1
|
Chubanava S, Karavaeva I, Ehrlich AM, Justicia RM, Basse AL, Kulik I, Dalbram E, Ahwazi D, Heaselgrave SR, Trošt K, Stocks B, Hodek O, Rodrigues RN, Havelund JF, Schlabs FL, Larsen S, Yonamine CY, Henriquez-Olguín C, Giustarini D, Rossi R, Gerhart-Hines Z, Moritz T, Zierath JR, Sakamoto K, Jensen TE, Færgeman NJ, Lavery GG, Deshmukh AS, Treebak JT. NAD depletion in skeletal muscle does not compromise muscle function or accelerate aging. Cell Metab 2025:S1550-4131(25)00212-8. [PMID: 40311622 DOI: 10.1016/j.cmet.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/27/2025] [Accepted: 04/08/2025] [Indexed: 05/03/2025]
Abstract
Nicotinamide adenine dinucleotide (NAD) is a ubiquitous electron carrier essential for energy metabolism and post-translational modification of numerous regulatory proteins. Dysregulations of NAD metabolism are widely regarded as detrimental to health, with NAD depletion commonly implicated in aging. However, the extent to which cellular NAD concentration can decline without adverse consequences remains unclear. To investigate this, we generated a mouse model in which nicotinamide phosphoribosyltransferase (NAMPT)-mediated NAD+ biosynthesis was disrupted in adult skeletal muscle. The intervention resulted in an 85% reduction in muscle NAD+ abundance while maintaining tissue integrity and functionality, as demonstrated by preserved muscle morphology, contractility, and exercise tolerance. This absence of functional impairments was further supported by intact mitochondrial respiratory capacity and unaltered muscle transcriptomic and proteomic profiles. Furthermore, lifelong NAD depletion did not accelerate muscle aging or impair whole-body metabolism. Collectively, these findings suggest that NAD depletion does not contribute to age-related decline in skeletal muscle function.
Collapse
Affiliation(s)
- Sabina Chubanava
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Iuliia Karavaeva
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amy M Ehrlich
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Roger M Justicia
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Astrid L Basse
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ivan Kulik
- Institute of Translational Stem Cell Research, Helmholtz Diabetes Center, Munich, Germany
| | - Emilie Dalbram
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Danial Ahwazi
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Samuel R Heaselgrave
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK; Centre for Systems Health and Integrated Metabolic Research, Department of Biosciences, Nottingham Trent University, Nottingham, UK
| | - Kajetan Trošt
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ben Stocks
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ondřej Hodek
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Raissa N Rodrigues
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper F Havelund
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Farina L Schlabs
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Steen Larsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Caio Y Yonamine
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Carlos Henriquez-Olguín
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark; Center for Exercise Physiology and Metabolism, Department of Kinesiology, Faculty of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - Daniela Giustarini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Ranieri Rossi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Zachary Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Moritz
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Juleen R Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Section of Integrative Physiology, Department of Molecular Medicine and Surgery and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas E Jensen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Nils J Færgeman
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK; Centre for Systems Health and Integrated Metabolic Research, Department of Biosciences, Nottingham Trent University, Nottingham, UK
| | - Atul S Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
2
|
Elliehausen CJ, Olszewski SS, Minton DM, Spiegelhoff AL, Shult CG, Zhu WG, Hornberger TA, Konopka AR. PoWeR elicits intracellular signaling, mitochondrial adaptations, and hypertrophy in multiple muscles consistent with endurance and resistance exercise training. J Appl Physiol (1985) 2025; 138:1034-1049. [PMID: 40100208 DOI: 10.1152/japplphysiol.00872.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/14/2024] [Accepted: 02/25/2025] [Indexed: 03/20/2025] Open
Abstract
Physical activity guidelines recommend both endurance and resistance exercise to improve and maintain overall health. Recently, progressive weighted wheel running (PoWeR), a voluntary, progressive, and high-volume exercise paradigm, was posited as a singular prototype of combined endurance and resistance exercise in mice as evident by enhanced capillarization and hypertrophy of select plantar flexor muscles. Despite growing interest in this model, it remains incompletely characterized if PoWeR resembles the acute and chronic responses to resistance and/or endurance exercise in humans. Therefore, the purpose of this study was to assess canonical signaling events, mitochondrial bioenergetics, and cellular adaptations across multiple extensor and flexor muscles of the fore- and hindlimbs that may be conducive for whole-body functional improvements as traditionally observed in humans. Eight weeks of PoWeR (∼8 km/day) improved glucose metabolism, exercise capacity, body composition, and bone mineral density as well as increased mass, myofiber cross-sectional area (CSA), and oxidative myofiber type distribution in the soleus, plantaris, and flexor digitorum longus (FDL). Using two ex vivo high-resolution fluororespirometry protocols that model in vivo physiological conditions, PoWeR decreased mitochondrial ADP sensitivity which was accompanied by greater mitochondrial H2O2 emissions, respiration, conductance, and protein content in the vastus lateralis, gastrocnemius, and triceps in muscle-specific fashion. Three days of short-term PoWeR stimulated mTOR complex 1 (mTORC1) and AMP activated protein kinase (AMPK) signaling in soleus, plantaris, and/or FDL in line with the hypertrophic and metabolic adaptations observed with long-term training. Collectively, these data support PoWeR as a suitable paradigm in mice to model the acute signaling and chronic adaptations associated with endurance and resistance exercise in humans.NEW & NOTEWORTHY Using PoWeR, we evaluated skeletal muscle mitochondrial and hypertrophic adaptions revealing muscle-specific adaptations across fore and hind limbs consistent with endurance and resistance exercise in humans. We present a short-term PoWeR paradigm that identifies muscle-specific signaling responses thought to support long-term adaptions to PoWeR. These data provide further support for PoWeR as a model to resemble the metabolic and anabolic adaptions to endurance and resistance exercise in humans.
Collapse
Affiliation(s)
- Christian J Elliehausen
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States
| | - Szczepan S Olszewski
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States
| | - Dennis M Minton
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States
| | - Audrey L Spiegelhoff
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States
| | - Carolyn G Shult
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States
| | - Wenyuan G Zhu
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Troy A Hornberger
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Adam R Konopka
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States
| |
Collapse
|
3
|
Pattamaprapanont P, Nava RC, Formato M, Cooney EM, Pinto AP, Alves-Wagner AB, Das A, Guan Y, Lessard SJ. A ketogenic diet enhances aerobic exercise adaptation and promotes muscle mitochondrial remodeling in hyperglycemic mice. RESEARCH SQUARE 2025:rs.3.rs-5814971. [PMID: 39975925 PMCID: PMC11838742 DOI: 10.21203/rs.3.rs-5814971/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
VO2peak is a key health benefit of aerobic exercise; however, chronic hyperglycemia is associated with persistently low VO2peak due to an impaired adaptive response to training. Here, we tested whether reducing blood glucose with a low-carbohydrate/high-fat "ketogenic" diet could restore aerobic exercise adaptation in a mouse model of hyperglycemia. Hyperglycemia was induced by streptozotocin (STZ) and mice were stratified to standard chow (STZ-CHOW), or a ketogenic diet (STZ-KETO), which rapidly normalized blood glucose. After aerobic exercise training, improvements in VO2peak were blunted in STZ-CHOW, but exercise response was restored in STZ-KETO. Improved VO2peak in STZ-KETO was associated with enhanced aerobic remodeling of skeletal muscle, including a more oxidative fiber-type and increased capillary density, along with restoration of circulating angiogenic markers. Moreover, KETO induced exercise-independent effects on muscle mitochondrial remodeling and mitochondrial dynamics, significantly increasing fatty acid oxidation. Our results identify a ketogenic diet as a potential therapy to improve aerobic exercise response in the growing population with hyperglycemia due to diabetes and other metabolic conditions.
Collapse
Affiliation(s)
- Pattarawan Pattamaprapanont
- Research Division; Joslin Diabetes Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Center for Exercise Medicine Research; Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Roberto C. Nava
- Research Division; Joslin Diabetes Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Mia Formato
- Research Division; Joslin Diabetes Center, Boston, MA, USA
| | | | | | - Ana B. Alves-Wagner
- Research Division; Joslin Diabetes Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Anamica Das
- Research Division; Joslin Diabetes Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yuntian Guan
- Research Division; Joslin Diabetes Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Sarah J. Lessard
- Research Division; Joslin Diabetes Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Center for Exercise Medicine Research; Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
- Department of Human Nutrition, Foods, and Exercise; Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| |
Collapse
|
4
|
Shahsavarnajand Bonab H, Tolouei Azar J, Soraya H, Nouri Habashi A. Aerobic interval training preconditioning protocols inhibit isoproterenol-induced pathological cardiac remodeling in rats: Implications on oxidative balance, autophagy, and apoptosis. SPORTS MEDICINE AND HEALTH SCIENCE 2024; 6:344-357. [PMID: 39309465 PMCID: PMC11411311 DOI: 10.1016/j.smhs.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 09/25/2024] Open
Abstract
This study aimed to investigate the potential cardioprotective effects of moderate and high-intensity aerobic interval training (MIIT and HIIT) preconditioning. The focus was on histological changes, pro-oxidant-antioxidant balance, autophagy initiation, and apoptosis in myocardial tissue incited by isoproterenol-induced pathological cardiac remodeling (ISO-induced PCR). Male Wistar rats were randomly divided into control (n = 6), ISO (n = 8), MIIT (n = 4), HIIT (n = 4), MIIT + ISO (n = 8), and HIIT + ISO (n = 8) groups. The MIIT and HIIT protocols were administered for 10 weeks, followed by the induction of cardiac remodeling using subcutaneous injection of ISO (100 mg/kg for two consecutive days). Alterations in heart rate (HR), mean arterial pressure (MAP), rate pressure product (RPP), myocardial oxygen consumption (MV ˙ O2), cardiac hypertrophy, histopathological changes, pro-oxidant-antioxidant balance, autophagy biomarkers (Beclin-1, Atg7, p62, LC3 I/II), and apoptotic cell distribution were measured. The findings revealed that the MIIT + ISO and HIIT + ISO groups demonstrated diminished myocardial damage, hemorrhage, immune cell infiltration, edema, necrosis, and apoptosis compared to ISO-induced rats. MIIT and HIIT preconditioning mitigated HR, enhanced MAP, and preserved MV ˙ O2 and RPP. The pro-oxidant-antioxidant balance was sustained in both MIIT + ISO and HIIT + ISO groups, with MIIT primarily inhibiting pro-apoptotic autophagy progression through maintaining pro-oxidant-antioxidant balance, and HIIT promoting pro-survival autophagy. The results demonstrated the beneficial effects of both MIIT and HIIT as AITs preconditioning in ameliorating ISO-induced PCR by improving exercise capacity, hemodynamic parameters, and histopathological changes. Some of these protective effects can be attributed to the modulation of cardiac apoptosis, autophagy, and oxidative stress.
Collapse
Affiliation(s)
- Hakimeh Shahsavarnajand Bonab
- Department of Exercise Physiology and Corrective Exercises, Faculty of Sport Sciences, Urmia University, Urmia, Iran
| | - Javad Tolouei Azar
- Department of Exercise Physiology and Corrective Exercises, Faculty of Sport Sciences, Urmia University, Urmia, Iran
| | - Hamid Soraya
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Akbar Nouri Habashi
- Department of Exercise Physiology and Corrective Exercises, Faculty of Sport Sciences, Urmia University, Urmia, Iran
| |
Collapse
|
5
|
Damasceno TR, Tanaka DM, Magnani EF, Oliveira RDB, Pereira DAG, Vieira-Alves I, Lemos VS, Cabeza JM, Fabricio CG, Resende AA, Gonçalves DAP, Zanetti GDO, de Carvalho EEV, Simões MV, Oliveira LFL. Exercise Training Reduces Inflammation and Fibrosis and Preserves Myocardial Function and Perfusion in a Model of Chronic Chagas Cardiomyopathy. Arq Bras Cardiol 2024; 121:e20230707. [PMID: 39258653 PMCID: PMC11495816 DOI: 10.36660/abc.20230707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/05/2024] [Accepted: 04/24/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Chronic Chagas cardiomyopathy (CCC) is caused by an inflammatory process induced by Trypanosoma cruzi, which leads to myocarditis with reactive and reparative fibrosis. CCC progresses with myocardial perfusion abnormalities and histopathological events that affect cardiorespiratory fitness (CRF). OBJECTIVES We evaluated the effects of aerobic physical training (APT) on myocardial perfusion and on morphological and functional impairments related with inflammation and fibrosis in Syrian hamsters with CCC. As a secondary objective, we analyzed the cross-sectional areas of the skeletal muscle. METHODS Hamsters with CCC and their respective controls were divided into four groups: CCC sedentary, CCC-APT, sedentary control and APT control. Seven months after infection, the animals underwent echocardiography, myocardial perfusion scintigraphy and cardiopulmonary exercise testing. Moderate-intensity APT was performed for fifty minutes, five times a week, for eight weeks. Subsequently, the animals were reassessed. Histopathological analysis was conducted after the above-mentioned procedures. The level of significance was set at 5% in all analyses (p<0.05). RESULTS CCC sedentary animals presented worse myocardial perfusion defects (MPD) over time, reduced left ventricle ejection fraction (LVEF) and showed more inflammation and fibrosis when compared to other groups (mixed ANOVA analysis). Conversely, APT was able to mitigate the progression of MPD, ameliorate inflammation and fibrosis and improve CRF efficiency in CCC-APT animals. CONCLUSIONS Our study demonstrated that APT ameliorated cardiac dysfunction, MPD, and reduced inflammation and fibrosis in CCC hamster models. Additionally, CCC-SED animals presented skeletal muscle atrophy while CCC-APT animals showed preserved skeletal muscle CSA. Understanding APT's effects on CCC's pathophysiological dimensions is crucial for future research and therapeutic interventions.
Collapse
Affiliation(s)
- Thayrine R. Damasceno
- Universidade Federal de Minas GeraisBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais, Belo Horizonte, MG – Brasil
| | - Denise M. Tanaka
- Universidade de São PauloFaculdade de Medicina de Ribeirão PretoRibeirão PretoSPBrasilFaculdade de Medicina de Ribeirão Preto – Universidade de São Paulo, Ribeirão Preto, SP – Brasil
| | - Enrico F. Magnani
- Universidade Federal de Minas GeraisBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais, Belo Horizonte, MG – Brasil
| | - Rafael D. B. Oliveira
- Universidade Federal de Minas GeraisBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais, Belo Horizonte, MG – Brasil
| | - Danielle A. G. Pereira
- Universidade Federal de Minas GeraisBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais, Belo Horizonte, MG – Brasil
| | - Ildernandes Vieira-Alves
- Universidade Federal de Minas GeraisBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais, Belo Horizonte, MG – Brasil
| | - Virginia S. Lemos
- Universidade Federal de Minas GeraisBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais, Belo Horizonte, MG – Brasil
| | - Jorge M. Cabeza
- Hospital Israelita Albert EinsteinSão PauloSPBrasilHospital Israelita Albert Einstein, São Paulo, SP – Brasil
| | - Camila G. Fabricio
- Universidade de São PauloFaculdade de Medicina de Ribeirão PretoRibeirão PretoSPBrasilFaculdade de Medicina de Ribeirão Preto – Universidade de São Paulo, Ribeirão Preto, SP – Brasil
| | - Alessandra A. Resende
- Universidade de São PauloFaculdade de Medicina de Ribeirão PretoRibeirão PretoSPBrasilFaculdade de Medicina de Ribeirão Preto – Universidade de São Paulo, Ribeirão Preto, SP – Brasil
| | - Dawit A. P. Gonçalves
- Universidade Federal de Minas GeraisBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais, Belo Horizonte, MG – Brasil
| | - Gustavo de Oliveira Zanetti
- Universidade Federal de Minas GeraisBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais, Belo Horizonte, MG – Brasil
| | - Eduardo E. Vieira de Carvalho
- Universidade Federal do Triângulo MineiroUberabaMGBrasilUniversidade Federal do Triângulo Mineiro, Uberaba, MG – Brasil
| | - Marcus V. Simões
- Universidade de São PauloFaculdade de Medicina de Ribeirão PretoRibeirão PretoSPBrasilFaculdade de Medicina de Ribeirão Preto – Universidade de São Paulo, Ribeirão Preto, SP – Brasil
| | - Luciano F. L. Oliveira
- Universidade Federal de Minas GeraisBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais, Belo Horizonte, MG – Brasil
- Universidade Federal do Triângulo MineiroUberabaMGBrasilUniversidade Federal do Triângulo Mineiro, Uberaba, MG – Brasil
| |
Collapse
|
6
|
Pan L, He X, Xu R, Bhattarai U, Niu Z, do Carmo J, Sun Y, Zeng H, Clemmer JS, Chen JX, Chen Y. Endothelial specific prolyl hydroxylase domain-containing protein 2 deficiency attenuates aging-related obesity and exercise intolerance. GeroScience 2024; 46:3945-3956. [PMID: 38462569 PMCID: PMC11226575 DOI: 10.1007/s11357-024-01108-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 02/20/2024] [Indexed: 03/12/2024] Open
Abstract
Obesity and exercise intolerance greatly reduce the life quality of older people. Prolyl hydroxylase domain-containing protein 2 (PHD2) is an important enzyme in modulating hypoxia-inducible factor-alpha (HIF) protein. Using vascular endothelial cell-specific PHD2 gene knockout (PHD2 ECKO) mice, we investigated the role of endothelial PHD2 in aging-related obesity and exercise capacity. Briefly, PHD2 ECKO mice were obtained by crossing PHD2-floxed mice with VE-Cadherin (Cdh5)-Cre transgenic mice. The effect of PHD2 ECKO on obesity and exercise capacity in PHD2 ECKO mice and control PHD2f/f mice were determined in young mice (6 to 7 months) and aged mice (16-18 months). We found that aged PHD2 ECKO mice, but not young mice, exhibited a lean phenotype, characterized by lower fat mass, and its ratio to lean weight, body weight, or tibial length, while their food uptake was not reduced compared with controls. Moreover, as compared with aged control mice, aged PHD2 ECKO mice exhibited increased oxygen consumption at rest and during exercise, and the maximum rate of oxygen consumption (VO2 max) during exercise. Furthermore, as compared with corresponding control mice, both young and aged PHD2 ECKO mice demonstrated improved glucose tolerance and lower insulin resistance. Together, these data demonstrate that inhibition of vascular endothelial PHD2 signaling significantly attenuates aging-related obesity, exercise intolerance, and glucose intolerance.
