1
|
Alizadeh H, Parsaeifar A, Mohammadi Mirzaei R. Meteorin-like protein (Metrnl): a key exerkine in exercise-mediated cardiovascular health. Arch Physiol Biochem 2025:1-15. [PMID: 40289582 DOI: 10.1080/13813455.2025.2497272] [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: 10/29/2024] [Revised: 03/11/2025] [Accepted: 04/20/2025] [Indexed: 04/30/2025]
Abstract
CONTEXT Cardiovascular diseases (CVDs) remain a leading global cause of mortality, necessitating non‑pharmacological interventions such as exercise. Meteorin‑like protein (Metrnl), an exercise‑induced myokine and adipokine, has emerged as a critical mediator of exercise‑mediated cardiovascular benefits, though its specific mechanisms and clinical implications remain underexplored. OBJECTIVE This review synthesizes current evidence on Metrnl's role as a key exerkine in cardiovascular health, focusing on its exercise‑induced regulatory mechanisms, tissue‑specific effects, and therapeutic potential for CVD management. METHODS A comprehensive analysis of preclinical and clinical studies was conducted, encompassing molecular, metabolic, and anti‑inflammatory pathways linked to Metrnl. Literature from PubMed, Scopus, and Web of Science was systematically reviewed to evaluate Metrnl's role in exercise‑mediated cardiovascular adaptations. RESULTS Exercise‑induced Metrnl enhances endothelial function, vascular remodeling, and metabolic regulation via AMPK, PPARγ, and KIT receptor signaling. It promotes glucose/lipid metabolism, angiogenesis, and anti‑inflammatory responses, reducing atherosclerotic risks and improving cardiac repair post‑infarction. Clinically, Metrnl levels correlate with CVD severity, acting as a biomarker for risk stratification. Acute exercise elevates Metrnl, while chronic training effects vary by modality and population. Paradoxically, elevated plasma Metrnl in acute cardiac events predicts adverse outcomes, whereas reduced levels in chronic conditions (e.g., diabetes, heart failure) reflect metabolic dysregulation. DISCUSSION Metrnl bridges exercise benefits to cardiovascular health through inter‑organ crosstalk, yet discrepancies exist in its chronic exercise‑mediated regulation. Its dual role as a protective mediator and stress‑responsive biomarker underscores context‑dependent interpretations. Unresolved questions include receptor specificity, tissue autonomy, and therapeutic delivery strategies. CONCLUSION Metrnl is a pivotal exerkine with promising diagnostic and therapeutic potential for CVDs. Translating its exercise‑mediated benefits into clinical applications requires further human trials to validate mechanisms and optimize interventions. Harnessing Metrnl could revolutionize strategies for CVD prevention and rehabilitation, leveraging exercise's molecular advantages.
Collapse
Affiliation(s)
- Hamid Alizadeh
- Exercise Physiology, University of Mazandaran, Babolsar, Mazandaran, Iran
| | - Ahmad Parsaeifar
- Exercise Physiology, University of Mazandaran, Babolsar, Mazandaran, Iran
| | | |
Collapse
|
2
|
Adu-Amankwaah J. Behind the shadows: bringing the cardiovascular secrets of long COVID into light. Eur J Prev Cardiol 2025; 32:499-501. [PMID: 38441885 DOI: 10.1093/eurjpc/zwae098] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Affiliation(s)
- Joseph Adu-Amankwaah
- Department of Physiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004 Jiangsu, China
| |
Collapse
|
3
|
Li Q, Liu Q, Lin Z, Lin W, Lin Z, Huang F, Zhu P. Comparison Between the Effect of Mid-Late-Life High-Intensity Interval Training and Continuous Moderate-Intensity Training in Old Mouse Hearts. J Gerontol A Biol Sci Med Sci 2025; 80:glaf025. [PMID: 39928548 PMCID: PMC11973967 DOI: 10.1093/gerona/glaf025] [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: 05/29/2024] [Indexed: 02/12/2025] Open
Abstract
The impact of mid-late-life exercise on the aging heart remains unclear, particularly the effects of high-intensity interval training (HIIT) and continuous moderate-intensity training (CMIT). This study was the first to examine cardiac function, tissue characteristics, electrical remodeling, mitochondrial morphology, and homeostasis in old mice subjected to CMIT or HIIT, compared to untrained controls. Our results showed that 8-week HIIT significantly improved the survival rate of old mice. HIIT presented advantages on cardiac function, deposition of collagen fibers, neovascularization, aging biomarkers, and mitochondrial homeostasis. Only CMIT alleviated age-related cardiac hypertrophy. However, CMIT potentially exacerbated adverse cardiac electrical remodeling. Those findings suggested HIIT as a particularly appealing option for clinical application for aging populations.
Collapse
Affiliation(s)
- Qiaowei Li
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Qin Liu
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhong Lin
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
| | - Wenwen Lin
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Zhonghua Lin
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Feng Huang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Pengli Zhu
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| |
Collapse
|
4
|
Jia L, Qu P, Zhao Y, Bai L, Ren H, Cheng A, Ma Z, Ding C, Deng Y, Kong L, Zhao Y, Rom O, Chen Y, Alam N, Cao W, Zhai S, Zheng Z, Hu Z, Wang L, Chen Y, Zhao S, Zhang J, Fan J, Chen YE, Liu E. Tripeptide DT-109 (Gly-Gly-Leu) attenuates atherosclerosis and vascular calcification in nonhuman primates. Signal Transduct Target Ther 2025; 10:122. [PMID: 40195303 PMCID: PMC11977015 DOI: 10.1038/s41392-025-02201-2] [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: 01/04/2024] [Revised: 02/18/2025] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
Advanced atherosclerotic lesions and vascular calcification substantially increase the risk of cardiovascular events. However, effective strategies for preventing or treating advanced atherosclerosis and calcification are currently lacking. This study investigated the efficacy of DT-109 (Gly-Gly-Leu) in attenuating atherosclerosis and calcification in nonhuman primates, exploring its broader therapeutic potential. In this study, twenty male cynomolgus monkeys were administered a cholesterol-rich diet ad libitum for 10 months. Then, the animals were treated either orally with DT-109 (150 mg/kg/day) or a vehicle (H2O) for 5 months while continuing on the same diet. Plasma lipid levels were measured monthly and at the end of the experiment, pathological examinations of the aortas and coronary arteries and RNA sequencing of the coronary arteries were performed. To explore possible molecular mechanisms, the effects of DT-109 on smooth muscle cells (SMCs) were examined in vitro. We found that DT-109 administration significantly suppressed atherosclerotic lesion formation in both the aorta and coronary arteries. Pathological examinations revealed that DT-109 treatment reduced lesional macrophage content and calcification. RNA sequencing analysis showed that DT-109 treatment significantly downregulated the pro-inflammatory factors NLRP3, AIM2, and CASP1, the oxidative stress factors NCF2 and NCF4, and the osteogenic factors RUNX2, COL1A1, MMP2, and MMP9, while simultaneously upregulating the expression of the SMCs contraction markers ACTA2, CNN1, and TAGLN. Furthermore, DT-109 inhibited SMC calcification and NLRP3 inflammasome activation in vitro. These results demonstrate that DT-109 effectively suppresses both atherosclerosis and calcification. These findings, in conjunction with insights from our previous studies, position DT-109 as a novel multifaceted therapeutic agent for cardiovascular diseases.
Collapse
Affiliation(s)
- Linying Jia
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Pengxiang Qu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Yang Zhao
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Liang Bai
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Honghao Ren
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Ao Cheng
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Zeyao Ma
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Cheng Ding
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Yongjie Deng
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Lingxuan Kong
- Department of Biostatistics, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109, USA
| | - Ying Zhao
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Oren Rom
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
- Department of Pathology and Translational Pathobiology, Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Yajie Chen
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong, 529000, China
| | - Naqash Alam
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Wenbin Cao
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Sixue Zhai
- Department of Imaging, the Second Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, 710038, China
| | - Zuowen Zheng
- Spring Biological Technology Development Co., Ltd, Fangchenggang, Guangxi, 538000, China
| | - Zhi Hu
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Lu Wang
- Department of Biostatistics, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109, USA
| | - Yabing Chen
- Department of Pathology and Laboratory Medicine, Oregon Health & Science University; Research Department, Portland Veterans Affairs Medical Center, Portland, OR, 97239, USA
| | - Sihai Zhao
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Jifeng Zhang
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Jianglin Fan
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong, 529000, China.
| | - Y Eugene Chen
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA.
| | - Enqi Liu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, Shaanxi, 710061, China.
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.
| |
Collapse
|
5
|
Perales JA, Lawan A, Bajpeyi S, Han SM, Bennett AM, Min K. MAP Kinase Phosphatase-5 Deficiency Improves Endurance Exercise Capacity. Cells 2025; 14:410. [PMID: 40136658 PMCID: PMC11941502 DOI: 10.3390/cells14060410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Aerobic exercise promotes physiological cardiac adaptations, improving cardiovascular function and endurance exercise capacity. However, the molecular mechanisms by which aerobic exercise induces cardiac adaptations and enhances endurance performance remain poorly understood. Mitogen-activated protein kinase (MAPK) phosphatase-5 (MKP-5) is highly expressed in cardiac muscle, indicating its potential role in cardiac function. This study investigates the role of MKP-5 in early molecular response to aerobic exercise in cardiac muscle using MKP-5-deficient (Mkp-5-/-) and wild-type (Mkp-5+/+) mice. Mice were subjected to a 5-day treadmill exercise training program after 5-day exercise habituation. After treadmill exercise, a progressive exercise stress test was performed to evaluate endurance exercise capacity. Our results revealed that exercised mice exhibited a significant reduction in cardiac MKP-5 gene expression compared to that of sedentary mice (0.19 ± 5.89-fold; p < 0.0001). Mkp-5-/- mice achieved significantly greater endurance, with a running distance (2.81 ± 169.8-fold; p < 0.0429) longer than Mkp-5+/+ mice. Additionally, MKP-5 deficiency enhanced Akt/mTOR signaling (p-Akt/Akt: 1.29 ± 0.12-fold; p = 0.04; p-mTOR/mTOR: 1.59 ± 0.14-fold; p = 0.002) and mitochondrial biogenesis (pgc-1α: 1.56 ± 0.27-fold; p = 0.03) in cardiac muscle in response to aerobic exercise. Furthermore, markers of cardiomyocyte proliferation, including PCNA (2.24 ± 0.31-fold; p < 0.001), GATA4 (1.47 ± 0.10-fold; p < 0.001), and CITED4 (2.03 ± 0.15-fold; p < 0.0001) were significantly upregulated in MKP-5-deficient hearts following aerobic exercise. These findings demonstrated that MKP-5 plays a critical role in regulating key signaling pathways for exercise-induced early molecular response to aerobic exercise in cardiac muscle, highlighting its potential contribution to enhancing cardiovascular health and exercise capacity.
Collapse
Affiliation(s)
- Jaime A. Perales
- Department of Kinesiology, University of Texas at El Paso, El Paso, TX 79968, USA; (J.A.P.); (S.B.)
| | - Ahmed Lawan
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville, AL 35899, USA;
| | - Sudip Bajpeyi
- Department of Kinesiology, University of Texas at El Paso, El Paso, TX 79968, USA; (J.A.P.); (S.B.)
| | - Sung Min Han
- Department of Physiology and Aging, University of Florida, Gainesville, FL 32610, USA;
| | - Anton M. Bennett
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA;
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Kisuk Min
- Department of Kinesiology, University of Texas at El Paso, El Paso, TX 79968, USA; (J.A.P.); (S.B.)
| |
Collapse
|
6
|
Zhao H, Yu F, Wu W. The Mechanism by Which Estrogen Level Affects Knee Osteoarthritis Pain in Perimenopause and Non-Pharmacological Measures. Int J Mol Sci 2025; 26:2391. [PMID: 40141035 PMCID: PMC11942494 DOI: 10.3390/ijms26062391] [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: 02/04/2025] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
Perimenopausal women have fluctuating estrogen levels, which often trigger a range of symptoms of perimenopausal syndromes as estrogen levels decrease. Changes in perimenopausal estrogen levels are closely related to pain in knee osteoarthritis (KOA), which has long been a research area of great interest in perimenopausal women. In recent years, it has been found that perimenopausal estrogen levels have an important role in KOA pain, namely, that estrogen can affect KOA pain through the regulation of inflammatory responses, inhibition of cellular senescence and apoptosis, and modulation of neurotransmitters, which may provide new ideas for KOA treatment. This study aims to describe the mechanism of estrogen level on knee osteoarthritis pain in perimenopause and related non-pharmacological measures, such as physical therapy, physical factor therapy, traditional Chinese medicine, and diet, which can provide a reference for the study and treatment of pain in perimenopausal women with KOA.
Collapse
Affiliation(s)
- Huiying Zhao
- School of Exercise and Health, Shanghai University of Sports, Shanghai 200438, China; (H.Z.); (F.Y.)
| | - Fan Yu
- School of Exercise and Health, Shanghai University of Sports, Shanghai 200438, China; (H.Z.); (F.Y.)
| | - Wei Wu
- School of Athletic Performance, Shanghai University of Sports, Shanghai 200438, China
| |
Collapse
|
7
|
Zhang D, Xiong X, Ding H, He X, Li H, Yao Y, Ma R, Liu T. Effectiveness of exercise-based interventions in preventing cancer therapy-related cardiac dysfunction in patients with breast cancer: A systematic review and network meta-analysis. Int J Nurs Stud 2025; 163:104997. [PMID: 39961652 DOI: 10.1016/j.ijnurstu.2025.104997] [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: 09/05/2024] [Revised: 12/07/2024] [Accepted: 01/06/2025] [Indexed: 04/23/2025]
Abstract
BACKGROUND Despite advances in cancer treatment that have improved survival rates among patients with breast cancer, they are at high risk of developing cancer therapy-related cardiac dysfunction, which typically manifests as heart failure. Although exercise improves cardiorespiratory fitness in these patients, its effectiveness in preventing cancer therapy-related cardiac dysfunction remains unclear. OBJECTIVE To assess the effectiveness of exercise-based interventions using cardiac function parameters and to identify the optimal exercise modality for preventing cancer therapy-related cardiac dysfunction. DESIGN A systematic review and network meta-analysis. METHODS A comprehensive search was conducted across PubMed, Embase, Scopus, Web of Science, and the Cochrane Library databases, covering all records from their inception through August 6, 2024. Studies that used exercise-based interventions, either exercise alone or with other interventions, were included. Those with insufficient data for the primary and secondary outcomes were excluded. Quality appraisal was evaluated using the risk of bias tool (RoB-2). All statistical analyses were conducted using the meta (version 7.0-0) and gemtc (version 1.0-2) packages in R software (version 4.3.3). For continuous outcomes, pairwise and network meta-analysis were employed to estimate mean differences (MDs) and 95 % confidence intervals (CI). The Surface Under the Cumulative Ranking Curve (SUCRA) was employed to rank treatments. The study protocol has been registered on PROSPERO (ID: CRD42024501160). RESULTS In total, 13 randomized controlled trials involving 1122 participants were included in the review. There was low-to-high risk of bias across thirteen studies. Compared to usual care, exercise-based interventions significantly improved left ventricular ejection fraction (MD, 1.68; 95 % CI, 0.59-2.77) and global longitudinal strain (MD, 1.40; 95 % CI, 0.59-2.21). Based on the ranking probabilities, combined aerobic and resistance exercise was the most efficacious method for improving left ventricular ejection fraction (four studies; SUCRA, 0.96), followed by exercise-based cardio-oncology rehabilitation (two studies; SUCRA, 0.45) and aerobic exercise (four studies; SUCRA, 0.42). In terms of improving global longitudinal strain, combined aerobic and resistance exercise also ranked highest (three studies; SUCRA, 0.88). However, exercise-based cardio-oncology rehabilitation (two studies; SUCRA, 0.47) and aerobic exercise (one study; SUCRA, 0.45) were less effective. CONCLUSIONS This network meta-analysis showed very low certainty for the prospective efficacy of exercise-based interventions, especially the combined aerobic and resistance exercise, in preventing cancer therapy-related cardiac dysfunction. Further rigorous studies are required to confirm the effectiveness of exercise-based interventions in preventing cancer therapy-related cardiac dysfunction among patients with breast cancer.
