1
|
Calabrese EJ, Mattson MP. The catabolic - anabolic cycling hormesis model of health and resilience. Ageing Res Rev 2024; 102:102588. [PMID: 39551161 DOI: 10.1016/j.arr.2024.102588] [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: 07/22/2024] [Revised: 10/31/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
A major goal of aging research is to identify ways of extending productive and disease-free lifespans. Here we present the catabolic - anabolic cycling hormesis (CACH) model for optimizing health. The CACH model is based on the concept that cells and organ systems respond to catabolic challenges in ways that bolster their resilience and that an anabolic recovery period is required to effectuate the benefits of the catabolic challenge. As two prominent real-world examples we highlight the literature on the molecular and cellular mechanisms by which physical exercise and intermittent fasting bolster cellular and organismal performance and resilience, and suppress disease processes. Over periods of weeks and months the CACH of exercise and fasting promote optimal health. The hormesis concept is integral to the CACH model and predicts an upper limit to the beneficial effects of catabolic - anabolic cycling that reflects a limit of biological plasticity. This paper extends the hormesis model of health by proposing that 1) it is comprised of two complementary phases: catabolic (adaptive stress responses and conservation of resources) and anabolic (growth and plasticity) and, 2) that CACH is metabolically integrated, quantitatively flexible and dynamically regulated. This model has important implications for future basic and translational research in the fields of aging and related disease processes.
Collapse
Affiliation(s)
- Edward J Calabrese
- Department of Environmental Health Sciences, University of Massachusetts, Amherst, MA 01003. USA.
| | - Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205. USA.
| |
Collapse
|
2
|
Maruyama N, Ogata T, Kasahara T, Hamaoka T, Higuchi Y, Tsuji Y, Tomita S, Sakamoto A, Nakanishi N, Matoba S. Loss of Cavin-2 destabilizes phosphatase and tensin homologue and enhances Akt signalling pathway in cardiomyocytes. Cardiovasc Res 2024; 120:1562-1576. [PMID: 38861679 DOI: 10.1093/cvr/cvae130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 03/19/2024] [Accepted: 05/03/2024] [Indexed: 06/13/2024] Open
Abstract
AIMS Specific cavins and caveolins, known as caveola-related proteins, have been implicated in cardiac hypertrophy and myocardial injury. Cavin-2 forms complexes with other caveola-related proteins, but the role of Cavin-2 in cardiomyocytes (CMs) is poorly understood. Here, we investigated an unknown function of Cavin-2 in CMs. METHODS AND RESULTS Under cardiac stress-free conditions, systemic Cavin-2 knockout (KO) induced mild and significant CM hypertrophy. Cavin-2 KO suppressed phosphatase and tensin homologue (PTEN) associated with Akt signalling, whereas there was no difference in Akt activity between the hearts of the wild-type and the Cavin-2 KO mice under cardiac stress-free conditions. However, after swim training, CM hypertrophy was more facilitated with enhanced phosphoinositide 3-kinase (PI3K)-Akt activity in the hearts of Cavin-2 KO mice. Cavin-2 knockdown neonatal rat CMs (NRCMs) using adenovirus expressing Cavin-2 short hairpin RNA were hypertrophied and resistant to hypoxia and H2O2-induced apoptosis. Cavin-2 knockdown increased Akt phosphorylation in NRCMs, and an Akt inhibitor inhibited Cavin-2 knockdown-induced anti-apoptotic responses in a dose-dependent manner. Cavin-2 knockdown increased phosphatidylinositol-3,4,5-triphosphate production and attenuated PTEN at the membrane fraction of NRCMs. Immunostaining and immunoprecipitation showed that Cavin-2 was associated with PTEN at the plasma membrane of NRCMs. A protein stability assay showed that Cavin-2 knockdown promoted PTEN destabilization in NRCMs. In an Angiotensin II (2-week continuous infusion)-induced pathological cardiac hypertrophy model, CM hypertrophy and CM apoptosis were suppressed in CM-specific Cavin-2 conditional KO (Cavin-2 cKO) mice. Because Cavin-2 cKO mouse hearts showed increased Akt activity but not decreased extracellular signal-regulated kinase activity, suppression of pathological hypertrophy by Cavin-2 loss may be due to increased survival of healthy CMs. CONCLUSION Cavin-2 plays a negative regulator in the PI3K-Akt signalling in CMs through interaction with PTEN. Loss of Cavin-2 enhances Akt activity by promoting PTEN destabilization, which promotes physiological CM hypertrophy and may enhance Akt-mediated cardioprotective effects against pathological CM hypertrophy.
Collapse
Affiliation(s)
- Naoki Maruyama
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Takehiro Ogata
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
- Department of Pathology and Cell Regulation, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Takeru Kasahara
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Tetsuro Hamaoka
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yusuke Higuchi
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yumika Tsuji
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Shinya Tomita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Akira Sakamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Naohiko Nakanishi
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
3
|
Zhang J. Non-coding RNAs and angiogenesis in cardiovascular diseases: a comprehensive review. Mol Cell Biochem 2024; 479:2921-2953. [PMID: 38306012 DOI: 10.1007/s11010-023-04919-5] [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: 10/27/2023] [Accepted: 12/18/2023] [Indexed: 02/03/2024]
Abstract
Non-coding RNAs (ncRNAs) have key roles in the etiology of many illnesses, including heart failure, myocardial infarction, stroke, and in physiological processes like angiogenesis. In transcriptional regulatory circuits that control heart growth, signaling, and stress response, as well as remodeling in cardiac disease, ncRNAs have become important players. Studies on ncRNAs and cardiovascular disease have made great progress recently. Here, we go through the functions of non-coding RNAs (ncRNAs) like circular RNAs (circRNAs), and microRNAs (miRNAs) as well as long non-coding RNAs (lncRNAs) in modulating cardiovascular disorders.
Collapse
Affiliation(s)
- Jie Zhang
- Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
4
|
Lopes Soares L, Portes AMO, Costa SFF, Leite LB, Natali AJ. Autonomic Dysregulation in Pulmonary Hypertension: Role of Physical Exercise. Hypertension 2024; 81:2228-2236. [PMID: 39234679 DOI: 10.1161/hypertensionaha.124.23573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
Pulmonary hypertension (PH) is a rare and severe condition characterized by increased pressure in the pulmonary circulation, often resulting in right ventricular failure and death. The autonomic nervous system (ANS) plays a crucial role in the cardiovascular and pulmonary controls. Dysfunction of ANS has been implicated in the pathogenesis of cardiopulmonary diseases. Conversely, dysfunctions in ANS can arise from these diseases, impacting cardiac and pulmonary autonomic functions and contributing to disease progression. The complex interaction between ANS dysfunction and PH plays a crucial role in the disease progression, making it essential to explore interventions that modulate ANS, such as physical exercise, to improve the treatment and prognosis of patients with PH. This review addresses autonomic dysfunctions found in PH and their implications for the cardiopulmonary system. Furthermore, we discuss how physical exercise, a significant modulator of ANS, may contribute to the prognosis of PH. Drawing from evidence of aerobic and resistance exercise training in patients and experimental models of PH, potential cardiovascular benefits of exercise are presented. Finally, we highlight emerging therapeutic targets and perspectives to better cope with the complex condition. A comprehensive understanding of the interaction between ANS and PH, coupled with targeted physical exercise interventions, may pave the way for innovative therapeutic strategies and significantly improve the treatment and prognosis of vulnerable patients.
Collapse
Affiliation(s)
- Leôncio Lopes Soares
- Department of Physical Education, Federal University of Viçosa, Minas Gerais, Brazil (L.L.S., S.F.F.C., L.B.L., A.J.N.)
| | | | | | - Luciano Bernardes Leite
- Department of Physical Education, Federal University of Viçosa, Minas Gerais, Brazil (L.L.S., S.F.F.C., L.B.L., A.J.N.)
| | - Antônio José Natali
- Department of Physical Education, Federal University of Viçosa, Minas Gerais, Brazil (L.L.S., S.F.F.C., L.B.L., A.J.N.)
| |
Collapse
|
5
|
Suematsu Y, Morita H, Abe M, Uehara Y, Koyoshi R, Fujimi K, Ideishi A, Takata K, Kato Y, Hirata T, Yahiro E, Morito N, Kitajima K, Yano Y, Satoh A, Yoshimura C, Ishida S, Okutsu S, Takahashi K, Shinohara Y, Sakaguchi T, Katsuki S, Tada K, Fujii T, Funakoshi S, Hu Y, Satoh T, Ohnishi H, Okamura K, Mizuno H, Arakawa K, Asayama K, Ohtsubo T, Ishigami T, Shibata S, Fujita T, Munakata M, Ohishi M, Ichihara A, Katsuya T, Mukoyama M, Rakugi H, Node K, Arima H, Miura SI. Differences in the effects of exercise on blood pressure depending on the physical condition of the subject and the type of exercise: a systematic review and meta-analysis. Hypertens Res 2024:10.1038/s41440-024-01974-3. [PMID: 39487318 DOI: 10.1038/s41440-024-01974-3] [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: 08/26/2024] [Revised: 09/29/2024] [Accepted: 10/16/2024] [Indexed: 11/04/2024]
Abstract
Although hypertension is a major cause of cardiovascular disease, the control of blood pressure (BP) is insufficient worldwide. Exercise is an effective treatment for reducing BP, but the differences in the blood pressure lowering effects of exercise according to the underlying pathophysiological condition, the type of exercise, and the geographic region are not fully understood. An umbrella review with a meta-analysis of 435 randomized controlled trials that investigated the BP-lowering effects of exercise was performed using Ovid MEDLINE and the Cochrane Library, covering the period from inception to August 1, 2023. A random effects model meta-analysis was performed to estimate the effect size across multiple studies. Exercise significantly reduced systolic BP in healthy subjects (-3.51 mmHg, 95% confidence interval: -3.90, -3.11; p < 0.001) and in those with lifestyle-related diseases including hypertension (-5.48 mmHg, -6.51, -4.45; p < 0.001), but not in those with cardiovascular diseases (-1.16 mmHg, -4.08, 1.76; p = 0.44). According to the type of exercise, all types significantly reduced systolic BP in healthy subjects and in those with lifestyle-related diseases, but not in those with cardiovascular diseases. According to the region, in Oceania, there were no reductions in systolic BP. In Asia, systolic BP was reduced in patients with cardiovascular diseases. In conclusion, any type of exercise reduced BP in healthy subjects and in those with lifestyle-related diseases, but not in those with cardiovascular diseases, and the region affected the effect of exercise. When using exercise to reduce hypertension, it is important to consider the patient's pathophysiological condition and the region.
Collapse
Affiliation(s)
- Yasunori Suematsu
- Department of Cardiology, Fukuoka University Hospital, Fukuoka, Japan
| | - Hidetaka Morita
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Makiko Abe
- Department of Preventive Medicine and Public Health, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Yoshinari Uehara
- Faculty of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Rie Koyoshi
- Division of Medical Safety Management, Fukuoka University Hospital, Fukuoka, Japan
| | - Kanta Fujimi
- Department of Rehabilitation, Fukuoka University Hospital, Fukuoka, Japan
| | - Akihito Ideishi
- Department of Cardiology, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Kohei Takata
- Department of Clinical Laboratory Medicine, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Yuta Kato
- Department of Cardiology, Fukuoka University Hospital, Fukuoka, Japan
| | - Tetsuo Hirata
- Department of Cardiology, Fukuoka University Hospital, Fukuoka, Japan
| | - Eiji Yahiro
- Postgraduate Clinical Training Center, Fukuoka University Hospital, Fukuoka, Japan
| | - Natsumi Morito
- Department of Clinical Laboratory Medicine, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Ken Kitajima
- Medical Education Center, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | | | - Atsushi Satoh
- Laboratory of Epidemiology and Prevention, Kobe Pharmaceutical University, Kobe, Japan
| | - Chikara Yoshimura
- Department of Preventive Medicine and Public Health, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Shintaro Ishida
- Department of Preventive Medicine and Public Health, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Shota Okutsu
- Department of General Medicine, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Koji Takahashi
- Division of Nephrology and Rheumatology, Department of Internal Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Yukiko Shinohara
- Department of Preventive Medicine and Public Health, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Takashi Sakaguchi
- Division of Nephrology and Rheumatology, Department of Internal Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Shiori Katsuki
- Division of Nephrology and Rheumatology, Department of Internal Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Kazuhiro Tada
- Division of Nephrology and Rheumatology, Department of Internal Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Takako Fujii
- Department of Preventive Medicine and Public Health, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
- NCD Epidemiology Research Center, Shiga University of Medical Science, Otsu, Japan
| | - Shunsuke Funakoshi
- Department of Preventive Medicine and Public Health, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Yaopeng Hu
- Department of Physiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Tomonori Satoh
- Research Center for the Promotion of Health and Employment Support, Tohoku Rosai Hospital, Sendai, Japan
| | - Hirofumi Ohnishi
- Department of Public Health, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Keisuke Okamura
- Department of Cardiology and Cardiovascular Center, Imamura Hospital, Tosu, Japan
| | - Hiroyuki Mizuno
- Division of Cardiovascular Medicine, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Kimika Arakawa
- National Hospital Organization, Kyushu Medical Center, Department of Clinical Laboratory, Fukuoka, Japan
| | - Kei Asayama
- Department of Hygiene and Public Health, Teikyo University School of Medicine, Tokyo, Japan
| | - Toshio Ohtsubo
- Department of Hypertension Internal Medicine, Fukuoka Red Cross Hospital, Fukuoka, Japan
| | - Tomoaki Ishigami
- Department of Cardiology, Yokohama City University Hospital, Yokohama, Japan
| | - Shigeru Shibata
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Takayuki Fujita
- Department of Physiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Masanori Munakata
- Research Center for the Promotion of Health and Employment Support, Tohoku Rosai Hospital, Sendai, Japan
| | - Mitsuru Ohishi
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Atsuhiro Ichihara
- Department of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | | | - Masashi Mukoyama
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | | | - Koichi Node
- Department of Cardiovascular Medicine, Saga University, Saga, Japan
| | - Hisatomi Arima
- Department of Preventive Medicine and Public Health, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Shin-Ichiro Miura
- Department of Cardiology, Fukuoka University Faculty of Medicine, Fukuoka, Japan.
| |
Collapse
|
6
|
Deng C, Wu Y. Vitamin D-Parathyroid Hormone-Fibroblast Growth Factor 23 Axis and Cardiac Remodeling. Am J Cardiovasc Drugs 2024:10.1007/s40256-024-00688-8. [PMID: 39392562 DOI: 10.1007/s40256-024-00688-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/26/2024] [Indexed: 10/12/2024]
Abstract
Cardiac remodeling is a compensatory adaptive response to chronic heart failure (HF) altering the structure, function, and metabolism of the heart. Many nutritional and metabolic diseases can aggravate the pathophysiological development of cardiac remodeling. Vitamin D deficiency leads to cardiac remodeling by activating the renin-angiotensin-aldosterone system (RAAS), resulting in enhanced inflammation and directly promoting cardiac fibrosis and extracellular matrix deposition. Hyperparathyroidism upregulates protein kinase A or protein kinase C, enhances intracellular calcium influx, promotes oxidative stress, activates RAAS, and increases aldosterone levels, thereby aggravating cardiac remodeling. Besides, fibroblast growth factor 23 (FGF23) plays a direct role in the heart, resulting in ventricular hypertrophy and myocardial fibrosis. Vitamin D deficiency leads to hyperparathyroidism, which in turn increases the level of FGF23. Elevated levels of FGF23 further inhibit vitamin D synthesis. Evidence exists that vitamin D deficiency, hyperparathyroidism, and marked elevations in FGF23 concentration form a vicious cycle and are believed to contribute directly to cardiac remodeling. Therefore, the purpose of this article is to introduce the specific effects of the above substances on the heart and to explain the significance of understanding the vitamin D-parathyroid hormone-FGF23 axis in improving or even reversing cardiac remodeling, thus contributing to the treatment of patients with HF.