Collapse
Affiliation(s)
- Lihong Pan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, 2500 North State Street, Jackson, MS, 39216, USA
| | - Xiaochen He
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, 2500 North State Street, Jackson, MS, 39216, USA
| | - Rui Xu
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, 2500 North State Street, Jackson, MS, 39216, USA
| | - Umesh Bhattarai
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, 2500 North State Street, Jackson, MS, 39216, USA
| | - Ziru Niu
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, 2500 North State Street, Jackson, MS, 39216, USA
| | - Jussara do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, 2500 North State Street, Jackson, MS, 39216, USA
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Heng Zeng
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - John S Clemmer
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, 2500 North State Street, Jackson, MS, 39216, USA
| | - Jian-Xiong Chen
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Yingjie Chen
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, 2500 North State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
7
|
Pattamaprapanont P, Cooney EM, MacDonald TL, Paulo JA, Pan H, Dreyfuss JM, Lessard SJ. Matrisome proteomics reveals novel mediators of muscle remodeling with aerobic exercise training. Matrix Biol Plus 2024; 23:100159. [PMID: 39220302 PMCID: PMC11363848 DOI: 10.1016/j.mbplus.2024.100159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Skeletal muscle has a unique ability to remodel in response to stimuli such as contraction and aerobic exercise training. Phenotypic changes in muscle that occur with training such as a switch to a more oxidative fiber type, and increased capillary density contribute to the well-known health benefits of aerobic exercise. The muscle matrisome likely plays an important role in muscle remodeling with exercise. However, due to technical limitations in studying muscle ECM proteins, which are highly insoluble, little is known about the muscle matrisome and how it contributes to muscle remodeling. Here, we utilized two-fraction methodology to extract muscle proteins, combined with multiplexed tandem mass tag proteomic technology to identify 161 unique ECM proteins in mouse skeletal muscle. In addition, we demonstrate that aerobic exercise training induces remodeling of a significant proportion of the muscle matrisome. We performed follow-up experiments to validate exercise-regulated ECM targets in a separate cohort of mice using Western blotting and immunofluorescence imaging. Our data demonstrate that changes in several key ECM targets are strongly associated with muscle remodeling processes such as increased capillary density in mice. We also identify LOXL1 as a novel muscle ECM target associated with aerobic capacity in humans. In addition, publically available data and databases were used for in silico modeling to determine the likely cellular sources of exercise-induced ECM remodeling targets and identify ECM interaction networks. This work greatly enhances our understanding of ECM content and function in skeletal muscle and demonstrates an important role for ECM remodeling in the adaptive response to exercise. The raw MS data have been deposited to the ProteomeXchange with identifier PXD053003.
Collapse
Affiliation(s)
| | | | - Tara L. MacDonald
- Research Division, Joslin Diabetes Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Hui Pan
- Research Division, Joslin Diabetes Center, Boston, MA, USA
| | - Jonathan M. Dreyfuss
- Research Division, Joslin Diabetes Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Sarah J. Lessard
- Research Division, Joslin Diabetes Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
8
|
Aishwarya R, Abdullah CS, Remex NS, Bhuiyan MAN, Lu XH, Dhanesha N, Stokes KY, Orr AW, Kevil CG, Bhuiyan MS. Diastolic dysfunction in Alzheimer's disease model mice is associated with Aβ-amyloid aggregate formation and mitochondrial dysfunction. Sci Rep 2024; 14:16715. [PMID: 39030247 PMCID: PMC11271646 DOI: 10.1038/s41598-024-67638-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024] Open
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disease caused by the deposition of Aβ aggregates or neurofibrillary tangles. AD patients are primarily diagnosed with the concurrent development of several cardiovascular dysfunctions. While few studies have indicated the presence of intramyocardial Aβ aggregates, none of the studies have performed detailed analyses for pathomechanism of cardiac dysfunction in AD patients. This manuscript used aged APPSWE/PS1 Tg and littermate age-matched wildtype (Wt) mice to characterize cardiac dysfunction and analyze associated pathophysiology. Detailed assessment of cardiac functional parameters demonstrated the development of diastolic dysfunction in APPSWE/PS1 Tg hearts compared to Wt hearts. Muscle function evaluation showed functional impairment (decreased exercise tolerance and muscle strength) in APPSWE/PS1 Tg mice. Biochemical and histochemical analysis revealed Aβ aggregate accumulation in APPSWE/PS1 Tg mice myocardium. APPSWE/PS1 Tg mice hearts also demonstrated histopathological remodeling (increased collagen deposition and myocyte cross-sectional area). Additionally, APPSWE/PS1 Tg hearts showed altered mitochondrial dynamics, reduced antioxidant protein levels, and impaired mitochondrial proteostasis compared to Wt mice. APPSWE/PS1 Tg hearts also developed mitochondrial dysfunction with decreased OXPHOS and PDH protein complex expressions, altered ETC complex dynamics, decreased complex activities, and reduced mitochondrial respiration. Our results indicated that Aβ aggregates in APPSWE/PS1 Tg hearts are associated with defects in mitochondrial respiration and complex activities, which may collectively lead to cardiac diastolic dysfunction and myocardial pathological remodeling.
Collapse
Affiliation(s)
- Richa Aishwarya
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
| | - Chowdhury S Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
| | - Naznin Sultana Remex
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Mohammad Alfrad Nobel Bhuiyan
- Department of Medicine, Division of Clinical Informatics, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Xiao-Hong Lu
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Nirav Dhanesha
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
| | - Karen Y Stokes
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - A Wayne Orr
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Christopher G Kevil
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Md Shenuarin Bhuiyan
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA.
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA.
| |
Collapse
|
9
|
Raph SM, Calderin EP, Nong Y, Brittian K, Garrett L, Zhang D, Nystoriak MA. Kv beta complex facilitates exercise-induced augmentation of myocardial perfusion and cardiac growth. Front Cardiovasc Med 2024; 11:1411354. [PMID: 38978788 PMCID: PMC11228310 DOI: 10.3389/fcvm.2024.1411354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/21/2024] [Indexed: 07/10/2024] Open
Abstract
The oxygen sensitivity of voltage-gated potassium (Kv) channels regulates cardiovascular physiology. Members of the Kv1 family interact with intracellular Kvβ proteins, which exhibit aldo-keto reductase (AKR) activity and confer redox sensitivity to Kv channel gating. The Kvβ proteins contribute to vasoregulation by controlling outward K+ currents in smooth muscle upon changes in tissue oxygen consumption and demand. Considering exercise as a primary physiological stimulus of heightened oxygen demand, the current study tested the role of Kvβ proteins in exercise performance, exercise-induced adaptations in myocardial perfusion, and physiological cardiac growth. Our findings reveal that genetic ablation of Kvβ2 proteins diminishes baseline exercise capacity in mice and attenuates the enhancement in exercise performance observed after long-term training. Moreover, we demonstrate that Kvβ2 proteins are critical for exercise-mediated enhancement in myocardial perfusion during cardiac stress as well as adaptive changes in cardiac structure. Our results underscore the importance of Kvβ proteins in metabolic vasoregulation, highlighting their role in modulating both exercise capacity and cardiovascular benefits associated with training. Furthermore, our study sheds light on a novel molecular target for enhancing exercise performance and improving the health benefits associated with exercise training in patients with limited capacity for physical activity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Matthew A. Nystoriak
- Center for Cardiometabolic Science, Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY, United States
| |
Collapse
|
10
|
Wongong R, Kijtawornrat A, Srichomthong C, Tongkobpeth S, Od-Ek P, Assawapitaksakul A, Caengprasath N, Khongphatthanayothin A, Porntaveetus T, Shotelersuk V. A novel BAG5 variant impairs the ER stress response pathway, causing dilated cardiomyopathy and arrhythmia. Sci Rep 2024; 14:11980. [PMID: 38796549 PMCID: PMC11127938 DOI: 10.1038/s41598-024-62764-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024] Open
Abstract
Pathogenic BAG5 variants recently linked to dilated cardiomyopathy (DCM) prompt further investigation into phenotypic, mutational, and pathomechanistic aspects. We explored the clinical and molecular characteristics of DCM associated with BAG5 variants, uncovering the consistently severe manifestations of the disease and its impact on the endoplasmic reticulum (ER) stress response. The analysis involved three siblings affected by DCM and arrhythmia, along with their four unaffected siblings, their unaffected father, and their mother who exhibited arrhythmia. The parents were consanguineous. Exome and Sanger sequencing identified a novel BAG5 variant, c.444_445delGA (p.Lys149AsnfsTer6), homozygous in affected siblings and heterozygous in parents and unaffected siblings. We generated heterozygous and homozygous Bag5 point mutant knock-in (KI) mice and evaluated cardiac pathophysiology under stress conditions, including tunicamycin (TN) administration. Bag5-/- mice displayed no abnormalities up to 12 months old and showed no anomalies during an exercise stress test. However, following TN injection, Bag5-/- mice exhibited significantly reduced left ventricular fractional shortening (LVFS) and ejection fraction (LVEF). Their cardiac tissues exhibited a notable increase in apoptotic cells, despite non-distinctive changes in CHOP and GRP78 levels. Interestingly, only Bag5 KI male mice demonstrated arrhythmia, which was more pronounced in Bag5-/- than in Bag5+/-males. Here, our study reveals a novel BAG5 mutation causing DCM by impairing the ER stress response, with observed sex-specific arrhythmia differences.
Collapse
Affiliation(s)
- Rutairat Wongong
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand
| | - Anusak Kijtawornrat
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Chalurmpon Srichomthong
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand
| | - Siraprapa Tongkobpeth
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Phichittra Od-Ek
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand
| | - Adjima Assawapitaksakul
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand
| | - Natarin Caengprasath
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand
| | - Apichai Khongphatthanayothin
- Center of Excellence in Arrhythmia Research Chulalongkorn University, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Bangkok Heart Hospital, Bangkok, Thailand
| | - Thantrira Porntaveetus
- Center of Excellence in Genomics and Precision Dentistry, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
- Graduate Program in Geriatric and Special Patients Care, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| | - Vorasuk Shotelersuk
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand
| |
Collapse
|
11
|
Herrera JJ, McAllister CM, Szczesniak D, Goddard R, Day SM. High-intensity exercise training using a rotarod instrument (RotaHIIT) significantly improves exercise capacity in mice. Physiol Rep 2024; 12:e15997. [PMID: 38697937 PMCID: PMC11065697 DOI: 10.14814/phy2.15997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/21/2024] [Accepted: 03/21/2024] [Indexed: 05/05/2024] Open
Abstract
Voluntary or forced exercise training in mice is used to assess functional capacity as well as potential disease-modifying effects of exercise over a range of cardiovascular disease phenotypes. Compared to voluntary wheel running, forced exercise training enables precise control of exercise workload and volume, and results in superior changes in cardiovascular performance. However, the use of a shock grid with treadmill-based training is associated with stress and risk of injury, and declining compliance with longer periods of training time for many mouse strains. With these limitations in mind, we designed a novel, high-intensity interval training modality (HIIT) for mice that is carried out on a rotarod. Abbreviated as RotaHIIT, this protocol establishes interval workload intensities that are not time or resource intensive, maintains excellent training compliance over time, and results in improved exercise capacity independent of sex when measured by treadmill graded exercise testing (GXT) and rotarod specific acceleration and endurance testing. This protocol may therefore be useful and easily implemented for a broad range of research investigations. As RotaHIIT training was not associated cardiac structural or functional changes, or changes in oxidative capacity in cardiac or skeletal muscle tissue, further studies will be needed to define the physiological adaptations and molecular transducers that are driving the training effect of this exercise modality.
Collapse
Affiliation(s)
- Jonathan J. Herrera
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
- Medical Scientist Training ProgramUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Christopher M. McAllister
- Department of Medicine, Division of Cardiovascular MedicineUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Danielle Szczesniak
- Department of Medicine, Division of Cardiovascular MedicineUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Rose‐Carmel Goddard
- Department of Medicine, Division of Cardiovascular MedicineUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Sharlene M. Day
- Department of Medicine, Division of Cardiovascular MedicineUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| |
Collapse
|
12
|
Legault EP, Ribeiro PAB, Petrenyov DR, Drumeva GO, Leduc C, Khullar S, DaSilva JN, Comtois AS, Tournoux FB. Effect of acute high-intensity interval exercise on a mouse model of doxorubicin-induced cardiotoxicity: a pilot study. BMC Sports Sci Med Rehabil 2024; 16:95. [PMID: 38671464 PMCID: PMC11046902 DOI: 10.1186/s13102-024-00881-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 04/12/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND It is unknown whether high-intensity interval exercise (HIIE) may potentiate or attenuate the cardiotoxic effect of chemotherapy agents such as doxorubicin (DOX) when performed shortly after treatment. The study aimed to investigate the effect of acute HIIE on cardiac function and structure performed either 1, 2 or 3 days after DOX injection in an animal model. METHODS Female C57bl/6 mice (n = 28), 70 days old, received a bolus 20 mg/kg intravenous tail vein DOX injection. Three exercise groups performed 1 HIIE session (16 sets of 1 min at 85-90% of peak running speed) at 1 (n = 7), 2 (n = 7), and 3 days (n = 8) following the DOX injection. A sedentary (SED) group of mice (n = 6) did not exercise. Animals underwent echocardiography under light anesthesia (isoflurane 0.5-1%) before and 7 days after the DOX injection. Animals were sacrificed on day 9 and hearts were collected for morphometric and histological analysis. RESULTS Animals exercising on day 3 had the smallest pre-post reduction in left ventricular fractional shortening (LVFS) (MΔ= -1.7 ± 3.3; p = 0.406) and the SED group had the largest reduction (MΔ=-6.8 ± 7.5; p = 0.009). After reclassification of animals according to their exercise compliance (performing > 8/16 of high-intensity bouts), LVFS in compliant mice was unchanged over time (LVFS MΔ= -1.3 ± 5.6; p = 0.396) while non-compliant animals had a LVFS reduction similar to sedentary animals. There were no significant differences in myocardial histology between groups. CONCLUSIONS In this pilot murine study, one single HIIE session did not exacerbate acute doxorubicin-induced cardiotoxicity. The timing of the HIIE session following DOX injection and the level of compliance to exercise could influence the negative impact of DOX on cardiac function.
Collapse
Affiliation(s)
- Elise P Legault
- @coeurlab research unit, Centre de recherche du Centre Hospitalier de l'Université de Montréal, 900 St Denis Street, Montréal, Québec, Canada.