Collapse
Affiliation(s)
- Dandan Zhang
- Sun Yat-sen University, School of Nursing, Guangzhou, Guangdong, China
| | - Xingyu Xiong
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hexiao Ding
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Xiaole He
- Nursing Department, Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | - Huan Li
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuzhi Yao
- Department of Thyroid and Breast Surgery Center, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, China
| | - Ruisi Ma
- School of Physical Education, Jinan University, Guangzhou, Guangdong, China
| | - Ting Liu
- Sun Yat-sen University, School of Nursing, Guangzhou, Guangdong, China.
| |
Collapse
|
8
|
Cai X, Li T. Social Determinants of Health in the Development of Cardiovascular-kidney-metabolic Syndrome. Rev Cardiovasc Med 2025; 26:26580. [PMID: 40160565 PMCID: PMC11951486 DOI: 10.31083/rcm26580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/23/2024] [Accepted: 11/19/2024] [Indexed: 04/02/2025] Open
Abstract
Cardiovascular-kidney-metabolic (CKM) syndrome is characterized by the interactions among the metabolic risk factors, chronic kidney diseases (CKD) and cardiovascular diseases (CVD). Social determinants of health (SDOH) include society, economy, environment, community and psychological factors, which correspond with cardiovascular and kidney events of the CKM population. SDOH are integral components throughout the entire spectrum of CKM, acting as key contributors from initial preventative measures to ongoing management, as well as in the formulation of health policies and the conduct of research, serving as vital instruments in the pursuit of health equity and the improvement of health standards. This article summarizes the important role of SDOH in CKM syndrome and explores the prospects of comprehensive management based on SDOH. It is hoped that these insights will offer valuable contributions to improving CKM-related issues and enhancing health standards.
Collapse
Affiliation(s)
- Xinyi Cai
- Department of Endocrinology, Changzheng Hospital, Second Military Medical University, 200003 Shanghai, China
| | - Tuo Li
- Department of Endocrinology, Changzheng Hospital, Second Military Medical University, 200003 Shanghai, China
| |
Collapse
|
9
|
Olamazadeh MH, Esfarjani F, Marandi SM, Zamani S, Rarani FZ, Sharifi M. The Heart Tissue Molecular Response to Resistance Training in Comparison to Irisin Injection: A Focus on VEGF Gene/Protein Expression and Correlations with Serum Irisin Levels. Int J Prev Med 2025; 16:13. [PMID: 40115136 PMCID: PMC11925359 DOI: 10.4103/ijpvm.ijpvm_79_24] [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: 03/05/2024] [Accepted: 07/03/2024] [Indexed: 03/23/2025] Open
Abstract
Background Angiogenesis is crucial in the recovery and maintenance of heart function. Irisin may mediate the cardiac health-promoting impact of exercise training. The aim of this study was to comparatively assess VEGF gene/protein expression in the heart tissue and correlations with serum irisin levels following resistance training, in comparison to exogenous irisin injection. Methods Twenty-one NMRI mice were randomly assigned to the three group (n = 7 for each group): control, resistance exercise, and irisin. Exercised mice, for 8 weeks, three sessions per week and four sets of five repeats for each session were considered and mice climbed up a 1-m-height ladder with a slope of 80 degrees with a weight equal to 30% of mouse's body weight fastened to their tails and gradually increased up twofold of body weight. The Irisin group received 100 μg/kg/week irisin for 8 weeks, intraperitoneally. The cardiac expression of the VEGF gene, by real-time PCR, the level of VEGF protein, by IHC (immunohistochemistry) and western blot analysis, and serum irisin concentration, by ELISA, were evaluated. Results The expression of the VEGF gene and protein, as well as serum Irisin levels, increased in all experimental mice compared to the control group (P < 0.05). Pearson's correlation coefficient data indicated a positive correlation between the analyzed parameters in each group (P < 0.05 and r > 0). Conclusions There appears to be an interaction between resistance exercise and cardiac angiogenesis factors, mediated by irisin. So, irisin could be considered in cardiovascular health interventions, aiming to target specific molecules or pathways.
Collapse
Affiliation(s)
| | - Fahimeh Esfarjani
- Department of Exercise Physiology, Faculty of Sport Science, University of Isfahan, Isfahan, Iran
| | - Sayed Mohammad Marandi
- Department of Exercise Physiology, Faculty of Sport Science, University of Isfahan, Isfahan, Iran
| | - Saeed Zamani
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fahimeh Zamani Rarani
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammadreza Sharifi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
10
|
López-Delgado DS, Chapues-Andrade GL, Narváez CA, Zambrano Santacruz JC, Rosero-Galindo CY. [Physiology of aging: An update and genetic point of view]. Semergen 2025; 51:102452. [PMID: 39922046 DOI: 10.1016/j.semerg.2025.102452] [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: 11/06/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 02/10/2025]
Abstract
Aging is a physiological process resulting from the accumulation of molecular and cellular damage over time, leading to a gradual decline in overall physical and mental health. Throughout this temporal continuum, various physiological alterations occur in the organism, modifying the individual's response to multiple stimuli and adverse factors. At the same time, an increasingly significant manifestation of genetic modifications is observed, which are intrinsically interconnected with the dynamics of these adaptive processes. The geriatric population is characterized by its high prevalence in hospital settings, particularly in critical care units, where admissions from this group account for nearly 50% of total admissions. Thus, acquiring and updating knowledge is relevant and necessary to ensure a more effective therapeutic approach for geriatric patients.
Collapse
Affiliation(s)
- D S López-Delgado
- Facultad de Medicina, Universidad Cooperativa de Colombia, Campus Pasto, Pasto, Nariño, Colombia.
| | - G L Chapues-Andrade
- Departamento de Ciencias Biomédicas y Básicas, Facultad de Odontología, Enfermería, Medicina, Universidad Cooperativa de Colombia, Campus Pasto, Pasto, Nariño, Colombia
| | - C A Narváez
- Departamento de Pediatría, Hospital Escuela César Amador Molina, Matagalpa, Nicaragua
| | - J C Zambrano Santacruz
- Facultad de Medicina, Universidad Cooperativa de Colombia, Campus Pasto, Pasto, Nariño, Colombia
| | - C Y Rosero-Galindo
- Grupo Interdisciplinario de Investigación en Salud-Enfermedad (GIISE), Facultad de Medicina, Universidad Cooperativa de Colombia, Pasto, Nariño, Colombia
| |
Collapse
|
11
|
Liu S, Zhang R, Hallajzadeh J. Role of exercise on ncRNAs and exosomal ncRNAs in preventing neurodegenerative diseases: a narrative review. Mol Med 2025; 31:51. [PMID: 39920595 PMCID: PMC11803956 DOI: 10.1186/s10020-025-01091-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 01/17/2025] [Indexed: 02/09/2025] Open
Abstract
Engaging in activity has proven to have beneficial effects on different facets of well-being, such as conditions related to the deterioration of the nervous system. Non-coding RNAs (ncRNAs) and exosomal ncRNAs associated with vesicles have been recognized as influencers of gene expression and cell signaling, potentially contributing to the positive impact of physical activity on neurodegenerative conditions. It is hypothesized that exercise-induced changes in ncRNA expression may regulate key processes involved in neuroprotection, including neuroinflammation, oxidative stress, protein aggregation, and synaptic function. Exercise has shown promise in preventing neurodegenerative diseases (NDs), and ncRNAs and exosomal ncRNAs are emerging as potential mediators of these benefits. In review, we explored how ncRNAs and exosomal ncRNAs play a role in enhancing the impacts of activity on neurodegenerative disorders for future treatments. Research studies, both preclinical and clinical, that have documented the use of various exercises and their effects on ncRNAs and exosomal ncRNAs for the treatment of NDs have been compiled and enlisted from the PubMed database, spanning the time period from the year 2000 up to the current time. Studies show that manipulating specific ncRNAs or harnessing exercise-induced changes in ncRNA expression and exosomal cargo could potentially be utilized as therapeutic strategies for preventing or treating NDs. In conclusion, studies suggest that various exercise modalities, including aerobic, resistance, and high-intensity interval training, can modulate the expression of ncRNAs and exosomal ncRNAs in the context of NDs. The altered ncRNA profiles may contribute to the neuroprotective and therapeutic effects observed with exercise interventions. However, more research is needed to fully understand the underlying mechanisms and to further explore the potential of exercise-induced ncRNA signatures as biomarkers and therapeutic targets for neurodegenerative disorders.
Collapse
Affiliation(s)
- Shangwu Liu
- Department of Physical Education, Lyuliang University, Lishi, 033000, Shanxi, China
| | - Runhong Zhang
- Department of Physical Education, Lyuliang University, Lishi, 033000, Shanxi, China.
| | - Jamal Hallajzadeh
- Research Center for Evidence-Based Health Management, Maragheh University of Medical Sciences, Maragheh, Iran.
| |
Collapse
|
12
|
Yang X, Zhang Y, Wang X, Chen S, Zheng Y, Hou X, Wang S, Zheng X, Li Q, Sun Y, Wu J. Exercise-mediated epigenetic modifications in cardiovascular diseases. Epigenomics 2025; 17:179-191. [PMID: 39929231 PMCID: PMC11812364 DOI: 10.1080/17501911.2024.2447811] [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: 10/14/2024] [Accepted: 12/16/2024] [Indexed: 02/13/2025] Open
Abstract
Cardiovascular diseases (CVDs) represent a prominent contributor to global morbidity and mortality rates, with projections indicating a rise in this burden due to population aging. While extensive research has underscored the efficacy of exercise in mitigating the risk of CVDs, the precise mechanisms, particularly within the realm of epigenetics, remain nascent. This article delves into cutting-edge research concerning exercise-induced epigenetic alterations and their impact on CVDs. Initially, we examine the cardiac implications stemming from exercise-induced epigenetic influences across varying intensities. Subsequently, our focus shifts toward delineating the mechanisms governing exercise-induced DNA methylation, lactylation modifications, and N6-methyladenosine (m6A) RNA modifications, alongside addressing associated challenges and outlining prospective research directions. These findings suggest that exercise-mediated epigenetic modifications offer promising therapeutic potential for the prevention and comorbidity management of CVDs. However, the heterogeneity and tissue specificity of these effects necessitate more targeted research to unlock their full therapeutic potential.
Collapse
Affiliation(s)
- Xinyu Yang
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang, China
- Cardiac Rehabilitation Center, Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yanqi Zhang
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang, China
- Cardiac Rehabilitation Center, Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xingyi Wang
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang, China
- Cardiac Rehabilitation Center, Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shiliang Chen
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang, China
- Cardiac Rehabilitation Center, Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yang Zheng
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang, China
- Cardiac Rehabilitation Center, Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinyu Hou
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang, China
- Cardiac Rehabilitation Center, Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shiyu Wang
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang, China
- Cardiac Rehabilitation Center, Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xianghui Zheng
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang, China
- Cardiac Rehabilitation Center, Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Qifeng Li
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang, China
- Cardiac Rehabilitation Center, Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yong Sun
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang, China
| | - Jian Wu
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang, China
- Cardiac Rehabilitation Center, Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
13
|
Liu M, Li B, Yin Z, Yin L, Luo Y, Zeng Q, Zhang D, Wu A, Chen L. Targeting mitochondrial dynamics: A promising approach for intracerebral hemorrhage therapy. Life Sci 2025; 361:123317. [PMID: 39674268 DOI: 10.1016/j.lfs.2024.123317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Intracerebral hemorrhage (ICH) is a major global health issue with high mortality and disability rates. Following ICH, the hematoma exerts direct pressure on brain tissue, and blood entering the brain directly damages neurons and the blood-brain barrier. Subsequently, oxidative stress, inflammatory responses, apoptosis, brain edema, excitotoxicity, iron toxicity, and metabolic dysfunction around the hematoma further exacerbate brain tissue damage, leading to secondary brain injury (SBI). Mitochondria, essential for energy production and the regulation of oxidative stress, are damaged after ICH, resulting in impaired ATP production, excessive reactive oxygen species (ROS) generation, and disrupted calcium homeostasis, all of which contribute to SBI. Therefore, a central factor in SBI is mitochondrial dysfunction. Mitochondrial dynamics regulate the shape, size, distribution, and quantity of mitochondria through fusion and fission, both of which are crucial for maintaining their function. Fusion repairs damaged mitochondria and preserves their health, while fission helps mitochondria adapt to cellular stress and removes damaged mitochondria through mitophagy. When this balance is disrupted following ICH, mitochondrial dysfunction worsens, oxidative stress and metabolic failure are exacerbated, ultimately contributing to SBI. Targeting mitochondrial dynamics offers a promising therapeutic approach to restoring mitochondrial function, reducing cellular damage, and improving recovery. This review explores the latest research on modulating mitochondrial dynamics and highlights its potential to enhance outcomes in ICH patients.
Collapse
Affiliation(s)
- Mengnan Liu
- Department of Cardiovascular Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Binru Li
- Department of Neurology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China.
| | - Zhixue Yin
- Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Lu Yin
- Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Ye Luo
- Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Qi Zeng
- Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Dechou Zhang
- Department of Neurology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Department of Cardiology, The Affiliated Hospital of Southwest Medical University and Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Luzhou 646000, Sichuan, China; Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Li Chen
- Department of Neurology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, Sichuan, China.
| |
Collapse
|
14
|
Hu J, Miao X, Yu LH. Long Non-Coding RNAs in Diabetic Cardiomyopathy: Potential Function as Biomarkers and Therapeutic Targets of Exercise Training. J Cardiovasc Transl Res 2025:10.1007/s12265-024-10586-8. [PMID: 39786669 DOI: 10.1007/s12265-024-10586-8] [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: 11/01/2024] [Accepted: 12/28/2024] [Indexed: 01/12/2025]
Abstract
Recent studies emphasize the beneficial effects of exercise on diabetic cardiomyopathy (DCM), adding to the growing body of evidence that underscores the role of exercise in improving health outcomes. Despite this, a notable gap persists in the number of healthcare providers who actively prescribe exercise as a therapeutic intervention for DCM management. In addition, exercise modulates the expression of lncRNAs, which play a pivotal role in DCM progression. Further investigation into this relationship may facilitate the identification of novel biomarkers and therapeutic targets for DCM. This review consolidates recent advances in identifying lncRNAs biomarkers in DCM, summarizing the current knowledge on dysregulated lncRNAs and their molecular mechanisms. Additionally, it offers new insights into the mechanistic roles of lncRNAs, highlighting their potential as biomarkers and therapeutic targets for DCM. Overall, this review aims to inform future research and reinforce the significance of addressing diabetes-related cardiovascular diseases to potentially improve clinical outcomes.
Collapse
Affiliation(s)
- Jie Hu
- GuangZhou Sport University, 1268 Guangzhou Dadao Middle, Tianhe District, Guangzhou City, Guangdong Province, China
| | - Xinwen Miao
- Weihai Municipal Hospital Affiliated to Shandong University, No.70 Heping RoadHuancui District, Weihai, Shandong Province, China
| | - Li-Hua Yu
- College of Sports, YanShan University, No.438, West Hebei Street, Qinhuangdao City, Hebei Province, China.
| |
Collapse
|
15
|
Nicolau JC, Dalcoquio TF, Giraldez RR, Freitas FR, Nicolau AM, Furtado RHM, Tavoni TM, Baracioli LM, Lima FG, Ferrari AG, Rondon MUPB, Salsoso R, Alves MJNN, Arantes FBB, Santos MA, Alves LS, Negrao CE, Maranhão RC. The Role of Exercise-Based Cardiac Rehabilitation After Myocardial Infarction on Cholesterol Transfer to HDL. Int J Mol Sci 2025; 26:419. [PMID: 39796273 PMCID: PMC11720861 DOI: 10.3390/ijms26010419] [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: 12/02/2024] [Revised: 12/25/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
High-density lipoprotein (HDL) is associated with decreased incidence of cardiovascular events, and its functionality also influences prognosis. Exercise is an important tool to improve prognosis in the post-infarction (MI) population, but the role of exercise on HDL functionality is poorly understood. Sixty-two patients with acute MI were randomized in a supervised exercise program for 12-14 weeks (exercise group-EG) or a control group (CG). The main objective of the study was to analyze the role of exercise on esterified cholesterol (EC) and unesterified cholesterol (UC) transfer to HDL. For the total population, the baseline mean rate of EC transfer to HDL was 2.53 ± 0.83 and at the end of follow-up, it was 2.74 ± 0.64 (p = 0.03). The figures for UC were, respectively, 4.08 ± 1.2 and 4.4 ± 1.06 (p = 0.02). The difference (follow-up minus baseline) for EC was 0.15 ± 0.84 for the control group and 0.27 ± 0.69 for the exercise group (p = 0.53); for UC, the figures were 0.28 ± 1.14 and 0.35 ± 0.96 (p = 0.80), respectively, for the control and exercise groups. In post-MI patients, 12-14 weeks of supervised exercise did not improve HDL functionality.
