1
|
Li J, Zhou X, Chen J, Zhu S, Mateus A, Eliasson P, Kingham PJ, Backman LJ. Impact of Static Myoblast Loading on Protein Secretion Linked to Tenocyte Migration. J Proteome Res 2025; 24:2529-2541. [PMID: 40202163 PMCID: PMC12053940 DOI: 10.1021/acs.jproteome.5c00068] [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: 01/22/2025] [Revised: 03/24/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025]
Abstract
Exercise has been shown to promote wound healing, including tendon repair. Myokines released from the exercised muscles are believed to play a significant role in this process. In our previous study, we used an in vitro coculture and loading model to demonstrate that 2% static loading of myoblasts increased the migration and proliferation of cocultured tenocytes─two crucial aspects of wound healing. IGF-1, released from myoblasts in response to 2% static loading, was identified as a contributor to the increased proliferation. However, the factors responsible for the enhanced migration remained unknown. In the current study, we subjected myoblasts in single culture conditions to 2, 5, and 10% static loading and performed proteomic analysis of the cell supernatants. Gene Ontology (GO) analysis revealed that 2% static loading induced the secretion of NBL1, C5, and EFEMP1, which is associated with cell migration and motility. Further investigation by adding exogenous recombinant proteins to human tenocytes showed that NBL1 increased tenocyte migration but not proliferation. This effect was not observed with treatments using C5 and EFEMP1.
Collapse
Affiliation(s)
- Junhong Li
- Department
of Medical and Translational Biology, UmeÅ
University, 90187 UmeÅ, Sweden
- Department
of Community Medicine and Rehabilitation, Physiotherapy, UmeÅ University, 90187 UmeÅ, Sweden
| | - Xin Zhou
- Department
of Medical and Translational Biology, UmeÅ
University, 90187 UmeÅ, Sweden
| | - Jialin Chen
- School
of Medicine, Southeast University, 210009 Nanjing, China
- Department
of Ophthalmology, Zhongda Hospital, Southeast
University, 210009 Nanjing, China
| | - Shaochun Zhu
- Department
of Chemistry, Umeå University, 90187 Umeå, Sweden
| | - Andre Mateus
- Department
of Chemistry, Umeå University, 90187 Umeå, Sweden
- The Laboratory
for Molecular Infection Medicine Sweden, Umeå University, 90187 Umeå, Sweden
| | - Pernilla Eliasson
- Department
of Orthopedics, Sahlgrenska University Hospital, 43180 Gothenburg, Sweden
| | - Paul J. Kingham
- Department
of Medical and Translational Biology, UmeÅ
University, 90187 UmeÅ, Sweden
| | - Ludvig J. Backman
- Department
of Medical and Translational Biology, UmeÅ
University, 90187 UmeÅ, Sweden
- Department
of Community Medicine and Rehabilitation, Physiotherapy, UmeÅ University, 90187 UmeÅ, Sweden
| |
Collapse
|
2
|
Shirai T, Uemichi K, Iwai R, Shinkai H, Iwata T, Tanimura R, Sugiyama S, Takemasa T. Systemic effect of combined functional overload and endurance-type swimming exercise on whole body metabolism in mice. Am J Physiol Endocrinol Metab 2025; 328:E695-E710. [PMID: 40248969 DOI: 10.1152/ajpendo.00433.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/06/2025] [Accepted: 03/26/2025] [Indexed: 04/19/2025]
Abstract
In this study, we examined the effects of concurrent functional overload and endurance exercise on muscle hypertrophy, mitochondrial function, and systemic adaptations in male mice. The mice were assigned to three groups: Sham (Sham), overload-induced hypertrophy (OL), and overload with concurrent 60-min free swimming (5 times/wk) (OL + Swim), for 4 wk. Although OL promoted muscle hypertrophy and protein synthesis through the Akt/mammalian/mechanistic target of rapamycin (mTOR) signaling pathway, the addition of swimming (OL + Swim) attenuated these effects, resulting in less pronounced muscle growth and a smaller increase in myofiber cross-sectional area. Notably, the OL + Swim group exhibited enhanced mitochondrial activity and glycogen content compared with the OL group. Both the OL and OL + Swim groups showed elevated rates of protein synthesis, with a significant upregulation of AMP-activated kinase (AMPK) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) in the OL + Swim group, suggesting enhanced mitochondrial biogenesis and adaptation. Concurrent training also resulted in systemic benefits, including reduced inguinal and epididymal white adipocyte size, improved mitochondrial enzyme activities in adipose and liver tissues, and higher levels of fibronectin type III domain containing protein 5 (FNDC5), fibroblast growth factor 21 (FGF21), and brain-derived neurotrophic factor (BDNF) in serum, which contributed to enhanced muscle protein synthesis in cultured muscle cells. These results highlight the trade-offs between muscle hypertrophy and metabolic health in mice and underscore the importance of balanced training regimens to optimize overall metabolic health and muscle function. Our results provide further insight into how concurrent strength and endurance training can be optimized for health and performance benefits.NEW & NOTEWORTHY This study provides novel insights into the mechanisms underlying the interference effect that occurs in concurrent training, highlighting the potential systemic benefits of combining resistance and endurance exercises. Despite a reduction in muscle hypertrophy, concurrent training enhances metabolic adaptations and systemic health markers and offers a comprehensive approach to improving both muscle and metabolic fitness.
