51
|
Jasińska-Stroschein M. Training programs in preclinical studies. The example of pulmonary hypertension. Systematic review and meta-analysis. PLoS One 2022; 17:e0276875. [PMCID: PMC9665399 DOI: 10.1371/journal.pone.0276875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
Background
Exercise and cardiopulmonary exercise testing are essential in the evaluation of physiological, biochemical, and molecular responses in the experimental studies on chronic diseases such as diabetes, heart failure and hypertension. The exercise tolerance and seem to be a valuable contribution to the experiments that are performed in animal models of pulmonary hypertension (PH), as well. The current survey uses detailed quantitative analyses to assess the advantages of exercise training programs performed in preclinical studies based on outcomes such as exercise capacity, cardiopulmonary hemodynamics, and mortality.
Methods
Articles were identified through search engines in the online electronic databases Pubmed/Medline, Web of Science following the PRISMA Protocol. Studies conducted between 1991 and 2022 without language restrictions were included in this study. Heterogeneity was assessed using the Cochrane Q-test and I2 test statistics. Subgroup analysis was employed with evidence of heterogeneity. Quality assessment was carried out using SYRCLE’s risk of bias tool for animal studies. Publication bias across studies was determined using the funnel plot and Egger’s regression test statistic.
Results
The available protocols typically included treadmill running, swimming, and voluntary wheel running with a different series of intensities, times and durations; these were also used in studies examining the efficacy of chronic training programs. In 66 interventions, PH induction reduced exercise endurance by half compared to healthy subjects, while exposure to tested medical agents normalized exercise capacity. The other 58 interventions demonstrated the advantages of various exercise training programs for PH. Induction of PH reduced exercise endurance by half compared to healthy subjects (R = 0.52; 0.48 − 0.55 95%CI; P<0.0001; I2 = 98.9%), while the exposure to tested medical agents normalized exercise capacity (R = 1.75; 1.61 − 1.91 95%CI; P<0.0001; I2 = 97.8%).
Conclusion
Despite a wide spectrum of study protocols to measure exercise endurance in animals with PH, there is a significant correlation between worsening of exercise-related parameters and PH development, manifested by alterations in haemodynamic and remodeling parameters. Familiarization with exercise, training program schedule, method used for PH induction, or detailed training parameters such as slope, exercise intensity or individualization, can influence the final outcome. This in turn can impact on the diversity and reproducibility of results being obtained in particular experimental studies.
Collapse
|
52
|
Riyono A, Tinduh D, Othman Z, Herawati L. Moderate intensity continuous and interval training affect visceral fat and insulin resistance model in female rat exposed high calorie diet. COMPARATIVE EXERCISE PHYSIOLOGY 2022. [DOI: 10.3920/cep220013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Chronic high-calorie diet (HCD) combined with physical inactivity promotes obesity and insulin resistance (IR). This study aimed to analyse the comparable effect of moderate-intensity continuous training (MICT) and moderate-intensity interval training (MIIT) on visceral fat weight and IR in subjects exposed to HCD. This randomised post-test research used only a control group design with female rats (Wistar norvegicus), 8 weeks old and 100-200 g of bodyweight. They were randomly divided into four groups: standard diet group (C), HCD group (C1), HCD combined with MICT group (C2) and HCD combined with MIIT group (C3). Each group consisted of six rats. HCD consisted of ad libitum standard diet plus dextrose solution by oral gavage for 4 weeks. The MICT was conducted by swimming plus 6% load of body weight for 10 min in the first week, for 20 min in the second week and 30 min in the third and fourth week. The MIIT was conducted by swimming in a ratio between swimming and rest time at 2:1 plus 6% load of BW, performed 5×/week for 4 weeks, and increased progressively. The mean body weight pre-intervention was 152.79±13.280 g and 150.12±9.195 g post-intervention (P=0.115). The mean fasting blood glucose pre-intervention was 79±8.668 mg/dl, and post-intervention 86.29±12.142 mg/dl (P=0.142). The mean visceral fat weight between C (1.94±0.66 g), C1 (1.45±0.47 g), C2 (1.41±0.44 g), and C3 (1.22±0.59 g) was not significant (P=0.179). The mean triglyceride level for C (173.33±30.30 mg/dl), C1 (157.16±47.32 mg/dl), C2 (112.83±25.49 mg/dl), and C3 (80.33±23.47 mg/dl) was significant (P=0.000). The mean IR model for C (4.796±0.070), C1 (4.728±0.125), C2 (4.620±0.123), C3 (4.360±0.143) was significant (P=0.000). In conclusion, both MICT and MIIT have an effect to improve IR and TG. The MIIT was more effective to improve IR compared to MICT in the female rats exposed to an HCD.
Collapse
Affiliation(s)
- A. Riyono
- Master Program of Basic Medicine Science, Faculty of Medicine, Universitas Airlangga, Jalan Mayjend Prof. Dr. Moestopo No. 4-8, Surabaya 60131, Indonesia
| | - D. Tinduh
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Universitas Airlangga, Jalan Mayjend Prof. Dr. Moestopo No. 4-8, Surabaya 60131, Indonesia
| | - Z. Othman
- Faculty of Health and Life Science, Management and Science University, Shah Alam, Selangor 40100, Malaysia
| | - L. Herawati
- Departement of Physiology, Faculty of Medicine, Universitas Airlangga, Jl Prof Dr Moestopo 47, Surabaya 60131, Indonesia
| |
Collapse
|
53
|
Borzykh AA, Gaynullina DK, Shvetsova AA, Kiryukhina OO, Kuzmin IV, Selivanova EK, Nesterenko AM, Vinogradova OL, Tarasova OS. Voluntary wheel exercise training affects locomotor muscle, but not the diaphragm in the rat. Front Physiol 2022; 13:1003073. [PMID: 36388097 PMCID: PMC9643685 DOI: 10.3389/fphys.2022.1003073] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/14/2022] [Indexed: 12/07/2024] Open
Abstract
Introduction: Functional tests and training regimens intensity-controlled by an individual are used in sport practice, clinical rehabilitation, and space medicine. The model of voluntary wheel running in rats can be used to explore molecular mechanisms of such training regimens in humans. Respiratory and locomotor muscles demonstrate diverse adaptations to treadmill exercise, but the effects of voluntary exercise training on these muscle types have not been compared yet. Therefore, this work aimed at the effects of voluntary ET on rat triceps brachii and diaphragm muscles with special attention to reactive oxygen species, which regulate muscle plasticity during exercise. Methods: Male Wistar rats were distributed into exercise trained (ET) and sedentary (Sed) groups. ET group had free access to running wheels, running activity was continuously recorded and analyzed using the original hardware/software complex. After 8 weeks, muscle protein contents were studied using Western blotting. Results: ET rats had increased heart ventricular weights but decreased visceral/epididymal fat weights and blood triglyceride level compared to Sed. The training did not change corticosterone, testosterone, and thyroid hormone levels, but decreased TBARS content in the blood. ET rats demonstrated higher contents of OXPHOS complexes in the triceps brachii muscle, but not in the diaphragm. The content of SOD2 increased, and the contents of NOX2 and SOD3 decreased in the triceps brachii muscle of ET rats, while there were no such changes in the diaphragm. Conclusion: Voluntary wheel running in rats is intensive enough to govern specific adaptations of muscle fibers in locomotor, but not respiratory muscle.
Collapse
Affiliation(s)
- Anna A. Borzykh
- State Research Center of the Russian Federation, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Dina K. Gaynullina
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | | | - Oxana O. Kiryukhina
- Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia
| | - Ilya V. Kuzmin
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | | | - Alexey M. Nesterenko
- Federal Center of Brain Research and Biotechnologies FMBA, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Olga L. Vinogradova
- State Research Center of the Russian Federation, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Olga S. Tarasova
- State Research Center of the Russian Federation, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
54
|
Bartra C, Jager LA, Alcarraz A, Meza-Ramos A, Sangüesa G, Corpas R, Guasch E, Batlle M, Sanfeliu C. Antioxidant Molecular Brain Changes Parallel Adaptive Cardiovascular Response to Forced Running in Mice. Antioxidants (Basel) 2022; 11:1891. [PMID: 36290614 PMCID: PMC9598430 DOI: 10.3390/antiox11101891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/14/2022] [Accepted: 09/19/2022] [Indexed: 10/03/2023] Open
Abstract
Physically active lifestyle has huge implications for the health and well-being of people of all ages. However, excessive training can lead to severe cardiovascular events such as heart fibrosis and arrhythmia. In addition, strenuous exercise may impair brain plasticity. Here we investigate the presence of any deleterious effects induced by chronic high-intensity exercise, although not reaching exhaustion. We analyzed cardiovascular, cognitive, and cerebral molecular changes in young adult male mice submitted to treadmill running for eight weeks at moderate or high-intensity regimens compared to sedentary mice. Exercised mice showed decreased weight gain, which was significant for the high-intensity group. Exercised mice showed cardiac hypertrophy but with no signs of hemodynamic overload. No morphological changes in the descending aorta were observed, either. High-intensity training induced a decrease in heart rate and an increase in motor skills. However, it did not impair recognition or spatial memory, and, accordingly, the expression of hippocampal and cerebral cortical neuroplasticity markers was maintained. Interestingly, proteasome enzymatic activity increased in the cerebral cortex of all trained mice, and catalase expression was significantly increased in the high-intensity group; both first-line mechanisms contribute to maintaining redox homeostasis. Therefore, physical exercise at an intensity that induces adaptive cardiovascular changes parallels increases in antioxidant defenses to prevent brain damage.
Collapse
Affiliation(s)
- Clara Bartra
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Lars Andre Jager
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), 08036 Barcelona, Spain
| | - Anna Alcarraz
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Arrhythmia Unit, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Aline Meza-Ramos
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Arrhythmia Unit, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Gemma Sangüesa
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Arrhythmia Unit, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Rubén Corpas
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Eduard Guasch
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Arrhythmia Unit, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red-Cardiovascular (CIBERCV), 28029 Madrid, Spain
| | - Montserrat Batlle
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Arrhythmia Unit, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red-Cardiovascular (CIBERCV), 28029 Madrid, Spain
| | - Coral Sanfeliu
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| |
Collapse
|
55
|
但 晴, 白 婧, 蔡 钟, 林 琨, 李 泱. [Changes of myocardial calcium currents in rats with myocardial injury induced by running exercise during acute hypoxia]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:1359-1366. [PMID: 36210709 PMCID: PMC9550543 DOI: 10.12122/j.issn.1673-4254.2022.09.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the changes in myocardial calcium currents in rats subjected to forced running exercise during acute hypoxia and their association with myocardial injury. METHODS Forty SD rats were randomized into quiescent group and running group either in normal oxygen (NQ and NR groups, respectively) or in acute hypoxia (HQ and HR groups, respectively). Hypoxia was induced by keeping the rats in a hypobaric oxygen chamber (PaO2=61.6kpa) for 4 h a day; the rats in the two running groups were forced to run on running wheels for 4 h each day. Rat ventricular myocytes was isolated by enzymatic digestion for recording action potentials and currents using patch clamp technique, and confocal Ca2+ imaging was used to monitor intracellular Ca2+ levels. The expressions of Cav1.2 channel and the cardiac ryanodine receptor (RyR2) were determined using Western blotting. RESULTS Compared with those in NQ group, the rats in HR group showed significantly decreased SOD activity (P < 0.01), increased h-FABP, hs-CRP and IMA levels (P < 0.05 or 0.01), obvious myocardial pathology, and prolonged APD50 and APD90 (P < 0.05). Of the different stress conditions, forced running in acute hypoxia resulted in the most prominent increase of the densities of ICa, L currents, causing also a significant left shift of the steady state activation curve and a significant right shift of the steady state inactivation curve. Compared with those in NQ group, the rats in NR, HQ and HR groups all exhibited higher rates of spontaneous calcium wave events in the cardiac myocytes, increased frequency of calcium sparks with lowered amplitude, enhanced calcium release amplitude in the ventricular myocytes, and delayed calcium ion reabsorption; in particular, these changes were the most conspicuous in HR group (P < 0.05 or 0.01). There was also a significant increase in the protein levels of Cav1.2 channel and RyR2 receptor in HR group (P < 0.05 or 0.01). CONCLUSIONS The mechanism of myocardial injury in rats subjected to forced running in acute hypoxia may involve the increase of oxidative stress and calcium current and intracellular calcium overload.
