1
|
Furrer R, Handschin C. Biomarkers of aging: from molecules and surrogates to physiology and function. Physiol Rev 2025; 105:1609-1694. [PMID: 40111763 DOI: 10.1152/physrev.00045.2024] [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: 10/30/2024] [Revised: 01/10/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Many countries face an unprecedented challenge in aging demographics. This has led to an exponential growth in research on aging, which, coupled to a massive financial influx of funding in the private and public sectors, has resulted in seminal insights into the underpinnings of this biological process. However, critical validation in humans has been hampered by the limited translatability of results obtained in model organisms, additionally confined by the need for extremely time-consuming clinical studies in the ostensible absence of robust biomarkers that would allow monitoring in shorter time frames. In the future, molecular parameters might hold great promise in this regard. In contrast, biomarkers centered on function, resilience, and frailty are available at the present time, with proven predictive value for morbidity and mortality. In this review, the current knowledge of molecular and physiological aspects of human aging, potential antiaging strategies, and the basis, evidence, and potential application of physiological biomarkers in human aging are discussed.
Collapse
|
2
|
Hu C. Prevention of cardiovascular disease for healthy aging and longevity: A new scoring system and related "mechanisms-hallmarks-biomarkers". Ageing Res Rev 2025; 107:102727. [PMID: 40096912 DOI: 10.1016/j.arr.2025.102727] [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: 05/09/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025]
Abstract
Healthy "environment-sleep-emotion-exercise-diet" intervention [E(e)SEEDi] lifestyle can improve the quality of life, prolong aging and promote longevity due to improvement of human immunity and prevention of cardiovascular diseases (CVD). Here, the author reviewed the associations between these core elements with CVD and cardiovascular aging, and developed a new scoring system based on the healthy E(e)SEEDi lifestyle for prediction and evaluation of life expectancy. These core factors are assigned 20 points each (120 points in total), and a higher score predicts healthier aging and longevity. The E(e)SEEDi represents "a tree of life" bearing the fruits of longevity as well as "a rocket of anti-ageing" carrying people around the world on a journey of longevity. In conclusion, the E(e)SEEDi can delay aging and increase the life expectancy due to the role of a series of cellular and molecular "mechanisms-hallmarks-biomarkers". It's believed that the novel scoring system has a huge potential and beautiful prospects.
Collapse
Affiliation(s)
- Chunsong Hu
- Department of Cardiovascular Medicine, Nanchang University, Hospital of Nanchang University, Jiangxi Academy of Medical Science, No. 461 Bayi Ave, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
3
|
Leng M, Yang F, Zhao J, Xiong Y, Zhou Y, Zhao M, Jia S, Liu L, Zheng Q, Gan L, Ye J, Zheng M. Mitophagy-mediated S1P facilitates muscle adaptive responses to endurance exercise through SPHK1-S1PR1/S1PR2 in slow-twitch myofibers. Autophagy 2025:1-19. [PMID: 40181214 DOI: 10.1080/15548627.2025.2488563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 01/07/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025] Open
Abstract
Endurance exercise triggers adaptive responses especially in slow-twitch myofibers of skeletal muscles, leading to the remodeling of myofiber structure and the mitochondrial network. However, molecular mechanisms underlying these adaptive responses, with a focus on the fiber type-specific perspective, remains largely unknown. In this study we analyzed the alterations of transcriptomics and metabolomics in distinct skeletal myofibers in response to endurance exercise. We determined that genes associated with sphingolipid metabolism, namely those encoding SPHK1, S1PR1, and S1PR2, are enriched in slow-twitch but not fast-twitch myofibers from both mouse and human skeletal muscles, and found that the SPHK1-S1PR pathway is essential for adaptive responses of slow-twitch to endurance exercise. Importantly, we demonstrate that endurance exercise causes the accumulation of ceramides on stressed mitochondria, and the mitophagic degradation of ceramides results in an increase of the sphingosine-1-phosphate (S1P) level. The elevated S1P thereby facilitates mitochondrial adaptation and enhances endurance capacity via the SPHK1-S1PR1/S1PR2 axis in slow-twitch muscles. Moreover, administration of S1P improves endurance performance in muscle atrophy mice by emulating these adaptive responses. Our findings reveal that the SPHK1-S1P-S1PR1/S1PR2 axis through mitophagic degradation of ceramides in slow-twitch myofibers is the central mediator to endurance exercise and highlight a potential therapeutic target for ameliorating muscle atrophy diseases.Abbreviations CQ: chloroquine; DMD: Duchenne muscular dystrophy; EDL: extensor digitorum longus; FCCP: carbonyl cyanide p-trifluoromethoxyphenyl hydrazone; FUNDC1: FUN14 domain containing 1; GTEx: genotype-tissue expression; MYH: myosin heavy chain; mtDNA: mitochondrial DNA; PPARGC1A/PGC-1α: peroxisome proliferator activated receptor, gamma, coactivator 1 alpha; RG: red gastrocnemius; S1P: sphingosine-1-phosphate; S1PR: sphingosine-1-phosphate receptor; Sol: soleus; SPHK1: sphingosine kinase 1; TA: tibialis anterior; WG: white gastrocnemius.
Collapse
Affiliation(s)
- Minghong Leng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P.R. China
| | - Fenghe Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P.R. China
| | - Junhui Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P.R. China
| | - Yufei Xiong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P.R. China
| | - Yiqing Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P.R. China
| | - Mingyang Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P.R. China
| | - Shi Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P.R. China
| | - Limei Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P.R. China
| | - Qiaoxia Zheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P.R. China
| | - Lebin Gan
- Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University), National Center for Trauma Medicine, Trauma Medicine Center, Peking University People's Hospital, Beijing, P.R. China
| | - Jingjing Ye
- Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University), National Center for Trauma Medicine, Trauma Medicine Center, Peking University People's Hospital, Beijing, P.R. China
| | - Ming Zheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, and State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P.R. China
| |
Collapse
|
4
|
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
|
5
|
Guzzoni V, Emerich de Abreu ICM, Bertagnolli M, Mendes RH, Belló-Klein A, Casarini DE, Flues K, Cândido GO, Paulini J, De Angelis K, Marcondes FK, Irigoyen MC, Sousa Cunha T. Aerobic training increases renal antioxidant defence and reduces angiotensin II levels, mitigating the high mortality in SHR-STZ model. Arch Physiol Biochem 2024; 130:992-1004. [PMID: 39016681 DOI: 10.1080/13813455.2024.2377381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/26/2024] [Indexed: 07/18/2024]
Abstract
OBJECTVE The purpose of the research was to investigate the effects of aerobic training on renal function, oxidative stress, intrarenal renin-angiotensin system, and mortality of hypertensive and diabetic (SHR-STZ) rats. MATERIALS AND METHODS Blood pressure, creatinine, urea levels, urinary glucose, urine volume, and protein excretion were reduced in trained SHR-STZ rats. RESULTS Aerobic training not only attenuated oxidative stress but also elevated the activity of antioxidant enzymes in the kid'ney of SHR-STZ rats. Training increased intrarenal levels of angiotensin-converting enzymes (ACE and ACE2) as well as the neprilysin (NEP) activity, along with decreased intrarenal angiotensin II (Ang II) levels. Aerobic training significantly improved the survival of STZ-SHR rats. CONCLUSION The protective role of aerobic training was associated with improvements in the renal antioxidative capacity, reduced urinary protein excretion along with reduced intrarenal Ang II and increased NEP activity. These findings might reflect a better survival under the combined pathological conditions, hypertension, and diabetes.
Collapse
Affiliation(s)
- Vinicius Guzzoni
- Department of Medicine, School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | | | - Mariane Bertagnolli
- Laboratory of Maternal-child Health, Hospital Sacre-Coeur Research Center, CIUSSS Nord-de-l'Île-de-Montréal, Montreal, Canada
- School of Physical and Occupational Therapy, Faculty of Medicine, McGill University, Montreal, Canada
| | - Roberta Hack Mendes
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Adriane Belló-Klein
- Department of Physiology, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Dulce Elena Casarini
- Department of Medicine, School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Karin Flues
- Laboratory of Experimental Hypertension, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Geórgia Orsi Cândido
- Laboratory of Experimental Hypertension, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Janaína Paulini
- Laboratory of Experimental Hypertension, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Kátia De Angelis
- Department of Physiology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
- Nove de Julho University (UNINOVE), São Paulo, Brazil
| | - Fernanda Klein Marcondes
- Department of Biosciences, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas (FOP - UNICAMP), Piracicaba, Brazil
| | - Maria Cláudia Irigoyen
- Laboratory of Experimental Hypertension, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Tatiana Sousa Cunha
- Department of Science and Technology, Institute of Science and Technology, Federal University of São Paulo (UNIFESP), São José dos Campos, Brazil
| |
Collapse
|
6
|
Kugler BA, Lysaker CR, Franczak E, Hauger BM, Csikos V, Stopperan JA, Allen JA, Stanford JA, Koch LG, Britton SL, Thyfault JP, Wilkins HM. Intrinsic aerobic capacity modulates Alzheimer's disease pathological hallmarks, brain mitochondrial function and proteome during aging. GeroScience 2024; 46:4955-4967. [PMID: 38867031 PMCID: PMC11336007 DOI: 10.1007/s11357-024-01248-3] [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: 04/02/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024] Open
Abstract
Low aerobic capacity is strongly associated with all-cause mortality and risk for Alzheimer's disease (AD). Individuals with early dementia and AD have lower aerobic capacity compared to age-matched controls. The mechanism by which aerobic capacity influences AD risk is unknown but is likely mediated by sexual dimorphism and tissue-level differences in mitochondrial energetics. Here, we used rats selectively bred for large differences in intrinsic aerobic exercise capacity. Brain tissue from 18-month and 24-month-old female and male low-capacity runner (LCR) and high-capacity runner (HCR) rats were analyzed for markers of mitochondrial function and AD-associated pathologies. LCR rats, irrespective of sex, exhibited a greater increase in brain amyloid beta (Aβ42) and tau hyperphosphorylation (pTauthr181/total tau) with aging. In female LCR rats, brain mitochondrial respiration at states 3, 4, and FCCP-induced uncoupling, when stimulated with pyruvate/malate, was reduced at 18 and 24 months, leading to lower ATP-linked mitochondrial respiration compared to mitochondria from HCR rats. Male LCR rats also showed reduced complex II-stimulated mitochondrial respiration (succinate + rotenone) at 24 months compared to HCR rats. Differences in mitochondrial respiration were associated with tau hyperphosphorylation and Aβ42 alterations in both HCR and LCR strains. Proteomic analysis unveiled a distinct difference in the mitochondrial proteome, wherein female LCR rats displayed diminished mitochondrial translation and oxidative phosphorylation (OXPHOS) proteins at 18 months compared to female HCR rats. Conversely, male LCR rats exhibited increased OXPHOS protein abundance but reduced tricarboxylic acid (TCA) cycle proteins compared to male HCR rats. These findings underscore a robust association between intrinsic aerobic exercise capacity, brain mitochondrial function, and AD pathologies during aging.