Collapse
Affiliation(s)
- Cuiyun Deng
- Special Demand Medical Care Ward, Beijing Anzhen Hospital Jilin Hospital (Changchun Central Hospital), Changchun, China
| | - Yihang Wu
- Interventional Center of Valvular Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.
| |
Collapse
|
7
|
Fitts RH, Wang X, Kwok WM, Camara AKS. Cardiomyocyte Adaptation to Exercise: K+ Channels, Contractility and Ischemic Injury. Int J Sports Med 2024; 45:791-803. [PMID: 38648799 DOI: 10.1055/a-2296-7604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Cardiovascular disease is a leading cause of morbidity and mortality, and exercise-training (TRN) is known to reduce risk factors and protect the heart from ischemia and reperfusion injury. Though the cardioprotective effects of exercise are well-documented, underlying mechanisms are not well understood. This review highlights recent findings and focuses on cardiac factors with emphasis on K+ channel control of the action potential duration (APD), β-adrenergic and adenosine regulation of cardiomyocyte function, and mitochondrial Ca2+ regulation. TRN-induced prolongation and shortening of the APD at low and high activation rates, respectively, is discussed in the context of a reduced response of the sarcolemma delayed rectifier potassium channel (IK) and increased content and activation of the sarcolemma KATP channel. A proposed mechanism underlying the latter is presented, including the phosphatidylinositol-3kinase/protein kinase B pathway. TRN induced increases in cardiomyocyte contractility and the response to adrenergic agonists are discussed. The TRN-induced protection from reperfusion injury is highlighted by the increased content and activation of the sarcolemma KATP channel and the increased phosphorylated glycogen synthase kinase-3β, which aid in preventing mitochondrial Ca2+ overload and mitochondria-triggered apoptosis. Finally, a brief section is presented on the increased incidences of atrial fibrillation associated with age and in life-long exercisers.
Collapse
Affiliation(s)
- Robert H Fitts
- Biological Sciences, Marquette University, Milwaukee, United States
| | - Xinrui Wang
- Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States
| | - Wai-Meng Kwok
- Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States
- Anesthesiology, Medical College of Wisconsin, Milwaukee, United States
- Cancer Center, Medical College of Wisconsin, Milwaukee, United States
| | - Amadou K S Camara
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States
- Anesthesiology, Medical College of Wisconsin, Milwaukee, United States
- Cancer Center, Medical College of Wisconsin, Milwaukee, United States
- Physiology, Medical College of Wisconsin, Milwaukee, United States
| |
Collapse
|
8
|
Drăgoi CM, Diaconu CC, Nicolae AC, Dumitrescu IB. Redox Homeostasis and Molecular Biomarkers in Precision Therapy for Cardiovascular Diseases. Antioxidants (Basel) 2024; 13:1163. [PMID: 39456418 PMCID: PMC11504313 DOI: 10.3390/antiox13101163] [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: 07/31/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
Precision medicine is envisioned as the future of cardiovascular healthcare, offering a more tailored and effective method for managing cardiovascular diseases compared to the traditional one-size-fits-all approaches. The complex role of oxidative stress in chronic diseases within the framework of precision medicine was carefully explored, delving into the cellular redox status and its critical involvement in the pathophysiological complexity of cardiovascular diseases (CVDs). The review outlines the mechanisms of reactive oxygen species generation and the function of antioxidants in maintaining redox balance. It emphasizes the elevated reactive oxygen species concentrations observed in heart failure and their detrimental impact on cardiovascular health. Various sources of ROS within the cardiovascular system are examined, including mitochondrial dysfunction, which contributes to oxidative stress and mitochondrial DNA degradation. The article also addresses oxidative stress's role in myocardial remodeling, a process pivotal to the progression of heart diseases. By integrating these aspects, the review underscores the importance of redox homeostasis and identifies molecular biomarkers that can enhance precision therapy for CVDs. The insights provided aim to pave the way for targeted therapeutic strategies that mitigate oxidative stress, thereby improving patient outcomes in cardiovascular medicine.
Collapse
Affiliation(s)
- Cristina Manuela Drăgoi
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania; (C.M.D.); (I.-B.D.)
| | - Camelia Cristina Diaconu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania;
- Department of Internal Medicine, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
| | - Alina Crenguța Nicolae
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania; (C.M.D.); (I.-B.D.)
| | - Ion-Bogdan Dumitrescu
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania; (C.M.D.); (I.-B.D.)
| |
Collapse
|
9
|
Podgórska D, Cieśla M, Płonka A, Bajorek W, Czarny W, Król P, Podgórski R. Changes in Circulating MicroRNA Levels as Potential Indicators of Training Adaptation in Professional Volleyball Players. Int J Mol Sci 2024; 25:6107. [PMID: 38892295 PMCID: PMC11173131 DOI: 10.3390/ijms25116107] [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: 04/17/2024] [Revised: 05/22/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
The increasing demand placed on professional athletes to enhance their fitness and performance has prompted the search for new, more sensitive biomarkers of physiological ability. One such potential biomarker includes microRNA (miRNA) small regulatory RNA sequences. The study investigated the levels of the selected circulating miRNAs before and after a 10-week training cycle in 12 professional female volleyball players, as well as their association with cortisol, creatine kinase (CK), and interleukin 6 (IL-6), using the qPCR technique. Significant decreases in the miR-22 (0.40 ± 0.1 vs. 0.28 ± 0.12, p = 0.009), miR-17 (0.35 ± 0.13 vs. 0.23 ± 0.08; p = 0.039), miR-24 (0.09 ± 0.04 vs. 0.05 ± 0.02; p = 0.001), and miR-26a (0.11 ± 0.06 vs. 0.06 ± 0.04; p = 0.003) levels were observed after training, alongside reduced levels of cortisol and IL-6. The correlation analysis revealed associations between the miRNAs' relative quantity and the CK concentrations, highlighting their potential role in the muscle repair processes. The linear regression analysis indicated that miR-24 and miR-26a had the greatest impact on the CK levels. The study provides insights into the dynamic changes in the miRNA levels during training, suggesting their potential as biomarkers for monitoring the adaptive responses to exercise. Overall, the findings contribute to a better understanding of the physiological effects of exercise and the potential use of miRNAs, especially miR-24 and miR-26a, as biomarkers in sports science and medicine.
Collapse
Affiliation(s)
- Dominika Podgórska
- Department of Internal Diseases, Institute of Medical Sciences, College of Medical Sciences, University of Rzeszow, 35-310 Rzeszow, Poland
| | - Marek Cieśla
- Institute of Medical Sciences, College of Medical Sciences, University of Rzeszow, 35-310 Rzeszow, Poland;
| | - Artur Płonka
- Institute of Physical Culture Studies, College of Medical Sciences, University of Rzeszow, 35-310 Rzeszow, Poland; (A.P.); (W.B.); (W.C.); (P.K.)
| | - Wojciech Bajorek
- Institute of Physical Culture Studies, College of Medical Sciences, University of Rzeszow, 35-310 Rzeszow, Poland; (A.P.); (W.B.); (W.C.); (P.K.)
| | - Wojciech Czarny
- Institute of Physical Culture Studies, College of Medical Sciences, University of Rzeszow, 35-310 Rzeszow, Poland; (A.P.); (W.B.); (W.C.); (P.K.)
| | - Paweł Król
- Institute of Physical Culture Studies, College of Medical Sciences, University of Rzeszow, 35-310 Rzeszow, Poland; (A.P.); (W.B.); (W.C.); (P.K.)
| | - Rafał Podgórski
- Department of Biochemistry, Institute of Medical Sciences, College of Medical Sciences, University of Rzeszow, 35-310 Rzeszow, Poland;
| |
Collapse
|
10
|
Du B, Zhang J, Kong L, Shi H, Zhang D, Wang X, Yang C, Li P, Yao R, Liang C, Wu L, Huang Z. Ovarian Tumor Domain-Containing 7B Attenuates Pathological Cardiac Hypertrophy by Inhibiting Ubiquitination and Degradation of Krüppel-Like Factor 4. J Am Heart Assoc 2023; 12:e029745. [PMID: 38084712 PMCID: PMC10863784 DOI: 10.1161/jaha.123.029745] [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: 02/13/2023] [Accepted: 08/15/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Cardiac hypertrophy (CH) is a well-established risk factor for many cardiovascular diseases and a primary cause of mortality and morbidity among older adults. Currently, no pharmacological interventions have been specifically tailored to treat CH. OTUD7B (ovarian tumor domain-containing 7B) is a member of the ovarian tumor-related protease (OTU) family that regulates many important cell signaling pathways. However, the role of OTUD7B in the development of CH is unclear. Therefore, we investigated the role of OTUD7B in CH. METHODS AND RESULTS OTUD7B knockout mice were used to assay the role of OTUD7B in CH after transverse aortic coarctation surgery. We further assayed the specific functions of OTUD7B in isolated neonatal rat cardiomyocytes. We found that OTUD7B expression decreased in hypertrophic mice hearts and phenylephrine-stimulated neonatal rat cardiomyocytes. Furthermore, OTUD7B deficiency exacerbated transverse aortic coarctation surgery-induced myocardial hypertrophy, abnormal cardiac function, and fibrosis. In cardiac myocytes, OTUD7B knockdown promoted phenylephrine stimulation-induced myocardial hypertrophy, whereas OTUD7B overexpression had the opposite effect. An immunoprecipitation-mass spectrometry analysis showed that OTUD7B directly binds to KLF4 (Krüppel-like factor 4). Additional molecular experiments showed that OTUD7B impedes KLF4 degradation by inhibiting lysine residue at 48 site-linked ubiquitination and suppressing myocardial hypertrophy by activating the serine/threonine kinase pathway. CONCLUSIONS These results demonstrate that the OTUD7B-KLF4 axis is a novel molecular target for CH treatment.
Collapse
Affiliation(s)
- Bin‐Bin Du
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Jie‐Lei Zhang
- Department of EndocrinologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Ling‐Yao Kong
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Hui‐Ting Shi
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Dian‐Hong Zhang
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Xing Wang
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Chun‐Lei Yang
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Peng‐Cheng Li
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Rui Yao
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Cui Liang
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Lei‐Ming Wu
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Zhen Huang
- Cardiovascular Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
11
|
Guo C, Wu RY, Dou JH, Song SF, Sun XL, Hu YW, Guo FS, Wei J, Lin L, Wei J. Mitophagy-dependent cardioprotection of resistance training on heart failure. J Appl Physiol (1985) 2023; 135:1390-1401. [PMID: 37942531 DOI: 10.1152/japplphysiol.00674.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/24/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023] Open
Abstract
Resistance exercise is an indispensable mode of exercise rehabilitation for heart failure. Here we elucidate the cardiac effects of resistance training alone or combined with different aerobic trainings on heart failure and explore the critical regulation of mitophagy. The chronic heart failure model was constructed by transverse aortic constriction surgery, followed by 8 wk of resistance training (RT), moderate-intensity continuous training combined with resistance training (MRT), and high-intensity interval training combined with resistance training (HRT), and subsequently analyzed the changes of maximum load, cardiac structure and function, and myocardial mitophagic activity. The role and signaling of mitophagy in exercise protection of heart failure were investigated by knockdown of Hif1α and Parkin genes in primary neonatal cardiomyocytes. RT and especially MRT improved maximum load (P < 0.0001), myocardial morphology and fibrosis (P < 0.0001), reduced left ventricular diameter and enhanced left ventricular systolic function (P < 0.01), and enhanced myocardial mitophagic activity and HIF1α expression (P < 0.05) in heart failure mice. However, HRT had no obvious protective effect on ventricular diameter and function or mitophagy. The abilities of exercise stimulation to regulate reactive oxygen species, adenosine triphosphate, and brain natriuretic peptide were impaired after knockdown of Hif1α and Parkin genes inhibited mitophagy in failing cardiomyocytes (P < 0.05). Different exercise modalities provide discrepant cardiovascular effects on heart failure, and MRT exhibits optimal protection. The HIF1α-Parkin-mitophagy pathway is involved in the protection and regulation of exercise on heart failure.NEW & NOTEWORTHY Impaired myocardial mitophagy is implicated in the pathogenesis of heart failure. Resistance training alone or combined with different aerobic trainings provide discrepant cardiovascular effects on heart failure, and the cardioprotective function depends on HIF1α-Parkin-mitophagy pathway.