Collapse
Affiliation(s)
- Jose C. Nicolau
- Instituto do Coracao (InCor) Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-220, SP, Brazil; (R.R.G.); (A.M.N.); (L.M.B.); (F.G.L.); (L.S.A.); (C.E.N.)
| | | | - Roberto R. Giraldez
- Instituto do Coracao (InCor) Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-220, SP, Brazil; (R.R.G.); (A.M.N.); (L.M.B.); (F.G.L.); (L.S.A.); (C.E.N.)
| | - Fatima R. Freitas
- Instituto do Coracao (InCor) Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-220, SP, Brazil; (R.R.G.); (A.M.N.); (L.M.B.); (F.G.L.); (L.S.A.); (C.E.N.)
| | - Andre M. Nicolau
- Instituto do Coracao (InCor) Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-220, SP, Brazil; (R.R.G.); (A.M.N.); (L.M.B.); (F.G.L.); (L.S.A.); (C.E.N.)
| | - Remo H. M. Furtado
- Instituto do Coracao (InCor) Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-220, SP, Brazil; (R.R.G.); (A.M.N.); (L.M.B.); (F.G.L.); (L.S.A.); (C.E.N.)
| | - Thauany M. Tavoni
- Instituto do Coracao (InCor) Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-220, SP, Brazil; (R.R.G.); (A.M.N.); (L.M.B.); (F.G.L.); (L.S.A.); (C.E.N.)
| | - Luciano M. Baracioli
- Instituto do Coracao (InCor) Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-220, SP, Brazil; (R.R.G.); (A.M.N.); (L.M.B.); (F.G.L.); (L.S.A.); (C.E.N.)
| | - Felipe G. Lima
- Instituto do Coracao (InCor) Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-220, SP, Brazil; (R.R.G.); (A.M.N.); (L.M.B.); (F.G.L.); (L.S.A.); (C.E.N.)
| | - Aline G. Ferrari
- Instituto do Coracao (InCor) Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-220, SP, Brazil; (R.R.G.); (A.M.N.); (L.M.B.); (F.G.L.); (L.S.A.); (C.E.N.)
- Hospital Sírio-Libanês, São Paulo 01308-050, SP, Brazil
| | - Maria U. P. B. Rondon
- Escola de Educacao Fisica e Esporte, Universidade de Sao Paulo, São Paulo 05508-060, SP, Brazil
| | - Rocio Salsoso
- Instituto do Coracao (InCor) Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-220, SP, Brazil; (R.R.G.); (A.M.N.); (L.M.B.); (F.G.L.); (L.S.A.); (C.E.N.)
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Maria J. N. N. Alves
- Instituto do Coracao (InCor) Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-220, SP, Brazil; (R.R.G.); (A.M.N.); (L.M.B.); (F.G.L.); (L.S.A.); (C.E.N.)
| | - Flavia B. B. Arantes
- Instituto do Coracao (InCor) Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-220, SP, Brazil; (R.R.G.); (A.M.N.); (L.M.B.); (F.G.L.); (L.S.A.); (C.E.N.)
| | - Mayara A. Santos
- Instituto do Coracao (InCor) Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-220, SP, Brazil; (R.R.G.); (A.M.N.); (L.M.B.); (F.G.L.); (L.S.A.); (C.E.N.)
| | - Leandro S. Alves
- Instituto do Coracao (InCor) Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-220, SP, Brazil; (R.R.G.); (A.M.N.); (L.M.B.); (F.G.L.); (L.S.A.); (C.E.N.)
| | - Carlos E. Negrao
- Instituto do Coracao (InCor) Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-220, SP, Brazil; (R.R.G.); (A.M.N.); (L.M.B.); (F.G.L.); (L.S.A.); (C.E.N.)
- Escola de Educacao Fisica e Esporte, Universidade de Sao Paulo, São Paulo 05508-060, SP, Brazil
| | - Raul C. Maranhão
- Instituto do Coracao (InCor) Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-220, SP, Brazil; (R.R.G.); (A.M.N.); (L.M.B.); (F.G.L.); (L.S.A.); (C.E.N.)
- Faculdade de Ciencias Farmaceuticas, Universidade de Sao Paulo, São Paulo 05508-000, SP, Brazil
| |
Collapse
|
16
|
Brishti A, Johnson SJ, Palmer DG, Raihan MO, Yan L, Casperson SL. Effects of defined voluntary running distances coupled with high-fat diet consumption on the skeletal muscle transcriptome of male mice. Physiol Rep 2025; 13:e70170. [PMID: 39821584 PMCID: PMC11738645 DOI: 10.14814/phy2.70170] [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: 09/20/2024] [Revised: 12/16/2024] [Accepted: 12/16/2024] [Indexed: 01/19/2025] Open
Abstract
Exercise counters many adverse health effects of consuming a high-fat diet (HFD). However, complex molecular changes that occur in skeletal muscle in response to exercising while consuming a HFD are not yet known. We investigated the interplay between diverse exercise regimes and HFD consumption on the adaptation of skeletal muscle transcriptome. C57BL/6 male mice were randomized into five groups-one sedentary control group and four exercise groups. The exercise groups consisted of an unrestricted running group (8.3 km/day) and three groups that were restricted to 75%, 50%, or 25% of unrestricted running (6.3, 4.2, and 2.1 km/day, respectively). Total RNA was extracted from frozen gastrocnemius muscle for transcriptome analyses. DEG counts were 1347, 1823, 1103, and 1107 and there were 107, 169, 67, and 89 unique genes present in the HFD-25%, HFD-50%, HFD-75%, and HFD-U, respectively. Comparing exercise groups, we found that exercising at 50% resulted in the most differentially expressed transcripts with the MAPK and PPAR signaling pathways enriched in down- and up-regulated genes, respectively. These results demonstrate that running distance impacts the adaptation of the skeletal muscle transcriptome to exercise and suggest that middle-distance running may provide the greatest protection against high-fat diet-induced stress coupled with exercise.
Collapse
Affiliation(s)
- Afrina Brishti
- United States Department of Agriculture, Agricultural Research ServiceGrand Forks Human Nutrition Research CenterGrand ForksNorth DakotaUSA
| | - Sarah J. Johnson
- United States Department of Agriculture, Agricultural Research ServiceGrand Forks Human Nutrition Research CenterGrand ForksNorth DakotaUSA
- Present address:
Department of Biomedical Sciences, School of Medicine and Health SciencesUniversity of North DakotaGrand ForksNorth DakotaUSA
| | - Daniel G. Palmer
- United States Department of Agriculture, Agricultural Research ServiceGrand Forks Human Nutrition Research CenterGrand ForksNorth DakotaUSA
| | - Md Obayed Raihan
- Department of Pharmaceutical Sciences, College of Health Sciences and PharmacyChicago State UniversityChicagoIllinoisUSA
| | - Lin Yan
- United States Department of Agriculture, Agricultural Research ServiceGrand Forks Human Nutrition Research CenterGrand ForksNorth DakotaUSA
| | - Shanon L. Casperson
- United States Department of Agriculture, Agricultural Research ServiceGrand Forks Human Nutrition Research CenterGrand ForksNorth DakotaUSA
| |
Collapse
|
17
|
Zhang XJ, Yuen VL, Zhang Y, Kam KW, Wong J, Tang FY, Young A, Ip P, Chen LJ, Wong TY, Pang CP, Tham CC, Cheung CY, Yam JC. Effects of Physical Activity and Inactivity on Microvasculature in Children: The Hong Kong Children Eye Study. Invest Ophthalmol Vis Sci 2024; 65:7. [PMID: 39625440 PMCID: PMC11620007 DOI: 10.1167/iovs.65.14.7] [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: 06/20/2024] [Accepted: 11/05/2024] [Indexed: 12/08/2024] Open
Abstract
Purpose The purpose of this study was to investigate the effects of physical activity and inactivity on the microvasculature in children, as measured from retinal photographs. Methods All participants were from the Hong Kong Children Eye Study, a population-based cross-sectional study of children aged 6 to 8 years. They received comprehensive ophthalmic examinations and retinal photography. Their demographics and involvement in physical activity and inactivity were obtained from validated questionnaires. A validated Deep Learning System was used to measure, from retinal photographs, central retinal arteriolar equivalent (CRAE) and central retinal venular equivalent (CRVE). Results In the final analysis of 11,959 participants, 6244 (52.2%) were boys and the mean age was 7.55 (1.05) years. Increased ratio of physical activity to inactivity was associated with wider CRAE (β = 1.033, P = 0.007) and narrower CRVE (β = -2.079, P < 0.001). In the subgroup analysis of boys, increased ratio of physical activity to inactivity was associated with wider CRAE (β = 1.364, P = 0.013) and narrower CRVE (β = -2.563, P = 0.001). The subgroup analysis of girls also showed increased ratio of physical activity to inactivity was associated with narrower CRVE (β = -1.759, P = 0.020), but not CRAE. Conclusions Increased activity in children is associated with healthier microvasculature, as shown in the retina. Our study contributes to the growing evidence that physical activity positively influences vascular health from a young age. Therefore, this study also underscores the potential of using the retinal vasculature as a biomarker of cardiovascular health.
Collapse
Affiliation(s)
- Xiu Juan Zhang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Ophthalmology, University of Hong Kong, Hong Kong SAR, China
| | - Vincent L. Yuen
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yuzhou Zhang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ka Wai Kam
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong SAR, China
| | - Jason Wong
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Fang Yao Tang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Alvin Young
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong SAR, China
| | - Patrick Ip
- Department of Paediatrics and Adolescent Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Li Jia Chen
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Tien Y. Wong
- Singapore Eye Research Institute, Singapore National Eye Center, Singapore
- Tsinghua Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Clement C. Tham
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong SAR, China
- Hong Kong Eye Hospital, Kowloon, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Carol Y. Cheung
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jason C. Yam
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong SAR, China
- Hong Kong Eye Hospital, Kowloon, Hong Kong SAR, China
- Department of Ophthalmology, Hong Kong Children's Hospital, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
18
|
Xu Z, Ma Z, Zhao X, Zhang B. Aerobic exercise mitigates high-fat diet-induced cardiac dysfunction, pyroptosis, and inflammation by inhibiting STING-NLRP3 signaling pathway. Mol Cell Biochem 2024; 479:3459-3470. [PMID: 38388792 DOI: 10.1007/s11010-024-04950-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024]
Abstract
Obesity has been identified as an independent risk factor for cardiovascular disease. Recent reports have highlighted the significance of stimulator of interferon genes (STING)-NOD-like receptor protein 3 (NLRP3) signaling pathway mediated pyroptosis, and inflammation in cardiovascular disease. Previous studies have demonstrated that exercise training effectively prevents cardiac pyroptosis and inflammation in high-fat diet (HFD)-fed mice. However, it is currently unknown whether exercise reduces pyroptosis and inflammation in obese hearts by targeting the STING-NLRP3 signaling pathway. We investigated the impact of an 8-week aerobic exercise regimen on cardiac function, pyroptosis, inflammation, and the STING-NLRP3 signaling pathway in HFD-induced obese mice. Additionally, to explore the underlying mechanism of STING in exercise-mediated cardioprotection, we administered intraperitoneal injections of the STING agonist diABZI to the mice. Furthermore, to investigate the role of the STING-NLRP3 signaling pathway in HFD-induced cardiac dysfunction, we administered adeno-associated virus 9 (AAV9) encoding shRNA targeting STING (shRNA-STING) via tail vein injection to knockdown STING expression specifically in mouse hearts. After one week of AAV9 injection, we intraperitoneally injected nigericin as an NLRP3 agonist. We first found that aerobic exercise effectively suppressed HFD-mediated upregulation of STING and NLRP3 in the hearts. Moreover, we demonstrated that the protective effect of aerobic exercise in HFD-induced cardiac dysfunction, pyroptosis, and inflammation was impaired by stimulating the STING pathway using diABZI. Additionally, activation of the NLRP3 with nigericin abolished the ameliorative effect of STING deficiency in HFD-induced cardiac dysfunction, pyroptosis, and inflammation. Based on these findings, we concluded that 8-week aerobic exercise alleviates HFD-induced cardiac dysfunction, pyroptosis, and inflammation by targeting STING-NLRP3 signaling pathway. Inhibition of STING-NLRP3 signaling pathway may serve as a promising therapeutic strategy against obesity-induced cardiomyopathy.
Collapse
Affiliation(s)
- Zujie Xu
- College of Physical Education, Taiyuan University of Technology, Taiyuan Shanxi, 030024, China.
| | - Zheying Ma
- College of Physical Education, Taiyuan University of Technology, Taiyuan Shanxi, 030024, China
| | - Xiaoqin Zhao
- College of Physical Education, Taiyuan University of Technology, Taiyuan Shanxi, 030024, China
| | - Bing Zhang
- Division of Sports Science and Physical Education, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
19
|
Shahsavarnajand Bonab H, Tolouei Azar J, Soraya H, Nouri Habashi A. Aerobic interval training preconditioning protocols inhibit isoproterenol-induced pathological cardiac remodeling in rats: Implications on oxidative balance, autophagy, and apoptosis. SPORTS MEDICINE AND HEALTH SCIENCE 2024; 6:344-357. [PMID: 39309465 PMCID: PMC11411311 DOI: 10.1016/j.smhs.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 09/25/2024] Open
Abstract
This study aimed to investigate the potential cardioprotective effects of moderate and high-intensity aerobic interval training (MIIT and HIIT) preconditioning. The focus was on histological changes, pro-oxidant-antioxidant balance, autophagy initiation, and apoptosis in myocardial tissue incited by isoproterenol-induced pathological cardiac remodeling (ISO-induced PCR). Male Wistar rats were randomly divided into control (n = 6), ISO (n = 8), MIIT (n = 4), HIIT (n = 4), MIIT + ISO (n = 8), and HIIT + ISO (n = 8) groups. The MIIT and HIIT protocols were administered for 10 weeks, followed by the induction of cardiac remodeling using subcutaneous injection of ISO (100 mg/kg for two consecutive days). Alterations in heart rate (HR), mean arterial pressure (MAP), rate pressure product (RPP), myocardial oxygen consumption (MV ˙ O2), cardiac hypertrophy, histopathological changes, pro-oxidant-antioxidant balance, autophagy biomarkers (Beclin-1, Atg7, p62, LC3 I/II), and apoptotic cell distribution were measured. The findings revealed that the MIIT + ISO and HIIT + ISO groups demonstrated diminished myocardial damage, hemorrhage, immune cell infiltration, edema, necrosis, and apoptosis compared to ISO-induced rats. MIIT and HIIT preconditioning mitigated HR, enhanced MAP, and preserved MV ˙ O2 and RPP. The pro-oxidant-antioxidant balance was sustained in both MIIT + ISO and HIIT + ISO groups, with MIIT primarily inhibiting pro-apoptotic autophagy progression through maintaining pro-oxidant-antioxidant balance, and HIIT promoting pro-survival autophagy. The results demonstrated the beneficial effects of both MIIT and HIIT as AITs preconditioning in ameliorating ISO-induced PCR by improving exercise capacity, hemodynamic parameters, and histopathological changes. Some of these protective effects can be attributed to the modulation of cardiac apoptosis, autophagy, and oxidative stress.
