1
|
Shalaby YM, Nakhal MM, Afandi B, Al-Zohily B, Majed L, Kumar KK, Emerald BS, Sadek B, Akour A, Akawi N. Impact of sodium-glucose cotransporter-2 inhibitors on aging biomarkers and plasma ceramide levels in type 2 diabetes: beyond glycemic control. Ann Med 2025; 57:2496795. [PMID: 40289660 PMCID: PMC12039402 DOI: 10.1080/07853890.2025.2496795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 04/07/2025] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Aging is a complex biological process marked by the decline of physiological functions and heightened susceptibility to chronic illnesses, notably cardiometabolic disorders. Ceramides (Cer) are lipid derivatives linked to aging and metabolic diseases. Sodium-Glucose Cotransporter-2 inhibitors (SGLT2i), widely used in managing type 2 diabetes, have an unclear impact on aging biomarkers and Cer profiles. OBJECTIVE This study explored the association between SGLT2i use, plasma Cer levels (CerC16:0, CerC18:0, CerC22:0, CerC24:0, and CerC24:1), and aging biomarkers-Human Insulin-Like Growth Factor 1 (IGF-1), mammalian target of rapamycin (mTOR), 5-Methylcytosine (5MC), and Human H2AFX (Histone H2AX) in patients with type 2 diabetes mellitus (T2DM). METHODS In this retrospective study, 95 participants were divided into three groups: patients on SGLT2i (n = 34), patients on non-SGLT2i anti-diabetic treatments (n = 36), and healthy controls (n = 25). Plasma Cer and aging biomarkers were quantified using Liquid Chromatography with tandem mass spectrometry (LC-MS-MS) and ELISA, respectively. Principal component analysis (PCA) assessed group-based clustering, while ANCOVA evaluated group differences with confounder adjustment. RESULTS SGLT2i-treated patients showed significantly lower CerC16:0, CerC22:0, and CerC24:1 levels (p < 0.01) and decreased 5MC and H2AX (p < 0.05) compared to non-SGLT2i patients. IGF-1 was significantly elevated in the SGLT2i group (p < 0.01), suggesting a possible protective effect on metabolic health. PCA distinguished control from diabetic groups but revealed overlap between SGLT2i and non-SGLT2i groups. CONCLUSION Beyond glucose control, SGLT2i may improve plasma Cer and aging markers in diabetic patients, supporting their broader therapeutic potential in aging and age-related diseases. Further large-scale studies are warranted to confirm these effects and underlying mechanisms.
Collapse
Affiliation(s)
- Youssef M. Shalaby
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Mohammed Moutaz Nakhal
- Department of Pharmacology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Bachar Afandi
- Department of Endocrinology, Tawam Hospital, Al-Ain, UAE
- Department of Medicine, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Bashar Al-Zohily
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Lina Majed
- Department of Endocrinology, Tawam Hospital, Al-Ain, UAE
| | - Kukkala Kiran Kumar
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE
| | - Bassem Sadek
- Department of Pharmacology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Amal Akour
- Department of Pharmacology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Nadia Akawi
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK
| |
Collapse
|
2
|
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
|
3
|
Alotaibi G, Alkhammash A. Pharmacological landscape of endoplasmic reticulum stress: Uncovering therapeutic avenues for metabolic diseases. Eur J Pharmacol 2025; 998:177509. [PMID: 40089262 DOI: 10.1016/j.ejphar.2025.177509] [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: 11/02/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025]
Abstract
The endoplasmic reticulum (ER) plays a fundamental role in maintaining cellular homeostasis by ensuring proper protein folding, lipid metabolism, and calcium regulation. However, disruptions to ER function, known as ER stress, activate the unfolded protein response (UPR) to restore balance. Chronic or unresolved ER stress contributes to metabolic dysfunctions, including insulin resistance, non-alcoholic fatty liver disease (NAFLD), and neurodegenerative disorders such as Parkinson's disease and Alzheimer's disease. Recent studies have also highlighted the importance of mitochondria-ER contact sites (MERCs) and ER-associated inflammation in disease progression. This review explores the current pharmacological landscape targeting ER stress, focusing on therapeutic strategies for rare metabolic and neurodegenerative diseases. It examines small molecules such as tauroursodeoxycholic acid (TUDCA) and 4-phenylbutyric acid (4-PBA), repurposed drugs like 17-AAG (17-N-allylamino-17demethoxygeldanamycin (tanespimycin)) and berberine, and phytochemicals such as resveratrol and hesperidin. Additionally, it discusses emerging therapeutic areas, including soluble epoxide hydrolase (sEH) inhibitors for metabolic disorders and MERCs modulation for neurological diseases. The review emphasizes challenges in translating these therapies to clinical applications, such as toxicity, off-target effects, limited bioavailability, and the lack of large-scale randomized controlled trials (RCTs). It also highlights the potential of personalized medicine approaches and pharmacogenomics in optimizing ER stress-targeting therapies.
Collapse
Affiliation(s)
- Ghallab Alotaibi
- Department of Pharmacology, College of Pharmacy, Shaqra University, Shaqra, 11961, Saudi Arabia.
| | - Abdullah Alkhammash
- Department of Pharmacology, College of Pharmacy, Shaqra University, Shaqra, 11961, Saudi Arabia.
| |
Collapse
|
4
|
Kidess GG, Hamza M, Goru R, Basit J, Alraiyes M, Alraies MC. The Impact of Sodium-Glucose Cotransporter-2 Inhibitors on Atrial Fibrillation Burden in Diabetic Patients. Am J Cardiol 2025; 246:65-70. [PMID: 40058622 DOI: 10.1016/j.amjcard.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/22/2025] [Accepted: 03/03/2025] [Indexed: 03/26/2025]
Abstract
Atrial Fibrillation (AF) and Type 2 Diabetes Mellitus (T2DM) are comorbid conditions associated with increased adverse outcomes. Recent evidence suggests that antidiabetic therapies such as sodium-glucose co-transporter-2 inhibitors (SGLT2i) and glucagon-like peptide-1 agonists (GLP1a) may influence the risk of AF and stroke differently. This study aims to compare the risk of new-onset AF and stroke in T2DM patients treated with SGLT2i versus GLP1a. A systematic literature review was performed on Pubmed and Embase, including studies comparing the effect of SGLT2i or GLP1a on new-onset AF and stroke incidence in T2DM patients. A random effects model was used to pool relative risk and 95% confidence intervals to assess the study outcomes. Univariate metaregression analysis was performed for selected demographics and comorbidities. Six observational studies were included in the analysis comprising 847,028 patients. Our meta-analysis found a significantly lower risk of new-onset AF in patients with T2DM treated with SGLT2i compared to those receiving GLP1a (RR = 0.76, 95% CI: 0.65 to 0.89). There was no statistically significant difference in the risk of stroke between SGLT2i and GLP1a (RR = 1.09, 95% CI = 0.98 to 1.21). Univariate meta-regression indicated male sex was a significant negative effect modifier for new-onset AF (coefficient = -0.0191, p-value = 0.0158). In conclusion, SGLT2i may reduce AF risk in T2DM patients, while GLP1a may provide a modest, nonsignificant protective effect against stroke. Further research is needed to confirm these results and guide cardiovascular risk management in patients with T2DM.
Collapse
Affiliation(s)
- George G Kidess
- Department of Medicine, Wayne State University School of Medicine, Detroit, Michigan
| | - Mohammad Hamza
- Department of Medicine, Guthrie Medical Group, Cortland, New York
| | - Rohit Goru
- Department of Medicine, Wayne State University School of Medicine, Detroit, Michigan
| | - Jawad Basit
- Department of Medicine, Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Mowaffak Alraiyes
- Interventional Cardiology Research Department, Wayne State School of Medicine, Detroit Medical Center, Detroit, Michigan
| | - M Chadi Alraies
- Cardiovascular Institute, Detroit Medical Center, DMC Heart Hospital, Detroit, Michigan.
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
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
|
8
|
Skawratananond S, Xiong DX, Zhang C, Tonk S, Pinili A, Delacruz B, Pham P, Smith SC, Navab R, Reddy PH. Mitophagy in Alzheimer's disease and other metabolic disorders: A focus on mitochondrial-targeted therapeutics. Ageing Res Rev 2025; 108:102732. [PMID: 40122398 DOI: 10.1016/j.arr.2025.102732] [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: 11/15/2024] [Revised: 02/19/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Mitochondria, as central regulators of cellular processes such as energy production, apoptosis, and metabolic homeostasis, are essential to cellular function and health. The maintenance of mitochondrial integrity, especially through mitophagy-the selective removal of impaired mitochondria-is crucial for cellular homeostasis. Dysregulation of mitochondrial function, dynamics, and biogenesis is linked to neurodegenerative and metabolic diseases, notably Alzheimer's disease (AD), which is increasingly recognized as a metabolic disorder due to its shared pathophysiologic features: insulin resistance, oxidative stress, and chronic inflammation. In this review, we highlight recent advancements in pharmacological interventions, focusing on agents that modulate mitophagy, mitochondrial uncouplers that reduce oxidative phosphorylation, compounds that directly scavenge reactive oxygen species to alleviate oxidative stress, and molecules that ameliorate amyloid beta plaque accumulation and phosphorylated tau pathology. Additionally, we explore dietary and lifestyle interventions-MIND and ketogenic diets, caloric restriction, physical activity, hormone modulation, and stress management-that complement pharmacological approaches and support mitochondrial health. Our review underscores mitochondria's central role in the pathogenesis and potential treatment of neurodegenerative and metabolic diseases, particularly AD. By advocating for an integrated therapeutic model that combines pharmacological and lifestyle interventions, we propose a comprehensive approach aimed at mitigating mitochondrial dysfunction and improving clinical outcomes in these complex, interrelated diseases.
Collapse
Affiliation(s)
- Shadt Skawratananond
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Daniel X Xiong
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, United States.
| | - Charlie Zhang
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Sahil Tonk
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Aljon Pinili
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Brad Delacruz
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Patrick Pham
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Shane C Smith
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Rahul Navab
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Internal Medicine, PES Institute of Medical Sciences and Research, Kuppam, India.
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX 79409, United States; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
9
|
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
|
10
|
Lao XY, Sun YL, Zhao ZJ, Liu J, Ruan XF. Pharmacological effects of betulinic acid and its protective mechanisms on the cardiovascular system. Fitoterapia 2025; 183:106561. [PMID: 40288588 DOI: 10.1016/j.fitote.2025.106561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/09/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Betulinic acid (BA), a pentacyclic triterpenoid saponin widely found in plants, has attracted attention for its diverse pharmacological activities. Recent studies highlight its cardioprotective potential, promoting its relevance in cardiovascular research. AIM OF THE REVIEW This review summarizes BA's physicochemical properties, structure-activity relationships, natural sources, and synthesis strategies. It further discusses its pharmacokinetics and toxicity to evaluate its drug development potential, with emphasis on cardioprotective effects and related signaling pathways. METHODS Literature was collected from databases such as PubMed and Web of Science, focusing on studies addressing BA's chemical characteristics, biological activities, pharmacokinetics, and cardiovascular relevance. RESULTS BA exerts cardioprotective effects via multiple signaling pathways, including NRF2, NF-κB, MAPK, and NFAT. These contribute to its antioxidant, anti-inflammatory, anti-apoptotic, and anti-proliferative actions, as well as its enhancement of endothelial function through nitric oxide signaling. BA also reduces lipid accumulation. Combined with favorable physicochemical properties and synthetic accessibility, these findings support BA as a promising multifunctional lead compound in cardiovascular pharmacology. CONCLUSION BA shows strong potential as a cardioprotective natural compound. Although further research is needed to validate its clinical efficacy and safety, its multi-target actions and structural versatility provide a solid basis for development in cardiovascular drug discovery.
