101
|
Vega RB, Konhilas JP, Kelly DP, Leinwand LA. Molecular Mechanisms Underlying Cardiac Adaptation to Exercise. Cell Metab 2017; 25:1012-1026. [PMID: 28467921 PMCID: PMC5512429 DOI: 10.1016/j.cmet.2017.04.025] [Citation(s) in RCA: 199] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/13/2017] [Accepted: 04/18/2017] [Indexed: 02/06/2023]
Abstract
Exercise elicits coordinated multi-organ responses including skeletal muscle, vasculature, heart, and lung. In the short term, the output of the heart increases to meet the demand of strenuous exercise. Long-term exercise instigates remodeling of the heart including growth and adaptive molecular and cellular re-programming. Signaling pathways such as the insulin-like growth factor 1/PI3K/Akt pathway mediate many of these responses. Exercise-induced, or physiologic, cardiac growth contrasts with growth elicited by pathological stimuli such as hypertension. Comparing the molecular and cellular underpinnings of physiologic and pathologic cardiac growth has unveiled phenotype-specific signaling pathways and transcriptional regulatory programs. Studies suggest that exercise pathways likely antagonize pathological pathways, and exercise training is often recommended for patients with chronic stable heart failure or following myocardial infarction. Herein, we summarize the current understanding of the structural and functional cardiac responses to exercise as well as signaling pathways and downstream effector molecules responsible for these adaptations.
Collapse
Affiliation(s)
- Rick B Vega
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL 32827, USA
| | - John P Konhilas
- Department of Physiology, Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85724, USA
| | - Daniel P Kelly
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL 32827, USA
| | - Leslie A Leinwand
- Molecular, Cellular and Developmental Biology, BioFrontiers Institute, University of Colorado, Boulder, CO 80309, USA.
| |
Collapse
|
102
|
Effects of high-intensity interval training and moderate-intensity continuous training on glycaemic control and skeletal muscle mitochondrial function in db/db mice. Sci Rep 2017; 7:204. [PMID: 28303003 PMCID: PMC5427962 DOI: 10.1038/s41598-017-00276-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 02/15/2017] [Indexed: 12/22/2022] Open
Abstract
Physical activity is known as an effective strategy for prevention and treatment of Type 2 Diabetes. The aim of this work was to compare the effects of a traditional Moderate Intensity Continuous Training (MICT) with a High Intensity Interval Training (HIIT) on glucose metabolism and mitochondrial function in diabetic mice. Diabetic db/db male mice (N = 25) aged 6 weeks were subdivided into MICT, HIIT or control (CON) group. Animals in the training groups ran on a treadmill 5 days/week during 10 weeks. MICT group ran for 80 min (0° slope) at 50-60% of maximal speed (Vmax) reached during an incremental test. HIIT group ran thirteen times 4 minutes (20° slope) at 85-90% of Vmax separated by 2-min-rest periods. HIIT lowered fasting glycaemia and HbA1c compared with CON group (p < 0.05). In all mitochondrial function markers assessed, no differences were noted between the three groups except for total amount of electron transport chain proteins, slightly increased in the HIIT group vs CON. Western blot analysis revealed a significant increase of muscle Glut4 content (about 2 fold) and higher insulin-stimulated Akt phosphorylation ratios in HIIT group. HIIT seems to improve glucose metabolism more efficiently than MICT in diabetic mice by mechanisms independent of mitochondrial adaptations.
Collapse
|
103
|
Gibb AA, McNally LA, Riggs DW, Conklin DJ, Bhatnagar A, Hill BG. FVB/NJ Mice Are a Useful Model for Examining Cardiac Adaptations to Treadmill Exercise. Front Physiol 2016; 7:636. [PMID: 28066267 PMCID: PMC5174104 DOI: 10.3389/fphys.2016.00636] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/05/2016] [Indexed: 12/17/2022] Open
Abstract
Mice are commonly used to examine the mechanisms by which exercise improves cardiometabolic health; however, exercise compliance and adaptations are often strain-dependent or are variable due to inconsistency in exercise training protocols. In this study, we examined nocturnal/diurnal behavior, treadmill exercise compliance, and systemic as well as cardiac-specific exercise adaptations in two commonly used mouse strains, C57BL/6J, and FVB/NJ mice. Metabolic cage analysis indicated a strong nocturnal nature of C57BL/6J mice, whereas FVB/NJ mice showed no circadian element to activity, food or water intake, VO2, or VCO2. Initial exercise capacity tests revealed that, compared with C57BL/6J mice, FVB/NJ mice are capable of achieving nearly 2-fold higher workloads prior to exhaustion. FVB/NJ mice tested during the day were capable of achieving significantly more work compared with their night-tested counterparts. Following 4 weeks of training, FVB/NJ mice showed significant increases in exercise capacity as well as physiologic cardiac growth characterized by enlarged myocytes and higher mitochondrial DNA content. C57BL/6J mice showed no increases in exercise capacity or cardiac growth regardless of whether they exercised during the day or the night. This lack of adaptation in C57BL/6J mice was attributable, at least in part, to their progressive loss of compliance to the treadmill training protocol. We conclude that the FVB/NJ strain is a useful and robust mouse model for examining cardiac adaptations to treadmill exercise and that treadmill training during daytime hours does not negatively affect exercise compliance or capacity.
Collapse
Affiliation(s)
- Andrew A Gibb
- Department of Medicine, Institute of Molecular Cardiology, University of LouisvilleLouisville, KY, USA; Diabetes and Obesity Center, University of LouisvilleLouisville, KY, USA; Department of Physiology, University of LouisvilleLouisville, KY, USA
| | - Lindsey A McNally
- Department of Medicine, Institute of Molecular Cardiology, University of LouisvilleLouisville, KY, USA; Diabetes and Obesity Center, University of LouisvilleLouisville, KY, USA
| | - Daniel W Riggs
- Department of Medicine, Institute of Molecular Cardiology, University of LouisvilleLouisville, KY, USA; Diabetes and Obesity Center, University of LouisvilleLouisville, KY, USA
| | - Daniel J Conklin
- Department of Medicine, Institute of Molecular Cardiology, University of LouisvilleLouisville, KY, USA; Diabetes and Obesity Center, University of LouisvilleLouisville, KY, USA
| | - Aruni Bhatnagar
- Department of Medicine, Institute of Molecular Cardiology, University of LouisvilleLouisville, KY, USA; Diabetes and Obesity Center, University of LouisvilleLouisville, KY, USA; Department of Physiology, University of LouisvilleLouisville, KY, USA; Department of Biochemistry and Molecular Genetics, University of LouisvilleLouisville, KY, USA
| | - Bradford G Hill
- Department of Medicine, Institute of Molecular Cardiology, University of LouisvilleLouisville, KY, USA; Diabetes and Obesity Center, University of LouisvilleLouisville, KY, USA; Department of Physiology, University of LouisvilleLouisville, KY, USA; Department of Biochemistry and Molecular Genetics, University of LouisvilleLouisville, KY, USA
| |
Collapse
|
104
|
Ivarsson N, Schiffer TA, Hernández A, Lanner JT, Weitzberg E, Lundberg JO, Westerblad H. Dietary nitrate markedly improves voluntary running in mice. Physiol Behav 2016; 168:55-61. [PMID: 27794435 DOI: 10.1016/j.physbeh.2016.10.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 09/05/2016] [Accepted: 10/21/2016] [Indexed: 11/16/2022]
Abstract
Nitrate supplementation is shown to increase submaximal force in human and mouse skeletal muscles. In this study, we test the hypothesis that the increased submaximal force induced by nitrate supplementation reduces the effort of submaximal voluntary running, resulting in increased running speed and distance. C57Bl/6N male mice were fed nitrate in the drinking water and housed with or without access to an in-cage running wheel. Nitrate supplementation in sedentary mice had no effect on endurance in a treadmill test, nor did it enhance mitochondrial function. However, after three weeks with in-cage running wheel, mice fed nitrate ran on average 20% faster and 30% further than controls (p<0.01). Compared to running controls, this resulted in ~13% improved endurance on a subsequent treadmill test (p<0.05) and increased mitochondrial oxidative capacity, as judged from a mean increase in citrate synthase activity of 14% (p<0.05). After six weeks with nitrate, the mice were running 58% longer distances per night. When nitrate supplementation was removed from the diet, the running distance and speed decreased to the control level, despite the improved endurance achieved during nitrate supplementation. In conclusion, low-frequency force improvement due to nitrate supplementation facilitates submaximal exercise such as voluntary running.
Collapse
Affiliation(s)
- Niklas Ivarsson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| | - Tomas A Schiffer
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Andrés Hernández
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Johanna T Lanner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Håkan Westerblad
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
105
|
Rodrigues NA, Torsoni AS, Fante T, Dos Reis IGM, Gobatto CA, Manchado-Gobatto FB. Lactate minimum underestimates the maximal lactate steady-state in swimming mice. Appl Physiol Nutr Metab 2016; 42:46-52. [PMID: 28006434 DOI: 10.1139/apnm-2016-0198] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The intensity of lactate minimum (LM) has presented a good estimate of the intensity of maximal lactate steady-state (MLSS); however, this relationship has not yet been verified in the mouse model. We proposed validating the LM protocol for swimming mice by investigating the relationship among intensities of LM and MLSS as well as differences between sexes, in terms of aerobic capacity. Nineteen mice (male: 10, female: 9) were submitted to the evaluation protocols for LM and MLSS. The LM protocol consisted of hyperlactatemia induction (30 s exercise (13% body mass (bm)), 30 s resting pause and exhaustive exercise (13% bm), 9 min resting pause and incremental test). The LM underestimated MLSS (mice: 17.6%; male: 13.5%; female: 21.6%). Pearson's analysis showed a strong correlation among intensities of MLSS and LM (male (r = 0.67, p = 0.033); female (r = 0.86, p = 0.003)), but without agreement between protocols. The Bland-Altman analysis showed that bias was higher for females (1.5 (0.98) % bm; mean (MLSS and LM): 4.4%-6.4% bm) as compared with males (0.84 (1.24) % bm; mean (MLSS and LM): 4.5%-7.5% bm). The error associated with the estimated of intensity for males was lower when compared with the range of means for MLSS and LM. Therefore, the LM test could be used to determine individual aerobic intensity for males (considering the bias) but not females. Furthermore, the females supported higher intensities than the males. The differences in body mass between sexes could not explain the higher intensities supported by the females.
Collapse
Affiliation(s)
- Natalia Almeida Rodrigues
- a Laboratory of Applied Sports Physiology, School of Applied Sciences, University of Campinas, Limeira, São Paulo, Brazil
| | - Adriana Souza Torsoni
- b Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas, Limeira, São Paulo, Brazil
| | - Thais Fante
- b Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas, Limeira, São Paulo, Brazil
| | - Ivan Gustavo Masselli Dos Reis
- a Laboratory of Applied Sports Physiology, School of Applied Sciences, University of Campinas, Limeira, São Paulo, Brazil
| | - Claudio Alexandre Gobatto
- a Laboratory of Applied Sports Physiology, School of Applied Sciences, University of Campinas, Limeira, São Paulo, Brazil
| | - Fúlvia Barros Manchado-Gobatto
- a Laboratory of Applied Sports Physiology, School of Applied Sciences, University of Campinas, Limeira, São Paulo, Brazil
| |
Collapse
|
106
|
Bowen TS, Eisenkolb S, Drobner J, Fischer T, Werner S, Linke A, Mangner N, Schuler G, Adams V. High-intensity interval training prevents oxidant-mediated diaphragm muscle weakness in hypertensive mice. FASEB J 2016; 31:60-71. [PMID: 27650398 DOI: 10.1096/fj.201600672r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/07/2016] [Indexed: 01/07/2023]
Abstract
Hypertension is a key risk factor for heart failure, with the latter characterized by diaphragm muscle weakness that is mediated in part by increased oxidative stress. In the present study, we used a deoxycorticosterone acetate (DOCA)-salt mouse model to determine whether hypertension could independently induce diaphragm dysfunction and further investigated the effects of high-intensity interval training (HIIT). Sham-treated (n = 11), DOCA-salt-treated (n = 11), and DOCA-salt+HIIT-treated (n = 15) mice were studied over 4 wk. Diaphragm contractile function, protein expression, enzyme activity, and fiber cross-sectional area and type were subsequently determined. Elevated blood pressure confirmed hypertension in DOCA-salt mice independent of HIIT (P < 0.05). Diaphragm forces were impaired by ∼15-20% in DOCA-salt vs. sham-treated mice (P < 0.05), but this effect was prevented after HIIT. Myosin heavy chain (MyHC) protein expression tended to decrease (∼30%; P = 0.06) in DOCA-salt vs. sham- and DOCA-salt+HIIT mice, whereas oxidative stress increased (P < 0.05). Enzyme activity of NADPH oxidase was higher, but superoxide dismutase was lower, with MyHC oxidation elevated by ∼50%. HIIT further prevented direct oxidant-mediated diaphragm contractile dysfunction (P < 0.05) after a 30 min exposure to H2O-2 (1 mM). Our data suggest that hypertension induces diaphragm contractile dysfunction via an oxidant-mediated mechanism that is prevented by HIIT.-Bowen, T. S., Eisenkolb, S., Drobner, J., Fischer, T., Werner, S., Linke, A., Mangner, N., Schuler, G., Adams, V. High-intensity interval training prevents oxidant-mediated diaphragm muscle weakness in hypertensive mice.