- Département des sciences de l'activité physique, Université du Québec à Montréal, Montréal, Québec, Canada.
| | - Paula A B Ribeiro
- @coeurlab research unit, Centre de recherche du Centre Hospitalier de l'Université de Montréal, 900 St Denis Street, Montréal, Québec, Canada
| | - Daniil R Petrenyov
- @coeurlab research unit, Centre de recherche du Centre Hospitalier de l'Université de Montréal, 900 St Denis Street, Montréal, Québec, Canada
| | - Gergana O Drumeva
- @coeurlab research unit, Centre de recherche du Centre Hospitalier de l'Université de Montréal, 900 St Denis Street, Montréal, Québec, Canada
- Département de pharmacologie et physiologie, Université de Montréal, Montréal, Québec, Canada
| | - Charles Leduc
- @coeurlab research unit, Centre de recherche du Centre Hospitalier de l'Université de Montréal, 900 St Denis Street, Montréal, Québec, Canada
- Département de pathologie et biologie cellulaire de l'Université de Montréal, Montréal, Québec, Canada
| | - Sharmila Khullar
- @coeurlab research unit, Centre de recherche du Centre Hospitalier de l'Université de Montréal, 900 St Denis Street, Montréal, Québec, Canada
- Département de pathologie et biologie cellulaire de l'Université de Montréal, Montréal, Québec, Canada
| | - Jean N DaSilva
- @coeurlab research unit, Centre de recherche du Centre Hospitalier de l'Université de Montréal, 900 St Denis Street, Montréal, Québec, Canada
- Département de pharmacologie et physiologie, Université de Montréal, Montréal, Québec, Canada
- Département de radiologie, radio-oncologie et médecine nucléaire, Université de Montréal, Montréal, Québec, Canada
| | - Alain Steve Comtois
- Département des sciences de l'activité physique, Université du Québec à Montréal, Montréal, Québec, Canada
| | - François B Tournoux
- @coeurlab research unit, Centre de recherche du Centre Hospitalier de l'Université de Montréal, 900 St Denis Street, Montréal, Québec, Canada
- Service de Cardiologie du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
13
|
Gilbert CJ, Rabolli CP, Golubeva VA, Sattler KM, Wang M, Ketabforoush A, Arnold WD, Lepper C, Accornero F. YTHDF2 governs muscle size through a targeted modulation of proteostasis. Nat Commun 2024; 15:2176. [PMID: 38467649 PMCID: PMC10928198 DOI: 10.1038/s41467-024-46546-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 02/28/2024] [Indexed: 03/13/2024] Open
Abstract
The regulation of proteostasis is fundamental for maintenance of muscle mass and function. Activation of the TGF-β pathway drives wasting and premature aging by favoring the proteasomal degradation of structural muscle proteins. Yet, how this critical post-translational mechanism is kept in check to preserve muscle health remains unclear. Here, we reveal the molecular link between the post-transcriptional regulation of m6A-modified mRNA and the modulation of SMAD-dependent TGF-β signaling. We show that the m6A-binding protein YTHDF2 is essential to determining postnatal muscle size. Indeed, muscle-specific genetic deletion of YTHDF2 impairs skeletal muscle growth and abrogates the response to hypertrophic stimuli. We report that YTHDF2 controls the mRNA stability of the ubiquitin ligase ASB2 with consequences on anti-growth gene program activation through SMAD3. Our study identifies a post-transcriptional to post-translational mechanism for the coordination of gene expression in muscle.
Collapse
Affiliation(s)
- Christopher J Gilbert
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Charles P Rabolli
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Volha A Golubeva
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Kristina M Sattler
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Meifang Wang
- NextGen Precision Health, University of Missouri, Columbia, MO, USA
- Department of Physical Medicine and Rehabilitation, University of Missouri, Columbia, MO, USA
| | - Arsh Ketabforoush
- NextGen Precision Health, University of Missouri, Columbia, MO, USA
- Department of Physical Medicine and Rehabilitation, University of Missouri, Columbia, MO, USA
| | - W David Arnold
- NextGen Precision Health, University of Missouri, Columbia, MO, USA
- Department of Physical Medicine and Rehabilitation, University of Missouri, Columbia, MO, USA
- Department of Neurology, University of Missouri, Columbia, MO, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
- Division of Neuromuscular Disorders, Department of Neurology, The Ohio State University, Columbus, OH, USA
- Department of Physical Medicine and Rehabilitation, The Ohio State University, Columbus, OH, USA
| | - Christoph Lepper
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Federica Accornero
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA.
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA.
| |
Collapse
|
14
|
Tai YK, Iversen JN, Chan KKW, Fong CHH, Abdul Razar RB, Ramanan S, Yap LYJ, Yin JN, Toh SJ, Wong CJK, Koh PFA, Huang RYJ, Franco-Obregón A. Secretome from Magnetically Stimulated Muscle Exhibits Anticancer Potency: Novel Preconditioning Methodology Highlighting HTRA1 Action. Cells 2024; 13:460. [PMID: 38474424 PMCID: PMC10930715 DOI: 10.3390/cells13050460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
Briefly (10 min) exposing C2C12 myotubes to low amplitude (1.5 mT) pulsed electromagnetic fields (PEMFs) generated a conditioned media (pCM) that was capable of mitigating breast cancer cell growth, migration, and invasiveness in vitro, whereas the conditioned media harvested from unexposed myotubes, representing constitutively released secretome (cCM), was less effective. Administering pCM to breast cancer microtumors engrafted onto the chorioallantoic membrane of chicken eggs reduced tumor volume and vascularity. Blood serum collected from PEMF-exposed or exercised mice allayed breast cancer cell growth, migration, and invasiveness. A secretome preconditioning methodology is presented that accentuates the graded anticancer potencies of both the cCM and pCM harvested from myotubes, demonstrating an adaptive response to pCM administered during early myogenesis that emulated secretome-based exercise adaptations observed in vivo. HTRA1 was shown to be upregulated in pCM and was demonstrated to be necessary and sufficient for the anticancer potency of the pCM; recombinant HTRA1 added to basal media recapitulated the anticancer effects of pCM and antibody-based absorption of HTRA1 from pCM precluded its anticancer effects. Brief and non-invasive PEMF stimulation may represent a method to commandeer the secretome response of muscle, both in vitro and in vivo, for clinical exploitation in breast and other cancers.
Collapse
Affiliation(s)
- Yee Kit Tai
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.I.); (C.J.K.W.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Jan Nikolas Iversen
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.I.); (C.J.K.W.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore
| | - Karen Ka Wing Chan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.I.); (C.J.K.W.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore
| | - Charlene Hui Hua Fong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.I.); (C.J.K.W.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore
| | - Rafhanah Banu Abdul Razar
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.I.); (C.J.K.W.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore
| | - Sharanya Ramanan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.I.); (C.J.K.W.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore
| | - Lye Yee Jasmine Yap
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.I.); (C.J.K.W.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore
| | - Jocelyn Naixin Yin
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.I.); (C.J.K.W.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore
| | - Shi Jie Toh
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.I.); (C.J.K.W.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore
| | - Craig Jun Kit Wong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.I.); (C.J.K.W.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore
| | - Pei Fern Angele Koh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore (R.Y.J.H.)
| | - Ruby Yun Ju Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore (R.Y.J.H.)
- Department of Obstetrics & Gynaecology, National University of Singapore, Singapore 119228, Singapore
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
- School of Medicine, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
| | - Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.I.); (C.J.K.W.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- Competence Center for Applied Biotechnology and Molecular Medicine, University of Zürich, 8057 Zürich, Switzerland
| |
Collapse
|
15
|
Zanetti GDO, Pessoa PWM, Vieira TS, Garcia RDA, Santos Barbosa NH, Arantes RME, Kettelhut IDC, Navegantes LCC, Wanner SP, Soares DD, Gonçalves DAP. Long-term heat acclimation training in mice: Similar metabolic and running performance adaptations despite a lower absolute intensity than training at temperate conditions. J Therm Biol 2024; 119:103797. [PMID: 38340467 DOI: 10.1016/j.jtherbio.2024.103797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/06/2023] [Accepted: 01/12/2024] [Indexed: 02/12/2024]
Abstract
This study investigated the impact of long-term heat acclimation (HA) training on mouse thermoregulation, metabolism, and running performance in temperate (T) and hot (H) environments. Male Swiss mice were divided into 1) Sedentary (SED) mice kept in T (22 °C; SED/T), 2) Endurance Trained mice (ET, 1 h/day, 5 days/week, 8 weeks, 60 % of maximum speed) in T (ET/T), 3) SED kept in H (32 °C; SED/H), and 4) ET in H (ET/H). All groups performed incremental load tests (ILT) in both environments before (pre-ET) and after four and eight weeks of ET. In the pre-ET period, H impaired (∼30 %) performance variables (maximum speed and external work) and increased (1.3 °C) maximum abdominal body temperature compared with T. In T, after four weeks, although ET/H exercised at a lower (∼30 %) absolute intensity than ET/T, performance variables and aerobic power (peak oxygen uptake, VO2peak) were similarly increased in both ET groups compared with SED/T. After eight weeks, the external work was higher in both ET groups compared with SED/T. Only ET/T significantly increased VO2peak (∼11 %) relative to its pre-ET period. In H, only after eight weeks, both ET groups improved (∼19 %) maximum speed and reduced (∼46 %) post-ILT blood lactate concentrations compared with their respective pre-ET values. Liver glycogen content increased (34 %) in both ET groups and SED/H compared with SED/T. Thus, ET/H was performed at a lower absolute intensity but promoted similar effects to ET/T on metabolism, aerobic power, and running performance. Our findings open perspectives for applying HA training as part of a training program or orthopedic and metabolic rehabilitation programs in injured or even obese animals, reducing mechanical load with equivalent or higher physiological demand.
Collapse
Affiliation(s)
- Gustavo de Oliveira Zanetti
- Exercise Physiology Laboratory (LAFISE), School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Pedro William Martins Pessoa
- Exercise Physiology Laboratory (LAFISE), School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Tales Sambrano Vieira
- Exercise Physiology Laboratory (LAFISE), School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rodrigo de Almeida Garcia
- Exercise Physiology Laboratory (LAFISE), School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Nicolas Henrique Santos Barbosa
- Exercise Physiology Laboratory (LAFISE), School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rosa Maria Esteves Arantes
- Department of Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Isis do Carmo Kettelhut
- Departments of Biochemistry & Immunology, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | - Samuel Penna Wanner
- Exercise Physiology Laboratory (LAFISE), School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Danusa Dias Soares
- Exercise Physiology Laboratory (LAFISE), School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Dawit Albieiro Pinheiro Gonçalves
- Exercise Physiology Laboratory (LAFISE), School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Section of Sports Physiology (SFE), Sports Training Center (CTE), Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
16
|
Yoshikawa A, Iizuka M, Kanamaru M, Kamijo S, Ohtaki H, Izumizaki M. Exercise evaluation with metabolic and ventilatory responses and blood lactate concentration in mice. Respir Physiol Neurobiol 2023; 318:104163. [PMID: 37734454 DOI: 10.1016/j.resp.2023.104163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 08/17/2023] [Accepted: 09/16/2023] [Indexed: 09/23/2023]
Abstract
This study aimed to clarify the differential exercise capacity between 2-month-old and 10-month-old mice using an incremental running test. Metabolic and ventilatory responses and blood lactate concentration were measured to evaluate exercise capacity. We examined whether incremental running test results reflected metabolic and ventilatory responses and blood lactate concentration observed during the steady-state running test. Metabolic response significantly declined with age, whereas ventilatory response was similar between the groups. A low-intensity/moderate exercise load of 10/min in an incremental running test was performed on both mice for 30 min. They showed a characteristic pattern in ventilatory response in 10-month mice. The results of incremental running tests didn't necessarily reflect the steady-state metabolic and ventilatory responses because some parameters showed an approximation and others did not in incremental and steady-state tests, which changed with age. Our study suggests metabolic and ventilatory responses depending on age and provides basic knowledge regarding the objective and quantitative assessment of treadmill running in an animal model.
Collapse
Affiliation(s)
- Akira Yoshikawa
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan; Division of Health Science Education, Showa University School of Nursing and Rehabilitation Sciences, Yokohama, Japan.
| | - Makito Iizuka
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan
| | - Mitsuko Kanamaru
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan; Faculty of Arts and Sciences at Fujiyoshida, Showa University, Yamanashi, Japan
| | - Shotaro Kamijo
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan; Department of Physiology, Showa University School of Pharmacy, Tokyo, Japan
| | - Hirokazu Ohtaki
- Department of Functional Neurobiology, Tokyo University of Pharmacy and Life Sciences, School of Pharmacy, Hachioji, Japan; Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| | - Masahiko Izumizaki
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
17
|
Nichtová Z, Fernandez-Sanz C, De La Fuente S, Yuan Y, Hurst S, Lanvermann S, Tsai HY, Weaver D, Baggett A, Thompson C, Bouchet-Marquis C, Várnai P, Seifert EL, Dorn GW, Sheu SS, Csordás G. Enhanced Mitochondria-SR Tethering Triggers Adaptive Cardiac Muscle Remodeling. Circ Res 2023; 132:e171-e187. [PMID: 37057625 PMCID: PMC10213149 DOI: 10.1161/circresaha.122.321833] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 03/29/2023] [Indexed: 04/15/2023]
Abstract
BACKGROUND Cardiac contractile function requires high energy from mitochondria, and Ca2+ from the sarcoplasmic reticulum (SR). Via local Ca2+ transfer at close mitochondria-SR contacts, cardiac excitation feedforward regulates mitochondrial ATP production to match surges in demand (excitation-bioenergetics coupling). However, pathological stresses may cause mitochondrial Ca2+ overload, excessive reactive oxygen species production and permeability transition, risking homeostatic collapse and myocyte loss. Excitation-bioenergetics coupling involves mitochondria-SR tethers but the role of tethering in cardiac physiology/pathology is debated. Endogenous tether proteins are multifunctional; therefore, nonselective targets to scrutinize interorganelle linkage. Here, we assessed the physiological/pathological relevance of selective chronic enhancement of cardiac mitochondria-SR tethering. METHODS We introduced to mice a cardiac muscle-specific engineered tether (linker) transgene with a fluorescent protein core and deployed 2D/3D electron microscopy, biochemical approaches, fluorescence imaging, in vivo and ex vivo cardiac performance monitoring and stress challenges to characterize the linker phenotype. RESULTS Expressed in the mature cardiomyocytes, the linker expanded and tightened individual mitochondria-junctional SR contacts; but also evoked a marked remodeling with large dense mitochondrial clusters that excluded dyads. Yet, excitation-bioenergetics coupling remained well-preserved, likely due to more longitudinal mitochondria-dyad contacts and nanotunnelling between mitochondria exposed to junctional SR and those sealed away from junctional SR. Remarkably, the linker decreased female vulnerability to acute massive β-adrenergic stress. It also reduced myocyte death and mitochondrial calcium-overload-associated myocardial impairment in ex vivo ischemia/reperfusion injury. CONCLUSIONS We propose that mitochondria-SR/endoplasmic reticulum contacts operate at a structural optimum. Although acute changes in tethering may cause dysfunction, upon chronic enhancement of contacts from early life, adaptive remodeling of the organelles shifts the system to a new, stable structural optimum. This remodeling balances the individually enhanced mitochondrion-junctional SR crosstalk and excitation-bioenergetics coupling, by increasing the connected mitochondrial pool and, presumably, Ca2+/reactive oxygen species capacity, which then improves the resilience to stresses associated with dysregulated hyperactive Ca2+ signaling.
Collapse
Affiliation(s)
- Zuzana Nichtová
- MitoCare, Pathology and Genomic Medicine, TJUH, Philadelphia, PA, USA
| | - Celia Fernandez-Sanz
- Center of Translational Medicine, TJUH, Philadelphia, PA, USA
- These authors contributed equally
| | - Sergio De La Fuente
- Center of Translational Medicine, TJUH, Philadelphia, PA, USA
- These authors contributed equally
| | - Yuexing Yuan
- Center of Translational Medicine, TJUH, Philadelphia, PA, USA
| | - Stephen Hurst
- MitoCare, Pathology and Genomic Medicine, TJUH, Philadelphia, PA, USA
| | | | - Hui-Ying Tsai
- Center of Translational Medicine, TJUH, Philadelphia, PA, USA
| | - David Weaver
- MitoCare, Pathology and Genomic Medicine, TJUH, Philadelphia, PA, USA
| | - Ariele Baggett
- MitoCare, Pathology and Genomic Medicine, TJUH, Philadelphia, PA, USA
| | | | | | - Péter Várnai
- Department of Physiology, Faculty of Medicine, Semmelweis Univ., Budapest, Hungary
| | - Erin L Seifert
- MitoCare, Pathology and Genomic Medicine, TJUH, Philadelphia, PA, USA
| | - Gerald W Dorn
- Center for Pharmacogenomics, John T. Milliken Dep. Med., WUSM, St Louis, MO, USA
| | - Shey-Shing Sheu
- Center of Translational Medicine, TJUH, Philadelphia, PA, USA
| | - György Csordás
- MitoCare, Pathology and Genomic Medicine, TJUH, Philadelphia, PA, USA
| |
Collapse
|
18
|
Bonilla IM, Baine S, Pokrass A, Mariángelo JIE, Kalyanasundaram A, Bogdanov V, Mezache L, Sakuta G, Beard CM, Belevych A, Tikunova S, Terentyeva R, Terentyev D, Davis J, Veeraraghavan R, Carnes CA, Györke S. STIM1 ablation impairs exercise-induced physiological cardiac hypertrophy and dysregulates autophagy in mouse hearts. J Appl Physiol (1985) 2023; 134:1287-1299. [PMID: 36995910 PMCID: PMC10190841 DOI: 10.1152/japplphysiol.00363.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 03/13/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023] Open
Abstract
Cardiac stromal interaction molecule 1 (STIM1), a key mediator of store-operated Ca2+ entry (SOCE), is a known determinant of cardiomyocyte pathological growth in hypertrophic cardiomyopathy. We examined the role of STIM1 and SOCE in response to exercise-dependent physiological hypertrophy. Wild-type (WT) mice subjected to exercise training (WT-Ex) showed a significant increase in exercise capacity and heart weight compared with sedentary (WT-Sed) mice. Moreover, myocytes from WT-Ex hearts displayed an increase in length, but not width, compared with WT-Sed myocytes. Conversely, exercised cardiac-specific STIM1 knock-out mice (cSTIM1KO-Ex), although displaying significant increase in heart weight and cardiac dilation, evidenced no changes in myocyte size and displayed a decreased exercise capacity, impaired cardiac function, and premature death compared with sedentary cardiac-specific STIM1 knock-out mice (cSTIM1KO-Sed). Confocal Ca2+ imaging demonstrated enhanced SOCE in WT-Ex myocytes compared with WT-Sed myocytes with no measurable SOCE detected in cSTIM1KO myocytes. Exercise training induced a significant increase in cardiac phospho-Akt Ser473 in WT mice but not in cSTIM1KO mice. No differences were observed in phosphorylation of mammalian target of rapamycin (mTOR) and glycogen synthase kinase (GSK) in exercised versus sedentary cSTIM1KO mice hearts. cSTIM1KO-Sed mice showed increased basal MAPK phosphorylation compared with WT-Sed that was not altered by exercise training. Finally, histological analysis revealed exercise resulted in increased autophagy in cSTIM1KO but not in WT myocytes. Taken together, our results suggest that adaptive cardiac hypertrophy in response to exercise training involves STIM1-mediated SOCE. Our results demonstrate that STIM1 is involved in and essential for the myocyte longitudinal growth and mTOR activation in response to endurance exercise training.NEW & NOTEWORTHY Store-operated Ca2+ entry (SOCE) has been implicated in pathological cardiac hypertrophy; however, its role in physiological hypertrophy is unknown. Here we report that SOCE is also essential for physiological cardiac hypertrophy and functional adaptations in response to endurance exercise. These adaptations were associated with activation of AKT/mTOR pathway and curtailed cardiac autophagy and degeneration. Thus, SOCE is a common mechanism and an important bifurcation point for signaling paths involved in physiological and pathological hypertrophy.