Collapse
Affiliation(s)
- Takanaga Shirai
- Department of Human Sciences, Kanagawa University, Kanagawa, Japan
- Research Fellow of Japan Society for Promotion Science, Tokyo, Japan
- Institute of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
| | - Kazuki Uemichi
- Research Fellow of Japan Society for Promotion Science, Tokyo, Japan
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Ryoto Iwai
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Hayato Shinkai
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Tomohiro Iwata
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Riku Tanimura
- Research Fellow of Japan Society for Promotion Science, Tokyo, Japan
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Shunsuke Sugiyama
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Tohru Takemasa
- Institute of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
3
|
Peng F, Chen X, Wu L, He J, Li Z, Hong Q, Zhao Q, Qian M, Wang X, Shen W, Qi T, Huang Y, Cai G, Zhang C, Chen X. Nitric oxide-primed engineered extracellular vesicles restore bioenergetics in acute kidney injury via mitochondrial transfer. Theranostics 2025; 15:5499-5517. [PMID: 40303326 PMCID: PMC12036870 DOI: 10.7150/thno.113741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/18/2025] [Indexed: 05/02/2025] Open
Abstract
Background: The disruption of mitochondrial homeostasis in acute kidney injury (AKI) is an important factor that drives persistent renal dysfunction. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have shown great therapeutic potential in AKI, but insufficient specificity of targeting the impaired mitochondrial function. Herein, we developed an engineered nitric oxide (NO)-primed MSC-EVs (pEVs) to restore mitochondrial homeostasis for AKI therapy. Methods: A cisplatin-induced AKI model was established to investigate the therapeutic effects of MSC-EVs. Proteomic and Western blot analyses compared mitochondrial cargos and functional assays included mitochondrial complex I activity and Adenosine triphosphate (ATP) quantification. Mitochondrial transfer was tracked using flow cytometry and confocal imaging. Mitochondrial dynamics, oxidative stress, and apoptosis were evaluated through ATP measurement, western blotting and rotenone-mediated respiratory chain inhibition. Results: Our data indicated that pEVs outperformed cEVs in restoring renal function and histopathology. Additionally, a reduction in mitochondria-associated oxidative stress and cell death was observed. Proteomic profiling revealed that NO priming enriched pEVs with mitochondrial complex I components, which directly enhanced their bioenergetic capacity, as evidenced by higher mitochondrial complex I activity and elevated ATP production compared to cEVs. In vivo tracking confirmed targeted delivery of pEV-derived mitochondrial contents to renal tubular cells, reducing mitochondrial reactive oxygen species (ROS) and restoring mitochondrial mass. Crucially, mitochondria-depleted pEVs abolished these therapeutic effects, establishing mitochondrial cargos as the primary therapeutic driver. Furthermore, pEVs activated a pro-survival cascade in recipient cells, showing superior efficacy in promoting mitochondrial biogenesis, dynamics, and mitophagy, thereby restoring renal mitochondrial homeostasis. Conclusion: Our study elucidated a mitochondria-targeted therapeutic strategy enabled by engineered EVs that deliver functional cargo to restore mitochondrial homeostasis. These advances provide transformative potential for AKI and other mitochondrial disorders.
Collapse
Affiliation(s)
- Fei Peng
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Xiaoniao Chen
- Department of Ophthalmology, Third Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Lingling Wu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Jiayi He
- Clinical Research Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Zongjin Li
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Quan Hong
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Qiang Zhao
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Meng Qian
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xu Wang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Wanjun Shen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Tingting Qi
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Yiyu Huang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Guangyan Cai
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Chuyue Zhang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
- Department of Nephrology and Institute of Kidney Diseases, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiangmei Chen
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| |
Collapse
|
4
|
Liu Y, Zhou R, Guo Y, Hu B, Xie L, An Y, Wen J, Liu Z, Zhou M, Kuang W, Xiao Y, Wang M, Xie G, Zhou H, Lu R, Peng H, Huang Y. Muscle-derived small extracellular vesicles induce liver fibrosis during overtraining. Cell Metab 2025; 37:824-841.e8. [PMID: 39879982 DOI: 10.1016/j.cmet.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/24/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025]
Abstract
The benefits of exercise for metabolic health occur in a dose-dependent manner. However, the adverse effects of overtraining and their underlying mechanisms remain unclear. Here, we show that overtraining induces hepatic fibrosis. Mechanistically, we find that excessive lactate accumulation in skeletal muscle leads to the lactylation of SH3 domain-containing 3 (SORBS3), triggering its liquid-liquid phase separation (LLPS). LLPS of SORBS3 enhances its interaction with flotillin 1 and selectively facilitates the sorting of F-box protein 2 (FBXO2) into small extracellular vesicles, referred to as "lactate bodies." Lactate bodies induce hepatocyte apoptosis followed by hepatic stellate cell activation via myeloid cell leukemia sequence 1 (MCL1)-BAX/BAK signaling. Inhibition of SORBS3 lactylation or FBXO2 disrupts lactate bodies formation and alleviates overtraining-triggered liver fibrosis. Likewise, reduction of muscle lactate bodies formation by salidroside attenuates overtraining-induced liver fibrosis. Collectively, we identify a process by which overtraining induces hepatic fibrosis, highlighting a potential therapeutic target for liver health.
Collapse
Affiliation(s)
- Ya Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Rui Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Yifan Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Biao Hu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Lingqi Xie
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Yuze An
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Jie Wen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Zheyu Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Min Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Weihong Kuang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Yao Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Min Wang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Genqing Xie
- Department of Endocrinology, The First People's Hospital of Xiangtan City, 411100 Xiangtan, Hunan, China
| | - Haiyan Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Renbin Lu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China.
| | - Hui Peng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China.
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 410008 Changsha, Hunan, China; FuRong Laboratory, 410078 Changsha, Hunan, China.
| |
Collapse
|
5
|
Wang X, Cao J, Wang Y, Li M, Hou S, Zhao Z, Yang M, Ju P, Jiang Y, Xiao J, Tang R, Liu H, Liu B, Zhang X, Wang B. Association of Muscle Radiodensity and Muscle Mass With Thoracic Aortic Calcification Progression in Dialysis Patients. J Cachexia Sarcopenia Muscle 2025; 16:e13813. [PMID: 40241470 PMCID: PMC12003956 DOI: 10.1002/jcsm.13813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Recent findings have spotlighted sarcopenia as a critical factor exacerbating cardiovascular risk in dialysis patients. However, no studies have investigated the relationship of muscle characteristics with thoracic aortic calcification (TAC). We explored whether skeletal muscle radiodensity (SMD) and skeletal muscle index (SMI) are associated with TAC in dialysis patients. METHODS In this study, 2517 dialysis patients (between January 2020 and June 2023) from four centres with chest computed tomography (CT) scans were analysed cross-sectionally. A cohort of 544 initial-dialysis patients (between January 2014 and December 2020) was followed for TAC progression. Chest CT images were used to assess SMD and SMI at the L1 level, as well as to measure the scores of TAC, including ascending TAC (ATAC), aortic arch calcification (AoAC) and descending TAC (DTAC). Multivariable linear regression models were employed to assess the effects of SMD and SMI on TAC and its progression. Restricted cubic spline was used to assess the potential non-linear relationships of SMD and SMI with TAC progression. RESULTS The mean (SD) age for the cross-sectional study was 54.8 (14.0) years, with males accounting for 58.2%. Over a mean (SD) follow-up duration of 3.45 (1.82) years, 85.7% showed TAC progression. Comparing the highest quartile of SMD to the lowest quartile, a significant inverse association was observed with TAC (β, -1.08 [-1.42 to -0.75]; p < 0.001); similar trends were noted for SMI (β, -0.42 [-0.74 to -0.10]; p = 0.011). SMD and SMI as continuous variables were also both significantly negatively correlated with TAC. In the longitudinal study, multivariable linear regression models revealed that an increase of 1 SD in SMD resulted in a decrease of 0.10 SD (95% CI, -0.17 to -0.02; p = 0.011) in TAC progression, and an increase of 1 SD in SMI resulted in a decrease of 0.12 SD (95% CI, -0.20 to -0.04; p = 0.003) in TAC progression. Restricted cubic spline models excluded non-linear trends for the relationships of SMD and SMI with TAC progression. The associations of SMD and SMI with DTAC were consistent with those observed for TAC, but neither showed a significant association with ATAC. CONCLUSIONS Higher SMD and higher SMI were significantly associated with lower TAC and its progression in dialysis patients. Improving SMD and SMI could be a new approach for reducing TAC.
