45
|
Lu LL, Liu LZ, Li L, Hu YY, Xian XH, Li WB. Sodium butyrate improves cognitive dysfunction in high-fat diet/ streptozotocin-induced type 2 diabetic mice by ameliorating hippocampal mitochondrial damage through regulating AMPK/PGC-1α pathway. Neuropharmacology 2024; 261:110139. [PMID: 39233201 DOI: 10.1016/j.neuropharm.2024.110139] [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: 07/15/2024] [Revised: 08/17/2024] [Accepted: 08/31/2024] [Indexed: 09/06/2024]
Abstract
Cognitive dysfunction is an important comorbidity of type 2 diabetes mellitus (T2DM). Sodium butyrate (NaB) is a short-chain fatty acid and has an effect improving T2DM-associated cognitive dysfunction. Using a high-fat diet (HFD)/streptozotocin (STZ)-induced T2DM mouse model, the present study investigated the mechanism involved in the beneficial effect of butyrate on diabetic cognitive dysfunction, with a focus on ameliorating mitochondrial damage through regulating the adenosine monophosphate-activated protein kinase/peroxisome proliferator-activated receptor gamma coactivator 1α (AMPK/PGC-1α) pathway considering the important role of mitochondrial impairments in the occurrence of T2DM-associated cognitive dysfunction. We found, based on reconfirmation of the improvement of NaB on cognitive impairment, that NaB treatment improved damaged synaptic structural plasticity including the decrease in dendritic spine density and downregulation in the expression of postsynaptic density protein 95 and synaptophysin in the hippocampus in the model mice. NaB treatment also ameliorated mitochondrial ultrastructural damage, increased mitochondrial membrane potential and adenosine 5'-triphosphate content, and improved mitochondrial biogenesis and dynamics in the model mice. Furthermore, the expression of phosphorylated AMPK and PGC-1α was upregulated after NaB treatment in the model mice. In particular, the above beneficial effects of NaB were blocked by the inhibition of either AMPK or PGC-1α. In conclusion, NaB treatment improved cognitive impairment and damaged synaptic structural plasticity in the hippocampus by ameliorating damage to mitochondrial morphology and function through regulating the AMPK/PGC-1α pathway in HFD/STZ-induced T2DM mice.
Collapse
Affiliation(s)
- Li-Li Lu
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, PR China; Department of Pathology, The Third Hospital of Shijiazhuang, 15 Tiyu South Avenue, Shijiazhuang, 050011, PR China
| | - Li-Zhe Liu
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, PR China
| | - Li Li
- Central Laboratory, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, PR China
| | - Yu-Yan Hu
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, PR China
| | - Xiao-Hui Xian
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, PR China.
| | - Wen-Bin Li
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, PR China.
| |
Collapse
|
46
|
Robles-Rivera RR, Pacheco-Moisés FP, Olvera-Montaño C, Castellanos-González JA, Barley-Villaseñor AL, Cardona-Muñoz EG, Rodríguez-Carrizalez AD. Mitochondrial Function and Oxidative Stress Biomarkers in Diabetic Retinopathy Development: An Analytical Cross-Sectional Study. Int J Mol Sci 2024; 25:13084. [PMID: 39684793 DOI: 10.3390/ijms252313084] [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: 11/04/2024] [Revised: 11/30/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
DR is a complex complication of DM with multiple biochemical pathways implicated in its genesis and progression. Circulating OS and mitochondrial function biomarkers represent potential candidates in the DR staging system. We conducted a comparative cross-sectional study comparing the OS biomarkers: TAC, GR, NOS, CARB, and hydroperoxydes, as well as mitochondrial function biomarkers: ATP synthase and ATPase activity in healthy volunteers, DM w/o DR, Moderate and Severe NPDR, and PDR. TAC is progressively diminished the more DR progresses to its proliferative stages. GR and NOS may function as biomarkers to differentiate the progression from S NPDR to PDR. CARB may correlate with the progression from M NPDR to S NPDR. Hydroperoxide levels were higher in patients with DR compared to DM w/o DR expressing OS in the early development of DR. ATPase activity is increasingly augmented the more DR progresses and may function as a biomarker that reflects the difference between N PDR and PDR, and ATP synthesis was lower the more DR progressed, being significantly lower compared to DM w/o DR. The behavior of OS and mitochondrial function in several stages of DR may aid in the staging and the prognosis of DR.