Collapse
Affiliation(s)
- 晴 但
- 解放军总医院第一医学中心心血管内科,北京 100039Department of Cardiology, Fist Medical Center, Chinese PLA General Hospital, Beijing 100000, China
| | - 婧 白
- 郑州大学第一附属医院心血管内科,河南 郑州 450052Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - 钟奇 蔡
- 中国人民解放军南部战区总医院干部病房,广东 广州 510014Cadre's Ward, General Hospital of Southern Theater Command of PLA, Guangzhou 510000, China
| | - 琨 林
- 解放军总医院第一医学中心心血管内科,北京 100039Department of Cardiology, Fist Medical Center, Chinese PLA General Hospital, Beijing 100000, China
| | - 泱 李
- 解放军总医院第六医学中心心血管 病医学部,北京 100048Department of Cardiology, Sixth Medical Center, Chinese PLA General Hospital, Beijing 100048, China
| |
Collapse
|
56
|
Ivonin AG, Smirnova SL, Roshchevskaya IM. Body Surface Potential Mapping during Ventricular Depolarization in Rats after Acute Exhaustive Exercise. Arq Bras Cardiol 2022; 119:S0066-782X2022005014203. [PMID: 36102423 PMCID: PMC9750213 DOI: 10.36660/abc.20211058] [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: 12/30/2021] [Revised: 04/03/2022] [Accepted: 06/01/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Exhaustive physical exercise can cause substantial changes in the electrical properties of the myocardium. OBJECTIVE To evaluate, using body surface potential mapping, the electrical activity of the heart in rats during ventricular depolarization after acute exhaustive exercise. METHODS Twelve-week-old male rats were submitted to acute treadmill exercise at 36 m/min until exhaustion. Unipolar electrocardiograms (ECGs) from the torso surface were recorded in zoletil-anesthetized rats three to five days before (Pre-Ex), 5 and 10 minutes after exhaustive exercise (Post-Ex 5 and Post-Ex 10, respectively) simultaneously with ECGs in limb leads. The instantaneous body surface potential maps (BSPMs) were analyzed during ventricular depolarization. P values <0.05 were considered statistically significant. RESULTS Compared with Pre-Ex, an early completion of the second inversion of potential distributions, an early completion of ventricular depolarization, as well as a decrease in the duration of the middle phase and the total duration of ventricular depolarization on BSPMs were revealed at Post-Ex 5. Also, compared with Pre-Ex, an increase in the amplitude of negative BSPM extremum at the R-wave peak on the ECG in lead II (RII-peak) and a decrease in the amplitude of negative BSPM extremum at 3 and 4 ms after RII-peak were showed at Post-Ex 5. At Post-Ex 10, parameters of BSPMs did not differ from those at Pre-Ex. CONCLUSION In rats, acute exhaustive exercise causes reversible changes in the temporal and amplitude characteristics of BSPMs during ventricular depolarization, most likely related to alterations in the excitation of the main mass of the ventricular myocardium.
Collapse
Affiliation(s)
- Alexey G. Ivonin
- Department of Comparative CardiologyKomi Scientific Centre of the Ural BranchRussian Academy of SciencesSyktyvkarFederação Russa Department of Comparative Cardiology – Komi Scientific Centre of the Ural Branch of the Russian Academy of Sciences , Syktyvkar – Federação Russa
| | - Svetlana L. Smirnova
- Department of Comparative CardiologyKomi Scientific Centre of the Ural BranchRussian Academy of SciencesSyktyvkarFederação Russa Department of Comparative Cardiology – Komi Scientific Centre of the Ural Branch of the Russian Academy of Sciences , Syktyvkar – Federação Russa
| | - Irina M. Roshchevskaya
- Department of Comparative CardiologyKomi Scientific Centre of the Ural BranchRussian Academy of SciencesSyktyvkarFederação Russa Department of Comparative Cardiology – Komi Scientific Centre of the Ural Branch of the Russian Academy of Sciences , Syktyvkar – Federação Russa
- Laboratory of Pharmacological ScreeningResearch Zakusov Institute of PharmacologyMoscowFederação Russa Laboratory of Pharmacological Screening – Research Zakusov Institute of Pharmacology , Moscow – Federação Russa
| |
Collapse
|
57
|
Ivonin AG, Smirnova SL, Roshchevskaya IM. Heart Electrical Activity during Ventricular Repolarization in Rats after Acute Exhaustive Treadmill Running. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022050313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
58
|
Chen H, Chen C, Spanos M, Li G, Lu R, Bei Y, Xiao J. Exercise training maintains cardiovascular health: signaling pathways involved and potential therapeutics. Signal Transduct Target Ther 2022; 7:306. [PMID: 36050310 PMCID: PMC9437103 DOI: 10.1038/s41392-022-01153-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/22/2022] [Accepted: 08/12/2022] [Indexed: 11/09/2022] Open
Abstract
Exercise training has been widely recognized as a healthy lifestyle as well as an effective non-drug therapeutic strategy for cardiovascular diseases (CVD). Functional and mechanistic studies that employ animal exercise models as well as observational and interventional cohort studies with human participants, have contributed considerably in delineating the essential signaling pathways by which exercise promotes cardiovascular fitness and health. First, this review summarizes the beneficial impact of exercise on multiple aspects of cardiovascular health. We then discuss in detail the signaling pathways mediating exercise's benefits for cardiovascular health. The exercise-regulated signaling cascades have been shown to confer myocardial protection and drive systemic adaptations. The signaling molecules that are necessary for exercise-induced physiological cardiac hypertrophy have the potential to attenuate myocardial injury and reverse cardiac remodeling. Exercise-regulated noncoding RNAs and their associated signaling pathways are also discussed in detail for their roles and mechanisms in exercise-induced cardioprotective effects. Moreover, we address the exercise-mediated signaling pathways and molecules that can serve as potential therapeutic targets ranging from pharmacological approaches to gene therapies in CVD. We also discuss multiple factors that influence exercise's effect and highlight the importance and need for further investigations regarding the exercise-regulated molecules as therapeutic targets and biomarkers for CVD as well as the cross talk between the heart and other tissues or organs during exercise. We conclude that a deep understanding of the signaling pathways involved in exercise's benefits for cardiovascular health will undoubtedly contribute to the identification and development of novel therapeutic targets and strategies for CVD.
Collapse
Affiliation(s)
- Huihua Chen
- School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chen Chen
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Michail Spanos
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Rong Lu
- School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Yihua Bei
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China. .,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China.
| | - Junjie Xiao
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China. .,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
59
|
Suarez PZ, Natali AJ, Mill JG, de Rezende LMT, Soares LL, Drummond FR, Cardoso LCC, Reis ECC, Lavorato VN, Carneiro-Júnior MA. Effects of moderate-continuous and high-intensity interval aerobic training on cardiac function of spontaneously hypertensive rats. Exp Biol Med (Maywood) 2022; 247:1691-1700. [PMID: 35880885 PMCID: PMC9597206 DOI: 10.1177/15353702221110823] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The aim of this study was to verify the effects of moderate-intensity continuous (MICT) and high-intensity interval (HIIT) aerobic training on cardiac morphology and function and the mechanical properties of single cardiomyocytes in spontaneously hypertensive rats (SHR) in the compensated phase of hypertension. Sixteen-week-old male SHR and normotensive Wistar (WIS) rats were allocated to six groups of six animals each: SHR CONT or WIS CONT (control); SHR MICT or WIS MICT (underwent MICT, 30 min/day, five days per week for eight weeks); and SHR HIIT or WIS HIIT (underwent HIIT, 30 min/day, five days per week for eight weeks). Total exercise time until fatigue and maximum running speed were determined using a maximal running test before and after the experimental period. Systolic (SAP), diastolic (DAP), and mean (MAP) blood pressures were measured using tail plethysmography before and after the experimental period. Echocardiographic evaluations were performed at the end of the experimental period. The rats were euthanized after in vivo assessments, and left ventricular myocytes were isolated to evaluate global intracellular Ca2+ transient ([Ca2+]i) and contractile function. Cellular measurements were performed at basal temperature (~37°C) at 3, 5, and 7 Hz. The results showed that both training programs increased total exercise time until fatigue and, consequently, maximum running speed. In hypertensive rats, MICT decreased SAP, DAP, MAP, interventricular septal thickness during systole and diastole, and the contraction amplitude at 5 Hz. HIIT increased heart weight and left ventricular wall thickness during systole and diastole and reduced SAP, MAP, and the time to peak [Ca2+]i at all pacing frequencies. In conclusion, both aerobic training protocols promoted beneficial adaptations to cardiac morphology, function, and mechanical properties of single cardiomyocytes in SHR.
Collapse
Affiliation(s)
- Pedro Z Suarez
- Laboratory of Exercise Biology,
Department of Physical Education, Universidade Federal de Viçosa (UFV), Viçosa
36570-000, Brazil
| | - Antônio J Natali
- Laboratory of Exercise Biology,
Department of Physical Education, Universidade Federal de Viçosa (UFV), Viçosa
36570-000, Brazil
| | - José G Mill
- Department of Physiological Sciences,
Universidade Federal do Espírito Santo (UFES), Vitória 29075-210, Brazil
| | - Leonardo MT de Rezende
- Laboratory of Exercise Biology,
Department of Physical Education, Universidade Federal de Viçosa (UFV), Viçosa
36570-000, Brazil
| | - Leôncio L Soares
- Laboratory of Exercise Biology,
Department of Physical Education, Universidade Federal de Viçosa (UFV), Viçosa
36570-000, Brazil
| | - Filipe R Drummond
- Department of General Biology,
Universidade Federal de Viçosa (UFV), Viçosa 36570-000, Brazil
| | - Lucas CC Cardoso
- Laboratory of Exercise Biology,
Department of Physical Education, Universidade Federal de Viçosa (UFV), Viçosa
36570-000, Brazil
| | - Emily CC Reis
- Department of Veterinary Medicine,
Universidade Federal de Viçosa (UFV), Viçosa 36570-000, Brazil
| | - Victor N Lavorato
- Department of Physical Education,
Centro Universitário Governador Ozanam Coelho (UNIFAGOC), Ubá 36506-022,
Brazil
| | - Miguel A Carneiro-Júnior
- Laboratory of Exercise Biology,
Department of Physical Education, Universidade Federal de Viçosa (UFV), Viçosa
36570-000, Brazil,Miguel A Carneiro-Júnior.
| |
Collapse
|
60
|
Luo M, Cao S, Lv D, He L, He Z, Li L, Li Y, Luo S, Chang Q. Aerobic Exercise Training Improves Renal Injury in Spontaneously Hypertensive Rats by Increasing Renalase Expression in Medulla. Front Cardiovasc Med 2022; 9:922705. [PMID: 35898283 PMCID: PMC9309879 DOI: 10.3389/fcvm.2022.922705] [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: 04/18/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
We aimed to examine the effects of aerobic exercise training on renal function in spontaneously hypertensive rats (SHR) and elucidate their possible mechanisms. Adult male SHR and age-matched Wistar-Kyoto rats (WKY) were divided into four groups: WKY sedentary group, SHR sedentary group, low-intensity training group, and medium-intensity training group. Using molecular and biochemical approaches, we investigated the effects of 14-week training on renalase (RNLS) protein levels, renal function, and apoptosis and oxidative stress modulators in kidney tissues. In vitro, angiotensin II (Ang II)-induced human kidney proximal epithelial cells (HK-2) were treated with RNLS, and changes in apoptosis and oxidative stress levels were observed. Our results show that moderate training improved renal function decline in SHR. In addition, aerobic exercise therapy significantly increased levels of RNLS in the renal medulla of SHR. We observed in vitro that RNLS significantly inhibited the increase of Ang II-inducedapoptosis and oxidative stress levels in HK-2. In conclusion, aerobic exercise training effectively improved renal function in SHR by promoting RNLS expression in the renal medulla. These results explain the possible mechanism in which exercise improves renal injury in hypertensive patients and suggest RNLS as a novel therapy for kidney injury patients.
Collapse
Affiliation(s)
- Minghao Luo
- The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, China
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical UniversityChongqing, China
| | - Shuyuan Cao
- The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, China
| | - Dingyi Lv
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical UniversityChongqing, China
| | - Longlin He
- The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, China
| | - Zhou He
- The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, China
| | - Lingang Li
- The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, China
| | - Yongjian Li
- The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, China
| | - Suxin Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical UniversityChongqing, China
| | - Qing Chang
- The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, China
- The College of Exercise Medicine, Chongqing Medical University, Chongqing, China
- *Correspondence: Qing Chang
| |
Collapse
|
61
|
Hyatt JK. MOTS-c increases in skeletal muscle following long-term physical activity and improves acute exercise performance after a single dose. Physiol Rep 2022; 10:e15377. [PMID: 35808870 PMCID: PMC9270643 DOI: 10.14814/phy2.15377] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/08/2022] [Accepted: 06/17/2022] [Indexed: 05/03/2023] Open
Abstract
Skeletal muscle adapts to aerobic exercise training, in part, through fast-to-slow phenotypic shifts and an expansion of mitochondrial networks. Recent research suggests that the local and systemic benefits of exercise training also may be modulated by the mitochondrial-derived peptide, MOTS-c. Using a combination of acute and chronic exercise challenges, the goal of the present study was to characterize the interrelationship between MOTS-c and exercise. Compared to sedentary controls, 4-8 weeks of voluntary running increased MOTS-c protein expression ~1.5-5-fold in rodent plantaris, medial gastrocnemius, and tibialis anterior muscles and is sustained for 4-6 weeks of detraining. This MOTS-c increase coincides with elevations in mtDNA reflecting an expansion of the mitochondrial genome to aerobic training. In a second experiment, a single dose (15 mg/kg) of MOTS-c administered to untrained mice improved total running time (12% increase) and distance (15% increase) during an acute exercise test. In a final experiment, MOTS-c protein translocated from the cytoplasm into the nucleus in two of six mouse soleus muscles 1 h following a 90-min downhill running challenge; no nuclear translocation was observed in the plantaris muscles from the same animals. These findings indicate that MOTS-c protein accumulates within trained skeletal muscle likely through a concomitant increase in mtDNA. Furthermore, these data suggest that the systemic benefits of exercise are, in part, mediated by an expansion of the skeletal muscle-derived MOTS-c protein pool. The benefits of training may persist into a period of inactivity (e.g., detraining) resulting from a sustained increase in intramuscular MOTS-c proteins levels.