Collapse
Affiliation(s)
- T Scott Bowen
- Department of Internal Medicine and Cardiology, Leipzig University Heart Center, Leipzig, Germany
| | - Sophia Eisenkolb
- Department of Internal Medicine and Cardiology, Leipzig University Heart Center, Leipzig, Germany
| | - Juliane Drobner
- Department of Internal Medicine and Cardiology, Leipzig University Heart Center, Leipzig, Germany
| | - Tina Fischer
- Department of Internal Medicine and Cardiology, Leipzig University Heart Center, Leipzig, Germany
| | - Sarah Werner
- Department of Internal Medicine and Cardiology, Leipzig University Heart Center, Leipzig, Germany
| | - Axel Linke
- Department of Internal Medicine and Cardiology, Leipzig University Heart Center, Leipzig, Germany
| | - Norman Mangner
- Department of Internal Medicine and Cardiology, Leipzig University Heart Center, Leipzig, Germany
| | - Gerhard Schuler
- Department of Internal Medicine and Cardiology, Leipzig University Heart Center, Leipzig, Germany
| | - Volker Adams
- Department of Internal Medicine and Cardiology, Leipzig University Heart Center, Leipzig, Germany
| |
Collapse
|
107
|
Carpentieri A, Gamberi T, Modesti A, Amoresano A, Colombini B, Nocella M, Bagni MA, Fiaschi T, Barolo L, Gulisano M, Magherini F. Profiling Carbonylated Proteins in Heart and Skeletal Muscle Mitochondria from Trained and Untrained Mice. J Proteome Res 2016; 15:3666-3678. [PMID: 27571187 DOI: 10.1021/acs.jproteome.6b00475] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Understanding the relationship between physical exercise, reactive oxygen species, and skeletal muscle modification is important in order to better identify the benefits or the damages that appropriate or inappropriate exercise can induce. Heart and skeletal muscles have a high density of mitochondria with robust energetic demands, and mitochondria plasticity has an important role in both the cardiovascular system and skeletal muscle responses. The aim of this study was to investigate the influence of regular physical activity on the oxidation profiles of mitochondrial proteins from heart and tibialis anterior muscles. To this end, we used the mouse as animal model. Mice were divided into two groups: untrained and regularly trained. The carbonylated protein pattern was studied by two-dimensional gel electrophoresis followed by Western blot with anti-dinitrophenyl hydrazone antibodies. Mass spectrometry analysis allowed the identification of several different protein oxidation sites, including methionine, cysteine, proline, and leucine residues. A large number of oxidized proteins were found in both untrained and trained animals. Moreover, mitochondria from skeletal muscles and heart showed almost the same carbonylation pattern. Interestingly, exercise training seems to increase the carbonylation level mainly of mitochondrial proteins from skeletal muscle.
Collapse
Affiliation(s)
- Andrea Carpentieri
- Department of Chemical Sciences, Federico II University, Complesso Universitario Monte Sant'Angelo , Via Cinthia 4, 80126 Naples, Italy
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence , Viale G.B. Morgagni 50, Florence, 50134 Italy
| | - Alessandra Modesti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence , Viale G.B. Morgagni 50, Florence, 50134 Italy
| | - Angela Amoresano
- Department of Chemical Sciences, Federico II University, Complesso Universitario Monte Sant'Angelo , Via Cinthia 4, 80126 Naples, Italy
| | - Barbara Colombini
- Department of Experimental and Clinical Medicine, University of Florence , Viale G.B. Morgagni 63, 50134 Florence, Italy
| | - Marta Nocella
- Department of Experimental and Clinical Medicine, University of Florence , Viale G.B. Morgagni 63, 50134 Florence, Italy
| | - Maria Angela Bagni
- Department of Experimental and Clinical Medicine, University of Florence , Viale G.B. Morgagni 63, 50134 Florence, Italy
| | - Tania Fiaschi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence , Viale G.B. Morgagni 50, Florence, 50134 Italy
| | - Lorenzo Barolo
- Department of Chemical Sciences, Federico II University, Complesso Universitario Monte Sant'Angelo , Via Cinthia 4, 80126 Naples, Italy
| | - Massimo Gulisano
- Department of Experimental and Clinical Medicine, University of Florence , Viale G.B. Morgagni 63, 50134 Florence, Italy
| | - Francesca Magherini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence , Viale G.B. Morgagni 50, Florence, 50134 Italy
| |
Collapse
|
108
|
Ayachi M, Niel R, Momken I, Billat VL, Mille-Hamard L. Validation of a Ramp Running Protocol for Determination of the True VO2max in Mice. Front Physiol 2016; 7:372. [PMID: 27621709 PMCID: PMC5002025 DOI: 10.3389/fphys.2016.00372] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 08/12/2016] [Indexed: 01/11/2023] Open
Abstract
In the field of comparative physiology, it remains to be established whether the concept of VO2max is valid in the mouse and, if so, how this value can be accurately determined. In humans, VO2max is generally considered to correspond to the plateau observed when VO2 no longer rises with an increase in workload. In contrast, the concept of VO2peak tends to be used in murine studies. The objectives of the present study were to determine whether (i) a continuous ramp protocol yielded a higher VO2peak than a stepwise, incremental protocol, and (ii) the VO2peak measured in the ramp protocol corresponded to VO2max. The three protocols (based on intensity-controlled treadmill running until exhaustion with eight female FVB/N mice) were performed in random order: (a) an incremental protocol that begins at 10 m.min−1 speed and increases by 3 m.min−1 every 3 min. (b) a ramp protocol with slow acceleration (3 m.min−2), and (c) a ramp protocol with fast acceleration (12 m.min−2). Each protocol was performed with two slopes (0 and 25°). Hence, each mouse performed six exercise tests. We found that the value of VO2peak was protocol-dependent (p < 0.05) and was highest (59.0 ml.kg 0.75.min−1) for the 3 m.min−2 0° ramp protocol. In the latter, the presence of a VO2max plateau was associated with the fulfillment of two secondary criteria (a blood lactate concentration >8 mmol.l−1 and a respiratory exchange ratio >1). The total duration of the 3 m.min−2 0° ramp protocol was shorter than that of the incremental protocol. Taken as a whole, our results suggest that VO2max in the mouse is best determined by applying a ramp exercise protocol with slow acceleration and no treadmill slope.
Collapse
Affiliation(s)
- Mohamed Ayachi
- Unité de Biologie Intégrative des Adaptations à l'Exercice, Université d'Evry Val d'Essonne Evry, France
| | - Romain Niel
- Unité de Biologie Intégrative des Adaptations à l'Exercice, Université d'Evry Val d'Essonne Evry, France
| | - Iman Momken
- Unité de Biologie Intégrative des Adaptations à l'Exercice, Université d'Evry Val d'Essonne Evry, France
| | - Véronique L Billat
- Unité de Biologie Intégrative des Adaptations à l'Exercice, Université d'Evry Val d'Essonne Evry, France
| | - Laurence Mille-Hamard
- Unité de Biologie Intégrative des Adaptations à l'Exercice, Université d'Evry Val d'Essonne Evry, France
| |
Collapse
|
109
|
de Carvalho FP, Benfato ID, Moretto TL, Barthichoto M, de Oliveira CAM. Voluntary running decreases nonexercise activity in lean and diet-induced obese mice. Physiol Behav 2016; 165:249-56. [PMID: 27497922 DOI: 10.1016/j.physbeh.2016.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 08/02/2016] [Accepted: 08/03/2016] [Indexed: 12/18/2022]
Abstract
PURPOSE Determine whether voluntary wheel running triggers compensatory changes in nonexercise activity in lean and high-fat diet fed mice. METHODS C57Bl/6 mice received a control (C) or a high-fat diet (H) and half of them had free access to a running wheel 5days/week (CE and HE, respectively) for 10weeks. Energy intake, nonexercise activity (global activity, distance covered and average speed of displacement in the home cage) and energy expenditure (EE) were evaluated at weeks 5 and 10 during the 2days without the wheels. RESULTS High-fat diet increased weight gain in H (110%) and HE (60%) groups compared to C and CE groups, respectively, with no effect of exercise. Wheel running increased energy intake (26% CE, 11% HE in week 5; 7% CE, 45% HE in week 10) and decreased distance covered (26% for both CE and HE in week 5; 35% CE and 13% HE in week 10) and average speed (35% CE and 13% HE in week 5; 45% CE and 18% HE in week 10) compared to the respective nonexercised groups. In week 10 there was an interaction between diet and exercise for global activity, which was reduced nearly 18% in CE, H, and HE groups compared to C. Access to a running wheel increased EE in week 5 (11% CE and 16% HE) but not in week 10, which is consistent with the period of highest running (number of turns: weeks 1-5 nearly 100%>weeks 6-10 for CE and HE groups). EE was reduced in H (19%) and HE (12%) groups compared to C and CE, in week 10. CONCLUSION Voluntary running causes a compensatory decrease in nonexercise activity and an increase in energy intake, both contributing to the lack of effect of exercise on body mass.
Collapse
Affiliation(s)
| | - Izabelle Dias Benfato
- Interdisciplinary Graduate Program in Health Sciences, Federal University of Sao Paulo, Santos, SP, Brazil
| | - Thaís Ludmilla Moretto
- Interdisciplinary Graduate Program in Health Sciences, Federal University of Sao Paulo, Santos, SP, Brazil
| | - Marcela Barthichoto
- Graduate Program in Food, Nutrition and Health, Federal University of Sao Paulo, Santos, SP, Brazil
| | | |
Collapse
|
110
|
Cattin ME, Ferry A, Vignaud A, Mougenot N, Jacquet A, Wahbi K, Bertrand AT, Bonne G. Mutation in lamin A/C sensitizes the myocardium to exercise-induced mechanical stress but has no effect on skeletal muscles in mouse. Neuromuscul Disord 2016; 26:490-9. [DOI: 10.1016/j.nmd.2016.05.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 05/18/2016] [Indexed: 12/11/2022]
|
111
|
Constans A, Pin-Barre C, Temprado JJ, Decherchi P, Laurin J. Influence of Aerobic Training and Combinations of Interventions on Cognition and Neuroplasticity after Stroke. Front Aging Neurosci 2016; 8:164. [PMID: 27445801 PMCID: PMC4928497 DOI: 10.3389/fnagi.2016.00164] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 06/21/2016] [Indexed: 12/17/2022] Open
Abstract
Stroke often aggravated age-related cognitive impairments that strongly affect several aspects of quality of life. However, few studies are, to date, focused on rehabilitation strategies that could improve cognition. Among possible interventions, aerobic training is well known to enhance cardiovascular and motor functions but may also induce beneficial effects on cognitive functions. To assess the effectiveness of aerobic training on cognition, it seems necessary to know whether training promotes the neuroplasticity in brain areas involved in cognitive functions. In the present review, we first explore in both human and animal how aerobic training could improve cognition after stroke by highlighting the neuroplasticity mechanisms. Then, we address the potential effect of combinations between aerobic training with other interventions, including resistance exercises and pharmacological treatments. In addition, we postulate that classic recommendations for aerobic training need to be reconsidered to target both cognition and motor recovery because the current guidelines are only focused on cardiovascular and motor recovery. Finally, methodological limitations of training programs and cognitive function assessment are also developed in this review to clarify their effectiveness in stroke patients.
Collapse
Affiliation(s)
| | - Caroline Pin-Barre
- Aix-Marseille Université, CNRS, ISM, UMR 7287Marseille, France; Université Nice Sophia Antipolis, LAMHESS, UPRES EA 6309Nice, France
| | | | | | - Jérôme Laurin
- Aix-Marseille Université, CNRS, ISM, UMR 7287 Marseille, France
| |
Collapse
|
112
|
Høydal MA, Stølen TO, Kettlewell S, Maier LS, Brown JH, Sowa T, Catalucci D, Condorelli G, Kemi OJ, Smith GL, Wisløff U. Exercise training reverses myocardial dysfunction induced by CaMKIIδC overexpression by restoring Ca2+ homeostasis. J Appl Physiol (1985) 2016; 121:212-20. [PMID: 27231311 DOI: 10.1152/japplphysiol.00188.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 05/20/2016] [Indexed: 11/22/2022] Open
Abstract
Several conditions of heart disease, including heart failure and diabetic cardiomyopathy, are associated with upregulation of cytosolic Ca(2+)/calmodulin-dependent protein kinase II (CaMKIIδC) activity. In the heart, CaMKIIδC isoform targets several proteins involved in intracellular Ca(2+) homeostasis. We hypothesized that high-intensity endurance training activates mechanisms that enable a rescue of dysfunctional cardiomyocyte Ca(2+) handling and thereby ameliorate cardiac dysfunction despite continuous and chronic elevated levels of CaMKIIδC CaMKIIδC transgenic (TG) and wild-type (WT) mice performed aerobic interval exercise training over 6 wk. Cardiac function was measured by echocardiography in vivo, and cardiomyocyte shortening and intracellular Ca(2+) handling were measured in vitro. TG mice had reduced global cardiac function, cardiomyocyte shortening (47% reduced compared with WT, P < 0.01), and impaired Ca(2+) homeostasis. Despite no change in the chronic elevated levels of CaMKIIδC, exercise improved global cardiac function, restored cardiomyocyte shortening, and reestablished Ca(2+) homeostasis to values not different from WT. The key features to explain restored Ca(2+) homeostasis after exercise training were increased L-type Ca(2+) current density and flux by 79 and 85%, respectively (P < 0.01), increased sarcoplasmic reticulum (SR) Ca(2+)-ATPase (SERCA2a) function by 50% (P < 0.01), and reduced diastolic SR Ca(2+) leak by 73% (P < 0.01), compared with sedentary TG mice. In conclusion, exercise training improves global cardiac function as well as cardiomyocyte function in the presence of a maintained high CaMKII activity. The main mechanisms of exercise-induced improvements in TG CaMKIIδC mice are mediated via increased L-type Ca(2+) channel currents and improved SR Ca(2+) handling by restoration of SERCA2a function in addition to reduced diastolic SR Ca(2+) leak.