Collapse
Affiliation(s)
- Ingrid M Bonilla
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
- Veterans Affairs Caribbean Healthcare System, San Juan, Puerto Rico, United States
- Department of Pharmacology and Toxicology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico, United States
| | - Stephen Baine
- Department of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States
| | - Anastasia Pokrass
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Juan Ignacio Elio Mariángelo
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Anuradha Kalyanasundaram
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart & Lung Research Institute, Columbus, Ohio, United States
| | - Vladimir Bogdanov
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Louisa Mezache
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Galina Sakuta
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Casey M Beard
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Andriy Belevych
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Svetlana Tikunova
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Radmila Terentyeva
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Dmitry Terentyev
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Jonathan Davis
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Rengasayee Veeraraghavan
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Cynthia A Carnes
- Department of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States
| | - Sandor Györke
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| |
Collapse
|
19
|
Aishwarya R, Abdullah CS, Remex NS, Nitu S, Hartman B, King J, Bhuiyan MAN, Rom O, Miriyala S, Panchatcharam M, Orr AW, Kevil CG, Bhuiyan MS. Pathological Sequelae Associated with Skeletal Muscle Atrophy and Histopathology in G93A*SOD1 Mice. MUSCLES (BASEL, SWITZERLAND) 2023; 2:51-74. [PMID: 38516553 PMCID: PMC10956373 DOI: 10.3390/muscles2010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a complex systemic disease that primarily involves motor neuron dysfunction and skeletal muscle atrophy. One commonly used mouse model to study ALS was generated by transgenic expression of a mutant form of human superoxide dismutase 1 (SOD1) gene harboring a single amino acid substitution of glycine to alanine at codon 93 (G93A*SOD1). Although mutant-SOD1 is ubiquitously expressed in G93A*SOD1 mice, a detailed analysis of the skeletal muscle expression pattern of the mutant protein and the resultant muscle pathology were never performed. Using different skeletal muscles isolated from G93A*SOD1 mice, we extensively characterized the pathological sequelae of histological, molecular, ultrastructural, and biochemical alterations. Muscle atrophy in G93A*SOD1 mice was associated with increased and differential expression of mutant-SOD1 across myofibers and increased MuRF1 protein level. In addition, high collagen deposition and myopathic changes sections accompanied the reduced muscle strength in the G93A*SOD1 mice. Furthermore, all the muscles in G93A*SOD1 mice showed altered protein levels associated with different signaling pathways, including inflammation, mitochondrial membrane transport, mitochondrial lipid uptake, and antioxidant enzymes. In addition, the mutant-SOD1 protein was found in the mitochondrial fraction in the muscles from G93A*SOD1 mice, which was accompanied by vacuolized and abnormal mitochondria, altered OXPHOS and PDH complex protein levels, and defects in mitochondrial respiration. Overall, we reported the pathological sequelae observed in the skeletal muscles of G93A*SOD1 mice resulting from the whole-body mutant-SOD1 protein expression.
Collapse
Affiliation(s)
- Richa Aishwarya
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Chowdhury S. Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Naznin Sultana Remex
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Sadia Nitu
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Brandon Hartman
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Judy King
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | | | - Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Sumitra Miriyala
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Manikandan Panchatcharam
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - A. Wayne Orr
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Christopher G. Kevil
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Md. Shenuarin Bhuiyan
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| |
Collapse
|
20
|
Peres Valgas Da Silva C, Shettigar VK, Baer LA, Abay E, Pinckard KM, Vinales J, Sturgill SL, Vidal P, Ziolo MT, Stanford KI. Exercise training after myocardial infarction increases survival but does not prevent adverse left ventricle remodeling and dysfunction in high-fat diet fed mice. Life Sci 2022; 311:121181. [PMID: 36372212 PMCID: PMC9712172 DOI: 10.1016/j.lfs.2022.121181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
AIMS Aerobic exercise is an important component of rehabilitation after cardiovascular injuries including myocardial infarction (MI). In human studies, the beneficial effects of exercise after an MI are blunted in patients who are obese or glucose intolerant. Here, we investigated the effects of exercise on MI-induced cardiac dysfunction and remodeling in mice chronically fed a high-fat diet (HFD). MAIN METHODS C57Bl/6 male mice were fed either a standard (Chow; 21% kcal/fat) or HFD (60% kcal/fat) for 36 weeks. After 24 weeks of diet, the HFD mice were randomly subjected to an MI (MI) or a sham surgery (Sham). Following the MI or sham surgery, a subset of mice were subjected to treadmill exercise. KEY FINDINGS HFD resulted in obesity and glucose intolerance, and this was not altered by exercise or MI. MI resulted in decreased ejection fraction, increased left ventricle mass, increased end systolic and diastolic diameters, increased cardiac fibrosis, and increased expression of genes involved in cardiac hypertrophy and heart failure in the MI-Sed and MI-Exe mice. Exercise prevented HFD-induced cardiac fibrosis in Sham mice (Sham-Exe) but not in MI-Exe mice. Exercise did, however, reduce post-MI mortality. SIGNIFICANCE These data indicate that exercise significantly increased survival after MI in a model of diet-induced obesity independent of effects on cardiac function. These data have important translational ramifications because they demonstrate that environmental interventions, including diet, need to be carefully evaluated and taken into consideration to support the effects of exercise in the cardiac rehabilitation of patients who are obese.
Collapse
Affiliation(s)
- Carmem Peres Valgas Da Silva
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Vikram K Shettigar
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Lisa A Baer
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Eaman Abay
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Kelsey M Pinckard
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Jorge Vinales
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Sarah L Sturgill
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Pablo Vidal
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Mark T Ziolo
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, United States of America; Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, United States of America; Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, United States of America.
| |
Collapse
|
21
|
Gupta S, Khanal S, Bhavnani N, Mathias A, Lallo J, Kiriakou A, Ferrell J, Raman P. Sex-specific differences in atherosclerosis, thrombospondin-1, and smooth muscle cell differentiation in metabolic syndrome versus non-metabolic syndrome mice. Front Cardiovasc Med 2022; 9:1020006. [PMID: 36505365 PMCID: PMC9727198 DOI: 10.3389/fcvm.2022.1020006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 11/04/2022] [Indexed: 11/24/2022] Open
Abstract
Introduction Metabolic syndrome (MetS) amplifies the risks of atherosclerosis. Despite well-known sexual dimorphism in atherosclerosis, underlying mechanisms are poorly understood. Our previous findings highlight a proatherogenic protein, thrombospondin-1 (TSP-1), in hyperglycemia- or hyperleptinemia (mimicking obesity)-induced atherosclerosis. However, the role of TSP-1 in the development of atherosclerosis prompted by co-existing hyperglycemia and obesity, characteristic of MetS, is unknown. The goal of this study was to examine sex-specific differences in lesion progression in a model of combined MetS and atherosclerosis (KKAyApoE) and interrogate how these differences relate to TSP-1 expression. Methods Male and female KKAy+/-ApoE-/- (with ectopic agouti gene expression) and age-matched non-agouti KKAy-/-ApoE-/- littermates were placed on a standard laboratory diet from 4 to 24 weeks age followed by blood and tissue harvests for biochemical, molecular, and aortic root morphometric studies. Results Metabolic profiling confirmed MetS phenotype of KKAy+/-ApoE-/-; however, only male genotypes were glucose intolerant with elevated VLDL-cholesterol and VLDL-triglyceride levels. Aortic root morphometry demonstrated profound lipid-filled lesions, increased plaque area, and augmented inflammatory and SMC abundance in MetS vs non-MetS males. This increase in lesion burden was accompanied with elevated TSP-1 and attenuated LMOD-1 (SM contractile marker) and SRF (transcriptional activator of SM differentiation) expression in male MetS aortic vessels. In contrast, while lipid burden, plaque area, and TSP-1 expression increased in MetS and non-MetS female mice, there was no significant difference between these genotypes. Increased collagen content was noted in MetS and non-MetS genotypes, specific to female mice. Measurement of plasma testosterone revealed a link between the atherogenic phenotype and abnormally high or low testosterone levels. To interrogate whether TSP-1 plays a direct role in SMC de-differentiation in MetS, we generated KKAy+/- mice with and without global TSP-1 deletion. Immunoblotting showed increased SM contractile markers in male KKAy+/-TSP-1-/- aortic vessels vs male KKAy+/-TSP-1+/ +. In contrast, TSP-1 deletion had no effect on SM contractile marker expression in female genotypes. Conclusion Together, the current study implicates a role of plasma testosterone in sex-specific differences in atherosclerosis and TSP-1 expression in MetS vs non-MetS mice. Our data suggest a sex-dependent differential role of TSP-1 on SMC de-differentiation in MetS. Collectively, these findings underscore a fundamental link between TSP-1 and VSMC phenotypic transformation in MetS.
Collapse
Affiliation(s)
- Shreya Gupta
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States,School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Saugat Khanal
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States,School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Neha Bhavnani
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States,School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Amy Mathias
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Jason Lallo
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Ariana Kiriakou
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Jessica Ferrell
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States,School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Priya Raman
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States,School of Biomedical Sciences, Kent State University, Kent, OH, United States,*Correspondence: Priya Raman,
| |
Collapse
|
22
|
Omoto ACM, do Carmo JM, Nelson B, Aitken N, Dai X, Moak S, Flynn E, Wang Z, Mouton AJ, Li X, Hall JE, da Silva AA. Central Nervous System Actions of Leptin Improve Cardiac Function After Ischemia–Reperfusion: Roles of Sympathetic Innervation and Sex Differences. J Am Heart Assoc 2022; 11:e027081. [DOI: 10.1161/jaha.122.027081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background
Therapeutic strategies for preventing paradoxical reperfusion injury after myocardial ischemia are limited. We tested whether central nervous system actions of leptin induce important protective effects on cardiac function and metabolism after myocardial ischemia/reperfusion (I/R) injury, the role of cardiac sympathetic innervation in mediating these effects, and whether there are major sex differences in the cardioprotective effects of chronic central nervous system leptin infusion.
Methods and Results
Myocardial I/R was induced by temporary ligation of the left descending coronary artery in male and female Wistar rats instrumented with intracerebroventricular cannula in the lateral ventricle. Vehicle or leptin (0.62 μg/h) infusion was started immediately after reperfusion and continued for 28 days using osmotic minipumps connected to the intracerebroventricular cannula. Cardiac function was assessed by echocardiography, ventricular pressures, and exercise performance. Intracerebroventricular leptin treatment markedly attenuated cardiac dysfunction post‐I/R as evidenced by improved ejection fraction (56.7±1.9 versus 22.6%±1.1%), maximal rate of left ventricle rise (11 680±2122 versus 5022±441 mm Hg) and exercise performance (−4.2±7.9 versus −68.2±3.8 Δ%) compared with vehicle‐treated rats. Intracerebroventricular leptin infusion reduced infarct size in females, but not males, when compared with ad‐lib fed or pair‐fed saline‐treated rats. Intracerebroventricular leptin treatment also increased cardiac NAD
+
/NADH content (≈10‐fold) and improved mitochondrial function when compared with vehicle treatment. Cervical ganglia denervation did not attenuate the cardiac protective effects of leptin after I/R injury.
Conclusions
These data indicate that leptin, via its central nervous system actions, markedly improves overall heart function and mitochondrial metabolism after I/R injury regardless of sex, effects that are largely independent of cardiac sympathetic innervation.
Collapse
Affiliation(s)
- Ana C. M. Omoto
- Department of Physiology and Biophysics Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center Jackson MS
| | - Jussara M. do Carmo
- Department of Physiology and Biophysics Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center Jackson MS
| | - Benjamin Nelson
- Department of Physiology and Biophysics Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center Jackson MS
| | - Nikaela Aitken
- Department of Physiology and Biophysics Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center Jackson MS
| | - Xuemei Dai
- Department of Physiology and Biophysics Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center Jackson MS
| | - Sydney Moak
- Department of Physiology and Biophysics Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center Jackson MS
| | - Elizabeth Flynn
- Department of Physiology and Biophysics Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center Jackson MS
| | - Zhen Wang
- Department of Physiology and Biophysics Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center Jackson MS
| | - Alan J. Mouton
- Department of Physiology and Biophysics Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center Jackson MS
| | - Xuan Li
- Department of Physiology and Biophysics Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center Jackson MS
| | - John E. Hall
- Department of Physiology and Biophysics Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center Jackson MS
| | - Alexandre A. da Silva
- Department of Physiology and Biophysics Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center Jackson MS
| |
Collapse
|
23
|
Friedman MA, Kohn DH. Calcium and phosphorus supplemented diet increases bone volume after thirty days of high speed treadmill exercise in adult mice. Sci Rep 2022; 12:14616. [PMID: 36028525 PMCID: PMC9418142 DOI: 10.1038/s41598-022-19016-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 08/23/2022] [Indexed: 11/22/2022] Open
Abstract
Weight-bearing exercise increases bone mass and strength. Increasing bone loading frequency during exercise can strengthen bone. Combining exercise with a calcium- and phosphorus-supplemented diet increases cortical area more than exercise alone in mice. Thus, we hypothesized that combining high-speed treadmill exercise while feeding mice a mineral-supplemented diet would lead to greater cortical area than high-speed exercise on a standard diet and low-speed exercise on a supplemented diet. Fifteen-week old male C57BL/6 mice were assigned to seven groups—(1) baseline, (2) non-exercise fed a control diet, (3) non-exercise fed a supplemented diet, (4) low-speed exercise fed a control diet, (5) low-speed exercise fed a supplemented diet, (6) high-speed exercise fed a control diet, and (7) high-speed exercise fed a supplemented diet. Mice exercised thirty days for 20 min/day at 12 m/min or 20 m/min. Tibiae were assessed by micro-CT and 4-point bending. Cortical area fraction and trabecular bone volume fraction (BV/TV) were significantly increased by the supplemented diet. High-speed exercised mice had significantly lower body weight, with no detrimental effects to bone health. Increasing running speed can decrease body weight while maintaining the benefits of exercise and nutrition on bone health. Running can lower body weight without harming bone health.
Collapse
Affiliation(s)
- Michael A Friedman
- The University of Michigan, 1011 N University Ave., Ann Arbor, MI, 48109, USA
| | - David H Kohn
- The University of Michigan, 1011 N University Ave., Ann Arbor, MI, 48109, USA.
| |
Collapse
|
24
|
Hastings MH, Herrera JJ, Guseh JS, Atlason B, Houstis NE, Abdul Kadir A, Li H, Sheffield C, Singh AP, Roh JD, Day SM, Rosenzweig A. Animal Models of Exercise From Rodents to Pythons. Circ Res 2022; 130:1994-2014. [PMID: 35679366 PMCID: PMC9202075 DOI: 10.1161/circresaha.122.320247] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Acute and chronic animal models of exercise are commonly used in research. Acute exercise testing is used, often in combination with genetic, pharmacological, or other manipulations, to study the impact of these manipulations on the cardiovascular response to exercise and to detect impairments or improvements in cardiovascular function that may not be evident at rest. Chronic exercise conditioning models are used to study the cardiac phenotypic response to regular exercise training and as a platform for discovery of novel pathways mediating cardiovascular benefits conferred by exercise conditioning that could be exploited therapeutically. The cardiovascular benefits of exercise are well established, and, frequently, molecular manipulations that mimic the pathway changes induced by exercise recapitulate at least some of its benefits. This review discusses approaches for assessing cardiovascular function during an acute exercise challenge in rodents, as well as practical and conceptual considerations in the use of common rodent exercise conditioning models. The case for studying feeding in the Burmese python as a model for exercise-like physiological adaptation is also explored.