Collapse
Affiliation(s)
- Xiao‐xu Wang
- Department of Nephrology, Zhongda HospitalSoutheast University School of MedicineNanjingChina
- School of MedicineSoutheast UniversityNanjingChina
| | - Jing‐yuan Cao
- Department of Nephrology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical MedicineNanjing Medical UniversityTaizhouChina
| | - Yao Wang
- Department of Nephrology, The Affiliated Hospital of Yangzhou UniversityYangzhou UniversityYangzhouChina
| | - Min Li
- Department of Nephrology, The Third Affiliated Hospital of Soochow UniversitySoochow UniversityChangzhouChina
| | - Shi‐mei Hou
- Department of Nephrology, The Third Affiliated Hospital of Soochow UniversitySoochow UniversityChangzhouChina
| | - Zhen Zhao
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda HospitalSoutheast University School of MedicineNanjingChina
| | - Min Yang
- Department of Nephrology, The Third Affiliated Hospital of Soochow UniversitySoochow UniversityChangzhouChina
| | - Ping‐ping Ju
- Department of Nephrology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical MedicineNanjing Medical UniversityTaizhouChina
| | - Yu‐jia Jiang
- Department of Nephrology, The Affiliated Hospital of Yangzhou UniversityYangzhou UniversityYangzhouChina
| | - Jing‐jie Xiao
- Covenant Health Palliative InstituteEdmontonAlbertaCanada
| | - Ri‐ring Tang
- Department of Nephrology, Zhongda HospitalSoutheast University School of MedicineNanjingChina
| | - Hong Liu
- Department of Nephrology, Zhongda HospitalSoutheast University School of MedicineNanjingChina
| | - Bi‐cheng Liu
- Department of Nephrology, Zhongda HospitalSoutheast University School of MedicineNanjingChina
| | - Xiao‐liang Zhang
- Department of Nephrology, Zhongda HospitalSoutheast University School of MedicineNanjingChina
| | - Bin Wang
- Department of Nephrology, Zhongda HospitalSoutheast University School of MedicineNanjingChina
| |
Collapse
|
6
|
Wu J, Liu Y, Zong J, Qiu M, Zhou Y, Li Y, Aili T, Zhao X, Hu B. TTK Inhibition Alleviates Postinjury Neointimal Formation and Atherosclerosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409250. [PMID: 39716891 PMCID: PMC11809377 DOI: 10.1002/advs.202409250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/19/2024] [Indexed: 12/25/2024]
Abstract
Atherosclerosis and its associated cardio-cerebrovascular complications remain the leading causes of mortality worldwide. Current lipid-lowering therapies reduce only approximately one-third of the cardiovascular risk. Furthermore, vascular restenosis and thrombotic events following surgical interventions for severe vascular stenosis significantly contribute to treatment failure. This highlights the urgent need for novel therapeutic targets to manage atherosclerosis and prevent restenosis and thrombosis after vascular injury. This study identifies TTK protein kinase (TTK) as a key regulator of vascular smooth muscle cell (VSMC) phenotypic switching in the context of postinjury neointimal formation and atherosclerosis. Mechanistically, TTK upregulation in VSMCs phosphorylates p120-catenin, leading to β-catenin nuclear accumulation and dissociation of the myocardin (MYOCD)/serum response factor (SRF) complex. Deletion of TTK specifically in VSMCs reduces postinjury neointimal formation in vascular injury models and attenuates atherosclerotic lesions in ApoE-/- mice. Notably, oral administration of the TTK inhibitor CFI-402257 mitigated neointimal formation without impairing reendothelialization and reduced atherosclerotic lesions in ApoE-/- mice without altering lipid levels. These findings suggest that targeting TTK, through inhibitors or alternative strategies, represents a promising approach to simultaneously prevent postinjury restenosis and treat atherosclerosis.
Collapse
Affiliation(s)
- Jie‐Hong Wu
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Yu‐Xiao Liu
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Jia‐Bin Zong
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Min Qiu
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Yi‐Fan Zhou
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Ya‐Nan Li
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Tuersun Aili
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Xin‐Ran Zhao
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Bo Hu
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| |
Collapse
|
7
|
Fu L, Cheng H, Xiong J, Xiao P, Shan X, Li Y, Li Y, Zhao X, Mi J. Effect of life course body composition on lipids and coronary atherosclerosis mediated by inflammatory biomarkers. Free Radic Biol Med 2025; 227:157-165. [PMID: 39638265 DOI: 10.1016/j.freeradbiomed.2024.12.011] [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: 09/16/2024] [Revised: 11/11/2024] [Accepted: 12/03/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE To investigate the mediating role of inflammatory biomarkers in the causal effect of body composition on lipids and atherosclerosis. METHODS Retrospective observational study and Mendelian randomization (MR) study were used. Observational analyses were undertaken using data from 4717 children and adolescents aged 6-18 years from Chinese who underwent dual-energy x-ray absorptiometry for body composition. MR analyses were based on summary statistics from UK Biobank, deCODE2021, GLGC, FinnGen and other large consortiums. Inflammatory biomarkers included leptin, insulin, adiponectin, osteocalcin, fibroblast growth factor 23 (FGF23) and parathyroid hormone (PTH). RESULTS In retrospective observational study, through osteocalcin, body composition had effects on total cholesterol (TC), triglyceride and low-density lipoprotein cholesterol (LDL). Conversely, fat mass vs. fat-free mass demonstrated opposing effects. Insulin played a role in the association of fat mass with TC and LDL (all P < 0.05). Mediation MR results indicated the causal effect of fat-free mass on coronary atherosclerosis via insulin (indirect effect, OR (odds ratio): 0.95 [95%CI, 0.92-0.98]) and adiponectin (OR: 0.96 [95%CI, 0.93-0.99]). Adiponectin also mediated the causal association of fat mass with coronary heart disease (OR: 1.06 [95%CI, 1.02-1.10]) and coronary atherosclerosis (OR: 1.05 [95%CI, 1.01, 1.09]). Leptin, adiponectin and insulin played roles in mediating the casual effects of body composition on triglyceride and high-density lipoprotein cholesterol. CONCLUSIONS Our findings suggest different body composition exert varying influences on lipids and atherosclerosis through distinct inflammatory biomarkers. The findings may be helpful in tailoring management of body composition based on inflammatory biomarkers with different lipid profiles.