Collapse
Affiliation(s)
- Morten A Høydal
- Norwegian University of Science and Technology, K. G. Jebsen Centre of Exercise in Medicine, Trondheim, Norway;
| | - Tomas O Stølen
- Norwegian University of Science and Technology, K. G. Jebsen Centre of Exercise in Medicine, Trondheim, Norway
| | - Sarah Kettlewell
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Lars S Maier
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | | | - Tomas Sowa
- Heart Center of the University of Göttingen, Göttingen, Germany; and
| | - Daniele Catalucci
- National Research Council, Institute of Genetic and Biomedical Research-UOS Milan and Humanitas Research Hospital, Milan, Italy
| | - Gianluigi Condorelli
- National Research Council, Institute of Genetic and Biomedical Research-UOS Milan and Humanitas Research Hospital, Milan, Italy
| | - Ole J Kemi
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Godfrey L Smith
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Ulrik Wisløff
- Norwegian University of Science and Technology, K. G. Jebsen Centre of Exercise in Medicine, Trondheim, Norway
| |
Collapse
|
113
|
Dual Modulation of Nociception and Cardiovascular Reflexes during Peripheral Ischemia through P2Y1 Receptor-Dependent Sensitization of Muscle Afferents. J Neurosci 2016; 36:19-30. [PMID: 26740646 DOI: 10.1523/jneurosci.2856-15.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
UNLABELLED Numerous musculoskeletal pain disorders are based in dysfunction of peripheral perfusion and are often comorbid with altered cardiovascular responses to muscle contraction/exercise. We have recently found in mice that 24 h peripheral ischemia induced by a surgical occlusion of the brachial artery (BAO) induces increased paw-guarding behaviors, mechanical hypersensitivity, and decreased grip strength. These behavioral changes corresponded to increased heat sensitivity as well as an increase in the numbers of chemosensitive group III/IV muscle afferents as assessed by an ex vivo forepaw muscles/median and ulnar nerves/dorsal root ganglion (DRG)/spinal cord (SC) recording preparation. Behaviors also corresponded to specific upregulation of the ADP-responsive P2Y1 receptor in the DRGs. Since group III/IV muscle afferents have separately been associated with regulating muscle nociception and exercise pressor reflexes (EPRs), and P2Y1 has been linked to heat responsiveness and phenotypic switching in cutaneous afferents, we sought to determine whether upregulation of P2Y1 was responsible for the observed alterations in muscle afferent function, leading to modulation of muscle pain-related behaviors and EPRs after BAO. Using an afferent-specific siRNA knockdown strategy, we found that inhibition of P2Y1 during BAO not only prevented the increased mean blood pressure after forced exercise, but also significantly reduced alterations in pain-related behaviors. Selective P2Y1 knockdown also prevented the increased firing to heat stimuli and the BAO-induced phenotypic switch in chemosensitive muscle afferents, potentially through regulating membrane expression of acid sensing ion channel 3. These results suggest that enhanced P2Y1 in muscle afferents during ischemic-like conditions may dually regulate muscle nociception and cardiovascular reflexes. SIGNIFICANCE STATEMENT Our current results suggest that P2Y1 modulates heat responsiveness and chemosensation in muscle afferents to play a key role in the development of pain-related behaviors during ischemia. At the same time, under these pathological conditions, the changes in muscle sensory neurons appear to modulate an increase in mean systemic blood pressure after exercise. This is the first report of the potential peripheral mechanisms by which group III/IV muscle afferents can dually regulate muscle nociception and the exercise pressor reflex. These data provide evidence related to the potential underlying reasons for the comorbidity of muscle pain and altered sympathetic reflexes in disease states that are based in problems with peripheral perfusion and may indicate a potential target for therapeutic intervention.
Collapse
|
114
|
Manotheepan R, Danielsen TK, Sadredini M, Anderson ME, Carlson CR, Lehnart SE, Sjaastad I, Stokke MK. Exercise training prevents ventricular tachycardia in CPVT1 due to reduced CaMKII-dependent arrhythmogenic Ca2+ release. Cardiovasc Res 2016; 111:295-306. [PMID: 27161030 DOI: 10.1093/cvr/cvw095] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 05/01/2016] [Indexed: 01/05/2023] Open
Abstract
AIMS Catecholaminergic polymorphic ventricular tachycardia type 1 (CPVT1) is caused by mutations in the cardiac ryanodine receptor (RyR2) that lead to disrupted Ca(2+) handling in cardiomyocytes and ventricular tachycardia. The aim of this study was to test whether exercise training could reduce the propensity for arrhythmias in mice with the CPVT1-causative missense mutation Ryr2-R2474S by restoring normal Ca(2+) handling. METHODS AND RESULTS Ryr2-R2474S mice (RyR-RS) performed a 2 week interval treadmill exercise training protocol. Each exercise session comprised five 8 min intervals at 80-90% of the running speed at maximal oxygen uptake (VO2max) and 2 min active rest periods at 60%. VO2max increased by 10 ± 2% in exercise trained RyR-RS (ET), while no changes were found in sedentary controls (SED). RyR-RS ET showed fewer episodes of ventricular tachycardia compared with RyR-RS SED, coinciding with fewer Ca(2+) sparks and waves, less diastolic Ca(2+) leak from the sarcoplasmic reticulum, and lower phosphorylation levels at RyR2 sites associated with Ca(2) (+)-calmodulin-dependent kinase type II (CaMKII) compared with RyR-RS SED. The CaMKII inhibitor autocamtide-2-related inhibitory peptide and also the antioxidant N-acetyl-l-cysteine reduced Ca(2+) wave frequency in RyR-RS equally to exercise training. Protein analysis as well as functional data indicated a mechanism depending on reduced levels of oxidized CaMKII after exercise training. Two weeks of detraining reversed the beneficial effects of the interval treadmill exercise training protocol in RyR-RS ET. CONCLUSION Long-term effects of interval treadmill exercise training reduce ventricular tachycardia episodes in mice with a CPVT1-causative Ryr2 mutation through lower CaMKII-dependent phosphorylation of RyR2.
Collapse
Affiliation(s)
- Ravinea Manotheepan
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Tore K Danielsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Mani Sadredini
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Mark E Anderson
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cathrine R Carlson
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Stephan E Lehnart
- Clinic of Cardiology and Pulmonology, Heart Research Center Göttingen, Göttingen, Germany
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Mathis K Stokke
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway Center for Heart Failure Research, University of Oslo, Oslo, Norway Clinic for Internal Medicine, Lovisenberg Diakonale Hospital, Oslo, Norway
| |
Collapse
|
115
|
Physical activity and exercise attenuate neuroinflammation in neurological diseases. Brain Res Bull 2016; 125:19-29. [PMID: 27021169 DOI: 10.1016/j.brainresbull.2016.03.012] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 03/17/2016] [Accepted: 03/22/2016] [Indexed: 12/15/2022]
Abstract
Major depressive disorder (MDD), schizophrenia (SCH), Alzheimer's disease (AD), and Parkinson's disease (PD) are devastating neurological disorders, which increasingly contribute to global morbidity and mortality. Although the pathogenic mechanisms of these conditions are quite diverse, chronic neuroinflammation is one underlying feature shared by all these diseases. Even though the specific root causes of these diseases remain to be identified, evidence indicates that the observed neuroinflammation is initiated by unique pathological features associated with each specific disease. If the initial acute inflammation is not resolved, a chronic neuroinflammatory state develops and ultimately contributes to disease progression. Chronic neuroinflammation is characterized by adverse and non-specific activation of glial cells, which can lead to collateral damage of nearby neurons and other glia. This misdirected neuroinflammatory response is hypothesized to contribute to neuropathology in MDD, SCH, AD, and PD. Physical activity (PA), which is critical for maintenance of whole body and brain health, may also beneficially modify neuroimmune responses. Since PA has neuroimmune-modifying properties, and the common underlying feature of MDD, SCH, AD, and PD is chronic neuroinflammation, we hypothesize that PA could minimize brain diseases by modifying glia-mediated neuroinflammation. This review highlights current evidence supporting the disease-altering potential of PA and exercise through modifications of neuroimmune responses, specifically in MDD, SCH, AD and PD.
Collapse
|
116
|
Hasumura T, Meguro S. Exercise quantity-dependent muscle hypertrophy in adult zebrafish (Danio rerio). J Comp Physiol B 2016; 186:603-14. [PMID: 26951149 DOI: 10.1007/s00360-016-0977-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 02/16/2016] [Accepted: 02/24/2016] [Indexed: 01/28/2023]
Abstract
Exercise is very important for maintaining and increasing skeletal muscle mass, and is particularly important to prevent and care for sarcopenia and muscle disuse atrophy. However, the dose-response relationship between exercise quantity, duration/day, and overall duration and muscle mass is poorly understood. Therefore, we investigated the effect of exercise duration on skeletal muscle to reveal the relationship between exercise quantity and muscle hypertrophy in zebrafish forced to exercise. Adult male zebrafish were exercised 6 h/day for 4 weeks, 6 h/day for 2 weeks, or 3 h/day for 2 weeks. Flow velocity was adjusted to maximum velocity during continual swimming (initial 43 cm/s). High-speed consecutive photographs revealed that zebrafish mainly drove the caudal part. Additionally, X-ray micro computed tomography measurements indicated muscle hypertrophy of the mid-caudal half compared with the mid-cranial half part. The cross-sectional analysis of the mid-caudal half muscle revealed that skeletal muscle (red, white, or total) mass increased with increasing exercise quantity, whereas that of white muscle and total muscle increased only under the maximum exercise load condition of 6 h/day for 4 weeks. Additionally, the muscle fiver size distributions of exercised fish were larger than those from non-exercised fish. We revealed that exercise quantity, duration/day, and overall duration were correlated with skeletal muscle hypertrophy. The forced exercise model enabled us to investigate the relationship between exercise quantity and skeletal muscle mass. These results open up the possibility for further investigations on the effects of exercise on skeletal muscle in adult zebrafish.
Collapse
Affiliation(s)
- Takahiro Hasumura
- Biological Science Research, Kao Corporation, 2606 Akabane, Ichikai-machi, Haga-gun, Tochigi, 321-3497, Japan
| | - Shinichi Meguro
- Biological Science Research, Kao Corporation, 2606 Akabane, Ichikai-machi, Haga-gun, Tochigi, 321-3497, Japan.
| |
Collapse
|
117
|
Gabriel BM, Hamilton DL, Tremblay AM, Wackerhage H. The Hippo signal transduction network for exercise physiologists. J Appl Physiol (1985) 2016; 120:1105-17. [PMID: 26940657 DOI: 10.1152/japplphysiol.01076.2015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/02/2016] [Indexed: 12/20/2022] Open
Abstract
The ubiquitous transcriptional coactivators Yap (gene symbol Yap1) and Taz (gene symbol Wwtr1) regulate gene expression mainly by coactivating the Tead transcription factors. Being at the center of the Hippo signaling network, Yap and Taz are regulated by the Hippo kinase cassette and additionally by a plethora of exercise-associated signals and signaling modules. These include mechanotransduction, the AKT-mTORC1 network, the SMAD transcription factors, hypoxia, glucose homeostasis, AMPK, adrenaline/epinephrine and angiotensin II through G protein-coupled receptors, and IL-6. Consequently, exercise should alter Hippo signaling in several organs to mediate at least some aspects of the organ-specific adaptations to exercise. Indeed, Tead1 overexpression in muscle fibers has been shown to promote a fast-to-slow fiber type switch, whereas Yap in muscle fibers and cardiomyocytes promotes skeletal muscle hypertrophy and cardiomyocyte adaptations, respectively. Finally, genome-wide association studies in humans have linked the Hippo pathway members LATS2, TEAD1, YAP1, VGLL2, VGLL3, and VGLL4 to body height, which is a key factor in sports.