Collapse
|
62
|
Hastings MH, Herrera JJ, Guseh JS, Atlason B, Houstis NE, Abdul Kadir A, Li H, Sheffield C, Singh AP, Roh JD, Day SM, Rosenzweig A. Animal Models of Exercise From Rodents to Pythons. Circ Res 2022; 130:1994-2014. [PMID: 35679366 PMCID: PMC9202075 DOI: 10.1161/circresaha.122.320247] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Acute and chronic animal models of exercise are commonly used in research. Acute exercise testing is used, often in combination with genetic, pharmacological, or other manipulations, to study the impact of these manipulations on the cardiovascular response to exercise and to detect impairments or improvements in cardiovascular function that may not be evident at rest. Chronic exercise conditioning models are used to study the cardiac phenotypic response to regular exercise training and as a platform for discovery of novel pathways mediating cardiovascular benefits conferred by exercise conditioning that could be exploited therapeutically. The cardiovascular benefits of exercise are well established, and, frequently, molecular manipulations that mimic the pathway changes induced by exercise recapitulate at least some of its benefits. This review discusses approaches for assessing cardiovascular function during an acute exercise challenge in rodents, as well as practical and conceptual considerations in the use of common rodent exercise conditioning models. The case for studying feeding in the Burmese python as a model for exercise-like physiological adaptation is also explored.
Collapse
Affiliation(s)
- Margaret H Hastings
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Jonathan J Herrera
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor (J.J.H.)
| | - J Sawalla Guseh
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Bjarni Atlason
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Nicholas E Houstis
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Azrul Abdul Kadir
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Haobo Li
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Cedric Sheffield
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Anand P Singh
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Jason D Roh
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Sharlene M Day
- Cardiovascular Medicine, Perelman School of Medicine' University of Pennsylvania, Philadelphia (S.M.D.)
| | - Anthony Rosenzweig
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| |
Collapse
|
63
|
Roh J, Hill JA, Singh A, Valero-Muñoz M, Sam F. Heart Failure With Preserved Ejection Fraction: Heterogeneous Syndrome, Diverse Preclinical Models. Circ Res 2022; 130:1906-1925. [PMID: 35679364 PMCID: PMC10035274 DOI: 10.1161/circresaha.122.320257] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) represents one of the greatest challenges facing cardiovascular medicine today. Despite being the most common form of heart failure worldwide, there has been limited success in developing therapeutics for this syndrome. This is largely due to our incomplete understanding of the biology driving its systemic pathophysiology and the heterogeneity of clinical phenotypes, which are increasingly being recognized as distinct HFpEF phenogroups. Development of efficacious therapeutics fundamentally relies on robust preclinical models that not only faithfully recapitulate key features of the clinical syndrome but also enable rigorous investigation of putative mechanisms of disease in the context of clinically relevant phenotypes. In this review, we propose a preclinical research strategy that is conceptually grounded in model diversification and aims to better align with our evolving understanding of the heterogeneity of clinical HFpEF. Although heterogeneity is often viewed as a major obstacle in preclinical HFpEF research, we challenge this notion and argue that embracing it may be the key to demystifying its pathobiology. Here, we first provide an overarching guideline for developing HFpEF models through a stepwise approach of comprehensive cardiac and extra-cardiac phenotyping. We then present an overview of currently available models, focused on the 3 leading phenogroups, which are primarily based on aging, cardiometabolic stress, and chronic hypertension. We discuss how well these models reflect their clinically relevant phenogroup and highlight some of the more recent mechanistic insights they are providing into the complex pathophysiology underlying HFpEF.
Collapse
Affiliation(s)
- Jason Roh
- Cardiovascular Research Center, Massachusetts General Hospital, Boston (J.R., A.S.)
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology) (J.A.H.), University of Texas Southwestern Medical Center, Dallas
- Department of Molecular Biology (J.A.H.), University of Texas Southwestern Medical Center, Dallas
| | - Abhilasha Singh
- Cardiovascular Research Center, Massachusetts General Hospital, Boston (J.R., A.S.)
| | - María Valero-Muñoz
- Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (M.V.-M., F.S.)
| | - Flora Sam
- Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (M.V.-M., F.S.)
| |
Collapse
|
64
|
Effects of Drosophila melanogaster regular exercise and apolipoprotein B knockdown on abnormal heart rhythm induced by a high-fat diet. PLoS One 2022; 17:e0262471. [PMID: 35657779 PMCID: PMC9165823 DOI: 10.1371/journal.pone.0262471] [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: 06/29/2021] [Accepted: 12/23/2021] [Indexed: 11/19/2022] Open
Abstract
Abnormal heart rhythm is a common cardiac dysfunction in obese patients, and its pathogenesis is related to systemic lipid accumulation. The cardiomyocyte-derived apoLpp (homologous gene in Drosophila of the human apolipoprotein B) plays an important role in whole-body lipid metabolism of Drosophila under a high-fat diet (HFD). Knockdown of apoLpp derived from cardiomyocytes can reduce HFD-induced weight gain and abdominal lipid accumulation. In addition, exercise can reduce the total amount of apoLpp in circulation. However, the relationship between regular exercise, cardiomyocyte-derived apoLpp and abnormal heart rhythm is unclear. We found that an HFD increased the level of triglyceride (TG) in the whole-body, lipid accumulation and obesity in Drosophila. Moreover, the expression of apoLpp in the heart increased sharply, the heart rate and arrhythmia index increased and fibrillation occurred. Conversely, regular exercise or cardiomyocyte-derived apoLpp knockdown reduced the TG level in the whole-body of Drosophila. This significantly reduced the arrhythmia induced by obesity, including the reduction of heart rate, arrhythmia index, and fibrillation. Under HFD conditions, flies with apoLpp knockdown in the heart could resist the abnormal cardiac rhythm caused by obesity after receiving regular exercise. HFD-induced obesity and abnormal cardiac rhythm may be related to the acute increase of cardiomyocyte-derived apoLpp. Regular exercise and inhibition of cardiomyocyte-derived apoLpp can reduce the HFD-induced abnormal cardiac rhythm.
Collapse
|
65
|
van Ham WB, Kessler EL, Oerlemans MI, Handoko ML, Sluijter JP, van Veen TA, den Ruijter HM, de Jager SC. Clinical Phenotypes of Heart Failure With Preserved Ejection Fraction to Select Preclinical Animal Models. JACC Basic Transl Sci 2022; 7:844-857. [PMID: 36061340 PMCID: PMC9436760 DOI: 10.1016/j.jacbts.2021.12.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/20/2021] [Accepted: 12/31/2021] [Indexed: 11/21/2022]
Abstract
To better define HFpEF clinically, patients are nowadays often clustered into phenogroups, based on their comorbidities and symptoms Many animal models claim to mimic HFpEF, but phenogroups are not yet regularly used to cluster them HFpEF animals models often lack reports of clinical symptoms of HF, therefore mainly presenting as extended models of LVDD, clinically seen as a prestate of HFpEF We investigated if clinically relevant phenogroups can guide selection of animal models aiming at better defined animal research
At least one-half of the growing heart failure population consists of heart failure with preserved ejection fraction (HFpEF). The limited therapeutic options, the complexity of the syndrome, and many related comorbidities emphasize the need for adequate experimental animal models to study the etiology of HFpEF, as well as its comorbidities and pathophysiological changes. The strengths and weaknesses of available animal models have been reviewed extensively with the general consensus that a “1-size-fits-all” model does not exist, because no uniform HFpEF patient exists. In fact, HFpEF patients have been categorized into HFpEF phenogroups based on comorbidities and symptoms. In this review, we therefore study which animal model is best suited to study the different phenogroups—to improve model selection and refinement of animal research. Based on the published data, we extrapolated human HFpEF phenogroups into 3 animal phenogroups (containing small and large animals) based on reports and definitions of the authors: animal models with high (cardiac) age (phenogroup aging); animal models focusing on hypertension and kidney dysfunction (phenogroup hypertension/kidney failure); and models with hypertension, obesity, and type 2 diabetes mellitus (phenogroup cardiometabolic syndrome). We subsequently evaluated characteristics of HFpEF, such as left ventricular diastolic dysfunction parameters, systemic inflammation, cardiac fibrosis, and sex-specificity in the different models. Finally, we scored these parameters concluded how to best apply these models. Based on our findings, we propose an easy-to-use classification for future animal research based on clinical phenogroups of interest.
Collapse
Affiliation(s)
- Willem B. van Ham
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Elise L. Kessler
- Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University of Utrecht, Utrecht, the Netherlands
| | | | - M. Louis Handoko
- Department of Cardiology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Joost P.G. Sluijter
- Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University of Utrecht, Utrecht, the Netherlands
| | - Toon A.B. van Veen
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hester M. den Ruijter
- Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Saskia C.A. de Jager
- Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Address for correspondence: Dr Saskia C.A. de Jager, Laboratory for Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands.
| |
Collapse
|
66
|
Brown C, Pemberton S, Babin A, Abdulhameed N, Noonan C, Brown MB, Banks WA, Rhea EM. Insulin blood-brain barrier transport and interactions are greater following exercise in mice. J Appl Physiol (1985) 2022; 132:824-834. [PMID: 35175106 PMCID: PMC8917914 DOI: 10.1152/japplphysiol.00866.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Exercise has multiple beneficial effects including improving peripheral insulin sensitivity, improving central function such as memory, and restoring a dysregulated blood-brain barrier (BBB). Central nervous system (CNS) insulin resistance is a common feature of cognitive impairment, including Alzheimer's disease. Delivery of insulin to the brain can improve memory. Endogenous insulin must cross the BBB to directly act within the CNS and this transport system can be affected by various physiological states and serum factors. Therefore, the current study sought to investigate whether exercise could enhance insulin BBB transport as a mechanism for the underlying benefits of exercise on cognition. We investigated radioactive insulin BBB pharmacokinetics following an acute bout of exercise in young, male and female CD-1 mice. In addition, we investigated changes in serum levels of substrates that are known to affect insulin BBB transport. Finally, we measured the basal level of a downstream protein involved in insulin receptor signaling in various brain regions as well as muscle. We found insulin BBB transport in males was greater following exercise, and in males and females to both enhance the level of insulin vascular binding and alter CNS insulin receptor signaling, independent of changes in serum factors known to alter insulin BBB transport.NEW & NOTEWORTHY Central nervous system (CNS) insulin and exercise are beneficial for cognition. CNS insulin resistance is present in Alzheimer's disease. CNS insulin levels are regulated by transport across the blood-brain barrier (BBB). We show that exercise can enhance insulin BBB transport and binding of insulin to the brain's vasculature in mice. There were no changes in serum factors known to alter insulin BBB pharmacokinetics. We conclude exercise could impact cognition through regulation of insulin BBB transport.