Collapse
Affiliation(s)
- Margaret H Hastings
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Jonathan J Herrera
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor (J.J.H.)
| | - J Sawalla Guseh
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Bjarni Atlason
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Nicholas E Houstis
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Azrul Abdul Kadir
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Haobo Li
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Cedric Sheffield
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Anand P Singh
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Jason D Roh
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Sharlene M Day
- Cardiovascular Medicine, Perelman School of Medicine' University of Pennsylvania, Philadelphia (S.M.D.)
| | - Anthony Rosenzweig
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| |
Collapse
|
25
|
Li H, Xia YY, Xia CL, Li Z, Shi Y, Li XB, Zhang JX. Mimicking Metabolic Disturbance in Establishing Animal Models of Heart Failure With Preserved Ejection Fraction. Front Physiol 2022; 13:879214. [PMID: 35592030 PMCID: PMC9110887 DOI: 10.3389/fphys.2022.879214] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 03/30/2022] [Indexed: 01/10/2023] Open
Abstract
Heart failure (HF), the terminal state of different heart diseases, imposed a significant health care burden worldwide. It is the last battlefield in dealing with cardiovascular diseases. HF with preserved ejection fraction (HFpEF) is a type of HF in which the symptoms and signs of HF are mainly ascribed to diastolic dysfunction of left ventricle, whereas systolic function is normal or near-normal. Compared to HF with reduced ejection fraction (HFrEF), the diagnosis and treatment of HFpEF have made limited progress, partly due to the lack of suitable animal models for translational studies in the past. Given metabolic disturbance and inflammatory burden contribute to HFpEF pathogenesis, recent years have witnessed emerging studies focusing on construction of animal models with HFpEF phenotype by mimicking metabolic disorders. These models prefer to recapitulate the metabolic disorders and endothelial dysfunction, leading to the more detailed understanding of the entity. In this review, we summarize the currently available animal models of HFpEF with metabolic disorders, as well as their advantages and disadvantages as tools for translational studies.
Collapse
Affiliation(s)
- Hui Li
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yi-Yuan Xia
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Chun-Lei Xia
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Intensive Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Zheng Li
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yi Shi
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao-Bo Li
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Xiao-Bo Li, ; Jun-Xia Zhang,
| | - Jun-Xia Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Xiao-Bo Li, ; Jun-Xia Zhang,
| |
Collapse
|
26
|
MacDonald TL, Pattamaprapanont P, Cooney EM, Nava RC, Mitri J, Hafida S, Lessard SJ. Canagliflozin Prevents Hyperglycemia-Associated Muscle Extracellular Matrix Accumulation and Improves the Adaptive Response to Aerobic Exercise. Diabetes 2022; 71:881-893. [PMID: 35108373 PMCID: PMC9044131 DOI: 10.2337/db21-0934] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/27/2022] [Indexed: 02/03/2023]
Abstract
Chronic hyperglycemia is associated with low response to aerobic exercise training in rodent models and humans, including reduced aerobic exercise capacity and impaired oxidative remodeling in skeletal muscle. Here, we investigated whether glucose lowering with the sodium-glucose cotransporter 2 inhibitor (SGLT2i), canagliflozin (Cana; 30 mg/kg/day), could restore exercise training response in a model of hyperglycemia (low-dose streptozotocin [STZ]). Cana effectively prevented increased blood glucose in STZ-treated mice. After 6 weeks of voluntary wheel running, Cana-treated mice displayed improvements in aerobic exercise capacity, higher capillary density in striated muscle, and a more oxidative fiber-type in skeletal muscle. In contrast, these responses were blunted or absent in STZ-treated mice. Recent work implicates glucose-induced accumulation of skeletal muscle extracellular matrix (ECM) and hyperactivation of c-Jun N-terminal kinase (JNK)/SMAD2 mechanical signaling as potential mechanisms underlying poor exercise response. In line with this, muscle ECM accretion was prevented by Cana in STZ-treated mice. JNK/SMAD2 signaling with acute exercise was twofold higher in STZ compared with control but was normalized by Cana. In human participants, ECM accumulation was associated with increased JNK signaling, low VO2peak, and impaired metabolic health (oral glucose tolerance test-derived insulin sensitivity). These data demonstrate that hyperglycemia-associated impairments in exercise adaptation can be ameliorated by cotherapy with SGLT2i.
Collapse
Affiliation(s)
- Tara L. MacDonald
- Research Division, Joslin Diabetes Center, Boston, MA
- Harvard Medical School, Boston, MA
| | | | | | - Roberto C. Nava
- Research Division, Joslin Diabetes Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Joanna Mitri
- Research Division, Joslin Diabetes Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Samar Hafida
- Research Division, Joslin Diabetes Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Sarah J. Lessard
- Research Division, Joslin Diabetes Center, Boston, MA
- Harvard Medical School, Boston, MA
- Corresponding author: Sarah J. Lessard,
| |
Collapse
|
27
|
McElroy GS, Chakrabarty RP, D'Alessandro KB, Hu YS, Vasan K, Tan J, Stoolman JS, Weinberg SE, Steinert EM, Reyfman PA, Singer BD, Ladiges WC, Gao L, Lopéz-Barneo J, Ridge K, Budinger GRS, Chandel NS. Reduced expression of mitochondrial complex I subunit Ndufs2 does not impact healthspan in mice. Sci Rep 2022; 12:5196. [PMID: 35338200 PMCID: PMC8956724 DOI: 10.1038/s41598-022-09074-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/16/2022] [Indexed: 01/01/2023] Open
Abstract
Aging in mammals leads to reduction in genes encoding the 45-subunit mitochondrial electron transport chain complex I. It has been hypothesized that normal aging and age-related diseases such as Parkinson’s disease are in part due to modest decrease in expression of mitochondrial complex I subunits. By contrast, diminishing expression of mitochondrial complex I genes in lower organisms increases lifespan. Furthermore, metformin, a putative complex I inhibitor, increases healthspan in mice and humans. In the present study, we investigated whether loss of one allele of Ndufs2, the catalytic subunit of mitochondrial complex I, impacts healthspan and lifespan in mice. Our results indicate that Ndufs2 hemizygous mice (Ndufs2+/−) show no overt impairment in aging-related motor function, learning, tissue histology, organismal metabolism, or sensitivity to metformin in a C57BL6/J background. Despite a significant reduction of Ndufs2 mRNA, the mice do not demonstrate a significant decrease in complex I function. However, there are detectable transcriptomic changes in individual cell types and tissues due to loss of one allele of Ndufs2. Our data indicate that a 50% decline in mRNA of the core mitochondrial complex I subunit Ndufs2 is neither beneficial nor detrimental to healthspan.
Collapse
Affiliation(s)
- Gregory S McElroy
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ram P Chakrabarty
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Karis B D'Alessandro
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yuan-Shih Hu
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Karthik Vasan
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jerica Tan
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Joshua S Stoolman
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Samuel E Weinberg
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Elizabeth M Steinert
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Paul A Reyfman
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Benjamin D Singer
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Warren C Ladiges
- Department of Comparative Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Lin Gao
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - José Lopéz-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Karen Ridge
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - G R Scott Budinger
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Navdeep S Chandel
- Department of Medicine Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA. .,Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
28
|
Janzen NR, Whitfield J, Murray-Segal L, Kemp BE, Hawley JA, Hoffman NJ. Disrupting AMPK-Glycogen Binding in Mice Increases Carbohydrate Utilization and Reduces Exercise Capacity. Front Physiol 2022; 13:859246. [PMID: 35392375 PMCID: PMC8980720 DOI: 10.3389/fphys.2022.859246] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
The AMP-activated protein kinase (AMPK) is a central regulator of cellular energy balance and metabolism and binds glycogen, the primary storage form of glucose in liver and skeletal muscle. The effects of disrupting whole-body AMPK-glycogen interactions on exercise capacity and substrate utilization during exercise in vivo remain unknown. We used male whole-body AMPK double knock-in (DKI) mice with chronic disruption of AMPK-glycogen binding to determine the effects of DKI mutation on exercise capacity, patterns of whole-body substrate utilization, and tissue metabolism during exercise. Maximal treadmill running speed and whole-body energy utilization during submaximal running were determined in wild type (WT) and DKI mice. Liver and skeletal muscle glycogen and skeletal muscle AMPK α and β2 subunit content and signaling were assessed in rested and maximally exercised WT and DKI mice. Despite a reduced maximal running speed and exercise time, DKI mice utilized similar absolute amounts of liver and skeletal muscle glycogen compared to WT. DKI skeletal muscle displayed reduced AMPK α and β2 content versus WT, but intact relative AMPK phosphorylation and downstream signaling at rest and following exercise. During submaximal running, DKI mice displayed an increased respiratory exchange ratio, indicative of greater reliance on carbohydrate-based fuels. In summary, whole-body disruption of AMPK-glycogen interactions reduces maximal running capacity and skeletal muscle AMPK α and β2 content and is associated with increased skeletal muscle glycogen utilization. These findings highlight potential unappreciated roles for AMPK in regulating tissue glycogen dynamics and expand AMPK’s known roles in exercise and metabolism.
Collapse
Affiliation(s)
- Natalie R. Janzen
- Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, Australia
| | - Jamie Whitfield
- Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, Australia
| | - Lisa Murray-Segal
- St Vincent’s Institute of Medical Research and Department of Medicine, University of Melbourne, Fitzroy, VIC, Australia
| | - Bruce E. Kemp
- Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, Australia
- St Vincent’s Institute of Medical Research and Department of Medicine, University of Melbourne, Fitzroy, VIC, Australia
| | - John A. Hawley
- Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, Australia
| | - Nolan J. Hoffman
- Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, Australia
- *Correspondence: Nolan J. Hoffman,
| |
Collapse
|
29
|
Wilson RJ, Lyons SP, Koves TR, Bryson VG, Zhang H, Li T, Crown SB, Ding JD, Grimsrud PA, Rosenberg PB, Muoio DM. Disruption of STIM1-mediated Ca 2+ sensing and energy metabolism in adult skeletal muscle compromises exercise tolerance, proteostasis, and lean mass. Mol Metab 2022; 57:101429. [PMID: 34979330 PMCID: PMC8814391 DOI: 10.1016/j.molmet.2021.101429] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Stromal interaction molecule 1 (STIM1) is a single-pass transmembrane endoplasmic/sarcoplasmic reticulum (E/SR) protein recognized for its role in a store operated Ca2+ entry (SOCE), an ancient and ubiquitous signaling pathway. Whereas STIM1 is known to be indispensable during development, its biological and metabolic functions in mature muscles remain unclear. METHODS Conditional and tamoxifen inducible muscle STIM1 knock-out mouse models were coupled with multi-omics tools and comprehensive physiology to understand the role of STIM1 in regulating SOCE, mitochondrial quality and bioenergetics, and whole-body energy homeostasis. RESULTS This study shows that STIM1 is abundant in adult skeletal muscle, upregulated by exercise, and is present at SR-mitochondria interfaces. Inducible tissue-specific deletion of STIM1 (iSTIM1 KO) in adult muscle led to diminished lean mass, reduced exercise capacity, and perturbed fuel selection in the settings of energetic stress, without affecting whole-body glucose tolerance. Proteomics and phospho-proteomics analyses of iSTIM1 KO muscles revealed molecular signatures of low-grade E/SR stress and broad activation of processes and signaling networks involved in proteostasis. CONCLUSION These results show that STIM1 regulates cellular and mitochondrial Ca2+ dynamics, energy metabolism and proteostasis in adult skeletal muscles. Furthermore, these findings provide insight into the pathophysiology of muscle diseases linked to disturbances in STIM1-dependent Ca2+ handling.
Collapse
Affiliation(s)
- Rebecca J Wilson
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA
| | - Scott P Lyons
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA
| | - Timothy R Koves
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA; Department of Medicine, Division of Geriatrics, Duke University School of Medicine, Durham, NC 27705, USA
| | - Victoria G Bryson
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA; Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, NC 27705, USA
| | - Hengtao Zhang
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA; Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, NC 27705, USA
| | - TianYu Li
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA; Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, NC 27705, USA
| | - Scott B Crown
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA
| | - Jin-Dong Ding
- Department of Medicine, Division of Ophthalmology, Duke University School of Medicine, Durham, NC 27705, USA
| | - Paul A Grimsrud
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University School of Medicine, Durham, NC 27705, USA
| | - Paul B Rosenberg
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA; Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, NC 27705, USA
| | - Deborah M Muoio
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University School of Medicine, Durham, NC 27705, USA; Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27705, USA.
| |
Collapse
|
30
|
The m 6A methyltransferase METTL3 regulates muscle maintenance and growth in mice. Nat Commun 2022; 13:168. [PMID: 35013323 PMCID: PMC8748755 DOI: 10.1038/s41467-021-27848-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 12/14/2021] [Indexed: 01/05/2023] Open
Abstract
Skeletal muscle serves fundamental roles in organismal health. Gene expression fluctuations are critical for muscle homeostasis and the response to environmental insults. Yet, little is known about post-transcriptional mechanisms regulating such fluctuations while impacting muscle proteome. Here we report genome-wide analysis of mRNA methyladenosine (m6A) dynamics of skeletal muscle hypertrophic growth following overload-induced stress. We show that increases in METTL3 (the m6A enzyme), and concomitantly m6A, control skeletal muscle size during hypertrophy; exogenous delivery of METTL3 induces skeletal muscle growth, even without external triggers. We also show that METTL3 represses activin type 2 A receptors (ACVR2A) synthesis, blunting activation of anti-hypertrophic signaling. Notably, myofiber-specific conditional genetic deletion of METTL3 caused spontaneous muscle wasting over time and abrogated overload-induced hypertrophy; a phenotype reverted by co-administration of a myostatin inhibitor. These studies identify a previously unrecognized post-transcriptional mechanism promoting the hypertrophic response of skeletal muscle via control of myostatin signaling. Muscle undergoes hypertrophy and atrophy in response to physiological stimuli or in pathological conditions, which is partially controlled through altered gene expression. Here the authors report that m6A methyltransferase METTL3 and mRNA m6A post-transcriptional modifications as a mechanism that regulates muscle hypertrophy and atrophy via myostatin signalling in mice.
Collapse
|
31
|
Aishwarya R, Abdullah CS, Remex NS, Alam S, Morshed M, Nitu S, Hartman B, King J, Bhuiyan MAN, Orr AW, Kevil CG, Bhuiyan MS. Molecular Characterization of Skeletal Muscle Dysfunction in Sigma 1 Receptor (Sigmar1) Knockout Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:160-177. [PMID: 34710383 PMCID: PMC8759042 DOI: 10.1016/j.ajpath.2021.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/11/2021] [Accepted: 10/04/2021] [Indexed: 01/03/2023]
Abstract
Sigma 1 receptor (Sigmar1) is a widely expressed, multitasking molecular chaperone protein that plays functional roles in several cellular processes. Mutations in the Sigmar1 gene are associated with several distal neuropathies with strong manifestation in skeletal muscle dysfunction with phenotypes like muscle wasting and atrophy. However, the physiological function of Sigmar1 in skeletal muscle remains unknown. Herein, the physiological role of Sigmar1 in skeletal muscle structure and function in gastrocnemius, quadriceps, soleus, extensor digitorum longus, and tibialis anterior muscles was determined. Quantification of myofiber cross-sectional area showed altered myofiber size distribution and changes in myofiber type in the skeletal muscle of the Sigmar1-/- mice. Interestingly, ultrastructural analysis by transmission electron microscopy showed the presence of abnormal mitochondria, and immunostaining showed derangements in dystrophin localization in skeletal muscles from Sigmar1-/- mice. In addition, myopathy in Sigmar1-/- mice was associated with an increased number of central nuclei, increased collagen deposition, and fibrosis. Functional studies also showed reduced endurance and exercise capacity in the Sigmar1-/- mice without any changes in voluntary locomotion, markers for muscle denervation, and muscle atrophy. Overall, this study shows, for the first time, a potential physiological function of Sigmar1 in maintaining healthy skeletal muscle structure and function.
Collapse
Affiliation(s)
- Richa Aishwarya
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Chowdhury S Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Naznin S Remex
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Shafiul Alam
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Mahboob Morshed
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Sadia Nitu
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Brandon Hartman
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Judy King
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | | | - A Wayne Orr
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Christopher G Kevil
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Md Shenuarin Bhuiyan
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana.
| |
Collapse
|
32
|
Peres Valgas da Silva C, Shettigar VK, Baer LA, Abay E, Madaris KL, Mehling MR, Hernandez-Saavedra D, Pinckard KM, Seculov NP, Ziolo MT, Stanford KI. Brown adipose tissue prevents glucose intolerance and cardiac remodeling in high-fat-fed mice after a mild myocardial infarction. Int J Obes (Lond) 2022; 46:350-358. [PMID: 34716427 PMCID: PMC8794788 DOI: 10.1038/s41366-021-00999-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Obesity increases the risk of developing impaired glucose tolerance (IGT) and type 2 diabetes (T2D) after myocardial infarction (MI). Brown adipose tissue (BAT) is important to combat obesity and T2D, and increasing BAT mass by transplantation improves glucose metabolism and cardiac function. The objective of this study was to determine if BAT had a protective effect on glucose tolerance and cardiac function in high-fat diet (HFD) fed mice subjected to a mild MI. METHODS Male C57BL/6 mice were fed a HFD for eight weeks and then divided into Sham (Sham-operated) and +BAT (mice receiving 0.1 g BAT into their visceral cavity). Sixteen weeks post-transplantation, mice were further subdivided into ±MI (Sham; Sham-MI; +BAT; +BAT-MI) and maintained on a HFD. Cardiac (echocardiography) and metabolic function (glucose and insulin tolerance tests, body composition and exercise tolerance) were assessed throughout 22 weeks post-MI. Quantitative PCR (qPCR) was performed to determine the expression of genes related to metabolic function of perigonadal adipose tissue (pgWAT), subcutaneous white adipose tissue (scWAT), liver, heart, tibialis anterior skeletal muscle (TA); and BAT. RESULTS +BAT prevented the increase in left ventricle mass (LVM) and exercise intolerance in response to MI. Similar to what is observed in humans, Sham-MI mice developed IGT post-MI, but this was negated in +BAT-MI mice. IGT was independent of changes in body composition. Genes involved in inflammation, insulin resistance, and metabolism were significantly altered in pgWAT, scWAT, and liver in Sham-MI mice compared to all other groups. CONCLUSIONS BAT transplantation prevents IGT, the increase in LVM, and exercise intolerance following MI. MI alters the expression of several metabolic-related genes in WAT and liver in Sham-MI mice, suggesting that these tissues may contribute to the impaired metabolic response. Increasing BAT may be an important intervention to prevent the development of IGT or T2D and cardiac remodeling in obese patients post-MI.