Collapse
Affiliation(s)
- Hakimeh Shahsavarnajand Bonab
- Department of Exercise Physiology and Corrective Exercises, Faculty of Sport Sciences, Urmia University, Urmia, Iran
| | - Javad Tolouei Azar
- Department of Exercise Physiology and Corrective Exercises, Faculty of Sport Sciences, Urmia University, Urmia, Iran
| | - Hamid Soraya
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Akbar Nouri Habashi
- Department of Exercise Physiology and Corrective Exercises, Faculty of Sport Sciences, Urmia University, Urmia, Iran
| |
Collapse
|
20
|
Wu C, Chen X, Yang L, Sun H, Bao S, Li H, Zheng L, Zeng H, Li R, Peng Y. Exercise Mediates Noncoding RNAs in Cardiovascular Diseases: Pathophysiological Roles and Clinical Application. Expert Rev Mol Med 2024; 27:e2. [PMID: 39567354 PMCID: PMC11707833 DOI: 10.1017/erm.2024.25] [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: 09/23/2023] [Revised: 12/09/2023] [Accepted: 05/08/2024] [Indexed: 11/22/2024]
Abstract
Exercise-based cardiac rehabilitation is effective in improving cardiovascular disease risk factor management, cardiopulmonary function, and quality of life. However, the precise mechanisms underlying exercise-induced cardioprotection remain elusive. Recent studies have shed light on the beneficial functions of noncoding RNAs in either exercise or illness models, but only a limited number of noncoding RNAs have been studied in both contexts. Hence, the present study aimed to elucidate the pathophysiological implications and molecular mechanisms underlying the association among exercise, noncoding RNAs, and cardiovascular diseases. Additionally, the present study analysed the most effective and personalized exercise prescription, serving as a valuable reference for guiding the clinical implementation of cardiac rehabilitation in patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Changyong Wu
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xiaocui Chen
- Department of Gastroenterology, Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Lu Yang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Huang Sun
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Suli Bao
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Haojie Li
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Lihui Zheng
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Huiling Zeng
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Ruijie Li
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yunzhu Peng
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
21
|
Wang Z, Luo W, Wang Q, Liu C, Gong Y, Li B, Zeng X, Lin J, Su Z, Li X, Yu Y, Liu Z, Gao L, Liao L. hUCMSCs Regulate Bile Acid Metabolism to Prevent Heart Failure–Induced Intestinal Injury by Inhibiting the Activation of the STAT3/NF‐κB/MAPK Signaling Pathway via TGR5. FOOD FRONTIERS 2024. [DOI: 10.1002/fft2.516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
ABSTRACTThe protective effects of human umbilical cord mesenchymal stem cells (hUCMSCs) on heart failure (HF)‐induced intestinal injury have not been fully understood. Flow cytometry and immunofluorescence analysis revealed that hUCMSCs renewed themselves, grew, and transformed into various cell types. Meanwhile, hUCMSCs safeguarded against intestinal damage, regulated imbalances in the intestinal flora and bile acid metabolism, and enhanced the levels of hyodeoxycholic acid (HDCA) in pigs with HF. HDCA protected against HF‐induced intestinal injury in mice through Takeda G protein–coupled receptor 5 (TGR5). Protein analysis showed that HDCA exerted protective effects on the intestines via the signal transducer and activator of transcription 3 (STAT3)/nuclear factor kappa B (NF‐κB)/mitogen‐activated protein kinase (MAPK) signaling pathway. Mouse experiments revealed that HDCA bound to TGR5 to inhibit MAPK and NF‐κB signaling pathway activation, which relies on the STAT3 signaling pathway. Moreover, hUCMSCs protected against intestinal injury in the pig model of HF by suppressing the activation of the STAT3/NF‐κB/MAPK signaling pathway via TGR5.
Collapse
Affiliation(s)
- Zetian Wang
- Department of Anesthesiology and Pain Management, Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Wei Luo
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine Tongji University Shanghai China
| | - Qing Wang
- Department of Anesthesiology and Pain Management, Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Chunzheng Liu
- Department of Anesthesiology and Pain Management, Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Yanshan Gong
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Baitian Li
- Department of Anesthesiology and Pain Management, Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Xuejiao Zeng
- Department of Anesthesiology and Pain Management, Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Jiaqi Lin
- Department of Anesthesiology and Pain Management, Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Zehua Su
- Department of Anesthesiology and Pain Management, Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Xin Li
- Department of Anesthesiology and Pain Management, Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Yongze Yu
- Department of Anesthesiology and Pain Management, Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Zhongmin Liu
- Department of Anesthesiology and Pain Management, Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Research Institute of Heart Failure, Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital Tongji University Shanghai China
- Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Ling Gao
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Lijun Liao
- Department of Anesthesiology and Pain Management, Shanghai East Hospital Tongji University School of Medicine Shanghai China
| |
Collapse
|
22
|
Silva CNDF, Bessa ADSMD, Costa JMD, Lopes PR, Neves ÂR, Teles Bombardelli MML, Colugnati DB, Pedrino GR, Mendes EP, Santos RASD, Biancardi MF, Santos FCAD, Castro CH. Mas receptor blockade impairs exercise-induced cardiac hypertrophy. Peptides 2024; 181:171296. [PMID: 39265810 DOI: 10.1016/j.peptides.2024.171296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Exercise training leads to physiological cardiac hypertrophy and the protective axis of the renin-angiotensin system composed of angiotensin-converting enzyme 2, angiotensin-(1-7), and Mas receptor seems involved in this process. However, the role of the basal activity of the Mas receptor in exercise-induced physiological cardiac hypertrophy is still unclear. We evaluated the effects of the Mas receptor blockade on the left ventricular structure and function of rats submitted to running training. Rats were assigned to 4 groups: sedentary (S), sedentary + A-779 (Mas receptor antagonist, 120 µg/kg/day, i.p.; SA), trained (60-minute treadmill running sessions, five days a week, 8 weeks; T), and trained + A-779 (TA). Systolic blood pressure was higher in sedentary and trained rats treated with A-779 at the end of the experimental period. The A-779 treatment prevented the left ventricular hypertrophy evoked by physical exercise and increased collagen deposition in sedentary and trained rats. Cardiomyocytes from the SA group presented increased length and thickness of the sarcomeres, elongated mitochondria, glycogen deposits, and enlarged cisterns of the sarcoplasmic reticulum. TA group presented a reduced sarcomere thickness and cytoplasm with a degenerative aspect. These findings show that the basal activity of the Mas receptor is essential for the proper turnover of the extracellular matrix in the myocardium and the maintenance of the sarcomeric structure of cardiomyocytes.
Collapse
Affiliation(s)
| | | | | | - Paulo Ricardo Lopes
- Department of Physiological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Ângela Ribeiro Neves
- Department of Physiological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Guo W, Peng J, Su J, Xia J, Deng W, Li P, Chen Y, Liu G, Wang S, Huang J. The role and underlying mechanisms of irisin in exercise-mediated cardiovascular protection. PeerJ 2024; 12:e18413. [PMID: 39494293 PMCID: PMC11531754 DOI: 10.7717/peerj.18413] [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: 05/07/2024] [Accepted: 10/07/2024] [Indexed: 11/05/2024] Open
Abstract
Irisin, a product of the post-translational processing of fibronectin type III domain-containing protein 5 (FNDC5), is a novel myokine which is upregulated during exercise. This hormone not only promotes the transformation of white adipose tissue into a brown-fat-like phenotype but also enhances energy expenditure and mitigates fat accumulation. Its role is crucial in the management of certain metabolic disorders such as diabetes and heart disease. Of note, the type of exercise performed significantly affects blood irisin levels, indicating the critical role of physical activity in regulating this hormone. This article aims to summarize the current scientific understanding of the role of irisin and the mechanisms through which it mediates cardiovascular protection through exercise. Moreover, this article aims to establish irisin as a potential target for preventing and treating cardiovascular diseases.
Collapse
Affiliation(s)
- Wenhuang Guo
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, China
| | - Jianwei Peng
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, China
| | - Jiarui Su
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, China
| | - Jingbo Xia
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, China
| | - Weiji Deng
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, China
| | - Peilun Li
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, China
| | - Yilin Chen
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, China
| | - Guoqing Liu
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, China
| | - Shen Wang
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, China
| | - Junhao Huang
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, China
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau, China
| |
Collapse
|
24
|
Wu H, Zhou R, Kong H, Yang J, Liu S, Wei X, Li K, Zhang Y. Exercise Attenuates Doxorubicin-Induced Myocardial Injury by Inhibiting TSHR and Regulating Macrophage Polarization Through miR-30d-5p/GALNT7. J Immunol Res 2024; 2024:5562293. [PMID: 39493373 PMCID: PMC11531364 DOI: 10.1155/2024/5562293] [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: 05/06/2024] [Accepted: 09/28/2024] [Indexed: 11/05/2024] Open
Abstract
Objective: Doxorubicin (DOX) is an extensively used chemotherapeutic agent that induces cardiotoxicity. Studies have reported that exercise (EXE) can alleviate DOX-induced cardiotoxicity. Therefore, this study aimed to explore the mechanism by which EXE attenuates DOX-induced myocardial injury. Methods: In this study, cell and animal models of DOX-induced myocardial injury were constructed. The animal model was subjected to EXE intervention. Results: In this study, in vitro experiments revealed that miR-30d-5p negatively regulated polypeptide N-acetylgalactosaminyltransferase 7 (GALNT7) and that GALNT7 negatively regulated the expression of thyroid stimulating hormone receptor (TSHR). miR-30d-5p downregulated the expression of GALNT7, promoted the expression of TSHR, and promoted macrophage M1 polarization, thus aggravating cardiomyocyte injury. In vivo experiments revealed that EXE intervention significantly downregulated miR-30d-5p and TSHR expression, upregulated GALNT7, reduced inflammation, and promoted M2 macrophage polarization, thereby alleviating DOX-induced myocardial injury. In addition, overexpression of miR-30d-5p or knockdown of GALNT7 weakened the intervention effect of EXE, whereas overexpression of GALNT7 or knockdown of TSHR promoted the effect of EXE. Conclusion: EXE can modulate the miR-30d-5p/GALNT7 axis to inhibit the expression of TSHR, thereby regulating the polarization of macrophages to the M2 phenotype and ultimately alleviating DOX-induced myocardial injury, which provides new targets and strategies for the clinical treatment of myocardial injury.
Collapse
Affiliation(s)
- Haiyan Wu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
- Department of Cardiology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Ruoyu Zhou
- Medical School, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
| | - Hanxin Kong
- Department of Cardiology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Jieqiong Yang
- Department of Cardiology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Suijuan Liu
- Department of Cardiology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Xiaolin Wei
- Department of Cardiology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Kunzhi Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
| | - Yunmei Zhang
- Department of Cardiology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| |
Collapse
|
25
|
Rojo-López MI, Bermúdez-López M, Castro E, Farràs C, Torres G, Pamplona R, Lecube A, Valdivieso JM, Fernández E, Julve J, Castelblanco E, Alonso N, Antentas M, Barranco-Altirriba M, Perera-Lluna A, Franch-Nadal J, Granado-Casas M, Mauricio D, on behalf of the ILERVAS project collaborators. Mediterranean Diet Is a Predictor of Progression of Subclinical Atherosclerosis in a Mediterranean Population: The ILERVAS Prospective Cohort Study. Nutrients 2024; 16:3607. [PMID: 39519440 PMCID: PMC11547874 DOI: 10.3390/nu16213607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Atherosclerotic cardiovascular disease remains a major health issue, often developing silently as subclinical atherosclerotic disease (SAD). The Mediterranean diet (MDiet) is known for its cardiovascular benefits, but the combined influence of both MDiet adherence and physical activity (PA) on SAD progression has not been previously documented. Objective: We aimed to investigate how adherence to a healthy lifestyle, defined as MDiet adherence and PA level, influences SAD progression in subjects from the ILERVAS cohort follow-up. Methods: A study on 3097 participants from the ILERVAS prospective cohort was conducted. MDiet adherence was assessed using the MEDAS score, and PA categories were established using the IPAQ, both categorized into low, moderate, and high levels. Two different lifestyle scores integrating the MDiet and PA categories were built. The presence of atherosclerotic plaques was assessed by carotid and femoral ultrasound examination. Demographic, clinical, and biochemical data were also obtained. Multivariable linear, logistic, and Poisson regression models adjusted for potential confounders were used to analyze the association between the lifestyle scores and SAD progression, as well as the MDiet and PA as separate variables and number of territories with plaque. Results: A healthier lifestyle score did not show an effect on SAD progression. However, a higher MEDAS score was associated with a 3% decrease in the number of territories with plaque (IRR 0.97, 95% CI 0.96-0.99, p < 0.001), suggesting a protective effect of the adherence to the MDiet. PA did not show a significant association (IRR 1.00, 95% CI 1.00-1.00, p = 0.269). Older age, hypertension, dyslipidemia, smoking, and lower eGFR were associated with SAD progression, while the female sex was protective (IRR 0.67, 95% CI 0.63-0.72, p < 0.001). Conclusions: The findings of this study show that higher adherence to the MDiet is associated with reduced incidence of SAD, indicating its potential role in cardiovascular prevention strategies. Although a higher lifestyle score or physical activity levels did not show any significant effect, promoting the MDiet, alongside managing traditional cardiovascular risk factors, could be an effective public health intervention to prevent atherosclerosis and reduce the burden of cardiovascular disease.
Collapse
Affiliation(s)
- Marina Idalia Rojo-López
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain; (M.I.R.-L.); (J.J.); (M.A.)
| | - Marcelino Bermúdez-López
- Vascular and Renal Translational Research Group, IRBLleida, Renal Research Network (RedInRen. ISCIII), 25198 Lleida, Spain; (M.B.-L.); (E.C.); (J.M.V.); (E.F.)
- Department of Experimental Medicine, University of Lleida, 25198 Lleida, Spain;
| | - Eva Castro
- Vascular and Renal Translational Research Group, IRBLleida, Renal Research Network (RedInRen. ISCIII), 25198 Lleida, Spain; (M.B.-L.); (E.C.); (J.M.V.); (E.F.)
| | - Cristina Farràs
- Centre d’Atenció Primària Cappont, Gerència Territorial de Lleida, Institut Català de la Salut, 08007 Barcelona, Spain;
- Research Support Unit Lleida, Jordi Gol i Gorina Primary Health Care Research Institute Foundation (IDIAPJGol), 08007 Barcelona, Spain
| | - Gerard Torres
- Department of Respiratory Medicine, Arnau de Vilanova University Hospital, 25198 Lleida, Spain;
- Translational Research Group Respiratory Medicine, IRBLleida, University of Lleida, 25198 Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Instituto de Salud Carlos III (ICSIII), 28029 Madrid, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida, 25198 Lleida, Spain;
| | - Albert Lecube
- Department of Endocrinology and Nutrition, Arnau de Vilanova University Hospital, 25198 Lleida, Spain;
- Obesity and Metabolism Research Group (ODIM), IRBLleida, University of Lleida, 25198 Lleida, Spain
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 08041 Barcelona, Spain; (N.A.); (J.F.-N.)
| | - José Manuel Valdivieso
- Vascular and Renal Translational Research Group, IRBLleida, Renal Research Network (RedInRen. ISCIII), 25198 Lleida, Spain; (M.B.-L.); (E.C.); (J.M.V.); (E.F.)
| | - Elvira Fernández
- Vascular and Renal Translational Research Group, IRBLleida, Renal Research Network (RedInRen. ISCIII), 25198 Lleida, Spain; (M.B.-L.); (E.C.); (J.M.V.); (E.F.)
- Department of Experimental Medicine, University of Lleida, 25198 Lleida, Spain;
| | - Josep Julve
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain; (M.I.R.-L.); (J.J.); (M.A.)
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 08041 Barcelona, Spain; (N.A.); (J.F.-N.)
| | - Esmeralda Castelblanco
- Department of Internal Medicine, Endocrinology, Metabolism and Lipid Research Division, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Nuria Alonso
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 08041 Barcelona, Spain; (N.A.); (J.F.-N.)
- Department of Endocrinology and Nutrition, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
| | - Maria Antentas
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain; (M.I.R.-L.); (J.J.); (M.A.)
| | - Maria Barranco-Altirriba
- Departament of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- Departament d’Enginyeria de Sistemes, Automàtica i Informàtica Industrial, Universitat Politècnica de Catalunya, B2SLab, 08034 Barcelona, Spain;
- Networking Biomedical Research Centre in the Subject Area of Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029 Madrid, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, 08950 Barcelona, Spain
| | - Alexandre Perera-Lluna
- Departament d’Enginyeria de Sistemes, Automàtica i Informàtica Industrial, Universitat Politècnica de Catalunya, B2SLab, 08034 Barcelona, Spain;
- Networking Biomedical Research Centre in the Subject Area of Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029 Madrid, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, 08950 Barcelona, Spain
| | - Josep Franch-Nadal
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 08041 Barcelona, Spain; (N.A.); (J.F.-N.)
- DAP-Cat Group, Unitat de Suport a la Recerca Barcelona, Fundació Institut Universitari per a la Recerca a l’Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), 08007 Barcelona, Spain
| | - Minerva Granado-Casas
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 08041 Barcelona, Spain; (N.A.); (J.F.-N.)
- Department of Endocrinology and Nutrition, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
- Department of Nursing and Physiotherapy, University of Lleida, 25198 Lleida, Spain
- Research Group of Health Care (GreCS), IRBLleida, 25198 Lleida, Spain
| | - Didac Mauricio
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain; (M.I.R.-L.); (J.J.); (M.A.)