Collapse
Affiliation(s)
- Brendan M Gabriel
- School of Medicine, Dentistry and Nutrition, University of Aberdeen, Scotland, UK; The Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Integrative Physiology, University of Copenhagen, Denmark; and Integrative physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Annie M Tremblay
- Stem Cell Program, Children's Hospital, Boston, Massachusetts; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts; Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Henning Wackerhage
- School of Medicine, Dentistry and Nutrition, University of Aberdeen, Scotland, UK; Faculty of Sport and Health Science, Technical University Munich, Germany;
| |
Collapse
|
118
|
Andrew JR, Saltzman W, Chappell MA, Garland T. Consequences of Fatherhood in the Biparental California Mouse (Peromyscus californicus): Locomotor Performance, Metabolic Rate, and Organ Masses. Physiol Biochem Zool 2016; 89:130-40. [PMID: 27082723 DOI: 10.1086/685435] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although effects of motherhood on mothers have been well documented in mammals, the effects of fatherhood on fathers are not well known. We evaluated effects of being a father on key metabolic and performance measures in the California mouse, Peromyscus californicus. California mice are genetically monogamous in the wild, and fathers show similar parental behavior to mothers, with the exception of lactation. To investigate the impact of fatherhood on fathers, focal males were paired with an intact female (breeding males), a tubally ligated female (nonbreeding males), or another male (virgins). Starting 3-5 d after the birth of each breeding pair's first litter, males were tested for locomotor performance (maximum sprint speed, treadmill endurance), basal metabolic rate (BMR), and maximum oxygen consumption ([Formula: see text]). At the end of the 11-d test period, mice were euthanized, hematocrit was determined, and organs were weighed. Speed, endurance, and [Formula: see text] were significantly repeatable between two replicate measurement days but did not differ among groups, nor did BMR. Breeding males had significantly larger hind limb muscles than did nonbreeding males, whereas virgin males had heavier subcutaneous fat pads than did nonbreeding and breeding males. Several correlations were observed at the level of individual variation (residuals from ANCOVA models), including positive correlations for endurance with [Formula: see text], [Formula: see text] with testes mass, and some of the digestion-related organs with each other. These results indicate that fatherhood may not have pronounced performance, metabolic, or morphological effects on fathers, at least under standard laboratory conditions and across a single breeding cycle.
Collapse
|
119
|
Petrosino JM, Heiss VJ, Maurya SK, Kalyanasundaram A, Periasamy M, LaFountain RA, Wilson JM, Simonetti OP, Ziouzenkova O. Graded Maximal Exercise Testing to Assess Mouse Cardio-Metabolic Phenotypes. PLoS One 2016; 11:e0148010. [PMID: 26859763 PMCID: PMC4747552 DOI: 10.1371/journal.pone.0148010] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 01/12/2016] [Indexed: 12/22/2022] Open
Abstract
Functional assessments of cardiovascular fitness (CVF) are needed to establish animal models of dysfunction, test the effects of novel therapeutics, and establish the cardio-metabolic phenotype of mice. In humans, the graded maximal exercise test (GXT) is a standardized diagnostic for assessing CVF and mortality risk. These tests, which consist of concurrent staged increases in running speed and inclination, provide diagnostic cardio-metabolic parameters, such as, VO2max, anaerobic threshold, and metabolic crossover. Unlike the human-GXT, published mouse treadmill tests have set, not staged, increases in inclination as speed progress until exhaustion (PXT). Additionally, they often lack multiple cardio-metabolic parameters. Here, we developed a mouse-GXT with the intent of improving mouse-exercise testing sensitivity and developing translatable parameters to assess CVF in healthy and dysfunctional mice. The mouse-GXT, like the human-GXT, incorporated staged increases in inclination, speed, and intensity; and, was designed by considering imitations of the PXT and differences between human and mouse physiology. The mouse-GXT and PXTs were both tested in healthy mice (C57BL/6J, FVBN/J) to determine their ability to identify cardio-metabolic parameters (anaerobic threshold, VO2max, metabolic crossover) observed in human-GXTs. Next, theses assays were tested on established diet-induced (obese-C57BL/6J) and genetic (cardiac isoform Casq2-/-) models of cardiovascular dysfunction. Results showed that both tests reported VO2max and provided reproducible data about performance. Only the mouse-GXT reproducibly identified anaerobic threshold, metabolic crossover, and detected impaired CVF in dysfunctional models. Our findings demonstrated that the mouse-GXT is a sensitive, non-invasive, and cost-effective method for assessing CVF in mice. This new test can be used as a functional assessment to determine the cardio-metabolic phenotype of various animal models or the effects of novel therapeutics.
Collapse
Affiliation(s)
- Jennifer M. Petrosino
- Department of Human Sciences, The Ohio State University, College of Education & Human Ecology, Columbus, Ohio, United States of America
- Biomedical Sciences Program, The Ohio State University, College of Medicine, Columbus, Ohio, United States of America
| | - Valerie J. Heiss
- Department of Human Sciences, The Ohio State University, College of Education & Human Ecology, Columbus, Ohio, United States of America
| | - Santosh K. Maurya
- Cardiovascular Pathobiology Program, Sanford Burnham Medical Research Institute at Lake Nona, Orland, Florida, United States of America
| | - Anuradha Kalyanasundaram
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
| | - Muthu Periasamy
- Cardiovascular Pathobiology Program, Sanford Burnham Medical Research Institute at Lake Nona, Orland, Florida, United States of America
| | - Richard A. LaFountain
- Department of Human Sciences, The Ohio State University, College of Education & Human Ecology, Columbus, Ohio, United States of America
| | - Jacob M. Wilson
- Department of Human Performance, The University of Tampa, Tampa, Florida, United States of America
| | - Orlando P. Simonetti
- Department of Radiology, The Ohio State University, College of Medicine, Columbus, Ohio, United States of America
- Department of Cardiovascular Medicine, The Ohio State University, College of Medicine, Columbus, Ohio, United States of America
| | - Ouliana Ziouzenkova
- Department of Human Sciences, The Ohio State University, College of Education & Human Ecology, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
120
|
Wilson MG, Ellison GM, Cable NT. Basic science behind the cardiovascular benefits of exercise. Br J Sports Med 2016; 50:93-9. [DOI: 10.1136/bjsports-2014-306596rep] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
121
|
Wilson MG, Ellison GM, Cable NT. Republished: Basic science behind the cardiovascular benefits of exercise. Postgrad Med J 2015; 91:704-11. [DOI: 10.1136/postgradmedj-2014-306596rep] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
122
|
Toedebusch RG, Ruegsegger GN, Braselton JF, Heese AJ, Hofheins JC, Childs TE, Thyfault JP, Booth FW. AMPK agonist AICAR delays the initial decline in lifetime-apex V̇o2 peak, while voluntary wheel running fails to delay its initial decline in female rats. Physiol Genomics 2015; 48:101-15. [PMID: 26578698 DOI: 10.1152/physiolgenomics.00078.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 11/11/2015] [Indexed: 11/22/2022] Open
Abstract
There has never been an outcome measure for human health more important than peak oxygen consumption (V̇o2 peak), yet little is known regarding the molecular triggers for its lifetime decline with aging. We examined the ability of physical activity or 5 wk of 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAR) administration to delay the initial aging-induced decline in lifetime-apex V̇o2 peak and potential underlying molecular mechanisms. Experiment 1 consisted of female rats with (RUN) and without (NO RUN) running wheels, while experiment 2 consisted of female nonrunning rats getting the AMPK agonist AICAR (0.5 mg/g/day) subcutaneously for 5 wk beginning at 17 wk of age. All rats underwent frequent, weekly or biweekly V̇o2 peak tests beginning at 10 wk of age. In experiment 1, lifetime-apex V̇o2 peak occurred at 19 wk of age in both RUN and NO RUN and decreased thereafter. V̇o2 peak measured across experiment 1 was ∼25% higher in RUN than in NO RUN. In experiment 2, AICAR delayed the chronological age observed in experiment 1 by 1 wk, from 19 wk to 20 wk of age. RUN and NO RUN showed different skeletal muscle transcriptomic profiles both pre- and postapex. Additionally, growth and development pathways are differentially regulated between RUN and NO RUN. Angiomotin mRNA was downregulated postapex in RUN and NO RUN. Furthermore, strong significant correlations to V̇o2 peak and trends for decreased protein concentration supports angiomotin's potential importance in our model. Contrary to our primary hypothesis, wheel running was not sufficient to delay the chronological age of lifetime-apex V̇o2 peak decline, whereas AICAR delayed it 1 wk.
Collapse
Affiliation(s)
- Ryan G Toedebusch
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | | | - Joshua F Braselton
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Alexander J Heese
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - John C Hofheins
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Tom E Childs
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - John P Thyfault
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas; and
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri; Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
123
|
Davis RT, Simon JN, Utter M, Mungai P, Alvarez MG, Chowdhury SAK, Heydemann A, Ke Y, Wolska BM, Solaro RJ. Knockout of p21-activated kinase-1 attenuates exercise-induced cardiac remodelling through altered calcineurin signalling. Cardiovasc Res 2015; 108:335-47. [PMID: 26464331 DOI: 10.1093/cvr/cvv234] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 10/03/2015] [Indexed: 01/14/2023] Open
Abstract
AIMS Despite its known cardiovascular benefits, the intracellular signalling mechanisms underlying physiological cardiac growth remain poorly understood. Therefore, the purpose of this study was to investigate a novel role of p21-activated kinase-1 (Pak1) in the regulation of exercise-induced cardiac hypertrophy. METHODS AND RESULTS Wild-type (WT) and Pak1 KO mice were subjected to 6 weeks of treadmill endurance exercise training (ex-training). Cardiac function was assessed via echocardiography, in situ haemodynamics, and the pCa-force relations in skinned fibre preparations at baseline and at the end of the training regimen. Post-translational modifications to the sarcomeric proteins and expression levels of calcium-regulating proteins were also assessed following ex-training. Heart weight/tibia length and echocardiography data revealed that there was marked hypertrophy following ex-training in the WT mice, which was not evident in the KO mice. Additionally, following ex-training, WT mice demonstrated an increase in cardiac contractility, myofilament calcium sensitivity, and phosphorylation of cardiac myosin-binding protein C, cardiac TnT, and tropomyosin compared with KO mice. With ex-training in WT mice, there were also increased protein levels of calcineurin and increased phosphorylation of phospholamban. CONCLUSIONS Our data suggest that Pak1 is essential for adaptive physiological cardiac remodelling and support previous evidence that demonstrates Pak1 signalling is important for cardiac growth and survival.
Collapse
Affiliation(s)
- Robert T Davis
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Jillian N Simon
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Megan Utter
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Paul Mungai
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Manuel G Alvarez
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Shamim A K Chowdhury
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Ahlke Heydemann
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Yunbo Ke
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| | - Beata M Wolska
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA Department of Medicine, Section of Cardiology, Center for Cardiovascular Research, University of Illinois, Chicago, IL 60612, USA
| | - R John Solaro
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave-Rm. E202, Chicago, IL 60612, USA
| |
Collapse
|
124
|
Kim TK, Park JY, Han PL. Physiological Parameters in the Blood of a Murine Stress-Induced Depression Model before and after Repeated Passive Exercise. Endocrinol Metab (Seoul) 2015; 30:371-80. [PMID: 25559715 PMCID: PMC4595363 DOI: 10.3803/enm.2015.30.3.371] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 10/13/2014] [Accepted: 10/17/2014] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Animal models are necessary to study the mechanism underlying the effects of exercise on depression but an effective procedure for exercise treatment and exercise effects on physiological parameters in a specific depression model need to be characterized. METHODS Physiological parameters including lactate, partial pressue of O₂ (pO₂) and CO₂ (pCO₂) saturated O₂ (sO₂), pH, HCO₃, total CO₂ (TCO₂), and base excess extracellular fluid (BEecf) levels in the blood were measured after treatment with passive exercise in normal mice and a stress-induced depression model. RESULTS Normal mice or mice that were subjected to daily 2-hour restraint for 14 days (2 hours × 14 days of restraint) were placed on a running wheel that was rotating at a speed of 9 m/min for 1 hour per day for 1 to 21 days. After repeated exercise in mice that were previously subjected to 2 hours × 14 days restraint, plasma lactate levels decreased, the levels of pO₂, sO₂, and pH tended to increase, and the levels of pCO₂ decreased in the absence of significant changes in HCO₃, TCO₂, and BEecf. However, none of these changes were additive to the stress effects or were much more severe than those induced after repeated passive exercise in normal mice. CONCLUSION These results suggest that passive exercise for 1 hour daily for 14 to 21 consecutive days on a running wheel rotating at a speed of 9 m/min may be used as an exercise protocol without inducing severe additive effects on physiological burdens.
Collapse
Affiliation(s)
- Tae Kyung Kim
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, Korea
| | - Jin Young Park
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, Korea
| | - Pyung Lim Han
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, Korea.
- Department of Chemistry and Nano Science, Ewha Womans University, Seoul, Korea
- Brain Disease Research Institute, Ewha Womans University, Seoul, Korea
| |
Collapse
|
125
|
|
126
|
Binning SA, Ros AFH, Nusbaumer D, Roche DG. Physiological plasticity to water flow habitat in the damselfish, Acanthochromis polyacanthus: linking phenotype to performance. PLoS One 2015; 10:e0121983. [PMID: 25807560 PMCID: PMC4373956 DOI: 10.1371/journal.pone.0121983] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/06/2015] [Indexed: 11/18/2022] Open
Abstract
The relationships among animal form, function and performance are complex, and vary across environments. Therefore, it can be difficult to identify morphological and/or physiological traits responsible for enhancing performance in a given habitat. In fishes, differences in swimming performance across water flow gradients are related to morphological variation among and within species. However, physiological traits related to performance have been less well studied. We experimentally reared juvenile damselfish, Acanthochromis polyacanthus, under different water flow regimes to test 1) whether aspects of swimming physiology and morphology show plastic responses to water flow, 2) whether trait divergence correlates with swimming performance and 3) whether flow environment relates to performance differences observed in wild fish. We found that maximum metabolic rate, aerobic scope and blood haematocrit were higher in wave-reared fish compared to fish reared in low water flow. However, pectoral fin shape, which tends to correlate with sustained swimming performance, did not differ between rearing treatments or collection sites. Maximum metabolic rate was the best overall predictor of individual swimming performance; fin shape and fish total length were 3.3 and 3.7 times less likely than maximum metabolic rate to explain differences in critical swimming speed. Performance differences induced in fish reared in different flow environments were less pronounced than in wild fish but similar in direction. Our results suggest that exposure to water motion induces plastic physiological changes which enhance swimming performance in A. polyacanthus. Thus, functional relationships between fish morphology and performance across flow habitats should also consider differences in physiology.