Collapse
Affiliation(s)
- Caitlin Brown
- 1Research and Development, Veterans Affairs Puget Sound Health Care System, Seattle, Washington
| | - Sarah Pemberton
- 1Research and Development, Veterans Affairs Puget Sound Health Care System, Seattle, Washington,2Department of Biology, University of Washington, Seattle, Washington
| | - Alice Babin
- 1Research and Development, Veterans Affairs Puget Sound Health Care System, Seattle, Washington
| | - Noor Abdulhameed
- 1Research and Development, Veterans Affairs Puget Sound Health Care System, Seattle, Washington,2Department of Biology, University of Washington, Seattle, Washington
| | - Cassidy Noonan
- 1Research and Development, Veterans Affairs Puget Sound Health Care System, Seattle, Washington,2Department of Biology, University of Washington, Seattle, Washington
| | - Mary Beth Brown
- 3Division of Physical Therapy, Department of Rehabilitation Medicine, University of Washington, Seattle, Washington
| | - William A. Banks
- 4Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington,5Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington
| | - Elizabeth M. Rhea
- 4Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington,5Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington
| |
Collapse
|
67
|
Holcomb LE, Rowe P, O’Neill CC, DeWitt EA, Kolwicz SC. Sex differences in endurance exercise capacity and skeletal muscle lipid metabolism in mice. Physiol Rep 2022; 10:e15174. [PMID: 35133078 PMCID: PMC8822869 DOI: 10.14814/phy2.15174] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 05/03/2023] Open
Abstract
Previous studies suggest that sex differences in lipid metabolism exist with females demonstrating a higher utilization of lipids during exercise, which is mediated partly by increased utilization of muscle triglycerides. However, whether these changes in lipid metabolism contribute directly to endurance exercise performance is unclear. Therefore, the objective of this study was to investigate the contribution of exercise substrate metabolism to sex differences in endurance exercise capacity (EEC) in mice. Male and female C57BL/6-NCrl mice were subjected to an EEC test until exhaustion on a motorized treadmill. The treadmill was set at a 10% incline, and the speed gradually increased from 10.2 m/min to 22.2 m/min at fixed intervals for up to 2.5 h. Tissues and blood were harvested in mice immediately following the EEC. A cohort of sedentary, non-exercised male and female mice were used as controls. Females outperformed males by ~25% on the EEC. Serum levels of both fatty acids and ketone bodies were ~50% higher in females at the end of the EEC. In sedentary female mice, skeletal muscle triglyceride content was significantly greater compared to sedentary males. Gene expression analysis demonstrated that genes involved in skeletal muscle fatty acid oxidation were significantly higher in females with no changes in genes associated with glucose uptake or ketone body oxidation. The findings suggest that female mice have a higher endurance exercise capacity and a greater ability to mobilize and utilize fatty acids for energy.
Collapse
Affiliation(s)
- Lola E. Holcomb
- Heart and Muscle Metabolism LaboratoryDepartment of Health and Exercise PhysiologyUrsinus CollegeCollegevillePennsylvaniaUSA
| | - Patrick Rowe
- Heart and Muscle Metabolism LaboratoryDepartment of Health and Exercise PhysiologyUrsinus CollegeCollegevillePennsylvaniaUSA
| | - Caitlin C. O’Neill
- Heart and Muscle Metabolism LaboratoryDepartment of Health and Exercise PhysiologyUrsinus CollegeCollegevillePennsylvaniaUSA
| | - Elizabeth A. DeWitt
- Heart and Muscle Metabolism LaboratoryDepartment of Health and Exercise PhysiologyUrsinus CollegeCollegevillePennsylvaniaUSA
| | - Stephen C. Kolwicz
- Heart and Muscle Metabolism LaboratoryDepartment of Health and Exercise PhysiologyUrsinus CollegeCollegevillePennsylvaniaUSA
| |
Collapse
|
68
|
Massett MP, Matejka C, Kim H. Systematic Review and Meta-Analysis of Endurance Exercise Training Protocols for Mice. Front Physiol 2021; 12:782695. [PMID: 34950054 PMCID: PMC8691460 DOI: 10.3389/fphys.2021.782695] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/05/2021] [Indexed: 11/13/2022] Open
Abstract
Inbred and genetically modified mice are frequently used to investigate the molecular mechanisms responsible for the beneficial adaptations to exercise training. However, published paradigms for exercise training in mice are variable, making comparisons across studies for training efficacy difficult. The purpose of this systematic review and meta-analysis was to characterize the diversity across published treadmill-based endurance exercise training protocols for mice and to identify training protocol parameters that moderate the adaptations to endurance exercise training in mice. Published studies were retrieved from PubMed and EMBASE and reviewed for the following inclusion criteria: inbred mice; inclusion of a sedentary group; and exercise training using a motorized treadmill. Fifty-eight articles met those inclusion criteria and also included a "classical" marker of training efficacy. Outcome measures included changes in exercise performance, V ˙ O2max, skeletal muscle oxidative enzyme activity, blood lactate levels, or exercise-induced cardiac hypertrophy. The majority of studies were conducted using male mice. Approximately 48% of studies included all information regarding exercise training protocol parameters. Meta-analysis was performed using 105 distinct training groups (i.e., EX-SED pairs). Exercise training had a significant effect on training outcomes, but with high heterogeneity (Hedges' g=1.70, 95% CI=1.47-1.94, Tau2=1.14, I2 =80.4%, prediction interval=-0.43-3.84). Heterogeneity was partially explained by subgroup differences in treadmill incline, training duration, exercise performance test type, and outcome variable. Subsequent analyses were performed on subsets of studies based on training outcome, exercise performance, or biochemical markers. Exercise training significantly improved performance outcomes (Hedges' g=1.85, 95% CI=1.55-2.15). Subgroup differences were observed for treadmill incline, training duration, and exercise performance test protocol on improvements in performance. Biochemical markers also changed significantly with training (Hedges' g=1.62, 95% CI=1.14-2.11). Subgroup differences were observed for strain, sex, exercise session time, and training duration. These results demonstrate there is a high degree of heterogeneity across exercise training studies in mice. Training duration had the most significant impact on training outcome. However, the magnitude of the effect of exercise training varies based on the marker used to assess training efficacy.
Collapse
Affiliation(s)
- Michael P Massett
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX, United States
| | - Caitlyn Matejka
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX, United States
| | - Hyoseon Kim
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX, United States
| |
Collapse
|
69
|
Qiu Y, Pan X, Chen Y, Xiao J. Hallmarks of exercised heart. J Mol Cell Cardiol 2021; 164:126-135. [PMID: 34914934 DOI: 10.1016/j.yjmcc.2021.12.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/29/2022]
Abstract
The benefits of exercise in humans on the heart have been well recognized for many years. Long-term endurance exercise training can induce physiologic cardiac hypertrophy with normal or enhanced heart function, and provide protective benefits in preventing heart failure. The heart-specific responses that occur during exercise are complex and highly variable. This review mainly focuses on the current understanding of the structural and functional cardiac adaptations to exercise as well as molecular pathways and signaling proteins responsible for these changes. Here, we summarize eight tentative hallmarks that represent common denominators of the exercised heart. These hallmarks are: cardiomyocyte growth, cardiomyocyte fate reprogramming, angiogenesis and lymphangiogenesis, mitochondrial remodeling, epigenetic alteration, enhanced endothelial function, quiescent cardiac fibroblast, and improved cardiac metabolism. A major challenge is to explore the underlying molecular mechanisms for cardio-protective effects of exercise, and to identify therapeutic targets for heart diseases.
Collapse
Affiliation(s)
- Yan Qiu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Xue Pan
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Yiwen Chen
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
70
|
Multiple Applications of Different Exercise Modalities with Rodents. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3898710. [PMID: 34868454 PMCID: PMC8639251 DOI: 10.1155/2021/3898710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/14/2021] [Accepted: 11/12/2021] [Indexed: 12/29/2022]
Abstract
A large proportion of chronic diseases can be derived from a sedentary lifestyle. Raising physical activity awareness is indispensable, as lack of exercise is the fourth most common cause of death worldwide. Animal models in different research fields serve as important tools in the study of acute or chronic noncommunicable disorders. With the help of animal-based exercise research, exercise-mediated complex antioxidant and inflammatory pathways can be explored, which knowledge can be transferred to human studies. Whereas sustained physical activity has an enormous number of beneficial effects on many organ systems, these animal models are easily applicable in several research areas. This review is aimed at providing an overall picture of scientific research studies using animal models with a focus on different training modalities. Without wishing to be exhaustive, the most commonly used forms of exercise are presented.
Collapse
|
71
|
Bass-Stringer S, Tai CMK, McMullen JR. IGF1-PI3K-induced physiological cardiac hypertrophy: Implications for new heart failure therapies, biomarkers, and predicting cardiotoxicity. JOURNAL OF SPORT AND HEALTH SCIENCE 2021; 10:637-647. [PMID: 33246162 PMCID: PMC8724616 DOI: 10.1016/j.jshs.2020.11.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/28/2020] [Accepted: 11/13/2020] [Indexed: 05/30/2023]
Abstract
Heart failure represents the end point of a variety of cardiovascular diseases. It is a growing health burden and a leading cause of death worldwide. To date, limited treatment options exist for the treatment of heart failure, but exercise has been well-established as one of the few safe and effective interventions, leading to improved outcomes in patients. However, a lack of patient adherence remains a significant barrier in the implementation of exercise-based therapy for the treatment of heart failure. The insulin-like growth factor 1 (IGF1)-phosphoinositide 3-kinase (PI3K) pathway has been recognized as perhaps the most critical pathway for mediating exercised-induced heart growth and protection. Here, we discuss how modulating activity of the IGF1-PI3K pathway may be a valuable approach for the development of therapies that mimic the protective effects of exercise on the heart. We outline some of the promising approaches being investigated that utilize PI3K-based therapy for the treatment of heart failure. We discuss the implications for cardiac pathology and cardiotoxicity that arise in a setting of reduced PI3K activity. Finally, we discuss the use of animal models of cardiac health and disease, and genetic mice with increased or decreased cardiac PI3K activity for the discovery of novel drug targets and biomarkers of cardiovascular disease.
Collapse
Affiliation(s)
- Sebastian Bass-Stringer
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC 3086, Australia
| | - Celeste M K Tai
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC 3086, Australia; Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Physiology and Department of Medicine Alfred Hospital, Monash University, Melbourne, VIC 3004, Australia.
| |
Collapse
|
72
|
Liu D, Liu DC, Fan H, Wang Y. Lactobacillus fermentum CQPC08 Attenuates Exercise-Induced Fatigue in Mice Through Its Antioxidant Effects and Effective Intervention of Galactooligosaccharide. Drug Des Devel Ther 2021; 15:5151-5164. [PMID: 34992351 PMCID: PMC8714972 DOI: 10.2147/dddt.s317456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/18/2021] [Indexed: 12/02/2022] Open
Abstract
Aim The purpose of this study is to study the antioxidant effect of Lactobacillus fermentum CQPC08 (CQPC08) on exercise-induced fatigue, and the beneficial intervention of GOS on CQPC08. Methods We use the treadmill to establish a fatigue model caused by exercise, and perform drug treatment after exercise. We tested the exhaustive exercise time of mice; investigated the changes of mice body weight, liver index, histopathology, serum biochemical indicators and mRNA expression levels of oxidative and inflammation-related genes; and assessed the potential fatigue inhibitory effect of CQPC08, and the anti-oxidation effect of the combination of GOS and CQPC08. Results The results suggest that CQPC08 and combination with GOS reduces fatigue-induced oxidative damage of the liver, and it decreases blood urea nitrogen (BUN), lactic acid (LA), glutamic-oxaloacetic transaminase (GOT), glutamic-pyruvic transaminase (GPT), malonaldehyde (MDA), inducible nitric oxide synthase (iNOS), tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6 in serum. Higher levels of serum catalase (CAT), glutathione peroxidase (GSH-Px) and superoxide dismutase (SOD) were found. Treatment with the CQPC08 and combination with GOS correlates with lower relative mRNA expression levels of neuronal NOS (nNOS), iNOS, and TNF-α, and with higher mRNA expression levels of catalase and copper/zinc (Cu/Zn) and manganese (Mn) SOD enzymes in the liver and muscles. Conclusion These results suggest that CQPC08 can resolve exercise-induced fatigue by improving antioxidant ability in mice, and the combination of GOS and CQPC08 enhances this ability of CQPC08.
Collapse
Affiliation(s)
- Dong Liu
- Development Chongqing University of Education, Chongqing, People’s Republic of China
- Education Major in Physical Education, University of Perpetual Help System DALTA Las Pinas, Manila, Philippines
| | - Da Chuan Liu
- Student Affairs Department, Jiangmen Preschool Education College, Jiangmen, Guangdong, People’s Republic of China
| | - Hao Fan
- School of Tourism and Service Management, Chongqing University of Education, Chongqing, People’s Republic of China
- Cultural Industries and Cultural Policy, Yuan Ze University, Taoyuan, Taiwan
| | - Yu Wang
- Orthopedics Department, General Hospital of Northern Theatre Command, Liaoning Province, People’s Republic of China
- Correspondence: Yu Wang Orthopedics Department, General Hospital of Northern Theatre Command, No. 83 Wenhua Road, Shenhe District, Shenyang, Liaoning Province, 110016, People’s Republic of ChinaTel +86-18609886338 Email
| |
Collapse
|
73
|
Lindsey ML, Brunt KR, Kirk JA, Kleinbongard P, Calvert JW, de Castro Brás LE, DeLeon-Pennell KY, Del Re DP, Frangogiannis NG, Frantz S, Gumina RJ, Halade GV, Jones SP, Ritchie RH, Spinale FG, Thorp EB, Ripplinger CM, Kassiri Z. Guidelines for in vivo mouse models of myocardial infarction. Am J Physiol Heart Circ Physiol 2021; 321:H1056-H1073. [PMID: 34623181 PMCID: PMC8834230 DOI: 10.1152/ajpheart.00459.2021] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 12/11/2022]
Abstract
Despite significant improvements in reperfusion strategies, acute coronary syndromes all too often culminate in a myocardial infarction (MI). The consequent MI can, in turn, lead to remodeling of the left ventricle (LV), the development of LV dysfunction, and ultimately progression to heart failure (HF). Accordingly, an improved understanding of the underlying mechanisms of MI remodeling and progression to HF is necessary. One common approach to examine MI pathology is with murine models that recapitulate components of the clinical context of acute coronary syndrome and subsequent MI. We evaluated the different approaches used to produce MI in mouse models and identified opportunities to consolidate methods, recognizing that reperfused and nonreperfused MI yield different responses. The overall goal in compiling this consensus statement is to unify best practices regarding mouse MI models to improve interpretation and allow comparative examination across studies and laboratories. These guidelines will help to establish rigor and reproducibility and provide increased potential for clinical translation.