Collapse
Affiliation(s)
- Chen Guo
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, People's Republic of China
| | - Rui-Yun Wu
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, People's Republic of China
| | - Jia-Hao Dou
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, People's Republic of China
| | - Shou-Fang Song
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, People's Republic of China
| | - Xue-Lu Sun
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, People's Republic of China
| | - Yi-Wei Hu
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, People's Republic of China
| | - Fan-Shun Guo
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, People's Republic of China
| | - Jia Wei
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, People's Republic of China
| | - Lin Lin
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, People's Republic of China
| | - Jin Wei
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, People's Republic of China
- Clinical Research Center for Endemic Disease of Shaanxi Province, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, People's Republic of China
| |
Collapse
|
12
|
Soares LL, Leite LB, Ervilha LOG, Pelozin BRA, Pereira NP, da Silva BAF, Portes AMO, Drummond FR, de Rezende LMT, Fernandes T, Oliveira EM, Neves MM, Reis ECC, Natali AJ. Resistance exercise training benefits pulmonary, cardiac, and muscular structure and function in rats with stable pulmonary artery hypertension. Life Sci 2023; 332:122128. [PMID: 37769805 DOI: 10.1016/j.lfs.2023.122128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/01/2023]
Abstract
AIM We tested the effects of low- to moderate-intensity resistance exercise training (RT) on the structure and function of pulmonary, right ventricle (RV), and skeletal muscle tissues in rats with stable pulmonary artery hypertension (PAH). MAIN METHODS After the first monocrotaline (MCT; 20 mg/kg) injection, male rats were submitted to a RT program (Ladder climbing; 55-65 % intensity), 5 times/week. Seven days later rats received the second MCT dose. Physical effort tolerance test and echocardiographic examination were performed. After euthanasia, lung, heart, and biceps brachii were processed for histological, single myocyte, and biochemical analysis. KEY FINDINGS RT improved survival and physical effort tolerance (i.e., maximum carrying load), mitigated the pulmonary artery resistance increase (i.e., TA/TE), and preserved cardiac function (i.e., fractional shortening, ejection fraction, stroke volume and TAPSE). RT counteracted oxidative stress (i.e., CAT, SOD, GST, MDA and NO) and adverse remodeling in lung (i.e., collapsed alveoli) and in biceps brachii (i.e., atrophy and total collagen) tissues. RT delayed RV adverse remodeling (i.e., hypertrophy, extracellular matrix, collagen types I and III, and fibrosis) and impairments in single RV myocyte contractility (i.e., amplitude and velocity to peak and relaxation). RT improved the expression of gene (i.e., miRNA 214) and intracellular Ca2+ cycling regulatory proteins (i.e., PLBser16); and of pathological (i.e., α/β-MHC and Foxo3) and physiological (i.e., Akt, p-Akt, mTOR, p-mTOR, and Bcl-xL) hypertrophy pathways markers in RV tissue. SIGNIFICANCE Low- to moderate-intensity RT benefits the structure and function of pulmonary, RV, and skeletal muscle tissues in rats with stable pulmonary artery hypertension.
Collapse
Affiliation(s)
- Leôncio Lopes Soares
- Universidade Federal de Viçosa, Departamento de Educação Física, Viçosa, Brazil.
| | | | | | | | - Noemy Pinto Pereira
- Universidade de São Paulo, Escola de Educação Física e Esportes, São Paulo, Brazil
| | | | | | | | | | - Tiago Fernandes
- Universidade de São Paulo, Escola de Educação Física e Esportes, São Paulo, Brazil
| | | | | | | | - Antônio José Natali
- Universidade Federal de Viçosa, Departamento de Educação Física, Viçosa, Brazil
| |
Collapse
|
13
|
Yusifov A, Borders MO, DeHoff MA, Polson SM, Schmitt EE, Bruns DR. Juvenile physical activity protects against isoproterenol-induced cardiac dysfunction later in life. J Appl Physiol (1985) 2023; 135:572-583. [PMID: 37439235 PMCID: PMC10538985 DOI: 10.1152/japplphysiol.00010.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 07/14/2023] Open
Abstract
Cardiovascular disease is an enormous public health problem, particularly in older populations. Exercise is the most potent cardioprotective intervention identified to date, with exercise in the juvenile period potentially imparting greater protection, given the plasticity of the developing heart. To test the hypothesis that voluntary wheel running early in life would be cardioprotective later in life when risk for disease is high, we provided male and female juvenile (3 wk old) mice access to a running wheel for 2 wk. Mice then returned to a home cage to age to adulthood (4-6 mo) before exposure to isoproterenol (ISO) to induce cardiac stress. Cardiac function and remodeling were compared with sedentary control mice, sedentary mice exposed to ISO, and mice that exercised in adulthood immediately before ISO. Early in life activity protected against ISO-induced stress as evidenced by attenuated cardiac mass, myocyte size, and fibrosis compared with sedentary mice exposed to ISO. ISO-induced changes in cardiac function were ameliorated in male mice that engaged in wheel running, with ejection fraction and fractional shortening reversed to control values. Adrenergic receptor expression was downregulated in juvenile male runners. This suppression persisted in adulthood following ISO, providing a putative mechanism by which exercise in the young male heart provides resilience to cardiac stress later in life. Together, we show that activity early in life induces persistent cardiac changes that attenuate ISO-induced stress in adulthood. Identification of the mechanisms by which early in life exercise is protective will yield valuable insights into how exercise is medicine across the life course.NEW & NOTEWORTHY Voluntary wheel running activity early in life induces persistent changes in the heart that attenuate isoproterenol-induced hypertrophy and fibrosis in adulthood. Though the mechanisms of this protection remain incompletely understood, activity-induced downregulation of adrenergic receptor expression early in life may contribute to later protection against adrenergic stress. Together these data suggest that efforts to maintain an active lifestyle early in life may have long-lasting cardioprotective benefits.
Collapse
Affiliation(s)
- Aykhan Yusifov
- Division of Kinesiology and Health, University of Wyoming, Laramie, Wyoming, United States
| | - Megan O Borders
- Division of Kinesiology and Health, University of Wyoming, Laramie, Wyoming, United States
| | - MacKenzie A DeHoff
- Division of Kinesiology and Health, University of Wyoming, Laramie, Wyoming, United States
| | - Sydney M Polson
- Division of Kinesiology and Health, University of Wyoming, Laramie, Wyoming, United States
| | - Emily E Schmitt
- Division of Kinesiology and Health, University of Wyoming, Laramie, Wyoming, United States
- WWAMI Medical Education Program, University of Wyoming, Laramie, Wyoming, United States
| | - Danielle R Bruns
- Division of Kinesiology and Health, University of Wyoming, Laramie, Wyoming, United States
- WWAMI Medical Education Program, University of Wyoming, Laramie, Wyoming, United States
| |
Collapse
|
14
|
Mannozzi J, Al-Hassan MH, Kaur J, Lessanework B, Alvarez A, Massoud L, Aoun K, Spranger M, O'Leary DS. Blood flow restriction training activates the muscle metaboreflex during low-intensity sustained exercise. J Appl Physiol (1985) 2023; 135:260-270. [PMID: 37348015 PMCID: PMC10393340 DOI: 10.1152/japplphysiol.00274.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/09/2023] [Indexed: 06/24/2023] Open
Abstract
Blood flow restriction training (BFRT) employs partial vascular occlusion of exercising muscle and has been shown to increase muscle performance while using reduced workload and training time. Numerous studies have demonstrated that BFRT increases muscle hypertrophy, mitochondrial function, and beneficial vascular adaptations. However, changes in cardiovascular hemodynamics during the exercise protocol remain unknown, as most studies measured blood pressure before the onset and after the cessation of exercise. With reduced perfusion to the exercising muscle during BFRT, the resultant accumulation of metabolites within the ischemic muscle could potentially trigger a large reflex increase in blood pressure, termed the muscle metaboreflex. At low workloads, this pressor response occurs primarily via increases in cardiac output. However, when increases in cardiac output are limited (e.g., heart failure or during severe exercise), the reflex shifts to peripheral vasoconstriction as the primary mechanism to increase blood pressure, potentially increasing the risk of a cardiovascular event. Using our chronically instrumented conscious canine model, we utilized a 60% reduction in femoral blood pressure applied to the hindlimbs during steady-state treadmill exercise (3.2 km/h) to reproduce the ischemic environment observed during BFRT. We observed significant increases in heart rate (+19 ± 3 beats/min), stroke volume (+2.52 ± 1.2 mL), cardiac output (+1.21 ± 0.2 L/min), mean arterial pressure (+18.2 ± 2.4 mmHg), stroke work (+1.93 ± 0.2 L/mmHg), and nonischemic vascular conductance (+3.62 ± 1.7 mL/mmHg), indicating activation of the muscle metaboreflex.NEW & NOTEWORTHY Blood flow restriction training (BFRT) increases muscle mass, strength, and endurance. There has been minimal consideration of the reflex cardiovascular responses that could be elicited during BFRT sessions. We showed that during low-intensity exercise BFRT may trigger large reflex increases in blood pressure and sympathetic activity due to muscle metaboreflex activation. Thus, we urge caution when employing BFRT, especially in patients in whom exaggerated cardiovascular responses may occur that could cause sudden, adverse cardiovascular events.
Collapse
Affiliation(s)
- Joseph Mannozzi
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Mohamed-Hussein Al-Hassan
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Jasdeep Kaur
- Department of Kinesiology and Health Education, University of Texas at Austin, Austin, Texas, United States
| | - Beruk Lessanework
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Alberto Alvarez
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Louis Massoud
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Kamel Aoun
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Marty Spranger
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States
| | - Donal S O'Leary
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, United States
| |
Collapse
|
15
|
Bonilla IM, Baine S, Pokrass A, Mariángelo JIE, Kalyanasundaram A, Bogdanov V, Mezache L, Sakuta G, Beard CM, Belevych A, Tikunova S, Terentyeva R, Terentyev D, Davis J, Veeraraghavan R, Carnes CA, Györke S. STIM1 ablation impairs exercise-induced physiological cardiac hypertrophy and dysregulates autophagy in mouse hearts. J Appl Physiol (1985) 2023; 134:1287-1299. [PMID: 36995910 PMCID: PMC10190841 DOI: 10.1152/japplphysiol.00363.2022] [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: 07/05/2022] [Revised: 03/13/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023] Open
Abstract
Cardiac stromal interaction molecule 1 (STIM1), a key mediator of store-operated Ca2+ entry (SOCE), is a known determinant of cardiomyocyte pathological growth in hypertrophic cardiomyopathy. We examined the role of STIM1 and SOCE in response to exercise-dependent physiological hypertrophy. Wild-type (WT) mice subjected to exercise training (WT-Ex) showed a significant increase in exercise capacity and heart weight compared with sedentary (WT-Sed) mice. Moreover, myocytes from WT-Ex hearts displayed an increase in length, but not width, compared with WT-Sed myocytes. Conversely, exercised cardiac-specific STIM1 knock-out mice (cSTIM1KO-Ex), although displaying significant increase in heart weight and cardiac dilation, evidenced no changes in myocyte size and displayed a decreased exercise capacity, impaired cardiac function, and premature death compared with sedentary cardiac-specific STIM1 knock-out mice (cSTIM1KO-Sed). Confocal Ca2+ imaging demonstrated enhanced SOCE in WT-Ex myocytes compared with WT-Sed myocytes with no measurable SOCE detected in cSTIM1KO myocytes. Exercise training induced a significant increase in cardiac phospho-Akt Ser473 in WT mice but not in cSTIM1KO mice. No differences were observed in phosphorylation of mammalian target of rapamycin (mTOR) and glycogen synthase kinase (GSK) in exercised versus sedentary cSTIM1KO mice hearts. cSTIM1KO-Sed mice showed increased basal MAPK phosphorylation compared with WT-Sed that was not altered by exercise training. Finally, histological analysis revealed exercise resulted in increased autophagy in cSTIM1KO but not in WT myocytes. Taken together, our results suggest that adaptive cardiac hypertrophy in response to exercise training involves STIM1-mediated SOCE. Our results demonstrate that STIM1 is involved in and essential for the myocyte longitudinal growth and mTOR activation in response to endurance exercise training.NEW & NOTEWORTHY Store-operated Ca2+ entry (SOCE) has been implicated in pathological cardiac hypertrophy; however, its role in physiological hypertrophy is unknown. Here we report that SOCE is also essential for physiological cardiac hypertrophy and functional adaptations in response to endurance exercise. These adaptations were associated with activation of AKT/mTOR pathway and curtailed cardiac autophagy and degeneration. Thus, SOCE is a common mechanism and an important bifurcation point for signaling paths involved in physiological and pathological hypertrophy.
Collapse
Affiliation(s)
- Ingrid M Bonilla
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
- Veterans Affairs Caribbean Healthcare System, San Juan, Puerto Rico, United States
- Department of Pharmacology and Toxicology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico, United States
| | - Stephen Baine
- Department of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States
| | - Anastasia Pokrass
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Juan Ignacio Elio Mariángelo
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Anuradha Kalyanasundaram
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart & Lung Research Institute, Columbus, Ohio, United States
| | - Vladimir Bogdanov
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Louisa Mezache
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Galina Sakuta
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Casey M Beard
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Andriy Belevych
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Svetlana Tikunova
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Radmila Terentyeva
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Dmitry Terentyev
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Jonathan Davis
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Rengasayee Veeraraghavan
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Cynthia A Carnes
- Department of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States
| | - Sandor Györke
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| |
Collapse
|
16
|
Cinato M, Mardani I, Miljanovic A, Drevinge C, Laudette M, Bollano E, Henricsson M, Tolö J, Bauza Thorbrügge M, Levin M, Lindbom M, Arif M, Pacher P, Andersson L, Olofsson CS, Borén J, Levin MC. Cardiac Plin5 interacts with SERCA2 and promotes calcium handling and cardiomyocyte contractility. Life Sci Alliance 2023; 6:e202201690. [PMID: 36717246 PMCID: PMC9887753 DOI: 10.26508/lsa.202201690] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 01/17/2023] [Accepted: 01/17/2023] [Indexed: 01/31/2023] Open
Abstract
The adult heart develops hypertrophy to reduce ventricular wall stress and maintain cardiac function in response to an increased workload. Although pathological hypertrophy generally progresses to heart failure, physiological hypertrophy may be cardioprotective. Cardiac-specific overexpression of the lipid-droplet protein perilipin 5 (Plin5) promotes cardiac hypertrophy, but it is unclear whether this response is beneficial. We analyzed RNA-sequencing data from human left ventricle and showed that cardiac PLIN5 expression correlates with up-regulation of cardiac contraction-related processes. To investigate how elevated cardiac Plin5 levels affect cardiac contractility, we generated mice with cardiac-specific overexpression of Plin5 (MHC-Plin5 mice). These mice displayed increased left ventricular mass and cardiomyocyte size but preserved heart function. Quantitative proteomics identified sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2 (SERCA2) as a Plin5-interacting protein. In situ proximity ligation assay further confirmed the Plin5/SERCA2 interaction. Live imaging showed increases in intracellular Ca2+ release during contraction, Ca2+ removal during relaxation, and SERCA2 function in MHC-Plin5 versus WT cardiomyocytes. These results identify a role of Plin5 in improving cardiac contractility through enhanced Ca2+ signaling.