Collapse
Affiliation(s)
- Xu Yuan Lao
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuan Long Sun
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Zhe Jun Zhao
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jie Liu
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiao Fen Ruan
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
11
|
Gjermeni E, Fiebiger R, Bundalian L, Garten A, Schöneberg T, Le Duc D, Blüher M. The impact of dietary interventions on cardiometabolic health. Cardiovasc Diabetol 2025; 24:234. [PMID: 40450314 DOI: 10.1186/s12933-025-02766-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 04/29/2025] [Indexed: 06/03/2025] Open
Abstract
Obesity and cardiometabolic diseases are leading causes of morbidity and mortality among adults worldwide. These conditions significantly contribute to and exacerbate other major causes of illness and death, including cancer, neurodegenerative diseases, and chronic kidney disease. The growing burden of these diseases has increased the interest of modern medicine in understanding metabolic processes and health, with diet emerging as a pivotal modifiable factor, alongside physical inactivity and smoking. In this review, we discuss the pathophysiological and evolutionary foundations of metabolic processes that may link "unhealthy" nutrition to obesity and cardiometabolic diseases and review the current literature to assess the effects of various diet interventions and patterns on cardiometabolic parameters. Special emphasis is placed on summarizing the latest, albeit partially contradictory, evidence to offer balanced dietary recommendations with the ultimate aim to improve cardiometabolic health.
Collapse
Affiliation(s)
- Erind Gjermeni
- Department of Cardiology, Median Center for Rehabilitation Schmannewitz, 04774, Dahlen, Germany.
| | - Raluca Fiebiger
- Department of Cardiology, Median Center for Rehabilitation Schmannewitz, 04774, Dahlen, Germany
| | - Linnaeus Bundalian
- Institute of Human Genetics, University Medical Center Leipzig, 04103, Leipzig, Germany
| | - Antje Garten
- Pediatric Research Center, University Hospital for Children and Adolescents, Leipzig University, 04103, Leipzig, Germany
| | - Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
- School of Medicine, University of Global Health Equity, Kigali, Rwanda
| | - Diana Le Duc
- Institute of Human Genetics, University Medical Center Leipzig, 04103, Leipzig, Germany
- Department of Genetics, Center for Diagnostics at Chemnitz Clinics, 09116, Chemnitz, Germany
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, 04103, Leipzig, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103, Leipzig, Germany
| |
Collapse
|
12
|
Zhang X, Peng N, Zhang X, Zhu Z, Miao Y, Wu Y, Ling J, Li C, Gu W, Zhang J, Ayiguli A, Zheng Z, Yu P, Liu X. Association of glucagon-like peptide-1 receptor agonists with atrial fibrillation, cardiac arrest, and ventricular fibrillation: Casual evidence from a drug target Mendelian randomization. Diabetol Metab Syndr 2025; 17:179. [PMID: 40442813 PMCID: PMC12123724 DOI: 10.1186/s13098-025-01712-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/22/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Glucagon-like peptide-1 receptor agonists (GLP-1RAs) have shown benefits for cardiorenal outcomes in patients with type 2 diabetes mellitus. Although some observational studies suggest that GLP-1RAs protect against arrhythmias, the evidence remains inconclusive. METHODS This study aimed to assess the causal relationship between GLP-1RAs and arrhythmias, including atrial fibrillation (AF), cardiac arrest, and ventricular fibrillation. We performed a two-sample Mendelian randomization (MR) analysis to examine the associations between genetically proxied GLP-1RAs and the risk of arrhythmias. Genetic instruments for GLP-1RAs were obtained from the cis-expression quantitative trait loci of the GLP1R gene, on the basis of data from the eQTLGen Consortium. Genome-wide association study (GWAS) data for AF were sourced from FinnGen10, whereas data for cardiac arrest and ventricular fibrillation came from the GWAS Catalog. Bayesian colocalization and multivariable Mendelian randomization (MVMR) analyses were conducted as supplementary analyses. RESULTS Twelve independent single nucleotide polymorphisms were identified as genetic instruments for GLP-1RAs. MR analysis indicated that genetically proxied GLP-1RAs were associated with a reduced risk of AF (odds ratio [OR] = 0.78, 95% confidence interval [CI] = 0.71-0.85, p = 4.45E-08, posterior probability of hypothesis 4 [PP.H4] = 0.007) and a lower risk of cardiac arrest and ventricular fibrillation (OR = 0.60, 95% CI = 0.42-0.85, p = 0.0039, PP.H4 = 0.018). Bayesian colocalization analysis revealed that genetically proxied GLP-1RAs did not share genetic variation with arrhythmias. MVMR analysis revealed that, after adjusting for body mass index and type 2 diabetes mellitus, genetically proxied GLP-1RAs did not have a significant effect on the risk of arrhythmias. CONCLUSIONS Our findings suggest that genetically proxied GLP-1RAs are causally associated with a reduced risk of AF, cardiac arrest, and ventricular fibrillation. Further randomized controlled trials are needed to confirm these results.
Collapse
Affiliation(s)
- Xinyi Zhang
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Nanqin Peng
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xiaoyue Zhang
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zicheng Zhu
- Department of Anesthesiology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yan Miao
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yuting Wu
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Chen Li
- Department of Pharmacy, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wenli Gu
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Jing Zhang
- Department of Anesthesiology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Abudukeremu Ayiguli
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ziheng Zheng
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Peng Yu
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| | - Xiao Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China.
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore.
| |
Collapse
|
13
|
Sato Y, Kato Y, Kanoke A, Sun JY, Nishijima Y, Wang RK, Stryker M, Endo H, Liu J. Type 2 diabetes abates retrograde collateral flow and promotes leukocyte adhesion following ischemic stroke. J Cereb Blood Flow Metab 2025:271678X251338203. [PMID: 40439073 PMCID: PMC12122487 DOI: 10.1177/0271678x251338203] [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: 10/22/2024] [Revised: 02/25/2025] [Accepted: 04/07/2025] [Indexed: 06/02/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is associated with impaired leptomeningeal collateral compensation and poor stroke outcome. Neutrophils tethering and rolling on endothelium after stroke can also independently reduce flow velocity. However, the chronology and topological changes in collateral circulation in T2DM is not yet defined. Here, we describe the spatial and temporal blood flow dynamics and vessel diameter changes in pial arteries and veins and leukocyte-endothelial adhesion following middle cerebral artery (MCA) stroke using two-photon microscopy in awake control and T2DM mice. Relative to control mice, T2DM mice already exhibited smaller pial vessels with reduced flow velocity prior to stroke. Following stroke, T2DM mice displayed persistently reduced blood flow in pial arteries and veins, resulting in a poor recovery of downstream penetrating arterial flow and a sustained deficit in microvascular flow. There was also persistent increase of leukocyte adhesion to the endothelium of veins, coincided with elevated neutrophils infiltration into brain parenchyma in T2DM mice compared to control mice after stroke. Our data suggest that T2DM-induced increase in inflammation and chronic remodeling of leptomeningeal vessels may contribute to the observed hemodynamics deficiency after stroke and subsequent poor stroke outcome.
Collapse
Affiliation(s)
- Yoshimichi Sato
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yuya Kato
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Atsushi Kanoke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Jennifer Y Sun
- Institute of Ophthalmology, University College London, London, UK
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Yasuo Nishijima
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ruikang K Wang
- Department of Bioengineering, College of Engineering and School of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Michael Stryker
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Hidenori Endo
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Jialing Liu
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
| |
Collapse
|
14
|
Reshadmanesh T, Mohebi R, Behnoush AH, Reshadmanesh A, Khalaji A, Norouzi M, Javanmardi E, Pishdad R, Jafarzadeh SR, Ghondaghsaz E, Chaparro S. The effects of sodium-glucose cotransporter-2 inhibitors in chemotherapy-induced cardiotoxicity and mortality in patients with cancer: a systematic review and meta-analysis. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:50. [PMID: 40426171 PMCID: PMC12107967 DOI: 10.1186/s40959-025-00343-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 05/01/2025] [Indexed: 05/29/2025]
Abstract
BACKGROUND The effects of sodium-glucose cotransporter-2 (SGLT2) inhibitors on reducing cardiovascular events in different subgroups of diabetic patients are under investigation. The current systematic review and meta-analysis investigated the effects of SGLT2 inhibitors on preventing cardiovascular events and mortality and their adverse events in patients with active cancer and diabetes undergoing cardiotoxic cancer treatment. METHODS We searched PubMed, Embase, Web of Science, and Scopus to find studies investigating the effects of SGLT2 inhibitors on patients with diabetes and confirmed cancer until 19 August 2024. Meta-analyses were conducted using the random-effects model to compare all-cause mortality, cancer-associated mortality, heart failure (HF) hospitalization, arrhythmia, and adverse event rates such as ketoacidosis, hypoglycemia, urinary tract infection, and sepsis between patients with or without SGLT2 inhibitors use. Risk ratios (RRs) with 95% confidence intervals (CI) were used to compare outcomes between SGLT2 inhibitors and non-SGLT2 inhibitors groups. RESULTS Eleven studies were included with 88,096 patients with confirmed cancer (49% male). Among the total population, 20,538 received SGLT2 inhibitors (age 61.68 ± 10.71), while 67,558 did not receive SGLT2 inhibitors (age 68.24 ± 9.48). The meta-analysis found that the patients who received SGLT2 inhibitors had a significantly lower mortality rate than those who did not receive SGLT2 inhibitors (RR 0.46, 95% CI 0.34 to 0.63, p-value < 0.0001). Similarly, the cancer-associated mortality rate was also lower (RR 0.29, 95% CI 0.27 to 0.30, p-value < 0.0001). Further analysis found that the SGLT2 inhibitor group had a lower rate of HF hospitalization, compared to controls (RR 0.44, 95% CI 0.27 to 0.70, p-value = 0.0007). Moreover, patients receiving SGLT2 inhibitors had a statistically lower rate of arrhythmia (RR 0.38, 95% CI 0.26 to 0.56, p-value < 0.0001). Finally, patients in the SGLT2 inhibitors group had a lower rate of adverse events (RR 0.51, 95% CI 0.42 to 0.62, p-value < 0.0001). CONCLUSIONS SGLT2 inhibitors are effective in reducing mortality (all-cause and cancer-associated), HF hospitalization, arrhythmia, and drug adverse events in patients with cancer. If confirmed in future studies, these agents could be a potentially ideal candidate to prevent cardiotoxicity of cancer therapies.
Collapse
Affiliation(s)
- Tara Reshadmanesh
- School of Medicine, Zanjan University of Medical Science, Zanjan, Iran
| | - Reza Mohebi
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Amir Hossein Behnoush
- School of Medicine, Tehran University of Medical Sciences, Poursina St., Keshavarz Blvd, Tehran, 1417613151, Iran.
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Azadeh Reshadmanesh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amirmohammad Khalaji
- School of Medicine, Tehran University of Medical Sciences, Poursina St., Keshavarz Blvd, Tehran, 1417613151, Iran
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mitra Norouzi
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Elmira Javanmardi
- School of Medicine, Zanjan University of Medical Science, Zanjan, Iran
| | - Reza Pishdad
- Division of Endocrinology, Diabetes, and Metabolism, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - S Reza Jafarzadeh
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Elina Ghondaghsaz
- Undergraduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Sandra Chaparro
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Miami Cardiac and Vascular Institute, Baptist Health South Florida, Miami, FL, USA
| |
Collapse
|
15
|
Yoo TT, Baek IH, Stoletniy L, Hilliard A, Sakr A, Doycheva D. Impact of sodium-glucose transport protein-2 (SGLT2) inhibitors on the inflammasome pathway in acute myocardial infarction in type 2 diabetes mellitus: a comprehensive review. Cardiovasc Diabetol 2025; 24:227. [PMID: 40420176 PMCID: PMC12105141 DOI: 10.1186/s12933-025-02777-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 05/06/2025] [Indexed: 05/28/2025] Open
Abstract
Sodium-glucose transport protein-2 (SGLT2) inhibitors, initially developed for glycemic control in type 2 diabetes mellitus (T2DM), have emerged as potential cardioprotective agents, reducing cardiovascular mortality and improving heart failure outcomes. Recent evidence suggests that SGLT2 inhibitors exert anti-inflammatory effects, particularly through modulating the inflammasome pathway. This review explores the role of the inflammasome in acute myocardial infarction (AMI) in T2DM and discusses the mechanisms by which SGLT2 inhibitors influence this pathway. We evaluate current studies on the impact of SGLT2 inhibitors on key inflammatory mediators, particularly the NLRP3 inflammasome, and discuss their potential therapeutic implications for reducing inflammation and myocardial injury in patients with T2DM experiencing AMI. In summary, the key novelties in this review lie in its focused mechanistic approach on the inflammasome pathway, its integration of diabetes and cardiovascular research, and its potential to influence future therapeutic strategies for AMI in T2DM patients. It offers a novel angle by tying together molecular mechanisms of inflammation with clinical implications in a specific patient population that faces high cardiovascular risk.