Collapse
Affiliation(s)
- Benjamin A Kugler
- University of Kansas Medical Center Department of Cell Biology and Physiology and Internal Medicine, Kansas City, KS, USA
| | - Colton R Lysaker
- University of Kansas Alzheimer's Disease Center and Department of Neurology, Kansas City, KS, USA
| | - Edziu Franczak
- University of Kansas Medical Center Department of Cell Biology and Physiology and Internal Medicine, Kansas City, KS, USA
| | - Brittany M Hauger
- University of Kansas Alzheimer's Disease Center and Department of Neurology, Kansas City, KS, USA
| | - Vivien Csikos
- University of Kansas Alzheimer's Disease Center and Department of Neurology, Kansas City, KS, USA
| | - Julia A Stopperan
- University of Kansas Alzheimer's Disease Center and Department of Neurology, Kansas City, KS, USA
| | - Julie A Allen
- University of Kansas Medical Center Department of Cell Biology and Physiology and Internal Medicine, Kansas City, KS, USA
| | - John A Stanford
- University of Kansas Medical Center Department of Cell Biology and Physiology and Internal Medicine, Kansas City, KS, USA
| | - Lauren G Koch
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, USA
| | - Steven L Britton
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - John P Thyfault
- University of Kansas Alzheimer's Disease Center and Department of Neurology, Kansas City, KS, USA
- University of Kansas Medical Center Department of Cell Biology and Physiology and Internal Medicine, Kansas City, KS, USA
- Research Service, Kansas City VA Medical Center Department of Veterans Affairs, University of Kansas Diabetes Center, Kansas City, KS, USA
- University of Kansas Medical Center Department of Molecular Biology and Biochemistry, Kansas City, KS, USA
| | - Heather M Wilkins
- University of Kansas Alzheimer's Disease Center and Department of Neurology, Kansas City, KS, USA.
- University of Kansas Medical Center Department of Molecular Biology and Biochemistry, Kansas City, KS, USA.
- Department of Neurology University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
7
|
Kunimura K, Nakamoto M, Ushijima M. S-1-Propenylcysteine Enhances Endurance Capacity of Mice by Stimulating Fatty Acid Metabolism via Muscle Isoform of Carnitine Acyltransferase-1. J Nutr 2024; 154:2707-2716. [PMID: 39053609 DOI: 10.1016/j.tjnut.2024.07.027] [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: 03/11/2024] [Revised: 05/31/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Endurance is an important capacity to sustain healthy lifestyles. Aged garlic extract (AGE) has been reported to exert an endurance-enhancing effect in clinical and animal studies, although little is known about its active ingredients and mechanism of action. OBJECTIVES This study investigated the potential effect of S-1-propenylcysteine (S1PC), a characteristic sulfur amino acid in AGE, on the swimming endurance of mice, and examined its mechanism of action by a metabolomics-based approach. METHODS Male Institute of Cancer Research (ICR) mice (6 wk old) were orally administered either water (control) or S1PC (6.5 mg/kg/d) for 2 wk. The swimming duration to exhaustion was measured at 24 h after the final administration. Nontargeted metabolomic analysis was conducted on the plasma samples obtained from mice after 40-min submaximal swimming bouts. Subsequently, the enzyme activity of carnitine acyltransferase-1 (CPT-1) and the content of malonyl-coenzyme A (CoA), acetyl-CoA, and adenosine triphosphate (ATP) were quantified in heart, skeletal muscles, and liver of mice. RESULTS The duration time of swimming was substantially increased in the S1PC-treated mice as compared with the control group. Metabolomic analysis revealed significant alterations in the plasma concentration of the metabolites involved in fatty acid metabolism, in particular medium- or long-chain acylcarnitines in the mice treated with S1PC. Moreover, the administration of S1PC significantly enhanced the CPT-1 activity with the concomitant decrease in the malonyl-CoA content in the heart and skeletal muscles. These effects of S1PC were accompanied by the elevation of the acetyl-CoA and ATP levels to enhance the energy production in those tissues. CONCLUSIONS S1PC is a key constituent responsible for the endurance-enhancing effect of AGE. This study suggests that S1PC helps provide energy during endurance exercise by increasing fatty acid metabolism via CPT-1 activation in the heart and skeletal muscles.
Collapse
Affiliation(s)
- Kayo Kunimura
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Hiroshima, Japan.
| | - Masato Nakamoto
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Hiroshima, Japan
| | - Mitsuyasu Ushijima
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Hiroshima, Japan
| |
Collapse
|
8
|
Bubak MP, Davidyan A, O'Reilly CL, Mondal SA, Keast J, Doidge SM, Borowik AK, Taylor ME, Volovičeva E, Kinter MT, Britton SL, Koch LG, Stout MB, Lewis TL, Miller BF. Metformin treatment results in distinctive skeletal muscle mitochondrial remodeling in rats with different intrinsic aerobic capacities. Aging Cell 2024; 23:e14235. [PMID: 38923664 PMCID: PMC11488331 DOI: 10.1111/acel.14235] [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: 03/19/2024] [Revised: 05/07/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
The rationale for the use of metformin as a treatment to slow aging was largely based on data collected from metabolically unhealthy individuals. For healthspan extension metformin will also be used in periods of good health. To understand the potential context specificity of metformin treatment on skeletal muscle, we used a rat model (high-capacity runner/low-capacity runner [HCR/LCR]) with a divide in intrinsic aerobic capacity. Outcomes of metformin treatment differed based on baseline intrinsic mitochondrial function, oxidative capacity of the muscle (gastroc vs soleus), and the mitochondrial population (intermyofibrillar vs. subsarcolemmal). Metformin caused lower ADP-stimulated respiration in LCRs, with less of a change in HCRs. However, a washout of metformin resulted in an unexpected doubling of respiratory capacity in HCRs. These improvements in respiratory capacity were accompanied by mitochondrial remodeling that included increases in protein synthesis and changes in morphology. Our findings raise questions about whether the positive findings of metformin treatment are broadly applicable.
Collapse
Affiliation(s)
- Matthew P. Bubak
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Arik Davidyan
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
- Department of Biological SciencesCalifornia State University SacramentoSacramentoCaliforniaUSA
| | - Colleen L. O'Reilly
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Samim A. Mondal
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Jordan Keast
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Stephen M. Doidge
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Agnieszka K. Borowik
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Michael E. Taylor
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Evelina Volovičeva
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Michael T. Kinter
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Steven L. Britton
- Department of AnesthesiologyUniversity of MichiganAnn ArborMichiganUSA
- Department of Molecular & Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Lauren G. Koch
- Department of Physiology and Pharmacology, College of Medicine and Life SciencesThe University of ToledoToledoOhioUSA
| | - Michael B. Stout
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Tommy L. Lewis
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Benjamin F. Miller
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
- The Oklahoma VA Medical CenterOklahoma CityOklahomaUSA
| |
Collapse
|
9
|
Nakayama Y, Kobayashi S, Masihuddin A, Abdali SA, Seneviratne AMPB, Ishii S, Iida J, Liang Q, Yoshioka J. Systemic Deletion of ARRDC4 Improves Cardiac Reserve and Exercise Capacity in Diabetes. Circ Res 2024; 135:416-433. [PMID: 38946541 PMCID: PMC11257811 DOI: 10.1161/circresaha.123.323158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 06/18/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND Exercise intolerance is an independent predictor of poor prognosis in diabetes. The underlying mechanism of the association between hyperglycemia and exercise intolerance remains undefined. We recently demonstrated that the interaction between ARRDC4 (arrestin domain-containing protein 4) and GLUT1 (glucose transporter 1) regulates cardiac metabolism. METHODS To determine whether this mechanism broadly impacts diabetic complications, we investigated the role of ARRDC4 in the pathogenesis of diabetic cardiac/skeletal myopathy using cellular and animal models. RESULTS High glucose promoted translocation of MondoA into the nucleus, which upregulated Arrdc4 transcriptional expression, increased lysosomal GLUT1 trafficking, and blocked glucose transport in cardiomyocytes, forming a feedback mechanism. This role of ARRDC4 was confirmed in human muscular cells from type 2 diabetic patients. Prolonged hyperglycemia upregulated myocardial Arrdc4 expression in multiple types of mouse models of diabetes. We analyzed hyperglycemia-induced cardiac and skeletal muscle abnormalities in insulin-deficient mice. Hyperglycemia increased advanced glycation end-products and elicited oxidative and endoplasmic reticulum stress leading to apoptosis in the heart and peripheral muscle. Deletion of Arrdc4 augmented tissue glucose transport and mitochondrial respiration, protecting the heart and muscle from tissue damage. Stress hemodynamic analysis and treadmill exhaustion test uncovered that Arrdc4-knockout mice had greater cardiac inotropic/chronotropic reserve with higher exercise endurance than wild-type animals under diabetes. While multiple organs were involved in the mechanism, cardiac-specific overexpression using an adenoassociated virus suggests that high levels of myocardial ARRDC4 have the potential to contribute to exercise intolerance by interfering with cardiac metabolism through its interaction with GLUT1 in diabetes. Importantly, the ARRDC4 mutation mouse line exhibited greater exercise tolerance, showing the potential therapeutic impact on diabetic cardiomyopathy by disrupting the interaction between ARRDC4 and GLUT1. CONCLUSIONS ARRDC4 regulates hyperglycemia-induced toxicities toward cardiac and skeletal muscle, revealing a new molecular framework that connects hyperglycemia to cardiac/skeletal myopathy to exercise intolerance.