Collapse
Affiliation(s)
- Mathieu Cinato
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy at University of Gothenburgand Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ismena Mardani
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy at University of Gothenburgand Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Azra Miljanovic
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy at University of Gothenburgand Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Christina Drevinge
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy at University of Gothenburgand Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Marion Laudette
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy at University of Gothenburgand Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Entela Bollano
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy at University of Gothenburgand Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Marcus Henricsson
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy at University of Gothenburgand Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Johan Tolö
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Marcos Bauza Thorbrügge
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Max Levin
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy at University of Gothenburgand Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Malin Lindbom
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy at University of Gothenburgand Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Muhammad Arif
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Linda Andersson
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy at University of Gothenburgand Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Charlotta S Olofsson
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy at University of Gothenburgand Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Malin C Levin
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy at University of Gothenburgand Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
17
|
Yang Y, Xu Y, Qian S, Tang T, Wang K, Feng J, Ding R, Yao J, Huang J, Wang J. Systematic investigation of the multi-scale mechanisms of herbal medicine on treating ventricular remodeling: Theoretical and experimental studies. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 112:154706. [PMID: 36796187 DOI: 10.1016/j.phymed.2023.154706] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 01/17/2023] [Accepted: 02/05/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND To explore the underlying molecule mechanism of herbal medicine in preventing ventricular remodeling (VR), we take a herbal formula that is clinically effective for preventing VR as an example, which composed of Pachyma hoelen Rumph, Atractylodes macrocephala Koidz., Cassia Twig and Licorice. Due to multi-components and multi-targets in herbal medicine, it is extremely difficult to systematically explain its mechanisms of action. METHODS An innovative systematic investigation framework which combines with pharmacokinetic screening, target fishing, network pharmacology, DeepDDI algorithm, computational chemistry, molecular thermodynamics, in vivo and in vitro experiments was performed for deciphering the underlying molecular mechanisms of herbal medicine for treating VR. RESULTS ADME screening and SysDT algorithm determined 75 potentially active compounds and 109 corresponding targets. Then, systematic analysis of networks reveals the crucial active ingredients and key targets in herbal medicine. Additionally, transcriptomic analysis identifies 33 key regulators during VR progression. Moreover, PPI network and biological function enrichment present four crucial signaling pathways, i.e. NF-κB and TNF, PI3K-AKT and C-type lectin receptor signaling pathways involved in VR. Besides, both molecular experiments at animal and cell levels reveal the beneficial effect of herbal medicine on preventing VR. Finally, MD simulations and binding free energy validate the reliability of drug-target interactions. CONCLUSION Our novelty is to build a systematic strategy which combines various theoretical methods combined with experimental approaches. This strategy provides a deep understanding for the study of molecular mechanisms of herbal medicine on treating diseases from systematic level, and offers a new idea for modern medicine to explore drug interventions for complex diseases as well.
Collapse
Affiliation(s)
- Yinfeng Yang
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Yuan Xu
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Shanna Qian
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Tongjuan Tang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Kangyong Wang
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Jie Feng
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Ran Ding
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Juan Yao
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Jinling Huang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China.
| | - Jinghui Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China.
| |
Collapse
|
18
|
Yuan J, Xu B, Ma J, Pang X, Fu Y, Liang M, Wang M, Pan Y, Duan Y, Tang M, Zhu B, Laher I, Li S. MOTS-c and aerobic exercise induce cardiac physiological adaptation via NRG1/ErbB4/CEBPβ modification in rats. Life Sci 2023; 315:121330. [PMID: 36584915 DOI: 10.1016/j.lfs.2022.121330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/16/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022]
Abstract
AIMS To determine the effects of the mitochondrial open reading frame of the 12S ribosomal RNA type-c (MOTS-c) and aerobic exercise on cardiac structure and function and explore the role of neuregulin-1 (NRG1)- ErbB2 receptor tyrosine kinase 4(ErbB4)- CCAAT-enhancer binding protein β (C/EBPβ) in cardiac physiological adaptation induced by MOTS-c and aerobic training. MAIN METHODS We used Hematoxylin-Eosin staining(HE)and Transmission Electron Microscope (TEM) to observe the cardiac myocardial structure, carotid artery catheterization to test cardiac function, and real-time quantitative polymerase chain reaction (qRT-PCR) and Western blotting to describe the changes of NRG1, ErbB4, C/EBPβ, and Gata in cardiac physiological adaptation. KEY FINDINGS MOTS-c and aerobic training significantly increased heart weight and heart weight index (HWI) (all p < 0.05). Aerobic exercise and MOTS-c treatment thickened myocardial fibers, with a tendency of hypertrophy. Heart rate (HR) (p < 0.001, p = 0.010, p = 0.011), the isovolumic diastolic time constant (Tau) (p < 0.001, p < 0.001, p < 0.001) in exercised (E), MOST-c treated (M) and their combination (ME) decreased significantly, while the dP/dtmax (p < 0.001, p < 0.001, p = 0.039) and dP/dtmin (p < 0.001, p < 0.001, p = 0.001) in groups E, M and ME were significantly higher than those in group C, but EDP (p = 0.903, p = 0.708, p = 0.744) remained unchanged. Moreover, C/EBPβ gene levels were significantly decreased in the differential gene expression between groups C and M transcriptomics sequencing. The levels of ErbB4 mRNA (p < 0.001, p < 0.001, p < 0.001) and Gata4 mRNA (p < 0.001, p < 0.001, p = 0.001) in groups E, M and ME increased significantly, while C/EBPβ mRNA expression decreased significantly (p < 0.001, p = 0.002, p = 0.001), which was consistent with the results of transcriptome sequencing. NRG1 mRNA in group E was significantly higher than that in group C (p = 0.003), but there was no significant difference between groups M and ME (p = 0.804, p = 0.320). The protein expression of NRG1 (p = 0.026, p < 0.001, p < 0.001), ErbB4 (p < 0.001, p < 0.001, p < 0.001) and Gata4 (p = 0.014, p < 0.001, p = 0.006) in groups E, M and ME increased significantly, while C/EBPβ decreased significantly (p < 0.001, p = 0.001, p = 0.002). SIGNIFICANCE Our findings suggest that MOTS-c and aerobic exercise had similar effects, improving myocardial morphology and structure and enhancing cardiac function through activation of the NRG1-ErbB4-C/EBPβ pathway.
Collapse
Affiliation(s)
- Jinghan Yuan
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bowen Xu
- Faculty of Science and Engineering, University of Nottingham, Ningbo 315000, China
| | - Jiacheng Ma
- Institute of Sport Medicine and Health, Chengdu Sport University, Chengdu 610041, China
| | - Xiaoli Pang
- Institute of Sport Medicine and Health, Chengdu Sport University, Chengdu 610041, China
| | - Yu Fu
- Institute of Sport Medicine and Health, Chengdu Sport University, Chengdu 610041, China
| | - Min Liang
- Institute of Sport Medicine and Health, Chengdu Sport University, Chengdu 610041, China
| | - Manda Wang
- Institute of Sport Medicine and Health, Chengdu Sport University, Chengdu 610041, China
| | - Yanrong Pan
- Institute of Sport Medicine and Health, Chengdu Sport University, Chengdu 610041, China
| | - Yimei Duan
- Institute of Sport Medicine and Health, Chengdu Sport University, Chengdu 610041, China
| | - Mi Tang
- Institute of Sport Medicine and Health, Chengdu Sport University, Chengdu 610041, China
| | - Bingmei Zhu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ismail Laher
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Shunchang Li
- Institute of Sport Medicine and Health, Chengdu Sport University, Chengdu 610041, China.
| |
Collapse
|
19
|
CircAMOTL1 RNA and AMOTL1 Protein: Complex Functions of AMOTL1 Gene Products. Int J Mol Sci 2023; 24:ijms24032103. [PMID: 36768425 PMCID: PMC9916871 DOI: 10.3390/ijms24032103] [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: 11/09/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
The complexity of the cellular proteome facilitates the control of a wide range of cellular processes. Non-coding RNAs, including microRNAs and long non-coding RNAs, greatly contribute to the repertoire of tools used by cells to orchestrate various functions. Circular RNAs (circRNAs) constitute a specific class of non-coding RNAs that have recently emerged as a widely generated class of molecules produced from many eukaryotic genes that play essential roles in regulating cellular processes in health and disease. This review summarizes current knowledge about circRNAs and focuses on the functions of AMOTL1 circRNAs and AMOTL1 protein. Both products from the AMOTL1 gene have well-known functions in physiology, cancer, and other disorders. Using AMOTL1 as an example, we illustrate how focusing on both circRNAs and proteins produced from the same gene contributes to a better understanding of gene functions.
Collapse
|
20
|
Wang B, Gan L, Deng Y, Zhu S, Li G, Nasser MI, Liu N, Zhu P. Cardiovascular Disease and Exercise: From Molecular Mechanisms to Clinical Applications. J Clin Med 2022; 11:jcm11247511. [PMID: 36556132 PMCID: PMC9785879 DOI: 10.3390/jcm11247511] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 11/29/2022] [Accepted: 12/03/2022] [Indexed: 12/23/2022] Open
Abstract
Inactivity is a significant risk factor for cardiovascular disease. Exercise may greatly enhance the metabolism and function of the cardiovascular system, lower several risk factors, and prevent the development and treatment of cardiovascular disease while delivering easy, physical, and emotional enjoyment. Exercise regulates the cardiovascular system by reducing oxidative stress and chronic inflammation, regulating cardiovascular insulin sensitivity and the body's metabolism, promoting stem cell mobilization, strengthening autophagy and myocardial mitochondrial function, and enhancing cardiovascular damage resistance, among other effects. Appropriate exercise intervention has become an essential adjuvant therapy in clinical practice for treating and rehabilitating various cardiovascular diseases. However, the prescription of exercise for preventing and treating cardiovascular diseases, particularly the precise selection of individual exercise techniques and their volume, remains controversial. Using multiomics to explain further the molecular process underlying the positive effects of exercise on cardiovascular health will not only improve our understanding of the effects of exercise on health but also establish a scientific basis and supply new ideas for preventing and treating cardiovascular diseases by activating the endogenous protective mechanisms of the body and suggesting more specific exercise prescriptions for cardiovascular rehabilitation.
Collapse
Affiliation(s)
- Bo Wang
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China; (B.W.); (L.G.); (Y.D.); (S.Z.); (G.L.)
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou 510640, China
| | - Lin Gan
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China; (B.W.); (L.G.); (Y.D.); (S.Z.); (G.L.)
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou 510640, China
| | - Yuzhi Deng
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China; (B.W.); (L.G.); (Y.D.); (S.Z.); (G.L.)
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou 510640, China
| | - Shuoji Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China; (B.W.); (L.G.); (Y.D.); (S.Z.); (G.L.)
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou 510640, China
| | - Ge Li
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China; (B.W.); (L.G.); (Y.D.); (S.Z.); (G.L.)
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou 510640, China
| | - Moussa Ide Nasser
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China; (B.W.); (L.G.); (Y.D.); (S.Z.); (G.L.)
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou 510640, China
- Correspondence: (M.I.N.); (N.L.); (P.Z.)
| | - Nanbo Liu
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China; (B.W.); (L.G.); (Y.D.); (S.Z.); (G.L.)
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou 510640, China
- Correspondence: (M.I.N.); (N.L.); (P.Z.)
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China; (B.W.); (L.G.); (Y.D.); (S.Z.); (G.L.)
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou 510640, China
- Correspondence: (M.I.N.); (N.L.); (P.Z.)
| |
Collapse
|
21
|
Wang Y, Bai L, Wen J, Zhang F, Gu S, Wang F, Yin J, Wang N. Cardiac-specific renalase overexpression alleviates CKD-induced pathological cardiac remodeling in mice. Front Cardiovasc Med 2022; 9:1061146. [PMID: 36588579 PMCID: PMC9798007 DOI: 10.3389/fcvm.2022.1061146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Introduction CKD-induced pathological cardiac remodeling is characterized by myocardial hypertrophy and cardiac fibrosis. The available therapeutic options are limited, it is thus urgently needed to identify novel therapeutic targets. Renalase (RNLS) is a newly discovered protein secreted by the kidney and was found beneficial in many renal diseases. But whether it exerts protective effects on cardiac remodeling in CKD remains unclear. Methods RNLS knockout (KO) and wild-type (WT) mice were both used to build CKD models and the adeno-associated virus (AAV9) system was used to overexpress RNLS cardiac specifically. Echocardiography was performed to detect cardiac structural changes every 6 weeks until 18 weeks post-surgery. High throughput sequencing was performed to understand the underlying mechanisms and the effects of RNLS on cardiac fibroblasts were validated in vitro. Results Knockout of RNLS aggravated cardiac remodeling in CKD, while RNLS cardiac-specific overexpression significantly reduced left ventricular hypertrophy and cardiac fibrosis induced by CKD. The following RNA-sequencing analysis revealed that RNLS significantly downregulated the extracellular matrix (ECM) receptor interaction pathway, ECM organization, and several ECM-related proteins. GSEA results showed RNLS significantly downregulated several profibrotic biological processes of cardiac fibroblasts which were upregulated by CKD, including fibroblast proliferation, leukocyte migration, antigen presentation, cytokine production, and epithelial-mesenchymal transition (EMT). In vitro, we validated that RNLS reduced the primary cardiac fibroblast proliferation and α-SMA expression stimulated by TGF-β. Conclusion In this study, we examined the cardioprotective role of RNLS in CKD-induced cardiac remodeling. RNLS may be a potential therapeutic factor that exerts an anti-fibrotic effect in pathological cardiac remodeling.