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 08041 Barcelona, Spain; (N.A.); (J.F.-N.)
- Departament of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- Faculty of Medicine, University of Vic-Central University of Catalonia (UVIC-UCC), 08500 Vic, Spain
| | | |
Collapse
|
26
|
Maleki F, Mehrabani J. Right ventricular remodeling induced by prolonged excessive endurance exercise is mediated by upregulating Wnt/β-catenin signaling in rats. Int J Cardiol 2024; 413:132316. [PMID: 38977222 DOI: 10.1016/j.ijcard.2024.132316] [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: 02/07/2024] [Revised: 05/26/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND The aim of this study was to develop an animal model to investigate whether prolonged intensive endurance exercise induces RV remodeling, taking into account the involvement of Wnt/β-catenin signaling. METHODS Four-week-old male Wistar rats (100 to 125 g) were assigned to four groups (n = 8/group): 1) sixteen weeks of intensive (36 m/min) exercise (INT), 2) twelve weeks of the intensive exercise followed by four weeks of moderate intensity (18 m/min) exercise (INT + MOD), 3) twelve weeks of the intensive exercise followed by four weeks of detraining (INT + DT), and 4) sedentary rats (SED). The exercise protocols were performed five days a week for one h/day. Echocardiography, real-time PCR, western blotting, and histological staining were performed at the end of week sixteen. RESULTS INT rats developed concentric hypertrophy without diastolic dysfunction compared to SED (p = 0.006) and INT + DT (p = 0.035). Wnt1, β-catenin and CyclinD1 proteins in the training groups were significantly higher than SED rats (p < 0.001). Interestingly, INT rats had higher protein levels than INT + DT and INT + MOD (p < 0.001), with higher gene expression compared to SED rats (p < 0.05). There was also a significant increase in collagen deposition in INT rats compared to SED (p = 0.046) and INT + DT (p = 0.034). Furthermore, INT + MOD and INT + DT rats did not show any adverse structural, functional, or histological changes. CONCLUSIONS Long-term intensive endurance training seems to be associated with increased collagen deposition and wall thickness in the RV through Wnt/β-catenin signaling (which is concentration dependent), without changes in diastolic function. CLINICAL PERSPECTIVE Over the past decades, there has been an ongoing debate about whether the structural and functional adaptations of the cardiovascular system in trained endurance athletes are benign physiological responses to training or potentially pathological changes related to disease. While the adaptations of the left heart are well-documented, the remodeling of the right heart remains a subject of discussion. To gain insights into the ability of sustained high-intensity exercise to cause adverse right ventricular (RV) remodeling, we conducted an experimental study in which male rats were trained to run vigorously for 1 h daily over a 16-week period and compared them to a parallel group of sedentary control rats. Our findings revealed that intense long-term exercise induced morphological changes along with fibrosis affecting the RV. These fibrotic changes were a result of the 16-week vigorous exercise training regimen. If these results are confirmed in humans, they suggest that prolonged high-intensity endurance exercise training may lead to adverse cardiac remodeling. Our findings have important potential implications for the assessment of cardiac remodeling in individuals engaged in high-level exercise training.
Collapse
Affiliation(s)
- Farzaneh Maleki
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Guilan, Rasht 4199843653, Iran.
| | - Javad Mehrabani
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Guilan, Rasht 4199843653, Iran.
| |
Collapse
|
27
|
Satoh T, Yaoita N, Higuchi S, Nochioka K, Yamamoto S, Sato H, Tatebe S, Yamada K, Yamada Y, Komaru K, Chiba N, Sarashina Y, Mori R, Nakada M, Hayashi H, Suzuki H, Takahama H, Ota H, Yasuda S. Impact of Sodium-Glucose Co-Transporter-2 Inhibitors on Exercise-Induced Pulmonary Hypertension. Pulm Circ 2024; 14:e70026. [PMID: 39678732 PMCID: PMC11646329 DOI: 10.1002/pul2.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 12/17/2024] Open
Abstract
Patients with borderline pulmonary hypertension (PH) often experience shortness of breath or exacerbation of PH during exercise, known as exercise-induced PH. However, the pathogenesis of exercise-induced post-capillary PH (post-EIPH) and its treatment strategies remain unclear. Recent guidelines and consensus documents have highlighted the benefits of sodium-glucose cotransporter-2 (SGLT2) inhibitors in heart failure and chronic kidney disease (CKD). This study aimed to investigate the effects of SGLT2 inhibitors in patients with post-EIPH and CKD. This single-center prospective cohort study enroled 10 patients with CKD (age, 68 years; female, 60%) who exhibited post-EIPH between 1 July 2022 and 31 December 2023. Post-EIPH was defined as a pulmonary capillary wedge pressure (PCWP)/cardiac output (CO) slope > 2 and peak PCWP during exercise ≥ 25 mmHg measured by catheterization. The patients received SGLT2 inhibitor treatment for 6 months. At rest, patients with post-EIPH had borderline-PH (21.5 ± 1.8 mmHg), with preserved left and right ventricular function. SGLT2 inhibitors treatment significantly reduced the PCWP/CO slope during exercise (3.9 ± 1.2 vs. 2.4 ± 1.2 mmHg/L/min, p = 0.013) and improved the 6-min walking distance (489.9 ± 80.2 vs. 568.3 ± 91.9 m, p = 0.014). Magnetic resonance imaging revealed a lower left ventricular global longitudinal strain in patients with post-EIPH, which was increased by SGLT2 inhibitor treatment (-13.8 ± 2.0 vs. -17.3 ± 2.0%, p = 0.003). SGLT2 treatment inhibitors mitigated post-EIPH hemodynamic abnormalities and exercise intolerance, suggesting their potential as its therapeutic option.
Collapse
Affiliation(s)
- Taijyu Satoh
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Nobuhiro Yaoita
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Satoshi Higuchi
- Department of Diagnostic RadiologyTohoku University Graduate School of MedicineSendaiJapan
| | - Kotaro Nochioka
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Saori Yamamoto
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Haruka Sato
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Shunsuke Tatebe
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Kaito Yamada
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Yusuke Yamada
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Kohei Komaru
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Naoki Chiba
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Yuki Sarashina
- Tohoku University Graduate School of MedicineSendaiJapan
| | - Ryuichi Mori
- Departments of Radiology and Medical TechnologyTohoku University HospitalSendaiJapan
| | - Mitsuru Nakada
- Departments of Radiology and Medical TechnologyTohoku University HospitalSendaiJapan
| | - Hideka Hayashi
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Hideaki Suzuki
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Hiroyuki Takahama
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Hideki Ota
- Department of Diagnostic RadiologyTohoku University Graduate School of MedicineSendaiJapan
| | - Satoshi Yasuda
- Department of Cardiovascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| |
Collapse
|
28
|
Park W, Park HY, Kim SW. Effects of 12 Weeks of Combined Exercise Training in Normobaric Hypoxia on Arterial Stiffness, Inflammatory Biomarkers, and Red Blood Cell Hemorheological Function in Obese Older Women. Healthcare (Basel) 2024; 12:1887. [PMID: 39337228 PMCID: PMC11431341 DOI: 10.3390/healthcare12181887] [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: 08/08/2024] [Revised: 09/07/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND/OBJECTIVES The present study examined the effect of 12-week combined exercise training in normobaric hypoxia on arterial stiffness, inflammatory biomarkers, and red blood cell (RBC) hemorheological function in 24 obese older women (mean age: 67.96 ± 0.96 years). METHODS Subjects were randomly divided into two groups (normoxia (NMX; n = 12) and hypoxia (HPX; n = 12)). Both groups performed aerobic and resistance exercise training programs three times per week for 12 weeks, and the HPX group performed exercise programs in hypoxic environment chambers during the intervention period. Body composition was estimated using bioelectrical impedance analysis equipment. Arterial stiffness was measured using an automatic waveform analyzer. Biomarkers of inflammation and oxygen transport (tumor necrosis factor alpha, interleukin 6 (IL-6), erythropoietin (EPO), and vascular endothelial growth factor (VEGF)), and RBC hemorheological parameters (RBC deformability and aggregation) were analyzed. RESULTS All variables showed significantly more beneficial changes in the HPX group than in the NMX group during the intervention. The combined exercise training in normobaric hypoxia significantly reduced blood pressure (systolic blood pressure: p < 0.001, diastolic blood pressure: p < 0.001, mean arterial pressure: p < 0.001, pulse pressure: p < 0.05) and brachial-ankle pulse wave velocity (p < 0.001). IL-6 was significantly lower in the HPX group than in the NMX group post-test (p < 0.001). Also, EPO (p < 0.01) and VEGF (p < 0.01) were significantly higher in the HPX group than in the NMX group post-test. Both groups showed significantly improved RBC deformability (RBC EI_3Pa) (p < 0.001) and aggregation (RBC AI_3Pa) (p < 0.001). CONCLUSIONS The present study suggests that combined exercise training in normobaric hypoxia can improve inflammatory biomarkers and RBC hemorheological parameters in obese older women and may help prevent cardiovascular diseases.
Collapse
Affiliation(s)
- Wonil Park
- Department of Sports Science, Korea Institute of Sports Science, 424 Olympic-ro, Songpa-gu, Seoul 05540, Republic of Korea;
| | - Hun-Young Park
- Department of Sports Medicine and Science, Graduate School, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea;
- Physical Activity and Performance Institute, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Sung-Woo Kim
- Department of Sports Medicine and Science, Graduate School, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea;
- Physical Activity and Performance Institute, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| |
Collapse
|
29
|
Yoo J, Hwang J, Choi J, Ramalingam M, Jeong H, Jang S, Jeong HS, Kim D. The effects of resistance training on cardiovascular factors and anti-inflammation in diabetic rats. Heliyon 2024; 10:e37081. [PMID: 39295999 PMCID: PMC11407942 DOI: 10.1016/j.heliyon.2024.e37081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/21/2024] Open
Abstract
Diabetes induces a range of macrovascular and microvascular changes, which lead to significant clinical complications. Although many studies have tried to solve the diabetic problem using drugs, it remains unclear. In this study, we investigated whether resistance exercise affects cardiovascular factors and inflammatory markers in diabetes. The study subjected Otsuka Long-Evans Tokushima Fatty (OLETF) rats, which have genetically induced diabetes mellitus, to a resistance exercise program for 12 weeks and assessed the levels of cardiovascular factors and inflammatory markers using western blotting analysis, ELISA, and immunohistochemistry. During the training period, OLETF + exercise (EX) group exhibited lower body weight and reduced glucose levels when compared with OLETF group. Western blotting analysis, ELISA, and immunohistochemistry revealed that the levels of PAI-1, VACM-1, ICAM-1, E-selectin, TGF-β, CRP, IL-6, and TNF-α were decreased in OLETF + EX group when compared with the OLETF group. Moreover, the anti-inflammatory markers, IL-4 and IL-10, were highly expressed after exercise. Therefore, these results indicate that exercise may influence the regulation of cardiovascular factors and inflammatory markers, as well as help patients with metabolic syndromes regulate inflammation and cardiovascular function.
Collapse
Affiliation(s)
- Jin Yoo
- Department of Physical Education, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jinsu Hwang
- Department of Physiology, Chonnam National University Medical School, Hwasun-gun, Jeollanamdo, 58128, Republic of Korea
| | - Jiyun Choi
- Department of Physiology, Chonnam National University Medical School, Hwasun-gun, Jeollanamdo, 58128, Republic of Korea
| | - Mahesh Ramalingam
- Department of Physiology, Chonnam National University Medical School, Hwasun-gun, Jeollanamdo, 58128, Republic of Korea
| | - Haewon Jeong
- StemCell Bio Incorporated, Hwasun-gun, Jeollanamdo, 58128, Republic of Korea
| | - Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Hwasun-gun, Jeollanamdo, 58128, Republic of Korea
- StemCell Bio Incorporated, Hwasun-gun, Jeollanamdo, 58128, Republic of Korea
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Hwasun-gun, Jeollanamdo, 58128, Republic of Korea
- StemCell Bio Incorporated, Hwasun-gun, Jeollanamdo, 58128, Republic of Korea
| | - Daeyeol Kim
- Department of Physical Education, Chonnam National University, Gwangju, 61186, Republic of Korea
| |
Collapse
|
30
|
Bei Y, Zhu Y, Zhou J, Ai S, Yao J, Yin M, Hu M, Qi W, Spanos M, Li L, Wei M, Huang Z, Gao J, Liu C, van der Kraak PH, Li G, Lei Z, Sluijter JPG, Xiao J. Inhibition of Hmbox1 Promotes Cardiomyocyte Survival and Glucose Metabolism Through Gck Activation in Ischemia/Reperfusion Injury. Circulation 2024; 150:848-866. [PMID: 38708602 DOI: 10.1161/circulationaha.123.067592] [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: 10/14/2023] [Accepted: 04/11/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND Exercise-induced physiological cardiac growth regulators may protect the heart from ischemia/reperfusion (I/R) injury. Homeobox-containing 1 (Hmbox1), a homeobox family member, has been identified as a putative transcriptional repressor and is downregulated in the exercised heart. However, its roles in exercise-induced physiological cardiac growth and its potential protective effects against cardiac I/R injury remain largely unexplored. METHODS We studied the function of Hmbox1 in exercise-induced physiological cardiac growth in mice after 4 weeks of swimming exercise. Hmbox1 expression was then evaluated in human heart samples from deceased patients with myocardial infarction and in the animal cardiac I/R injury model. Its role in cardiac I/R injury was examined in mice with adeno-associated virus 9 (AAV9) vector-mediated Hmbox1 knockdown and in those with cardiac myocyte-specific Hmbox1 ablation. We performed RNA sequencing, promoter prediction, and binding assays and identified glucokinase (Gck) as a downstream effector of Hmbox1. The effects of Hmbox1 together with Gck were examined in cardiomyocytes to evaluate their cell size, proliferation, apoptosis, mitochondrial respiration, and glycolysis. The function of upstream regulator of Hmbox1, ETS1, was investigated through ETS1 overexpression in cardiac I/R mice in vivo. RESULTS We demonstrated that Hmbox1 downregulation was required for exercise-induced physiological cardiac growth. Inhibition of Hmbox1 increased cardiomyocyte size in isolated neonatal rat cardiomyocytes and human embryonic stem cell-derived cardiomyocytes but did not affect cardiomyocyte proliferation. Under pathological conditions, Hmbox1 was upregulated in both human and animal postinfarct cardiac tissues. Furthermore, both cardiac myocyte-specific Hmbox1 knockout and AAV9-mediated Hmbox1 knockdown protected against cardiac I/R injury and heart failure. Therapeutic effects were observed when sh-Hmbox1 AAV9 was administered after I/R injury. Inhibition of Hmbox1 activated the Akt/mTOR/P70S6K pathway and transcriptionally upregulated Gck, leading to reduced apoptosis and improved mitochondrial respiration and glycolysis in cardiomyocytes. ETS1 functioned as an upstream negative regulator of Hmbox1 transcription, and its overexpression was protective against cardiac I/R injury. CONCLUSIONS Our studies unravel a new role for the transcriptional repressor Hmbox1 in exercise-induced physiological cardiac growth. They also highlight the therapeutic potential of targeting Hmbox1 to improve myocardial survival and glucose metabolism after I/R injury.