Collapse
Affiliation(s)
- Yoshinobu Nakayama
- Department of Molecular, Cellular & Biomedical Sciences, City University of New York School of Medicine, City College of New York, New York, NY
- Department of Anesthesiology and Intensive Care, Kindai University Faculty of Medicine, Osaka, Japan
| | - Satoru Kobayashi
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY
| | - Aliya Masihuddin
- Department of Molecular, Cellular & Biomedical Sciences, City University of New York School of Medicine, City College of New York, New York, NY
| | - Syed Amir Abdali
- Department of Molecular, Cellular & Biomedical Sciences, City University of New York School of Medicine, City College of New York, New York, NY
| | - A. M. Pramodh Bandara Seneviratne
- Department of Molecular, Cellular & Biomedical Sciences, City University of New York School of Medicine, City College of New York, New York, NY
| | - Sachiyo Ishii
- Department of Anesthesiology and Critical Care, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Jun Iida
- Department of Anesthesiology and Critical Care, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Qiangrong Liang
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY
| | - Jun Yoshioka
- Department of Molecular, Cellular & Biomedical Sciences, City University of New York School of Medicine, City College of New York, New York, NY
- The Graduate Center, City University of New York, New York, NY
| |
Collapse
|
10
|
Schenk S, Sagendorf TJ, Many GM, Lira AK, de Sousa LGO, Bae D, Cicha M, Kramer KS, Muehlbauer M, Hevener AL, Rector RS, Thyfault JP, Williams JP, Goodyear LJ, Esser KA, Newgard CB, Bodine SC. Physiological Adaptations to Progressive Endurance Exercise Training in Adult and Aged Rats: Insights from the Molecular Transducers of Physical Activity Consortium (MoTrPAC). FUNCTION 2024; 5:zqae014. [PMID: 38984994 PMCID: PMC11245678 DOI: 10.1093/function/zqae014] [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: 11/22/2023] [Revised: 01/31/2024] [Accepted: 03/19/2024] [Indexed: 07/11/2024] Open
Abstract
While regular physical activity is a cornerstone of health, wellness, and vitality, the impact of endurance exercise training on molecular signaling within and across tissues remains to be delineated. The Molecular Transducers of Physical Activity Consortium (MoTrPAC) was established to characterize molecular networks underlying the adaptive response to exercise. Here, we describe the endurance exercise training studies undertaken by the Preclinical Animal Sites Studies component of MoTrPAC, in which we sought to develop and implement a standardized endurance exercise protocol in a large cohort of rats. To this end, Adult (6-mo) and Aged (18-mo) female (n = 151) and male (n = 143) Fischer 344 rats were subjected to progressive treadmill training (5 d/wk, ∼70%-75% VO2max) for 1, 2, 4, or 8 wk; sedentary rats were studied as the control group. A total of 18 solid tissues, as well as blood, plasma, and feces, were collected to establish a publicly accessible biorepository and for extensive omics-based analyses by MoTrPAC. Treadmill training was highly effective, with robust improvements in skeletal muscle citrate synthase activity in as little as 1-2 wk and improvements in maximum run speed and maximal oxygen uptake by 4-8 wk. For body mass and composition, notable age- and sex-dependent responses were observed. This work in mature, treadmill-trained rats represents the most comprehensive and publicly accessible tissue biorepository, to date, and provides an unprecedented resource for studying temporal-, sex-, and age-specific responses to endurance exercise training in a preclinical rat model.
Collapse
Affiliation(s)
- Simon Schenk
- Department of Orthopaedic Surgery, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Tyler J Sagendorf
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Gina M Many
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Ana K Lira
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Luis G O de Sousa
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Dam Bae
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Michael Cicha
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Kyle S Kramer
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Michael Muehlbauer
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA
| | - Andrea L Hevener
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - R Scott Rector
- Research Service,
Harry S. Truman Memorial Veterans’ Medical Center, Columbia, MO 65201, USA
- NextGen Precision Health,
University of Missouri, Columbia, MO 65201, USA
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO 65211, USA
| | - John P Thyfault
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- KU Diabetes Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - John P Williams
- Division of Aging Biology, National Institute on Aging, National Institutes of Health, Bethesda, MD 20898, USA
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism,
Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Karyn A Esser
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Christopher B Newgard
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA
| | - Sue C Bodine
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| |
Collapse
|
11
|
Arroum T, Hish GA, Burghardt KJ, Ghamloush M, Bazzi B, Mrech A, Morse PT, Britton SL, Koch LG, McCully JD, Hüttemann M, Malek MH. Mitochondria Transplantation: Rescuing Innate Muscle Bioenergetic Impairment in a Model of Aging and Exercise Intolerance. J Strength Cond Res 2024; 38:1189-1199. [PMID: 38900170 PMCID: PMC11192236 DOI: 10.1519/jsc.0000000000004793] [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] [Indexed: 06/21/2024]
Abstract
ABSTRACT Arroum, T, Hish, GA, Burghardt, KJ, Ghamloush, M, Bazzi, B, Mrech, A, Morse, PT, Britton, SL, Koch, LG, McCully, JD, Hüttemann, M, and Malek, MH. Mitochondria transplantation: Rescuing innate muscle bioenergetic impairment in a model of aging and exercise intolerance. J Strength Cond Res 38(7): 1189-1199, 2024-Mitochondria, through oxidative phosphorylation, are crucial for energy production. Disease, genetic impairment, or deconditioning can harm muscle mitochondria, affecting energy production. Endurance training enhances mitochondrial function but assumes mobility. Individuals with limited mobility lack effective treatments for mitochondrial dysfunction because of disease or aging. Mitochondrial transplantation replaces native mitochondria that have been damaged with viable, respiration-competent mitochondria. Here, we used a rodent model selectively bred for low-capacity running (LCR), which exhibits innate mitochondrial dysfunction in the hind limb muscles. Hence, the purpose of this study was to use a distinct breed of rats (i.e., LCR) that display hereditary skeletal muscle mitochondrial dysfunction to evaluate the consequences of mitochondrial transplantation. We hypothesized that the transplantation of mitochondria would effectively alleviate mitochondrial dysfunction in the hind limb muscles of rats when compared with placebo injections. In addition, we hypothesized that rats receiving the mitochondrial transplantation would experience an improvement in their functional capacity, as evaluated through incremental treadmill testing. Twelve aged LCR male rats (18 months old) were randomized into 2 groups (placebo or mitochondrial transplantation). One LCR rat of the same age and sex was used as the donor to isolate mitochondria from the hindlimb muscles. Isolated mitochondria were injected into both hindlimb muscles (quadriceps femoris, tibialis anterior (TA), and gastrocnemius complex) of a subset LCR (n = 6; LCR-M) rats. The remaining LCR (n = 5; LCR-P) subset received a placebo injection containing only the vehicle without the isolated mitochondria. Four weeks after mitochondrial transplantation, rodents were euthanized and hindlimb muscles harvested. The results indicated a significant (p < 0.05) increase in mitochondrial markers for glycolytic (plantaris and TA) and mixed (quadricep femoris) muscles, but not oxidative muscle (soleus). Moreover, we found significant (p < 0.05) epigenetic changes (i.e., hypomethylation) at the global and site-specific levels for a key mitochondrial regulator (transcription factor A mitochondrial) between the placebo and mitochondrial transplantation groups. To our knowledge, this is the first study to examine the efficacy of mitochondrial transplantation in a rodent model of aging with congenital skeletal muscle dysfunction.
Collapse
Affiliation(s)
- Tasnim Arroum
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, 48201
| | - Gerald A. Hish
- Unit for Laboratory Animal Medicine (ULAM), University of Michigan, Ann Arbor, Ann Arbor, MI 48109
| | - Kyle J. Burghardt
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Detroit, MI 48201
| | - Mohamed Ghamloush
- Physical Therapy Program, Wayne State University, Eugene Applebaum College of Pharmacy and Health Sciences, Department of Health Care Sciences, Detroit, MI 48201
- Integrative Physiology of Exercise Laboratory, Wayne State University, Eugene Applebaum College of Pharmacy and Health Sciences, Department of Health Care Sciences, Detroit, MI 48201
| | - Belal Bazzi
- Physical Therapy Program, Wayne State University, Eugene Applebaum College of Pharmacy and Health Sciences, Department of Health Care Sciences, Detroit, MI 48201
- Integrative Physiology of Exercise Laboratory, Wayne State University, Eugene Applebaum College of Pharmacy and Health Sciences, Department of Health Care Sciences, Detroit, MI 48201
| | - Abdallah Mrech
- Physical Therapy Program, Wayne State University, Eugene Applebaum College of Pharmacy and Health Sciences, Department of Health Care Sciences, Detroit, MI 48201
- Integrative Physiology of Exercise Laboratory, Wayne State University, Eugene Applebaum College of Pharmacy and Health Sciences, Department of Health Care Sciences, Detroit, MI 48201
| | - Paul T. Morse
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, 48201
| | - Steven L. Britton
- Department of Anesthesiology, University of Michigan, Ann Arbor, Ann Arbor, MI 48109
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Ann Arbor, MI 48109
| | - Lauren G. Koch
- Department of Physiology and Pharmacology, The University of Toledo, College of Medicine and Life Sciences, Toledo, OH 43606
| | - James D. McCully
- Department of Cardiac Surgery, Boston Children’s Hospital Harvard Medical School, Boston, MA 02115
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, 48201
| | - Moh H. Malek
- Physical Therapy Program, Wayne State University, Eugene Applebaum College of Pharmacy and Health Sciences, Department of Health Care Sciences, Detroit, MI 48201
- Integrative Physiology of Exercise Laboratory, Wayne State University, Eugene Applebaum College of Pharmacy and Health Sciences, Department of Health Care Sciences, Detroit, MI 48201
| |
Collapse
|
12
|
Vaes AW, Burtin C, Casaburi R, Celli BR, Evans RA, Lareau SC, Nici L, Rochester CL, Troosters T. Prevalence and prognostic importance of exercise limitation and physical inactivity in COPD. Breathe (Sheff) 2024; 20:230179. [PMID: 38873237 PMCID: PMC11167648 DOI: 10.1183/20734735.0179-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/08/2024] [Indexed: 06/15/2024] Open
Abstract
Exercise limitation and physical inactivity are separate, but related constructs. Both are commonly present in individuals with COPD, contribute to disease burden over and above the respiratory impairments, and are independently predictive of adverse outcomes. Because of this, clinicians should consider assessing these variables in their patients with COPD. Field tests of exercise performance such as the 6-min walk test and the incremental and endurance shuttle walk tests require limited additional resources, and results correlate with negative outcomes. Laboratory measures of exercise performance using a treadmill or cycle ergometer assess exercise capacity, provide prognostic information and have the advantage of explaining physiological mechanisms (and their interactions) underpinning exercise limitation. Limitations in exercise capacity (i.e. "cannot do") and physical inactivity (i.e. "do not do") are both associated with mortality; exercise limitation appears to be the more important driver of this outcome.