Collapse
Affiliation(s)
- Ricardo Raúl Robles-Rivera
- Institute of Clinical and Experimental Therapeutics, Department of Physiology, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Fermín Paul Pacheco-Moisés
- Department of Chemistry, University Centre of Exact and Engineering Sciences, University of Guadalajara, Guadalajara 44430, Jalisco, Mexico
| | - Cecilia Olvera-Montaño
- Institute of Clinical and Experimental Therapeutics, Department of Physiology, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - José Alberto Castellanos-González
- Department of Ophthalmology, Specialties Hospital of the National Occidental Medical Center, Mexican Institute of Social Security, Guadalajara 44349, Jalisco, Mexico
| | - Andre Leonardo Barley-Villaseñor
- Institute of Clinical and Experimental Therapeutics, Department of Physiology, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Ernesto Germán Cardona-Muñoz
- Institute of Clinical and Experimental Therapeutics, Department of Physiology, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Adolfo Daniel Rodríguez-Carrizalez
- Institute of Clinical and Experimental Therapeutics, Department of Physiology, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
47
|
Wei X, Zhang X, Peng Y, Wu J, Mo H, An Z, Deng X, Peng Y, Liu L, Jiang W, Chen J, Hu Z, Wang Z, Zhuo L. Identification of a novel 10-hydroxyevodiamine prodrug as a potent topoisomerase inhibitor with improved aqueous solubility for treatment of hepatocellular carcinoma. Eur J Med Chem 2024; 279:116807. [PMID: 39243453 DOI: 10.1016/j.ejmech.2024.116807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/09/2024]
Abstract
Natural product evodiamine (Evo) and its synthetic derivatives represent an attractive dual Topo 1/2 inhibitors with broad-spectrum antitumor efficacy. However, the clinical applications of these compounds have been impeded by their poor aqueous solubility. Herein, a series of water-soluble 10-substituted-N(14)-phenylevodiamine derivatives were designed and synthesized. The most potent compound 45 featuring a quaternary ammonium salt fragment achieved robust aqueous solubility and nanomolar potency against a panel of human hepatoma cell lines Huh7, HepG2, SK-Hep-1, SMMC-7721, and SMMC-7721/DOX (doxorubicin-resistant cell). Further studies revealed that 45 could inhibit Topo 1 and Topo 2, induce apoptosis, arrest the cell cycle at the G2/M stage and inhibit the migration and invasion. Compound 45 exhibited potent antitumor activity (TGI = 51.1 %, 10 mg/kg) in the Huh7 xenograft model with acceptable safety profile. In addition, a 21-day long-term dose toxicity study confirmed that the maximum tolerated dose of compound 45 was 20 mg/kg. Overall, this study presented a promising Evo-derived candidate for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xiuzhen Wei
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xi Zhang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yan Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Junbo Wu
- Department of Colorectal Surgery, Hengyang Central Hospital, Hengyang, Hunan, 421001, China
| | - Hanxuan Mo
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zhigang An
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xinyu Deng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Ying Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Linyi Liu
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Weifan Jiang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jinjin Chen
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zecheng Hu
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Zhen Wang
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, 810008, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410008, China; MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, China.