Collapse
Affiliation(s)
- Sandra A Binning
- Division of Evolution, Ecology and Genetics, Research School of Biology, The Australian National University, Canberra, Australia; ARC Centre of Excellence for Coral Reef Studies, The Australian National University, Canberra, Australia; Eco-Éthologie, Institut de Biologie, Université de Neuchâtel, Neuchâtel, Switzerland
| | - Albert F H Ros
- Eco-Éthologie, Institut de Biologie, Université de Neuchâtel, Neuchâtel, Switzerland
| | - David Nusbaumer
- Eco-Éthologie, Institut de Biologie, Université de Neuchâtel, Neuchâtel, Switzerland
| | - Dominique G Roche
- Division of Evolution, Ecology and Genetics, Research School of Biology, The Australian National University, Canberra, Australia; ARC Centre of Excellence for Coral Reef Studies, The Australian National University, Canberra, Australia; Eco-Éthologie, Institut de Biologie, Université de Neuchâtel, Neuchâtel, Switzerland
| |
Collapse
|
127
|
Waring CD, Henning BJ, Smith AJ, Nadal-Ginard B, Torella D, Ellison GM. Cardiac adaptations from 4 weeks of intensity-controlled vigorous exercise are lost after a similar period of detraining. Physiol Rep 2015; 3:3/2/e12302. [PMID: 25713328 PMCID: PMC4393210 DOI: 10.14814/phy2.12302] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Intensity-controlled (relative to VO2max) treadmill exercise training in adult rats results in the activation and ensuing differentiation of endogenous c-kitpos cardiac stem/progenitor cells (eCSCs) into newly formed cardiomyocytes and capillaries. Whether these training-induced adaptations persist following detraining is undetermined. Twelve male Wistar rats (∼230 g) were exercised at 80–85% of their VO2max for 30 min day−1, 4 days week−1 for 4 weeks (TR;n = 6), followed by 4 weeks of detraining (DTR; n = 6). Twelve untrained rats acted as controls (CTRL). Exercise training significantly enhanced VO2max (11.34 mL kg−1 min−1) and wet heart weight (29%) above CTRL (P < 0.05). Echocardiography revealed that exercise training increased LV mass (∼32%), posterior and septal wall thickness (∼15%), ejection fraction and fractional shortening (∼10%) compared to CTRL (P < 0.05). Cardiomyocyte diameter (17.9 ± 0.1 μm vs. 14.9 ± 0.6 μm), newly formed (BrdUpos/Ki67pos) cardiomyocytes (7.2 ± 1.3%/1.9 ± 0.7% vs. 0.2 ± 0.1%/0.1 ± 0.1%), total cardiomyocyte number (45.6 ± 0.6 × 106 vs. 42.5 ± 0.4 × 106), c-kitpos eCSC number (884 ± 112 per 106 cardiomyocytes vs. 482 ± 132 per 106 cardiomyocytes), and capillary density (4123 ± 227 per mm2 vs. 2117 ± 118 per mm2) were significantly greater in the LV of trained animals (P < 0.05) than CTRL. Detraining removed the stimulus for c-kitpos eCSC activation (640 ± 98 per 106 cardiomyocytes) and resultant cardiomyocyte hyperplasia (0.4 ± 0.3% BrdUpos/0.2 ± 0.2% Ki67pos cardiomyocytes). Capillary density (3673 ± 374 per mm2) and total myocyte number (44.7 ± 0.5 × 106) remained elevated following detraining, but cardiomyocyte hypertrophy (15.0 ± 0.4 μm) was lost, resulting in a reduction of anatomical (wall thickness ∼4%; LV mass ∼10% and cardiac mass ∼8%, above CTRL) and functional (EF & FS ∼2% above CTRL) parameters gained through exercise training. These findings demonstrate that cardiac adaptations, produced by 4 weeks of intensity-controlled exercise training are lost after a similar period of detraining.
Collapse
Affiliation(s)
- Cheryl D Waring
- Stem Cell and Regenerative Biology Unit (BioStem), Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, L3 3AF, UK
| | - Beverley J Henning
- Stem Cell and Regenerative Biology Unit (BioStem), Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, L3 3AF, UK Centre of Human & Aerospace Physiological Sciences and Centre for Stem Cells & Regenerative Medicine, Faculty of Medicine & Life Sciences, King's College London, London, SE1 1UL, UK
| | - Andrew J Smith
- Centre of Human & Aerospace Physiological Sciences and Centre for Stem Cells & Regenerative Medicine, Faculty of Medicine & Life Sciences, King's College London, London, SE1 1UL, UK
| | - Bernardo Nadal-Ginard
- Centre of Human & Aerospace Physiological Sciences and Centre for Stem Cells & Regenerative Medicine, Faculty of Medicine & Life Sciences, King's College London, London, SE1 1UL, UK
| | - Daniele Torella
- Laboratory of Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, 88100, Italy
| | - Georgina M Ellison
- Centre of Human & Aerospace Physiological Sciences and Centre for Stem Cells & Regenerative Medicine, Faculty of Medicine & Life Sciences, King's College London, London, SE1 1UL, UK Laboratory of Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, 88100, Italy
| |
Collapse
|
128
|
Svensson M, Lexell J, Deierborg T. Effects of Physical Exercise on Neuroinflammation, Neuroplasticity, Neurodegeneration, and Behavior: What We Can Learn From Animal Models in Clinical Settings. Neurorehabil Neural Repair 2014; 29:577-89. [PMID: 25527485 DOI: 10.1177/1545968314562108] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Physical exercise is a cornerstone in the management of many neurodegenerative disorders, such as Parkinson's disease, dementia, and stroke. However, much of its beneficial effects on improving motor functions and cognition as well as decreasing neurodegeneration and neuroinflammation are not yet well understood. The obvious limitations of studying the protective mechanisms behind exercise, for example, brain plasticity and neurodegeneration, could be overcome by generating novel animal models of neurodegenerative disorders. In this narrative review, we discuss the beneficial effects of exercise performed in animal models of neurodegenerative disorders and how the results from animal studies can be used in clinical settings. From preclinical studies, the positive effects of exercise have been related to increased levels of neurotrophic factors, elevated expression of anti-inflammatory cytokines, and reduced levels of pro-inflammatory cytokines and activated microglia. It is clear that parameters influencing the effect of exercise, such as intensity, still remain to be investigated in animal studies in order to find the optimal program that can be translated into exercise interventions for patients with neurodegenerative diseases.
Collapse
Affiliation(s)
- Martina Svensson
- Department of Experimental Medical Sciences, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Jan Lexell
- Department of Health Sciences, Rehabilitation Medicine Research Group, Lund University, Lund, Sweden Department of Neurology and Rehabilitation Medicine, Skane University Hospital, Lund, Sweden
| | - Tomas Deierborg
- Department of Experimental Medical Sciences, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| |
Collapse
|
129
|
Effects of exercise training on pulmonary vessel muscularization and right ventricular function in an animal model of COPD. Respir Res 2014; 15:117. [PMID: 25261051 PMCID: PMC4181603 DOI: 10.1186/s12931-014-0117-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 09/17/2014] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Right ventricular dysfunction in COPD is common, even in the absence of pulmonary hypertension. The aim of the present study was to examine the effects of high intensity interval training (HIIT) on right ventricular (RV) function, as well as pulmonary blood vessel remodeling in a mouse model of COPD. METHODS 42 female A/JOlaHsd mice were randomized to exposure to either cigarette smoke or air for 6 hours/day, 5 days/week for 14 weeks. Mice from both groups were further randomized to sedentariness or HIIT for 4 weeks. Cardiac function was evaluated by echocardiography and muscularization of pulmonary vessel walls by immunohistochemistry. RESULTS Smoke exposure induced RV systolic dysfunction demonstrated by reduced tricuspid annular plane systolic excursion. HIIT in smoke-exposed mice reversed RV dysfunction. There were no significant effects on the left ventricle of neither smoke exposure nor HIIT. Muscularization of the pulmonary vessels was reduced after exercise intervention, but no significant effects on muscularization were observed from smoke exposure. CONCLUSIONS RV function was reduced in mice exposed to cigarette smoke. No Increase in pulmonary vessel muscularization was observed in these mice, implying that other mechanisms caused the RV dysfunction. HIIT attenuated the RV dysfunction in the smoke exposed mice. Reduced muscularization of the pulmonary vessels due to HIIT suggests that exercise training not only affects the heart muscle, but also has important effects on the pulmonary vasculature.
Collapse
|
130
|
Mouisel E, Relizani K, Mille-Hamard L, Denis R, Hourdé C, Agbulut O, Patel K, Arandel L, Morales-Gonzalez S, Vignaud A, Garcia L, Ferry A, Luquet S, Billat V, Ventura-Clapier R, Schuelke M, Amthor H. Myostatin is a key mediator between energy metabolism and endurance capacity of skeletal muscle. Am J Physiol Regul Integr Comp Physiol 2014; 307:R444-54. [DOI: 10.1152/ajpregu.00377.2013] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Myostatin (Mstn) participates in the regulation of skeletal muscle size and has emerged as a regulator of muscle metabolism. Here, we hypothesized that lack of myostatin profoundly depresses oxidative phosphorylation-dependent muscle function. Toward this end, we explored Mstn −/− mice as a model for the constitutive absence of myostatin and AAV-mediated overexpression of myostatin propeptide as a model of myostatin blockade in adult wild-type mice. We show that muscles from Mstn −/− mice, although larger and stronger, fatigue extremely rapidly. Myostatin deficiency shifts muscle from aerobic toward anaerobic energy metabolism, as evidenced by decreased mitochondrial respiration, reduced expression of PPAR transcriptional regulators, increased enolase activity, and exercise-induced lactic acidosis. As a consequence, constitutively reduced myostatin signaling diminishes exercise capacity, while the hypermuscular state of Mstn−/− mice increases oxygen consumption and the energy cost of running. We wondered whether these results are the mere consequence of the congenital fiber-type switch toward a glycolytic phenotype of constitutive Mstn −/− mice. Hence, we overexpressed myostatin propeptide in adult mice, which did not affect fiber-type distribution, while nonetheless causing increased muscle fatigability, diminished exercise capacity, and decreased Pparb/d and Pgc1a expression. In conclusion, our results suggest that myostatin endows skeletal muscle with high oxidative capacity and low fatigability, thus regulating the delicate balance between muscle mass, muscle force, energy metabolism, and endurance capacity.