Collapse
Affiliation(s)
- Anouk W. Vaes
- Department of Research and Development, Ciro, Horn, The Netherlands
| | - Chris Burtin
- REVAL - Rehabilitation Research Center, BIOMED - Biomedical Research Institute, Faculty of Rehabilitation Sciences, Hasselt University, Diepenbeek, Belgium
| | - Richard Casaburi
- Rehabilitation Clinical Trials Center, Lundquist Institute for Biomedical Innovation at Harbor-University of California, Los Angeles (UCLA) Medical Center, Torrance, CA, USA
| | - Bartolome R. Celli
- Pulmonary and Critical Care Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rachael A. Evans
- Department of Respiratory Science, University of Leicester, Leicester, UK
| | - Suzanne C. Lareau
- University of Colorado College of Nursing, Anschutz Medical Campus, Aurora, CO, USA
| | - Linda Nici
- Providence Veterans Affairs Medical Center, Providence, RI, USA
- The Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Carolyn L. Rochester
- Yale University School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
| | | |
Collapse
|
13
|
Vetr NG, Gay NR, Montgomery SB. The impact of exercise on gene regulation in association with complex trait genetics. Nat Commun 2024; 15:3346. [PMID: 38693125 PMCID: PMC11063075 DOI: 10.1038/s41467-024-45966-w] [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: 02/01/2023] [Accepted: 02/01/2024] [Indexed: 05/03/2024] Open
Abstract
Endurance exercise training is known to reduce risk for a range of complex diseases. However, the molecular basis of this effect has been challenging to study and largely restricted to analyses of either few or easily biopsied tissues. Extensive transcriptome data collected across 15 tissues during exercise training in rats as part of the Molecular Transducers of Physical Activity Consortium has provided a unique opportunity to clarify how exercise can affect tissue-specific gene expression and further suggest how exercise adaptation may impact complex disease-associated genes. To build this map, we integrate this multi-tissue atlas of gene expression changes with gene-disease targets, genetic regulation of expression, and trait relationship data in humans. Consensus from multiple approaches prioritizes specific tissues and genes where endurance exercise impacts disease-relevant gene expression. Specifically, we identify a total of 5523 trait-tissue-gene triplets to serve as a valuable starting point for future investigations [Exercise; Transcription; Human Phenotypic Variation].
Collapse
|
14
|
Arroum T, Hish GA, Burghardt KJ, McCully JD, Hüttemann M, Malek MH. Mitochondrial Transplantation's Role in Rodent Skeletal Muscle Bioenergetics: Recharging the Engine of Aging. Biomolecules 2024; 14:493. [PMID: 38672509 PMCID: PMC11048484 DOI: 10.3390/biom14040493] [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: 02/19/2024] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Mitochondria are the 'powerhouses of cells' and progressive mitochondrial dysfunction is a hallmark of aging in skeletal muscle. Although different forms of exercise modality appear to be beneficial to attenuate aging-induced mitochondrial dysfunction, it presupposes that the individual has a requisite level of mobility. Moreover, non-exercise alternatives (i.e., nutraceuticals or pharmacological agents) to improve skeletal muscle bioenergetics require time to be effective in the target tissue and have another limitation in that they act systemically and not locally where needed. Mitochondrial transplantation represents a novel directed therapy designed to enhance energy production of tissues impacted by defective mitochondria. To date, no studies have used mitochondrial transplantation as an intervention to attenuate aging-induced skeletal muscle mitochondrial dysfunction. The purpose of this investigation, therefore, was to determine whether mitochondrial transplantation can enhance skeletal muscle bioenergetics in an aging rodent model. We hypothesized that mitochondrial transplantation would result in sustained skeletal muscle bioenergetics leading to improved functional capacity. METHODS Fifteen female mice (24 months old) were randomized into two groups (placebo or mitochondrial transplantation). Isolated mitochondria from a donor mouse of the same sex and age were transplanted into the hindlimb muscles of recipient mice (quadriceps femoris, tibialis anterior, and gastrocnemius complex). RESULTS The results indicated significant increases (ranging between ~36% and ~65%) in basal cytochrome c oxidase and citrate synthase activity as well as ATP levels in mice receiving mitochondrial transplantation relative to the placebo. Moreover, there were significant increases (approx. two-fold) in protein expression of mitochondrial markers in both glycolytic and oxidative muscles. These enhancements in the muscle translated to significant improvements in exercise tolerance. CONCLUSIONS This study provides initial evidence showing how mitochondrial transplantation can promote skeletal muscle bioenergetics in an aging rodent model.
Collapse
Affiliation(s)
- Tasnim Arroum
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA; (T.A.); (M.H.)
| | - Gerald A. Hish
- Unit for Laboratory Animal Medicine (ULAM), University of Michigan, Ann Arbor, MI 48109, USA
| | - Kyle J. Burghardt
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - James D. McCully
- Department of Cardiac Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA; (T.A.); (M.H.)
| | - Moh H. Malek
- Physical Therapy Program, Department of Health Care Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
- Integrative Physiology of Exercise Laboratory, Department of Health Care Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
15
|
Fleischman JY, Casey JL, Meijer JL, Treutelaar MK, Rajendiran TM, Soni T, Evans CR, Burant CF. Sex modulates the diet-induced changes to the plasma lipidome in a rat model of cardiorespiratory fitness. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159451. [PMID: 38191091 DOI: 10.1016/j.bbalip.2024.159451] [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: 10/02/2023] [Revised: 01/01/2024] [Accepted: 01/03/2024] [Indexed: 01/10/2024]
Abstract
OBJECTIVE Individuals with higher intrinsic cardiorespiratory fitness (CRF) experience decreased rates of cardiometabolic disease and mortality, and high CRF is associated with increased utilization of fatty acids (FAs) for energy. Studies suggest a complex relationship between CRF, diet, and sex with health outcomes, but this interaction is understudied. We hypothesized that FA utilization differences by fitness and sex could be detected in the plasma metabolome when rats or humans were fed a high carbohydrate (HC) or high fat (HF) diet. METHODS Male and female rats selectively bred for low (LCR) and high (HCR) CRF were fed a chow diet or a sucrose-free HF (45 % fat) or HC (10 % fat) diet. Plasma samples were collected at days 0, 3, and 14. Human plasma data was collected from male and female participants who were randomized into a HC or HF diet for 21 days. Samples were analyzed using liquid chromatography-mass spectrometry and regression statistics were used to quantify the effect of diet, CRF, and sex on the lipidome. RESULTS In rats, the baseline lipidome is more significantly influenced by sex than by CRF, especially as elevated diglycerides, triglycerides, phosphatidylcholines, and lysophosphatidylcholines in males. A dynamic response to diet was observed 3 days after diet, but after 14 days of either diet, the lipidome was modulated by sex with a larger effect size than by diet. Data from the human study also suggests a sex-dependent response to diet with opposite directionality of affect compared to rats, highlighting species-dependent responses to dietary intervention.
Collapse
Affiliation(s)
- Johanna Y Fleischman
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States of America; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States of America
| | - James L Casey
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States of America
| | - Jennifer L Meijer
- Department of Medicine, Weight and Wellness Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States of America; Quantitative Biomedical Sciences, Geisel School of Medicine, Dartmouth College, Hanover, NH, United States of America
| | - Mary K Treutelaar
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States of America
| | - Thekkelnaycke M Rajendiran
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI, United States of America
| | - Tanu Soni
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI, United States of America
| | - Charles R Evans
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States of America; Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI, United States of America
| | - Charles F Burant
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States of America; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States of America; Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI, United States of America.
| |
Collapse
|
16
|
Franczak E, Maurer A, Drummond VC, Kugler BA, Wells E, Wenger M, Peelor FF, Crosswhite A, McCoin CS, Koch LG, Britton SL, Miller BF, Thyfault JP. Divergence in aerobic capacity and energy expenditure influence metabolic tissue mitochondrial protein synthesis rates in aged rats. GeroScience 2024; 46:2207-2222. [PMID: 37880490 PMCID: PMC10828174 DOI: 10.1007/s11357-023-00985-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/14/2023] [Indexed: 10/27/2023] Open
Abstract
Age-associated declines in aerobic capacity promote the development of various metabolic diseases. In rats selectively bred for high/low intrinsic aerobic capacity, greater aerobic capacity reduces susceptibility to metabolic disease while increasing longevity. However, little remains known how intrinsic aerobic capacity protects against metabolic disease, particularly with aging. Here, we tested the effects of aging and intrinsic aerobic capacity on systemic energy expenditure, metabolic flexibility and mitochondrial protein synthesis rates using 24-month-old low-capacity (LCR) or high-capacity runner (HCR) rats. Rats were fed low-fat diet (LFD) or high-fat diet (HFD) for eight weeks, with energy expenditure (EE) and metabolic flexibility assessed utilizing indirect calorimetry during a 48 h fast/re-feeding metabolic challenge. Deuterium oxide (D2O) labeling was used to assess mitochondrial protein fraction synthesis rates (FSR) over a 7-day period. HCR rats possessed greater EE during the metabolic challenge. Interestingly, HFD induced changes in respiratory exchange ratio (RER) in male and female rats, while HCR female rat RER was largely unaffected by diet. In addition, analysis of protein FSR in skeletal muscle, brain, and liver mitochondria showed tissue-specific adaptations between HCR and LCR rats. While brain and liver protein FSR were altered by aerobic capacity and diet, these effects were less apparent in skeletal muscle. Overall, we provide evidence that greater aerobic capacity promotes elevated EE in an aged state, while also regulating metabolic flexibility in a sex-dependent manner. Modulation of mitochondrial protein FSR by aerobic capacity is tissue-specific with aging, likely due to differential energetic requirements by each tissue.