Collapse
Affiliation(s)
- Carmem Peres Valgas da Silva
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Vikram K. Shettigar
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Lisa A. Baer
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Eaman Abay
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Kendra L. Madaris
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Mikayla R. Mehling
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Diego Hernandez-Saavedra
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Kelsey M. Pinckard
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Nickolai P. Seculov
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Mark T. Ziolo
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH USA
| | - Kristin I. Stanford
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH USA
| |
Collapse
|
33
|
Radhakrishnan J, Baetiong A, Gazmuri RJ. Enhanced Oxygen Utilization Efficiency With Concomitant Activation of AMPK-TBC1D1 Signaling Nexus in Cyclophilin-D Conditional Knockout Mice. Front Physiol 2021; 12:756659. [PMID: 34955879 PMCID: PMC8692870 DOI: 10.3389/fphys.2021.756659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/12/2021] [Indexed: 12/02/2022] Open
Abstract
We have previously reported in HEK 293 T cells and in constitutive cyclophilin-D (Cyp-D) knockout (KO) mice that Cyp-D ablation downregulates oxygen consumption (VO2) and triggers an adaptive response that manifest in higher exercise endurance with less VO2. This adaptive response involves a metabolic switch toward preferential utilization of glucose via AMPK-TBC1D1 signaling nexus. We now investigated whether a similar response could be triggered in mice after acute ablation of Cyp-D using tamoxifen-induced ROSA26-Cre-mediated (i.e., conditional KO, CKO) by subjecting them to treadmill exercise involving five running sessions. At their first treadmill running session, CKO mice and controls had comparable VO2 (208.4 ± 17.9 vs. 209.1 ± 16.8 ml/kg min−1), VCO2 (183.6 ± 17.2 vs. 184.8 ± 16.9 ml/kg min−1), and RER (0.88 ± 0.043 vs. 0.88 ± 0.042). With subsequent sessions, CKO mice displayed more prominent reduction in VO2 (genotype & session interaction p = 0.000) with less prominent reduction in VCO2 resulting in significantly increased RER (genotype and session interaction p = 0.013). The increase in RER was consistent with preferential utilization of glucose as respiratory substrate (4.6 ± 0.8 vs. 4.0 ± 0.9 mg/min, p = 0.003). CKO mice also performed a significantly higher treadmill work for given VO2 expressed as a power/VO2 ratio (7.4 ± 0.2 × 10−3 vs. 6.7 ± 0.2 10−3 ratio, p = 0.025). Analysis of CKO skeletal muscle tissue after completion of five treadmill running sessions showed enhanced AMPK activation (0.669 ± 0.06 vs. 0.409 ± 0.11 pAMPK/β-tubulin ratio, p = 0.005) and TBC1D1 inactivation (0.877 ± 0.16 vs. 0.565 ± 0.09 pTBC1D1/β-tubulin ratio, p < 0.05) accompanied by increased glucose transporter-4 levels consistent with activation of the AMPK-TBC1D1 signaling nexus enabling increased glucose utilization. Taken together, our study demonstrates that like constitutive Cyp-D ablation, acute Cyp-D ablation also induces a state of increased O2 utilization efficiency, paving the way for exploring the use of pharmacological approach to elicit the same response, which could be beneficial under O2 limiting conditions.
Collapse
Affiliation(s)
- Jeejabai Radhakrishnan
- Resuscitation Institute, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States.,Department of Clinical Sciences, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Alvin Baetiong
- Resuscitation Institute, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Raúl J Gazmuri
- Resuscitation Institute, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States.,Department of Clinical Sciences, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States.,Captain James A. Lovell Federal Health Care Center, North Chicago, IL, United States
| |
Collapse
|
34
|
Massett MP, Matejka C, Kim H. Systematic Review and Meta-Analysis of Endurance Exercise Training Protocols for Mice. Front Physiol 2021; 12:782695. [PMID: 34950054 PMCID: PMC8691460 DOI: 10.3389/fphys.2021.782695] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/05/2021] [Indexed: 11/13/2022] Open
Abstract
Inbred and genetically modified mice are frequently used to investigate the molecular mechanisms responsible for the beneficial adaptations to exercise training. However, published paradigms for exercise training in mice are variable, making comparisons across studies for training efficacy difficult. The purpose of this systematic review and meta-analysis was to characterize the diversity across published treadmill-based endurance exercise training protocols for mice and to identify training protocol parameters that moderate the adaptations to endurance exercise training in mice. Published studies were retrieved from PubMed and EMBASE and reviewed for the following inclusion criteria: inbred mice; inclusion of a sedentary group; and exercise training using a motorized treadmill. Fifty-eight articles met those inclusion criteria and also included a "classical" marker of training efficacy. Outcome measures included changes in exercise performance, V ˙ O2max, skeletal muscle oxidative enzyme activity, blood lactate levels, or exercise-induced cardiac hypertrophy. The majority of studies were conducted using male mice. Approximately 48% of studies included all information regarding exercise training protocol parameters. Meta-analysis was performed using 105 distinct training groups (i.e., EX-SED pairs). Exercise training had a significant effect on training outcomes, but with high heterogeneity (Hedges' g=1.70, 95% CI=1.47-1.94, Tau2=1.14, I2 =80.4%, prediction interval=-0.43-3.84). Heterogeneity was partially explained by subgroup differences in treadmill incline, training duration, exercise performance test type, and outcome variable. Subsequent analyses were performed on subsets of studies based on training outcome, exercise performance, or biochemical markers. Exercise training significantly improved performance outcomes (Hedges' g=1.85, 95% CI=1.55-2.15). Subgroup differences were observed for treadmill incline, training duration, and exercise performance test protocol on improvements in performance. Biochemical markers also changed significantly with training (Hedges' g=1.62, 95% CI=1.14-2.11). Subgroup differences were observed for strain, sex, exercise session time, and training duration. These results demonstrate there is a high degree of heterogeneity across exercise training studies in mice. Training duration had the most significant impact on training outcome. However, the magnitude of the effect of exercise training varies based on the marker used to assess training efficacy.
Collapse
Affiliation(s)
- Michael P Massett
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX, United States
| | - Caitlyn Matejka
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX, United States
| | - Hyoseon Kim
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX, United States
| |
Collapse
|
35
|
Fiedler A, Careau V. Individual (Co)variation in Resting and Maximal Metabolic Rates in Wild Mice. Physiol Biochem Zool 2021; 94:338-352. [PMID: 34343458 DOI: 10.1086/716042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
AbstractBasal metabolic rate (BMR) represents the lowest level of aerobic metabolism in a resting, postabsorptive endotherm as measured within the thermoneutral zone. By contrast, maximal metabolic rate ([Formula: see text]max) reflects the upper limit of aerobic metabolism achieved during intensive exercise. As BMR and [Formula: see text]max define the boundaries of the possible levels of aerobic metabolism expressed by a normothermic individual, a key question is whether BMR and [Formula: see text]max are correlated. In the present study, we took repeated paired measurements of thermoneutral resting metabolic rate (RMRt) and [Formula: see text]max on 165 white-footed mice (Peromyscus leucopus). Over a single summer (May-October), repeatability (R ± SE) was low but statistically significant ([Formula: see text]) for both RMRt and [Formula: see text]max ([Formula: see text] for RMRt; [Formula: see text] for [Formula: see text]max). Willingness to run during the forced-exercise trials was also significantly repeatable ([Formula: see text]). At the residual level (within individual), RMRt and [Formula: see text]max tended to be positively correlated ([Formula: see text], [Formula: see text]), suggesting the presence of correlated phenotypic plasticity. By contrast, RMRt and [Formula: see text]max were significantly negatively correlated at the among-individual level ([Formula: see text]). To the extent that variation in RMRt reflects variation in BMR, the negative among-individual correlation does not corroborate the idea that a costly metabolic machinery is needed to support a high [Formula: see text]max. Future research should investigate the (genetic) relationship between RMRt (and BMR) and other energetically expensive behaviors and activities to better understand how energy is allocated within individuals.
Collapse
|
36
|
Vecchiatto B, da Silva RC, Higa TS, Muller CR, Américo ALV, Fortunato-Lima VC, Ferreira MM, Martucci LF, Fonseca-Alaniz MH, Evangelista FS. Oxidative phenotype induced by aerobic physical training prevents the obesity-linked insulin resistance without changes in gastrocnemius muscle ACE2-Angiotensin(1-7)-Mas axis. Diabetol Metab Syndr 2021; 13:74. [PMID: 34229719 PMCID: PMC8262010 DOI: 10.1186/s13098-021-00693-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/23/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND We investigate the effect of aerobic physical training (APT) on muscle morphofunctional markers and Angiotensin Converting Enzyme 2/Angiotensin 1-7/Mas receptor (ACE2/Ang 1-7/Mas) axis in an obesity-linked insulin resistance (IR) animal model induced by cafeteria diet (CAF). METHODS Male C57BL/6J mice were assigned into groups CHOW-SED (chow diet, sedentary; n = 10), CHOW-TR (chow diet, trained; n = 10), CAF-SED (n = 10) and CAF-TR (n = 10). APT consisted in running sessions of 60 min at 60% of maximal speed, 5 days per week for 8 weeks. RESULTS Trained groups had lower body weight and adiposity compared with sedentary groups. CAF-TR improved the glucose and insulin tolerance tests compared with CAF-SED group (AUC = 28.896 ± 1589 vs. 35.200 ± 1076 mg dL-1 120 min-1; kITT = 4.1 ± 0.27 vs. 2.5 ± 0.28% min-1, respectively). CHOW-TR and CAF-TR groups increased exercise tolerance, running intensity at which VO2 max was reached, the expression of p-AMPK, p-ACC and PGC1-α proteins compared with CHOW-SED and CAF-SED. Mithocondrial protein expression of Mfn1, Mfn2 and Drp1 did not change. Lipid deposition reduced in CAF-TR compared with CAF-SED group (3.71 vs. 5.53%/area), but fiber typing, glycogen content, ACE2 activity, Ang 1-7 concentration and Mas receptor expression did not change. CONCLUSIONS The APT prevents obesity-linked IR by modifying the skeletal muscle phenotype to one more oxidative independent of changes in the muscle ACE2/Ang 1-7/Mas axis.
Collapse
Affiliation(s)
- Bruno Vecchiatto
- School of Arts, Science and Humanities, University of Sao Paulo, Av. Arlindo Bettio, 1000, Ermelino Mattarazzo, São Paulo, SP, CEP 03828-000, Brazil
| | - Rafael C da Silva
- School of Arts, Science and Humanities, University of Sao Paulo, Av. Arlindo Bettio, 1000, Ermelino Mattarazzo, São Paulo, SP, CEP 03828-000, Brazil
| | - Talita S Higa
- School of Arts, Science and Humanities, University of Sao Paulo, Av. Arlindo Bettio, 1000, Ermelino Mattarazzo, São Paulo, SP, CEP 03828-000, Brazil
| | - Cynthia R Muller
- Department of Experimental Pathophysiology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Anna Laura V Américo
- Department of Experimental Pathophysiology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Vanessa C Fortunato-Lima
- School of Arts, Science and Humanities, University of Sao Paulo, Av. Arlindo Bettio, 1000, Ermelino Mattarazzo, São Paulo, SP, CEP 03828-000, Brazil
| | - Marília M Ferreira
- School of Arts, Science and Humanities, University of Sao Paulo, Av. Arlindo Bettio, 1000, Ermelino Mattarazzo, São Paulo, SP, CEP 03828-000, Brazil
| | - Luiz Felipe Martucci
- Department of Experimental Pathophysiology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | | | - Fabiana S Evangelista
- School of Arts, Science and Humanities, University of Sao Paulo, Av. Arlindo Bettio, 1000, Ermelino Mattarazzo, São Paulo, SP, CEP 03828-000, Brazil.
| |
Collapse
|
37
|
Gomes-Santos IL, Jordão CP, Passos CS, Brum PC, Oliveira EM, Chammas R, Camargo AA, Negrão CE. Exercise Training Preserves Myocardial Strain and Improves Exercise Tolerance in Doxorubicin-Induced Cardiotoxicity. Front Cardiovasc Med 2021; 8:605993. [PMID: 33869297 PMCID: PMC8047409 DOI: 10.3389/fcvm.2021.605993] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 03/01/2021] [Indexed: 12/25/2022] Open
Abstract
Doxorubicin causes cardiotoxicity and exercise intolerance. Pre-conditioning exercise training seems to prevent doxorubicin-induced cardiac damage. However, the effectiveness of the cardioprotective effects of exercise training concomitantly with doxorubicin treatment remains largely unknown. To determine whether low-to-moderate intensity aerobic exercise training during doxorubicin treatment would prevent cardiotoxicity and exercise intolerance, we performed exercise training concomitantly with chronic doxorubicin treatment in mice. Ventricular structure and function were accessed by echocardiography, exercise tolerance by maximal exercise test, and cardiac biology by histological and molecular techniques. Doxorubicin-induced cardiotoxicity, evidenced by impaired ventricular function, cardiac atrophy, and fibrosis. Exercise training did not preserve left ventricular ejection fraction or reduced fibrosis. However, exercise training preserved myocardial circumferential strain alleviated cardiac atrophy and restored cardiomyocyte cross-sectional area. On the other hand, exercise training exacerbated doxorubicin-induced body wasting without affecting survival. Finally, exercise training blunted doxorubicin-induced exercise intolerance. Exercise training performed during doxorubicin-based chemotherapy can be a valuable approach to attenuate cardiotoxicity.
Collapse
Affiliation(s)
- Igor L Gomes-Santos
- Faculdade de Medicina, Heart Institute (InCor), Hospital das Clínicas, Universidade de São Paulo, São Paulo, Brazil
| | - Camila P Jordão
- Faculdade de Medicina, Heart Institute (InCor), Hospital das Clínicas, Universidade de São Paulo, São Paulo, Brazil
| | - Clevia S Passos
- Faculdade de Medicina, Heart Institute (InCor), Hospital das Clínicas, Universidade de São Paulo, São Paulo, Brazil
| | - Patricia C Brum
- School of Physical Education and Sport, Universidade de São Paulo, São Paulo, Brazil
| | - Edilamar M Oliveira
- School of Physical Education and Sport, Universidade de São Paulo, São Paulo, Brazil
| | - Roger Chammas
- Faculdade de Medicina, Cancer Institute of the State of São Paulo (ICESP), Hospital das Clínicas, Universidade de São Paulo, São Paulo, Brazil
| | - Anamaria A Camargo
- Centro de Oncologia Molecular, Hospital Sírio-Libanês, São Paulo, Brazil
| | - Carlos E Negrão
- Faculdade de Medicina, Heart Institute (InCor), Hospital das Clínicas, Universidade de São Paulo, São Paulo, Brazil.,School of Physical Education and Sport, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
38
|
Adamovich Y, Ezagouri S, Dandavate V, Asher G. Monitoring daytime differences in moderate intensity exercise capacity using treadmill test and muscle dissection. STAR Protoc 2021; 2:100331. [PMID: 33598660 PMCID: PMC7868630 DOI: 10.1016/j.xpro.2021.100331] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
There is growing interest in medicine and sports in uncovering exercise modifiers that enhance or limit exercise capacity. Here, we detail a protocol for testing the daytime effect on running capacity in mice using a moderate intensity treadmill effort test. Instructions for dissecting soleus, gastrocnemius plantaris, and quadriceps muscles for further analysis are provided as well. This experimental setup is optimized for addressing questions regarding the involvement of daytime and circadian clocks in regulating exercise capacity. For complete details on the use and execution of this protocol, please refer to Ezagouri et al. (2019). Exercise capacity is influenced by the time of day Protocol for determining moderate intensity exercise capacity using treadmill test Instructions for muscle dissection
Collapse
Affiliation(s)
- Yaarit Adamovich
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Saar Ezagouri
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Vaishnavi Dandavate
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Gad Asher
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| |
Collapse
|
39
|
Soori R, Ghram A, Zare Shahneh M, Choobineh S, Costa PB, Voltarelli FA. Effects of high intensity interval training and aging on cardiac muscle apoptosis markers in C57BL/6 Mice. SPORT SCIENCES FOR HEALTH 2021. [DOI: 10.1007/s11332-020-00670-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
40
|
Petrosino JM, Longenecker JZ, Ramkumar S, Xu X, Dorn LE, Bratasz A, Yu L, Maurya S, Tolstikov V, Bussberg V, Janssen PM, Periasamy M, Kiebish MA, Duester G, von Lintig J, Ziouzenkova O, Accornero F. Paracardial fat remodeling affects systemic metabolism through alcohol dehydrogenase 1. J Clin Invest 2021; 131:141799. [PMID: 33586683 PMCID: PMC7880313 DOI: 10.1172/jci141799] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/10/2020] [Indexed: 11/17/2022] Open
Abstract
The relationship between adiposity and metabolic health is well established. However, very little is known about the fat depot, known as paracardial fat (pCF), located superior to and surrounding the heart. Here, we show that pCF remodels with aging and a high-fat diet and that the size and function of this depot are controlled by alcohol dehydrogenase 1 (ADH1), an enzyme that oxidizes retinol into retinaldehyde. Elderly individuals and individuals with obesity have low ADH1 expression in pCF, and in mice, genetic ablation of Adh1 is sufficient to drive pCF accumulation, dysfunction, and global impairments in metabolic flexibility. Metabolomics analysis revealed that pCF controlled the levels of circulating metabolites affecting fatty acid biosynthesis. Also, surgical removal of the pCF depot was sufficient to rescue the impairments in cardiometabolic flexibility and fitness observed in Adh1-deficient mice. Furthermore, treatment with retinaldehyde prevented pCF remodeling in these animals. Mechanistically, we found that the ADH1/retinaldehyde pathway works by driving PGC-1α nuclear translocation and promoting mitochondrial fusion and biogenesis in the pCF depot. Together, these data demonstrate that pCF is a critical regulator of cardiometabolic fitness and that retinaldehyde and its generating enzyme ADH1 act as critical regulators of adipocyte remodeling in the pCF depot.