Collapse
Affiliation(s)
- Yi Wang
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Linnan Bai
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiejun Wen
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangfei Zhang
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sijie Gu
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Wang
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianyong Yin
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,*Correspondence: Jianyong Yin,
| | - Niansong Wang
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Niansong Wang,
| |
Collapse
|
22
|
Bonavida V, Ghassemi K, Ung G, Inouye K, Thankam FG, Agrawal DK. Novel Approaches to Program Cells to Differentiate into Cardiomyocytes in Myocardial Regeneration. Rev Cardiovasc Med 2022; 23:392. [PMID: 39076655 PMCID: PMC11270456 DOI: 10.31083/j.rcm2312392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/17/2022] [Accepted: 10/31/2022] [Indexed: 07/31/2024] Open
Abstract
With heart failure (HF) being one of the leading causes of hospitalization and death worldwide, multiple stem cell therapies have been attempted to accelerate the regeneration of the infarct zone. Versatile strategies have emerged to establish the cell candidates of cardiomyocyte lineage for regenerative cardiology. This article illustrates critical insights into the emerging technologies, current approaches, and translational promises on the programming of diverse cell types for cardiac regeneration.
Collapse
Affiliation(s)
- Victor Bonavida
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Kaitlyn Ghassemi
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Gwendolyn Ung
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Keiko Inouye
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Finosh G Thankam
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
23
|
Xuan R, Qiu W, Zhou Y, Magnuson JT, Luo S, Greer JB, Xu B, Liu J, Xu EG, Schlenk D, Zheng C. Parental transfer of an antibiotic mixture induces cardiotoxicity in early life-stage zebrafish: A cross-generational study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 849:157726. [PMID: 35914592 DOI: 10.1016/j.scitotenv.2022.157726] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 06/15/2023]
Abstract
Antibiotic residues in the aquatic environment have been shown to induce significant adverse effects on the early-life stage development of aquatic organisms, though the underlying molecular mechanisms of these effects have not been well characterized. In this study, we performed global mRNA-miRNA sequencing, canonical pathway analyses, morphological, physiological, immunohistochemical, and behavioral analyses to comprehensively assess the cross-generational cardiotoxicity and mechanisms of antibiotic mixtures in zebrafish. Following parental treatment to 1 and 100 μg/L antibiotic mixtures (15 of the most commonly detected antibiotics) for 150 days, all 15 assessed antibiotics were detected in the F1 eggs, indicating the cross-generational transfer of antibiotics. Global mRNA-miRNA sequencing functional analysis predicted cardiotoxicity in the F1 generation by using the F1 whole fish. Consistent with canonical pathway analyses, significant cardiotoxicity was observed in F1 larvae, as well as the apoptosis of cardiac cells. Furthermore, let-7a-5p regulated the cardiac hypertrophy signaling pathway, suggesting mechanisms of miRNA of let-7 family mediating cross-generational cardiotoxicity of antibiotics in zebrafish. This study lays some groundwork for developing interventions to prevent parental exposure to environmental pollutants such as antibiotics from adversely affecting offspring development.
Collapse
Affiliation(s)
- Rongrong Xuan
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China
| | - Wenhui Qiu
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen 518055, China; State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Yuping Zhou
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China
| | - Jason T Magnuson
- Department of Environmental Sciences, University of California, Riverside, CA 92521, United States
| | - Shusheng Luo
- State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Justin B Greer
- Department of Environmental Sciences, University of California, Riverside, CA 92521, United States
| | - Bentuo Xu
- School of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Jingyu Liu
- State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Elvis Genbo Xu
- Department of Biology, University of Southern Denmark, Odense 5230, Denmark
| | - Daniel Schlenk
- Department of Environmental Sciences, University of California, Riverside, CA 92521, United States
| | - Chunmiao Zheng
- State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
24
|
Wang Y, Liu H, He D, Zhang B, Liu Y, Xu K, Cao S, Huo Y, Liu J, Zeng L, Yan H, Dang S, Mi B. Association between physical activity and major adverse cardiovascular events in northwest China: A cross-sectional analysis from the Regional Ethnic Cohort Study. Front Public Health 2022; 10:1025670. [PMID: 36466532 PMCID: PMC9713839 DOI: 10.3389/fpubh.2022.1025670] [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: 08/23/2022] [Accepted: 11/02/2022] [Indexed: 11/18/2022] Open
Abstract
Background To examine the association between daily physical activity (PA) and major adverse cardiovascular events (MACEs) in northwest China. Methods The data in this analysis were part of the baseline survey of the Regional Ethnic Cohort Study in Northwest China from June 2018 to May 2019 in Shaanxi Province. This study used standardized self-reported total physical activity (continuous and categorical variables) and self-reported outcomes of MACEs. All analyses were conducted using the logistic regression model and stratified by age, sex, body mass index (BMI), and region. The dose-response relationships were assessed with a restricted cubic spline. Results The average level of total PA was 17.60 MET hours per day (MET-h/d). Every increase of four MET-h/d of total PA was associated with a lower risk of MACEs [adjusted OR = 0.95 (95% CI, 0.93~0.98)]. Compared with participants in the bottom quartile of total PA, a lower risk of MACEs was observed in the top quartile group [≥23.3 MET-h/d, 0.68 (0.55~0.83)]. Stratified analyses showed similar results in males, females, participants over 45 years old, participants in the rural region, and normal weight range participants (BMI < 24 kg/m2). Total participants also observed a dose-response relationship after adjusting for socioeconomic and lifestyle factors. Conclusions A higher level of PA was associated with a lower MACE risk. Future research should examine the longitudinal association of prospectively measured PA and the risk of MACEs.
Collapse
|
25
|
Chaboksafar M, Fakhr L, Kheirouri S, Alizadeh M. The effects of astaxanthin supplementation on expression of microRNAs involved in cardiovascular diseases: a systematic review of current evidence. Int J Food Sci Nutr 2022; 73:1019-1029. [DOI: 10.1080/09637486.2022.2123909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Affiliation(s)
- Maryam Chaboksafar
- Students Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Laleh Fakhr
- Students Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sorayya Kheirouri
- Department of Clinical Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Alizadeh
- Department of Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
26
|
Xue B, Yu Y, Beltz TG, Guo F, Wei SG, Johnson AK. Voluntary Exercise Eliminates Maternal Gestational Hypertension-Induced Hypertensive Response Sensitization to Postweaning High-Fat Diet in Male Adult Offspring. Hypertension 2022; 79:2016-2027. [PMID: 35730432 PMCID: PMC9378552 DOI: 10.1161/hypertensionaha.122.19608] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Exercise has profound effects on cardiovascular function and metabolism in both physiological and pathophysiological states. The present study tested whether voluntary exercise would protect male offspring against maternal gestational hypertension-induced hypertensive response sensitization elicited by post-weaning high-fat diet (HFD). METHODS AND RESULTS On low-lard-fat diet, offspring of both normotensive and hypertensive dams had comparable resting blood pressure, but HFD feeding elicited an enhanced increase in blood pressure (ie, hypertensive response sensitization) in sedentary offspring of hypertensive dams when compared with sedentary offspring of normotensive dams. The HFD fed sedentary offspring of hypertensive dams displayed greater sympathetic activity, enhanced pressor responses to centrally administered ANG II (angiotensin II) or leptin, and greater mRNA expression of proinflammatory cytokines, leptin, and a marker of blood-brain barrier leakage in the hypothalamic paraventricular nucleus. The enhanced blood pressure and central sympathetic activity in HFD-fed sedentary offspring of hypertensive dams were significantly reduced by exercise but fell only to levels comparable to HFD-fed exercising offspring of normotensive dams. HFD-induced increases in plasma IL-6 (interleukin-6) and sympathetic activity and greater pressor responses to central TNF (tumor necrosis factor)-α in offspring from both normotensive and hypertensive dams were also maintained after exercise. Nevertheless, exercise had remarkably beneficial effects on metabolic and autonomic function, brain reactivity to ANG II and leptin and gene expression of brain prohypertensive factors in all offspring. CONCLUSIONS Voluntary exercise plays a beneficial role in preventing maternal hypertension-induced hypertensive response sensitization, and that this is associated with attenuation of enhanced brain reactivity and centrally driven sympathetic activity.
Collapse
Affiliation(s)
- Baojian Xue
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
| | - Yang Yu
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Terry G. Beltz
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
| | - Fang Guo
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
| | - Shun-Guang Wei
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
- François M. Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA, USA
| | - Alan Kim Johnson
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
- Department of Health and Human Physiology, University of Iowa, Iowa City, IA, USA
- François M. Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
27
|
Zhang T, Ge J. Mechanism of CREB1 in cardiac function of rats with heart failure via regulating the microRNA-376a-3p/TRAF6 axis. Mamm Genome 2022; 33:490-501. [PMID: 35217880 DOI: 10.1007/s00335-022-09947-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: 10/28/2021] [Accepted: 01/14/2022] [Indexed: 10/19/2022]
Abstract
Heart failure (HF) is a complicated disease resulting from impaired heart function. CREB1 is a candidate target in heart-concerning diseases. This paper attempts to explore the role of CREB1 in HF. Initially, the HF rat model was established by constricted abdominal aortic surgery and the cardiac function of HF rats was assessed by ultrasonic cardiogram. Levels of CK-MB and LDH and activity of Caspase-3 and Caspase-9 in HF rats were determined. Subsequently, myocardium pathological injury and myocardium apoptosis were detected. Additionally, the interactions between CREB1 and miR-376a-3p and between miR-376a-3p and TRAF6 were verified. The roles of CREB1, miR-376a-3p, and TRAF6 in HF were evaluated. In HF rats, CREB1 and miR-376a-3p were both downregulated while TRAF6 was upregulated. Besides, HF rats had decreased values of EF and FS, elevated levels of CK-MB and LDH, inflammatory infiltration, promoted cardiomyocyte apoptosis, and elevated activity of Caspase-3 and Caspase-9, which were all reversed by CREB1. Additionally, CREB1 activated miR-376a-3p expression, and miR-376a-3p targeted TRAF6 transcription. Both miR-376a-3p knockdown and TRAF6 overexpression annulled the protective role of CREB1 overexpression in cardiac function of HF rats. CREB1 activated miR-376a-3p expression to suppress TRAF6, thereby promoting the cardiac function of HF rats.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Cardiac Surgery, The Affiliated First Hospital of USTC, No. 1 Swan Lake Road, Shushan District, Hefei, 230000, Anhui, China
| | - Jianjun Ge
- Department of Cardiac Surgery, The Affiliated First Hospital of USTC, No. 1 Swan Lake Road, Shushan District, Hefei, 230000, Anhui, China.
| |
Collapse
|
28
|
Zhou WW, Dai C, Liu WZ, Zhang C, Zhang Y, Yang GS, Guo QH, Li S, Yang HX, Li AY. Gentianella acuta improves TAC-induced cardiac remodelling by regulating the Notch and PI3K/Akt/FOXO1/3 pathways. Biomed Pharmacother 2022; 154:113564. [PMID: 35988427 DOI: 10.1016/j.biopha.2022.113564] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/10/2022] [Accepted: 08/14/2022] [Indexed: 12/18/2022] Open
Abstract
Cardiac remodelling mainly manifests as excessive myocardial hypertrophy and fibrosis, which are associated with heart failure. Gentianella acuta (G. acuta) is reportedly effective in cardiac protection; however, the mechanism by which it protects against cardiac remodelling is not fully understood. Here, we discuss the effects and mechanisms of G. acuta in transverse aortic constriction (TAC)-induced cardiac remodelling in rats. Cardiac function was analysed using echocardiography and electrocardiography. Haematoxylin and eosin, Masson's trichrome, and wheat germ agglutinin staining were used to observe pathophysiological changes. Additionally, real-time quantitative reverse transcription polymerase chain reaction and western blotting were used to measure protein levels and mRNA levels of genes related to myocardial hypertrophy and fibrosis. Immunofluorescence double staining was used to investigate the co-expression of endothelial and interstitial markers. Western blotting was used to estimate the expression and phosphorylation levels of the regulatory proteins involved in autophagy and endothelial-mesenchymal transition (EndMT). The results showed that G. acuta alleviated cardiac dysfunction and remodelling. The elevated levels of myocardial hypertrophy and fibrosis markers, induced by TAC, decreased significantly after G. acuta intervention. G. acuta decreased the expression of LC3 II and Beclin1, and increased p62 expression. G. acuta upregulated the expression of CD31 and vascular endothelial-cadherin, and prevented the expression of α-smooth muscle actin and vimentin. Furthermore, G. acuta inhibited the PI3K/Akt/FOXO1/3a pathway and activated the Notch signalling. These findings demonstrated that G. acuta has cardioprotective effects, such as alleviating myocardial fibrosis, inhibiting hypertrophy, reducing autophagy, and blocking EndMT by regulating the PI3K/Akt/FOXO1/3a and Notch signalling pathways.