Collapse
Affiliation(s)
- Edziu Franczak
- Department of Cell Biology and Physiology, Medical Center, The University of Kansas, Kansas City, KS, 66160, USA
- Kansas City Veterans Affairs Medical Center, Kansas City, MO, 64128, USA
| | - Adrianna Maurer
- Department of Cell Biology and Physiology, Medical Center, The University of Kansas, Kansas City, KS, 66160, USA
| | - Vivien Csikos Drummond
- Department of Cell Biology and Physiology, Medical Center, The University of Kansas, Kansas City, KS, 66160, USA
| | - Benjamin A Kugler
- Department of Cell Biology and Physiology, Medical Center, The University of Kansas, Kansas City, KS, 66160, USA
- Kansas Center for Metabolism and Obesity Research, Kansas City, MO, 64128, USA
- KU Diabetes Institute and Department of Internal Medicine-Division of Endocrinology and Metabolism, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Hemenway Life Sciences Innovation Center, Mailstop 3043, Kansas City, KS, 66160, USA
| | - Emily Wells
- Department of Cell Biology and Physiology, Medical Center, The University of Kansas, Kansas City, KS, 66160, USA
| | - Madi Wenger
- Department of Cell Biology and Physiology, Medical Center, The University of Kansas, Kansas City, KS, 66160, USA
- Kansas Center for Metabolism and Obesity Research, Kansas City, MO, 64128, USA
- KU Diabetes Institute and Department of Internal Medicine-Division of Endocrinology and Metabolism, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Hemenway Life Sciences Innovation Center, Mailstop 3043, Kansas City, KS, 66160, USA
| | | | - Abby Crosswhite
- Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Colin S McCoin
- Department of Cell Biology and Physiology, Medical Center, The University of Kansas, Kansas City, KS, 66160, USA
- Kansas City Veterans Affairs Medical Center, Kansas City, MO, 64128, USA
- Kansas Center for Metabolism and Obesity Research, Kansas City, MO, 64128, USA
- KU Diabetes Institute and Department of Internal Medicine-Division of Endocrinology and Metabolism, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Hemenway Life Sciences Innovation Center, Mailstop 3043, Kansas City, KS, 66160, USA
| | - Lauren G Koch
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43606, USA
| | - Steven L Britton
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Benjamin F Miller
- KU Diabetes Institute and Department of Internal Medicine-Division of Endocrinology and Metabolism, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Hemenway Life Sciences Innovation Center, Mailstop 3043, Kansas City, KS, 66160, USA
| | - John P Thyfault
- Department of Cell Biology and Physiology, Medical Center, The University of Kansas, Kansas City, KS, 66160, USA.
- Kansas City Veterans Affairs Medical Center, Kansas City, MO, 64128, USA.
- Kansas Center for Metabolism and Obesity Research, Kansas City, MO, 64128, USA.
- KU Diabetes Institute and Department of Internal Medicine-Division of Endocrinology and Metabolism, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Hemenway Life Sciences Innovation Center, Mailstop 3043, Kansas City, KS, 66160, USA.
| |
Collapse
|
17
|
Kemi OJ, Hoydal MA, Haram PM, Smith GL, Ellingsen O, Koch LG, Britton SL, Wisloff U. Inherited physical capacity: Widening divergence from young to adult to old. Ann N Y Acad Sci 2024; 1534:145-155. [PMID: 38520387 DOI: 10.1111/nyas.15130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2024]
Abstract
Cardiorespiratory performance segregates into rat strains of inherited low- and high-capacity runners (LCRs and HCRs); during adulthood, this segregation remains stable, but widens in senescence and is followed by segregated function, health, and mortality. However, this segregation has not been investigated prior to adulthood. We, therefore, assessed cardiorespiratory performance and cardiac cell (cardiomyocyte) structure-function in 1- and 4-month-old LCRs and HCRs. Maximal oxygen uptake was 23% less in LCRs at 1-month compared to HCRs at 1-month, and 72% less at 4 months. Cardiomyocyte contractility was 37-56% decreased, and Ca2+ release was 34-62% decreased, in 1- and 4-month LCRs versus HCRs. This occurred because HCRs had improved contractility and Ca2+ release during maturation, whereas LCRs did not. In quiescent cardiomyocytes, LCRs displayed 180% and 297% more Ca2+ sparks and 91% and 38% more Ca2+ waves at 1 and 4 months versus HCRs. Cell sizes were not different between LCRs and HCRs, but LCRs showed reduced transverse-tubules versus HCRs, though no discrepant transverse-tubule generation occurred during maturation. In conclusion, LCRs show reduced scores for aerobic capacity and cardiomyocyte structure-function compared to HCRs and there is a widening divergence between LCRs and HCRs during juvenile to near-adult maturation.
Collapse
Affiliation(s)
- Ole J Kemi
- School of Cardiovascular and Metabolic Health, University of Glasgow College of Medical, Veterinary and Life Sciences, Glasgow, UK
| | - Morten A Hoydal
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Faculty of Medicine and Health Sciences, Trondheim, Norway
| | - Per M Haram
- Department of Cardiothoracic Surgery, St Olav's Hospital, Trondheim, Norway
| | - Godfrey L Smith
- School of Cardiovascular and Metabolic Health, University of Glasgow College of Medical, Veterinary and Life Sciences, Glasgow, UK
| | - Oyvind Ellingsen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Faculty of Medicine and Health Sciences, Trondheim, Norway
- Department of Cardiology, St Olav's Hospital, Trondheim, Norway
| | - Lauren G Koch
- Department of Physiology and Pharmacology, University of Toledo, Toledo, Ohio, USA
| | - Steven L Britton
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Ulrik Wisloff
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Faculty of Medicine and Health Sciences, Trondheim, Norway
- School of Human Movement and Nutrition Science, University of Queensland, Saint Lucia, Queensland, Australia
| |
Collapse
|
18
|
Bubak MP, Davidyan A, O'Reilly CL, Mondal SA, Keast J, Doidge SM, Borowik AK, Taylor ME, Volovičeva E, Kinter MT, Britton SL, Koch LG, Stout MB, Lewis TL, Miller BF. Metformin treatment results in distinctive skeletal muscle mitochondrial remodeling in rats with different intrinsic aerobic capacities. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.582957. [PMID: 38496648 PMCID: PMC10942369 DOI: 10.1101/2024.03.01.582957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
The rationale for the use of metformin as a treatment to slow aging was largely based on data collected from metabolically unhealthy individuals. For healthspan extension metformin will also be used in periods of good health. To understand potential context specificity of metformin treatment on skeletal muscle, we used a rat model (HCR/LCR) with a divide in intrinsic aerobic capacity. Outcomes of metformin treatment differed based on baseline intrinsic mitochondrial function, oxidative capacity of the muscle (gastroc vs soleus), and the mitochondrial population (IMF vs SS). Metformin caused lower ADP-stimulated respiration in LCRs, with less of a change in HCRs. However, a washout of metformin resulted in an unexpected doubling of respiratory capacity in HCRs. These improvements in respiratory capacity were accompanied by mitochondrial remodeling that included increases in protein synthesis and changes in morphology. Our findings raise questions about whether the positive findings of metformin treatment are broadly applicable.
Collapse
|
19
|
Carazza-Kessler FG, Campos MS, Bittencourt RR, Rosa-Silva HTD, Brum PO, Silveira AK, Teixeira AA, Ribeiro CT, Peixoto DO, Santos L, Andrade G, Panzenhagen AC, Scheibel IM, Gelain DP, Fonseca Moreira JC. Transgenerational inheritance of methylmercury and vitamin A-induced toxicological effects in a Wistar rats environmental-based model. CHEMOSPHERE 2024; 351:141239. [PMID: 38272134 DOI: 10.1016/j.chemosphere.2024.141239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/22/2023] [Accepted: 01/15/2024] [Indexed: 01/27/2024]
Abstract
Mercury (Hg) and vitamin A (VitA) are two environmental factors with potential health impacts, especially during pregnancy and early childhood. Fish and seafood may present elevated levels of methylmercury (MeHg), the major Hg derivative, and VitA. This study aimed to evaluate the transgenerational effects of exposure to MeHg and/or VitA on epigenetic and toxicological parameters in a Wistar rat model. Our findings revealed persistent toxicological effects in generations F1 and F2 following low/mild doses of MeHg and/or VitA exposure during dams' (F0) gestation and breastfeeding. Toxicological effects observed in F2 included chronic DNA damage, bone marrow toxicity, altered microglial content, reduced neuronal signal, and diminished male longevity. Sex-specific patterns were also observed. Co-exposure to MeHg and VitA showed both synergistic and antagonistic effects. Additionally, the study demonstrated that MeHg and VitA affected histone methylation and caused consistent effects in F2. While MeHg exposure has been associated with transgenerational inheritance effects in other organisms, this study provides the first evidence of transgenerational inheritance of MeHg and VitA-induced toxicological effects in rodents. Although the exact mechanism is not yet fully understood, these findings suggest that MeHg and VitA may perpetuate their impacts across generations. The study highlights the need for remedial policies and interventions to mitigate the potential health problems faced by future generations exposed to MeHg or VitA. Further research is warranted to investigate the transgenerational effects beyond F2 and determine the matrilineal or patrilineal inheritance patterns.