Collapse
Affiliation(s)
- Merry L Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, Nebraska
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Keith R Brunt
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Saint John, New Brunswick, Canada
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - John W Calvert
- Carlyle Fraser Heart Center of Emory University Hospital Midtown, Atlanta, Georgia
- Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - Lisandra E de Castro Brás
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Kristine Y DeLeon-Pennell
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Dominic P Del Re
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Nikolaos G Frangogiannis
- Division of Cardiology, Department of Medicine, The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York
| | - Stefan Frantz
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Richard J Gumina
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, Florida
| | - Steven P Jones
- Department of Medicine, Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky
| | - Rebecca H Ritchie
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Victoria, Australia
| | - Francis G Spinale
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine and the Columbia Veteran Affairs Medical Center, Columbia, South Carolina
| | - Edward B Thorp
- Department of Pathology and Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California Davis School of Medicine, Davis, California
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
74
|
Bei Y, Wang L, Ding R, Che L, Fan Z, Gao W, Liang Q, Lin S, Liu S, Lu X, Shen Y, Wu G, Yang J, Zhang G, Zhao W, Guo L, Xiao J. Animal exercise studies in cardiovascular research: Current knowledge and optimal design-A position paper of the Committee on Cardiac Rehabilitation, Chinese Medical Doctors' Association. JOURNAL OF SPORT AND HEALTH SCIENCE 2021; 10:660-674. [PMID: 34454088 PMCID: PMC8724626 DOI: 10.1016/j.jshs.2021.08.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 05/09/2021] [Accepted: 07/11/2021] [Indexed: 05/02/2023]
Abstract
Growing evidence has demonstrated exercise as an effective way to promote cardiovascular health and protect against cardiovascular diseases However, the underlying mechanisms of the beneficial effects of exercise have yet to be elucidated. Animal exercise studies are widely used to investigate the key mechanisms of exercise-induced cardiovascular protection. However, standardized procedures and well-established evaluation indicators for animal exercise models are needed to guide researchers in carrying out effective, high-quality animal studies using exercise to prevent and treat cardiovascular diseases. In our review, we present the commonly used animal exercise models in cardiovascular research and propose a set of standard procedures for exercise training, emphasizing the appropriate measurements and analysis in these chronic exercise models. We also provide recommendations for optimal design of animal exercise studies in cardiovascular research, including the choice of exercise models, control of exercise protocols, exercise at different stages of disease, and other considerations, such as age, sex, and genetic background. We hope that this position paper will promote basic research on exercise-induced cardiovascular protection and pave the way for successful translation of exercise studies from bench to bedside in the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Yihua Bei
- Institute of Geriatrics, Affiliated Nantong Hospital of Shanghai University, Sixth People's Hospital of Nantong, School of Medicine, Shanghai University, Nantong 226011, China; Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Lei Wang
- Department of Rehabilitation Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Rongjing Ding
- Department of Cardiology, Peking University People's Hospital, Beijing 100044, China
| | - Lin Che
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai 200065, China
| | - Zhiqing Fan
- Department of Cardiology, Daqing Oilfield General Hospital, Daqing 163000, China
| | - Wei Gao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Qi Liang
- Department of Rehabilitation Medicine, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Shenghui Lin
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| | - Suixin Liu
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Xiao Lu
- Department of Rehabilitation Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yuqin Shen
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai 200065, China
| | - Guifu Wu
- Department of Cardiology, Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518033, China; Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Sun Yat-Sen University, Shenzhen 518033, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jian Yang
- Department of Rehabilitation Medicine, Shanghai Xuhui Central Hospital, Shanghai 200031, China
| | - Guolin Zhang
- Cardiac Rehabilitation Department, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Wei Zhao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Lan Guo
- Cardiac Rehabilitation Department, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.
| | - Junjie Xiao
- Institute of Geriatrics, Affiliated Nantong Hospital of Shanghai University, Sixth People's Hospital of Nantong, School of Medicine, Shanghai University, Nantong 226011, China; Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
75
|
Kelestimur H, Bulmus O, Serhatlioglu I, Ercan Z, Ozer Kaya S, Yardimci A, Ulker N, Kacar E, Canpolat S. Effects of treadmill exercise on sexual behavior and reproductive parameters in chronically stressed-male rats. Physiol Res 2021; 70:765-775. [PMID: 34505528 PMCID: PMC8820524 DOI: 10.33549/physiolres.934585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 07/09/2021] [Indexed: 11/25/2022] Open
Abstract
Exposure to chronic stress stimulates the hypothalamic-pituitary-adrenal (HPA) axis and then simultaneously inhibits hypothalamic-pituitary-gonadal axis (HPG) axis activity. The inhibition formed by the HPA axis is the main mechanism of action of stress on reproductive function. HPG axis activity is known to be changed by various factors, including exercise. Exercise has been found to have a number of positive effects on sexual behavior, reproductive hormones, and sperm parameters in studies with animal models for many years. The main aim of this study is to investigate the effects of chronic treadmill exercise on chronically stressed-male rats' sexual behavior, reproductive hormones, and sperm parameters. A total of 40 sexually adult male rats were randomly and equally divided into four groups as control, stress, exercise, and stress+exercise. Animals in the exercise group were subjected to the chronic treadmill exercise (moderate intensity) for 33 days with a periodic increase in speed and duration. Animals in the stress group were exposed to restraint stress for 1 h, 2 h, and 3 h during the first, second and third 15 days respectively. Sexual behavior parameters, hormone measurements, and sperm parameters were evaluated. The main effects of chronic exercise on sexual behavior were centered on a significant increase in the ejaculation frequency (EF) in the stress+exercise group. Also, sperm concentration and motility in the stress group significantly decreased, and then sperm motility was improved by exercise in the stress+exercise group. In sum, our results show that chronic treadmill exercise may improve the adverse effects of chronic stress on sexual behavior and sperm parameters in male rats in terms of some parameters.
Collapse
Affiliation(s)
- H Kelestimur
- Department of Physiology, Faculty of Medicine, Firat University, Elazig, Turkey.
| | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Sidorova YS, Shipelin VA, Petrov NA, Zorin SN, Mazo VK. Adaptogenic Properties of a Phytoecdysteroid-Rich Extract from the Leaves of Spinacia oleracea L. PLANTS (BASEL, SWITZERLAND) 2021; 10:2555. [PMID: 34961026 PMCID: PMC8704498 DOI: 10.3390/plants10122555] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 06/14/2023]
Abstract
Increasing the ability of the human body to adapt in conditions of physical or emotional stress is promising from the standpoint of the use of preventive nutrition containing functional food ingredients (FFI) with proven effectiveness in complex physiological in vivo studies. In this work, we developed FFI from spinach leaves (Spinacia oleracea L.) with a high content of polyphenols and adaptogens-phytoecdysteroids. Using in vivo models of increased physical activity and immobilization-induced emotional stress, we evaluated the nonspecific resistance of rats in response to the addition of the developed FFI to the diet. In the acute toxicity experiment, we found no signs of FFI toxicity up to 5000 mg/kg body weight. As a result of the daily 26-day consumption of FFI, we observed an anxiolytic effect in physiological studies. FFI prevented an increase in the content of biogenic amines in the blood, the main markers of the stress system, and had a positive effect on the lipid metabolism of the rats. The obtained results demonstrate a "smoothing" effect on the body's reaction in response to induced stress conditions.
Collapse
Affiliation(s)
| | - Vladimir A. Shipelin
- Federal Research Centre of Nutrition and Biotechnology, 109240 Moscow, Russia; (Y.S.S.); (N.A.P.); (S.N.Z.); (V.K.M.)
| | | | | | | |
Collapse
|
77
|
Lindsey ML, LeBlanc AJ, Ripplinger CM, Carter JR, Kirk JA, Hansell Keehan K, Brunt KR, Kleinbongard P, Kassiri Z. Reinforcing rigor and reproducibility expectations for use of sex and gender in cardiovascular research. Am J Physiol Heart Circ Physiol 2021; 321:H819-H824. [PMID: 34524922 DOI: 10.1152/ajpheart.00418.2021] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Merry L Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, Nebraska.,Research Service, Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Amanda J LeBlanc
- Department of Physiology and Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky
| | | | - Jason R Carter
- Department of Health and Human Development, Montana State University, Bozeman, Montana
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois
| | - Kara Hansell Keehan
- Strategic Journal Development, American Physiological Society, Rockville, Maryland.,AJP-Heart and Circulatory Physiology, American Physiological Society, Rockville, Maryland
| | - Keith R Brunt
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Saint John, New Brunswick, Canada
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
78
|
Buehler PW, Swindle D, Pak DI, Ferguson SK, Majka SM, Karoor V, Moldovan R, Sintas C, Black J, Gentinetta T, Buzzi RM, Vallelian F, Wassmer A, Edler M, Bain J, Schu D, Hassell K, Nuss R, Schaer DJ, Irwin DC. Hemopexin dosing improves cardiopulmonary dysfunction in murine sickle cell disease. Free Radic Biol Med 2021; 175:95-107. [PMID: 34478834 PMCID: PMC9231663 DOI: 10.1016/j.freeradbiomed.2021.08.238] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/26/2021] [Accepted: 08/29/2021] [Indexed: 01/13/2023]
Abstract
Hemopexin (Hpx) is a crucial defense protein against heme liberated from degraded hemoglobin during hemolysis. High heme stress creates an imbalance in Hpx bioavailability, favoring heme accumulation and downstream pathophysiological responses leading to cardiopulmonary disease progression in sickle cell disease (SCD) patients. Here, we evaluated a model of murine SCD, which was designed to accelerate red blood cell sickling, pulmonary hypertension, right ventricular dysfunction, and exercise intolerance by exposure of the mice to moderate hypobaric hypoxia. The sequence of pathophysiology in this model tracks with circulatory heme accumulation, lipid oxidation, extensive remodeling of the pulmonary vasculature, and fibrosis. We hypothesized that Hpx replacement for an extended period would improve exercise tolerance measured by critical speed as a clinically meaningful therapeutic endpoint. Further, we sought to define the effects of Hpx on upstream cardiopulmonary function, histopathology, and tissue oxidation. Our data shows that tri-weekly administrations of Hpx for three months dose-dependently reduced heme exposure and pulmonary hypertension while improving cardiac pressure-volume relationships and exercise tolerance. Furthermore, Hpx administration dose-dependently attenuated pulmonary fibrosis and oxidative modifications in the lung and myocardium of the right ventricle. Observations in our SCD murine model are consistent with pulmonary vascular and right ventricular pathology at autopsy in SCD patients having suffered from severe pulmonary hypertension, right ventricular dysfunction, and sudden cardiac death. This study provides a translational evaluation supported by a rigorous outcome analysis demonstrating therapeutic proof-of-concept for Hpx replacement in SCD.