Collapse
Affiliation(s)
- Wei-Wei Zhou
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei, China
| | - Cheng Dai
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei, China
| | - Wei-Zhe Liu
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei, China; Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Shijiazhuang 050091, Hebei, China
| | - Chuang Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei, China
| | - Yu Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei, China
| | - Gao-Shan Yang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei, China; Hebei Key Laboratory of Chinese Medicine Research on Cardio-cerebrovascular Disease, Shijiazhuang 050091, Hebei, China
| | - Qiu-Hong Guo
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei, China
| | - Si Li
- Department of Technology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Hong-Xia Yang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei, China.
| | - Ai-Ying Li
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei, China; Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Shijiazhuang 050091, Hebei, China; Hebei Key Laboratory of Chinese Medicine Research on Cardio-cerebrovascular Disease, Shijiazhuang 050091, Hebei, China.
| |
Collapse
|
29
|
Yin A, Yuan R, Xiao Q, Zhang W, Xu K, Yang X, Yang W, Xu L, Wang X, Zhuang F, Li Y, Cai Z, Sun Z, Zhou B, He B, Shen L. Exercise-derived peptide protects against pathological cardiac remodeling. EBioMedicine 2022; 82:104164. [PMID: 35843176 PMCID: PMC9297110 DOI: 10.1016/j.ebiom.2022.104164] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 11/22/2022] Open
Abstract
Background Exercise training protects the heart against pathological cardiac remodeling and confers cardioprotection from heart failure. However, the underlying mechanism is still elusive. Methods An integrative analysis of multi-omics data of the skeletal muscle in response to exercise is performed to search for potential exerkine. Then, CCDC80tide is examined in humans after acute exercise. The role of CCDC80tide is assessed in a mouse model of hypertensive cardiac remodeling and in hypertension-mediated cell injury models. The transcriptomic analysis and immunoprecipitation assay are conducted to explore the mechanism. Findings The coiled-coil domain-containing protein 80 (CCDC80) is found strongly positively associated with exercise. Interestingly, exercise stimuli induce the secretion of C-terminal CCDC80 (referred as CCDC80tide hereafter) via EVs-encapsulated CCDC80tide into the circulation. Importantly, cardiac-specific expression of CCDC80tide protects against angiotensin II (Ang II)-induced cardiac hypertrophy and fibrosis in mice. In in vitro studies, the expression of CCDC80tide reduces Ang II-induced cardiomyocyte hypertrophy, cardiac microvascular endothelial cell (CMEC) inflammation, and mitigated vascular smooth muscle cell (VSMC) proliferation and collagen formation. To understand the cardioprotective effect of CCDC80tide, a transcriptomic analysis reveals a dramatic inhibition of the STAT3 (Signal transducer and activator of transcription 3) signaling pathway in CCDC80tide overexpressing cells. Mechanistically, CCDC80tide selectively interacts with the kinase-active form of JAK2 (Janus kinase 2) and consequently inhibits its kinase activity to phosphorylate and activate STAT3. Interpretation The results provide new insights into exercise-afforded cardioprotection in pathological cardiac remodeling and highlight the therapeutic potential of CCDC80tide in heart failure treatment. Funding This work was supported by the National Natural Science Foundation of China [Grant/Award Numbers: 81770428, 81830010, 82130012, 81900438, 82100447); Shanghai Science and Technology Committee [Grant/Award Numbers: 21S11903000, 19JC1415702]; Emerging and Advanced Technology Programs of Hospital Development Center of Shanghai [Grant/Award Number: SHDC12018129]; China Postdoctoral Science Foundation [2021M692108]; and China National Postdoctoral Program for Innovative Talents [BX20200211].
Collapse
Affiliation(s)
- Anwen Yin
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Ruosen Yuan
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Qingqing Xiao
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Weifeng Zhang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Ke Xu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xiaoxiao Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Wentao Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Lei Xu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xia Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Fei Zhuang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Yi Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhaohua Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhe Sun
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Bin Zhou
- Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| |
Collapse
|
30
|
Luft C, da Costa MS, Antunes GL, de Oliveira JR, Donadio MVF. The role of maternal exercise on placental, behavioral and genetic alterations induced by prenatal stress. Neurochem Int 2022; 158:105384. [PMID: 35787396 DOI: 10.1016/j.neuint.2022.105384] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/03/2022] [Accepted: 06/27/2022] [Indexed: 12/11/2022]
Abstract
The present study aimed to evaluate the effects of treadmill maternal exercise on alterations induced by prenatal stress in neonatal mice. Female and male Balb/c mice were divided into five groups: control (CON), prenatal restraint stress (PNS), prenatal restraint stress and physical exercise before pregnancy (PNS + EX1), prenatal restraint stress and physical exercise during pregnancy (PNS + EX2), and prenatal restraint stress and physical exercise before and during pregnancy (PNS + EX3). Exercise was performed using a treadmill, at a speed of 10 m/min, for 60 minutes, 5 days a week. Maternal behavior was assessed on days 3, 4 and 5 postpartum (PPD). Placental gene expression of glucocorticoid receptor (GR), 11-β-hydroxysteroid dehydrogenase 2 (11β-HSD2), 5-hydroxytryptamine receptor 1A (5HT1AR), and corticotropin releasing hormone receptor 1 (CRHR1) were analyzed. In neonatal mice, the gene expression of GR, mineralocorticoid receptor (MR), CRHR1, 5HTr1, oxytocin Receptor 1 (OXTr1), tropomyosin related kinase B (TRκB), brain-derived neurotrophic factor exon I (BDNF I), and BDNF IV was analyzed in the brain (PND0) and hippocampus (PND10). Maternal exercise improved (p < 0.05) maternal care. In the placenta, maternal exercise prevented (p < 0.01) the increase in GR expression caused by PNS. In the brain from PND0, exercise before pregnancy prevented (p = 0.002) the decreased CRHR1 expression promoted by PNS. In the hippocampus of PND10 males, PNS decreased (p = 0.0005) GR expression, and exercise before pregnancy prevented (p = 0.003) this effect. In PND10 females, maternal exercise prevented (p < 0.05) the PNS-induced increase in MR expression. PNS + EX2 males showed increased (p < 0.01) BDNF I gene expression and PNS + EX1 females demonstrated increased (p = 0.03) BDNF IV expression. In conclusion, maternal physical exercise may play a role in modulating maternal-fetal health and may contribute to preventing neurodevelopmental changes induced by prenatal stress.
Collapse
Affiliation(s)
- Carolina Luft
- Laboratory of Pediatric Physical Activity, Infant Center, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Laboratory of Cellular Biophysics and Inflammation, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Mariana Severo da Costa
- Laboratory of Pediatric Physical Activity, Infant Center, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Laboratory of Cellular Biophysics and Inflammation, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Géssica Luana Antunes
- Laboratory of Cellular Biophysics and Inflammation, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Jarbas Rodrigues de Oliveira
- Laboratory of Cellular Biophysics and Inflammation, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Márcio Vinícius Fagundes Donadio
- Laboratory of Pediatric Physical Activity, Infant Center, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Laboratory of Cellular Biophysics and Inflammation, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Department of Physiotherapy, Facultad de Medicina y Ciencias de la Salud, Universitat Internacional de Catalunya (UIC), Barcelona, Spain.
| |
Collapse
|
31
|
Schmitt EE, McNair BD, Polson SM, Cook RF, Bruns DR. Mechanisms of Exercise-Induced Cardiac Remodeling Differ Between Young and Aged Hearts. Exerc Sport Sci Rev 2022; 50:137-144. [PMID: 35522248 PMCID: PMC9203913 DOI: 10.1249/jes.0000000000000290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aging induces physiological and molecular changes in the heart that increase the risk for heart disease. Several of these changes are targetable by exercise. We hypothesize that the mechanisms by which exercise improves cardiac function in the aged heart differ from those in the young exercised heart.
Collapse
Affiliation(s)
| | - Benjamin D McNair
- Division of Kinesiology & Health, University of Wyoming, Laramie, WY
| | - Sydney M Polson
- Division of Kinesiology & Health, University of Wyoming, Laramie, WY
| | - Ross F Cook
- Division of Kinesiology & Health, University of Wyoming, Laramie, WY
| | | |
Collapse
|
32
|
Nerve growth factor and post-infarction cardiac remodeling. ACTA BIOMEDICA SCIENTIFICA 2022. [DOI: 10.29413/abs.2022-7.2.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The prevalence of sudden death from chronic heart failure and cardiac arrhythmias caused by myocardial infarction is a complex problem in cardiology. Post-infarction cardiac remodeling occurs after myocardial infarction. This compensatory-adaptive reaction, regulated by mechanical, neurohumoral and genetic factors, includes the structural and functional changes of cardiomyocytes, stromal elements and extracellular matrix, geometry and architectonics of the left ventricular cavity. Adverse left ventricular remodeling is associated with heart failure and increased mortality. The concept of post-infarction cardiac remodeling is an urgent problem, since the mechanisms of development and progression of adverse post-infarction changes in the myocardium are completely unexplored. In recent years, the scientist attention has been focused on neurotrophic factors involved in the sympathetic nervous system and the vascular system remodeling after myocardial infarction. Nerve growth factor (NGF) is a protein from the neurotrophin family that is essential for the survival and development of sympathetic and sensory neurons, which also plays an important role in vasculogenesis. Acute myocardial infarction and heart failure are characterized by changes in the expression and activity of neurotrophic factors and their receptors, affecting the innervation of the heart muscle, as well as having a direct effect on cardiomyocytes, endothelial and smooth muscle vascular cells. The identification of the molecular mechanisms involved in the interactions between cardiomyocytes and neurons, as well as the study of the effects of NGF in the cardiovascular system, will improve understanding of the cardiac remodeling mechanism. This review summarizes the available scientific information (2019–2021) about mechanisms of the link between post-infarction cardiac remodeling and NGF functions.
Collapse
|
33
|
Tumor Necrosis Factor-α-Induced Protein 8-Like 2 Ameliorates Cardiac Hypertrophy by Targeting TLR4 in Macrophages. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9469143. [PMID: 35528518 PMCID: PMC9072033 DOI: 10.1155/2022/9469143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/04/2022] [Indexed: 11/28/2022]
Abstract
Background Tumor necrosis factor-α-induced protein 8-like 2 (TIPE2), a novel immunoregulatory protein, has been reported to regulate inflammation and apoptosis. The role of TIPE2 in cardiovascular disease, especially cardiac hypertrophy, has not been elucidated. Thus, the aim of the present study was to explore the role of TIPE2 in cardiac hypertrophy. Methods Mice were subjected to aortic banding (AB) to induce an adverse hypertrophic model. To overexpress TIPE2, mice were injected with a lentiviral vector expressing TIPE2. Echocardiographic and hemodynamic analyses were used to evaluate cardiac function. Neonatal rat cardiomyocytes (NRCMs) and mouse peritoneal macrophages (MPMs) were isolated and stimulated with angiotensin II. NRCMs and MPM were also cocultured and stimulated with angiotensin II. Cells were transfected with Lenti-TIPE2 to overexpress TIPE2. Results TIPE2 expression levels were downregulated in hypertrophic mouse hearts and in macrophages in heart tissue. TIPE2 overexpression attenuated pressure overload-induced cardiac hypertrophy, fibrosis, and cardiac dysfunction. Moreover, we found that TIPE2 overexpression in neonatal cardiomyocytes did not relieve the angiotensin II-induced hypertrophic response in vitro. Furthermore, TIPE2 overexpression downregulated TLR4 and NF-κB signaling in macrophages but not in cardiomyocytes, which led to diminished inflammation in macrophages and consequently reduced the activation of hypertrophic Akt signaling in cardiomyocytes. TLR4 inhibition by TAK-242 did not enhance the antihypertrophic effect of TIPE2 overexpression. Conclusions The present study indicated that TIPE2 represses macrophage activation by targeting TLR4, subsequently inhibiting cardiac hypertrophy.
Collapse
|
34
|
Chaulin AM. Metabolic Pathway of Cardiospecific Troponins: From Fundamental Aspects to Diagnostic Role (Comprehensive Review). Front Mol Biosci 2022; 9:841277. [PMID: 35517866 PMCID: PMC9062030 DOI: 10.3389/fmolb.2022.841277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/28/2022] [Indexed: 11/28/2022] Open
Abstract
Many molecules of the human body perform key regulatory functions and are widely used as targets for the development of therapeutic drugs or as specific diagnostic markers. These molecules undergo a significant metabolic pathway, during which they are influenced by a number of factors (biological characteristics, hormones, enzymes, etc.) that can affect molecular metabolism and, as a consequence, the serum concentration or activity of these molecules. Among the most important molecules in the field of cardiology are the molecules of cardiospecific troponins (Tns), which regulate the processes of myocardial contraction/relaxation and are used as markers for the early diagnosis of ischemic necrosis of cardiomyocytes (CMC) in myocardial infarction (MI). The diagnostic value and diagnostic capabilities of cardiospecific Tns have changed significantly after the advent of new (highly sensitive (HS)) detection methods. Thus, early diagnostic algorithms of MI were approved for clinical practice, thanks to which the possibility of rapid diagnosis and determination of optimal tactics for managing patients with MI was opened. Relatively recently, promising directions have also been opened for the use of cardiospecific Tns as prognostic markers both at the early stages of the development of cardiovascular diseases (CVD) (arterial hypertension (AH), heart failure (HF), coronary heart disease (CHD), etc.), and in non-ischemic extra-cardiac pathologies that can negatively affect CMC (for example, sepsis, chronic kidney disease (CKD), chronic obstructive pulmonary disease (COPD), etc.). Recent studies have also shown that cardiospecific Tns are present not only in blood serum, but also in other biological fluids (urine, oral fluid, pericardial fluid, amniotic fluid). Thus, cardiospecific Tns have additional diagnostic capabilities. However, the fundamental aspects of the metabolic pathway of cardiospecific Tns are definitively unknown, in particular, specific mechanisms of release of Tns from CMC in non-ischemic extra-cardiac pathologies, mechanisms of circulation and elimination of Tns from the human body, mechanisms of transport of Tns to other biological fluids and factors that may affect these processes have not been established. In this comprehensive manuscript, all stages of the metabolic pathway are consistently and in detail considered, starting from release from CMC and ending with excretion (removal) from the human body. In addition, the possible diagnostic role of individual stages and mechanisms, influencing factors is analyzed and directions for further research in this area are noted.
Collapse
Affiliation(s)
- Aleksey M. Chaulin
- Department of Cardiology and Cardiovascular Surgery, Department of Clinical Chemistry, Samara State Medical University, Samara, Russia
- Samara Regional Clinical Cardiological Dispensary, Samara, Russia
| |
Collapse
|
35
|
Preventive aerobic training preserves sympathovagal function and improves DNA repair capacity of peripheral blood mononuclear cells in rats with cardiomyopathy. Sci Rep 2022; 12:6422. [PMID: 35440673 PMCID: PMC9018832 DOI: 10.1038/s41598-022-09361-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 03/03/2022] [Indexed: 11/09/2022] Open
Abstract
To evaluate the effect of preventive aerobic exercise training on sympathovagal function, cardiac function, and DNA repair capacity in a preclinical model of doxorubicin (DOX)-induced cardiomyopathy. Forty male Wistar-Kyoto rats were allocated into four groups (n = 10/group): D (DOX-treated) and C (controls) remained sedentary, and DT (DOX-trained) and CT (control-trained) performed aerobic training 4 days/week, during 4 weeks before exposure to DOX (4 mg/kg/week during 4 weeks) or saline solution. We evaluated cardiac function (echocardiography), hemodynamic and sympathovagal modulation (artery-femoral cannulation), cardiac troponin T levels, and DNA repair capacity (comet assay). Exercise training preserved ejection fraction (D: − 14.44% vs. DT: − 1.05%, p < 0.001), fractional shortening (D: − 8.96% vs. DT: − 0.27%, p = 0.025) and troponin T levels (D: 6.4 ± 3.6 vs. DT: 2.8 ± 1.7 ng/mL, p = 0.010). DOX increased heart rate variability (C: 27.7 ± 7.9 vs. D: 7.5 ± 2.2 ms2, p < 0.001) and induced sympathovagal dysfunction (LF/HF, C: 0.37 ± 0.15 vs. D: 0.15 ± 0.15, p = 0.036) through exacerbation of sympathetic function (LF, C: 0.22 ± 0.01 vs. D: 0.48 ± 0.24 Hz, p = 0.019). Peripheral mononuclear blood cells of DT animals presented lower residual DNA damage (D: 43.4 ± 8.4% vs. DT: 26 ± 3.4%, p = 0.003 after 1 h). Cardioprotective effects of preventive aerobic exercise training are mediated by preservation of sympathovagal function and improvement of DNA repair capacity of peripheral blood mononuclear cells.