Collapse
Affiliation(s)
- Etienne Mouisel
- Institut National de la Santé et de la Recherche Médicale (INSERM)/Paul Sabatier University, UMR 1048, Institute of Metabolic and Cardiovascular Diseases, Obesity Research Laboratory, Toulouse, France
- Sorbonne Universités, Universités Européennes, l'Université Pierre et Marie Curie (UPMC), Paris 06, Myology Center of Research and Institut National de la Santé et de la Recherche Médicale, UMR S974 and Centre National de la Recherche Scientifique, FRE 3617 and Institut de Myologie, Paris, France
| | - Karima Relizani
- Department of Neuropediatrics and NeuroCure Clinical Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany
- Laboratoire “End:icap”, UFR des Sciences de la Santé, Université de Versailles Saint-Quentin-en-Yvelines, France
| | | | - Raphaël Denis
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS EAC 4413, Paris, France
| | - Christophe Hourdé
- Sorbonne Universités, Universités Européennes, l'Université Pierre et Marie Curie (UPMC), Paris 06, Myology Center of Research and Institut National de la Santé et de la Recherche Médicale, UMR S974 and Centre National de la Recherche Scientifique, FRE 3617 and Institut de Myologie, Paris, France
- Laboratory of Exercise Physiology, University of Savoie, Chambery, France
| | - Onnik Agbulut
- UPMC, Paris 06, Sorbonne Universités, UMR Centre National de la Recherche Scientifique (CNRS) Biological Adaptation and Ageing, Paris, France
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Ludovic Arandel
- Sorbonne Universités, Universités Européennes, l'Université Pierre et Marie Curie (UPMC), Paris 06, Myology Center of Research and Institut National de la Santé et de la Recherche Médicale, UMR S974 and Centre National de la Recherche Scientifique, FRE 3617 and Institut de Myologie, Paris, France
| | - Susanne Morales-Gonzalez
- Department of Neuropediatrics and NeuroCure Clinical Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | - Luis Garcia
- Laboratoire “End:icap”, UFR des Sciences de la Santé, Université de Versailles Saint-Quentin-en-Yvelines, France
- Laboratoire International Associé - Biothérapies Appliquées aux Handicaps Neuromusculaires, Centre Scientifique de Monaco, Monaco
| | - Arnaud Ferry
- Sorbonne Universités, Universités Européennes, l'Université Pierre et Marie Curie (UPMC), Paris 06, Myology Center of Research and Institut National de la Santé et de la Recherche Médicale, UMR S974 and Centre National de la Recherche Scientifique, FRE 3617 and Institut de Myologie, Paris, France
- Université Paris Descartes, Paris, France
| | - Serge Luquet
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS EAC 4413, Paris, France
- CNRS, EAC 4413, Paris, France; and
| | | | | | - Markus Schuelke
- Department of Neuropediatrics and NeuroCure Clinical Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Helge Amthor
- Sorbonne Universités, Universités Européennes, l'Université Pierre et Marie Curie (UPMC), Paris 06, Myology Center of Research and Institut National de la Santé et de la Recherche Médicale, UMR S974 and Centre National de la Recherche Scientifique, FRE 3617 and Institut de Myologie, Paris, France
- Laboratoire “End:icap”, UFR des Sciences de la Santé, Université de Versailles Saint-Quentin-en-Yvelines, France
- Laboratoire International Associé - Biothérapies Appliquées aux Handicaps Neuromusculaires, Centre Scientifique de Monaco, Monaco
| |
Collapse
|
131
|
De Souza MR, Pimenta L, Pithon-Curi TC, Bucci M, Fontinele RG, De Souza RR. Effects of aerobic training, resistance training, or combined resistance-aerobic training on the left ventricular myocardium in a rat model. Microsc Res Tech 2014; 77:727-34. [PMID: 24954110 DOI: 10.1002/jemt.22394] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 05/29/2014] [Accepted: 06/08/2014] [Indexed: 11/08/2022]
Abstract
This study follows the left ventricular (LV) hypertrophy in rats undergoing aerobic training alone (A), resistance training alone (R), or combined resistance and aerobic training (RA) (usually referred as concurrent training) program. A sedentary control group (C) was included. LV remodeling was evaluated using electron and light microscopy. The LV weight to body weight (LVW: BW) increased 11.4% in A group, 35% in the R group, and 18% in the RA group compared to the C group. The LV thickness increased 6% in the A group, 17% in the R group, and 10% in the RA group. The LV internal diameter increased 19% in the A group, 3% in the R group, and 8% in the RA group compared with the C group. The cross-sectional area of cardiomyocyte increased by 1% with the A group, 27% with R group, and 12% with RA training. The capillary density increased by 5.4% with A training, 11.0% with R training, and 7.7% with RA training compared with the C group. The volume fraction of interstitial collagen increased by 0.4% with training A, increased by 2.8% with R training, and 0.9% with RA training. In conclusion, except for the LV internal diameter, which increased more in the A group, the cardiac parameters increased more in the R group than in the other groups and in RA group than in A group. Collagen density increased from 5.4 ± 0.8% in the C group to 5.8 ± 0.6% in the A group (n. s.) (P > 0.05), to 8.2 ± 0.7% in the R group (P < 0.05), and to 6.3 ± 0.4% in the RA group (P < 0.05). These results demonstrate a significant increase for collagen content in the LV with R and RA exercise, but the increase was higher with R training alone than with RA training.
Collapse
|
132
|
Weissmann N, Peters DM, Klöpping C, Krüger K, Pilat C, Katta S, Seimetz M, Ghofrani HA, Schermuly RT, Witzenrath M, Seeger W, Grimminger F, Mooren FC. Structural and functional prevention of hypoxia-induced pulmonary hypertension by individualized exercise training in mice. Am J Physiol Lung Cell Mol Physiol 2014; 306:L986-95. [PMID: 24705723 DOI: 10.1152/ajplung.00275.2013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Pulmonary hypertension (PH) is a disease with a poor prognosis characterized by a vascular remodeling process and an increase in pulmonary vascular resistance. While a variety of reports demonstrated that exercise training exerts beneficial effects on exercise performance and quality of life in PH patients, it is not known how physical exercise affects vascular remodeling processes occurring in hypoxia-induced PH. Therefore, we investigated the effect of individualized exercise training on the development of hypoxia-induced PH in mice. Training effects were compared with pharmacological treatment with the phosphodiesterase 5 inhibitor Sildenafil or a combination of training plus Sildenafil. Trained mice who received Sildenafil showed a significantly improved walking distance (from 88.9 ± 8.1 to 146.4 ± 13.1 m) and maximum oxygen consumption (from 93.3 ± 2.9 to 105.5 ± 2.2% in combination with Sildenafil, to 102.2 ± 3.0% with placebo) compared with sedentary controls. Right ventricular systolic pressure, measured by telemetry, was at the level of healthy normoxic animals, whereas right heart hypertrophy did not benefit from training. Most interestingly, the increase in small pulmonary vessel muscularization was prevented by training. Respective counterregulatory processes were detected for the nitric oxide-soluble guanylate cyclase-phosphodiesterase system. We conclude that individualized daily exercise can prevent vascular remodeling in hypoxia-induced PH.
Collapse
Affiliation(s)
- Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany;
| | - Dorothea M Peters
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany
| | - Christina Klöpping
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany
| | - Karsten Krüger
- Department of Sports Medicine, Justus Liebig-University Giessen, Giessen, Germany
| | - Christian Pilat
- Department of Sports Medicine, Justus Liebig-University Giessen, Giessen, Germany
| | - Susmitha Katta
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany
| | - Michael Seimetz
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany
| | - Hossein A Ghofrani
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany
| | - Ralph T Schermuly
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany
| | - Martin Witzenrath
- Division of Infectiology and Pneumology, Charité-Universitätsmedizin Berlin Medical Clinic, Berlin, Germany; and
| | - Werner Seeger
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany; Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Friedrich Grimminger
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany
| | - Frank C Mooren
- Department of Sports Medicine, Justus Liebig-University Giessen, Giessen, Germany
| |
Collapse
|
133
|
Acute exercises induce disorders of the gastrointestinal integrity in a murine model. Eur J Appl Physiol 2013; 114:609-17. [PMID: 24352573 DOI: 10.1007/s00421-013-2791-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 12/05/2013] [Indexed: 01/16/2023]
Abstract
PURPOSE Many endurance athletes complain about gastrointestinal (GI) symptoms. It is assumed that exercise-induced shift of perfusion with consecutive hypoperfusion of the enteral vascular system leads to an increased GI permeability and tissue damage. Therefore, the aim of the study was to investigate permeability, apoptosis, electrogenic ion transport (Isc), and tissue conductance (Gt) of the small intestine in a murine exercise model. METHODS After spirometry, male Swiss CD-1 mice were subjected to an intensive treadmill exercise (80% VO2max). Sedentary mice served as controls. The small intestine was removed at several time intervals post-exercise. Apoptotic cells were determined by the TUNEL method, while fluorescein isothiocyanate dextran permeation indicated intestinal permeability. The Gt and Isc measurements were carried out in a modified Ussing chamber. RESULTS Apoptosis of epithelial cells increased continuously until 24 h post exercise (0.8 ± 0.42 versus 39.2 ± 26.0%; p < 0.05). Compared with the control group the permeability increased 2 h after exercise (0.47 ± 0.07 versus 0.67 ± 0.14 FU/min; p < 0.05). Isc measurements of the ileum were augmented after 24 h (3.33 ± 0.56 versus 5.77 ± 1.16 μEq/h/cm(2); p < 0.05). At this time the Gt increased as well (28.8 ± 3.37 versus 32.5 ± 2.59 mS/cm(2); p < 0.05). CONCLUSION In the murine exercise model there is evidence that after intense endurance exercise repair processes occur in small intestinal epithelial cells, which affect permeability, Gt, and Isc. The formation of lamellipodia to close the "leaky" tight junctions caused by apoptosis might be an underlying mechanism.
Collapse
|
134
|
Physical Exercise Induces Specific Adaptations Resulting in Reduced Organ Injury and Mortality During Severe Polymicrobial Sepsis. Crit Care Med 2013; 41:e246-55. [DOI: 10.1097/ccm.0b013e31828a2ae3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
135
|
Serradj N, Picquet F, Jamon M. Early postnatal motor experience shapes the motor properties of C57BL/6J adult mice. Eur J Neurosci 2013; 38:3281-91. [PMID: 23869740 DOI: 10.1111/ejn.12311] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 06/13/2013] [Indexed: 01/04/2023]
Abstract
This study aimed to evaluate the long-term consequences of early motor training on the muscle phenotype and motor output of middle-aged C57BL/6J mice. Neonatal mice were subjected to a variety of motor training procedures, for 3 weeks during the period of acquisition of locomotion. These procedures are widely used for motor training in adults; they include enriched environment, forced treadmill, chronic centrifugation, and hindlimb suspension. At 9 months, the mice reared in the enriched environment showed a slower type of fibre in slow muscles and a faster type in fast muscles, improved performance in motor tests, and a modified gait and body posture while walking. The proportion of fibres in the postural muscles of centrifuged mice did not change, but these mice showed improved resistance to fatigue. The suspended mice showed increased persistence of immature hybrid fibres in the tibialis, with a slower shift in the load-bearing soleus, without any behavioural changes. The forced treadmill was very stressful for the mice, but had limited effects on motor output, although a slower profile was observed in the tibialis. These results support the hypothesis that motor experience during a critical period of motor development shapes muscle phenotype and motor output. The different impacts of the various training procedures suggest that motor performance in adults can be optimized by appropriate training during a defined period of motor development.
Collapse
Affiliation(s)
- Nadjet Serradj
- Department of Physiology, Pharmacology & Neuroscience, Sophie Davis School of Biomedical Education, City College of New York/CCNY, New York, NY, USA
| | | | | |
Collapse
|
136
|
Jogiya R, Makowski M, Phinikaridou A, Patel AS, Jansen C, Zarinabad N, Chiribiri A, Botnar R, Nagel E, Kozerke S, Plein S. Hyperemic stress myocardial perfusion cardiovascular magnetic resonance in mice at 3 Tesla: initial experience and validation against microspheres. J Cardiovasc Magn Reson 2013; 15:62. [PMID: 23870734 PMCID: PMC3750232 DOI: 10.1186/1532-429x-15-62] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 07/07/2013] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Dynamic first pass contrast-enhanced myocardial perfusion is the standard CMR method for the estimation of myocardial blood flow (MBF) and MBF reserve in man, but it is challenging in rodents because of the high temporal and spatial resolution requirements. Hyperemic first pass myocardial perfusion CMR during vasodilator stress in mice has not been reported. METHODS Five C57BL/6 J mice were scanned on a clinical 3.0 Tesla Achieva system (Philips Healthcare, Netherlands). Vasodilator stress was induced via a tail vein catheter with an injection of dipyridamole. Dynamic contrast-enhanced perfusion imaging (Gadobutrol 0.1 mmol/kg) was based on a saturation recovery spoiled gradient echo method with 10-fold k-space and time domain undersampling (k-t PCA). One week later the mice underwent repeat anaesthesia and LV injections of fluorescent microspheres at rest and at stress. Microspheres were analysed using confocal microscopy and fluorescence-activated cell sorting. RESULTS Mean MBF at rest measured by Fermi-function constrained deconvolution was 4.1 ± 0.5 ml/g/min and increased to 9.6 ± 2.5 ml/g/min during dipyridamole stress (P = 0.005). The myocardial perfusion reserve was 2.4 ± 0.54. The mean count ratio of stress to rest microspheres was 2.4 ± 0.51 using confocal microscopy and 2.6 ± 0.46 using fluorescence. There was good agreement between cardiovascular magnetic resonance CMR and microspheres with no significant difference (P = 0.84). CONCLUSION First-pass myocardial stress perfusion CMR in a mouse model is feasible at 3 Tesla. Rest and stress MBF values were consistent with existing literature and perfusion reserve correlated closely to microsphere analysis. Data were acquired on a 3 Tesla scanner using an approach similar to clinical acquisition protocols, potentially facilitating translation of imaging findings between rodent and human studies.