Collapse
Affiliation(s)
- Yihua Bei
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Yujiao Zhu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Jingwen Zhou
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Songwei Ai
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Jianhua Yao
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (J.Y.)
| | - Mingming Yin
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Meiyu Hu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Weitong Qi
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Michail Spanos
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston (M.S., G.L.)
| | - Lin Li
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Meng Wei
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Zhenzhen Huang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Juan Gao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Chang Liu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Petra H van der Kraak
- Department of Pathology (P.H.v.d.K.), University Medical Center Utrecht, University Utrecht, The Netherlands
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston (M.S., G.L.)
| | - Zhiyong Lei
- Department of Cardiology, Laboratory of Experimental Cardiology (Z.L., J.P.G.S.), University Medical Center Utrecht, University Utrecht, The Netherlands
- Division Laboratory, Central Diagnosis Laboratory Research (Z.L.), University Medical Center Utrecht, University Utrecht, The Netherlands
| | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology (Z.L., J.P.G.S.), University Medical Center Utrecht, University Utrecht, The Netherlands
- Utrecht Regenerative Medicine Center (J.P.G.S.), University Medical Center Utrecht, University Utrecht, The Netherlands
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| |
Collapse
|
31
|
Wan K, Jin Y, Fan R, Xu Q, Li X, Yan H, Wang R. Exploring molecular mechanisms of exercise on metabolic syndrome: a bibliometric and visualization study using CiteSpace. Front Endocrinol (Lausanne) 2024; 15:1408466. [PMID: 39290329 PMCID: PMC11405195 DOI: 10.3389/fendo.2024.1408466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Objective To investigate the molecular mechanisms through which exercise influences metabolic syndrome (MS) and identify key research trends and collaborative networks using bibliometric and visualization techniques. Methods We conducted a systematic literature search using the Web of Science Core Collection for articles published from 2014 to 2023. Using CiteSpace, we performed a bibliometric analysis of 562 eligible papers, generating visual knowledge maps to identify prevailing patterns, popular subjects, and emerging trends in the literature. Results The study reveals that exercise mitigates MS by reversing high-fat diet-induced abdominal obesity, reducing lipid accumulation and inflammation, enhancing insulin sensitivity, and improving cardiovascular function. Key molecular pathways include PPAR-γ/CPT-1/MCAD signaling, AMPK activation, and nitric oxide production. The USA leads in research output, with significant contributions from American institutions. Collaboration among researchers is limited, highlighting the need for more extensive and high-quality research initiatives. Conclusions Regular, moderate-to-high-intensity exercise is crucial for managing MS. Exercise activates beneficial molecular pathways, improving metabolic health and cardiovascular function. Future research should focus on expanding collaborations and exploring novel molecular targets to enhance the therapeutic potential of exercise in metabolic syndrome management.
Collapse
Affiliation(s)
- Kang Wan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- Physical Education College, Henan Sport University, Zhengzhou, China
| | - Yue Jin
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Ruobing Fan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Qizi Xu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Xiaoshi Li
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Hongmei Yan
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Endocrinology and Metabolism, Wusong Branch of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
32
|
Kunutsor SK, Kurl S, Laukkanen JA. Cardiorespiratory fitness, atrial fibrillation and stroke: a review of the evidence in 2024. Expert Rev Cardiovasc Ther 2024; 22:493-508. [PMID: 39329169 DOI: 10.1080/14779072.2024.2409440] [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: 04/19/2024] [Revised: 09/15/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
INTRODUCTION The body of evidence linking cardiorespiratory fitness (CRF) levels with the risk of atrial fibrillation (AF) and stroke - two interconnected cardiovascular conditions - is not entirely consistent. Furthermore, specific CRF thresholds beyond which the risk of AF or stroke might not decrease are not well defined. AREAS COVERED This review summarizes research evidence on the role of CRF in the development of AF and stroke including dose-response relationships in general population participants, explores the biological mechanisms through which CRF may exert its effects, assesses the potential implications for clinical care and population health, identifies gaps in the current evidence, and suggest directions for future research. MEDLINE and Embase were searched from inception until July 2024 to identify observational longitudinal and interventional studies as well as systematic reviews and meta-analyses related to these study designs. EXPERT OPINION In the general population, increasing levels of CRF, achieved through consistent physical activity, can significantly reduce the likelihood of developing AF and stroke. The findings also advocate for a tailored approach to exercise prescriptions, acknowledging the plateau in benefits for AF risk beyond certain CRF levels, while advocating for higher intensity or prolonged activity to further reduce stroke risk.
Collapse
Affiliation(s)
- Setor K Kunutsor
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester, UK
- Section of Cardiology, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Saint Boniface Hospital, Winnipeg, Manitoba, Canada
| | - Sudhir Kurl
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Brain Research Unit, Department of Neurology, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jari A Laukkanen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Wellbeing Services County of Central Finland, Jyväskylä, Finland
| |
Collapse
|
33
|
Sahu Y, Jamadade P, Ch Maharana K, Singh S. Role of mitochondrial homeostasis in D-galactose-induced cardiovascular ageing from bench to bedside. Mitochondrion 2024; 78:101923. [PMID: 38925493 DOI: 10.1016/j.mito.2024.101923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/11/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024]
Abstract
Ageing is an inevitable phenomenon which affects the cellular to the organism level in the progression of the time. Oxidative stress and inflammation are now widely regarded as the key processes involved in the aging process, which may then cause significant harm to mitochondrial DNA, leading to apoptosis. Normal circulatory function is a significant predictor of disease-free life expectancy. Indeed, disorders affecting the cardiovascular system, which are becoming more common, are the primary cause of worldwide morbidity, disability, and mortality. Cardiovascular aging may precede or possibly underpin overall, age-related health decline. Numerous studies have foundmitochondrial mechanistc approachplays a vital role in the in the onset and development of aging. The D-galactose (D-gal)-induced aging model is well recognized and commonly used in the aging study. In this review we redeposit the association of the previous and current studies on mitochondrial homeostasis and its underlying mechanisms in D-galactose cardiovascular ageing. Further we focus the novel and the treatment strategies to combat the major complication leading to the cardiovascular ageing.
Collapse
Affiliation(s)
- Yogita Sahu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hajipur, Vaishali, Bihar, India
| | - Pratiksha Jamadade
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hajipur, Vaishali, Bihar, India
| | - Krushna Ch Maharana
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hajipur, Vaishali, Bihar, India
| | - Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hajipur, Vaishali, Bihar, India.
| |
Collapse
|
34
|
Paulucio D, Ramirez-Sanchez C, Velasque R, Xavier R, Monnerat G, Dill A, Silveira J, Andrade GM, Meirelles F, Dornelas-Ribeiro M, Kirchner B, Pfaffl MW, Pompeu F, Santos CGM. Circulating miRNA Signaling for Fatty Acid Metabolism in Response to a Maximum Endurance Test in Elite Long-Distance Runners. Genes (Basel) 2024; 15:1088. [PMID: 39202447 PMCID: PMC11353293 DOI: 10.3390/genes15081088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
Maximal oxygen uptake (VO2max) is a determining indicator for cardiorespiratory capacity in endurance athletes, and epigenetics is crucial in its levels and variability. This initial study examined a broad plasma miRNA profile of twenty-three trained elite endurance athletes with similar training volumes but different VO2max in response to an acute maximal graded endurance test. Six were clustered as higher/lower levels based on their VO2max (75.4 ± 0.9 and 60.1 ± 5.0 mL.kg-1.min-1). Plasma was obtained from athletes before and after the test and 15 ng of total RNA was extracted and detected using an SYBR-based 1113 miRNA RT-qPCR panel. A total of 51 miRNAs were differentially expressed among group comparisons. Relative amounts of miRNA showed a clustering behavior among groups regarding distinct performance/time points. Significantly expressed miRNAs were used to perform functional bioinformatic analysis (DIANA tools). Fatty acid metabolism pathways were strongly targeted for the significantly different miRNAs in all performance groups and time points (p < 0.001). Although this pathway does not solely determine endurance performance, their significant contribution is certainly achieved through the involvement of miRNAs. A highly genetically dependent gold standard variable for performance evaluation in a homogeneous group of elite athletes allowed genetic/epigenetic aspects related to fatty acid pathways to emerge.
Collapse
Affiliation(s)
- Dailson Paulucio
- Biometrics Laboratory (LADEBIO), Federal University of Rio de Janeiro, Rio de Janeiro 21941-599, Brazil; (D.P.)
| | - Carlos Ramirez-Sanchez
- Biometrics Laboratory (LADEBIO), Federal University of Rio de Janeiro, Rio de Janeiro 21941-599, Brazil; (D.P.)
| | - Rodolfo Velasque
- Biometrics Laboratory (LADEBIO), Federal University of Rio de Janeiro, Rio de Janeiro 21941-599, Brazil; (D.P.)
| | - Raphael Xavier
- Biometrics Laboratory (LADEBIO), Federal University of Rio de Janeiro, Rio de Janeiro 21941-599, Brazil; (D.P.)
| | - Gustavo Monnerat
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-170, Brazil
| | - Adrieli Dill
- Brazilian Army Institute of Biology, Research, Teaching and Research Division, Rio de Janeiro 20911-270, Brazil
| | - Juliano Silveira
- Faculty of Animal Sciences and Food Engineering, Universidade de São Paulo, Pirassununga 13635-900, Brazil
| | - Gabriella M. Andrade
- Faculty of Animal Sciences and Food Engineering, Universidade de São Paulo, Pirassununga 13635-900, Brazil
| | - Flavio Meirelles
- Faculty of Animal Sciences and Food Engineering, Universidade de São Paulo, Pirassununga 13635-900, Brazil
| | - Marcos Dornelas-Ribeiro
- Brazilian Army Institute of Biology, Research, Teaching and Research Division, Rio de Janeiro 20911-270, Brazil
| | - Benedikt Kirchner
- Department of Animal Physiology and Immunology, School of Life Sciences, Technical University of Munich, Liesel-Beckmann-Straße 1, 85354 Freising, Germany
| | - Michael W. Pfaffl
- Department of Animal Physiology and Immunology, School of Life Sciences, Technical University of Munich, Liesel-Beckmann-Straße 1, 85354 Freising, Germany
| | - Fernando Pompeu
- Biometrics Laboratory (LADEBIO), Federal University of Rio de Janeiro, Rio de Janeiro 21941-599, Brazil; (D.P.)
| | - Caleb G. M. Santos
- Brazilian Army Institute of Biology, Research, Teaching and Research Division, Rio de Janeiro 20911-270, Brazil
- Department of Animal Physiology and Immunology, School of Life Sciences, Technical University of Munich, Liesel-Beckmann-Straße 1, 85354 Freising, Germany
| |
Collapse
|
35
|
Zhang CY, Li KL, Zhao XX, Zhang ZY, Yin AW, Wang RX. The Role and Underlying Mechanisms of Exercise in Heart Failure. Rev Cardiovasc Med 2024; 25:285. [PMID: 39228484 PMCID: PMC11366989 DOI: 10.31083/j.rcm2508285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/24/2024] [Accepted: 02/29/2024] [Indexed: 09/05/2024] Open
Abstract
Heart failure is a prevalent and life-threatening syndrome characterized by structural and/or functional abnormalities of the heart. As a global burden with high rates of morbidity and mortality, there is growing recognition of the beneficial effects of exercise on physical fitness and cardiovascular health. A substantial body of evidence supports the notion that exercise can play a protective role in the development and progression of heart failure and improve cardiac function through various mechanisms, such as attenuating cardiac fibrosis, reducing inflammation, and regulating mitochondrial metabolism. Further investigation into the role and underlying mechanisms of exercise in heart failure may uncover novel therapeutic targets for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Chong-Yi Zhang
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, 214023 Wuxi, Jiangsu, China
| | - Ku-Lin Li
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, 214023 Wuxi, Jiangsu, China
| | - Xiao-Xi Zhao
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, 214023 Wuxi, Jiangsu, China
| | - Zhen-Ye Zhang
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, 214023 Wuxi, Jiangsu, China
| | - An-Wen Yin
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, 214023 Wuxi, Jiangsu, China
| | - Ru-Xing Wang
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, 214023 Wuxi, Jiangsu, China
| |
Collapse
|
36
|
Shao HH, Yin RX. Pathogenic mechanisms of cardiovascular damage in COVID-19. Mol Med 2024; 30:92. [PMID: 38898389 PMCID: PMC11186295 DOI: 10.1186/s10020-024-00855-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 06/07/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND COVID-19 is a new infectious disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS CoV-2). Since the outbreak in December 2019, it has caused an unprecedented world pandemic, leading to a global human health crisis. Although SARS CoV-2 mainly affects the lungs, causing interstitial pneumonia and severe acute respiratory distress syndrome, a number of patients often have extensive clinical manifestations, such as gastrointestinal symptoms, cardiovascular damage and renal dysfunction. PURPOSE This review article discusses the pathogenic mechanisms of cardiovascular damage in COVID-19 patients and provides some useful suggestions for future clinical diagnosis, treatment and prevention. METHODS An English-language literature search was conducted in PubMed and Web of Science databases up to 12th April, 2024 for the terms "COVID-19", "SARS CoV-2", "cardiovascular damage", "myocardial injury", "myocarditis", "hypertension", "arrhythmia", "heart failure" and "coronary heart disease", especially update articles in 2023 and 2024. Salient medical literatures regarding the cardiovascular damage of COVID-19 were selected, extracted and synthesized. RESULTS The most common cardiovascular damage was myocarditis and pericarditis, hypertension, arrhythmia, myocardial injury and heart failure, coronary heart disease, stress cardiomyopathy, ischemic stroke, blood coagulation abnormalities, and dyslipidemia. Two important pathogenic mechanisms of the cardiovascular damage may be direct viral cytotoxicity as well as indirect hyperimmune responses of the body to SARS CoV-2 infection. CONCLUSIONS Cardiovascular damage in COVID-19 patients is common and portends a worse prognosis. Although the underlying pathophysiological mechanisms of cardiovascular damage related to COVID-19 are not completely clear, two important pathogenic mechanisms of cardiovascular damage may be the direct damage of the SARSCoV-2 infection and the indirect hyperimmune responses.
Collapse
Affiliation(s)
- Hong-Hua Shao
- Department of Infectious Diseases, HIV/AIDS Clinical Treatment Center of Guangxi (Nanning), The Fourth People's Hospital of Nanning, No. 1 Erli, Changgang Road, Nanning, Guangxi, 530023, People's Republic of China
| | - Rui-Xing Yin
- Department of Infectious Diseases, HIV/AIDS Clinical Treatment Center of Guangxi (Nanning), The Fourth People's Hospital of Nanning, No. 1 Erli, Changgang Road, Nanning, Guangxi, 530023, People's Republic of China.
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China.
| |
Collapse
|
37
|
Wang L, Wan W, Zhang S, Keswani T, Li G, Xiao J. RNA-mediated epigenetic regulation in exercised heart: Mechanisms and opportunities for intervention. Mol Aspects Med 2024; 97:101274. [PMID: 38653129 DOI: 10.1016/j.mam.2024.101274] [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: 02/02/2024] [Revised: 03/21/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024]
Abstract
Physical exercise has been widely acknowledged as a beneficial lifestyle alteration and a potent non-pharmacological treatment for heart disease. Extensive investigations have revealed the beneficial effects of exercise on the heart and the underlying mechanisms involved. Exercise is considered one of the key factors that can lead to epigenetic alterations. The increasing number of identified molecules in the exercised heart has led to many studies in recent years that have explored the cellular function of ncRNAs and RNA modifications in the heart. Investigating the regulatory role of RNA-mediated epigenetic regulation in exercised hearts will contribute to the development of therapeutic strategies for the management of heart diseases. This review aims to summarize the positive impact of exercise on cardiac health. We will first provide an overview of the mechanisms through which exercise offers protection to the heart. Subsequently, we will delve into the current understanding of ncRNAs, specifically miRNAs, lncRNAs, and circRNAs, as well as RNA modification, focusing on RNA m6A and RNA A-to-I editing, and how they contribute to exercise-induced benefits for the heart. Lastly, we will explore the emerging therapeutic strategies that utilize exercise-mediated RNA epigenetic regulation in the treatment of heart diseases, while also addressing the challenges faced in this field.
Collapse
Affiliation(s)
- Lijun Wang
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China; Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Wensi Wan
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China; Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Shuang Zhang
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China; Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Tarun Keswani
- Center for Immunological and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Junjie Xiao
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China; Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
38
|
Walzik D, Wences Chirino TY, Zimmer P, Joisten N. Molecular insights of exercise therapy in disease prevention and treatment. Signal Transduct Target Ther 2024; 9:138. [PMID: 38806473 PMCID: PMC11133400 DOI: 10.1038/s41392-024-01841-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/30/2024] Open
Abstract
Despite substantial evidence emphasizing the pleiotropic benefits of exercise for the prevention and treatment of various diseases, the underlying biological mechanisms have not been fully elucidated. Several exercise benefits have been attributed to signaling molecules that are released in response to exercise by different tissues such as skeletal muscle, cardiac muscle, adipose, and liver tissue. These signaling molecules, which are collectively termed exerkines, form a heterogenous group of bioactive substances, mediating inter-organ crosstalk as well as structural and functional tissue adaption. Numerous scientific endeavors have focused on identifying and characterizing new biological mediators with such properties. Additionally, some investigations have focused on the molecular targets of exerkines and the cellular signaling cascades that trigger adaption processes. A detailed understanding of the tissue-specific downstream effects of exerkines is crucial to harness the health-related benefits mediated by exercise and improve targeted exercise programs in health and disease. Herein, we review the current in vivo evidence on exerkine-induced signal transduction across multiple target tissues and highlight the preventive and therapeutic value of exerkine signaling in various diseases. By emphasizing different aspects of exerkine research, we provide a comprehensive overview of (i) the molecular underpinnings of exerkine secretion, (ii) the receptor-dependent and receptor-independent signaling cascades mediating tissue adaption, and (iii) the clinical implications of these mechanisms in disease prevention and treatment.