Collapse
Affiliation(s)
- Paul W Buehler
- University of Maryland, Department of Pathology and the Center for Blood Oxygen Transport, Department of Pediatrics, School of Medicine, Baltimore, MD, USA.
| | - Delaney Swindle
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - David I Pak
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Scott K Ferguson
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; Department of Kinesiology and Exercise Sciences, College of Natural and Health Sciences, University of Hawaii at Hilo, Hilo, HI, USA
| | - Susan M Majka
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Hospital, Denver, CO, USA
| | - Vijaya Karoor
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Radu Moldovan
- Advanced Light Microscopy Core, CU Anschutz Medical Campus, Aurora,, CO, USA
| | - Chantal Sintas
- Department of Pathology and Laboratory Medicine at Children's Hospital Colorado, USA
| | - Jennifer Black
- Department of Pathology, Pediatrics, University of Colorado School of Medicine, USA
| | | | - Raphael M Buzzi
- Division of Internal Medicine, University and University Hospital of Zurich, Zurich, Switzerland
| | - Florence Vallelian
- Division of Internal Medicine, University and University Hospital of Zurich, Zurich, Switzerland
| | | | - Monika Edler
- CSL Behring AG, Research and Development, Bern, Switzerland
| | - Joseph Bain
- CSL Behring AG, Innovations GmbH, Marburg, Germany
| | - Daniel Schu
- CSL Behring AG, Innovations GmbH, Marburg, Germany
| | - Kathryn Hassell
- Division of Hematology Colorado Sickle Cell Treatment and Research Center, School of Medicine, Anschutz Medical Campus, University of Colorado-Denver School of Medicine, Aurora,, CO, USA
| | - Rachelle Nuss
- Division of Hematology Colorado Sickle Cell Treatment and Research Center, School of Medicine, Anschutz Medical Campus, University of Colorado-Denver School of Medicine, Aurora,, CO, USA
| | - Dominik J Schaer
- Division of Internal Medicine, University and University Hospital of Zurich, Zurich, Switzerland
| | - David C Irwin
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
79
|
Magalhães FDC, Fernandes T, Bassaneze V, Mattos KC, Schettert I, Marques FLN, Krieger JE, Nava R, Barauna VG, Menezes de Oliveira E. High-volume endurance exercise training stimulates hematopoiesis by increasing ACE NH2-terminal activity. Clin Sci (Lond) 2021; 135:2377-2391. [PMID: 34608942 DOI: 10.1042/cs20210739] [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: 07/19/2021] [Revised: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 11/17/2022]
Abstract
One of the health benefits of endurance exercise training (ET) is the stimulation of hematopoiesis. However, the mechanisms underlying ET-induced hematopoietic adaptations are understudied. N-Acetyl-Seryl-Aspartyl-Lysyl-Proline (Ac-SDKP) inhibits proliferation of early hematopoietic progenitor cells. The angiotensin I-converting enzyme (ACE) NH2-terminal promotes hematopoiesis by inhibiting the anti-hematopoietic effect of Ac-SDKP. Here we demonstrate for the first time the role of ACE NH2-terminal in ET-induced hematopoietic adaptations. Wistar rats were subjected to 10 weeks of moderate-(T1) and high-(T2) volume swimming-training. Although both protocols induced classical ET-associated adaptations, only T2 increased plasma ACE NH2-domain activity (by 40%, P=0.0003) and reduced Ac-SDKP levels (by 50%, P<0.0001). T2 increased the number of hematopoietic stem cells (HSCs; ∼200%, P=0.0008), early erythroid progenitor colonies (∼300%, P<0.0001) and reticulocytes (∼500%, P=0.0007), and reduced erythrocyte lifespan (∼50%, P=0.022). Following, Wistar rats were subjected to T2 or T2 combined with ACE NH2-terminal inhibition (captopril (Cap) treatment: 10 mg.kg-1.day-1). T2 combined with ACE NH2-terminal inhibition prevented Ac-SDKP decrease and attenuated ET-induced hematopoietic adaptations. Altogether, our findings show that ET-induced hematopoiesis was at least partially associated with increased ACE NH2-terminal activity and reduction in the hematopoietic inhibitor Ac-SDKP.
Collapse
Affiliation(s)
- Flávio de Castro Magalhães
- Laboratory of Biochemistry of the Motor Activity, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil
- Laboratory of Exercise Biology, Integrated Center of Health Research, Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas-Federal University of the Jequitinhonha and Mucuri Valleys, 5000 MGT 367 Road-km 583-Alto da Jacuba, 39100-000, Diamantina, Minas Gerais, Brazil
| | - Tiago Fernandes
- Laboratory of Biochemistry of the Motor Activity, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil
| | - Vinícius Bassaneze
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Katt Coelho Mattos
- Laboratory of Biochemistry of the Motor Activity, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil
| | - Isolmar Schettert
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Fabio Luiz Navarro Marques
- Radiopharmacy Laboratory, Nuclear Medicine Center, Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - José Eduardo Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Roberto Nava
- Department of Health, Exercise, and Sports Sciences, University of New Mexico, Albuquerque, NM 87131, U.S.A
| | - Valério Garrone Barauna
- Laboratory of Molecular and Cellular Physiology, Health Sciences Center, Federal University of Espírito Santo, Vitória, Brazil
| | - Edilamar Menezes de Oliveira
- Laboratory of Biochemistry of the Motor Activity, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
80
|
Liu Z, Yang B. CTRP6(C1q/Tumor Necrosis Factor (TNF)-related protein-6) alleviated the sevoflurane induced injury of mice central nervous system by promoting the expression of p-Akt (phosphorylated Akt). Bioengineered 2021; 12:5716-5726. [PMID: 34516328 PMCID: PMC8806630 DOI: 10.1080/21655979.2021.1967838] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Postoperative cognitive impairment and nervous system damage caused by anesthetics seriously affect patient’s postoperative recovery. Recent study has revealed that CTRP6 could alleviate apoptosis, inflammation and oxidative stress of nerve cells, thereby relieving nervous system damage induced by cerebral ischemia reperfusion. However, whether CTRP6 could relieve sevoflurane induced central nervous system injury is unclear. We stimulated C57BL/6 mice with sevoflurane and injected CTRP6 overexpression adenovirus vector. Next, H&E staining and TUNEL assays were performed to examine the effect of CTRP6 on sevoflurane induced injury of central nervous system. Finally, we isolated primary nerve cells of hippocampus. Flow cytometry and commercial kits were used for the detection of apoptosis and ROS levels of these cells. Western blotting was used for the detection of the expression level of p-Akt in central nervous tissues and primary cells. Results showed that sevoflurane induced injury and apoptosis of central nervous tissues. Overexpression of CTRP6 relieved apoptosis and injury of these tissues. CTRP6 inhibited the expression of cleaved caspase-3 and cleaved PARP in these tissues. Sevoflurane promoted apoptosis of primary cells and enhanced the expression of ROS and MDA in these cells. Overexpression of CTRP6 alleviated apoptosis and suppressed production of ROS and MDA in these cells. In addition, CTRP6 also enhanced the expression of p-Akt in primary cells. Taken together, our results suggested that CTRP6 relieved sevoflurane induced injury of central nervous tissues by promoting the expression of p-Akt. Therefore, the targeted drug of CTRP6 should be explored for the remission of these symptoms.
Collapse
Affiliation(s)
- Zhiwen Liu
- Department of Anesthesiology, The Second Hospital, University to South China Hengyang Cty, Hengyang City, Hunan Province, China
| | - Bin Yang
- Department of Anesthesiology, The Second Hospital, University to South China Hengyang Cty, Hengyang City, Hunan Province, China
| |
Collapse
|
81
|
Power Guerra N, Parveen A, Bühler D, Brauer DL, Müller L, Pilz K, Witt M, Glass Ä, Bajorat R, Janowitz D, Wolkenhauer O, Vollmar B, Kuhla A. Fibroblast Growth Factor 21 as a Potential Biomarker for Improved Locomotion and Olfaction Detection Ability after Weight Reduction in Obese Mice. Nutrients 2021; 13:nu13092916. [PMID: 34578793 PMCID: PMC8470262 DOI: 10.3390/nu13092916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 12/11/2022] Open
Abstract
Obesity is one of the most challenging diseases of the 21st century and is accompanied by behavioural disorders. Exercise, dietary adjustments, or time-restricted feeding are the only successful long-term treatments to date. Fibroblast growth factor 21 (FGF21) plays a key role in dietary regulation, but FGF21 resistance is prevalent in obesity. The aim of this study was to investigate in obese mice whether weight reduction leads to improved behaviour and whether these behavioural changes are associated with decreased plasma FGF21 levels. After establishing a model for diet-induced obesity, mice were subjected to three different interventions for weight reduction, namely dietary change, treadmill exercise, or time-restricted feeding. In this study, we demonstrated that only the combination of dietary change and treadmill exercise affected all parameters leading to a reduction in weight, fat, and FGF21, as well as less anxious behaviour, higher overall activity, and improved olfactory detection abilities. To investigate the interrelationship between FGF21 and behavioural parameters, feature selection algorithms were applied designating FGF21 and body weight as one of five highly weighted features. In conclusion, we concluded from the complementary methods that FGF21 can be considered as a potential biomarker for improved behaviour in obese mice after weight reduction.
Collapse
Affiliation(s)
- Nicole Power Guerra
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, Schillingallee 69a, 18057 Rostock, Germany; (N.P.G.); (A.P.); (D.B.); (L.M.); (B.V.)
- Department of Anatomy, Rostock University Medical Centre, Gertrudenstraße 9, 18057 Rostock, Germany;
| | - Alisha Parveen
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, Schillingallee 69a, 18057 Rostock, Germany; (N.P.G.); (A.P.); (D.B.); (L.M.); (B.V.)
| | - Daniel Bühler
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, Schillingallee 69a, 18057 Rostock, Germany; (N.P.G.); (A.P.); (D.B.); (L.M.); (B.V.)
| | - David Leon Brauer
- Department of Systems Biology and Bioinformatics, University of Rostock, Ulmenstraße 69, 18057 Rostock, Germany; (D.L.B.); (O.W.)
| | - Luisa Müller
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, Schillingallee 69a, 18057 Rostock, Germany; (N.P.G.); (A.P.); (D.B.); (L.M.); (B.V.)
- Department of Psychosomatic Medicine and Psychotherapy, Rostock University Medical Centre, Gehlsheimerstraße 20, 18147 Rostock, Germany
- Centre for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Centre, Gehlsheimerstraße 20, 18147 Rostock, Germany
| | - Kristin Pilz
- Department of Psychiatry, University of Greifswald, Ellernholzstraße 1-2, 17489 Greifswald, Germany; (K.P.); (D.J.)
| | - Martin Witt
- Department of Anatomy, Rostock University Medical Centre, Gertrudenstraße 9, 18057 Rostock, Germany;
| | - Änne Glass
- Institute for Biostatistics and Informatics, Rostock University Medical Centre, Ernst-Heydemann-Straße 8, 18057 Rostock, Germany;
| | - Rika Bajorat
- Department of Anesthesiology and Intensive Care Medicine, Rostock University Medical Centre, Schillingallee 35, 18057 Rostock, Germany;
| | - Deborah Janowitz
- Department of Psychiatry, University of Greifswald, Ellernholzstraße 1-2, 17489 Greifswald, Germany; (K.P.); (D.J.)
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock, Ulmenstraße 69, 18057 Rostock, Germany; (D.L.B.); (O.W.)
- Leibniz-Institute for Food Systems Biology, Technical University of Munich, Lise-Meitner-Straße 34, 85354 Freising, Germany
| | - Brigitte Vollmar
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, Schillingallee 69a, 18057 Rostock, Germany; (N.P.G.); (A.P.); (D.B.); (L.M.); (B.V.)
- Centre for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Centre, Gehlsheimerstraße 20, 18147 Rostock, Germany
| | - Angela Kuhla
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, Schillingallee 69a, 18057 Rostock, Germany; (N.P.G.); (A.P.); (D.B.); (L.M.); (B.V.)
- Centre for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Centre, Gehlsheimerstraße 20, 18147 Rostock, Germany
- Correspondence: ; Tel.: +49-381-494-2503
| |
Collapse
|
82
|
Luo M, Cao C, Niebauer J, Yan J, Ma X, Chang Q, Zhang T, Huang X, Liu G. Effects of different intensities of continuous training on vascular inflammation and oxidative stress in spontaneously hypertensive rats. J Cell Mol Med 2021; 25:8522-8536. [PMID: 34331512 PMCID: PMC8419160 DOI: 10.1111/jcmm.16813] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/05/2021] [Accepted: 07/09/2021] [Indexed: 12/17/2022] Open
Abstract
We aimed to study the effects and underlying mechanism of different intensities of continuous training (CT) on vascular inflammation and oxidative stress in spontaneously hypertensive rats (SHR). Rats were divided into five groups (n = 12): Wistar‐Kyoto rats sedentary group (WKY‐S), sedentary group (SHR‐S), low‐intensity CT group (SHR‐L), medium‐intensity CT group (SHR‐M) and high‐intensity CT group (SHR‐H). Changes in body mass, heart rate and blood pressure were recorded. The rats were euthanized after 14 weeks, and blood and vascular tissue samples were collected. Haematoxylin and Eosin staining was used to observe the aortic morphology, and Western blot was used to detect the expression of mesenteric artery proteins. After CT, the mean arterial pressures improved in SHR‐L and SHR‐M and increased in SHR‐H compared with those in SHR‐S. Vascular inflammation and oxidative stress levels significantly subsided in SHR‐L and SHR‐M (p < 0.05), whereas in SHR‐H, only vascular inflammation significantly subsided (p < 0.05), and oxidative stress remained unchanged (p > 0.05). AMPK and SIRT1/3 expressions in SHR‐L and SHR‐M were significantly up‐regulated than those in SHR‐S (p < 0.05). These results indicated that low‐ and medium‐intensity CT can effectively reduce the inflammatory response and oxidative stress of SHR vascular tissue, and high‐intensity CT can improve vascular tissue inflammation but not oxidative stress.