Collapse
|
36
|
The Importance of Cardiac Troponin Metabolism in the Laboratory Diagnosis of Myocardial Infarction (Comprehensive Review). BIOMED RESEARCH INTERNATIONAL 2022; 2022:6454467. [PMID: 35402607 PMCID: PMC8986381 DOI: 10.1155/2022/6454467] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 01/02/2023]
Abstract
The study of the metabolism of endogenous molecules is not only of great fundamental significance but also of high practical importance, since many molecules serve as drug targets and/or biomarkers for laboratory diagnostics of diseases. Thus, cardiac troponin molecules have long been used as the main biomarkers for confirmation of diagnosis of myocardial infarction, and with the introduction of high-sensitivity test methods, many of our ideas about metabolism of these cardiac markers have changed significantly. In clinical practice, there are opening new promising diagnostic capabilities of cardiac troponins, the understanding and justification of which are closely connected with the fundamental principles of the metabolism of these molecules. Our current knowledge about the metabolism of cardiac troponins is insufficient and extremely disconnected from various literary sources. Thus, many researchers do not sufficiently understand the potential importance of cardiac troponin metabolism in the laboratory diagnosis of myocardial infarction. The purpose of this comprehensive review is to systematize information about the metabolism of cardiac troponins and during the discussion to focus on the potential impact of cTns metabolism on the laboratory diagnosis of myocardial infarction. The format of this comprehensive review includes a sequential consideration and analysis of the stages of the metabolic pathway, starting from possible release mechanisms and ending with elimination mechanisms. This will allow doctors and researchers to understand the significant importance of cTns metabolism and its impact on the laboratory diagnosis of myocardial infarction.
Collapse
|
37
|
Chaulin AM. Biology of Cardiac Troponins: Emphasis on Metabolism. BIOLOGY 2022; 11:429. [PMID: 35336802 PMCID: PMC8945489 DOI: 10.3390/biology11030429] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/28/2022] [Accepted: 03/07/2022] [Indexed: 02/07/2023]
Abstract
Understanding of the biology of endo- and exogenous molecules, in particular their metabolism, is not only of great theoretical importance, but also of high practical significance, since many molecules serve as drug targets or markers for the laboratory diagnostics of many human diseases. Thus, cardiac troponin (cTns) molecules have long been used as key markers for the confirmation of diagnosis of myocardial infarction (MI), and with the introduction of contemporary (high sensitivity) test methods, many of our concepts related to the biology of these cardiac markers have changed significantly. In current clinical practice, there are opening new promising diagnostic capabilities of cTns, the understanding and justification of which is closely connected with the theoretical principles of the metabolism of these molecules. However, today, the biology and metabolism of cTns have not been properly investigated; in particular, we do not know the precise mechanisms of release of these molecules from the myocardial cells (MCs) of healthy people and the mechanisms of circulation, and the elimination of cTns from the bloodstream. The main purpose of this manuscript is to systematize information about the biology of cTns, with an emphasis on the metabolism of cTns. The format of this paper, starting with the release of cTns in the blood and concluding with the metabolism/filtration of troponins, provides a comprehensive yet logically easy way for the readers to approach our current knowledge in the framework of understanding the basic mechanisms by which cTns are produced and processed. Conclusions. Based on the analysis of the current literature, the important role of biology and all stages of metabolism (release, circulation, removal) of cTns in laboratory diagnostics should be noted. It is necessary to continue studying the biology and metabolism of cTns, because this will improve the differential diagnosis of MI and i a new application of cTns immunoassays in current clinical practice.
Collapse
Affiliation(s)
- Aleksey M Chaulin
- Department of Histology and Embryology, Samara State Medical University, 89 Chapaevskaya Street, Samara Region, 443099 Samara, Russia
- Department of Cardiology and Cardiovascular Surgery, Samara State Medical University, 89 Chapaevskaya Street, Samara Region, 443099 Samara, Russia
| |
Collapse
|
38
|
De Sousa RAL, Improta-Caria AC. Regulation of microRNAs in Alzheimer´s disease, type 2 diabetes, and aerobic exercise training. Metab Brain Dis 2022; 37:559-580. [PMID: 35075500 DOI: 10.1007/s11011-022-00903-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia. The evolution and aggregation of amyloid beta (β) oligomers is linked to insulin resistance in AD, which is also the major characteristic of type 2 diabetes (T2D). Being physically inactive can contribute to the development of AD and/or T2D. Aerobic exercise training (AET), a type of physical exercise, can be useful in preventing or treating the negative outcomes of AD and T2D. AD, T2D and AET can regulate the expression of microRNAs (miRNAs). Here, we review some of the changes in miRNAs expression regulated by AET, AD and T2D. MiRNAs play an important role in the gene regulation of key signaling pathways in both pathologies, AD and T2D. MiRNA dysregulation is evident in AD and has been associated with several neuropathological alterations, such as the development of a reactive gliosis. Expression of miRNAs are associated with many pathophysiological mechanisms involved in T2D like insulin synthesis, insulin resistance, glucose intolerance, hyperglycemia, intracellular signaling, and lipid profile. AET regulates miRNAs levels. We identified 5 miRNAs (miR-21, miR-29a/b, miR-103, miR-107, and miR-195) that regulate gene expression and are modulated by AET on AD and T2D. The identified miRNAs are potential targets to treat the symptoms of AD and T2D. Thus, AET is a non-pharmacological tool that can be used to prevent and fight the negative outcomes in AD and T2D.
Collapse
Affiliation(s)
- Ricardo Augusto Leoni De Sousa
- Programa Multicêntrico de Pós-Graduação Em Ciências Fisiológicas- Sociedade Brasileira de Fisiologia (SBFis), Universidade Federal Dos Vales Do Jequitinhonha E Mucuri (UFVJM), Campus JK, Rodovia MGT 367, Km 583, Alto da Jacuba, nº 5000, Diamantina, Minas Gerais, CEP 39100-000, Brazil.
| | - Alex Cleber Improta-Caria
- Post-Graduate Program in Medicine and Health, Faculty of Medicine, Federal University of Bahia, Bahia, Brazil
| |
Collapse
|
39
|
Chaulin A. Metabolic Pathway of Cardiac Troponins and Its Diagnostic Value. Vasc Health Risk Manag 2022; Volume 18:153-180. [DOI: 10.2147/vhrm.s335851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
|
40
|
Extracellular Vesicles and Thrombogenicity in Atrial Fibrillation. Int J Mol Sci 2022; 23:ijms23031774. [PMID: 35163695 PMCID: PMC8836440 DOI: 10.3390/ijms23031774] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 01/30/2022] [Accepted: 02/02/2022] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) are defined as a heterogenic group of lipid bilayer vesicular structures with a size in the range of 30–4000 nm that are released by all types of cultured cells. EVs derived from platelets, mononuclears, endothelial cells, and adipose tissue cells significantly increase in several cardiovascular diseases, including in atrial fibrillation (AF). EVs are engaged in cell-to-cell cooperation, endothelium integrity, inflammation, and immune response and are a cargo for several active molecules, such as regulatory peptides, receptors, growth factors, hormones, and lipids. Being transductors of the intercellular communication, EVs regulate angiogenesis, neovascularization, coagulation, and maintain tissue reparation. There is a large amount of evidence regarding the fact that AF is associated with elevated levels of EVs derived from platelets and mononuclears and a decreased number of EVs produced by endothelial cells. Moreover, some invasive procedures that are generally performed for the treatment of AF, i.e., pulmonary vein isolation, were found to be triggers for elevated levels of platelet and mononuclear EVs and, in turn, mediated the transient activation of the coagulation cascade. The review depicts the role of EVs in thrombogenicity in connection with a risk of thromboembolic complications, including ischemic stroke and systemic thromboembolism, in patients with various forms of AF.
Collapse
|
41
|
Xu Y, Gao H, Du Z, Liu H, Cheng Q, Zhang F, Ye J, Wang A, Dou Y, Ma B, Zhao N, Zhu F, Xu X, Shen N, Wu J, Xue B. A new approach for reducing pollutants level: a longitudinal cohort study of physical exercises in young people. BMC Public Health 2022; 22:223. [PMID: 35114971 PMCID: PMC8812347 DOI: 10.1186/s12889-022-12621-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 01/21/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The present study aimed to evaluate the elimination of three common pollutants (dimethoate, benzo(a)pyrene (BaP) and bisphenol A (BPA) by different physical exercises and to assess the possible factors which could affect the pollutants elimination. METHODS A total of 200 individuals who chose different kinds of exercises in accordance to their own wish were recruited. The levels of urinary pollutants were measured using β-glucuronidase hydrolysis followed by a high-performance liquid chromatography tandem mass spectrometry-based method. RESULTS Totally, the levels of dimethoate, BaP and BPA were reduced after physical exercises. However, the elimination of BaP in male was higher than that in female but the elimination of BPA in female was higher than that in male. Multivariate logistic regression showed that the degree of heart rate (HR) change was a protective factor affecting the improvement effect of dimethoate, BaP and BPA while BMI (body mass index) was a risk factor. Nevertheless, sex was a risk factor affecting the improvement of dimethoate and BaP but had a lower efficacy on BPA improvement. CONCLUSION The present findings indicate that physical exercises can be considered as a novel approach to eliminate pollutants level in human body and can also give suggestions for choosing specific physical exercises to male and female individuals. Moreover, those who are with higher BMI need to lose weight before eliminating pollutant level through physical exercises.
Collapse
Affiliation(s)
- Yujuan Xu
- Hohai University, Nanjing, 210098, China
| | - Hongliang Gao
- Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, China
| | - Zhixiang Du
- Department of Infectious Diseases, Taizhou People's Hospital, Taizhou, 225300, China
| | - He Liu
- General surgery department, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, China
| | - Qi Cheng
- Medical School of Nanjing University, Nanjing, 210093, China
| | - Furong Zhang
- Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, China
| | - Juan Ye
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | | | - Yanjun Dou
- Hohai University, Nanjing, 210098, China
| | - Bei Ma
- Hohai University, Nanjing, 210098, China
| | - Ningwei Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Feng Zhu
- General surgery department, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, China
| | - Xianlin Xu
- Department of Urology, Sir Run Run Hospital, Nanjing Medical University, 109 Longmian Road, Jiangning, Nanjing, 211100, Jiangsu, China
| | - Ning Shen
- China Exposomics Institute (CEI) Precision Medicine Co. Ltd, Shanghai, 200120, China
| | - Jing Wu
- Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, China
| | - Bin Xue
- Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
42
|
Hong JH, Zhang HG. Transcription Factors Involved in the Development and Prognosis of Cardiac Remodeling. Front Pharmacol 2022; 13:828549. [PMID: 35185581 PMCID: PMC8849252 DOI: 10.3389/fphar.2022.828549] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/14/2022] [Indexed: 01/09/2023] Open
Abstract
To compensate increasing workload, heart must work harder with structural changes, indicated by increasing size and changing shape, causing cardiac remodeling. However, pathological and unlimited compensated cardiac remodeling will ultimately lead to decompensation and heart failure. In the past decade, numerous studies have explored many signaling pathways involved in cardiac remodeling, but the complete mechanism of cardiac remodeling is still unrecognized, which hinders effective treatment and drug development. As gene transcriptional regulators, transcription factors control multiple cellular activities and play a critical role in cardiac remodeling. This review summarizes the regulation of fetal gene reprogramming, energy metabolism, apoptosis, autophagy in cardiomyocytes and myofibroblast activation of cardiac fibroblasts by transcription factors, with an emphasis on their potential roles in the development and prognosis of cardiac remodeling.
Collapse
|
43
|
Pretreatment of Nicorandil Protects the Heart from Exhaustive Exercise-Induced Myocardial Injury in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7550872. [PMID: 35035509 PMCID: PMC8758261 DOI: 10.1155/2022/7550872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 11/02/2021] [Accepted: 11/12/2021] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Nicorandil has been widely used for the treatment of angina pectoris and myocardial infarction. The purpose of this study was to investigate whether nicorandil plays a protective role in exhaustive exercise (EE)-induced myocardial injury. METHODS Here, we applied the rat EE model and treated them with exercise preconditioning (EP, reported to protect the heart) or different doses of nicorandil gavage, respectively, to explore whether there are protective effects of single EP or nicorandil or a combination of both and the potential mechanism. Forty-nine male Sprague Dawley rats were randomly divided into control, EE, EP + EE, nicorandil (with low, middle, and high dose) + EE, and EP + nicorandil (middle dose) + EE. Blood samples and myocardial tissues were collected to analyze the myocardial injury-related index. RESULTS EE induced myocardial structural damage and altered the myocardial injury markers, which were partially reversed by pretreatment of nicorandil. In addition, oxidative stress and inflammation lead to the accumulation of reactive oxygen species (ROS) products and further damage to the myocardium, while pretreatment of nicorandil reduces the oxidative stress response and inflammation. Moreover, nicorandil suppressed the myocardial apoptosis induced by EE, as indicated by a decrease of Bax and caspase-3 expression and an increase of Bcl-2 expression. Finally, the pathway in which nicorandil plays a role may be involved in the endothelial nitric oxide synthase (eNOS)/nitric oxide (NO) pathway. Pretreatment of nicorandil increased the protein level of myocardial eNOS and NO production. CONCLUSION Our result demonstrated that nicorandil has protective effects in EE-induced myocardial injury with dose-dependent effects. A combination of nicorandil and EP can further improve the protective effects. Taken together, nicorandil can be potentially used as an intervention method in EE-induced myocardial injury.