| | - Linsheng Zhuo
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
48
|
Xiang H, Lyu Q, Chen S, Ouyang J, Xiao D, Liu Q, Long H, Zheng X, Yang X, Lu H. PACS2/CPT1A/DHODH signaling promotes cardiomyocyte ferroptosis in diabetic cardiomyopathy. Cardiovasc Diabetol 2024; 23:432. [PMID: 39633391 PMCID: PMC11619700 DOI: 10.1186/s12933-024-02514-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/17/2024] [Indexed: 12/07/2024] Open
Abstract
OBJECTIVES The pathophysiology of diabetic cardiomyopathy (DCM) is a phenomenon of great interest, but its clinical problems have not yet been effectively addressed. Recently, the mechanism of ferroptosis in the pathophysiology of various diseases, including DCM, has attracted widespread attention. Here, we explored the role of PACS2 in ferroptosis in DCM through its downregulation of PACS2 expression. METHODS AND RESULTS Cardiomyocytes were treated with high glucose and palmitic acid (HGPA), and the detection of cardiomyocyte iron ions, lipid peroxides, and reactive oxygen species (ROS) revealed clear ferroptosis during these treatments. Silencing PACS2 downregulated CPT1A expression and upregulated DHODH expression significantly, reversing HGPA-induced ferroptosis. Further silencing of PACS2 with a CPT1A agonist exacerbated cardiomyocyte ferroptosis while promoting mitochondrial damage in cardiomyocytes. Using a mouse model of type 2 diabetes induced by streptozotocin (STZ) and a high-fat diet (HFD), we found that PACS2 deletion reversed these treatment-induced increases in cellular iron ions, impaired cardiac function, mitochondrial damage and ferroptosis in cardiac muscle tissues. CONCLUSIONS The PACS2/CPT1A/DHODH signalling pathway may be involved in ferroptosis in DCM by regulating cardiomyocyte mitochondrial function.
Collapse
MESH Headings
- Animals
- Ferroptosis/drug effects
- Diabetic Cardiomyopathies/pathology
- Diabetic Cardiomyopathies/metabolism
- Diabetic Cardiomyopathies/enzymology
- Diabetic Cardiomyopathies/physiopathology
- Diabetic Cardiomyopathies/genetics
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Signal Transduction
- Carnitine O-Palmitoyltransferase/metabolism
- Carnitine O-Palmitoyltransferase/genetics
- Mice, Inbred C57BL
- Diabetes Mellitus, Experimental/enzymology
- Male
- Mice, Knockout
- Mitochondria, Heart/pathology
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/drug effects
- Reactive Oxygen Species/metabolism
- Palmitic Acid/pharmacology
- Diabetes Mellitus, Type 2/enzymology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Mice
- Diet, High-Fat
Collapse
Affiliation(s)
- Hong Xiang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan, China
- Center for Experimental Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Qi Lyu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Shuhua Chen
- Department of Biochemistry, School of Life Sciences of Central South University, Changsha, Hunan, China
| | - Jie Ouyang
- Department of Cardiology, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Di Xiao
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Quanjun Liu
- Department of Cardiology, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - HaiJiao Long
- Department of Cardiology, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xinru Zheng
- Department of Cardiology, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiaoping Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan, China.
| | - Hongwei Lu
- Center for Experimental Medicine, The Third Xiangya Hospital of Central South University, Changsha, China.
- Department of Cardiology, Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
49
|
Al-Suhaimi E, AlQuwaie R, AlSaqabi R, Winarni D, Dewi FRP, AlRubaish AA, Shehzad A, Elaissari A. Hormonal orchestra: mastering mitochondria's role in health and disease. Endocrine 2024; 86:903-929. [PMID: 39172335 DOI: 10.1007/s12020-024-03967-1] [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: 05/17/2024] [Accepted: 07/10/2024] [Indexed: 08/23/2024]
Abstract
Mitochondria is a subcellular organelle involved in the pathogenesis of cellular stress, immune responses, differentiation, metabolic disorders, aging, and death by regulating process of fission, fusion, mitophagy, and transport. However, an increased interest in mitochondria as powerhouse for ATP production, the mechanisms of mitochondria-mediated cellular dysfunction in response to hormonal interaction remains unknown. Mitochondrial matrix contains chaperones and proteases that regulate intrinsic apoptosis pathway through pro-apoptotic Bcl-2 family's proteins Bax/Bak, and Cyt C release, and induces caspase-dependent and independent cells death. Energy and growth regulators such as thyroid hormones have profound effect on mitochondrial inner membrane protein and lipid compositions, ATP production by regulating oxidative phosphorylation system. Mitochondria contain cholesterol side-chain cleavage enzyme, P450scc, ferredoxin, and ferredoxin reductase providing an essential site for steroid hormones biosynthesis. In line with this, neurohormones such as oxytocin, vasopressin, and melatonin are correlated with mitochondrial integrity, displaying therapeutic implications for inflammatory and immune responses. Melatonin's also displayed protective role against oxidative stress and mitochondrial synthesis of ROS, suggesting a defense mechanism against aging-related diseases. An imbalance in mitochondrial bioenergetics can cause neurodegenerative disorders, cardiovascular diseases, and cancers. Hormone-induced PGC-1α stimulates mitochondrial biogenesis via activation of NRF1 and NRF2, which in turn triggers mtTFA in brown adipose and cardiac myocytes. Mitochondria can be transferred through cells merging, exosome-mediated transfer, and tunneling through nanotubes. By delineating the underlying molecular mechanism of hormonal mitochondrial interaction, this study reviews the dynamics mechanisms of mitochondria and its effects on cellular level, health, diseases, and therapeutic strategies targeting mitochondrial diseases.