Collapse
Affiliation(s)
- Liwan Fu
- Center for Non-Communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Hong Cheng
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Jingfan Xiong
- Child and Adolescent Chronic Disease Prevention and Control Department, Shenzhen Center for Chronic Disease Control, Shenzhen, China
| | - Pei Xiao
- Center for Non-Communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xinying Shan
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Yanyan Li
- Child and Adolescent Chronic Disease Prevention and Control Department, Shenzhen Center for Chronic Disease Control, Shenzhen, China
| | - Yan Li
- Child and Adolescent Chronic Disease Prevention and Control Department, Shenzhen Center for Chronic Disease Control, Shenzhen, China
| | - Xiaoyuan Zhao
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Jie Mi
- Center for Non-Communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China.
| |
Collapse
|
8
|
Zhang X, Huang DX, Xuan C, Li Y, Jiang Y, Wu X, Zhou W, Lei Y, Yang F, Ma H, Hou K, Han X, Li G. Aerobic exercise training attenuates ischemia-reperfusion injury in mice by decreasing the methylation level of METTL3-associated m6A RNA in cardiomyocytes. Life Sci 2025; 361:123294. [PMID: 39645164 DOI: 10.1016/j.lfs.2024.123294] [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: 09/04/2024] [Revised: 11/17/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND AND AIMS Ischemic heart disease (IHD) presents a significant global health challenge, with myocardial ischemia-reperfusion injury (MIRI) being a major pathophysiological contributor and lacking effective interventions. While aerobic exercise training (AET) enhances cardiovascular health, its protective mechanism in MIRI remains elusive. This study aims to elucidate the protective effect of AET in MIRI and its underlying mechanism. METHODS A mouse model of AET and MIRI was established to evaluate basic indices, cardiac ultrasound, and myocardial injury markers. Dot Blot, qRT-PCR, and Western blot were employed to assess m6A RNA methylation levels and related protein expression in myocardial tissue. In vitro, primary cardiomyocyte culture was utilized to mimic MIRI, evaluating cell viability, mitochondrial membrane potential, etc. Finally, myocardial tissues of MIRI mice were immunoprecipitated for m6A RNA methylation and sequenced to analyze related signaling pathways. KEY RESULTS AET significantly improved cardiac function and mitigated myocardial injury and fibrosis. Moreover, AET protected myocardium from MIRI by reducing m6A RNA methylation levels and modulating METTL3 expression. In vitro experiments demonstrated that the decrease in m6A RNA methylation levels and METTL3 expression conferred resistance to hypoxia/reoxygenation-induced injury. Furthermore, sequencing results indicated elevated myocardial tissue m6A RNA methylation levels during MIRI, activation of the Nrf2-related signaling pathway, and AET-mediated regulation of the Nrf2/HO-1 signaling pathway, thereby attenuating MIRI through modulation of METTL3-related m6A methylation. CONCLUSION AND SIGNIFICANCE AET attenuates MIRI by reducing the level of METTL3-related m6A RNA methylation in cardiomyocytes and activating the Nrf2/HO-1 antioxidant signaling pathway. This finding provides a novel insight and strategy for the prevention and treatment of IHD, holding significant clinical implications.
Collapse
Affiliation(s)
- Xinmin Zhang
- Department of Anesthesiology, First Hospital of Jilin University, Changchun 130021, China; The Public Laboratory Platform of First Hospital of Jilin University, Changchun 130021, China
| | - Dong-Xu Huang
- Department of Hand and Podiatric Surgery, Orthopedics Center, First Hospital of Jilin University, Changchun 130021, China; Jilin Province Key Laboratory on Tissue Repair, Reconstruction and Regeneration, First Hospital of Jilin University, Changchun 130021, China
| | - Chengluan Xuan
- Department of Anesthesiology, First Hospital of Jilin University, Changchun 130021, China
| | - Yanhui Li
- Department of Anesthesiology, First Hospital of Jilin University, Changchun 130021, China
| | - Yuting Jiang
- Reproductive Medicine Center, Prenatal Diagnosis Center, First Hospital of Jilin University, Changchun, 130021, China
| | - Xuehan Wu
- Department of Anesthesiology, First Hospital of Jilin University, Changchun 130021, China
| | - Wenqian Zhou
- Department of Cardiovascular Disease, First Hospital of Jilin University, Changchun 130021, China
| | - Yang Lei
- Department of Anesthesiology, First Hospital of Jilin University, Changchun 130021, China
| | - Fan Yang
- Department of Anesthesiology, First Hospital of Jilin University, Changchun 130021, China
| | - Haichun Ma
- Department of Anesthesiology, First Hospital of Jilin University, Changchun 130021, China
| | - Kun Hou
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Xue Han
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Guichen Li
- Department of Neurology, First Hospital of Jilin University, 1 Xinmin Avenue, 130021, Changchun, China.