Collapse
Affiliation(s)
- Thomas T Yoo
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, CA, 92354, USA
| | - In Hae Baek
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, CA, 92354, USA
| | - Liset Stoletniy
- Division of Cardiology, School of Medicine, Loma Linda University, 11234 Anderson St, Loma Linda, CA, 92354, USA
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, CA, 92354, USA
| | - Anthony Hilliard
- Division of Cardiology, School of Medicine, Loma Linda University, 11234 Anderson St, Loma Linda, CA, 92354, USA
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, CA, 92354, USA
| | - Antoine Sakr
- Division of Cardiology, School of Medicine, Loma Linda University, 11234 Anderson St, Loma Linda, CA, 92354, USA
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, CA, 92354, USA
| | - Desislava Doycheva
- Division of Cardiology, School of Medicine, Loma Linda University, 11234 Anderson St, Loma Linda, CA, 92354, USA.
- Department of Physiology and Pharmacology, Loma Linda University, 11175 Campus St, Loma Linda, CA, 92354, USA.
| |
Collapse
|
16
|
Nuñez-Selles AJ, Nuñez-Musa RA, Guillen-Marmolejos RA. Linking oxidative stress biomarkers to disease progression and antioxidant therapy in hypertension and diabetes mellitus. Front Mol Biosci 2025; 12:1611842. [PMID: 40492113 PMCID: PMC12146192 DOI: 10.3389/fmolb.2025.1611842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Accepted: 05/07/2025] [Indexed: 06/11/2025] Open
Abstract
Oxidative stress (OS) is increasingly recognized as a key factor linking hypertension (HTN) and diabetes mellitus (DM). This review summarizes recent evidence regarding the dual role of OS as both an instigator and an amplifier of cardiometabolic dysfunction. In HTN, reactive oxygen species (ROS) produced by NADPH oxidases (NOXs) and mitochondrial dysfunction contribute to endothelial impairment and vascular remodeling. In DM, hyperglycemia-induced ROS production worsens beta-cell failure and insulin resistance through pathways such as the AGE-RAGE signaling, protein kinase C (PKC) activation, and the polyol pathway. Clinically validated biomarkers of OS, such as F2-isoprostanes (which indicate lipid peroxidation), 8-OHdG (which indicates DNA damage), and the activities of redox enzymes like superoxide dismutase (SOD) and glutathione peroxidase (GPx), show strong correlations with disease progression and end-organ complications. Despite promising preclinical results, the application of antioxidant therapies in clinical settings has faced challenges due to inconsistent outcomes, highlighting the need for targeted approaches. Emerging strategies include: 1. Mitochondria-targeted antioxidants to enhance vascular function in resistant HTN; 2. Nrf2 activators to restore redox balance in early diabetes; and 3. Specific inhibitors of NOX isoforms. We emphasize three transformative areas of research: (i) the interaction between the microbiome and ROS, where modifying gut microbiota can reduce systemic OS; (ii) the use of nanotechnology to deliver antioxidants directly to pancreatic islets or atherosclerotic plaques; and (iii) phenotype-specific diagnosis and therapy guided by redox biomarkers and genetic profiling (for example, KEAP1/NRF2 polymorphisms). Integrating these advances with lifestyle modifications, such as following a Mediterranean diet and exercising regularly, may provide additional benefits. This review outlines a mechanistic framework for targeting OS in the comorbidity of HTN and DM while identifying critical knowledge gaps, particularly regarding the timing of antioxidant signaling and the development of personalized redox medicine, which may serve as a reference for researchers and clinicians working in this area.
Collapse
Affiliation(s)
- Alberto J. Nuñez-Selles
- Research Division, Universidad Nacional “Pedro Henríquez Ureña” (UNPHU), Santo Domingo, Dominican Republic
| | | | | |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Zhang F, Han Y, Li F, Guo B, Chen J, Zhou W, Xiao P, Ma H, Jin Y, Feng J, Min Y. Egg exosome miR-145-5p decreases mitochondrial ROS to protect chicken embryo hepatocytes against apoptosis through targeting MAPK10. J Anim Sci Biotechnol 2025; 16:74. [PMID: 40410809 PMCID: PMC12103047 DOI: 10.1186/s40104-025-01203-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/31/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND Higher embryonic mortality, especially in aged breeding hens, is associated with insufficient hepatic functionality in maintaining redox homeostasis. Our previous study demonstrated that egg exosome-derived miRNAs may play a key role in modulating embryonic oxidation-reduction process, whereas the exact function and mechanism were still poorly understood. The present study aimed to investigate the roles of egg exosome miRNAs in maintaining dynamic equilibrium of free radicals and peroxide agents in embryonic liver, as well as demonstrate the specific mechanism using oxidative stress-challenged hepatocytes. RESULTS Compared to 36-week-old breeding hens, decreased hatchability and increased embryonic mortality were observed in 65-week-old breeding hens. Meanwhile, the older group showed the increased MDA levels and decreased SOD and GSH-Px activities in embryonic liver, muscle and serum. Embryonic mortality was significantly positively correlated with MDA level and negatively correlated with GSH-Px activity in embryonic liver. In addition, 363 differentially expressed genes (DEGs) were identified in embryonic liver, 13 differentially expressed miRNAs (DE-miRNAs) were identified in egg exosomes. These DEGs and DE-miRNAs were involved in oxidoreductase activity, glutathione metabolic process, MAPK signaling pathway, apoptosis and autophagy. miRNA-mRNA network analysis further found that DEGs targeted by DE-miRNAs were mainly enriched in programmed cell death, such as apoptosis and autophagy. Wherein, MAPK10 with highest MCC and AUC values was significantly related to GSH-Px activity and MDA level, and served as the target gene of miR-145-5p based on dual luciferase reporter experiment and correlation analysis. Bioinformatics analysis found that miR-145-5p/MAPK10 axis might alleviate peroxide generation and apoptosis. In primary hepatocytes of chick embryos, miR-145-5p transfection significantly reversed H2O2-induced mitochondrial ROS increase, MAPK10, BAX and CASP3 overexpression and excessive apoptosis. CONCLUSION Exosome miR-145-5p in eggs could target MAPK10 and decrease mitochondrial ROS, attenuating oxidative damage and apoptosis in hepatocytes of chick embryos. These findings may provide new theoretical basis for the improvement of maternal physiological status to maintain embryonic redox homeostasis by nutritional or genetic modifications.
Collapse
Affiliation(s)
- Fengdong Zhang
- College of Animal Science and Technology, Northwest A&F University, Shaanxi, 712100, China
| | - Yongchang Han
- College of Animal Science and Technology, Northwest A&F University, Shaanxi, 712100, China
| | - Fan Li
- College of Animal Science and Technology, Northwest A&F University, Shaanxi, 712100, China
| | - Boya Guo
- College of Animal Science and Technology, Northwest A&F University, Shaanxi, 712100, China
| | - Jian Chen
- College of Animal Science and Technology, Northwest A&F University, Shaanxi, 712100, China
| | - Wenchuan Zhou
- College of Animal Science and Technology, Northwest A&F University, Shaanxi, 712100, China
| | - Pan Xiao
- College of Animal Science and Technology, Northwest A&F University, Shaanxi, 712100, China
| | - Hui Ma
- College of Animal Science and Technology, Northwest A&F University, Shaanxi, 712100, China
| | - Yongyan Jin
- College of Animal Science and Technology, Northwest A&F University, Shaanxi, 712100, China
| | - Jia Feng
- College of Animal Science and Technology, Northwest A&F University, Shaanxi, 712100, China.
| | - Yuna Min
- College of Animal Science and Technology, Northwest A&F University, Shaanxi, 712100, China.
| |
Collapse
|
20
|
Huang M, Liu Y, Chen C. SGLT2 inhibition mitigates intracerebral hemorrhage risk by modulating inflammation. Sci Rep 2025; 15:17770. [PMID: 40404687 PMCID: PMC12098824 DOI: 10.1038/s41598-024-84118-4] [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: 10/06/2024] [Accepted: 12/19/2024] [Indexed: 05/24/2025] Open
Abstract
Sodium-glucose cotransporter 2 (SGLT-2) inhibitors have been increasingly recognized for their potential neuroprotective properties. Nevertheless, their effects on intracerebral hemorrhage (ICH) are still debated, with the precise mechanisms involved remaining unclear. Recent studies indicate that these inhibitors might lower ICH risk through anti-inflammatory mechanisms. This study analyzed genome-wide association study (GWAS) data from individuals of European ancestry, focusing on the relationship between 92 inflammatory biomarkers and ICH. A two-sample, two-step Mendelian randomization (MR) approach was used to examine the potential connection between SGLT-2 inhibition and ICH, as well as to investigate whether inflammatory biomarkers mediate this relationship. Genetic proxies for SGLT-2 inhibition were determined based on variants linked to the expression of the SLC5A2 gene and levels of glycated hemoglobin (HbA1c). Odds ratios with 95% confidence intervals were calculated to assess the associations between SGLT-2 inhibition, inflammatory biomarkers, and the likelihood of ICH. The genetic prediction of SGLT-2 inhibition was found to be inversely related to the risk of ICH (OR = 0.152; 95% CI = 0.066-0.352; P < 0.001). Out of the 92 inflammatory biomarkers examined, 33 showed a significant association with SGLT-2 inhibition. Notably, levels of IL10 receptor subunit beta (IL10RB) were significantly correlated with both SGLT-2 inhibition and ICH. Moreover, IL10RB accounted for 9.167% of the total mediation effect of SGLT-2 inhibition on ICH. The findings of this study suggest a link between SGLT-2 inhibition and a decreased risk of ICH, with IL10RB emerging as a possible mediator. This presents a potential new strategy for ICH prevention and intervention. Further studies are needed to clarify the role of inflammatory pathways in the connection between SGLT-2 inhibition and ICH.
Collapse
Affiliation(s)
- Mingsheng Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiheng Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Cheng Chen
- Department of Intensive Care Unit, JianYang Hospital of Traditional Chinese Medicine, 421 Xiongzhou Avenue South Section, Jianyang, 641499, Sichuan Province, China.
| |
Collapse
|
21
|
Zahalka SJ, Akturk HK, Galindo RJ, Shah VN, Low Wang CC. Continuous Glucose Monitoring for Prediabetes - Roles, Evidence, and Gaps. Endocr Pract 2025:S1530-891X(25)00893-6. [PMID: 40409607 DOI: 10.1016/j.eprac.2025.05.742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/28/2025] [Accepted: 05/13/2025] [Indexed: 05/25/2025]
Abstract
Continuous glucose monitoring (CGM) has transformed the care of patients with diabetes, and there is great potential to extend these benefits to prediabetes. The recent FDA approval of over the counter CGMs has increased interest for use in individuals with prediabetes. It is of particular interest to use CGM to guide early individualized lifestyle interventions to prevent the progression of prediabetes to diabetes and support reversion to normoglycemia. In this review, we discuss published evidence regarding CGM metrics in normoglycemia, briefly review the use of CGM to diagnose prediabetes, and review available evidence for CGM use during lifestyle interventions in individuals with prediabetes. Future studies are needed to validate CGM metrics for prediabetes and evaluate effects of early intervention with CGM in this population.
Collapse
Affiliation(s)
- Salwa J Zahalka
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Mail Stop 8106, Aurora, CO 80045.
| | - Halis K Akturk
- Barbara Davis Center for Diabetes, University of Colorado, 1775 Aurora Ct #A140, Aurora, CO 80045.
| | - Rodolfo J Galindo
- University of Miami Miller School of Medicine, Division of Endocrinology, 1450 Northwest 10th Avenue, Miami, FL 33136.
| | - Viral N Shah
- Division of Endocrinology & Metabolism, Indiana University School of Medicine, 1120 W. Michigan Street CL380, Room 380F, Indianapolis, IN 46202-5209.
| | - Cecilia C Low Wang
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus School of Medicine, 12801 East 17(th) Avenue, MS8106, Aurora, CO 80045.