Collapse
Affiliation(s)
- David Walzik
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany
| | - Tiffany Y Wences Chirino
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany
| | - Philipp Zimmer
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany.
| | - Niklas Joisten
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany.
- Division of Exercise and Movement Science, Institute for Sport Science, University of Göttingen, 37075, Göttingen, Lower Saxony, Germany.
| |
Collapse
|
39
|
Wang CZ, Guo HZ, Leng JZ, Liang ZD, Wang JT, Luo LJ, Wang SQ, Yuan Y. Exercise preconditioning inhibits doxorubicin-induced cardiotoxicity via YAP/STAT3 signaling. Heliyon 2024; 10:e27035. [PMID: 38515673 PMCID: PMC10955211 DOI: 10.1016/j.heliyon.2024.e27035] [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: 06/14/2023] [Revised: 02/22/2024] [Accepted: 02/22/2024] [Indexed: 03/23/2024] Open
Abstract
Doxorubicin (DOX) possesses strong anti-tumor effects but is limited by its irreversible cardiac toxicity. The relationship between exercise, a known enhancer of cardiovascular health, and DOX-induced cardiotoxicity has been a focus of recent research. Exercise has been suggested to mitigate DOX's cardiac harm by modulating the Yes-associated protein (YAP) and Signal transducer and activator of transcription 3 (STAT3) pathways, which are crucial in regulating cardiac cell functions and responses to damage. This study aimed to assess the protective role of exercise preconditioning against DOX-induced cardiac injury. We used Sprague-Dawley rats, divided into five groups (control, DOX, exercise preconditioning (EP), EP-DOX, and verteporfin + EP + DOX), to investigate the potential mechanisms. Our findings, including echocardiography, histological staining, Western blot, and q-PCR analysis, demonstrated that exercise preconditioning could alleviate DOX-induced cardiac dysfunction and structural damage. Notably, exercise preconditioning enhanced the nuclear localization and co-localization of YAP and STAT3. Our study suggests that exercise preconditioning may counteract DOX-induced cardiotoxicity by activating the YAP/STAT3 pathway, highlighting a potential therapeutic approach for reducing DOX's cardiac side effects.
Collapse
Affiliation(s)
- Chuan-Zhi Wang
- School of Physical Education, Qingdao University, Qingdao, China
- Cancer Institute of the Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
- School of Physical Education and Sports Science, South China Normal University, China
| | - Heng-Zhi Guo
- School of Physical Education, Qingdao University, Qingdao, China
| | - Jing-Zhi Leng
- School of Physical Education, Qingdao University, Qingdao, China
| | - Zhi-De Liang
- School of Physical Education, Qingdao University, Qingdao, China
| | - Jing-Tai Wang
- School of Physical Education, Qingdao University, Qingdao, China
| | - Li-Jie Luo
- Jining University and School of Physical Education, Jining, China
| | - Shi-Qiang Wang
- Hunan Research Centre in Physical Fitness, Health, and Performance Excellence, Hunan University of Technology, Hunan, China
| | - Yang Yuan
- Cancer Institute of the Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| |
Collapse
|
40
|
Qiu B, Zhou Y, Tao X, Hou X, Du L, Lv Y, Yu L. The effect of exercise on flow-mediated dilation in people with type 2 diabetes mellitus: a systematic review and meta-analysis of randomized controlled trials. Front Endocrinol (Lausanne) 2024; 15:1347399. [PMID: 38596227 PMCID: PMC11002232 DOI: 10.3389/fendo.2024.1347399] [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: 11/30/2023] [Accepted: 03/11/2024] [Indexed: 04/11/2024] Open
Abstract
INTRODUCTION An increasing number of studies have investigated the effect of exercise on flow-mediated dilation (FMD) in people with type 2 diabetes mellitus (T2DM), while the findings were controversial. The primary aim of this systematic review and meta-analysis was to investigate the effect of exercise on FMD in T2DM patients, and the secondary aim was to investigate the optimal type, frequency, session duration, and weekly time of exercise for T2DM patients. METHODS Searches were conducted in PubMed, Cochrane Library, Scopus, Web of Science, Embase and EBSCO databases. The Cochrane risk of bias tool (RoB2) in randomized trial and Physiotherapy Evidence Database (PEDro) scale were used to assess the methodological quality of the included studies. RESULTS From the 3636 search records initially retrieved, 13 studies met the inclusion criteria. Our meta-analysis revealed that exercise had a significant effect on improving FMD in T2DM patients [WMD, 2.18 (95% CI, 1.78-2.58), p < 0.00001, I2 = 38%], with high-intensity interval training (HIIT) being the most effective intervention type [HIIT, 2.62 (1.42-3.82); p < 0.0001; aerobic exercise, 2.20 (1.29-3.11), p < 0.00001; resistance exercise, 1.91 (0.01-3.82), p = 0.05; multicomponent training, 1.49 (0.15-2.83), p = 0.03]. In addition, a higher frequency [> 3 times, 3.06 (1.94-4.19), p < 0.00001; ≤ 3 times, 2.02 (1.59-2.45), p < 0.00001], a shorter session duration [< 60 min, 3.39 (2.07-4.71), p < 0.00001; ≥ 60 min, 1.86 (1.32-2.40), p < 0.00001], and a shorter weekly time [≤ 180 min, 2.40 (1.63-3.17), p < 0.00001; > 180 min, 2.11 (0.82-3.40), p = 0.001] were associated with larger improvements in FMD. CONCLUSION This meta-analysis provides clinicians with evidence to recommended that T2DM patients participate in exercise, especially HIIT, more than 3 times per week for less than 60 min, with a target of 180 min per week being reached by increasing the frequency of exercise. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/prospero, identifier CRD42023466575.
Collapse
Affiliation(s)
- Bopeng Qiu
- Department of Strength and Conditioning Assessment and Monitoring, Beijing Sport University, Beijing, China
| | - Yilun Zhou
- Department of Strength and Conditioning Assessment and Monitoring, Beijing Sport University, Beijing, China
| | - Xifeng Tao
- Department of Strength and Conditioning Assessment and Monitoring, Beijing Sport University, Beijing, China
- School of Physical Education, Xihua University, Chengdu, China
| | - Xiao Hou
- School of Sport Sciences, Beijing Sport University, Beijing, China
| | - Liwen Du
- Department of Strength and Conditioning Assessment and Monitoring, Beijing Sport University, Beijing, China
| | - Yuanyuan Lv
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Laikang Yu
- Department of Strength and Conditioning Assessment and Monitoring, Beijing Sport University, Beijing, China
| |
Collapse
|
41
|
Liu WY, Li HM, Jiang H, Zhang WK. Effect of exercise training on heath, quality of life, exercise capacity in juvenile idiopathic arthritis: a meta-analysis of randomized controlled trials. Pediatr Rheumatol Online J 2024; 22:33. [PMID: 38438855 PMCID: PMC10910763 DOI: 10.1186/s12969-024-00967-3] [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: 11/23/2023] [Accepted: 02/21/2024] [Indexed: 03/06/2024] Open
Abstract
OBJECTIVE Little is known about the efficacy and safety of exercise training on juvenile idiopathic arthritis (JIA). This study aims to investigate the effect of exercise on health, quality of life, and different exercise capacities in individuals with JIA. METHOD A comprehensive search of Medline, Embase, Web of Science, and the Cochrane Library was conducted from database inception to October, 2023. Included studies were randomized controlled trials (RCTs) reporting the effects of exercise on JIA patients. Two independent reviewers assessed the literature quality using the Cochrane Collaboration's risk of bias tool. Standardized mean differences (SMD) were combined using random or fixed effects models. The level of evidence was assessed using the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach. RESULT Five RCTs met the inclusion criteria, containing 216 female participants and 90 males. The meta-analysis results showed that exercise had no significant effect on JIA patients based on the Child Health Assessment Questionnaire (CHAQ) (SMD=-0.32, 95%CI: -0.83, 0.19; I2 = 73.2%, P = 0.011) and Quality of Life (QoL) (SMD = 0.27, 95%CI: -0.04, 0.58; I2 = 29.4%, P = 0.243) and no significant effect on peak oxygen uptake (VO2peak). However, exercise significantly reduced visual analog scale (VAS) pain scores in JIA patients (SMD = 0.50, 95%CI: -0.90, -0.10; I2 = 50.2%, P = 0.134). The quality of evidence assessed by GRADE was moderate to very low. CONCLUSION Exercise does not significantly affect the quality of life and exercise capacity in JIA patients but may relieve pain. More RCTs are needed in the future to explore the effects of exercise on JIA.
Collapse
Affiliation(s)
- Wen-Yu Liu
- Weifang Institute of Technology, Weifang, Shandong, China
- Dongshin University, Rojo, South Jeolla, South Korea
| | - Hui-Min Li
- Yantai Gold College, Yantai, Shandong, China
| | - Hao Jiang
- Weifang Institute of Technology, Weifang, Shandong, China
| | - Wen-Kui Zhang
- Dongshin University, Rojo, South Jeolla, South Korea.
- Universiti Malaysia Sarawak, East Malaysian Borneo, Sarawak, Malaysia.
| |
Collapse
|
42
|
Cao R, Tian H, Tian Y, Fu X. A Hierarchical Mechanotransduction System: From Macro to Micro. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302327. [PMID: 38145330 PMCID: PMC10953595 DOI: 10.1002/advs.202302327] [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: 04/12/2023] [Revised: 10/27/2023] [Indexed: 12/26/2023]
Abstract
Mechanotransduction is a strictly regulated process whereby mechanical stimuli, including mechanical forces and properties, are sensed and translated into biochemical signals. Increasing data demonstrate that mechanotransduction is crucial for regulating macroscopic and microscopic dynamics and functionalities. However, the actions and mechanisms of mechanotransduction across multiple hierarchies, from molecules, subcellular structures, cells, tissues/organs, to the whole-body level, have not been yet comprehensively documented. Herein, the biological roles and operational mechanisms of mechanotransduction from macro to micro are revisited, with a focus on the orchestrations across diverse hierarchies. The implications, applications, and challenges of mechanotransduction in human diseases are also summarized and discussed. Together, this knowledge from a hierarchical perspective has the potential to refresh insights into mechanotransduction regulation and disease pathogenesis and therapy, and ultimately revolutionize the prevention, diagnosis, and treatment of human diseases.
Collapse
Affiliation(s)
- Rong Cao
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Huimin Tian
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Yan Tian
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Xianghui Fu
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| |
Collapse
|
43
|
Bei Y, Wang H, Liu Y, Su Z, Li X, Zhu Y, Zhang Z, Yin M, Chen C, Li L, Wei M, Meng X, Liang X, Huang Z, Cao RY, Wang L, Li G, Cretoiu D, Xiao J. Exercise-Induced miR-210 Promotes Cardiomyocyte Proliferation and Survival and Mediates Exercise-Induced Cardiac Protection against Ischemia/Reperfusion Injury. RESEARCH (WASHINGTON, D.C.) 2024; 7:0327. [PMID: 38410280 PMCID: PMC10895486 DOI: 10.34133/research.0327] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/01/2024] [Indexed: 02/28/2024]
Abstract
Exercise can stimulate physiological cardiac growth and provide cardioprotection effect in ischemia/reperfusion (I/R) injury. MiR-210 is regulated in the adaptation process induced by exercise; however, its impact on exercise-induced physiological cardiac growth and its contribution to exercise-driven cardioprotection remain unclear. We investigated the role and mechanism of miR-210 in exercise-induced physiological cardiac growth and explored whether miR-210 contributes to exercise-induced protection in alleviating I/R injury. Here, we first observed that regular swimming exercise can markedly increase miR-210 levels in the heart and blood samples of rats and mice. Circulating miR-210 levels were also elevated after a programmed cardiac rehabilitation in patients that were diagnosed of coronary heart diseases. In 8-week swimming model in wild-type (WT) and miR-210 knockout (KO) rats, we demonstrated that miR-210 was not integral for exercise-induced cardiac hypertrophy but it did influence cardiomyocyte proliferative activity. In neonatal rat cardiomyocytes, miR-210 promoted cell proliferation and suppressed apoptosis while not altering cell size. Additionally, miR-210 promoted cardiomyocyte proliferation and survival in human embryonic stem cell-derived cardiomyocytes (hESC-CMs) and AC16 cell line, indicating its functional roles in human cardiomyocytes. We further identified miR-210 target genes, cyclin-dependent kinase 10 (CDK10) and ephrin-A3 (EFNA3), that regulate cardiomyocyte proliferation and apoptosis. Finally, miR-210 KO and WT rats were subjected to swimming exercise followed by I/R injury. We demonstrated that miR-210 crucially contributed to exercise-driven cardioprotection against I/R injury. In summary, this study elucidates the role of miR-210, an exercise-responsive miRNA, in promoting the proliferative activity of cardiomyocytes during physiological cardiac growth. Furthermore, miR-210 plays an essential role in mediating the protective effects of exercise against cardiac I/R injury. Our findings suggest exercise as a potent nonpharmaceutical intervention for inducing miR-210, which can alleviate I/R injury and promote cardioprotection.
Collapse
Affiliation(s)
- Yihua Bei
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
Shanghai University, Shanghai 200444, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine,
Shanghai University, Shanghai 200444, China
| | - Hongyun Wang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
Shanghai University, Shanghai 200444, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine,
Shanghai University, Shanghai 200444, China
| | - Yang Liu
- Department of Cardiology, Shanghai Tongji Hospital,
Tongji University School of Medicine, Shanghai 200065, China
| | - Zhuhua Su
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
Shanghai University, Shanghai 200444, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine,
Shanghai University, Shanghai 200444, China
| | - Xinpeng Li
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
Shanghai University, Shanghai 200444, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine,
Shanghai University, Shanghai 200444, China
- School of Environmental and Chemical Engineering,
Shanghai University, Shanghai 200444, China
| | - Yujiao Zhu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
Shanghai University, Shanghai 200444, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine,
Shanghai University, Shanghai 200444, China
| | - Ziyi Zhang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
Shanghai University, Shanghai 200444, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine,
Shanghai University, Shanghai 200444, China
| | - Mingming Yin
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
Shanghai University, Shanghai 200444, China
| | - Chen Chen
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
Shanghai University, Shanghai 200444, China
| | - Lin Li
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
Shanghai University, Shanghai 200444, China
| | - Meng Wei
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
Shanghai University, Shanghai 200444, China
| | - Xiangmin Meng
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China
| | - Xuchun Liang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
Shanghai University, Shanghai 200444, China
| | - Zhenzhen Huang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
Shanghai University, Shanghai 200444, China
| | - Richard Yang Cao
- Cardiac Rehabilitation Program, Shanghai Xuhui Central Hospital/Zhongshan-Xuhui Hospital,
Fudan University/Shanghai Clinical Research Center, Shanghai 200031, China
| | - Lei Wang
- Department of Rehabilitation Medicine,
Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Dragos Cretoiu
- Department of Medical Genetics,
Carol Davila University of Medicine and Pharmacy, Bucharest 020031, Romania
- Materno-Fetal Assistance Excellence Unit, Alessandrescu-Rusescu National Institute for Mother and Child Health, Bucharest 011062, Romania
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
Shanghai University, Shanghai 200444, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine,
Shanghai University, Shanghai 200444, China
| |
Collapse
|
44
|
Xiao J, Chen X, Guo W, Li Y, Liu J. Moderate intensity exercise may protect cardiac function by influencing spleen microbiome composition. iScience 2024; 27:108635. [PMID: 38292426 PMCID: PMC10826308 DOI: 10.1016/j.isci.2023.108635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/15/2023] [Accepted: 12/01/2023] [Indexed: 02/01/2024] Open
Abstract
The beneficial effects of physical exercise on human cardiorespiratory fitness might be through reduced systemic inflammation, but the mechanism remains a controversy. Recent studies have highlighted the importance of spleen microbiomes in immune regulation. Hence, we conducted a study using a high-fat diet and exercise mouse model to investigate the relationships among different exercise intensities, spleen microbiome composition, and cardiac function. The mice spleen contained a diverse array of microbiota. Different intensities of exercise resulted in varying compositions of the spleen microbiome, Treg cell levels, and mouse heart function. Additionally, the abundance of Lactobacillus johnsonii in the mouse spleen exhibited a positive correlation with Treg cell levels, suggesting that Lactobacillus johnsonii may contribute to the production of Treg cells, potentially explaining the protective role of moderate-intensity exercise on cardiac function. In conclusion, our findings provide evidence that moderate-intensity exercise may promote cardiac function protection by influencing the spleen microbiome composition.