Collapse
Affiliation(s)
- Ebtesam Al-Suhaimi
- Vice presidency for Scientific Research and Innovation, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.
- King Abdulaziz and his Companions Foundation for Giftedness and Creativity "Mawhiba", Riyadh, Saudi Arabia.
| | - Rahaf AlQuwaie
- Master Program of Biotechnology, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Reem AlSaqabi
- Master Program of Biotechnology, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Dwi Winarni
- Department of Biology, Faculty of Science and Technology, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Firli Rahmah Primula Dewi
- Department of Biology, Faculty of Science and Technology, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Abdullah A AlRubaish
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Adeeb Shehzad
- Biodiversity Unit, Research Center, Dhofar University, Salalah, Oman
| | | |
Collapse
|
50
|
He J, Qin W, Zhang Y, Yan J, Han X, Gao J, Li Q, Jiao K. Upregulated Mitochondrial Dynamics Is Responsible for the Procatabolic Changes of Chondrocyte Induced by α2-Adrenergic Signal Activation. Cartilage 2024; 15:440-452. [PMID: 37646151 PMCID: PMC11520003 DOI: 10.1177/19476035231189841] [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] [Indexed: 09/01/2023] Open
Abstract
OBJECTIVE Activation of sympathetic tone is important for cartilage degradation in osteoarthritis (OA). Recent studies reported that sympathetic signals can affect the mitochondrial function of target cells. It is unknown whether this effect exits in chondrocytes and affects chondrocyte catabolism. The contribution of mitochondrial dynamics in the activation of α2-adrenergic signal-mediated chondrocyte catabolism was investigated in this study. DESIGN Primary chondrocytes were stimulated with norepinephrine (NE) alone, or pretreated with an α2-adrenergic receptor (Adra2) antagonist (yohimbine) and followed by stimulation with NE. Changes in chondrocyte metabolism and their mitochondrial dynamics were investigated. RESULTS We demonstrated that NE stimulation induced increased gene and protein expressions of matrix metalloproteinase-3 and decreased level of aggrecan by chondrocytes. This was accompanied by upregulated mitochondriogenesis and the number of mitochondria, when compared with the vehicle-treated controls. Mitochondrial fusion and fission, and mitophagy also increased significantly in response to NE stimulation. Inhibition of Adra2 attenuated chondrocyte catabolism and mitochondrial dynamics induced by NE. CONCLUSIONS The present findings indicate that upregulation of mitochondrial dynamics through mitochondriogenesis, fusion, fission, and mitophagy is responsible for activation of α2-adrenergic signal-mediated chondrocyte catabolism. The hypothesis that "α2-adrenergic signal activation promotes cartilage degeneration in temporomandibular joint osteoarthritis (TMJ-OA) by upregulating mitochondrial dynamics in chondrocytes" is validated. This represents a new regulatory mechanism in the chondrocytes of TMJ-OA that inhibits abnormal activation of mitochondrial fusion and fission is a potential regulator for improving mitochondrial function and inhibiting chondrocyte injury and contrives a potentially innovative therapeutic direction for the prevention of TMJ-OA.
Collapse
Affiliation(s)
- Jiaying He
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Wenpin Qin
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Yusong Zhang
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Jianfei Yan
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Xiaoxiao Han
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
- The College of Life Sciences, Northwest University, Xi’an, China
| | - Jialu Gao
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Qihong Li
- Department of Stomatology, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Kai Jiao
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| |
Collapse
|