| |
Collapse
|
9
|
György B, Szatmári R, Ditrói T, Torma F, Pálóczi K, Balbisi M, Visnovitz T, Koltai E, Nagy P, Buzás EI, Horvath S, Radák Z. The protein cargo of extracellular vesicles correlates with the epigenetic aging clock of exercise sensitive DNAmFitAge. Biogerontology 2025; 26:35. [PMID: 39775340 PMCID: PMC11711255 DOI: 10.1007/s10522-024-10177-9] [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: 11/10/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025]
Abstract
Extracellular vesicles (EVs) are implicated in inter-organ communication, which becomes particularly relevant during aging and exercise. DNA methylation-based aging clocks reflect lifestyle and environmental factors, while regular exercise is known to induce adaptive responses, including epigenetic adaptations. Twenty individuals with High-fitness (aged 57.7 ± 9.8 years) and twenty Medium-Low-fitness (aged 57.5 ± 9.7 years) subjects provided blood samples. EVs were isolated from the samples using a size exclusion chromatography (SEC)-based method, and their protein content was analyzed by mass spectrometry (MS). Acceleration of the biological age estimator DNAmFitAge (AgeAccelFit) was associated with the protein cargo of EVs, whereas PhenoAge and GrimAge acceleration did not show a significant relationship. This finding suggests that the epigenetic aging-modulating role of exercise may involve inter-organ communication via EVs. Set Enrichment Analysis was performed to identify enriched Gene Ontology (GO) terms for sets of proteins that were either correlated with AgeAccelFit or detected exclusively in individuals with high levels of aerobic fitness. The protein cargo of EVs further suggests that inter-organ communication influences inflammation, the immune system, cellular repair, adhesion, metabolism and coagulation. Our findings help to understand the preventive role of exercise, which could be mediated in part by EVs.
Collapse
Affiliation(s)
- Bernadett György
- Research Centre for Molecular Exercise Science, Hungarian University of Sport Science, Alkotás U. 42-48, Budapest, 1123, Hungary
| | - Réka Szatmári
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Ráth György U. 7-9, Budapest, 1122, Hungary
- Laki Kálmán Doctoral School, University of Debrecen, Nagyerdei Krt. 98, Debrecen, 4032, Hungary
- Chemistry Coordinating Institute, University of Debrecen, Egyetem Tér 1, Debrecen, 4032, Hungary
| | - Tamás Ditrói
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Ráth György U. 7-9, Budapest, 1122, Hungary
| | - Ferenc Torma
- Research Centre for Molecular Exercise Science, Hungarian University of Sport Science, Alkotás U. 42-48, Budapest, 1123, Hungary
| | - Krisztina Pálóczi
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Üllői Út 26, Budapest, 1085, Hungary
| | - Mirjam Balbisi
- Institute of Organic Chemistry, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, Budapest, 1117, Hungary
| | - Tamás Visnovitz
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Üllői Út 26, Budapest, 1085, Hungary
- Department of Plant Physiology and Molecular Plant Biology, ELTE Eötvös Loránd University, Pázmány Péter Sétány 1/C, Budapest, 1117, Hungary
| | - Erika Koltai
- Research Centre for Molecular Exercise Science, Hungarian University of Sport Science, Alkotás U. 42-48, Budapest, 1123, Hungary
| | - Péter Nagy
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Ráth György U. 7-9, Budapest, 1122, Hungary
- Chemistry Coordinating Institute, University of Debrecen, Egyetem Tér 1, Debrecen, 4032, Hungary
- Department of Anatomy and Histology, HUN-REN-UVMB Laboratory of Redox Biology Research Group, University of Veterinary Medicine, István Utca 2, Budapest, 1078, Hungary
| | - Edit I Buzás
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Üllői Út 26, Budapest, 1085, Hungary
- HUN-REN-SU Translational Extracellular Vesicle Research Group, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Steve Horvath
- Department of Biostatistics, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, 90095, USA
- San Diego Institute of Science, Altos Labs, San Diego, CA, 92121, USA
| | - Zsolt Radák
- Research Centre for Molecular Exercise Science, Hungarian University of Sport Science, Alkotás U. 42-48, Budapest, 1123, Hungary.
- Faculty of Sport Sciences, Waseda University, Tokorozawa, 2-579-15, Japan.
| |
Collapse
|
10
|
Shi X, Zhang L, Wu S, Zhang C, Mamtilahun M, Li Y, Zhang Z, Zuo C, Cui F, Li W, Yang G, Tang Y. A simple polydopamine-based platform for engineering extracellular vesicles with brain-targeting peptide and imaging probes to improve stroke outcome. J Extracell Vesicles 2025; 14:e70031. [PMID: 39783851 PMCID: PMC11714163 DOI: 10.1002/jev2.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/26/2024] [Accepted: 12/16/2024] [Indexed: 01/30/2025] Open
Abstract
Extracellular vesicles (EVs) have shown great potential for treating various diseases. Translating EVs-based therapy from bench to bedside remains challenging due to inefficient delivery of EVs to the injured area and lack of techniques to visualize the entire targeting process. Here we developed a dopamine surface functionalization platform that facilitates easy and simultaneous conjugation of targeting peptide and multi-mode imaging probes to the surface of EVs. Utilizing this platform we concurrently modified M2 microglia-derived EVs (M2-EVs) with neuronal targeting peptide rabies virus glycoprotein peptide 29 (RVG29) and multi-modal imaging tracers, resulting in the targeted delivery of M2-EVs to stroke mice brain and enabled the dynamic visualization of the targeting process from whole-body to cellular levels. We determined that intra-arterial injection achieved the highest efficiency of targeted delivery of engineered EVs to the stroke mice brain, improved therapeutic efficacy by reducing neuronal apoptosis. Mechanistically, EVs miRNA array revealed that a number of anti-apoptosis related miRNAs were significantly up-regulated, including miR-221-3p and miR-423-3p, both exerted anti-apoptotic effects through p38/ERK signalling pathways in stroke. Overall, this platform provides a facile and powerful tool for multifunctional engineering of EVs for multiscale therapeutic evaluation and enhancement of EV-based therapy, with valuable prospects for clinical translation.