Collapse
Affiliation(s)
- Flávio Gabriel Carazza-Kessler
- Centro de Estudos em Estresse Oxidativo - Laboratório 32, Programa de Pós-Graduação em Biologia Celular e Molecular - Instituto de Biociências - Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos 2600 - Prédio Anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.
| | - Marlene Soares Campos
- Centro de Estudos em Estresse Oxidativo - Laboratório 32, Programa de Pós-Graduação em Biologia Celular e Molecular - Instituto de Biociências - Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos 2600 - Prédio Anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.
| | - Reykla Ramon Bittencourt
- Centro de Estudos em Estresse Oxidativo - Laboratório 32, Programa de Pós-Graduação em Biologia Celular e Molecular - Instituto de Biociências - Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos 2600 - Prédio Anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.
| | - Helen Taís da Rosa-Silva
- Centro de Estudos em Estresse Oxidativo - Laboratório 32, Programa de Pós-Graduação em Biologia Celular e Molecular - Instituto de Biociências - Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos 2600 - Prédio Anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.
| | - Pedro Ozorio Brum
- Centro de Estudos em Estresse Oxidativo - Laboratório 32, Programa de Pós-Graduação em Biologia Celular e Molecular - Instituto de Biociências - Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos 2600 - Prédio Anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.
| | - Alexandre Kléber Silveira
- Centro de Estudos em Estresse Oxidativo - Laboratório 32, Programa de Pós-Graduação em Biologia Celular e Molecular - Instituto de Biociências - Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos 2600 - Prédio Anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.
| | - Alexsander Alves Teixeira
- Centro de Estudos em Estresse Oxidativo - Laboratório 32, Programa de Pós-Graduação em Biologia Celular e Molecular - Instituto de Biociências - Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos 2600 - Prédio Anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.
| | - Camila Tiefensee Ribeiro
- Centro de Estudos em Estresse Oxidativo - Laboratório 32, Programa de Pós-Graduação em Biologia Celular e Molecular - Instituto de Biociências - Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos 2600 - Prédio Anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.
| | - Daniel Oppermann Peixoto
- Centro de Estudos em Estresse Oxidativo - Laboratório 32, Programa de Pós-Graduação em Biologia Celular e Molecular - Instituto de Biociências - Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos 2600 - Prédio Anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.
| | - Lucas Santos
- Centro de Estudos em Estresse Oxidativo - Laboratório 32, Programa de Pós-Graduação em Biologia Celular e Molecular - Instituto de Biociências - Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos 2600 - Prédio Anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.
| | - Giovanni Andrade
- Centro de Estudos em Estresse Oxidativo - Laboratório 32, Programa de Pós-Graduação em Biologia Celular e Molecular - Instituto de Biociências - Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos 2600 - Prédio Anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.
| | - Alana Castro Panzenhagen
- Centro de Estudos em Estresse Oxidativo - Laboratório 32, Programa de Pós-Graduação em Biologia Celular e Molecular - Instituto de Biociências - Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos 2600 - Prédio Anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.
| | - Ingrid Matsubara Scheibel
- Centro de Estudos em Estresse Oxidativo - Laboratório 32, Programa de Pós-Graduação em Biologia Celular e Molecular - Instituto de Biociências - Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos 2600 - Prédio Anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.
| | - Daniel Pens Gelain
- Centro de Estudos em Estresse Oxidativo - Laboratório 32, Programa de Pós-Graduação em Biologia Celular e Molecular - Instituto de Biociências - Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos 2600 - Prédio Anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.
| | - José Cláudio Fonseca Moreira
- Centro de Estudos em Estresse Oxidativo - Laboratório 32, Programa de Pós-Graduação em Biologia Celular e Molecular - Instituto de Biociências - Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos 2600 - Prédio Anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.
| |
Collapse
|
20
|
Heyne E, Zeeb S, Junker C, Petzinna A, Schrepper A, Doenst T, Koch LG, Britton SL, Schwarzer M. Exercise Training Differentially Affects Skeletal Muscle Mitochondria in Rats with Inherited High or Low Exercise Capacity. Cells 2024; 13:393. [PMID: 38474357 PMCID: PMC10931189 DOI: 10.3390/cells13050393] [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: 12/31/2023] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Exercise capacity has been related to morbidity and mortality. It consists of an inherited and an acquired part and is dependent on mitochondrial function. We assessed skeletal muscle mitochondrial function in rats with divergent inherited exercise capacity and analyzed the effect of exercise training. Female high (HCR)- and low (LCR)-capacity runners were trained with individually adapted high-intensity intervals or kept sedentary. Interfibrillar (IFM) and subsarcolemmal (SSM) mitochondria from gastrocnemius muscle were isolated and functionally assessed (age: 15 weeks). Sedentary HCR presented with higher exercise capacity than LCR paralleled by higher citrate synthase activity and IFM respiratory capacity in skeletal muscle of HCR. Exercise training increased exercise capacity in both HCR and LCR, but this was more pronounced in LCR. In addition, exercise increased skeletal muscle mitochondrial mass more in LCR. Instead, maximal respiratory capacity was increased following exercise in HCRs' IFM only. The results suggest that differences in skeletal muscle mitochondrial subpopulations are mainly inherited. Exercise training resulted in different mitochondrial adaptations and in higher trainability of LCR. HCR primarily increased skeletal muscle mitochondrial quality while LCR increased mitochondrial quantity in response to exercise training, suggesting that inherited aerobic exercise capacity differentially affects the mitochondrial response to exercise training.
Collapse
Affiliation(s)
- Estelle Heyne
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany; (E.H.); (A.P.); (A.S.); (T.D.)
| | - Susanne Zeeb
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany; (E.H.); (A.P.); (A.S.); (T.D.)
| | - Celina Junker
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany; (E.H.); (A.P.); (A.S.); (T.D.)
| | - Andreas Petzinna
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany; (E.H.); (A.P.); (A.S.); (T.D.)
| | - Andrea Schrepper
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany; (E.H.); (A.P.); (A.S.); (T.D.)
| | - Torsten Doenst
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany; (E.H.); (A.P.); (A.S.); (T.D.)
| | - Lauren G. Koch
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University Toledo, Toledo, OH 43606, USA;
| | - Steven L. Britton
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA;
| | - Michael Schwarzer
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany; (E.H.); (A.P.); (A.S.); (T.D.)
| |
Collapse
|
21
|
Mäkinen EE, Lensu S, Wikgren J, Pekkala S, Koch LG, Britton SL, Nokia MS. Intrinsic running capacity associates with hippocampal electrophysiology and long-term potentiation in rats. Neurosci Lett 2024; 823:137665. [PMID: 38301912 DOI: 10.1016/j.neulet.2024.137665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 01/11/2024] [Accepted: 01/29/2024] [Indexed: 02/03/2024]
Abstract
Good aerobic and metabolic fitness associates with better cognitive performance and brain health. Conversely, poor metabolic health predisposes to neurodegenerative diseases. Our previous findings indicate that rats selectively bred for Low Capacity for Running (LCR) show less synaptic plasticity and more inflammation in the hippocampus and perform worse in tasks requiring flexible cognition than rats bred for High Capacity for Running (HCR). Here we aimed to determine whether hippocampal electrophysiological activity related to learning and memory would be impaired in LCR compared to HCR rats. We also studied whether an exercise intervention could even out the possible differences. We conducted in vivo recordings from the dorsal hippocampus under terminal urethane anesthesia in middle-aged sedentary males and female rats, and in females allowed to access running wheels for 6 weeks. Our results indicate stronger long-term potentiation (LTP) in the CA3-CA1 synapse in HCR than LCR rats, and in female than male rats. Compared to LCR rats, HCR rats had more dentate spikes and more gamma epochs, the occurrence of which also correlated positively with the magnitude of LTP. Voluntary running reduced the differences between female LCR and HCR rats. In conclusion, low innate fitness links to reduced hippocampal function and plasticity which can seems to improve with voluntary aerobic exercise even in middle age.
Collapse
Affiliation(s)
- Elina E Mäkinen
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland.
| | - Sanna Lensu
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland; Centre for Interdisciplinary Brain Research, Department of Psychology, University of Jyväskylä, Jyväskylä, Finland
| | - Jan Wikgren
- Centre for Interdisciplinary Brain Research, Department of Psychology, University of Jyväskylä, Jyväskylä, Finland
| | - Satu Pekkala
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Lauren G Koch
- Department of Physiology and Pharmacology, The University of Toledo, Toledo, OH, USA
| | - Steven L Britton
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Miriam S Nokia
- Centre for Interdisciplinary Brain Research, Department of Psychology, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
22
|
Huang T, Li H, Chen X, Chen D, Yu B, He J, Luo Y, Yan H, Zheng P, Yu J, Huang Z. Dietary Ferulic Acid Increases Endurance Capacity by Promoting Skeletal Muscle Oxidative Phenotype, Mitochondrial Function, and Antioxidant Capacity. Mol Nutr Food Res 2024; 68:e2200719. [PMID: 38193241 DOI: 10.1002/mnfr.202200719] [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: 10/22/2022] [Revised: 06/23/2023] [Indexed: 01/10/2024]
Abstract
SCOPE Endurance capacity is essential for endurance athletes' achievement and individuals' health. Nutritional supplements are a proven way to enhance endurance capacity. Previous studies have shown that ferulic acid (FA) enhances endurance capacity, but the underlying mechanism is unclear. The study is aimed to investigate the mechanism by which FA increases endurance capacity. METHODS AND RESULTS Forty mice are divided into control and 0.5% FA-supplemented groups, and an exhaustive swimming test demonstrates increased endurance capacity with FA supplementation. This study investigates the underlying mechanism for this effect of FA. Firstly, RT-PCR and western blot analysis find that FA increases the transformation from fast to slow muscle fiber. Additionally, adenosine triphosphate concentration, metabolic enzyme activity, and mitochondrial DNA analysis find that FA increases mitochondrial biogenesis and activates nuclear factor erythroid 2-related factor (NRF)1 signaling pathway in muscle. Besides, through antioxidant capacity analysis, this study finds that FA activates NRF2 signaling pathway and improves the antioxidant capacity in muscle. Moreover, inhibiting NRF2 eliminates FA's effect on muscle fiber transformation in C2C12 cells. CONCLUSION Our results suggest that FA increases endurance capacity by promoting skeletal muscle oxidative phenotype, mitochondrial function, and antioxidant capacity, which may be related to the NRF1 and NRF2 signaling pathways.