Collapse
Affiliation(s)
- Jennifer M. Petrosino
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Jacob Z. Longenecker
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | | | - Xianyao Xu
- Department of Biomedical Engineering, Dorothy M. Davis Heart and Lung Research Institute
| | - Lisa E. Dorn
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | | | - Lianbo Yu
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio, USA
| | - Santosh Maurya
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | | | - Valerie Bussberg
- BERG, Precision Medicine Department, Framingham, Massachusetts, USA
| | - Paul M.L. Janssen
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Muthu Periasamy
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
- Department of Internal Medicine, University of Central Florida, Orlando, Florida, USA
| | | | - Gregg Duester
- Development, Aging, and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Johannes von Lintig
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Ouliana Ziouzenkova
- Department of Human Sciences, College of Education and Human Ecology, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Federica Accornero
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
41
|
Tai YK, Ng C, Purnamawati K, Yap JLY, Yin JN, Wong C, Patel BK, Soong PL, Pelczar P, Fröhlich J, Beyer C, Fong CHH, Ramanan S, Casarosa M, Cerrato CP, Foo ZL, Pannir Selvan RM, Grishina E, Degirmenci U, Toh SJ, Richards PJ, Mirsaidi A, Wuertz‐Kozak K, Chong SY, Ferguson SJ, Aguzzi A, Monici M, Sun L, Drum CL, Wang J, Franco‐Obregón A. Magnetic fields modulate metabolism and gut microbiome in correlation with
Pgc‐1α
expression: Follow‐up to an in vitro magnetic mitohormetic study. FASEB J 2020; 34:11143-11167. [DOI: 10.1096/fj.201903005rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 06/07/2020] [Accepted: 06/15/2020] [Indexed: 01/07/2023]
Affiliation(s)
- Yee Kit Tai
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Charmaine Ng
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
| | - Kristy Purnamawati
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Jasmine Lye Yee Yap
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Jocelyn Naixin Yin
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Craig Wong
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Bharati Kadamb Patel
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
| | - Poh Loong Soong
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Pawel Pelczar
- Centre for Transgenic Models University of Basel Basel Switzerland
- Institute of Laboratory Animal Science University of Zürich Zürich Switzerland
| | | | - Christian Beyer
- Centre Suisse d'électronique et de microtechnique, CSEM SA Neuchatel Switzerland
| | - Charlene Hui Hua Fong
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Sharanya Ramanan
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Marco Casarosa
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio” University of Florence Florence Italy
- Institute for Biomechanics ETH Zürich Zürich Switzerland
| | | | - Zi Ling Foo
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Rina Malathi Pannir Selvan
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Elina Grishina
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Ufuk Degirmenci
- Institute of Molecular and Cell Biology, A*STAR Singapore Singapore
| | - Shi Jie Toh
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Pete J. Richards
- Competence Center for Applied Biotechnology and Molecular Medicine University of Zürich Zürich Switzerland
| | - Ali Mirsaidi
- Competence Center for Applied Biotechnology and Molecular Medicine University of Zürich Zürich Switzerland
| | - Karin Wuertz‐Kozak
- Competence Center for Applied Biotechnology and Molecular Medicine University of Zürich Zürich Switzerland
- Department of Biomedical Engineering Rochester Institute of Technology (RIT) Rochester NY USA
- Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS) Singapore Singapore
| | - Suet Yen Chong
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS) Singapore Singapore
| | - Stephen J. Ferguson
- Institute of Molecular and Cell Biology, A*STAR Singapore Singapore
- Competence Center for Applied Biotechnology and Molecular Medicine University of Zürich Zürich Switzerland
| | - Adriano Aguzzi
- Institut für Neuropathologie Universitätsspital Zürich Zürich Switzerland
| | - Monica Monici
- ASAcampus JL, ASA Res. Div. ‐ Dept. of Experimental and Clinical Biomedical Sciences “Mario Serio” University of Florence Florence Italy
| | - Lei Sun
- DUKE‐NUS Graduate Medical School Singapore Singapore Singapore
| | - Chester L. Drum
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS) Singapore Singapore
| | - Jiong‐Wei Wang
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS) Singapore Singapore
- Department of Physiology Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
| | - Alfredo Franco‐Obregón
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
- Institute of Molecular and Cell Biology, A*STAR Singapore Singapore
- Department of Physiology Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Institute for Health Innovation & Technology, iHealthtech National University of Singapore Singapore Singapore
| |
Collapse
|
42
|
Gálvez I, Martín-Cordero L, Hinchado MD, Ortega E. β2 Adrenergic Regulation of the Phagocytic and Microbicide Capacity of Circulating Monocytes: Influence of Obesity and Exercise. Nutrients 2020; 12:nu12051438. [PMID: 32429330 PMCID: PMC7284544 DOI: 10.3390/nu12051438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/08/2020] [Accepted: 05/13/2020] [Indexed: 01/14/2023] Open
Abstract
Obese individuals present anomalous immune/inflammatory responses with dysregulations in neuroendocrine responses and immune/stress feedback mechanisms. In this context, exercise and β2 adrenergic activation present monocyte-mediated anti-inflammatory effects that are modulated by obesity. However, these anti-inflammatory effects could immunocompromise the monocyte-mediated innate response against a pathogen challenge. Thus, the objective of this work was to evaluate the effect of obesity, and exercise in this condition, on the β2 adrenergic regulation of the phagocytic and microbicide capacity of circulating monocytes. C57BL/6J mice were allocated to different sedentary or exercised, lean or obese groups. Obese mice showed a lower monocyte-mediated innate response than that of lean mice. Globally, selective β2 adrenergic receptor agonist terbutaline decreased the innate response of monocytes from lean and obese sedentary animals, whereas exercise stimulated it. Exercise modulates β2 adrenergic regulation of the innate response in lean and obese animals, with a global stimulatory or neutral effect, thus abolishing the inhibitory effect of terbutaline occurring in sedentary animals. These effects cannot be explained only by changes in the surface expression of toll-like receptors. Therefore, in general, terbutaline does not hinder the effects of regular exercise, but regular exercise does abolish the effects of terbutaline in sedentary individuals.
Collapse
Affiliation(s)
- Isabel Gálvez
- Grupo de Investigación en Inmunofisiología, Departamento de Enfermería, Facultad de Medicina, Universidad de Extremadura, 06071 Badajoz, Spain;
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz, Spain; (L.M.-C.); (M.D.H.)
| | - Leticia Martín-Cordero
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz, Spain; (L.M.-C.); (M.D.H.)
- Grupo de Investigación en Inmunofisiología, Departamento de Enfermería, Centro Universitario de Plasencia, Universidad de Extremadura, 10600 Plasencia, Spain
| | - María Dolores Hinchado
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz, Spain; (L.M.-C.); (M.D.H.)
- Grupo de Investigación en Inmunofisiología, Departamento de Fisiología, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain
| | - Eduardo Ortega
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz, Spain; (L.M.-C.); (M.D.H.)
- Grupo de Investigación en Inmunofisiología, Departamento de Fisiología, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain
- Correspondence: ; Tel.: +34-924-289-300
| |
Collapse
|
43
|
Poole DC, Copp SW, Colburn TD, Craig JC, Allen DL, Sturek M, O'Leary DS, Zucker IH, Musch TI. Guidelines for animal exercise and training protocols for cardiovascular studies. Am J Physiol Heart Circ Physiol 2020; 318:H1100-H1138. [PMID: 32196357 DOI: 10.1152/ajpheart.00697.2019] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Whole body exercise tolerance is the consummate example of integrative physiological function among the metabolic, neuromuscular, cardiovascular, and respiratory systems. Depending on the animal selected, the energetic demands and flux through the oxygen transport system can increase two orders of magnitude from rest to maximal exercise. Thus, animal models in health and disease present the scientist with flexible, powerful, and, in some instances, purpose-built tools to explore the mechanistic bases for physiological function and help unveil the causes for pathological or age-related exercise intolerance. Elegant experimental designs and analyses of kinetic parameters and steady-state responses permit acute and chronic exercise paradigms to identify therapeutic targets for drug development in disease and also present the opportunity to test the efficacy of pharmacological and behavioral countermeasures during aging, for example. However, for this promise to be fully realized, the correct or optimal animal model must be selected in conjunction with reproducible tests of physiological function (e.g., exercise capacity and maximal oxygen uptake) that can be compared equitably across laboratories, clinics, and other proving grounds. Rigorously controlled animal exercise and training studies constitute the foundation of translational research. This review presents the most commonly selected animal models with guidelines for their use and obtaining reproducible results and, crucially, translates state-of-the-art techniques and procedures developed on humans to those animal models.
Collapse
Affiliation(s)
- David C Poole
- Department of Kinesiology, Kansas State University, Manhattan, Kansas.,Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas
| | - Steven W Copp
- Department of Kinesiology, Kansas State University, Manhattan, Kansas
| | - Trenton D Colburn
- Department of Kinesiology, Kansas State University, Manhattan, Kansas
| | - Jesse C Craig
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah.,Geriatric Research, Education, and Clinical Center, Veterans Affairs Medical Center, Salt Lake City, Utah
| | - David L Allen
- Department of Psychology and Neuroscience, University of Colorado, Boulder, Colorado
| | - Michael Sturek
- Department of Anatomy, Cell Biology and Physiology, Indiana University, Indianapolis, Indiana
| | - Donal S O'Leary
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Irving H Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Timothy I Musch
- Department of Kinesiology, Kansas State University, Manhattan, Kansas.,Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas
| |
Collapse
|
44
|
Speck AE, Aguiar AS. Letter to the Editor: Mechanisms of sex differences in exercise capacity. Am J Physiol Regul Integr Comp Physiol 2020; 318:R156-R157. [PMID: 31888353 DOI: 10.1152/ajpregu.00187.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Ana Elisa Speck
- Lab Biology of Exercise, Department of Health Sciences, ARA-Centro Araranguá, UFSC-Federal University of Santa Catarina, Araranguá, SC, Brazil
| | - Aderbal S Aguiar
- Lab Biology of Exercise, Department of Health Sciences, ARA-Centro Araranguá, UFSC-Federal University of Santa Catarina, Araranguá, SC, Brazil
| |
Collapse
|
45
|
Caru M, Lalonde F, Legault E, Curnier D, St-Pierre DH, Comtois AS, Tournoux F. Ethical consideration and feasibility demonstration of high-intensity interval training without the use of electrical shocks in mice with and without doxorubicin exposition. Am J Cancer Res 2019; 9:2813-2820. [PMID: 31911864 PMCID: PMC6943355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 09/30/2019] [Indexed: 06/10/2023] Open
Abstract
INTRODUCTION Most protocols intended to stimulate cardiovascular training in mice use electrical shocks that cause psychological stress and interfere with running performance. The aim of this study was to: 1) demonstrate the feasibility of a two-week high-intensity interval training (HIIT) program without the use of electric shocks in mice and 2) show that HIIT without electric shocks is feasible in the specific context of mice exposed to chemotherapy (i.e., doxorubicin). METHODS Ten C57bl/6 6-week-old female mice underwent a maximal exercise capacity test before and after two weeks of HIIT (five sessions per week) to measure their maximum running speed. The electrical stimulus was substituted by gently lifting the hind legs of the training mice using a tongue depressor. A second sample of ten C57bl/6 10-week-old female mice receiving a single intravenous injection of 20 mg/kg of doxorubicin underwent a single session of HIIT post-DOX using the same gentle stimulation method. RESULTS After two weeks of HIIT without the use of electric shocks, non-treated mice had a significant increase in their maximal speed (4.4 m•min-1; P = 0.019). In DOX-treated mice, the compliance rate to run went from 100% during the acclimation period prior to doxorubicin treatment to 100% when HIIT was performed after the DOX treatment. Doxorubicin treatment seemed to affect exercise compliance in DOX-treated mice. Our study demonstrated that a two-week HIIT program in non-treated mice and a single HIIT session in DOX-treated mice are feasible. CONCLUSION The use of electric shocks was not required to obtain acceptable exercise compliance and a significant change in mice physical capacity. Our technique to perform a treadmill maximal exercise capacity test was shown to be feasible, even in specific pathological conditions like chemotherapy infusion, and could become a reference for future research protocols aimed at reducing the impact of psychological stress caused by electric shocks in mice. This model of exercise training in mice introduces an alternative to ethical conduct standards in animal research.
Collapse
Affiliation(s)
- Maxime Caru
- Laboratory of Pathophysiology of EXercise (LPEX), School of Kinesiology and Physical Activity Sciences, Faculty of Medicine, University of MontrealMontreal, Quebec, Canada
- Laboratoire EA 4430 - Clinique Psychanalyse Developpement (CliPsyD), Department of Psychology, University of Paris NanterreNanterre, Ile-de-France, France
- Sainte-Justine University Health Center, Research CenterMontreal, Canada
| | - François Lalonde
- Laboratory of Pathophysiology of EXercise (LPEX), School of Kinesiology and Physical Activity Sciences, Faculty of Medicine, University of MontrealMontreal, Quebec, Canada
- University Hospital of Montreal, Research CenterMontreal, Canada
- Department of Exercise Sciences, Faculty of Sciences, Université du Québec à MontréalMontréal, Canada
| | - Elise Legault
- University Hospital of Montreal, Research CenterMontreal, Canada
- Department of Exercise Sciences, Faculty of Sciences, Université du Québec à MontréalMontréal, Canada
| | - Daniel Curnier
- Laboratory of Pathophysiology of EXercise (LPEX), School of Kinesiology and Physical Activity Sciences, Faculty of Medicine, University of MontrealMontreal, Quebec, Canada
- Sainte-Justine University Health Center, Research CenterMontreal, Canada
- University Hospital of Montreal, Research CenterMontreal, Canada
| | - David H St-Pierre
- Sainte-Justine University Health Center, Research CenterMontreal, Canada
- Department of Exercise Sciences, Faculty of Sciences, Université du Québec à MontréalMontréal, Canada
| | - Alain Steve Comtois
- Department of Exercise Sciences, Faculty of Sciences, Université du Québec à MontréalMontréal, Canada
| | | |
Collapse
|
46
|
Martín-Cordero L, Gálvez I, Hinchado MD, Ortega E. β2 Adrenergic Regulation of the Phagocytic and Microbicide Capacity of Macrophages from Obese and Lean Mice: Effects of Exercise. Nutrients 2019; 11:nu11112721. [PMID: 31717554 PMCID: PMC6893822 DOI: 10.3390/nu11112721] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/31/2019] [Accepted: 11/07/2019] [Indexed: 12/17/2022] Open
Abstract
Macrophages are crucial in the inflammation associated with obesity. Exercise is the main non-pharmacological strategy against obesity, not only for improving metabolic impairment, but also because of its anti-inflammatory effects, particularly those mediated by β2 adrenergic receptors (β2-AR). Nevertheless, these anti-inflammatory effects could immunocompromise the innate response against pathogen challenge. Thus, the objective of this work was to evaluate the effect of obesity, and of exercise in this condition, on the β2 adrenergic regulation of the innate function of macrophages. High fat diet-induced obese C57BL/6J mice were used to evaluate the effects of acute and regular exercise on the phagocytic and microbicide capacities of peritoneal macrophages. Selective β2-AR agonist terbutaline (1 µM) decreased the phagocytic and microbicide activities of macrophages from control lean and obese sedentary animals. While acute exercise did not modify the inhibitory capacity of terbutaline, regular exercise abolished this inhibitory effect. These effects cannot be explained only by changes in the surface expression of β2-AR. In conclusion, (1) obesity does not alter the β2-AR-mediated decrease of the innate response of macrophages and (2) regular exercise can revert the inhibitory effect of terbutaline on the phagocytic activity of macrophages, although obesity seems to hinder this immunophysiological adaptation.