Collapse
Affiliation(s)
- Xiaojing Shi
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
- McGovern Institute for Brain ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Lu Zhang
- Department of Nuclear MedicineChanghai Hospital Affiliated to Naval Medical UniversityShanghaiChina
| | - Shengju Wu
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Chunfu Zhang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Muyassar Mamtilahun
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Yongfang Li
- Department of Rehabilitation MedicineRuijin Hospital, Shanghai Jiao Tong UniversityShanghaiChina
| | - Zhijun Zhang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Changjing Zuo
- Department of Nuclear MedicineChanghai Hospital Affiliated to Naval Medical UniversityShanghaiChina
| | - Fengzhen Cui
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Wanlu Li
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Guo‐Yuan Yang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Yaohui Tang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
11
|
Perera N, De Blasio MJ, Febbraio MA. Harnessing the therapeutic potential of exercise in extracellular vesicle-based therapy in metabolic disease associated cardiovascular complications. Free Radic Biol Med 2025; 226:230-236. [PMID: 39549882 DOI: 10.1016/j.freeradbiomed.2024.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 10/10/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
Cardiovascular disease (CVD) is a leading cause of mortality, affecting ∼18 million individuals each year. Obesity and type 2 diabetes mellitus in particular, both chronic metabolic disorders, are risk factors for CVD. The salutary effects of physical activity in preventing and ameliorating CVD have long been acknowledged, as it improves glucose and lipid homeostasis, alongside attenuating oxidative damage, increasing mitochondrial function, and ultimately improving cardiac function. Exercise serves as a catalyst for the secretion of extracellular vesicles (EVs), facilitating inter-tissue communication, by which tissues can deliver important signals from one tissue to another. In recent years, an increasing number of studies have focused on the cargo encapsulated within exercise-derived EVs, as well as the orchestration of inter-tissue crosstalk aimed at modulating metabolism and tissue function in CVDs. The precise mechanisms underpinning the cardioprotective properties of exercise-derived EVs, however, remains only partially elucidated. This review explores novel EV based therapeutic options in CVD and, in particular, EVs derived from models of exercise to alter metabolism and enhance cardiovascular outcomes.
Collapse
Affiliation(s)
- Nimna Perera
- Monash Institute of Pharmaceutical Sciences, Parkville, Melbourne, Australia
| | - Miles J De Blasio
- Monash Institute of Pharmaceutical Sciences, Parkville, Melbourne, Australia
| | - Mark A Febbraio
- Monash Institute of Pharmaceutical Sciences, Parkville, Melbourne, Australia.
| |
Collapse
|
12
|
Tokizane K, Imai SI. Inter-organ communication is a critical machinery to regulate metabolism and aging. Trends Endocrinol Metab 2024:S1043-2760(24)00320-5. [PMID: 39694728 DOI: 10.1016/j.tem.2024.11.013] [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: 10/18/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024]
Abstract
Inter-organ communication (IOC) is a complex mechanism involved in maintaining metabolic homeostasis and healthy aging. Dysregulation of distinct forms of IOC is linked to metabolic derangements and age-related pathologies, implicating these processes as a potential target for therapeutic intervention to promote healthy aging. In this review, we delve into IOC mediated by hormonal signaling, circulating factors, organelle signaling, and neuronal networks and examine their roles in regulating metabolism and aging. Given the role of the hypothalamus as a high-order control center for aging and longevity, we particularly emphasize the importance of its communication with peripheral organs and pave the way for a better understanding of this critical machinery in metabolism and aging.
Collapse
Affiliation(s)
- Kyohei Tokizane
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, 63110, MO, USA
| | - Shin-Ichiro Imai
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, 63110, MO, USA; Department of Medicine, Washington University School of Medicine, St. Louis, 63110, MO, USA.
| |
Collapse
|
13
|
Szilágyi A, Takács B, Szekeres R, Tarjányi V, Nagy D, Priksz D, Bombicz M, Kiss R, Szabó AM, Lehoczki A, Gesztelyi R, Juhász B, Szilvássy Z, Varga B. Effects of voluntary and forced physical exercise on the retinal health of aging Wistar rats. GeroScience 2024; 46:4707-4728. [PMID: 38795184 PMCID: PMC11336036 DOI: 10.1007/s11357-024-01208-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 05/13/2024] [Indexed: 05/27/2024] Open
Abstract
Aging is accompanied by an increased prevalence of degenerative conditions, including those affecting ocular health, which significantly impact quality of life and increase the burden on healthcare systems. Among these, retinal aging is of particular concern due to its direct link to vision impairment, a leading cause of disability in the elderly. Vision loss in the aging population is associated with heightened risks of cognitive decline, social isolation, and morbidity. This study addresses the critical gap in our understanding of modifiable lifestyle factors, such as physical exercise, that may mitigate retinal aging and its related pathologies. We investigated the effects of different exercise regimens-voluntary (recreational-type) and forced (high-intensity)-on the retinal health of aging Wistar rats (18-month-old), serving as a model for studying the translational potential of exercise interventions in humans. Male Wistar rats were divided into four groups: a young control (3-month-old) for baseline comparison, an aged sedentary control, an aged group engaging in voluntary exercise via a running wheel in their cage, and an aged group subjected to forced exercise on a treadmill for six sessions of 20 min each per week. After a 6-month experimental period, we assessed retinal function via electroretinography (ERG), measured retinal thickness histologically, and analyzed protein expression changes relevant to oxidative stress, inflammation, and anti-aging mechanisms. Our findings reveal that voluntary exercise positively impacts retinal function and morphology, reducing oxidative stress and inflammation markers while enhancing anti-aging protein expression. In contrast, forced exercise showed diminished benefits. These insights underscore the importance of exercise intensity and preference in preserving retinal health during aging. The study highlights the potential of recreational physical activity as a non-invasive strategy to counteract retinal aging, advocating for further research into exercise regimens as preventative therapies for age-related ocular degenerations.
Collapse
Affiliation(s)
- Anna Szilágyi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Barbara Takács
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Réka Szekeres
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Vera Tarjányi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Dávid Nagy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Dániel Priksz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Mariann Bombicz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Rita Kiss
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Adrienn Mónika Szabó
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Andrea Lehoczki
- Departments of Hematology and Stem Cell Transplantation, South Pest Central Hospital, National Institute of Hematology and Infectious Diseases, Saint Ladislaus Campus, Budapest, Hungary
- Department of Public Health, Semmelweis University, Budapest, Hungary
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
| | - Rudolf Gesztelyi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Béla Juhász
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Zoltán Szilvássy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Balázs Varga
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary.
| |
Collapse
|
14
|
Lee JC, Ray RM, Scott TA. Prospects and challenges of tissue-derived extracellular vesicles. Mol Ther 2024; 32:2950-2978. [PMID: 38910325 PMCID: PMC11403234 DOI: 10.1016/j.ymthe.2024.06.025] [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: 01/14/2024] [Revised: 05/30/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024] Open
Abstract
Extracellular vesicles (EVs) are considered a vital component of cell-to-cell communication and represent a new frontier in diagnostics and a means to identify pathways for therapeutic intervention. Recently, studies have revealed the importance of tissue-derived EVs (Ti-EVs), which are EVs present in the interstitial spaces between cells, as they better represent the underlying physiology of complex, multicellular tissue microenvironments in biology and disease. EVs are native, lipid bilayer membraned nano-sized particles produced by all cells that are packaged with varied functional biomolecules including proteins, lipids, and nucleic acids. They are implicated in short- and long-range cellular communication and may elicit functional responses in recipient cells. To date, studies have often utilized cultured cells or biological fluids as a source for EVs that do not capture local molecular signatures of the tissue microenvironment. Recent work utilizing Ti-EVs has elucidated novel biomarkers for disease and provided insights into disease mechanisms that may lead to the development of novel therapeutic agents. Still, there are considerable challenges facing current studies. This review explores the vast potential and unique challenges for Ti-EV research and provides considerations for future studies that seek to advance this exciting field.