Collapse
Affiliation(s)
- Roy Jogiya
- King’s College London BHF Centre of Excellence, NIHR Biomedical Research Centre and Welcome Trust and EPSRC Medical Engineering Centre at Guy’s and St. Thomas’ NHS Foundation Trust, Division of Imaging Sciences, The Rayne Institute, London, UK
| | - Markus Makowski
- King’s College London BHF Centre of Excellence, NIHR Biomedical Research Centre and Welcome Trust and EPSRC Medical Engineering Centre at Guy’s and St. Thomas’ NHS Foundation Trust, Division of Imaging Sciences, The Rayne Institute, London, UK
| | - Alkystsis Phinikaridou
- King’s College London BHF Centre of Excellence, NIHR Biomedical Research Centre and Welcome Trust and EPSRC Medical Engineering Centre at Guy’s and St. Thomas’ NHS Foundation Trust, Division of Imaging Sciences, The Rayne Institute, London, UK
| | - Ashish S Patel
- Academic Department of Surgery, Cardiovascular Division, BHF Centre of Excellence, Kings College, London, UK
| | - Christian Jansen
- King’s College London BHF Centre of Excellence, NIHR Biomedical Research Centre and Welcome Trust and EPSRC Medical Engineering Centre at Guy’s and St. Thomas’ NHS Foundation Trust, Division of Imaging Sciences, The Rayne Institute, London, UK
| | - Niloufar Zarinabad
- King’s College London BHF Centre of Excellence, NIHR Biomedical Research Centre and Welcome Trust and EPSRC Medical Engineering Centre at Guy’s and St. Thomas’ NHS Foundation Trust, Division of Imaging Sciences, The Rayne Institute, London, UK
| | - Amedeo Chiribiri
- King’s College London BHF Centre of Excellence, NIHR Biomedical Research Centre and Welcome Trust and EPSRC Medical Engineering Centre at Guy’s and St. Thomas’ NHS Foundation Trust, Division of Imaging Sciences, The Rayne Institute, London, UK
| | - Rene Botnar
- King’s College London BHF Centre of Excellence, NIHR Biomedical Research Centre and Welcome Trust and EPSRC Medical Engineering Centre at Guy’s and St. Thomas’ NHS Foundation Trust, Division of Imaging Sciences, The Rayne Institute, London, UK
| | - Eike Nagel
- King’s College London BHF Centre of Excellence, NIHR Biomedical Research Centre and Welcome Trust and EPSRC Medical Engineering Centre at Guy’s and St. Thomas’ NHS Foundation Trust, Division of Imaging Sciences, The Rayne Institute, London, UK
| | - Sebastian Kozerke
- King’s College London BHF Centre of Excellence, NIHR Biomedical Research Centre and Welcome Trust and EPSRC Medical Engineering Centre at Guy’s and St. Thomas’ NHS Foundation Trust, Division of Imaging Sciences, The Rayne Institute, London, UK
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| | - Sven Plein
- King’s College London BHF Centre of Excellence, NIHR Biomedical Research Centre and Welcome Trust and EPSRC Medical Engineering Centre at Guy’s and St. Thomas’ NHS Foundation Trust, Division of Imaging Sciences, The Rayne Institute, London, UK
- Multidisciplinary Cardiovascular Research Centre & Leeds Institute of Genetics, Health and Therapeutics, University of Leeds, Leeds, LS2 9JT, UK
| |
Collapse
|
137
|
Carneiro-Júnior MA, Prímola-Gomes TN, Quintão-Júnior JF, Drummond LR, Lavorato VN, Drummond FR, Felix LB, Oliveira EM, Cruz JS, Natali AJ, Mill JG. Regional effects of low-intensity endurance training on structural and mechanical properties of rat ventricular myocytes. J Appl Physiol (1985) 2013; 115:107-15. [DOI: 10.1152/japplphysiol.00041.2013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
We tested the effects of low-intensity endurance training (LIET) on the structural and mechanical properties of right (RV) and left ventricular (LV) myocytes. Male Wistar rats (4 mo old) were randomly divided into control (C, n = 7) and trained (T, n = 7, treadmill running at 50–60% of maximal running speed for 8 wk) groups. Isolated ventricular myocyte dimensions, contractility, Ca2+ transients {intracellular Ca2+ concentration ([Ca2+]i)}, and ventricular [Ca2+]i regulatory proteins were measured. LIET augmented cell length (C, 152.5 ± 2.0 μm vs. T, 162.2 ± 2.1 μm; P < 0.05) and volume (C, 5,162 ± 131 μm3 vs. T, 5,506 ± 132 μm3; P < 0.05) in the LV but not in the RV. LIET increased cell shortening (C, 7.5 ± 0.3% vs. T, 8.6 ± 0.3%; P < 0.05), the [Ca2+]i transient amplitude (C, 2.49 ± 0.06 F/F0 vs. T, 2.82 ± 0.06 F/F0; P < 0.05), the expression of sarcoplasmic reticulum Ca2+-ATPase 2a (C, 1.07 ± 0.13 vs. T, 1.59 ± 0.12; P < 0.05), and the levels of phosphorylated phospholamban at serine 16 (C, 0.99 ± 0.11 vs. T, 1.34 ± 0.10; P < 0.05), and reduced the total phospholamban-to-sarcoplasmic reticulum Ca2+-ATPase 2a ratio (C, 1.19 ± 0.15 vs. T, 0.40 ± 0.16; P < 0.05) in the LV without changing such parameters in the RV. In conclusion, LIET affected the structure and improved the mechanical properties of LV but not of RV myocytes in rats, helping to characterize the functional and morphological changes that accompany the endurance training-induced cardiac remodeling.
Collapse
Affiliation(s)
| | - Thales Nicolau Prímola-Gomes
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - Judson Fonseca Quintão-Júnior
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - Lucas Rios Drummond
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - Victor Neiva Lavorato
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - Filipe Rios Drummond
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - Leonardo Bonato Felix
- Department of Electrical Engineering, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - Edilamar Menezes Oliveira
- School of Physical Education and Sport, Laboratory of Biochemistry and Molecular Biology of the Exercise, University of São Paulo, São Paulo, Brazil; and
| | - Jader Santos Cruz
- Department of Biochemistry and Immunology, Laboratory of Excitable Membranes and Cardiovascular Biology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Antonio José Natali
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - José Geraldo Mill
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| |
Collapse
|
138
|
Hafstad AD, Lund J, Hadler-Olsen E, Höper AC, Larsen TS, Aasum E. High- and moderate-intensity training normalizes ventricular function and mechanoenergetics in mice with diet-induced obesity. Diabetes 2013; 62:2287-94. [PMID: 23493573 PMCID: PMC3712042 DOI: 10.2337/db12-1580] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Although exercise reduces several cardiovascular risk factors associated with obesity/diabetes, the metabolic effects of exercise on the heart are not well-known. This study was designed to investigate whether high-intensity interval training (HIT) is superior to moderate-intensity training (MIT) in counteracting obesity-induced impairment of left ventricular (LV) mechanoenergetics and function. C57BL/6J mice with diet-induced obesity (DIO mice) displaying a cardiac phenotype with altered substrate utilization and impaired mechanoenergetics were subjected to a sedentary lifestyle or 8-10 weeks of isocaloric HIT or MIT. Although both modes of exercise equally improved aerobic capacity and reduced obesity, only HIT improved glucose tolerance. Hearts from sedentary DIO mice developed concentric LV remodeling with diastolic and systolic dysfunction, which was prevented by both HIT and MIT. Both modes of exercise also normalized LV mechanical efficiency and mechanoenergetics. These changes were associated with altered myocardial substrate utilization and improved mitochondrial capacity and efficiency, as well as reduced oxidative stress, fibrosis, and intracellular matrix metalloproteinase 2 content. As both modes of exercise equally ameliorated the development of diabetic cardiomyopathy by preventing LV remodeling and mechanoenergetic impairment, this study advocates the therapeutic potential of physical activity in obesity-related cardiac disorders.
Collapse
Affiliation(s)
- Anne D Hafstad
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway.
| | | | | | | | | | | |
Collapse
|
139
|
Arena R, Myers J, Forman DE, Lavie CJ, Guazzi M. Should high-intensity-aerobic interval training become the clinical standard in heart failure? Heart Fail Rev 2013; 18:95-105. [PMID: 22791516 DOI: 10.1007/s10741-012-9333-z] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Aerobic exercise training in the heart failure (HF) population is supported by an extensive body of literature. The clinically accepted model for exercise prescription is currently moderate-intensity-aerobic continuous training (MI-ACT). Documented benefits from the literature include improvements in various aspects of physiologic function, aerobic exercise capacity and quality of life while the impact on morbidity and mortality is promising but requires further investigation. Recently, however, a body of evidence has begun to emerge demonstrating high-intensity-aerobic interval training (HI-AIT) can be performed safely with impressive improvements in physiology, functional capacity and quality of life. These initial findings have led some to question the long-standing clinical approach to aerobic exercise training in patients with HF (i.e., MI-ACT), implying it should perhaps be replaced with a HI-AIT model. This is a potentially controversial paradigm shift given the potential increase in adverse event risk associated with exercising at higher intensities, particularly in the HF population where the likelihood of an untoward episode is already at a heightened state relative to the apparently healthy population. The present review therefore addresses key issues related to HI-AIT in the HF population and makes recommendations for future research and current clinical practice.
Collapse
Affiliation(s)
- Ross Arena
- Physical Therapy Program, Department of Orthopaedics and Rehabilitation, University of New Mexico School of Medicine, Albuquerque, NM 87131-0001, USA.
| | | | | | | | | |
Collapse
|
140
|
Johnsen AB, Høydal M, Røsbjørgen R, Stølen T, Wisløff U. Aerobic interval training partly reverse contractile dysfunction and impaired Ca2+ handling in atrial myocytes from rats with post infarction heart failure. PLoS One 2013; 8:e66288. [PMID: 23799089 PMCID: PMC3682943 DOI: 10.1371/journal.pone.0066288] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 05/05/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND There is limited knowledge about atrial myocyte Ca(2+) handling in the failing hearts. The aim of this study was to examine atrial myocyte contractile function and Ca(2+) handling in rats with post-infarction heart failure (HF) and to examine whether aerobic interval training could reverse a potential dysfunction. METHODS AND RESULTS Post-infarction HF was induced in Sprague Dawley rats by ligation of the left descending coronary artery. Atrial myocyte shortening was depressed (p<0.01) and time to relaxation was prolonged (p<0.01) in sedentary HF-rats compared to healthy controls. This was associated with decreased Ca(2+) amplitude, decreased SR Ca(2+) content, and slower Ca(2+) transient decay. Atrial myocytes from HF-rats had reduced sarcoplasmic reticulum Ca(2+) ATPase activity, increased Na(+)/Ca(2+)-exchanger activity and increased diastolic Ca(2+) leak through ryanodine receptors. High intensity aerobic interval training in HF-rats restored atrial myocyte contractile function and reversed changes in atrial Ca(2+) handling in HF. CONCLUSION Post infarction HF in rats causes profound impairment in atrial myocyte contractile function and Ca(2+) handling. The observed dysfunction in atrial myocytes was partly reversed after aerobic interval training.
Collapse
Affiliation(s)
- Anne Berit Johnsen
- K.G. Jebsen Center of Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Morten Høydal
- K.G. Jebsen Center of Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ragnhild Røsbjørgen
- K.G. Jebsen Center of Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Tomas Stølen
- K.G. Jebsen Center of Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ulrik Wisløff
- K.G. Jebsen Center of Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- * E-mail:
| |
Collapse
|
141
|
Wolff G, Esser KA. Scheduled exercise phase shifts the circadian clock in skeletal muscle. Med Sci Sports Exerc 2013; 44:1663-70. [PMID: 22460470 DOI: 10.1249/mss.0b013e318255cf4c] [Citation(s) in RCA: 191] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE It has been well established in mammals that circadian behavior as well as the molecular clockwork can be synchronized to the light-dark cycle via the suprachiasmatic nucleus of the hypothalamus (SCN). In addition to light, it has been demonstrated that nonphotic time cues, such as restricting the time of food availability, can alter circadian behavior and clock gene expression in selected peripheral tissues such as the liver. Studies have also suggested that scheduled physical activity (exercise) can alter circadian rhythms in behavior and clock gene expression; however, currently, the effects of exercise alone are largely unknown and have not been explored in skeletal muscle. METHODS Period2::Luciferase (Per2::Luc) mice were maintained under 12 h of light followed by 12 h of darkness then exposed to 2 h of voluntary or involuntary exercise during the light phase for 4 wk. Control mice were left in home cages or moved to the exercise environment (sham). A second group of mice had restricted access to food (4 h · d(-1) for 2 wk) to compare the effects of two nonphotic cues on PER2::LUC bioluminescence. Skeletal muscle, lung, and SCN tissue explants were cultured for 5-6 d to study molecular rhythms. RESULTS In the exercised mice, the phase of peak PER2::LUC bioluminescence was shifted in the skeletal muscle and lung explants but not in the SCN suggesting a specific synchronizing effect of exercise on the molecular clockwork in peripheral tissues. CONCLUSIONS These data provide evidence that the molecular circadian clock in peripheral tissues can respond to the time of exercise suggesting that physical activity contributes important timing information for synchronization of circadian clocks throughout the body.
Collapse
Affiliation(s)
- Gretchen Wolff
- Department of Physiology, Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| | | |
Collapse
|
142
|
Radovits T, Oláh A, Lux Á, Németh BT, Hidi L, Birtalan E, Kellermayer D, Mátyás C, Szabó G, Merkely B. Rat model of exercise-induced cardiac hypertrophy: hemodynamic characterization using left ventricular pressure-volume analysis. Am J Physiol Heart Circ Physiol 2013; 305:H124-34. [PMID: 23645462 DOI: 10.1152/ajpheart.00108.2013] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Long-term exercise training is associated with characteristic structural and functional changes of the myocardium, termed athlete's heart. Several research groups investigated exercise training-induced left ventricular (LV) hypertrophy in animal models; however, only sporadic data exist about detailed hemodynamics. We aimed to provide functional characterization of exercise-induced cardiac hypertrophy in a rat model using the in vivo method of LV pressure-volume (P-V) analysis. After inducing LV hypertrophy by swim training, we assessed LV morphometry by echocardiography and performed LV P-V analysis using a pressure-conductance microcatheter to investigate in vivo cardiac function. Echocardiography showed LV hypertrophy (LV mass index: 2.41 ± 0.09 vs. 2.03 ± 0.08 g/kg, P < 0.01), which was confirmed by heart weight data and histomorphometry. Invasive hemodynamic measurements showed unaltered heart rate, arterial pressure, and LV end-diastolic volume along with decreased LV end-systolic volume, thus increased stroke volume and ejection fraction (73.7 ± 0.8 vs. 64.1 ± 1.5%, P < 0.01) in trained versus untrained control rats. The P-V loop-derived sensitive, load-independent contractility indexes, such as slope of end-systolic P-V relationship or preload recruitable stroke work (77.0 ± 6.8 vs. 54.3 ± 4.8 mmHg, P = 0.01) were found to be significantly increased. The observed improvement of ventriculoarterial coupling (0.37 ± 0.02 vs. 0.65 ± 0.08, P < 0.01), along with increased LV stroke work and mechanical efficiency, reflects improved mechanoenergetics of exercise-induced cardiac hypertrophy. Despite the significant hypertrophy, we observed unaltered LV stiffness (slope of end-diastolic P-V relationship: 0.043 ± 0.007 vs. 0.040 ± 0.006 mmHg/μl) and improved LV active relaxation (τ: 10.1 ± 0.6 vs. 11.9 ± 0.2 ms, P < 0.01). According to our knowledge, this is the first study that provides characterization of functional changes and hemodynamic relations in exercise-induced cardiac hypertrophy.