Collapse
Affiliation(s)
- Tengteng Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, P. R. China
| | - Huawei Li
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, P. R. China
| | - Xiaoling Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, P. R. China
| | - Daiwen Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, P. R. China
| | - Bing Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, P. R. China
| | - Jun He
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, P. R. China
| | - Yuheng Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, P. R. China
| | - Hui Yan
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, P. R. China
| | - Ping Zheng
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, P. R. China
| | - Jie Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, P. R. China
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, P. R. China
| |
Collapse
|
23
|
Tobias GC, Gomes JLP, Fernandes LG, Voltarelli VA, de Almeida NR, Jannig PR, de Souza RWA, Negrão CE, Oliveira EM, Chammas R, Alves CRR, Brum PC. Aerobic exercise training mitigates tumor growth and cancer-induced splenomegaly through modulation of non-platelet platelet factor 4 expression. Sci Rep 2023; 13:21970. [PMID: 38081853 PMCID: PMC10713653 DOI: 10.1038/s41598-023-47217-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/10/2023] [Indexed: 12/18/2023] Open
Abstract
Exercise training reduces the incidence of several cancers, but the mechanisms underlying these effects are not fully understood. Exercise training can affect the spleen function, which controls the hematopoiesis and immune response. Analyzing different cancer models, we identified that 4T1, LLC, and CT26 tumor-bearing mice displayed enlarged spleen (splenomegaly), and exercise training reduced spleen mass toward control levels in two of these models (LLC and CT26). Exercise training also slowed tumor growth in melanoma B16F10, colon tumor 26 (CT26), and Lewis lung carcinoma (LLC) tumor-bearing mice, with minor effects in mammary carcinoma 4T1, MDA-MB-231, and MMTV-PyMT mice. In silico analyses using transcriptome profiles derived from these models revealed that platelet factor 4 (Pf4) is one of the main upregulated genes associated with splenomegaly during cancer progression. To understand whether exercise training would modulate the expression of these genes in the tumor and spleen, we investigated particularly the CT26 model, which displayed splenomegaly and had a clear response to the exercise training effects. RT-qPCR analysis confirmed that trained CT26 tumor-bearing mice had decreased Pf4 mRNA levels in both the tumor and spleen when compared to untrained CT26 tumor-bearing mice. Furthermore, exercise training specifically decreased Pf4 mRNA levels in the CT26 tumor cells. Aspirin treatment did not change tumor growth, splenomegaly, and tumor Pf4 mRNA levels, confirming that exercise decreased non-platelet Pf4 mRNA levels. Finally, tumor Pf4 mRNA levels are deregulated in The Cancer Genome Atlas Program (TCGA) samples and predict survival in multiple cancer types. This highlights the potential therapeutic value of exercise as a complementary approach to cancer treatment and underscores the importance of understanding the exercise-induced transcriptional changes in the spleen for the development of novel cancer therapies.
Collapse
Affiliation(s)
- Gabriel C Tobias
- School of Physical Education and Sport, Universidade de São Paulo, Avenida Professor Mello Moraes, 65-Butantã, São Paulo, SP, 05508-030, Brazil.
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
| | - João L P Gomes
- School of Physical Education and Sport, Universidade de São Paulo, Avenida Professor Mello Moraes, 65-Butantã, São Paulo, SP, 05508-030, Brazil
| | - Larissa G Fernandes
- School of Physical Education and Sport, Universidade de São Paulo, Avenida Professor Mello Moraes, 65-Butantã, São Paulo, SP, 05508-030, Brazil
| | - Vanessa A Voltarelli
- School of Physical Education and Sport, Universidade de São Paulo, Avenida Professor Mello Moraes, 65-Butantã, São Paulo, SP, 05508-030, Brazil
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Ney R de Almeida
- School of Physical Education and Sport, Universidade de São Paulo, Avenida Professor Mello Moraes, 65-Butantã, São Paulo, SP, 05508-030, Brazil
| | - Paulo R Jannig
- School of Physical Education and Sport, Universidade de São Paulo, Avenida Professor Mello Moraes, 65-Butantã, São Paulo, SP, 05508-030, Brazil
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Rodrigo W Alves de Souza
- School of Physical Education and Sport, Universidade de São Paulo, Avenida Professor Mello Moraes, 65-Butantã, São Paulo, SP, 05508-030, Brazil
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Carlos E Negrão
- School of Physical Education and Sport, Universidade de São Paulo, Avenida Professor Mello Moraes, 65-Butantã, São Paulo, SP, 05508-030, Brazil
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Edilamar M Oliveira
- School of Physical Education and Sport, Universidade de São Paulo, Avenida Professor Mello Moraes, 65-Butantã, São Paulo, SP, 05508-030, Brazil
| | - Roger Chammas
- Department of Radiology and Oncology, Faculdade de Medicine, Universidade de São Paulo, São Paulo, Brazil
| | - Christiano R R Alves
- School of Physical Education and Sport, Universidade de São Paulo, Avenida Professor Mello Moraes, 65-Butantã, São Paulo, SP, 05508-030, Brazil
| | - Patricia C Brum
- School of Physical Education and Sport, Universidade de São Paulo, Avenida Professor Mello Moraes, 65-Butantã, São Paulo, SP, 05508-030, Brazil.
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
24
|
Fleischman JY, Van den Bergh F, Collins NL, Bowers M, Beard DA, Burant CF. Higher mitochondrial oxidative capacity is the primary molecular differentiator in muscle of rats with high and low intrinsic cardiorespiratory fitness. Mol Metab 2023; 76:101793. [PMID: 37625738 PMCID: PMC10480665 DOI: 10.1016/j.molmet.2023.101793] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/07/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
OBJECTIVE Cardiorespiratory fitness (CRF) is tightly linked with health and longevity and is implicated in metabolic flexibility and substrate metabolism. The high capacity runner (HCR) and low capacity runner (LCR) rat lines are a genetically heterogeneous rat model selected and bred for CRF that reflect CRF in humans by exhibiting differences in nutrient handling. This study aims to differentiate the intrinsic substrate preference of the HCR compared to LCR rats to better understand the intersection of mitochondrial respiration and intrinsic CRF. METHODS We performed bulk skeletal muscle RNA-Sequencing on male and female HCR and LCR rats and assessed the effect of rat line on mitochondrial gene expression pathways using the MitoCarta3.0 database. In a separate cohort of rats, mitochondria were isolated from skeletal and cardiac muscle and maximal oxidation rates were measured using an Oroboros O2k when provided either pyruvate or fatty acid substrates. RESULTS The expression of mitochondrial genes are significantly upregulated in HCR skeletal muscle in both male and female rats. In respirometry experiments, fatty acid oxidative capacities were greater in HCR compared to LCR, and male compared to female rats, as a function of both mitochondrial quality and mitochondrial density. This effect was greater in the skeletal muscle than in the heart. Pyruvate oxidation did not differ significantly between lines. CONCLUSIONS The capacity for increased fatty acid oxidation in the HCR rat is a result of selection for running capacity and is likely a key contributor to the healthy metabolic phenotype of individuals with high CRF.
Collapse
Affiliation(s)
- Johanna Y Fleischman
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, USA
| | | | - Nicole L Collins
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Madelyn Bowers
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Daniel A Beard
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, USA.
| | - Charles F Burant
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, USA.
| |
Collapse
|
25
|
Tynkkynen NP, Törmäkangas T, Palviainen T, Hyvärinen M, Klevjer M, Joensuu L, Kujala U, Kaprio J, Bye A, Sillanpää E. Associations of polygenic inheritance of physical activity with aerobic fitness, cardiometabolic risk factors and diseases: the HUNT study. Eur J Epidemiol 2023; 38:995-1008. [PMID: 37603226 PMCID: PMC10501929 DOI: 10.1007/s10654-023-01029-w] [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: 04/05/2023] [Accepted: 07/10/2023] [Indexed: 08/22/2023]
Abstract
Physical activity (PA), aerobic fitness, and cardiometabolic diseases (CMD) are highly heritable multifactorial phenotypes. Shared genetic factors may underlie the associations between higher levels of PA and better aerobic fitness and a lower risk for CMDs. We aimed to study how PA genotype associates with self-reported PA, aerobic fitness, cardiometabolic risk factors and diseases. PA genotype, which combined variation in over one million of gene variants, was composed using the SBayesR polygenic scoring methodology. First, we constructed a polygenic risk score for PA in the Trøndelag Health Study (N = 47,148) using UK Biobank single nucleotide polymorphism-specific weights (N = 400,124). The associations of the PA PRS and continuous variables were analysed using linear regression models and with CMD incidences using Cox proportional hazard models. The results showed that genotypes predisposing to higher amount of PA were associated with greater self-reported PA (Beta [B] = 0.282 MET-h/wk per SD of PRS for PA, 95% confidence interval [CI] = 0.211, 0.354) but not with aerobic fitness. These genotypes were also associated with healthier cardiometabolic profile (waist circumference [B = -0.003 cm, 95% CI = -0.004, -0.002], body mass index [B = -0.002 kg/m2, 95% CI = -0.004, -0.001], high-density lipoprotein cholesterol [B = 0.004 mmol/L, 95% CI = 0.002, 0.006]) and lower incidence of hypertensive diseases (Hazard Ratio [HR] = 0.97, 95% CI = 0.951, 0.990), stroke (HR = 0.94, 95% CI = 0.903, 0.978) and type 2 diabetes (HR = 0.94, 95 % CI = 0.902, 0.970). Observed associations were independent of self-reported PA. These results support earlier findings suggesting small pleiotropic effects between PA and CMDs and provide new evidence about associations of polygenic inheritance of PA and intermediate cardiometabolic risk factors.
Collapse
Affiliation(s)
- Niko Paavo Tynkkynen
- Gerontology Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, P.O. Box 35 (VIV), Jyväskylä, FIN-40014, Finland
| | - Timo Törmäkangas
- Gerontology Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, P.O. Box 35 (VIV), Jyväskylä, FIN-40014, Finland
| | - Teemu Palviainen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, Helsinki, Finland
| | - Matti Hyvärinen
- Gerontology Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, P.O. Box 35 (VIV), Jyväskylä, FIN-40014, Finland
| | - Marie Klevjer
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Laura Joensuu
- Gerontology Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, P.O. Box 35 (VIV), Jyväskylä, FIN-40014, Finland
| | - Urho Kujala
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, Helsinki, Finland
| | - Anja Bye
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Elina Sillanpää
- Gerontology Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, P.O. Box 35 (VIV), Jyväskylä, FIN-40014, Finland.
- The Wellbeing Services County of Central Finland, Jyväskylä, Finland.
| |
Collapse
|
26
|
Ganji E, Lamia SN, Stepanovich M, Whyte N, Goulet RW, Abraham AC, Killian ML. Optogenetic-induced muscle loading leads to mechanical adaptation of the Achilles tendon enthesis in mice. SCIENCE ADVANCES 2023; 9:eadf4683. [PMID: 37352350 PMCID: PMC10289645 DOI: 10.1126/sciadv.adf4683] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/17/2023] [Indexed: 06/25/2023]
Abstract
Skeletal shape depends on the transmission of contractile muscle forces from tendon to bone across the enthesis. Loss of muscle loading impairs enthesis development, yet little is known if and how the postnatal enthesis adapts to increased loading. Here, we studied adaptations in enthesis structure and function in response to increased loading, using optogenetically induced muscle contraction in young (i.e., growth) and adult (i.e., mature) mice. Daily bouts of unilateral optogenetic loading in young mice led to radial calcaneal expansion and warping. This also led to a weaker enthesis with increased collagen damage in young tendon and enthisis, with little change in adult mice. We then used RNA sequencing to identify the pathways associated with increased mechanical loading during growth. In tendon, we found enrichment of glycolysis, focal adhesion, and cell-matrix interactions. In bone, we found enrichment of inflammation and cell cycle. Together, we demonstrate the utility of optogenetic-induced muscle contraction to elicit in vivo adaptation of the enthesis.