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
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
|
25
|
Koh JYJ, Tan CYH, Li M, Liu MH, Chew HSJ. The Effectiveness of Time-Restricted Eating as an Intermittent Fasting Approach on Shift Workers' Glucose Metabolism: A Systematic Review and Meta-Analysis. Nutrients 2025; 17:1689. [PMID: 40431429 PMCID: PMC12114545 DOI: 10.3390/nu17101689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Revised: 05/07/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Shift workers face higher risks of impaired glucose metabolism due to irregular eating habits and circadian misalignment. Time-restricted eating (TRE) could improve glucose metabolism by aligning food intake with the circadian clock, but its effectiveness remains unclear. Methods: Ten electronic databases (PubMed, EMBASE, Cochrane Library, CINAHL, PsycINFO, Scopus, Web of Science, ProQuest Dissertations and Theses, Science.gov, and ClinicalTrials.gov) were searched from journal inception to September 2024. Only randomized controlled trials (RCTs) involving shift workers were included. Meta-analyses with sensitivity analyses were conducted using a random-effects model to pool glucose metabolism and sleep outcomes, with heterogeneity and quality assessments performed. Results: Six RCTs were included. TRE demonstrated positive but non-significant effects on glucose metabolism outcomes: fasting blood glucose (weighted mean difference [WMD]: -0.02 mmol/L, 95% confidence interval [CI]: -0.13 to 0.10, I2 = 0%), fasting blood insulin (WMD: -5.77 pmol/L, 95% CI: -85.62 to 74.08, I2 = 92%), HOMA-IR (WMD: -0.50, 95% CI: -2.76 to 1.76, I2 = 82%), 2 h postprandial glucose (WMD: -0.65 mmol/L, 95% CI: -3.18 to 1.89, I2 = 86%), total sleep time (g = 0.07, 95% CI: -0.23 to 0.37, I2 = 0%), and sleep efficiency (g = -0.05, 95% CI: -0.63 to 0.53, I2 = 62%). Sensitivity analyses yielded similar findings, and overall certainty of evidence was rated 'very low'. Conclusions: While TRE shows potential for improving the glucose metabolism in shift workers, current evidence remains inconclusive due to small sample sizes and study limitations. Future research should prioritize well-powered TRE RCTs in shift workers that adhere to a 6-10 h eating window. Incorporating early-TRE schedules with sleep hygiene may optimize metabolic outcomes, with circadian biomarkers analyzed to better elucidate the mechanistic pathway implicated.
Collapse
Affiliation(s)
- Jia Ying Jennell Koh
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117543, Singapore; (J.Y.J.K.); (C.Y.H.T.)
| | - Celine Yu Han Tan
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117543, Singapore; (J.Y.J.K.); (C.Y.H.T.)
| | - Meng Li
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China;
| | - Mei Hui Liu
- Department of Food Science and Technology, Faculty of Science, National University of Singapore, Singapore 117543, Singapore;
| | - Han Shi Jocelyn Chew
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117543, Singapore; (J.Y.J.K.); (C.Y.H.T.)
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117543, Singapore
| |
Collapse
|
26
|
Carbone F, Després JP, Ioannidis JPA, Neeland IJ, Garruti G, Busetto L, Liberale L, Ministrini S, Vilahur G, Schindler TH, Macedo MP, Di Ciaula A, Krawczyk M, Geier A, Baffy G, Faienza MF, Farella I, Santoro N, Frühbeck G, Yárnoz-Esquiroz P, Gómez-Ambrosi J, Chávez-Manzanera E, Vázquez-Velázquez V, Oppert JM, Kiortsis DN, Sbraccia P, Zoccali C, Portincasa P, Montecucco F. Bridging the gap in obesity research: A consensus statement from the European Society for Clinical Investigation. Eur J Clin Invest 2025:e70059. [PMID: 40371883 DOI: 10.1111/eci.70059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/12/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Most forms of obesity are associated with chronic diseases that remain a global public health challenge. AIMS Despite significant advancements in understanding its pathophysiology, effective management of obesity is hindered by the persistence of knowledge gaps in epidemiology, phenotypic heterogeneity and policy implementation. MATERIALS AND METHODS This consensus statement by the European Society for Clinical Investigation identifies eight critical areas requiring urgent attention. Key gaps include insufficient long-term data on obesity trends, the inadequacy of body mass index (BMI) as a sole diagnostic measure, and insufficient recognition of phenotypic diversity in obesity-related cardiometabolic risks. Moreover, the socio-economic drivers of obesity and its transition across phenotypes remain poorly understood. RESULTS The syndemic nature of obesity, exacerbated by globalization and environmental changes, necessitates a holistic approach integrating global frameworks and community-level interventions. This statement advocates for leveraging emerging technologies, such as artificial intelligence, to refine predictive models and address phenotypic variability. It underscores the importance of collaborative efforts among scientists, policymakers, and stakeholders to create tailored interventions and enduring policies. DISCUSSION The consensus highlights the need for harmonizing anthropometric and biochemical markers, fostering inclusive public health narratives and combating stigma associated with obesity. By addressing these gaps, this initiative aims to advance research, improve prevention strategies and optimize care delivery for people living with obesity. CONCLUSION This collaborative effort marks a decisive step towards mitigating the obesity epidemic and its profound impact on global health systems. Ultimately, obesity should be considered as being largely the consequence of a socio-economic model not compatible with optimal human health.
Collapse
Affiliation(s)
- Federico Carbone
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Genoa, Italy
| | - Jean-Pierre Després
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Québec, Québec, Canada
- VITAM - Centre de Recherche en santé Durable, Centre intégré Universitaire de santé et de Services Sociaux de la Capitale-Nationale, Québec, Québec, Canada
| | - John P A Ioannidis
- Department of Medicine, Stanford Cardiovascular Institute, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, USA
- Department of Epidemiology and Population Health, Stanford Cardiovascular Institute, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, USA
- Department of Biomedical Science, Stanford Cardiovascular Institute, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, USA
| | - Ian J Neeland
- Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Cardiovascular Disease, Harrington Heart and Vascular Institute, Cleveland, Ohio, USA
| | - Gabriella Garruti
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Luca Busetto
- Department of Medicine, University of Padua, Padua, Italy
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Genoa, Italy
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Cardiology Department, Luzerner Kantonspital, Lucerne, Switzerland
| | - Gemma Vilahur
- Research Institute, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, IIB-Sant Pau, Barcelona, Spain
- CiberCV, Institute Carlos III, Madrid, Spain
| | - Thomas H Schindler
- Washington University in St. Louis, Mallinckrodt Institute of Radiology, Division of Nuclear Medicine, Cardiovascular Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Maria Paula Macedo
- APDP - Diabetes Portugal, Education and Research Center, Lisbon, Portugal
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS | FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Agostino Di Ciaula
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Marcin Krawczyk
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Essen, Germany
- Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver Surgery, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Andreas Geier
- Interdisciplinary Amyloidosis Center of Northern Bavaria, University Hospital of Würzburg, Würzburg, Germany
- Department of Internal Medicine II, Hepatology, University Hospital of Würzburg, Würzburg, Germany
| | - Gyorgy Baffy
- Department of Medicine, VA Boston Healthcare System, Harvard Medical School, Boston, Massachusetts, USA
| | - Maria Felicia Faienza
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Ilaria Farella
- Department of Medicine and Surgery, LUM University, Casamassima, Italy
| | - Nicola Santoro
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Medicine and Health Sciences, "V. Tiberio" University of Molise, Campobasso, Italy
| | - Gema Frühbeck
- Department of Endocrinology and Nutrition, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBERObn (CIBER Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Patricia Yárnoz-Esquiroz
- Department of Endocrinology and Nutrition, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBERObn (CIBER Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Gómez-Ambrosi
- Department of Endocrinology and Nutrition, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBERObn (CIBER Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Emma Chávez-Manzanera
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Jean-Michel Oppert
- Department of Nutrition, Pitié-Salpêtrière Hospital (AP-HP), Human Nutrition Research Center Ile-de-France (CRNH IdF), Sorbonne University, Paris, France
| | - Dimitrios N Kiortsis
- Atherothrombosis Research Centre, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Paolo Sbraccia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carmine Zoccali
- Renal Research Institute, New York, New York, USA
- Institute of Molecular Biology and Genetics (Biogem), Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia Trapianto Renale (IPNET), c/o Nefrologia, Grande Ospedale Metropolitano, Reggio Calabria, Italy
| | - Piero Portincasa
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Genoa, Italy
| |
Collapse
|
27
|
Koval A, Mamadalieva NZ, Mamadalieva R, Jalilov F, Katanaev VL. Success and Controversy of Natural Products as Therapeutic Modulators of Wnt Signaling and Its Interplay with Oxidative Stress: Comprehensive Review Across Compound Classes and Experimental Systems. Antioxidants (Basel) 2025; 14:591. [PMID: 40427472 PMCID: PMC12108534 DOI: 10.3390/antiox14050591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2025] [Revised: 05/05/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
The highly conserved Wnt signaling pathway, a complex network critical for embryonic development and adult tissue homeostasis, regulates diverse cellular processes, ultimately influencing tissue organization and organogenesis; its dysregulation is implicated in numerous diseases, and it is known to be affected by oxidative pathways. This report reviews the recent literature on major classes of natural products with pronounced anti-oxidant properties, such as cardiac glycosides, steroid saponins, ecdysteroids, withanolides, cucurbitacins, triterpenes, flavonoids, and iridoids, that modulate its activity in various pathological conditions, summarizing and critically analyzing their effects on the Wnt pathway in various therapeutically relevant experimental models and highlighting the role of ROS-mediated crosstalk with Wnt signaling in these studies. Models reviewed include not only cancer but also stroke, ischemia, bone or kidney diseases, and regenerative medicine, such as re-epithelialization, cardiac maintenance, and hair loss. It highlights the paramount importance of modulating this signaling by natural products to define future research directions. We also discuss controversies identified in the mode of action of several compounds in different models and directions on how to further improve the quality and depth of such studies.
Collapse
Affiliation(s)
- Alexey Koval
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland;
| | - Nilufar Z. Mamadalieva
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland;
- Faculty of Medicine, Alfraganus University, Yuqori Qoraqamish Str. 2a, Tashkent 100190, Uzbekistan;
- Institute of the Chemistry of Plant Substances, Uzbekistan Academy of Sciences, Mirzo Ulugbek Str. 77, Tashkent 100170, Uzbekistan
| | - Rano Mamadalieva
- Faculty of Medicine, Namangan University of Business and Science, Beshkapa Str. 111, Namangan 160100, Uzbekistan;
| | - Fazliddin Jalilov
- Faculty of Medicine, Alfraganus University, Yuqori Qoraqamish Str. 2a, Tashkent 100190, Uzbekistan;
| | - Vladimir L. Katanaev
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland;
- Translational Oncology Research Center, Qatar Biomedical Research Institute (QBRI), College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha P.O. Box 34110, Qatar
| |
Collapse
|
28
|
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
|
29
|
Machado-Junior PB, Lass A, de Bortolo J, Anizelli LB, Rocha MT, Proença HM, Carvalhal SR, Mehanna SH, Nagashima S, Fernandes LC, de Noronha L, de Moraes TP, Pinho RA. Sodium-Glucose Cotransporter-2 Inhibitor Improves Renal Injury by Regulating the Redox Profile, Inflammatory Parameters, and Pyroptosis in an Experimental Model of Diabetic Kidney Disease. ACS Pharmacol Transl Sci 2025; 8:1270-1281. [PMID: 40370980 PMCID: PMC12070321 DOI: 10.1021/acsptsci.4c00552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 03/30/2025] [Accepted: 04/08/2025] [Indexed: 05/16/2025]
Abstract
Inflammatory response, oxidative stress, and pyroptosis play important roles in the pathogenesis of diabetic kidney disease (DKD), and the NOD-like receptor protein 3 (NLRP3) inflammasome complex and pyroptosis are possible cellular regulators dependent on these processes. Treatment of DKD relies on sodium-glucose cotransporter-2 inhibitors (SGLT2is); however, its effects on oxidative stress and the NLRP3 complex have not yet been fully elucidated. This study aimed to evaluate the role of a SGLT2i in the regulation of the redox system, inflammatory profile, and NLRP3 inflammasome in an experimental model of DKD. Briefly, C57BL/6 mice were subjected to a DKD model induced by the combination of a high-caloric diet and streptozotocin (40 mg/kg). The animals were exposed to empagliflozin 35 mg/kg, and clinical (plasma glucose, water and caloric intake, and weight gain) and functional (glycosuria and albuminuria) parameters were subsequently evaluated. After 25 weeks, the animals were euthanized for evaluation of histological parameters, redox activity, NLRP3 complex activity, and pyroptosis. Our results showed that DKD model animals had clinical features of DKD, namely, high body mass index, glucose levels, albuminuria, and glomerular area. Empagliflozin reduced glycemia levels, glomerular area, H2O2 levels, IL-1β, IL-1α, and TNF-α levels, lipid peroxidation, and protein carbonylation. It also improved urinary albumin excretion and decreased gasdermin D levels. No changes were observed in the NLRP3 complex proteins. In conclusion, the SGLT2i empagliflozin improved glycemic control and reduced glomerular damage through control of the redox profile and inflammatory parameters, indicating its potential as a treatment for DKD.