Collapse
Affiliation(s)
- Leticia Martín-Cordero
- Grupo de Investigación en Inmunofisiología, Departamento de Enfermería, Centro Universitario de Plasencia, Universidad de Extremadura, 10600 Plasencia, Spain;
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz, Spain; (I.G.); (M.D.H.)
| | - Isabel Gálvez
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz, Spain; (I.G.); (M.D.H.)
- Grupo de Investigación en Inmunofisiología, Departamento de Fisiología, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain
| | - María Dolores Hinchado
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz, Spain; (I.G.); (M.D.H.)
- Grupo de Investigación en Inmunofisiología, Departamento de Fisiología, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain
| | - Eduardo Ortega
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz, Spain; (I.G.); (M.D.H.)
- Grupo de Investigación en Inmunofisiología, Departamento de Fisiología, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain
- Correspondence: ; Tel.: +34-924-289-300
| |
Collapse
|
47
|
Gálvez I, Martín-Cordero L, Hinchado MD, Álvarez-Barrientos A, Ortega E. Obesity Affects β2 Adrenergic Regulation of the Inflammatory Profile and Phenotype of Circulating Monocytes from Exercised Animals. Nutrients 2019; 11:nu11112630. [PMID: 31684076 PMCID: PMC6893831 DOI: 10.3390/nu11112630] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 12/20/2022] Open
Abstract
Anomalous immune/inflammatory responses in obesity take place along with alterations in the neuroendocrine responses and dysregulation in the immune/stress feedback mechanisms. Exercise is a potential anti-inflammatory strategy in this context, but the influence of exercise on the β2 adrenergic regulation of the monocyte-mediated inflammatory response in obesity remains completely unknown. The first objective of this study was to analyze the effect of exercise on the inflammatory profile and phenotype of monocytes from obese and lean animals, and the second aim was to determine whether obesity could affect monocytes' inflammatory response to β2 adrenergic activation in exercised animals. C57BL/6J mice were allocated to different lean or obese groups: sedentary, with acute exercise, or with regular exercise. The inflammatory profile and phenotype of their circulating monocytes were evaluated by flow cytometry in the presence or absence of the selective β2 adrenergic receptor agonist terbutaline. Exercise caused an anti-inflammatory effect in obese individuals and a pro-inflammatory effect in lean individuals. β2 adrenergic receptor stimulation exerted a global pro-inflammatory effect in monocytes from exercised obese animals and an anti-inflammatory effect in monocytes from exercised lean animals. Thus, β2 adrenergic regulation of inflammation in monocytes from exercised animals seems to depend on the inflammatory basal set-point.
Collapse
Affiliation(s)
- Isabel Gálvez
- Grupo de Investigación en Inmunofisiología, Departamento de Fisiología, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain.
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz, Spain.
| | - Leticia Martín-Cordero
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz, Spain.
- Grupo de Investigación en Inmunofisiología, Departamento de Enfermería, Centro Universitario de Plasencia, Universidad de Extremadura, 10600 Plasencia, Spain.
| | - María Dolores Hinchado
- Grupo de Investigación en Inmunofisiología, Departamento de Fisiología, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain.
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz, Spain.
| | - Alberto Álvarez-Barrientos
- Servicio de Técnicas Aplicadas a la Biociencia (STAB), Universidad de Extremadura, 06071 Badajoz, Spain.
| | - Eduardo Ortega
- Grupo de Investigación en Inmunofisiología, Departamento de Fisiología, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain.
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz, Spain.
| |
Collapse
|
48
|
Radhakrishnan J, Baetiong A, Kaufman H, Huynh M, Leschinsky A, Fresquez A, White C, DiMario JX, Gazmuri RJ. Improved exercise capacity in cyclophilin-D knockout mice associated with enhanced oxygen utilization efficiency and augmented glucose uptake via AMPK-TBC1D1 signaling nexus. FASEB J 2019; 33:11443-11457. [PMID: 31339770 DOI: 10.1096/fj.201802238r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We previously reported in HEK 293T cells that silencing the mitochondrial peptidyl prolyl isomerase cyclophilin-D (Cyp-D) reduces Vo2. We now report that in vivo Cyp-D ablation using constitutive Cyp-D knockout (KO) mice also reduces Vo2 both at rest (∼15%) and during treadmill exercise (∼12%). Yet, despite Vo2 reduction, these Cyp-D KO mice ran longer (1071 ± 77 vs. 785 ± 79 m; P = 0.002), for longer time (43 ± 3 vs. 34 ± 3 min; P = 0.004), and at higher speed (34 ± 1 vs. 29 ± 1 m/s; P ≤ 0.001), resulting in increased work (87 ± 6 vs. 58 ± 6 J; P ≤ 0.001). There were parallel reductions in carbon dioxide production, but of lesser magnitude, yielding a 2.3% increase in the respiratory exchange ratio consistent with increased glucose utilization as respiratory substrate. In addition, primary skeletal muscle cells of Cyp-D KO mice subjected to electrical stimulation exhibited higher glucose uptake (4.4 ± 0.55 vs. 2.6 ± 0.04 pmol/mg/min; P ≤ 0.001) with enhanced AMPK activation (0.58 ± 0.06 vs. 0.38 ± 0.03 pAMPK/β-tubulin ratio; P ≤ 0.01) and TBC1 (Tre-2/USP6, BUB2, Cdc16) domain family, member 1 (TBC1D1) inactivation. Likewise, pharmacological activation of AMPK also increased glucose uptake (3.2 ± 0.3 vs. 2.3 ± 0.2 pmol/mg/min; P ≤ 0.001). Moreover, lactate and ATP levels were increased in these cells. Taken together, Cyp-D ablation triggered an adaptive response resulting in increased exercise capacity despite less oxygen utilization associated with increased glucose uptake and utilization involving AMPK-TBC1D1 signaling nexus.-Radhakrishnan, J., Baetiong, A., Kaufman, H., Huynh, M., Leschinsky, A., Fresquez, A., White, C., DiMario, J. X., Gazmuri, R. J. Improved exercise capacity in cyclophilin-D knockout mice associated with enhanced oxygen utilization efficiency and augmented glucose uptake via AMPK-TBC1D1 signaling nexus.
Collapse
Affiliation(s)
- Jeejabai Radhakrishnan
- Resuscitation Institute, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
- Department of Clinical Sciences, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Alvin Baetiong
- Resuscitation Institute, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Harrison Kaufman
- Resuscitation Institute, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Michelle Huynh
- Resuscitation Institute, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Angela Leschinsky
- Resuscitation Institute, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Adriana Fresquez
- Discipline of Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
- Center for Cancer Cell Biology, Immunology, and Infection, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Carl White
- Discipline of Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
- Center for Cancer Cell Biology, Immunology, and Infection, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Joseph X DiMario
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
- Department of Biomedical Research, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Raúl J Gazmuri
- Resuscitation Institute, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
- Department of Clinical Sciences, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
- Captain James A. Lovell Federal Health Care Center, North Chicago, Illinois, USA
| |
Collapse
|
49
|
Beyhoff N, Lohr D, Foryst-Ludwig A, Klopfleisch R, Brix S, Grune J, Thiele A, Erfinanda L, Tabuchi A, Kuebler WM, Pieske B, Schreiber LM, Kintscher U. Characterization of Myocardial Microstructure and Function in an Experimental Model of Isolated Subendocardial Damage. Hypertension 2019; 74:295-304. [PMID: 31291149 PMCID: PMC6635061 DOI: 10.1161/hypertensionaha.119.12956] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Subendocardial damage is among the first cardiac manifestations of hypertension and is already present in asymptomatic disease states. Accordingly, markers of subendocardial impairment may facilitate early detection of cardiac damages and risk stratification under these conditions. This study aimed to investigate the impact of subendocardial damage on myocardial microstructure and function to elucidate early pathophysiologic processes and to identify corresponding diagnostic measures. Mice (n=38) were injected with isoproterenol to induce isolated subendocardial scarring or saline as corresponding control. Cardiac function and myocardial deformation were determined by high-frequency echocardiography. The cardiac stress response was assessed in a graded exercise test and during dobutamine stress echocardiography. Myocardial microstructure was studied ex vivo by 7 T diffusion tensor magnetic resonance imaging at a spatial resolution of 100×100×100 µm 3 . Results were correlated with histology and biomarker expression. Subendocardial fibrosis was accompanied by diastolic dysfunction, impaired longitudinal deformation (global peak longitudinal strain [LS]: -12.5±0.5% versus -15.6±0.5%; P<0.001) and elevated biomarker expression (ANP [atrial natriuretic peptide], Galectin-3, and ST2). Systolic function and cardiac stress response remained preserved. Diffusion tensor magnetic resonance imaging revealed a left-shift in helix angle towards lower values in isoproterenol-treated animals, which was mainly determined by subepicardial myofibers (mean helix angle: 2.2±0.8° versus 5.9±1.0°; P<0.01). Longitudinal strain and subepicardial helix angle were highly predictive for subendocardial fibrosis (sensitivity, 82%-92% and specificity, 89%-90%). The results indicate that circumscribed subendocardial damage alone can cause several hallmarks observed in cardiovascular high-risk patients. Microstructural remodeling under these conditions involves also remote regions, and corresponding changes in longitudinal strain and helix angle might serve as diagnostic markers.
Collapse
Affiliation(s)
- Niklas Beyhoff
- From the Charité-Universitätsmedizin Berlin, corporate member of Freie Universitaät Berlin, Humboldt-Universitaät zu Berlin, and Berlin Institute of Health, Institute of Pharmacology, Center for Cardiovascular Research, Berlin, Germany (N.B., A.F.-L., S.B., J.G., Arne Thiele, U.K.).,DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Germany (N.B., A.F.-L., S.B., J.G., Arne Thiele, L.E., Arata Tabuchi, W.M.K., B.P., U.K.)
| | - David Lohr
- Chair of Cellular and Molecular Imaging, Comprehensive Heart Failure Center (CHFC), University Hospital Wuerzburg, Germany (D.L., L.M.S.)
| | - Anna Foryst-Ludwig
- From the Charité-Universitätsmedizin Berlin, corporate member of Freie Universitaät Berlin, Humboldt-Universitaät zu Berlin, and Berlin Institute of Health, Institute of Pharmacology, Center for Cardiovascular Research, Berlin, Germany (N.B., A.F.-L., S.B., J.G., Arne Thiele, U.K.).,DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Germany (N.B., A.F.-L., S.B., J.G., Arne Thiele, L.E., Arata Tabuchi, W.M.K., B.P., U.K.)
| | - Robert Klopfleisch
- Department of Veterinary Pathology, College of Veterinary Medicine, Freie Universität Berlin, Germany (R.K.)
| | - Sarah Brix
- From the Charité-Universitätsmedizin Berlin, corporate member of Freie Universitaät Berlin, Humboldt-Universitaät zu Berlin, and Berlin Institute of Health, Institute of Pharmacology, Center for Cardiovascular Research, Berlin, Germany (N.B., A.F.-L., S.B., J.G., Arne Thiele, U.K.).,DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Germany (N.B., A.F.-L., S.B., J.G., Arne Thiele, L.E., Arata Tabuchi, W.M.K., B.P., U.K.)
| | - Jana Grune
- From the Charité-Universitätsmedizin Berlin, corporate member of Freie Universitaät Berlin, Humboldt-Universitaät zu Berlin, and Berlin Institute of Health, Institute of Pharmacology, Center for Cardiovascular Research, Berlin, Germany (N.B., A.F.-L., S.B., J.G., Arne Thiele, U.K.).,DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Germany (N.B., A.F.-L., S.B., J.G., Arne Thiele, L.E., Arata Tabuchi, W.M.K., B.P., U.K.).,Charité-Universitaätsmedizin Berlin, corporate member of Freie Universitaät Berlin, Humboldt-Universitaät zu Berlin, and Berlin Institute of Health, Institute of Physiology, Berlin, Germany (J.G., L.E., Arata Tabuchi, W.M.K.)
| | - Arne Thiele
- From the Charité-Universitätsmedizin Berlin, corporate member of Freie Universitaät Berlin, Humboldt-Universitaät zu Berlin, and Berlin Institute of Health, Institute of Pharmacology, Center for Cardiovascular Research, Berlin, Germany (N.B., A.F.-L., S.B., J.G., Arne Thiele, U.K.).,DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Germany (N.B., A.F.-L., S.B., J.G., Arne Thiele, L.E., Arata Tabuchi, W.M.K., B.P., U.K.)
| | - Lasti Erfinanda
- DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Germany (N.B., A.F.-L., S.B., J.G., Arne Thiele, L.E., Arata Tabuchi, W.M.K., B.P., U.K.).,Charité-Universitaätsmedizin Berlin, corporate member of Freie Universitaät Berlin, Humboldt-Universitaät zu Berlin, and Berlin Institute of Health, Institute of Physiology, Berlin, Germany (J.G., L.E., Arata Tabuchi, W.M.K.)
| | - Arata Tabuchi
- DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Germany (N.B., A.F.-L., S.B., J.G., Arne Thiele, L.E., Arata Tabuchi, W.M.K., B.P., U.K.).,Charité-Universitaätsmedizin Berlin, corporate member of Freie Universitaät Berlin, Humboldt-Universitaät zu Berlin, and Berlin Institute of Health, Institute of Physiology, Berlin, Germany (J.G., L.E., Arata Tabuchi, W.M.K.)
| | - Wolfgang M Kuebler
- DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Germany (N.B., A.F.-L., S.B., J.G., Arne Thiele, L.E., Arata Tabuchi, W.M.K., B.P., U.K.).,Charité-Universitaätsmedizin Berlin, corporate member of Freie Universitaät Berlin, Humboldt-Universitaät zu Berlin, and Berlin Institute of Health, Institute of Physiology, Berlin, Germany (J.G., L.E., Arata Tabuchi, W.M.K.)
| | - Burkert Pieske
- DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Germany (N.B., A.F.-L., S.B., J.G., Arne Thiele, L.E., Arata Tabuchi, W.M.K., B.P., U.K.).,Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin and Deutsches Herzzentrum Berlin (DHZB), Department of Cardiology, Berlin, Germany (B.P.)
| | - Laura M Schreiber
- Chair of Cellular and Molecular Imaging, Comprehensive Heart Failure Center (CHFC), University Hospital Wuerzburg, Germany (D.L., L.M.S.)
| | - Ulrich Kintscher
- From the Charité-Universitätsmedizin Berlin, corporate member of Freie Universitaät Berlin, Humboldt-Universitaät zu Berlin, and Berlin Institute of Health, Institute of Pharmacology, Center for Cardiovascular Research, Berlin, Germany (N.B., A.F.-L., S.B., J.G., Arne Thiele, U.K.).,DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Germany (N.B., A.F.-L., S.B., J.G., Arne Thiele, L.E., Arata Tabuchi, W.M.K., B.P., U.K.)
| |
Collapse
|
50
|
Son JS, Liu X, Tian Q, Zhao L, Chen Y, Hu Y, Chae SA, de Avila JM, Zhu MJ, Du M. Exercise prevents the adverse effects of maternal obesity on placental vascularization and fetal growth. J Physiol 2019; 597:3333-3347. [PMID: 31115053 DOI: 10.1113/jp277698] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS Maternal exercise improves the metabolic health of maternal mice challenged with a high-fat diet. Exercise intervention of obese mothers prevents fetal overgrowth. Exercise intervention reverses impaired placental vascularization in obese mice. Maternal exercise activates placental AMP-activated protein kinase, which was inhibited as a result of maternal obesity. ABSTRACT More than one-third of pregnant women in the USA are obese and maternal obesity (MO) negatively affects fetal development, which predisposes offspring to metabolic diseases. The placenta mediates nutrient delivery to fetuses and its function is impaired as a result of MO. Exercise ameliorates metabolic dysfunction resulting from obesity, although its effect on placental function of obese mothers has not been explored. In the present study, C57BL/6J female mice were randomly assigned into two groups fed either a control or a high-fat diet (HFD) and then the mice on each diet were further divided into two subgroups with/without exercise. In HFD-induced obese mice, daily treadmill exercise during pregnancy reduced body weight gain, lowered serum glucose and lipid concentration, and improved insulin sensitivity of maternal mice. Importantly, maternal exercise prevented fetal overgrowth (macrosomia) induced by MO. To further examine the preventive effects of exercise on fetal overgrowth, placental vascularization and nutrient transporters were analysed. Vascular density and the expression of vasculogenic factors were reduced as a result of MO but were recovered by maternal exercise. On the other hand, the contents of nutrient transporters were not substantially altered by MO or exercise, suggesting that the protective effects of exercise in MO-induced fetal overgrowth were primarily a result of the alteration of placental vascularization and improved maternal metabolism. Furthermore, exercise enhanced downstream insulin signalling and activated AMP-activated protein kinase in HFD placenta. In sum, maternal exercise prevented fetal overgrowth induced by MO, which was associated with improved maternal metabolism and placental vascularization in obese mothers with exercise.
Collapse
Affiliation(s)
- Jun Seok Son
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Xiangdong Liu
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Qiyu Tian
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Liang Zhao
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Yanting Chen
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Yun Hu
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Song Ah Chae
- Department of Movement Sciences, University of Idaho, Moscow, ID, USA
| | - Jeanene M de Avila
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA, USA
| | - Min Du
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| |
Collapse
|