Collapse
Affiliation(s)
- Justin C Lee
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Roslyn M Ray
- Gene Therapy Research, CSL Behring, Pasadena, CA 91106, USA
| | - Tristan A Scott
- Center for Gene Therapy, City of Hope, Beckman Research Institute and Hematological Malignancy and Stem Cell Transplantation Institute, Duarte, CA 91010, USA.
| |
Collapse
|
15
|
Wang Y, Ding S. Extracellular vesicles in cancer cachexia: deciphering pathogenic roles and exploring therapeutic horizons. J Transl Med 2024; 22:506. [PMID: 38802952 PMCID: PMC11129506 DOI: 10.1186/s12967-024-05266-9] [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: 03/12/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Cancer cachexia (CC) is a debilitating syndrome that affects 50-80% of cancer patients, varying in incidence by cancer type and significantly diminishing their quality of life. This multifactorial syndrome is characterized by muscle and fat loss, systemic inflammation, and metabolic imbalance. Extracellular vesicles (EVs), including exosomes and microvesicles, play a crucial role in the progression of CC. These vesicles, produced by cancer cells and others within the tumor environment, facilitate intercellular communication by transferring proteins, lipids, and nucleic acids. A comprehensive review of the literature from databases such as PubMed, Scopus, and Web of Science reveals insights into the formation, release, and uptake of EVs in CC, underscoring their potential as diagnostic and prognostic biomarkers. The review also explores therapeutic strategies targeting EVs, which include modifying their release and content, utilizing them for drug delivery, genetically altering their contents, and inhibiting key cachexia pathways. Understanding the role of EVs in CC opens new avenues for diagnostic and therapeutic approaches, potentially mitigating the syndrome's impact on patient survival and quality of life.
Collapse
Affiliation(s)
- Yifeng Wang
- Department of Thoracic Surgery, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital, Nantong, 226001, P.R. China
- School of Medicine, Nantong University, Nantong, 226001, P.R. China
| | - Shengguang Ding
- Department of Thoracic Surgery, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital, Nantong, 226001, P.R. China.
| |
Collapse
|
16
|
Pinto AC, Tavares P, Neves B, Oliveira PF, Vitorino R, Moreira-Gonçalves D, Ferreira R. Exploiting the therapeutic potential of contracting skeletal muscle-released extracellular vesicles in cancer: Current insights and future directions. J Mol Med (Berl) 2024; 102:617-628. [PMID: 38451309 PMCID: PMC11055777 DOI: 10.1007/s00109-024-02427-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/09/2024] [Accepted: 02/05/2024] [Indexed: 03/08/2024]
Abstract
The health benefits of exercise training in a cancer setting are increasingly acknowledged; however, the underlying molecular mechanisms remain poorly understood. It has been suggested that extracellular vesicles (EVs) released from contracting skeletal muscles play a key role in mediating the systemic benefits of exercise by transporting bioactive molecules, including myokines. Nevertheless, skeletal muscle-derived vesicles account for only about 5% of plasma EVs, with the immune cells making the largest contribution. Moreover, it remains unclear whether the contribution of skeletal muscle-derived EVs increases after physical exercise or how muscle contraction modulates the secretory activity of other tissues and thus influences the content and profile of circulating EVs. Furthermore, the destination of EVs after exercise is unknown, and it depends on their molecular composition, particularly adhesion proteins. The cargo of EVs is influenced by the training program, with acute training sessions having a greater impact than chronic adaptations. Indeed, there are numerous questions regarding the role of EVs in mediating the effects of exercise, the clarification of which is critical for tailoring exercise training prescriptions and designing exercise mimetics for patients unable to engage in exercise programs. This review critically analyzes the current knowledge on the effects of exercise on the content and molecular composition of circulating EVs and their impact on cancer progression.
Collapse
Affiliation(s)
- Ana Carolina Pinto
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Patrícia Tavares
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
- iBiMED, Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal
- CIAFEL, Faculty of Sports, University of Porto and Laboratory for Integrative and ITR, Translational Research in Population Health, 4200-450, Porto, Portugal
| | - Bruno Neves
- iBiMED, Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Pedro F Oliveira
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Rui Vitorino
- iBiMED, Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal.
| | - Daniel Moreira-Gonçalves
- CIAFEL, Faculty of Sports, University of Porto and Laboratory for Integrative and ITR, Translational Research in Population Health, 4200-450, Porto, Portugal
| | - Rita Ferreira
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
17
|
Du Y, Wu L, Wang L, Reiter RJ, Lip GYH, Ren J. Extracellular vesicles in cardiovascular diseases: From pathophysiology to diagnosis and therapy. Cytokine Growth Factor Rev 2023; 74:40-55. [PMID: 37798169 DOI: 10.1016/j.cytogfr.2023.09.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/07/2023]
Abstract
Extracellular vesicles (EVs), encompassing exosomes, microvesicles (MVs), and apoptotic bodies (ABs), are cell-derived heterogeneous nanoparticles with a pivotal role in intercellular communication. EVs are enclosed by a lipid-bilayer membrane to escape enzymatic degradation. EVs contain various functional molecules (e.g., nucleic acids, proteins, lipids and metabolites) which can be transferred from donor cells to recipient cells. EVs provide many advantages including accessibility, modifiability and easy storage, stability, biocompatibility, heterogeneity and they readily penetrate through biological barriers, making EVs ideal and promising candidates for diagnosis/prognosis biomarkers and therapeutic tools. Recently, EVs were implicated in both physiological and pathophysiological settings of cardiovascular system through regulation of cell-cell communication. Numerous studies have reported a role for EVs in the pathophysiological progression of cardiovascular diseases (CVDs) and have evaluated the utility of EVs for the diagnosis/prognosis and therapeutics of CVDs. In this review, we summarize the biology of EVs, evaluate the perceived biological function of EVs in different CVDs along with a consideration of recent progress for the application of EVs in diagnosis/prognosis and therapies of CVDs.