Collapse
Affiliation(s)
- Paulo
Andre Bispo Machado-Junior
- Laboratory
of Exercise Biochemistry in Health, Graduate Program in Health Sciences, Pontifical Catholic University of Paraná (PUCPR), 1555 Imaculada Conceição
Street, Curitiba, Parana 80215-901, Brazil
- Graduate
Program in Health Sciences, Pontifical Catholic
University of Paraná (PUCPR), 1555 Imaculada Conceição Street. Curitiba, Parana 80215-901, Brazil
| | - Andre Lass
- Laboratory
of Exercise Biochemistry in Health, Graduate Program in Health Sciences, Pontifical Catholic University of Paraná (PUCPR), 1555 Imaculada Conceição
Street, Curitiba, Parana 80215-901, Brazil
- Graduate
Program in Health Sciences, Pontifical Catholic
University of Paraná (PUCPR), 1555 Imaculada Conceição Street. Curitiba, Parana 80215-901, Brazil
| | - Julia de Bortolo
- Laboratory
of Exercise Biochemistry in Health, Graduate Program in Health Sciences, Pontifical Catholic University of Paraná (PUCPR), 1555 Imaculada Conceição
Street, Curitiba, Parana 80215-901, Brazil
| | - Leticia Bressan Anizelli
- Laboratory
of Exercise Biochemistry in Health, Graduate Program in Health Sciences, Pontifical Catholic University of Paraná (PUCPR), 1555 Imaculada Conceição
Street, Curitiba, Parana 80215-901, Brazil
| | - Mateus T. Rocha
- Laboratory
of Exercise Biochemistry in Health, Graduate Program in Health Sciences, Pontifical Catholic University of Paraná (PUCPR), 1555 Imaculada Conceição
Street, Curitiba, Parana 80215-901, Brazil
| | - Henrique Machado
Sousa Proença
- Nephrology
Division, Universidade Federal de São
Paulo (UNIFESP), 1500 Sena Madureira, São Paulo, São Paulo 04021-001, Brazil
| | | | - Samya Hamad Mehanna
- Graduate
Program in Health Sciences, Pontifical Catholic
University of Paraná (PUCPR), 1555 Imaculada Conceição Street. Curitiba, Parana 80215-901, Brazil
| | - Seigo Nagashima
- Laboratory
of Experimental Pathology, Graduate Program of Health Sciences, Pontifical Catholic University of Paraná (PUCPR), 1555 Imaculada Conceição
Street, Curitiba, Parana 80215-901, Brazil
| | - Luiz Claudio Fernandes
- Department
of Physiology, Federal University of Parana
(UFPR), Curitiba, Parana 81531-970, Brazil
| | - Lucia de Noronha
- Graduate
Program in Health Sciences, Pontifical Catholic
University of Paraná (PUCPR), 1555 Imaculada Conceição Street. Curitiba, Parana 80215-901, Brazil
- Laboratory
of Experimental Pathology, Graduate Program of Health Sciences, Pontifical Catholic University of Paraná (PUCPR), 1555 Imaculada Conceição
Street, Curitiba, Parana 80215-901, Brazil
| | - Thyago Proença de Moraes
- Graduate
Program in Health Sciences, Pontifical Catholic
University of Paraná (PUCPR), 1555 Imaculada Conceição Street. Curitiba, Parana 80215-901, Brazil
| | - Ricardo A. Pinho
- Laboratory
of Exercise Biochemistry in Health, Graduate Program in Health Sciences, Pontifical Catholic University of Paraná (PUCPR), 1555 Imaculada Conceição
Street, Curitiba, Parana 80215-901, Brazil
- Graduate
Program in Health Sciences, Pontifical Catholic
University of Paraná (PUCPR), 1555 Imaculada Conceição Street. Curitiba, Parana 80215-901, Brazil
| |
Collapse
|
30
|
Bin H, Wen W. Metformin attenuates endoplasmic reticulum stress in diabetic kidney disease: mechanistic insights and future perspectives. Int Urol Nephrol 2025:10.1007/s11255-025-04562-7. [PMID: 40343634 DOI: 10.1007/s11255-025-04562-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
Diabetic kidney disease (DKD) is a common microvascular complication of diabetes that can lead to end-stage renal failure. Emerging evidence suggests that endoplasmic reticulum (ER) stress plays a crucial role in the pathogenesis of DKD by affecting various renal parenchymal cells, including endothelial cells, podocytes, and mesangial cells. This review comprehensively examines the relationship between ER stress and DKD, focusing on how metformin, a first-line antidiabetic medication, ameliorates ER stress-induced kidney injury. Multiple factors, including reactive oxygen species (ROS), proteinuria, and advanced glycation end products (AGEs), contribute to ER stress in DKD. Metformin's renoprotective effects are primarily mediated through activation of the AMPK signaling pathway, which modulates ER stress response, reduction of oxidative stress and its impact on ER function, and improvement of mitochondrial function. These mechanisms collectively lead to decreased proteinuria, reduced cell apoptosis, and attenuated epithelial-mesenchymal transition in diabetic kidneys. Understanding these molecular mechanisms provides new insights into the therapeutic potential of metformin in DKD treatment. However, further research is needed to elucidate the precise molecular pathways through which metformin regulates ER stress in different renal cell types under diabetic conditions.
Collapse
Affiliation(s)
- Huang Bin
- Department of Endocrinology, Division of Life Science and Medicine, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, 230000, China
| | - Wenjie Wen
- Anhui Province Engineering Research Center for Dental Materials and Application, School of Stomatology, Wannan Medical College, Wuhu, 241002, China.
| |
Collapse
|
31
|
Tian Y, Kang L, Ha NT, Deng J, Liu D. Hydrogen peroxide in midbrain sleep neurons regulates sleep homeostasis. Cell Metab 2025:S1550-4131(25)00254-2. [PMID: 40378838 DOI: 10.1016/j.cmet.2025.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 01/13/2025] [Accepted: 04/22/2025] [Indexed: 05/19/2025]
Abstract
Sleep could protect animals from oxidative damage, yet the dynamic interplay between the redox state and sleep homeostasis remains unclear. Here, we show that acute sleep deprivation (SD) in mice caused a general increase in brain oxidation, particularly in sleep-promoting regions. In vivo imaging of intracellular hydrogen peroxide (H2O2) real-time dynamics revealed that in nigra sleep neurons, the increase in cytosolic but not mitochondrial H2O2 reflects sleep debt and tracks spontaneous wakefulness by positively correlating with wake duration. By controllably manipulating intraneuronal H2O2, we discovered that H2O2 elevation is required for compensatory sleep and causally promotes sleep initiation, at least partly dependent on transient receptor potential melastatin 2 (TRPM2) channel. However, excessive H2O2 induced brain inflammation and sleep fragmentation. Together, our study demonstrates intraneuronal H2O2 as a crucial signaling molecule that translates brain redox imbalance into sleep drive and underscores the significance of oxidative eustress in sleep homeostasis.
Collapse
Affiliation(s)
- Yujing Tian
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Luwei Kang
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ngoc T Ha
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Juan Deng
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Danqian Liu
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
32
|
Losarwar S, Pancholi B, Babu R, Garabadu D. Mitochondria-dependent innate immunity: A potential therapeutic target in Flavivirus infection. Int Immunopharmacol 2025; 154:114551. [PMID: 40158432 DOI: 10.1016/j.intimp.2025.114551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/10/2025] [Accepted: 03/22/2025] [Indexed: 04/02/2025]
Abstract
Mitochondria, known as the powerhouse of cells, play a crucial role in host innate immunity during flavivirus infections such as Dengue, Zika, West Nile, and Japanese Encephalitis Virus. Mitochondrial antiviral signaling protein (MAVS) resides on the outer mitochondrial membrane which is triggered by viral RNA recognition by RIG-I-like receptors (RLRs). This activation induces IRF3 and NF-κB signaling, resulting in type I interferon (IFN) production and antiviral responses. Upon flavivirus infection, mitochondrial stress and dysfunction may lead to the release of mitochondrial DNA (mtDNA) into the cytoplasm, which serves as a damage-associated molecular pattern (DAMP). Cytosolic mtDNA is sensed by cGAS (cyclic GMP-AMP synthase), leading to the activation of the STING (Stimulator of Interferon Genes) pathway to increase IFN production and expand inflammation. Flaviviral proteins control mitochondrial morphology by controlling mitochondrial fission (MF) and fusion (MFu), disrupting mitochondrial dynamics (MD) to inhibit MAVS signaling and immune evasion. Flaviviral proteins also cause oxidative stress, resulting in the overproduction of reactive oxygen species (ROS), which triggers NLRP3 inflammasome activation and amplifies inflammation. Additionally, flaviviruses drive metabolic reprogramming by shifting host cell metabolism from oxidative phosphorylation (OxPhos) to glycolysis and fatty acid synthesis, creating a pro-replicative environment that supports viral replication and persistence. Thus, the present review explores the complex interaction between MAVS, mtDNA, and the cGAS-STING pathway, which is key to the innate immune response against flavivirus infections. Understanding these mechanisms opens new avenues in therapeutic interventions in targeting mitochondrial pathways to enhance antiviral immunity and mitigate viral infection.
Collapse
Affiliation(s)
- Saurabh Losarwar
- Department of Pharmacology, Central University of Punjab, Bhatinda 151401, India
| | | | - Raja Babu
- Department of Pharmacology, Central University of Punjab, Bhatinda 151401, India
| | - Debapriya Garabadu
- Department of Pharmacology, Central University of Punjab, Bhatinda 151401, India.
| |
Collapse
|
33
|
Sun M, Wang X, Lu Z, Yang Y, Lv S, Miao M, Chen WM, Wu SY, Zhang J. Long-term delirium and survival outcomes in patients treated with GLP-1 receptor agonists versus metformin in type 2 diabetes: A population-based cohort study. Diabetes Obes Metab 2025. [PMID: 40331425 DOI: 10.1111/dom.16434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/19/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025]
Abstract
AIM Type 2 diabetes mellitus (T2DM) is associated with an increased risk of delirium and mortality. While glucagon-like peptide-1 receptor agonists (GLP-1 RAs) provide metabolic and neuroprotective benefits, their long-term impact on delirium risk remains uncertain. This study compares GLP-1 RAs and metformin in relation to delirium and mortality in T2DM patients using real-world data. METHODS A retrospective cohort study was conducted using the TriNetX global federated research network, which primarily comprises U.S.-based healthcare organisations (approximately 85%), with additional sites in Europe, Asia-Pacific and the Middle East. Adults (≥18 years) with T2DM who initiated GLP-1 RAs or metformin were included. Propensity score matching (PSM) balances baseline characteristics. The primary outcome was incident delirium; the secondary outcome was all-cause mortality. Kaplan-Meier survival curves and time-dependent Cox models assessed associations. RESULTS After 1:1 PSM (N = 63 096 per group), GLP-1 RAs showed no overall reduction in delirium risk (AHR: 0.98, 95% CI: 0.94-1.02, p = 0.3628). However, they were protective in the first 5 years (AHR: 0.89, 95% CI: 0.86-0.92, p < 0.0001) but increased delirium risk between 5 and 10 years (AHR: 1.15, 95% CI: 1.04-1.26, p = 0.0046). Subgroup analysis revealed lower delirium risk with GLP-1 RAs in middle-aged patients (40-79 years) and those with HbA1c <7.5%. Higher risk was observed in Asian and Native Hawaiian/Pacific Islander populations. However, these findings should be interpreted with caution due to the relatively small subgroup sizes and the limited representativeness of these groups within the predominantly U.S.-based database, in which Asian and Native Hawaiian/Pacific Islander patients together accounted for less than 5% of the overall cohort. Mortality risk was lower in absolute terms for GLP-1 RAs (6.28% vs. 9.95%) but higher in long-term hazard (AHR: 1.16, 95% CI: 1.12-1.21, p < 0.001). CONCLUSIONS GLP-1 RA use was initially associated with a lower risk of delirium, but this association reversed over time. Subgroup variations suggest individualised treatment considerations. Metformin remains a preferred option given its stable cognitive and survival benefits.