Collapse
Affiliation(s)
- Minghao Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunmei Cao
- Division of Sports Science and Physical Education, Tsinghua University, Beijing, China
| | - Josef Niebauer
- University Institute of Sports Medicine, Prevention and Rehabilitation, Paracelsus Medical University, Salzburg, Austria
| | - Jianghong Yan
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Xindong Ma
- Division of Sports Science and Physical Education, Tsinghua University, Beijing, China
| | - Qing Chang
- The College of Exercise Medicine, Chongqing Medical University, Chongqing, China
| | - Ting Zhang
- The Fifth Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Xiaoxiao Huang
- The College of Exercise Medicine, Chongqing Medical University, Chongqing, China
| | - Guochun Liu
- The College of Exercise Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
83
|
Miguel-Dos-Santos R, Moreira JBN, Loennechen JP, Wisløff U, Mesquita T. Exercising immune cells: The immunomodulatory role of exercise on atrial fibrillation. Prog Cardiovasc Dis 2021; 68:52-59. [PMID: 34274371 DOI: 10.1016/j.pcad.2021.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 07/12/2021] [Indexed: 12/17/2022]
Abstract
Exercise training is generally beneficial for cardiovascular health, improving stroke volume, cardiac output, and aerobic capacity. Despite these benefits, some evidence indicates that endurance training may increase the risk of atrial fibrillation (AF), particularly in highly trained individuals. Among multiple mechanisms, autonomic tone changes and atrial remodeling have been proposed as main contributors for exercise-induced AF. However, the contribution of local and systemic immunity is poorly understood in the development of atrial arrhythmogenic substrates. Here we aim to update the field of immunomodulation in the context of exercise and AF by compiling and reconciling the most recent evidence from preclinical and human studies and rationalize the applicability of "lone" AF terminology in athletes.
Collapse
Affiliation(s)
- Rodrigo Miguel-Dos-Santos
- Department of Physiology, Federal University of Sergipe, Sergipe, Brazil; Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - José Bianco Nascimento Moreira
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Jan Pål Loennechen
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; Department of Cardiology, St. Olav's University Hospital, Trondheim, Norway
| | - Ulrik Wisløff
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; School of Human Movement and Nutrition Science, University of Queensland, Queensland, Australia.
| | - Thássio Mesquita
- Smidt Heart Institute, Cedars-Sinai Medical Center, California, United States..
| |
Collapse
|
84
|
Aicher BO, Zhang J, Muratoglu SC, Galisteo R, Arai AL, Gray VL, Lal BK, Strickland DK, Ucuzian AA. Moderate aerobic exercise prevents matrix degradation and death in a mouse model of aortic dissection and aneurysm. Am J Physiol Heart Circ Physiol 2021; 320:H1786-H1801. [PMID: 33635167 PMCID: PMC8163659 DOI: 10.1152/ajpheart.00229.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 11/22/2022]
Abstract
Thoracic aortic aneurysm and dissection (TAAD) is a deadly disease characterized by intimal disruption induced by hemodynamic forces of the circulation. The effect of exercise in patients with TAAD is largely unknown. β-Aminopropionitrile (BAPN) is an irreversible inhibitor of lysyl oxidase that induces TAAD in mice. The objective of this study was to investigate the effect of aerobic exercise on BAPN-induced TAAD. Upon weaning, mice were given either BAPN-containing water or standard drinking water and subjected to either conventional cage activity (BAPN-CONV) or forced treadmill exercise (BAPN-EX) for up to 26 wk. Mortality was 23.5% (20/85) for BAPN-CONV mice versus 0% (0/22) for BAPN-EX mice (hazard ratio 3.8; P = 0.01). BAPN induced significant elastic lamina fragmentation and intimal-medial thickening compared with BAPN-untreated controls, and aneurysms were identified in 50% (5/10) of mice that underwent contrast-enhanced CT scanning. Exercise significantly decreased BAPN-induced wall thickening, calculated circumferential wall tension, and lumen diameter, with 0% (0/5) of BAPN-EX demonstrating chronic aortic aneurysm formation on CT scan. Expression of selected genes relevant to vascular diseases was analyzed by qRT-PCR. Notably, exercise normalized BAPN-induced increases in TGF-β pathway-related genes Cd109, Smad4, and Tgfβr1; inflammation-related genes Vcam1, Bcl2a1, Ccr2, Pparg, Il1r1, Il1r1, Itgb2, and Itgax; and vascular injury- and response-related genes Mmp3, Fn1, and Vwf. Additionally, exercise significantly increased elastin expression in BAPN-treated animals compared with controls. This study suggests that moderate aerobic exercise may be safe and effective in preventing the most devastating outcomes in TAAD.NEW & NOTEWORTHY Moderate aerobic exercise was shown to significantly reduce mortality, extracellular matrix degradation, and thoracic aortic aneurysm and dissection formation associated with lysyl oxidase inhibition in a mouse model. Gene expression suggested a reversal of TGF-β, inflammation, and extracellular matrix remodeling pathway dysregulation, along with augmented elastogenesis with exercise.
Collapse
MESH Headings
- Aminopropionitrile
- Aortic Dissection/chemically induced
- Aortic Dissection/metabolism
- Aortic Dissection/pathology
- Aortic Dissection/therapy
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Aortic Aneurysm, Thoracic/chemically induced
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/therapy
- Aortic Rupture/chemically induced
- Aortic Rupture/metabolism
- Aortic Rupture/pathology
- Aortic Rupture/prevention & control
- Dilatation, Pathologic
- Disease Models, Animal
- Disease Progression
- Exercise Therapy
- Extracellular Matrix/metabolism
- Extracellular Matrix/pathology
- Extracellular Matrix Proteins/metabolism
- Gene Expression Regulation
- Hemodynamics
- Male
- Mice, Inbred C57BL
- Proteolysis
- Signal Transduction
- Vascular Remodeling
- Mice
Collapse
Affiliation(s)
- Brittany O Aicher
- Center for Vascular & Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jackie Zhang
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Selen C Muratoglu
- Center for Vascular & Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Rebeca Galisteo
- Center for Vascular & Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland
| | - Allison L Arai
- Center for Vascular & Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland
| | - Vicki L Gray
- Department of Physical Therapy and Rehabilitation Science, University of Maryland School of Medicine, Baltimore, Maryland
| | - Brajesh K Lal
- Center for Vascular & Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
- Baltimore Veterans Affairs Medical Center, Vascular Service, Baltimore, Maryland
| | - Dudley K Strickland
- Center for Vascular & Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Physical Therapy and Rehabilitation Science, University of Maryland School of Medicine, Baltimore, Maryland
| | - Areck A Ucuzian
- Center for Vascular & Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
- Baltimore Veterans Affairs Medical Center, Vascular Service, Baltimore, Maryland
| |
Collapse
|
85
|
Craig JC, Broxterman RM, Cerbie JF, La Salle DT, Roundy CS, Jarrett CL, Richardson RS, Trinity JD. The dynamic adjustment of mean arterial pressure during exercise: a potential tool for discerning cardiovascular health status. J Appl Physiol (1985) 2021; 130:1544-1554. [PMID: 33830814 DOI: 10.1152/japplphysiol.00057.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The regulation of mean arterial pressure (MAP) during exercise has important physiological and clinical implications. Kinetics analysis on numerous physiological variables following the transition from unloaded-to-loaded exercise has revealed important information regarding their control. Surprisingly, the dynamic response of MAP during this transition remains to be quantified. Therefore, ten healthy participants (5/5 M/F, 24 ± 3 yr) completed repeated transitions from unloaded to moderate- and heavy-intensity dynamic single-leg knee-extensor exercise to investigate the on-kinetics of MAP. Following the transition to loaded exercise, MAP increased in a first-order dynamic manner, subsequent to a time delay (moderate: 23 ± 10; heavy: 19 ± 9 s, P > 0.05) at a speed (τ, moderate: 59 ± 30; heavy: 66 ± 19 s, P > 0.05), which did not differ between intensities, but the MAP amplitude was doubled during heavy-intensity exercise (moderate: 12 ± 5; heavy: 24 ± 8 mmHg, P < 0.001). The reproducibility [coefficient of variation (CV)] during heavy intensity for unloaded baseline, amplitude, and mean response time, when assessed as individual transitions, was 7 ± 1%, 18 ± 2%, and 25 ± 4%, respectively. Averaging two transitions improved the CVs to 4 ± 1%, 8 ± 2%, and 13 ± 3%, respectively. Preliminary findings supporting the clinical relevance of evaluating MAP kinetics in middle-aged hypertensive (n = 5) and, age-matched, normotensive (n = 5) participants revealed an exaggerated MAP response in both older groups (P < 0.05), but the MAP response was slowed only for the patients with hypertension (P < 0.05). It is concluded that kinetics modeling of MAP is practical for heavy-intensity knee-extensor exercise and may provide insight into cardiovascular health and the effect of aging.NEW & NOTEWORTHY Kinetics analysis of physiological variables following workload transitions provides important information, but this has not been performed on mean arterial pressure (MAP), despite the clear clinical importance of this variable. This investigation reveals that kinetic modeling of MAP following unloaded-to-loaded knee-extensor exercise is practical and repeatable. Additional preliminary findings in hypertensive and, age-matched, normotensive subjects suggest that MAP kinetics may provide insight into cardiovascular health and the effect of aging.
Collapse
Affiliation(s)
- Jesse C Craig
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah.,Geriatric Research, Education, and Clinical Center, Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Ryan M Broxterman
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah.,Geriatric Research, Education, and Clinical Center, Veterans Affairs Medical Center, Salt Lake City, Utah
| | - James F Cerbie
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| | - D Taylor La Salle
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| | - Caleb S Roundy
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah
| | - Catherine L Jarrett
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah.,Geriatric Research, Education, and Clinical Center, Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Russell S Richardson
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah.,Geriatric Research, Education, and Clinical Center, Veterans Affairs Medical Center, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| | - Joel D Trinity
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah.,Geriatric Research, Education, and Clinical Center, Veterans Affairs Medical Center, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| |
Collapse
|
86
|
Lindsey ML, Kassiri Z, Hansell Keehan K, Brunt KR, Carter JR, Kirk JA, Kleinbongard P, LeBlanc AJ, Ripplinger CM. We are the change we seek. Am J Physiol Heart Circ Physiol 2021; 320:H1411-H1414. [PMID: 33710925 PMCID: PMC8260391 DOI: 10.1152/ajpheart.00090.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Merry L. Lindsey
- 1Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, Nebraska,2Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Zamaneh Kassiri
- 3Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada
| | - Kara Hansell Keehan
- 4American Journal of Physiology-Heart and Circulatory Physiology, American Physiological Society, Rockville, Maryland
| | - Keith R. Brunt
- 5Department of Pharmacology, Faculty of Medicine, Dalhousie University, Saint John, New Brunswick, Canada
| | - Jason R. Carter
- 6Department of Health and Human Development, Montana State University, Bozeman, Montana
| | - Jonathan A. Kirk
- 7Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois
| | - Petra Kleinbongard
- 8Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - Amanda J. LeBlanc
- 9Department of Physiology and Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky
| | - Crystal M. Ripplinger
- 10Department of Pharmacology, University of California Davis School of Medicine, Davis, California
| |
Collapse
|
87
|
Sousa Neto I, Fontes W, Prestes J, Marqueti R. Impact of paternal exercise on physiological systems in the offspring. Acta Physiol (Oxf) 2021; 231:e13620. [PMID: 33606364 DOI: 10.1111/apha.13620] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/04/2021] [Accepted: 02/01/2021] [Indexed: 02/06/2023]
Abstract
A significant number of studies have demonstrated that paternal exercise modulates future generations via effects on the sperm epigenome. However, comprehensive information regarding the effects of exercise performed by the father on different tissues and their clinical relevance has not yet been explored in detail. This narrative review is focused on the effects of paternal exercise training on various physiological systems of offspring. A detailed mechanistic understanding of these effects could provide crucial clues for the exercise physiology field and aid the development of therapeutic approaches to mitigate disorders in future generations. Non-coding RNA and DNA methylation are major routes for transmitting epigenetic information from parents to offspring. Resistance and treadmill exercise are the most frequently used modalities of planned and structured exercise in controlled experiments. Paternal exercise orchestrated protective effects over changes in fetus development and placenta inflammatory status. Moreover paternal exercise promoted modifications in the ncRNA profiles, gene and protein expression in the hippocampus, left ventricle, skeletal muscle, tendon, liver and pancreas in the offspring, while the transgenerational effects are unknown. Paternal exercise demonstrates clinical benefits to the offspring and provides a warning on the harmful effects of a paternal unhealthy lifestyle. Exercise in fathers is presented as one of the most logical and cost-effective ways of restoring health in the offspring and, consequently, modifying the phenotype. It is important to consider that paternal programming might have unique significance in the developmental origins of offspring diseases.