Collapse
Affiliation(s)
- Yuxin Du
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Lin Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Litao Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, TX, USA
| | - Gregory Y H Lip
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China; Department of Laboratory Medicine and Pathology, University of Washington, Seattle WA98195, USA.
| |
Collapse
|
18
|
Ungvari Z, Fazekas-Pongor V, Csiszar A, Kunutsor SK. The multifaceted benefits of walking for healthy aging: from Blue Zones to molecular mechanisms. GeroScience 2023; 45:3211-3239. [PMID: 37495893 PMCID: PMC10643563 DOI: 10.1007/s11357-023-00873-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/11/2023] [Indexed: 07/28/2023] Open
Abstract
Physical activity, including walking, has numerous health benefits in older adults, supported by a plethora of observational and interventional studies. Walking decreases the risk or severity of various health outcomes such as cardiovascular and cerebrovascular diseases, type 2 diabetes mellitus, cognitive impairment and dementia, while also improving mental well-being, sleep, and longevity. Dose-response relationships for walking duration and intensity are established for adverse cardiovascular outcomes. Walking's favorable effects on cardiovascular risk factors are attributed to its impact on circulatory, cardiopulmonary, and immune function. Meeting current physical activity guidelines by walking briskly for 30 min per day for 5 days can reduce the risk of several age-associated diseases. Additionally, low-intensity physical exercise, including walking, exerts anti-aging effects and helps prevent age-related diseases, making it a powerful tool for promoting healthy aging. This is exemplified by the lifestyles of individuals in Blue Zones, regions of the world with the highest concentration of centenarians. Walking and other low-intensity physical activities contribute significantly to the longevity of individuals in these regions, with walking being an integral part of their daily lives. Thus, incorporating walking into daily routines and encouraging walking-based physical activity interventions can be an effective strategy for promoting healthy aging and improving health outcomes in all populations. The goal of this review is to provide an overview of the vast and consistent evidence supporting the health benefits of physical activity, with a specific focus on walking, and to discuss the impact of walking on various health outcomes, including the prevention of age-related diseases. Furthermore, this review will delve into the evidence on the impact of walking and low-intensity physical activity on specific molecular and cellular mechanisms of aging, providing insights into the underlying biological mechanisms through which walking exerts its beneficial anti-aging effects.
Collapse
Affiliation(s)
- Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary.
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | | | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Setor K Kunutsor
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Gwendolen Road, Leicester, LE5 4WP, UK.
| |
Collapse
|
19
|
Salvioli S, Basile MS, Bencivenga L, Carrino S, Conte M, Damanti S, De Lorenzo R, Fiorenzato E, Gialluisi A, Ingannato A, Antonini A, Baldini N, Capri M, Cenci S, Iacoviello L, Nacmias B, Olivieri F, Rengo G, Querini PR, Lattanzio F. Biomarkers of aging in frailty and age-associated disorders: State of the art and future perspective. Ageing Res Rev 2023; 91:102044. [PMID: 37647997 DOI: 10.1016/j.arr.2023.102044] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/01/2023]
Abstract
According to the Geroscience concept that organismal aging and age-associated diseases share the same basic molecular mechanisms, the identification of biomarkers of age that can efficiently classify people as biologically older (or younger) than their chronological (i.e. calendar) age is becoming of paramount importance. These people will be in fact at higher (or lower) risk for many different age-associated diseases, including cardiovascular diseases, neurodegeneration, cancer, etc. In turn, patients suffering from these diseases are biologically older than healthy age-matched individuals. Many biomarkers that correlate with age have been described so far. The aim of the present review is to discuss the usefulness of some of these biomarkers (especially soluble, circulating ones) in order to identify frail patients, possibly before the appearance of clinical symptoms, as well as patients at risk for age-associated diseases. An overview of selected biomarkers will be discussed in this regard, in particular we will focus on biomarkers related to metabolic stress response, inflammation, and cell death (in particular in neurodegeneration), all phenomena connected to inflammaging (chronic, low-grade, age-associated inflammation). In the second part of the review, next-generation markers such as extracellular vesicles and their cargos, epigenetic markers and gut microbiota composition, will be discussed. Since recent progresses in omics techniques have allowed an exponential increase in the production of laboratory data also in the field of biomarkers of age, making it difficult to extract biological meaning from the huge mass of available data, Artificial Intelligence (AI) approaches will be discussed as an increasingly important strategy for extracting knowledge from raw data and providing practitioners with actionable information to treat patients.
Collapse
Affiliation(s)
- Stefano Salvioli
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy; IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | | | - Leonardo Bencivenga
- Department of Translational Medical Sciences, University of Naples Federico II, Napoli, Italy
| | - Sara Carrino
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Maria Conte
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Sarah Damanti
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milano, Italy
| | - Rebecca De Lorenzo
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milano, Italy
| | - Eleonora Fiorenzato
- Parkinson's Disease and Movement Disorders Unit, Center for Rare Neurological Diseases (ERN-RND), Department of Neurosciences, University of Padova, Padova, Italy
| | - Alessandro Gialluisi
- Department of Epidemiology and Prevention, IRCCS NEUROMED, Pozzilli, Italy; EPIMED Research Center, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Assunta Ingannato
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Angelo Antonini
- Parkinson's Disease and Movement Disorders Unit, Center for Rare Neurological Diseases (ERN-RND), Department of Neurosciences, University of Padova, Padova, Italy; Center for Neurodegenerative Disease Research (CESNE), Department of Neurosciences, University of Padova, Padova, Italy
| | - Nicola Baldini
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Miriam Capri
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Simone Cenci
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milano, Italy
| | - Licia Iacoviello
- Department of Epidemiology and Prevention, IRCCS NEUROMED, Pozzilli, Italy; EPIMED Research Center, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica Delle Marche, Ancona, Italy; Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples Federico II, Napoli, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Scientific Institute of Telese Terme, Telese Terme, Italy
| | | | | |
Collapse
|