Collapse
Affiliation(s)
- Mingyang Sun
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Xiaoling Wang
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Zhongyuan Lu
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Yitian Yang
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Shuang Lv
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Mengrong Miao
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Wan-Ming Chen
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, Taipei, Taiwan
- Artificial Intelligence Development Center, Fu Jen Catholic University, Taipei, Taiwan
| | - Szu-Yuan Wu
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, Taipei, Taiwan
- Artificial Intelligence Development Center, Fu Jen Catholic University, Taipei, Taiwan
- Department of Food Nutrition and Health Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
- Big Data Center, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan
- Division of Radiation Oncology, Department of Medicine, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan
- Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung, Taiwan
- Cancer Center, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan
- Centers for Regional Anesthesia and Pain Medicine, Taipei Municipal Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Jiaqiang Zhang
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| |
Collapse
|
34
|
Kupczyk D, Bilski R, Szeleszczuk Ł, Mądra-Gackowska K, Studzińska R. The Role of Diet in Modulating Inflammation and Oxidative Stress in Rheumatoid Arthritis, Ankylosing Spondylitis, and Psoriatic Arthritis. Nutrients 2025; 17:1603. [PMID: 40362911 PMCID: PMC12073256 DOI: 10.3390/nu17091603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Revised: 05/02/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
Rheumatic diseases such as rheumatoid arthritis (RA), ankylosing spondylitis (AS), and psoriatic arthritis (PsA) are chronic autoimmune disorders characterized by persistent inflammation and oxidative stress, leading to joint damage and reduced quality of life. In recent years, increasing attention has been given to diet as a modifiable environmental factor that can complement pharmacological therapy. This review summarizes current evidence on how key dietary components-such as omega-3 fatty acids, fiber, polyphenols, and antioxidant vitamins-affect inflammatory pathways and oxidative balance. Special emphasis is placed on the Mediterranean diet, low-starch diets, and hypocaloric regimens, which have shown potential in improving disease activity. The gut microbiota emerges as a critical mediator between diet and immune function, with dietary interventions capable of restoring eubiosis and strengthening the intestinal barrier. Additionally, this paper discusses challenges in the clinical implementation of diet therapy, the need for personalized nutritional strategies, and the importance of integrating diet into holistic patient care. Collectively, findings suggest that dietary interventions may reduce disease activity, mitigate systemic inflammation, and enhance patients' overall well-being.
Collapse
Affiliation(s)
- Daria Kupczyk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland
| | - Rafał Bilski
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland
| | - Łukasz Szeleszczuk
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-093 Warsaw, Poland;
| | - Katarzyna Mądra-Gackowska
- Department of Geriatrics, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 Skłodowskiej Curie Str., 85-094 Bydgoszcz, Poland;
| | - Renata Studzińska
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 2 Jurasza Str., 85-089 Bydgoszcz, Poland;
| |
Collapse
|
35
|
Abdel-Lah ES, Sherkawy HS, Mohamed WH, Fawy MA, Hasan AA, Muhammed AA, Taha AF, Tony AA, Hamad N, Gamea MG. Empagliflozin and memantine combination ameliorates cognitive impairment in scopolamine + heavy metal mixture-induced Alzheimer's disease in rats: role of AMPK/mTOR, BDNF, BACE-1, neuroinflammation, and oxidative stress. Inflammopharmacology 2025:10.1007/s10787-025-01755-5. [PMID: 40325262 DOI: 10.1007/s10787-025-01755-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Accepted: 04/02/2025] [Indexed: 05/07/2025]
Abstract
One of the major consequences of diabetes mellitus that has gained attention due to its rising incidence is cognitive impairment. Recent research suggested that sodium-glucose cotransporter-2 (SGLT-2) inhibitors can mitigate memory impairment linked to Alzheimer's disease and are now being explored for their cognitive benefits. However, their mechanisms were not thoroughly studied. This research investigates the hypothesis of the neuroprotective effect of empagliflozin administration against scopolamine-heavy metal mixture (SCO + HMM)-treated Alzheimer's rat models in comparison with memantine as a reference drug and the impact of their combination. Yet, the neuroprotective effects of memantine and empagliflozin combination against cognitive impairment have not been previously explored. This study employed adult male albino rats categorized into five groups. The impact of empagliflozin, memantine, and their concomitant administration on cognitive performance was assessed in a scopolamine and heavy metal mixture-treated Alzheimer's disease model in rats. The assessment of rats' cognitive behavior, memory, and spatial learning was conducted, followed by an evaluation of hippocampal brain-derived neurotrophic factor (BDNF), beta-secretase (BACE-1), oxidative stress (OS), and inflammatory marker activity. And, a western blot analysis was conducted to detect phosphorylated 5' AMP-activated protein kinase (p-AMPK), phosphorylated mammalian target of rapamycin (p-mTOR), and heme oxygenase-1 (HO-1). Hippocampal and cerebellar histopathology were thoroughly examined, in addition to the expressions of amyloid β (Aβ). The current data demonstrate the involvement of the pAMPK/mTOR/HO-1 signaling pathway in empagliflozin neuroprotection against SCO + HMM-induced AD. In addition, it reduces AD hallmarks (Aβ and BACE1), neuro-inflammation, and oxidative stress sequelae, and enhances neurogenesis and synaptic density via BDNF. This study proposes that EMPA, especially when co-administered with other conventional anti-Alzheimer therapy, may be formulated into an innovative therapeutic strategy for the enhancement of cognitive impairments associated with neurodegenerative disorders.
Collapse
Affiliation(s)
- Ebtsam S Abdel-Lah
- Department of Pharmacology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt.
- Department of Pharmacology, School of Veterinary Medicine, Badr University, Assiut, 11829, Egypt.
| | - Hoda S Sherkawy
- Department of Medical Biochemistry, Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Wafaa H Mohamed
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Aswan University, Aswan, Egypt
| | - Mariam A Fawy
- Department of Zoology, Faculty of Science, South Valley University, Qena, 83523, Egypt
| | - Asmaa A Hasan
- Department of Human Anatomy and Embryology, Faculty of Medicine, Aswan University, Aswan, 81528, Egypt
| | - Asmaa A Muhammed
- Department of Medical Physiology, Faculty of Medicine, Aswan University, Aswan, 81528, Egypt
| | - Amira F Taha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| | - Abeer A Tony
- Department of Neuropsychiatry, Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Nashwa Hamad
- Department of Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71515, Egypt
| | - Marwa G Gamea
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, 71526, Egypt
- Basic Medical Science Department, Badr University, Assiut, Egypt
| |
Collapse
|
36
|
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
|
37
|
Cuttone A, Cannavò V, Abdullah RMS, Fugazzotto P, Arena G, Brancati S, Muscarà A, Morace C, Quartarone C, Ruggeri D, Squadrito G, Russo GT. Expanding the Use of SGLT2 Inhibitors in T2D Patients Across Clinical Settings. Cells 2025; 14:668. [PMID: 40358192 PMCID: PMC12071329 DOI: 10.3390/cells14090668] [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: 03/26/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025] Open
Abstract
Sodium-glucose cotransporter-2 inhibitors (SGLT2i) are currently recommended in patients with type 2 diabetes (T2D) to reduce serum glucose levels. Moreover, robust evidence has clearly demonstrated their beneficial cardiovascular and renal effects, making this class of drugs pivotal for the treatment of T2D, especially when complicated by diabetic kidney disease or heart failure. However, several other comorbidities are frequently encountered in T2D patients beyond these long-term diabetes complications, especially in the internal medicine setting. For some of these comorbidities, such as MAFLD and cognitive impairment, the association with diabetes is increasingly recognized, with the hypothesis of a common pathophysiologic background, whereas, for others, a coincident epidemiology linked to the ageing of populations, including that of T2D subjects, may be advocated. In the effort of personalizing T2D treatment, evidence on the potential effects of SGLT2i in these different clinical conditions is accumulating. The purpose of this narrative review is to update current literature on the effects of SGLT2i for the treatment of T2D in different clinical settings beyond glycaemic control, and to elucidate potential molecular mechanisms by which they exert these effects.
Collapse
Affiliation(s)
- Alessandro Cuttone
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Vittorio Cannavò
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Raouf Mastan Sheik Abdullah
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Pierluigi Fugazzotto
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Giada Arena
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Simona Brancati
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Andrea Muscarà
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Carmela Morace
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Cristina Quartarone
- Internal Medicine and Diabetology Unit, University Hospital of Messina, 98124 Messina, Italy; (C.Q.); (D.R.)
| | - Domenica Ruggeri
- Internal Medicine and Diabetology Unit, University Hospital of Messina, 98124 Messina, Italy; (C.Q.); (D.R.)
| | - Giovanni Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Giuseppina Tiziana Russo
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| |
Collapse
|
38
|
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
|
39
|
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
|
40
|
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
|
41
|
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
|
42
|
Márquez‐Arrico CF, Silvestre FJ, Fernández‐Reyes M, López‐Domènech S, Hermenejildo J, Abad‐Jiménez Z, Silvestre‐Rangil J, Fernández‐Collazo P, Morillas C, Montiel‐Company JM, Víctor VM, Rocha M. Gingival inflammation and leukocyte-endothelium cell interactions in women with polycystic ovary syndrome. J Periodontol 2025; 96:478-489. [PMID: 39403884 PMCID: PMC12123394 DOI: 10.1002/jper.24-0148] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 09/12/2024] [Accepted: 09/24/2024] [Indexed: 06/02/2025]
Abstract
BACKGROUND Given the link between chronic inflammation and periodontal pathologies and increased cardiovascular risk, this study aims to investigate if gingivitis exacerbates the inflammatory response and subclinical atherosclerotic markers in women with polycystic ovary syndrome (PCOS). METHODS For this case-control study, women were assigned to three groups: two PCOS groups (with and without gingivitis) and a control group. Anthropometric and biochemical variables were determined, along with periodontal parameters (probing pocket depth [PPD], clinical attachment level [CAL], bleeding on probing [BOP], plaque index, calculus index, and tooth loss), systemic and neutrophil inflammatory markers (tumor necrosis factor alpha [TNFα], C-reactive protein [CRP], and c-Jun N-terminal kinase [JNK]), systemic oxidative stress mediators (myeloperoxidase [MPO] and glutathione), soluble cellular adhesion molecules, and neutrophil-endothelium cell interactions (rolling flux, velocity, and adhesion). RESULTS Of 104 women recruited, 68 had PCOS, 24 of whom presented gingivitis, and 36 were controls. PCOS patients presented altered sexual hormone, lipid, and carbohydrate profiles. Levels of systemic inflammatory markers, MPO, and soluble platelet selectin (sP-selectin) were higher, and glutathione levels were lower in PCOS patients. BOP, plaque, and calculus index values were higher in PCOS patients with gingivitis. Neutrophils from PCOS patients showed increased JNK and decreased adhesion under flow conditions, with reduced rolling velocity and increased rolling flux and cellular adhesion, all of which were more pronounced in those with gingivitis. BOP was independently associated with rolling velocity, rolling flux, and cellular adhesion. CONCLUSION Neutrophils of PCOS patients with gingivitis exhibit hyperactivity, promoting interaction with the endothelium and potentially contributing to atherosclerotic disease. PLAIN LANGUAGE SUMMARY Currently, there is a high prevalence of diseases that affect tooth-supporting tissues (periodontal diseases) and negatively influence the oral health and quality of life of the adult population. These pathologies lead to movement of the teeth and impairment of chewing function, eventually resulting in the loss of teeth. In recent years, the concept of periodontal medicine has arisen and consists of studying how periodontal diseases can influence our general inflammatory system and how systemic inflammatory pathologies can affect our oral health. In the present study, we evaluate a group of women with polycystic ovary syndrome (PCOS), a condition characterized by alterations of sex hormones and lipid profile and weight gain (body mass index). Our results show a high prevalence of gum inflammation among women with PCOS, which affects the interaction of their leukocytes and endothelial cells. The leukocytes of these women are hyper-responsive, presenting greater endothelial adhesion, lower flow velocity and enhanced rolling compared to those in a PCOS group without gum inflammation or controls. This study has generated a new line of research to analyze how neutrophils from patients with gingivitis exhibit hyperactivity, which promotes their interaction with the endothelium, thus contributing to the development of atherosclerotic disease.