Collapse
|
44
|
Ghanemi A, Yoshioka M, St-Amand J. Exercise, Diet and Sleeping as Regenerative Medicine Adjuvants: Obesity and Ageing as Illustrations. MEDICINES (BASEL, SWITZERLAND) 2022; 9:medicines9010007. [PMID: 35049940 PMCID: PMC8778846 DOI: 10.3390/medicines9010007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 12/21/2022]
Abstract
Regenerative medicine uses the biological and medical knowledge on how the cells and tissue regenerate and evolve in order to develop novel therapies. Health conditions such as ageing, obesity and cancer lead to an impaired regeneration ability. Exercise, diet choices and sleeping pattern have significant impacts on regeneration biology via diverse pathways including reducing the inflammatory and oxidative components. Thus, exercise, diet and sleeping management can be optimized towards therapeutic applications in regenerative medicine. It could allow to prevent degeneration, optimize the biological regeneration and also provide adjuvants for regenerative medicine.
Collapse
Affiliation(s)
- Abdelaziz Ghanemi
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (A.G.); (M.Y.)
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada
| | - Mayumi Yoshioka
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (A.G.); (M.Y.)
| | - Jonny St-Amand
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (A.G.); (M.Y.)
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada
- Correspondence: ; Tel.: +1-418-654-2296
| |
Collapse
|
45
|
Li Q, Zhang J, Liu S, Zhang F, Zhuang J, Chen Y. MicroRNA-17-3p is upregulated in psoriasis and regulates keratinocyte hyperproliferation and pro-inflammatory cytokine secretion by targeting <em>CTR9</em>. Eur J Histochem 2022; 66. [PMID: 35016493 PMCID: PMC8764465 DOI: 10.4081/ejh.2022.3275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 12/31/2021] [Indexed: 11/23/2022] Open
Abstract
Psoriasis is a chronic inflammatory skin disease. Although miRNAs are reported to be associated with the pathogenesis of psoriasis, the contribution of individual microRNAs toward psoriasis remains unclear. The miR-17-92 cluster regulates cell growth and immune functions that are associated with psoriasis. miR-17-3p is a member of miR-17-92 cluster; however, its role in dermatological diseases remains unclear. Our study aims at investigating the effects of miR-17-3p and its potential target gene on keratinocytes proliferation and secretion of pro-inflammatory cytokine and their involvement in psoriasis. Initially, we found that miR-17-3p was upregulated in psoriatic skin lesions, and bioinformatic analyses suggested that CTR9 is likely to be a target gene of miR-17-3p. Quantitative reverse-transcriptase PCR and immunohistochemical analysis revealed that CTR9 expression was downregulated in psoriatic lesions. Using dual-luciferase reporter assays, we identified CTR9 as a direct target of miR-17-3p. Further functional experiments demonstrated that miR-17-3p promoted the proliferation and pro-inflammatory cytokine secretion of keratinocytes, whereas CTR9 exerted the opposite effects. Gain-of-function studies confirmed that CTR9 suppression partially accounted for the effects of miR- 17-3p in keratinocytes. Furthermore, Western blot revealed that miR-17-3p activates the downstream STAT3 signaling pathway while CTR9 inactivates the STAT3 signaling pathway. Together, these findings indicate that miR-17-3p regulates keratinocyte proliferation and pro-inflammatory cytokine secretion partially by targeting the CTR9, which inactivates the downstream STAT3 protein, implying that miR-17-3p might be a novel therapeutic target for psoriasis.
Collapse
Affiliation(s)
- Qingwen Li
- Dermatology Hospital, Southern Medical University, Guangzhou.
| | - Jiao Zhang
- Dermatology Hospital, Southern Medical University, Guangzhou.
| | - Shougang Liu
- Dermatology Hospital, Southern Medical University, Guangzhou.
| | - Fangfei Zhang
- Dermatology Hospital, Southern Medical University, Guangzhou.
| | - Jiayi Zhuang
- Dermatology Hospital, Southern Medical University, Guangzhou.
| | - Yongfeng Chen
- Dermatology Hospital, Southern Medical University, Guangzhou.
| |
Collapse
|
46
|
Bordignon C, dos Santos BS, Rosa DD. Impact of Cancer Cachexia on Cardiac and Skeletal Muscle: Role of Exercise Training. Cancers (Basel) 2022; 14:cancers14020342. [PMID: 35053505 PMCID: PMC8773522 DOI: 10.3390/cancers14020342] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/27/2021] [Accepted: 01/07/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Cachexia is a syndrome that can be present in many patients diagnosed with cancer, especially in those with metastatic or very advanced tumors. The patient may present with weight loss, loss of muscle mass, and even cardiac dysfunction as a result of it. The aim of this review is to understand how cachexia manifests and whether physical exercise has any role in trying to prevent or reverse this syndrome in cancer patients. Abstract Cachexia is a multifactorial syndrome that presents with, among other characteristics, progressive loss of muscle mass and anti-cardiac remodeling effect that may lead to heart failure. This condition affects about 80% of patients with advanced cancer and contributes to worsening patients’ tolerance to anticancer treatments and to their premature death. Its pathogenesis involves an imbalance in metabolic homeostasis, with increased catabolism and inflammatory cytokines levels, leading to proteolysis and lipolysis, with insufficient food intake. A multimodal approach is indicated for patients with cachexia, with the aim of reducing the speed of muscle wasting and improving their quality of life, which may include nutritional, physical, pharmacologic, and psychological support. This review aims to outline the mechanisms of muscle loss, as well as to evaluate the current clinical evidence of the use of physical exercise in patients with cachexia.
Collapse
Affiliation(s)
- Cláudia Bordignon
- Oncology Center, Hospital Moinhos de Vento, Porto Alegre 90560-030, Brazil;
- Graduate Program in Pathology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-070, Brazil
| | - Bethânia S. dos Santos
- Department of Clinical Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro 20560-121, Brazil;
- Rede D’Or São Luiz, Rio de Janeiro 22271-110, Brazil
| | - Daniela D. Rosa
- Oncology Center, Hospital Moinhos de Vento, Porto Alegre 90560-030, Brazil;
- Graduate Program in Pathology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-070, Brazil
- Brazilian Breast Cancer Study Group (GBECAM), Porto Alegre 90619-900, Brazil
- Correspondence:
| |
Collapse
|
47
|
Nijholt KT, Sánchez-Aguilera PI, Voorrips SN, de Boer RA, Westenbrink BD. Exercise: a molecular tool to boost muscle growth and mitochondrial performance in heart failure? Eur J Heart Fail 2021; 24:287-298. [PMID: 34957643 PMCID: PMC9302125 DOI: 10.1002/ejhf.2407] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/15/2021] [Accepted: 12/22/2021] [Indexed: 11/30/2022] Open
Abstract
Impaired exercise capacity is the key symptom of heart failure (HF) and is associated with reduced quality of life and higher mortality rates. Unfortunately, current therapies, although generally lifesaving, have only small or marginal effects on exercise capacity. Specific strategies to alleviate exercise intolerance may improve quality of life, while possibly improving prognosis as well. There is overwhelming evidence that physical exercise improves performance in cardiac and skeletal muscles in health and disease. Unravelling the mechanistic underpinnings of exercise‐induced improvements in muscle function could provide targets that will allow us to boost exercise performance in HF. With the current review we discuss: (i) recently discovered signalling pathways that govern physiological muscle growth as well as mitochondrial quality control mechanisms that underlie metabolic adaptations to exercise; (ii) the mechanistic underpinnings of exercise intolerance in HF and the benefits of exercise in HF patients on molecular, functional and prognostic levels; and (iii) potential molecular therapeutics to improve exercise performance in HF. We propose that novel molecular therapies to boost adaptive muscle growth and mitochondrial quality control in HF should always be combined with some form of exercise training.
Collapse
Affiliation(s)
- Kirsten T Nijholt
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Pablo I Sánchez-Aguilera
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Suzanne N Voorrips
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - B Daan Westenbrink
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
48
|
Li D, Wang P, Wei W, Wang C, Zhong Y, Lv L, Wang J. Serum MicroRNA Expression Patterns in Subjects After the 5-km Exercise Are Strongly Associated With Cardiovascular Adaptation. Front Physiol 2021; 12:755656. [PMID: 34912238 PMCID: PMC8667031 DOI: 10.3389/fphys.2021.755656] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/01/2021] [Indexed: 12/02/2022] Open
Abstract
Circulating microRNAs (miRNAs) have been reported dysregulated during exercise. However, the changes of specific serum miRNAs during the 5-km run test with intensity of 51–52% maximum oxygen uptake (V̇O2max) and their association with traditional cardiovascular-related indicators remain well-characterized. Levels of miR-1, miR-21, miR-146a, miR-155, miR-181, and miR-210 were detected in 120 young subjects before and after the exercise training by quantitative reverse-transcription PCR (RT-qPCR). Besides, the levels of cardiac troponin I (cTNI), myoglobin (Myo), creatine kinase (CK), creatine kinase-MB (CK-MB), aspartate aminotransferase (AST), lactate dehydrogenase (LDH), ischemia-modified albumin (IMA), interleukin-6 (IL-6), and C-reactive protein (CRP) were measured and the correlation between levels of serum miRNAs and biochemical parameters was also analyzed. Compared with resting state, the serum levels of miR-1, miR-146a, miR-155, miR-181, and miR-210 were significantly increased after exercise training. Serum levels of miR-146a, miR-155, and miR-210 after exercise training were positively correlated with Myo, CK-MB, and LDH, respectively, while miR-1, miR-146a, miR-181, and miR-155 were positively correlated with the levels of IL-6. Additionally, all the five miRNAs were negatively correlated with IMA levels. The multivariate logistic regression analysis showed that high levels of miR-146a, AST, LDH, and IL-6 in serum were risk factors, while low IMA contents were a protective factor for cardiovascular adaptation during exercise. In conclusion, the dynamic changes of miRNAs under the condition of the 5-km continuous running contribute to the adaptive regulation of the cardiovascular function of the body.
Collapse
Affiliation(s)
- Dandan Li
- Department of Clinical Laboratory, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Pingping Wang
- Department of Clinical Laboratory, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Wenyan Wei
- Department of Clinical Laboratory, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Cheng Wang
- Department of Clinical Laboratory, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yong Zhong
- Department of Health Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Lei Lv
- Department of Geriatric Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Junjun Wang
- Department of Clinical Laboratory, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| |
Collapse
|
49
|
Sanchis-Gomar F, Arnau-Moyano M, Daimiel L, Lippi G, Leischik R, Vallecillo N, Yvert T, Jiménez SL, Santiago C, Pareja-Galeano H. Circulating microRNAs fluctuations in exercise-induced cardiac remodeling: A systematic review. Am J Transl Res 2021; 13:13298-13309. [PMID: 35035676 PMCID: PMC8748080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/30/2021] [Indexed: 06/14/2023]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that participate in gene expression regulation. It has been observed that circulating levels of miRNAs may fluctuate during exercise, showing numerous cardiac biological and physiological effects such as structural and functional adaptations. We aimed to provide an overview of the currently available information concerning the role of circulating miRNAs in cardiovascular adaptations in response to acute and/or chronic exercise training. Relevant studies published were searched in three databases: PubMed, Web of Science and Scopus. A combination of the following keywords was used: ("microRNA" OR "miRNA" OR "miR" AND "exercise" OR "training" OR "physical activity") AND "(heart hypertrophy" OR "cardiac remodeling" OR "cardiac muscle mass" OR "cardiac hypertrophy"). Only experimental studies, written in English and conducted in healthy individuals were included. Five articles met the inclusion criteria and were finally included in this systematic review after reviewing both title, abstract and full-text. A total of thirty-six circulating cardiac-related miRNAs were analyzed, but only five of them (miR-1, miR-133a, miR-146a, miR-206 and miR-221) were directly associated with cardiac adaptations parameters, while two of them (miR-1 and miR-133a) were related to cardiac hypertrophy. Most of them were upregulated immediately after a marathon and returned to basal levels at longer times. Therefore, we conclude that, although evidence is still limited, and long-term studies are needed to obtain more robust evidence, exercise is more likely to affect circulating cardiac-related miRNAs levels.
Collapse
Affiliation(s)
- Fabian Sanchis-Gomar
- Department of Physiology, Faculty of Medicine, University of Valencia and INCLIVA Biomedical Research InstituteValencia, Spain
| | | | - Lidia Daimiel
- Nutritional Control of The Epigenome Group, Instituto Madrileño de Estudios Avanzados (IMDEA) Food, CEI UAM+CSICMadrid 28049, Spain
| | - Giuseppe Lippi
- Section of Clinical Biochemistry, University of VeronaVerona, Italy
| | - Roman Leischik
- School of Medicine, Faculty of Health, Witten/Herdecke UniversityWitten, Germany
| | - Néstor Vallecillo
- Faculty of Biomedical and Health Sciences, Universidad Europea de MadridMadrid, Spain
| | - Thomas Yvert
- Faculty of Sport Sciences, Universidad Europea de MadridMadrid, Spain
| | - Sergio L Jiménez
- Centre for Sport Studies, Rey Juan Carlos UniversityMadrid, Spain
| | - Catalina Santiago
- Faculty of Sport Sciences, Universidad Europea de MadridMadrid, Spain
| | - Helios Pareja-Galeano
- Department of Physical Education, Sport and Human Movement, Universidad Autónoma de MadridMadrid, Spain
| |
Collapse
|
50
|
Ageing and Obesity Shared Patterns: From Molecular Pathogenesis to Epigenetics. Diseases 2021; 9:diseases9040087. [PMID: 34940025 PMCID: PMC8700721 DOI: 10.3390/diseases9040087] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/23/2021] [Accepted: 11/28/2021] [Indexed: 12/22/2022] Open
Abstract
In modern societies, ageing and obesity represent medical challenges for healthcare professionals and caregivers. Obesity and ageing share common features including the related cellular and molecular pathways as well as the impacts they have as risk factors for a variety of diseases and health problems. Both of these health problems also share exercise and a healthy lifestyle as the best therapeutic options. Importantly, ageing and obesity also have common epigenetic changes (histone modification, DNA methylation, noncoding RNAs, and chromatin remodeling) that are also impacted by exercise. This suggests that epigenetic pathways are among the mechanisms via which exercise induces its benefits, including ageing and obesity improvements. Exploring these interrelations and based on the fact that both ageing and obesity represent risk factors for each other, would lead to optimizing the available therapeutic approaches towards improved obesity management and healthy ageing.
Collapse
|