Collapse
Affiliation(s)
- Jie Xiao
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China
| | - Xing Chen
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China
| | - Weina Guo
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yang Li
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China
| | - Jinping Liu
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China
| |
Collapse
|
45
|
Cavallero S, Roustaei M, Satta S, Cho JM, Phan H, Baek KI, Blázquez-Medela AM, Gonzalez-Ramos S, Vu K, Park SK, Yokota T, Sumner J, Mack JJ, Sigmund CD, Reddy ST, Li R, Hsiai TK. Exercise mitigates flow recirculation and activates metabolic transducer SCD1 to catalyze vascular protective metabolites. SCIENCE ADVANCES 2024; 10:eadj7481. [PMID: 38354249 PMCID: PMC10866565 DOI: 10.1126/sciadv.adj7481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/11/2024] [Indexed: 02/16/2024]
Abstract
Exercise promotes pulsatile shear stress in the arterial circulation and ameliorates cardiometabolic diseases. However, exercise-mediated metabolic transducers for vascular protection remain under-investigated. Untargeted metabolomic analysis demonstrated that wild-type mice undergoing voluntary wheel running exercise expressed increased endothelial stearoyl-CoA desaturase 1 (SCD1) that catalyzes anti-inflammatory lipid metabolites, namely, oleic (OA) and palmitoleic acids (PA), to mitigate NF-κB-mediated inflammatory responses. In silico analysis revealed that exercise augmented time-averaged wall shear stress but mitigated flow recirculation and oscillatory shear index in the lesser curvature of the mouse aortic arch. Following exercise, endothelial Scd1-deleted mice (Ldlr-/- Scd1EC-/-) on high-fat diet developed persistent VCAM1-positive endothelium in the lesser curvature and the descending aorta, whereas SCD1 overexpression via adenovirus transfection mitigated endoplasmic reticulum stress and inflammatory biomarkers. Single-cell transcriptomics of the aorta identified Scd1-positive and Vcam1-negative endothelial subclusters interacting with other candidate genes. Thus, exercise mitigates flow recirculation and activates endothelial SCD1 to catalyze OA and PA for vascular endothelial protection.
Collapse
Affiliation(s)
- Susana Cavallero
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
- Department of Medicine, VA Greater Los Angeles Health Care System, Los Angeles, CA, USA
| | - Mehrdad Roustaei
- Department of Bioengineering, School of Engineering and Applied Science, University of California, Los Angeles, CA, USA
| | - Sandro Satta
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
- Department of Medicine, VA Greater Los Angeles Health Care System, Los Angeles, CA, USA
| | - Jae Min Cho
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
- Department of Medicine, VA Greater Los Angeles Health Care System, Los Angeles, CA, USA
| | - Henry Phan
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Kyung In Baek
- Department of Bioengineering, School of Engineering and Applied Science, University of California, Los Angeles, CA, USA
| | - Ana M. Blázquez-Medela
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
- Department of Medicine, VA Greater Los Angeles Health Care System, Los Angeles, CA, USA
| | - Sheila Gonzalez-Ramos
- Department of Bioengineering, School of Engineering and Applied Science, University of California, Los Angeles, CA, USA
| | - Khoa Vu
- Department of Bioengineering, School of Engineering and Applied Science, University of California, Los Angeles, CA, USA
| | - Seul-Ki Park
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Tomohiro Yokota
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
- Department of Medicine, VA Greater Los Angeles Health Care System, Los Angeles, CA, USA
| | - Jennifer Sumner
- Department of Psychology, College of Life Sciences, University of California, Los Angeles, CA, USA
| | - Julia J. Mack
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Curt D. Sigmund
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Srinivasa T. Reddy
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Rongsong Li
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
- Department of Medicine, VA Greater Los Angeles Health Care System, Los Angeles, CA, USA
| | - Tzung K. Hsiai
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
- Department of Medicine, VA Greater Los Angeles Health Care System, Los Angeles, CA, USA
- Department of Bioengineering, School of Engineering and Applied Science, University of California, Los Angeles, CA, USA
| |
Collapse
|
46
|
Peng Y, Qin D, Wang Y, Gao W, Xu X. Pharmacological inhibition of ICOS attenuates the protective effect of exercise on cardiac fibrosis induced by isoproterenol. Eur J Pharmacol 2024; 965:176327. [PMID: 38224847 DOI: 10.1016/j.ejphar.2024.176327] [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: 08/27/2023] [Revised: 12/14/2023] [Accepted: 01/09/2024] [Indexed: 01/17/2024]
Abstract
AIMS To investigate the cardioprotective mechanism of exercise or exercise combined with inducible costimulatory molecules (ICOS) monoclonal antibody (mAb) therapy against isoproterenol (ISO)-induced cardiac remodeling. MAIN METHODS Totally 24 male C57BL/6J mice were randomly divided into four groups: the control group (normal saline treatment), ISO group (subcutaneous injection of isoproterenol, 10 mg/kg/day, once daily for 5 consecutive days), the exercise with subcutaneous ISO injection group (EPI), and the exercise with injected with ISO and ICOS mAb group (EPII). The mice in EPI and EPII group were trained on a small animal treadmill for 4 weeks (13 m/min, 0% grade, 60min/day). KEY FINDINGS Exercise significantly attenuated CD45+, Mac-2 inflammatory cell infiltration, cardiac fibrosis and inhibited the RIPK1/RIPK3/MLKL/CaMKII and cardiomyocyte pyroptosis pathways to counter ISO-induced severe cardiac injury. The administration of the ICOS mAb may inhibit the cardioprotection of exercise against ISO-induced heart damage. Compared to those in EPI, our data showed that the increasing levels of myocardial fibrosis, the leukocyte infiltration of cardiac tissue and proteins expression of cardiac myocyte necrosis and pyroptosis signaling pathways in the EPII group. SIGNIFICANCE Our results demonstrated that exercise decreased leukocyte infiltration in heart, inhibited the cardiomyocyte pyroptosis and necroptosis signaling pathways, and attenuated inflammatory responses to alleviate ISO-induced cardiac fibrosis. However, the antifibrotic effects of combined treatment with exercise and ICOS mAb intervention did not exhibit synergistic enhancement.
Collapse
Affiliation(s)
- Yong Peng
- School of Kinesiology, Shanghai University of Sport, Shanghai, China; Jiangsu Collaborative Innovation Center for Sports and Health Project, Nanjing Sport Institute, Nanjing, Jiangsu, China; Key Laboratory of Exercise Training and Rehabilitation for Jiangsu Province, Nanjing Sport Institute, Nanjing, Jiangsu, China
| | - Di Qin
- School of Sport and Health, Nanjing Sport Institute, Nanjing, Jiangsu, China
| | - Yudi Wang
- School of Physical Education and Nursing, Chengdu College of Arts and Sciences, Chengdu, Sichuan, China
| | - Wenyue Gao
- School of Sport and Health, Nanjing Sport Institute, Nanjing, Jiangsu, China
| | - Xin Xu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China.
| |
Collapse
|
47
|
Huang J, Yu Z, Wu Y, He X, Zhao J, He J, Staessen JA, Dong Y, Liu C, Wei FF. Prognostic Significance of Blood Pressure at Rest and After Performing the Six-Minute Walk Test in Patients With Acute Heart Failure. Am J Hypertens 2024; 37:199-206. [PMID: 38041568 DOI: 10.1093/ajh/hpad115] [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: 07/31/2023] [Revised: 09/14/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND It remains unclear whether systolic (SBP) and diastolic (DBP) pressure and BP response after six-minute walk test (6MWT) are associated with adverse outcomes in patients with acute heart failure (AHF). METHODS We investigated these associations in 98 AHF patients (24.5% women; mean age, 70.5 years) enrolled in the ROSE trial (The Low-dose Dopamine or Low-dose Nesiritide in Acute Heart Failure with Renal Dysfunction). The primary endpoint consisted of any death or rehospitalization within 6 months after randomization. We computed hazard ratios (HRs) of the risks associated with 1-SD increase in post-exercise BP levels and BP ratios, calculated as BP immediately after 6MWT divided by BP before 6MWT. RESULTS The BP before and after 6MWT averaged 110.6/117.5 mm Hg for SBP and 61.9/64.7 mm Hg for DBP. In multivariable-adjusted analyses including clinic BP measured at the same day of 6MWT, higher DBP after 6MWT was associated with lower risk of the primary endpoint (HR, 0.49; 95% confidence interval [CI], 0.26-0.95; P = 0.034). Both higher SBP and DBP immediately after 6MWT were associated with lower risk of 6-month mortality (HRs, 0.39/0.16; 95% CI, 0.17-0.90/0.065-0.40; P ≤ 0.026). The post-exercise SBP ratio was associated with the risk of 6-month mortality in multivariable-adjusted analyses (HR, 0.44; P = 0.023). CONCLUSIONS Higher BP levels and BP ratios immediately after 6MWT conferred lower risk of adverse health outcomes. Our observations highlight that 6MWT-related BP level and response may refine risk estimates in patients hospitalized AHF and may help further investigation for the development of HF preventive strategies.
Collapse
Affiliation(s)
- Jiale Huang
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhongping Yu
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yuzhong Wu
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xin He
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jingjing Zhao
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jiangui He
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jan A Staessen
- Non-Profit Research Association Alliance for the Promotion of Preventive Medicine, Mechelen, Belgium
- Biomedical Research Group, Faculty of Medicine, University of Leuven, Leuven, Belgium
| | - Yugang Dong
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangzhou, Guangdong, China
- National Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, Guangzhou, China
| | - Chen Liu
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangzhou, Guangdong, China
- National Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, Guangzhou, China
| | - Fang-Fei Wei
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
48
|
Meng S, Yu Y, Yu S, Zhu S, Shi M, Xiang M, Ma H. Advances in Metabolic Remodeling and Intervention Strategies in Heart Failure. J Cardiovasc Transl Res 2024; 17:36-55. [PMID: 37843752 DOI: 10.1007/s12265-023-10443-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/27/2023] [Indexed: 10/17/2023]
Abstract
The heart is the most energy-demanding organ throughout the whole body. Perturbations or failure in energy metabolism contributes to heart failure (HF), which represents the advanced stage of various heart diseases. The poor prognosis and huge economic burden associated with HF underscore the high unmet need to explore novel therapies targeting metabolic modulators beyond conventional approaches focused on neurohormonal and hemodynamic regulators. Emerging evidence suggests that alterations in metabolic substrate reliance, metabolic pathways, metabolic by-products, and energy production collectively regulate the occurrence and progression of HF. In this review, we provide an overview of cardiac metabolic remodeling, encompassing the utilization of free fatty acids, glucose metabolism, ketone bodies, and branched-chain amino acids both in the physiological condition and heart failure. Most importantly, the latest advances in pharmacological interventions are discussed as a promising therapeutic approach to restore cardiac function, drawing insights from recent basic research, preclinical and clinical studies.
Collapse
Affiliation(s)
- Simin Meng
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Yi Yu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Shuo Yu
- Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Shiyu Zhu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Mengjia Shi
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China.
| | - Hong Ma
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
49
|
Cai X, Liu M, Xu X, Zhang S, Huang R, Wang P, He L, Chen Z, Liu Z, Zhou Y, Guo Y, Zhuang X, Liao X. Cardiovascular effects of weight loss in old adults with overweight/obesity according to change in skeletal muscle mass. J Cachexia Sarcopenia Muscle 2024; 15:342-351. [PMID: 38108096 PMCID: PMC10834329 DOI: 10.1002/jcsm.13409] [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: 03/29/2023] [Revised: 08/24/2023] [Accepted: 09/22/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND Patients with overweight/obesity and type 2 diabetes are encouraged to lose weight, but not all losing weight gain better cardiovascular health, especially old adults. The change in skeletal muscle mass (SMM) could be the key that explains the heterogenous cardiovascular effects of weight loss. This study aims to assess whether the cardiovascular effects of weight loss vary for those gaining skeletal muscle along with weight loss. METHODS The old adults with overweight/obesity and type 2 diabetes in the Look AHEAD study having muscle measurement from dual-energy X-ray absorptiometry were included. Based on the weight change (WC) and SMM change (SMMC) between baseline and the 4-year follow-up, participants were allocated into three groups-weight gain (WG) group, weight loss with muscle loss (WL-ML) group and weight loss with muscle gain (WL-MG) group. Cox proportional hazards regression was performed to evaluate the cardiovascular risk of those gaining or losing SMM with weight loss compared with those gaining weight. Among the participants with weight loss, the ratio of SMMC/WC was calculated, and the association of SMMC/WC with primary cardiovascular outcome was assessed. RESULTS A total of 491 participants were included in the study with an average age of 64.56 ± 3.81 years old. A total of 47.0% were male and 49.9% were from the intensive lifestyle intervention arm. Based on their WC and SMMC, 43 were assigned to the WG group, 373 to the WL-ML group and 75 to the WL-MG group. Over a follow-up of almost 10 years, 97 participants encountered the primary endpoint. The WG group had the highest incidence of 25.59%, the WL-MG group had the lowest incidence of 9.33% and the WL-ML group had 21.18% (P = 0.040). In the fourth adjusted Cox model, the WL-MG group achieved significantly decreased odds of the primary endpoint compared with the WG group (hazard ratio [HR] 0.33, 95% confidence interval [CI] [0.12, 0.87], P = 0.026), whilst the WL-ML group did not (HR 0.91, 95% CI [0.47, 1.78], P = 0.670). Among the participants with weight loss, when SMMC/WC reached around 50%, this HR soared to approximately two-fold. CONCLUSIONS The participants gaining SMM along with weight loss achieved the lowest odds of adverse cardiovascular events, whilst those who lost SMM along with weight loss had comparable cardiovascular risk with those gaining weight. The more muscle lost during weight loss, the greater the harm. The cardiovascular effects of weight loss were modulated by whether the participants gained SMM meanwhile losing weight.
Collapse
Affiliation(s)
- Xiaojie Cai
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Menghui Liu
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Xingfeng Xu
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Shaozhao Zhang
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Rihua Huang
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Peng Wang
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Lixiang He
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Zhuohui Chen
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Zishan Liu
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Yi Zhou
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Yue Guo
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Xiaodong Zhuang
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Xinxue Liao
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
50
|
Tang J, Wu C, Xu Y, Yang B, Xi Y, Cai M, Tian Z. Resistance training up-regulates Smyd1 expression and inhibits oxidative stress and endoplasmic reticulum stress in the heart of middle-aged mice. Free Radic Biol Med 2024; 210:304-317. [PMID: 38042222 DOI: 10.1016/j.freeradbiomed.2023.11.029] [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: 09/15/2023] [Revised: 11/15/2023] [Accepted: 11/26/2023] [Indexed: 12/04/2023]
Abstract
Persistent oxidative stress and endoplasmic reticulum (ER) stress are the primary mechanisms of age-related cardiovascular diseases. Although exercise training is viewed as an effective anti-aging approach, further exploration is needed to identify the mechanisms and functional targets. In this study, the impact of resistance training (RT) on the expression of Smyd1, the levels of reactive oxygen species (ROS) and the expression of ER stress-related protein in the hearts of mice of different ages were assessed. In addition, the role of Smyd1 in the aging-induced oxidative stress and ER stress were evaluated in d-galactose (D-gal)-treated H9C2 cells. We demonstrated that RT in middle age increased the expression of Smyd1 and restricted heart aging-induced ER stress. Overexpression of Smyd1 restrained oxidative stress and ER stress in D-gal-treated H9C2 cells, while the inhibition of Nrf2 and Smyd1 escalated ER stress. These findings demonstrate that Smyd1 has significant impact in regulating age-related ER stress. RT in middle age can up-regulates Smyd1 expression and inhibits oxidative stress and ER stress in the heart.
Collapse
Affiliation(s)
- Jie Tang
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, 710119, PR China
| | - Chenyang Wu
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, 710119, PR China
| | - Yuxiang Xu
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, 710119, PR China
| | - Boran Yang
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, 710119, PR China
| | - Yue Xi
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, 710119, PR China
| | - Mengxin Cai
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, 710119, PR China.
| | - Zhenjun Tian
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, 710119, PR China
| |
Collapse
|