Collapse
Affiliation(s)
| | - Francisco Javier Silvestre
- Department of StomatologyUniversity of ValenciaValenciaSpain
- Department of StomatologyUniversity Hospital Doctor PesetFoundation for the Promotion of Health and Biomedical ResearchValenciaSpain
| | - Meylin Fernández‐Reyes
- Department of Endocrinology and NutritionUniversity Hospital Doctor PesetFoundation for the Promotion of Health and Biomedical ResearchValenciaSpain
| | | | - Jonathan Hermenejildo
- Department of Endocrinology and NutritionUniversity Hospital Doctor PesetFoundation for the Promotion of Health and Biomedical ResearchValenciaSpain
| | - Zaida Abad‐Jiménez
- Department of Endocrinology and NutritionUniversity Hospital Doctor PesetFoundation for the Promotion of Health and Biomedical ResearchValenciaSpain
| | | | - Pablo Fernández‐Collazo
- Department of Endocrinology and NutritionUniversity Hospital Doctor PesetFoundation for the Promotion of Health and Biomedical ResearchValenciaSpain
| | - Carlos Morillas
- Department of Endocrinology and NutritionUniversity Hospital Doctor PesetFoundation for the Promotion of Health and Biomedical ResearchValenciaSpain
| | | | - Víctor M. Víctor
- Department of PhysiologyUniversity of ValenciaBiomedical Research Institute Valencia (INCLIVA)ValenciaSpain
- National Network of Biomedical Research on Hepatic and Digestive DiseasesCIBERehdValenciaSpain
| | - Milagros Rocha
- Department of Endocrinology and NutritionUniversity Hospital Doctor PesetFoundation for the Promotion of Health and Biomedical ResearchValenciaSpain
- National Network of Biomedical Research on Hepatic and Digestive DiseasesCIBERehdValenciaSpain
| |
Collapse
|
43
|
Lolescu BM, Furdui-Lința AV, Ilie CA, Sturza A, Zară F, Muntean DM, Blidișel A, Crețu OM. Adipose tissue as target of environmental toxicants: focus on mitochondrial dysfunction and oxidative inflammation in metabolic dysfunction-associated steatotic liver disease. Mol Cell Biochem 2025; 480:2863-2879. [PMID: 39704874 PMCID: PMC12048461 DOI: 10.1007/s11010-024-05165-z] [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: 10/12/2024] [Accepted: 11/10/2024] [Indexed: 12/21/2024]
Abstract
Obesity, diabetes, and their cardiovascular and hepatic comorbidities are alarming public health issues of the twenty-first century, which share mitochondrial dysfunction, oxidative stress, and chronic inflammation as common pathophysiological mechanisms. An increasing body of evidence links the combined exposure to multiple environmental toxicants with the occurrence and severity of metabolic diseases. Endocrine disruptors (EDs) are ubiquitous chemicals or mixtures with persistent deleterious effects on the living organisms beyond the endocrine system impairment; in particular, those known as metabolism-disrupting chemicals (MDCs), increase the risk of the metabolic pathologies in adult organism or its progeny. Being largely lipophilic, MDCs mainly target the adipose tissue and elicit mitochondrial dysfunction by interfering with mitochondrial bioenergetics, biogenesis, dynamics and/or other functions. Plastics, when broken down into micro- and nano-plastics (MNPs), have been detected in several human tissues, including the liver. The harmful interplay between inflammatory and redox processes, which mutually interact in a positive feed-back loop, hence the term oxidative inflammation ("OxInflammation"), occurs both at systemic and organ level. In both liver and adipose tissue, oxinflammation contributes to the progression of the metabolic dysfunction-associated steatotic liver disease (MASLD). Moreover, it has been reported that individuals with MASLD may be more susceptible to the harmful effects of toxicants (mainly, those related to mitochondria) and that chronic exposure to EDs/MDCs or MNPs may play a role in the development of the disease. While liver has been systematically investigated as major target organ for ambient chemicals, surprisingly, less information is available in the literature with respect to the adipose tissue. In this narrative review, we delve into the current literature on the most studied environmental toxicants (bisphenols, polychlorinated biphenyls, phthalates, tolylfluanid and tributyltin, per-fluoroalkyl and polyfluoroalkyl substances, heavy metals and MNPs), summarize their deleterious effects on adipose tissue, and address the role of dysregulated mitochondria and oxinflammation, particularly in the setting of MASLD.
Collapse
Affiliation(s)
- Bogdan M Lolescu
- Doctoral School Medicine, Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Adina V Furdui-Lința
- Doctoral School Medicine, Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department III Functional Sciences-Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Cosmin A Ilie
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department III Functional Sciences-Chair of Public Health & Sanitary Management, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Adrian Sturza
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department III Functional Sciences-Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Flavia Zară
- Department II Microscopic Morphology-Chair of Histology, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department of Pathology, Timisoara Municipal Emergency Clinical Hospital, Timișoara, Romania
| | - Danina M Muntean
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
- Department III Functional Sciences-Chair of Pathophysiology, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Timișoara, Romania
| | - Alexandru Blidișel
- Department of Surgery I-Clinic of Surgical Semiotics & Thoracic Surgery, Center for Hepato-Biliary and Pancreatic Surgery, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Eftimie Murgu Sq., No.2, 300041, Timișoara, Romania.
| | - Octavian M Crețu
- Department of Surgery I-Clinic of Surgical Semiotics & Thoracic Surgery, Center for Hepato-Biliary and Pancreatic Surgery, "Victor Babeș" University of Medicine and Pharmacy of Timișoara, Eftimie Murgu Sq., No.2, 300041, Timișoara, Romania
| |
Collapse
|
44
|
Wang C, Li H, Li F, Yang Y, Xu Z, Gao T, Li R, Zhang R, Mu Y, Guo Z, Guo Q, Liu S. The mitochondrial protectant SS31 optimized decellularized Wharton's jelly scaffold improves allogeneic chondrocyte implantation-mediated articular cartilage repair. J Orthop Translat 2025; 52:126-137. [PMID: 40291636 PMCID: PMC12032180 DOI: 10.1016/j.jot.2025.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/24/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Background The process of allogeneic chondrocyte implantation entails obtaining donor chondrocytes, culturing them in a medium enriched with growth factors, and then introducing them-either individually or in conjunction with biocompatible scaffolds-into areas of cartilage damage. While promising, this approach is hindered by mitochondrial dysfunction in the implanted chondrocytes. Methods This research introduced an innovative approach by creating a new type of scaffold derived from Decellularized Umbilical Cord Wharton's Jelly (DUCWJ) extracted from human umbilical cords. The scaffold was manufactured using procedures involving decellularization and lyophilization. The resulting scaffold demonstrated superior characteristics, including high porosity, hydrophilic properties, and excellent biocompatibility. To enhance its function, SS31 peptides, known for their mitochondrial-protective properties, were chemically bonded to the scaffold surface, creating an SS31@DUCWJ system. This system aims to protect chondrocytes and regulate the mitochondrial respiratory chain (MRC), thereby improving cartilage repair mediated by allogeneic chondrocyte implantation. Results In vitro studies have shown that SS31 effectively attenuates metabolic dysfunction, extracellular matrix degradation, oxidative stress, inflammation, and mitochondrial damage induced by serial cell passages. Complementary in vivo experiments showed that the SS31@DUCWJ scaffold promoted regeneration of healthy articular cartilage in femoral condylar defects in rabbits. Conclusions This SS31-modified porous decellularized scaffold represents an innovative biomaterial with anti-inflammatory properties and targeted mitochondrial regulation. It offers a promising new approach for treating articular cartilage injuries. The translational potential of this article Our study was the first to successfully load the mitochondrial protectant SS31 onto a DUCWJ hydrogel scaffold for localized drug delivery. This method is highly efficacious in repairing cartilage defects and offers a promising new avenue for the treatment of such conditions.
Collapse
Affiliation(s)
- Chao Wang
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Hao Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Fakai Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Yongkang Yang
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Ziheng Xu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Tianze Gao
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Runmeng Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Ruiyang Zhang
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yuhao Mu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Zheng Guo
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Quanyi Guo
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Shuyun Liu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| |
Collapse
|
45
|
Zhao X, Liu Y, Wang D, Li T, Xu Z, Li Z, Bai X, Wang Y. Role of GLP‑1 receptor agonists in sepsis and their therapeutic potential in sepsis‑induced muscle atrophy (Review). Int J Mol Med 2025; 55:74. [PMID: 40052580 PMCID: PMC11936484 DOI: 10.3892/ijmm.2025.5515] [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: 01/17/2025] [Indexed: 03/27/2025] Open
Abstract
Sepsis‑induced myopathy (SIM) is a common complication in intensive care units, which is often associated with adverse outcomes, primarily manifested as skeletal muscle weakness and atrophy. Currently, the management of SIM focuses on prevention strategies, as effective therapeutic options remain elusive. Glucagon‑like peptide‑1 (GLP‑1) receptor agonists (GLP‑1RAs) have garnered attention as hypoglycemic and weight‑loss agents, with an increasing body of research focusing on the extrapancreatic effects of GLP‑1. In preclinical settings, GLP‑1RAs exert protective effects against sepsis‑related multiple organ dysfunction through anti‑inflammatory and antioxidant mechanisms. Based on the existing research, we hypothesized that GLP‑1RAs may serve potential protective roles in the repair and regeneration of skeletal muscle affected by sepsis. The present review aimed to explore the relationship between GLP‑1RAs and sepsis, as well as their impact on muscle atrophy‑related myopathy. Furthermore, the potential mechanisms and therapeutic benefits of GLP‑1RAs are discussed in the context of muscle atrophy induced by sepsis.
Collapse
Affiliation(s)
- Xuan Zhao
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yukun Liu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Dongfang Wang
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Tonghan Li
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhikai Xu
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhanfei Li
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiangjun Bai
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yuchang Wang
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
46
|
Wang Q, Wang S, Zhuang Z, Wu X, Gao H, Zhang T, Zou G, Ge X, Liu Y. Identification of potential crucial genes and mechanisms associated with metabolically unhealthy obesity based on the gene expression profile. Front Genet 2025; 16:1540721. [PMID: 40376303 PMCID: PMC12078199 DOI: 10.3389/fgene.2025.1540721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/10/2025] [Indexed: 05/18/2025] Open
Abstract
Background Obesity is an epidemic and systemic metabolic disease that seriously endangers human health. This study aimed to understand the transcriptomic characteristics of the blood of metabolically unhealthy obesity (MUO) and provide insight into the target genes of differently expressed microRNAs in the occurrence and development of MUO. Methods The GSE146869, GSE145412, GSE23561, and GSE169290 datasets were analyzed to understand the transcriptome characteristics of the blood of MUO and provide insights into the target genes of differently expressed microRNAs (DEMs) in MUO. Functional and pathway enrichment analyses and gene interaction network analyses were performed to profile the function of differentially expressed genes (DEGs). In addition, miRNet 2.0, TransmiR v2.0, RNA22, TargetScan 7.2, miRDB, and miRWalk databases were used to predict the target genes of effective microRNAs. Results A total of 189 co-DEGs were identified in at least two datasets. The 156 co-upregulated genes were enriched into 29 biological process (BP) terms and 12 KEGG pathways. Among the 29 BP terms, the immune- and metabolism-related BP terms were enriched. The 33 co-downregulated genes were enriched into two BP terms, including apoptotic process and regulation of the apoptotic process, with no KEGG pathway. The hub genes EGF, STAT3, IL1B, PF4, SELP, and ITGA2B in the gene interaction network might play important roles in abnormal BP in MUO. Among the 19 DEMs identified in the blood of the MUO group by the GSE169290 dataset, 18 microRNAs targeted 85 genes as risk factors in MUO. Conclusion A network consisting of 18 microRNAs and 85 target genes might serve as a risk factor for metabolically unhealthy obesity.