Collapse
Affiliation(s)
- Ivo Sousa Neto
- Laboratory of Molecular Analysis Graduate Program of Sciences and Technology of Health Faculdade de Ceilândia ‐ Universidade de Brasília Brasília Distrito Federal Brazil
| | - Wagner Fontes
- Laboratory of Protein Chemistry and Biochemistry Department of Cell Biology Institute of Biology Universidade de Brasília Brasília Distrito Federal Brazil
| | - Jonato Prestes
- Graduate Program on Physical Education Universidade Católica de Brasília Brasília Distrito Federal Brazil
| | - Rita Marqueti
- Laboratory of Molecular Analysis Graduate Program of Sciences and Technology of Health Faculdade de Ceilândia ‐ Universidade de Brasília Brasília Distrito Federal Brazil
| |
Collapse
|
88
|
Butenas ALE, Colburn TD, Baumfalk DR, Ade CJ, Hageman KS, Copp SW, Poole DC, Musch TI. Angiotensin converting enzyme inhibition improves cerebrovascular control during exercise in male rats with heart failure. Respir Physiol Neurobiol 2021; 286:103613. [PMID: 33421609 DOI: 10.1016/j.resp.2020.103613] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/21/2020] [Accepted: 12/27/2020] [Indexed: 01/08/2023]
Abstract
We investigated the effects of chronic (∼7 weeks) treatment with the angiotensin converting enzyme (ACE) inhibitor Captopril in rats with heart failure with reduced ejection fraction (HF-rEF) on brain blood flow (BF; radiolabeled microspheres) at rest and during submaximal exercise. We hypothesized that middle cerebral, posterior cerebral, and cerebellar BF during submaximal exercise (20 m/min, 5% incline) would be reduced in rats with HF-rEF (n = 10) compared to healthy (SHAM, n = 10) controls and HF-rEF rats chronically treated with Captopril (HF-rEF + Cap., n = 20). During submaximal exercise middle cerebral (HF-rEF + Cap.: 274 ± 12; HF-rEF: 234 ± 23; SHAM: 248 ± 24 ml/min/100 g) and cerebellar (HF-rEF + Cap.: 222 ± 14; HF-rEF: 243 ± 22; SHAM: 214 ± 23 ml/min/100 g) BF increased from rest in all groups with no difference among groups (P > 0.24). Posterior cerebral BF increased from rest in all groups but was lower than SHAM (394 ± 46 ml/min/100 g; P = 0.03) in HF-rEF (298 ± 19 ml/min/100 g) but not HF-rEF + Cap. (356 ± 18 ml/min/100 g; P = 0.14), supporting the concept that ACE inhibition in HF-rEF elevates brain BF increases, at least to the posterior cerebral region, during moderate intensity exercise/physical activity.
Collapse
Affiliation(s)
- Alec L E Butenas
- Department of Kinesiology, Kansas State University, Manhattan, KS, United States
| | - Trenton D Colburn
- Department of Kinesiology, Kansas State University, Manhattan, KS, United States
| | - Dryden R Baumfalk
- Department of Kinesiology, Kansas State University, Manhattan, KS, United States
| | - Carl J Ade
- Department of Kinesiology, Kansas State University, Manhattan, KS, United States
| | - K Sue Hageman
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, United States
| | - Steven W Copp
- Department of Kinesiology, Kansas State University, Manhattan, KS, United States
| | - David C Poole
- Department of Kinesiology, Kansas State University, Manhattan, KS, United States; Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, United States
| | - Timothy I Musch
- Department of Kinesiology, Kansas State University, Manhattan, KS, United States; Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, United States.
| |
Collapse
|
89
|
Sangha GS, Goergen CJ, Prior SJ, Ranadive SM, Clyne AM. Preclinical techniques to investigate exercise training in vascular pathophysiology. Am J Physiol Heart Circ Physiol 2021; 320:H1566-H1600. [PMID: 33385323 PMCID: PMC8260379 DOI: 10.1152/ajpheart.00719.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Atherosclerosis is a dynamic process starting with endothelial dysfunction and inflammation and eventually leading to life-threatening arterial plaques. Exercise generally improves endothelial function in a dose-dependent manner by altering hemodynamics, specifically by increased arterial pressure, pulsatility, and shear stress. However, athletes who regularly participate in high-intensity training can develop arterial plaques, suggesting alternative mechanisms through which excessive exercise promotes vascular disease. Understanding the mechanisms that drive atherosclerosis in sedentary versus exercise states may lead to novel rehabilitative methods aimed at improving exercise compliance and physical activity. Preclinical tools, including in vitro cell assays, in vivo animal models, and in silico computational methods, broaden our capabilities to study the mechanisms through which exercise impacts atherogenesis, from molecular maladaptation to vascular remodeling. Here, we describe how preclinical research tools have and can be used to study exercise effects on atherosclerosis. We then propose how advanced bioengineering techniques can be used to address gaps in our current understanding of vascular pathophysiology, including integrating in vitro, in vivo, and in silico studies across multiple tissue systems and size scales. Improving our understanding of the antiatherogenic exercise effects will enable engaging, targeted, and individualized exercise recommendations to promote cardiovascular health rather than treating cardiovascular disease that results from a sedentary lifestyle.
Collapse
Affiliation(s)
- Gurneet S Sangha
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana.,Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - Steven J Prior
- Department of Kinesiology, University of Maryland School of Public Health, College Park, Maryland.,Baltimore Veterans Affairs Geriatric Research, Education, and Clinical Center, Baltimore, Maryland
| | - Sushant M Ranadive
- Department of Kinesiology, University of Maryland School of Public Health, College Park, Maryland
| | - Alisa M Clyne
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| |
Collapse
|
90
|
Colburn TD, Weber RE, Hageman KS, Caldwell JT, Schulze KM, Ade CJ, Behnke BJ, Poole DC, Musch TI. Vascular ATP-sensitive K + channels support maximal aerobic capacity and critical speed via convective and diffusive O 2 transport. J Physiol 2020; 598:4843-4858. [PMID: 32798233 PMCID: PMC7874302 DOI: 10.1113/jp280232] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/13/2020] [Indexed: 12/26/2022] Open
Abstract
KEY POINTS Oral sulphonylureas, widely prescribed for diabetes, inhibit pancreatic ATP-sensitive K+ (KATP ) channels to increase insulin release. However, KATP channels are also located within vascular (endothelium and smooth muscle) and muscle (cardiac and skeletal) tissue. We evaluated left ventricular function at rest, maximal aerobic capacity ( V ̇ O2 max) and submaximal exercise tolerance (i.e. speed-duration relationship) during treadmill running in rats, before and after systemic KATP channel inhibition via glibenclamide. Glibenclamide impaired critical speed proportionally more than V ̇ O2 max but did not alter resting cardiac output. Vascular KATP channel function (topical glibenclamide superfused onto hindlimb skeletal muscle) resolved a decreased blood flow and interstitial PO2 during twitch contractions reflecting impaired O2 delivery-to-utilization matching. Our findings demonstrate that systemic KATP channel inhibition reduces V ̇ O2 max and critical speed during treadmill running in rats due, in part, to impaired convective and diffusive O2 delivery, and thus V ̇ O2 , especially within fast-twitch oxidative skeletal muscle. ABSTRACT Vascular ATP-sensitive K+ (KATP ) channels support skeletal muscle blood flow and microvascular oxygen delivery-to-utilization matching during exercise. However, oral sulphonylurea treatment for diabetes inhibits pancreatic KATP channels to enhance insulin release. Herein we tested the hypotheses that: i) systemic KATP channel inhibition via glibenclamide (GLI; 10 mg kg-1 i.p.) would decrease cardiac output at rest (echocardiography), maximal aerobic capacity ( V ̇ O2 max) and the speed-duration relationship (i.e. lower critical speed (CS)) during treadmill running; and ii) local KATP channel inhibition (5 mg kg-1 GLI superfusion) would decrease blood flow (15 µm microspheres), interstitial space oxygen pressures (PO2 is; phosphorescence quenching) and convective and diffusive O2 transport ( Q ̇ O2 and DO2 , respectively; Fick Principle and Law of Diffusion) in contracting fast-twitch oxidative mixed gastrocnemius muscle (MG: 9% type I+IIa fibres). At rest, GLI slowed left ventricular relaxation (2.11 ± 0.59 vs. 1.70 ± 0.23 cm s-1 ) and decreased heart rate (321 ± 23 vs. 304 ± 22 bpm, both P < 0.05) while cardiac output remained unaltered (219 ± 64 vs. 197 ± 39 ml min-1 , P > 0.05). During exercise, GLI reduced V ̇ O2 max (71.5 ± 3.1 vs. 67.9 ± 4.8 ml kg-1 min-1 ) and CS (35.9 ± 2.4 vs. 31.9 ± 3.1 m min-1 , both P < 0.05). Local KATP channel inhibition decreased MG blood flow (52 ± 25 vs. 34 ± 13 ml min-1 100 g tissue-1 ) and PO2 isnadir (5.9 ± 0.9 vs. 4.7 ± 1.1 mmHg) during twitch contractions. Furthermore, MG V ̇ O2 was reduced via impaired Q ̇ O2 and DO2 (P < 0.05 for each). Collectively, these data support that vascular KATP channels help sustain submaximal exercise tolerance in healthy rats. For patients taking sulfonylureas, KATP channel inhibition may exacerbate exercise intolerance.
Collapse
Affiliation(s)
- Trenton D Colburn
- Department of Kinesiology, Kansas State University, Manhattan, KS, 66506, USA
| | - Ramona E Weber
- Department of Kinesiology, Kansas State University, Manhattan, KS, 66506, USA
| | - K Sue Hageman
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, 66506, USA
| | - Jacob T Caldwell
- Department of Kinesiology, Kansas State University, Manhattan, KS, 66506, USA
| | - Kiana M Schulze
- Department of Kinesiology, Kansas State University, Manhattan, KS, 66506, USA
| | - Carl J Ade
- Department of Kinesiology, Kansas State University, Manhattan, KS, 66506, USA
| | - Brad J Behnke
- Department of Kinesiology, Kansas State University, Manhattan, KS, 66506, USA
| | - David C Poole
- Department of Kinesiology, Kansas State University, Manhattan, KS, 66506, USA
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, 66506, USA
| | - Timothy I Musch
- Department of Kinesiology, Kansas State University, Manhattan, KS, 66506, USA
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, 66506, USA
| |
Collapse
|
91
|
Butenas ALE, Smith JR, Copp SW, Sue Hageman K, Poole DC, Musch TI. Type II diabetes accentuates diaphragm blood flow increases during submaximal exercise in the rat. Respir Physiol Neurobiol 2020; 281:103518. [PMID: 32777269 DOI: 10.1016/j.resp.2020.103518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 12/25/2022]
Abstract
We investigated the effect of type 2 diabetes mellitus (T2DM) on respiratory muscle blood flow (BF) during exercise. Using the Goto-Kakizaki (GK) rat model of T2DM, we hypothesized that diaphragm, intercostal and transverse abdominis BFs (radiolabeled microspheres) would be higher in male GK rats (n = 10) compared to healthy male Wistar controls (CON; n = 8) during submaximal exercise (20 m/min, 10 % grade). Blood glucose was significantly higher in GK (246 ± 29 mg/dL) compared to CON (103 ± 4 mg/dL; P < 0.01). Respiratory muscle BFs were not different at rest (P> 0.50). From rest to submaximal exercise, respiratory muscle BFs increased in both groups to all muscles (P < 0.01). During submaximal exercise GK rats had higher diaphragm BFs (GK: 189 ± 13; CON: 138 ± 14 mL/min/100 g, P < 0.01), and vascular conductance (GK: 1.4 ± 0.1; CON: 1.0 ± 0.1 mL/min/mmHg/100 g; P < 0.01) compared to CON. There were no differences in intercostal or transverse abdominis BF or VC during exercise (P> 0.15). These findings suggest that submaximal exercise requires a higher diaphragm BF and VC in T2DM compared to healthy counterparts.
Collapse
Affiliation(s)
- Alec L E Butenas
- Department of Kinesiology, Kansas State University, Manhattan, KS, United States.
| | - Joshua R Smith
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Steven W Copp
- Department of Kinesiology, Kansas State University, Manhattan, KS, United States
| | - K Sue Hageman
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, United States
| | - David C Poole
- Department of Kinesiology, Kansas State University, Manhattan, KS, United States; Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, United States
| | - Timothy I Musch
- Department of Kinesiology, Kansas State University, Manhattan, KS, United States; Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, United States
| |
Collapse
|