Collapse
|
143
|
Pastore S, Hood DA. Endurance training ameliorates the metabolic and performance characteristics of circadian Clock mutant mice. J Appl Physiol (1985) 2013; 114:1076-84. [PMID: 23429867 DOI: 10.1152/japplphysiol.01505.2012] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Circadian locomotor output cycles kaput (CLOCK) is a nuclear transcription factor that is a component of the central autoregulatory feedback loop that governs the generation of biological rhythms. Homozygous Clock mutant mice contain a truncated CLOCK(Δ19) protein within somatic cells, subsequently causing an impaired ability to rhythmically transactivate circadian genes. The present study sought to investigate whether the Clock mutation affects mitochondrial physiology within skeletal muscle, as well as the responsiveness of these mutant animals to adapt to a chronic voluntary endurance training protocol. Within muscle, Clock mutant mice displayed 44% and 45% reductions in peroxisome proliferator-activated receptor-γ coactivator 1-α (PGC-1α) and mitochondrial transcription factor-A protein content, respectively, and an accompanying 16% decrease in mitochondrial content, as determined by cytochrome c oxidase enzyme activity. These decrements contributed to a 50% decrease in exercise tolerance in Clock mutant mice. Interestingly, the Clock mutation did not appear to alter subsarcolemmal or intermyofibrillar mitochondrial respiration within muscle or systemic glucose tolerance. Daily locomotor activity levels were similar between wild-type and Clock mutant mice throughout the training protocol. Endurance training ameliorated the decrease in PGC-1α protein expression and mitochondrial content in the Clock mutant mice, eliciting a 2.9-fold improvement in exercise tolerance. Thus our data suggest that a functional CLOCK protein is essential to ensure the maintenance of mitochondrial content within muscle although the absence of a functional CLOCK protein does not impair the ability of animals to adapt to chronic exercise.
Collapse
Affiliation(s)
- Stephen Pastore
- Muscle Health Research Center, York University, Toronto, Ontario, Canada
| | | |
Collapse
|
144
|
Scott JM, Lakoski S, Mackey JR, Douglas PS, Haykowsky MJ, Jones LW. The potential role of aerobic exercise to modulate cardiotoxicity of molecularly targeted cancer therapeutics. Oncologist 2013; 18:221-31. [PMID: 23335619 PMCID: PMC3579607 DOI: 10.1634/theoncologist.2012-0226] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 09/05/2012] [Indexed: 01/03/2023] Open
Abstract
Molecularly targeted therapeutics (MTT) are the future of cancer systemic therapy. They have already moved from palliative therapy for advanced solid malignancies into the setting of curative-intent treatment for early-stage disease. Cardiotoxicity is a frequent and potentially serious adverse complication of some targeted therapies, leading to a broad range of potentially life-threatening complications, therapy discontinuation, and poor quality of life. Low-cost pleiotropic interventions are therefore urgently required to effectively prevent and/or treat MTT-induced cardiotoxicity. Aerobic exercise therapy has the unique capacity to modulate, without toxicity, multiple gene expression pathways in several organ systems, including a plethora of cardiac-specific molecular and cell-signaling pathways implicated in MTT-induced cardiac toxicity. In this review, we examine the molecular signaling of antiangiogenic and HER2-directed therapies that may underpin cardiac toxicity and the hypothesized molecular mechanisms underlying the cardioprotective properties of aerobic exercise. It is hoped that this knowledge can be used to maximize the benefits of small molecule inhibitors, while minimizing cardiac damage in patients with solid malignancies.
Collapse
Affiliation(s)
- Jessica M Scott
- Exercise Physiology and Countermeasures, NASA Johnson Space Center, Universities Space Research Association, 2101 NASA Parkway, Houston, TX 77058, USA.
| | | | | | | | | | | |
Collapse
|
145
|
Casuso RA, Martínez-Amat A, Martínez-López EJ, Camiletti-Moirón D, Porres JM, Aranda P. Ergogenic effects of quercetin supplementation in trained rats. J Int Soc Sports Nutr 2013; 10:3. [PMID: 23316871 PMCID: PMC3549753 DOI: 10.1186/1550-2783-10-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 12/19/2012] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED BACKGROUND Quercetin is a natural polyphenolic compound currently under study for its ergogenic capacity to improve mitochondrial biogenesis. Sedentary mice have exhibited increased endurance performance, but results are contradictory in human models. METHODS We examined the effects of six weeks of endurance training and quercetin supplementation on markers of endurance performance and training in a rodent model. Rats were randomly assigned to one of the following groups: placebo+sedentary (PS), quercetin+sedentary (QS), placebo+endurance training (PT) and quercetin+endurance training (QT). Quercetin was administered at a dose of 25 mg/kg on alternate days. During six weeks of treatment volume parameters of training were recorded, and after six weeks all groups performed a maximal graded VO2 max test and a low-intensity endurance run-to-fatigue test. RESULTS No effects were found in VO2 peak (p>0.999), nor in distance run during low-intensity test, although it was 14% greater in QT when compared with PT (P = 0.097). Post-exercise blood lactate was increased in QT when compared with PT (p=0.023) and also in QS compared with PS (p=0.024). CONCLUSIONS This study showed no effects in VO2 peak, speed at VO2 peak or endurance time to exhaustion after six weeks of quercetin supplementation compared with placebo in trained rats. Quercetin was show to increase blood lactate production after high-intensity exercise.
Collapse
Affiliation(s)
- Rafael A Casuso
- Department of Health Sciences, University of Jaén, Jaén, E-23071, Spain.
| | | | | | | | | | | |
Collapse
|
146
|
Dlugosz EM, Chappell MA, Meek TH, Szafrańska P, Zub K, Konarzewski M, Jones JH, Bicudo E, Nespolo RF, Careau V, Garland T. Phylogenetic analysis of mammalian maximal oxygen consumption during exercise. J Exp Biol 2013; 216:4712-21. [DOI: 10.1242/jeb.088914] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Summary
We compiled published values of mammalian maximum oxygen consumption during exercise (VO2max) and supplemented these data with new measurements of VO2max for the largest rodent (capybara), 20 species of smaller-bodied rodents, two species of weasels, and one small marsupial. Many of the new data were obtained with running-wheel respirometers instead of the treadmill systems used in most previous measurements of mammalian VO2max. We used both conventional and phylogenetically informed allometric regression models to analyze VO2max of 77 ‘species’ (including subspecies or separate populations within species) in relation to body size, phylogeny, diet, and measurement method. Both body mass and allometrically mass-corrected VO2max showed highly significant phylogenetic signal (i.e., related species tended to resemble each other). The Akaike Information Criterion corrected for sample size was used to compare 27 candidate models predicting VO2max (all of which included body mass). In addition to mass, the two best-fitting models (cumulative Akaike weight = 0.93) included dummy variables coding for three species previously shown to have high VO2max (pronghorn, horse, and a bat), and incorporated a transformation of the phylogenetic branch lengths under an Ornstein-Uhlenbeck model of residual variation (thus indicating phylogenetic signal in the residuals). We found no statistical difference between wheel- and treadmill-elicited values, and diet had no predictive ability for VO2max. Averaged across all models, the allometric scaling exponent was 0.839, with 95% confidence limits of 0.795 and 0.883, which does not provide support for a scaling exponent of 0.67, 0.75 or unity.
Collapse
|
147
|
Colombo R, Siqueira R, Becker CU, Fernandes TG, Pires KM, Valença SS, Souza-Rabbo MP, Araujo AS, Belló-Klein A. Effects of exercise on monocrotaline-induced changes in right heart function and pulmonary artery remodeling in rats. Can J Physiol Pharmacol 2013; 91:38-44. [DOI: 10.1139/cjpp-2012-0261] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Pulmonary arterial hypertension (PAH) induced by monocrotaline (MCT) is an experimental protocol of right heart failure. We analyzed the role of exercise training on the right ventricle structure and function, pulmonary artery remodeling, and GSK-3β expression. Rats were divided among the following groups: sedentary control (SC), sedentary monocrotaline (SM), trained control (TC), and trained monocrotaline (TM). Rats underwent exercise training for a period of 5 weeks, with 3 weeks post-MCT injection. Rats in the SM and TM groups presented with an increase in right ventricle hypertrophy indexes and lung congestion. The right ventricular end diastolic pressure (RVEDP), right ventricular systolic pressure (RVSP), and its minimum and maximal pressure derivates were increased in the SM and TM groups. The right ventricle interstitial volume pulmonary artery thickness and p-GSK-3β/GSK-3β were increased in the MCT groups as compared with the control groups. The TM group had a reduction in interstitial volume, p-GSK-3β/GSK-3β ratio, pulmonary artery thickness, RVEDP, and an increase in intramyocardial vessels volume as compared with the SM group. The overall results have shown that the exercise protocol used promoted positive changes in right ventricle and pulmonary artery remodeling. These observations also suggest that structural remodeling may be influenced by signaling proteins, such as GSK-3β.
Collapse
Affiliation(s)
- Rafael Colombo
- Laboratório de Fisiologia Cardiovascular e Espécies Ativas de Oxigênio, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, CEP 90050-170, Porto Alegre, Brasil
| | - Rafaela Siqueira
- Laboratório de Fisiologia Cardiovascular e Espécies Ativas de Oxigênio, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, CEP 90050-170, Porto Alegre, Brasil
| | - Cristiano Urbano Becker
- Laboratório de Fisiologia Cardiovascular e Espécies Ativas de Oxigênio, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, CEP 90050-170, Porto Alegre, Brasil
| | - Tânia Gatelli Fernandes
- Laboratório de Fisiologia Cardiovascular e Espécies Ativas de Oxigênio, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, CEP 90050-170, Porto Alegre, Brasil
| | - Karla Maria Pires
- Laboratório de Inflamação, Estresse Oxidativo e Câncer, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Samuel Santos Valença
- Laboratório de Inflamação, Estresse Oxidativo e Câncer, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | | | - Alex Sander Araujo
- Laboratório de Fisiologia Cardiovascular e Espécies Ativas de Oxigênio, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, CEP 90050-170, Porto Alegre, Brasil
| | - Adriane Belló-Klein
- Laboratório de Fisiologia Cardiovascular e Espécies Ativas de Oxigênio, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, CEP 90050-170, Porto Alegre, Brasil
| |
Collapse
|
148
|
Hadden H. High frequency mechanical ventilation affects respiratory system mechanics differently in C57BL/6J and BALB/c adult mice. Respir Physiol Neurobiol 2013; 185:472-6. [DOI: 10.1016/j.resp.2012.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 09/16/2012] [Accepted: 09/21/2012] [Indexed: 11/30/2022]
|
149
|
Guiraud T, Nigam A, Gremeaux V, Meyer P, Juneau M, Bosquet L. High-intensity interval training in cardiac rehabilitation. Sports Med 2012; 42:587-605. [PMID: 22694349 DOI: 10.2165/11631910-000000000-00000] [Citation(s) in RCA: 183] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
High-intensity interval training (HIIT) is frequently used in sports training. The effects on cardiorespiratory and muscle systems have led scientists to consider its application in the field of cardiovascular diseases. The objective of this review is to report the effects and interest of HIIT in patients with coronary artery disease (CAD) and heart failure (HF), as well as in persons with high cardiovascular risk. A non-systematic review of the literature in the MEDLINE database using keywords 'exercise', 'high-intensity interval training', 'interval training', 'coronary artery disease', 'coronary heart disease', 'chronic heart failure' and 'metabolic syndrome' was performed. We selected articles concerning basic science research, physiological research, and randomized or non-randomized interventional clinical trials published in English. To summarize, HIIT appears safe and better tolerated by patients than moderate-intensity continuous exercise (MICE). HIIT gives rise to many short- and long-term central and peripheral adaptations in these populations. In stable and selected patients, it induces substantial clinical improvements, superior to those achieved by MICE, including beneficial effects on several important prognostic factors (peak oxygen uptake, ventricular function, endothelial function), as well as improving quality of life. HIIT appears to be a safe and effective alternative for the rehabilitation of patients with CAD and HF. It may also assist in improving adherence to exercise training. Larger randomized interventional studies are now necessary to improve the indications for this therapy in different populations.
Collapse
Affiliation(s)
- Thibaut Guiraud
- Montreal Heart Institute, Cardiovascular Prevention Centre-Centre PIC, Universit de Montral, Montral, Qubec, Canada.
| | | | | | | | | | | |
Collapse
|
150
|
Yi CX, Al-Massadi O, Donelan E, Lehti M, Weber J, Ress C, Trivedi C, Müller TD, Woods SC, Hofmann SM. Exercise protects against high-fat diet-induced hypothalamic inflammation. Physiol Behav 2012; 106:485-90. [DOI: 10.1016/j.physbeh.2012.03.021] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 03/02/2012] [Accepted: 03/21/2012] [Indexed: 12/16/2022]
|