Collapse
Affiliation(s)
- Qingqing Wang
- Department of Nephrology, Xuzhou Children’s Hospital, Xuzhou, Jiangsu, China
| | - Silu Wang
- School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhanyu Zhuang
- Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education (Xuzhou Medical University), Xuzhou, Jiangsu, China
| | - Xueting Wu
- Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education (Xuzhou Medical University), Xuzhou, Jiangsu, China
| | - Hongkun Gao
- School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Tianyi Zhang
- School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education (Xuzhou Medical University), Xuzhou, Jiangsu, China
| | - Guorong Zou
- Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education (Xuzhou Medical University), Xuzhou, Jiangsu, China
| | - Xing Ge
- School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education (Xuzhou Medical University), Xuzhou, Jiangsu, China
| | - Yapeng Liu
- Yunlong District Center for Disease Control and Prevention, Xuzhou, Jiangsu, China
| |
Collapse
|
47
|
Choi RH, Karasawa T, Meza CA, Maschek JA, Manuel AM, Nikolova LS, Fisher‐Wellman KH, Cox JE, Chaix A, Funai K. Semaglutide-induced weight loss improves mitochondrial energy efficiency in skeletal muscle. Obesity (Silver Spring) 2025; 33:974-985. [PMID: 40254778 PMCID: PMC12015655 DOI: 10.1002/oby.24274] [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: 11/15/2024] [Revised: 02/01/2025] [Accepted: 02/05/2025] [Indexed: 04/22/2025]
Abstract
OBJECTIVE Glucagon-like peptide-1 receptor agonists (e.g., semaglutide) potently induce weight loss, thereby reducing obesity-related complications. However, weight regain occurs when treatment is discontinued. An increase in skeletal muscle oxidative phosphorylation (OXPHOS) efficiency upon diet-mediated weight loss has been described, which may contribute to reduced systemic energy expenditure and weight regain. We set out to determine the unknown effect of semaglutide on muscle OXPHOS efficiency. METHODS C57BL/6J mice were fed a high-fat diet for 12 weeks before receiving semaglutide or vehicle for 1 or 3 weeks. The rates of ATP production and oxygen (O2) consumption were measured via high-resolution respirometry and fluorometry to determine OXPHOS efficiency in muscle at these two time points. RESULTS Semaglutide treatment led to significant reductions in fat and lean mass. Semaglutide improved skeletal muscle OXPHOS efficiency, measured as ATP produced per O2 consumed in permeabilized muscle fibers. Mitochondrial proteomic analysis revealed changes restricted to two proteins linked to complex III assembly (LYRM7 and TTC19; p < 0.05 without multiple corrections) without substantial changes in the abundance of OXPHOS subunits. CONCLUSIONS These data indicate that weight loss with semaglutide treatment increases skeletal muscle mitochondrial efficiency. Future studies could test whether it contributes to weight regain.
Collapse
Affiliation(s)
- Ran Hee Choi
- Diabetes & Metabolism Research Center, University of UtahSalt Lake CityUtahUSA
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
| | - Takuya Karasawa
- Diabetes & Metabolism Research Center, University of UtahSalt Lake CityUtahUSA
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
- Research Institute of Sport Science, Nippon Sport Science UniversitySetagayaJapan
| | - Cesar A. Meza
- Diabetes & Metabolism Research Center, University of UtahSalt Lake CityUtahUSA
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
| | - J. Alan Maschek
- Diabetes & Metabolism Research Center, University of UtahSalt Lake CityUtahUSA
- Mass Spectrometry and Proteomics CoreUniversity of UtahSalt Lake CityUtahUSA
| | - Allison M. Manuel
- Mass Spectrometry and Proteomics CoreUniversity of UtahSalt Lake CityUtahUSA
| | - Linda S. Nikolova
- Electron Microscopy Core FacilityUniversity of UtahSalt Lake CityUtahUSA
| | - Kelsey H. Fisher‐Wellman
- Department of Cancer BiologyComprehensive Cancer Center of Wake Forest Baptist HealthWinston‐SalemNorth CarolinaUSA
| | - James E. Cox
- Diabetes & Metabolism Research Center, University of UtahSalt Lake CityUtahUSA
- Mass Spectrometry and Proteomics CoreUniversity of UtahSalt Lake CityUtahUSA
- Department of BiochemistryUniversity of UtahSalt Lake CityUtahUSA
| | - Amandine Chaix
- Diabetes & Metabolism Research Center, University of UtahSalt Lake CityUtahUSA
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
- Molecular Medicine ProgramUniversity of UtahSalt Lake CityUtahUSA
| | - Katsuhiko Funai
- Diabetes & Metabolism Research Center, University of UtahSalt Lake CityUtahUSA
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
- Department of BiochemistryUniversity of UtahSalt Lake CityUtahUSA
- Molecular Medicine ProgramUniversity of UtahSalt Lake CityUtahUSA
| |
Collapse
|
48
|
Schwegel N, Strohhofer C, Kolesnik E, Oltean S, Hüttmair A, Pipp C, Benedikt M, Verheyen N, Gollmer J, Ablasser K, Wallner M, Santner V, Tripolt N, Pferschy P, Zechner P, Alber H, Siller-Matula JM, Kopp K, Zirlik A, Aziz F, Sourij H, von Lewinski D. Impact of empagliflozin on cardiac structure and function assessed by echocardiography after myocardial infarction: a post-hoc sub-analysis of the emmy trial. Clin Res Cardiol 2025; 114:629-639. [PMID: 39297940 PMCID: PMC12058928 DOI: 10.1007/s00392-024-02523-1] [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: 01/03/2024] [Accepted: 08/14/2024] [Indexed: 09/21/2024]
Abstract
BACKGROUND Empagliflozin administered after acute myocardial infarction proofed to improve cardiometabolic parameters and biomarkers, but the impact on cardiac function is still largely unknown. The aim of this post-hoc echocardiographic sub-analysis of the EMMY trial was to provide in-depth echocardiographic analysis on the effects of empagliflozin versus placebo on standard and novel echocardiographic structural and functional parameters after acute myocardial infarction. METHODS In this post-hoc analysis of the EMMY trial a subset of 313 patients (157 empagliflozin vs. 156 placebo) was enrolled for post-processing analysis of echocardiographic structural and functional parameters. On top of two-dimensional and Doppler parameters, myocardial deformation analyses were performed to assess ventricular and atrial strain values. RESULTS Left ventricular volumes showed significant differences in favor of empagliflozin over the course of the trial (change in left ventricular end-diastolic volume median [interquartile range] 8 [-3;19]% versus 13 [0;29]%, p = 0.048; left ventricular end-systolic volume -3 [-15;12]% versus 4 [-12;18]%, p = 0.044). This effect persisted after adjusting for baseline values, age, and sex. Left ventricular systolic and diastolic function overall improved over the course of the trial and parameters for diastolic function showed a distinct trend between groups but did not meet statistical significance in this cohort. CONCLUSION In this post-hoc analysis among patients with acute myocardial infarction, treatment with empagliflozin resulted in a significant beneficial effect on left ventricular end-diastolic and end-systolic volume, without significantly improving left ventricular or right ventricular functional parameters compared to placebo after 26 weeks. CLINICALTRIALS GOV REGISTRATION NCT03087773.
Collapse
Affiliation(s)
- Nora Schwegel
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Christoph Strohhofer
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Ewald Kolesnik
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria.
| | - Sabrina Oltean
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Alexander Hüttmair
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Christian Pipp
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Martin Benedikt
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Nicolas Verheyen
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Johannes Gollmer
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Klemens Ablasser
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Markus Wallner
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Viktoria Santner
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Norbert Tripolt
- Trials Unit for Interdisciplinary Metabolic Medicine, Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Peter Pferschy
- Trials Unit for Interdisciplinary Metabolic Medicine, Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Peter Zechner
- Department of Cardiology and Intensive Care Medicine, Hospital Graz II, West Location, Graz, Austria
| | - Hannes Alber
- Department of Cardiology, Public Hospital Klagenfurt Am Woerthersee, Klagenfurt Am Woerthersee, Austria
| | | | - Kristen Kopp
- Division of Cardiology and Internal Intensive Care Medicine, Department of Internal Medicine II, Paracelsus Medical Private University of Salzburg, Salzburg, Austria
| | - Andreas Zirlik
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| | - Faisal Aziz
- Trials Unit for Interdisciplinary Metabolic Medicine, Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Harald Sourij
- Trials Unit for Interdisciplinary Metabolic Medicine, Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Dirk von Lewinski
- Division of Cardiology, University Heart Center Graz, Medical University of Graz, Graz, Austria
| |
Collapse
|
49
|
Lin YH, Lin KJ, Shao IH. Androgen Deprivation Therapy: Mitochondrial Dysfunction and Metabolic Impacts in Prostate Cancer. Prostate 2025; 85:719-720. [PMID: 40235168 DOI: 10.1002/pros.24866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 01/22/2025] [Indexed: 04/17/2025]
Affiliation(s)
- Yu-Hsiang Lin
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Taoyuan City, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kuo-Jen Lin
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Taoyuan City, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - I-Hung Shao
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Taoyuan City, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
50
|
Alami M, Zerif E, Khalil A, Hajji N, Ramassamy C, Lacombe G, Laurent B, Cohen AA, Wikowski JM, Gris D, Bunt T, van Tellingen O, Hirokawa K, Fulop T, Berrougui H. Neuroprotective effects of SGLT2 inhibitors empagliflozin and dapagliflozin on Aβ 1-42-induced neurotoxicity and neuroinflammation in cellular models of Alzheimer's disease. J Alzheimers Dis 2025; 105:464-480. [PMID: 40111935 DOI: 10.1177/13872877251329474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
BackgroundAlzheimer's disease (AD) is a chronic brain degenerative disease that leads to dementia.ObjectiveThe aim of the present study is to investigate the neuroprotective impact of sodium-glucose cotransporter-2 inhibitors (SGLT2i) (empagliflozin and dapagliflozin) on tau phosphorylation, oxidative stress, and neuroinflammation.MethodsWe used MTT (3-(4, 5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide) assay, annexin-V-FITC kit, and DCFH-DA (dichloro-dihydro-fluorescein diacetate) to respectively evaluate the effect of the SGLT2i (empagliflozin and dapagliflozin) on amyloid-β (Aβ)1-42-induced neuronal death, apoptosis, and oxidative stress. The expression of NLRP3-inflammasome, phospho-Tau181, glycogen synthase kinase-3 beta (GSK-3β), cyclin-dependent kinase 5 (CdK5), and histone deacetylase 6 (HDAC6), was quantified by flow cytometry. Drug distribution in the mice's brains was assessed by liquid chromatography-mass spectrometry (LC-MS).ResultsAβ1-42 significantly reduced cell viability and increased apoptosis, which was reversed by using gliflozins. SGLT2i significantly reduced Aβ1-42-induced reactive oxygen species generation, downregulated NLRP3-inflammasome, and diminished tau pathology. Mechanistically, the last effect involved the modulation of GSK-3β and CdK5 protein expression. However, the tested treatments did not modify the Aβ1-42-stimulating effect of HDAC6. Gliflozins are substrates of drug transporters ATP-binding cassette sub-family B member 1 and/or ATP binding cassette subfamily G member 2 (ABCB1 and ABCG2), and Elacridar significantly enhances their brain distribution.ConclusionsSGLT2i empagliflozin and dapagliflozin exhibited neuroprotective actions against human Aβ1-42-induced neurotoxicity.
Collapse
Affiliation(s)
- Mehdi Alami
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, Beni Mellal, Morocco
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Canada
| | - Echarki Zerif
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Canada
| | - Abdelouahed Khalil
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Canada
| | - Nabil Hajji
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, Seville, Spain
| | - Charles Ramassamy
- INRS-Centre Armand-Frappier Santé-biotechnologie, Laval, Montréal, Québec, Canada
| | - Guy Lacombe
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Canada
| | - Benoit Laurent
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Research Center on Aging, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | | | - Jacek M Wikowski
- Department of Embryology, Medical University of Gdańsk, Gdańsk, Poland
| | - Denis Gris
- Immunology Division, Faculty of Medicine and Health Sciences, Centre de Recherche du CHUS, University of Sherbrooke, Sherbrooke, Québec, Canada
| | - Ton Bunt
- Izumi Biosciences, Inc., Lexington, MA, USA
| | - Olaf van Tellingen
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Katsuiku Hirokawa
- Department of Pathology, Institute Health and Life Science, Tokyo Medical and Dental University, Tokyo and Nito-memory Nakanosogo Hospital, Itabashi-ku, Tokyo, Japan
| | - Tamas Fulop
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Canada
| | - Hicham Berrougui
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, Beni Mellal, Morocco
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Canada
| |
Collapse
|