Collapse
Affiliation(s)
- Elahe Ganji
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, MI 48109, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 North Mathews Ave., Urbana, IL 61801, USA
- Department of Biomedical Engineering, University of Delaware, 540 S. College Ave., Newark, DE 19713, USA
| | - Syeda N. Lamia
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, MI 48109, USA
- Department of Mechanical Engineering, University of Michigan, 2350 Hayward St., Ann Arbor, MI 48109, USA
| | - Matthew Stepanovich
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, MI 48109, USA
| | - Noelle Whyte
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, MI 48109, USA
| | - Robert W. Goulet
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, MI 48109, USA
| | - Adam C. Abraham
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, MI 48109, USA
| | - Megan L. Killian
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Delaware, 540 S. College Ave., Newark, DE 19713, USA
| |
Collapse
|
27
|
Ganji E, Lamia SN, Stepanovich M, Whyte N, Abraham AC, Killian ML. Optogenetic-Induced Muscle Loading Leads to Mechanical Adaptation of the Achilles Tendon Enthesis in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.11.536376. [PMID: 37090593 PMCID: PMC10120626 DOI: 10.1101/2023.04.11.536376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
The growth of the skeleton depends on the transmission of contractile muscle forces from tendon to bone across the extracellular matrix-rich enthesis. Loss of muscle loading leads to significant impairments in enthesis development. However, little is known about how the enthesis responds to increased loading during postnatal growth. To study the cellular and matrix adaptations of the enthesis in response to increased muscle loading, we used optogenetics to induce skeletal muscle contraction and unilaterally load the Achilles tendon and enthesis in young (i.e., during growth) and adult (i.e., mature) mice. In young mice, daily bouts of unilateral optogenetic loading led to expansion of the calcaneal apophysis and growth plate, as well as increased vascularization of the normally avascular enthesis. Daily loading bouts, delivered for 3 weeks, also led to a mechanically weaker enthesis with increased molecular-level accumulation of collagen damage in young mice. However, adult mice did not exhibit impaired mechanical properties or noticeable structural adaptations to the enthesis. We then focused on the transcriptional response of the young tendon and bone following optogenetic-induced loading. After 1 or 2 weeks of loading, we identified, in tendon, transcriptional activation of canonical pathways related to glucose metabolism (glycolysis) and inhibited pathways associated with cytoskeletal remodeling (e.g., RHOA and CREB signaling). In bone, we identified activation of inflammatory signaling (e.g., NFkB and STAT3 signaling) and inhibition of ERK/MAPK and PTEN signaling. Thus, we have demonstrated the utility of optogenetic-induced skeletal muscle contraction to elicit structural, functional, and molecular adaptation of the enthesis in vivo especially during growth.
Collapse
Affiliation(s)
- Elahe Ganji
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, Michigan, 48109
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 North Mathews Ave., Urbana, Illinois, 61801
| | - Syeda N Lamia
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, Michigan, 48109
- Department of Mechanical Engineering, University of Michigan, 2350 Hayward St., Ann Arbor, Michigan, 48109
| | - Matthew Stepanovich
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, Michigan, 48109
| | - Noelle Whyte
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, Michigan, 48109
| | - Adam C Abraham
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, Michigan, 48109
| | - Megan L Killian
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, Michigan, 48109
- Department of Biomedical Engineering, University of Delaware, 540 S. College Ave., Newark, Delaware, 19713
| |
Collapse
|
28
|
Fleischman JY, Qi NR, Treutelaar MK, Britton SL, Koch LG, Li JZ, Burant CF. Intrinsic cardiorespiratory fitness modulates clinical and molecular response to caloric restriction. Mol Metab 2023; 68:101668. [PMID: 36642218 PMCID: PMC9938335 DOI: 10.1016/j.molmet.2023.101668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/21/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVE Caloric restriction (CR) is one extrinsic intervention that can improve metabolic health, and it shares many phenotypical parallels with intrinsic high cardiorespiratory fitness (CRF), including reduced adiposity, increased cardiometabolic health, and increased longevity. CRF is a highly heritable trait in humans and has been established in a genetic rat model selectively bred for high (HCR) and low (LCR) CRF, in which the HCR live longer and have reduced body weight compared to LCR. This study addresses whether the inherited high CRF phenotype occurs through similar mechanisms by which CR promotes health and longevity. METHODS We compared HCR and LCR male rats fed ad libitum (AL) or calorically restricted (CR) for multiple physiological, metabolic, and molecular traits, including running capacity at 2, 8, and 12 months; per-hour metabolic cage activity over daily cycles at 6 and 12 months; and plasma lipidomics, liver and muscle transcriptomics, and body composition after 12 months of treatment. RESULTS LCR-CR developed a physiological profile that mirrors the high-CRF phenotype in HCR-AL, including reduced adiposity and increased insulin sensitivity. HCR show higher spontaneous activity than LCR. Temporal modeling of hourly energy expenditure (EE) dynamics during the day, adjusted for body weight and hourly activity levels, suggest that CR has an EE-suppressing effect, and high-CRF has an EE-enhancing effect. Pathway analysis of gene transcripts indicates that HCR and LCR both show a response to CR that is similar in the muscle and different in the liver. CONCLUSIONS CR provides LCR a health-associated positive effect on physiological parameters that strongly resemble HCR. Analysis of whole-body EE and transcriptomics suggests that HCR and LCR show line-dependent responses to CR that may be accreditable to difference in genetic makeup. The results do not preclude the possibility that CRF and CR pathways may converge.
Collapse
Affiliation(s)
- Johanna Y Fleischman
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Nathan R Qi
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Michigan Mouse Metabolic Phenotyping Center, University of Michigan, Ann Arbor, MI, USA
| | - Mary K Treutelaar
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Steven L Britton
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA; Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - Lauren G Koch
- Department of Physiology and Pharmacology, The University of Toledo, Toledo, OH, USA
| | - Jun Z Li
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Charles F Burant
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
29
|
Stierwalt HD, Morris EM, Maurer A, Apte U, Phillips K, Li T, Meers GME, Koch LG, Britton SL, Graf G, Rector RS, Mercer K, Shankar K, Thyfault JP. Rats with high aerobic capacity display enhanced transcriptional adaptability and upregulation of bile acid metabolism in response to an acute high-fat diet. Physiol Rep 2022; 10:e15405. [PMID: 35923133 PMCID: PMC9350427 DOI: 10.14814/phy2.15405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 07/01/2022] [Accepted: 07/13/2022] [Indexed: 06/09/2023] Open
Abstract
Rats selectively bred for the high intrinsic aerobic capacity runner (HCR) or low aerobic capacity runner (LCR) show pronounced differences in susceptibility for high-fat/high sucrose (HFHS) diet-induced hepatic steatosis and insulin resistance, replicating the protective effect of high aerobic capacity in humans. We have previously shown multiple systemic differences in energy and substrate metabolism that impacts steatosis between HCR and LCR rats. This study aimed to investigate hepatic-specific mechanisms of action via changes in gene transcription. Livers of HCR rats had a greater number of genes that significantly changed in response to 3-day HFHS compared with LCR rats (171 vs. 75 genes: >1.5-fold, p < 0.05). HCR and LCR rats displayed numerous baseline differences in gene expression while on a low-fat control diet (CON). A 3-day HFHS diet resulted in greater expression of genes involved in the conversion of excess acetyl-CoA to cholesterol and bile acid (BA) synthesis compared with the CON diet in HCR, but not LCR rats. These results were associated with higher fecal BA loss and lower serum BA concentrations in HCR rats. Exercise studies in rats and mice also revealed higher hepatic expression of cholesterol and BA synthesis genes. Overall, these results suggest that high aerobic capacity and exercise are associated with upregulated BA synthesis paired with greater fecal excretion of cholesterol and BA, an effect that may play a role in protection against hepatic steatosis in rodents.
Collapse
Affiliation(s)
- Harrison D. Stierwalt
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
- Research ServiceKansas City VA Medical CenterKansas CityMissouriUSA
| | - E. Matthew Morris
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
| | - Adrianna Maurer
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
| | - Udayan Apte
- Department of Pharmacology, Toxicology, and TherapeuticsUniversity of Kansas Medical CenterKansas CityMissouriUSA
| | | | - Tiangang Li
- Department of PhysiologyUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| | - Grace M. E. Meers
- Division of Gastroenterology and HepatologyUniversity of MissouriColumbiaMissouriUSA
- Division of Nutrition and Exercise PhysiologyColumbiaMissouriUSA
| | - Lauren G. Koch
- Physiology and PharmacologyThe University of ToledoToledoOhioUSA
| | | | - Greg Graf
- Department of Pharmaceutical SciencesSaha Cardiovascular Research Center, University of KentuckyLexingtonKentuckyUSA
| | - R. Scott Rector
- Division of Gastroenterology and HepatologyUniversity of MissouriColumbiaMissouriUSA
- Division of Nutrition and Exercise PhysiologyColumbiaMissouriUSA
- Research ServiceHarry S Truman Memorial VA HospitalColumbiaMissouriUSA
| | - Kelly Mercer
- Arkansas Children's Nutrition CenterUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Kartik Shankar
- Section of Nutrition, Department of PediatricsUniversity of Colorado School of Medicine Anschutz Medical CampusAuroraColoradoUSA
| | - John P. Thyfault
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
- Research ServiceKansas City VA Medical CenterKansas CityMissouriUSA
| |
Collapse
|
30
|
Pantiya P, Thonusin C, Sumneang N, Ongnok B, Chunchai T, Kerdphoo S, Jaiwongkam T, Arunsak B, Siri-Angkul N, Sriwichaiin S, Chattipakorn N, Chattipakorn SC. High Cardiorespiratory Fitness Protects against Molecular Impairments of Metabolism, Heart, and Brain with Higher Efficacy in Obesity-Induced Premature Aging. Endocrinol Metab (Seoul) 2022; 37:630-640. [PMID: 35927067 PMCID: PMC9449107 DOI: 10.3803/enm.2022.1430] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 06/08/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGRUOUND High cardiorespiratory fitness (CRF) protects against age-related diseases. However, the mechanisms mediating the protective effect of high intrinsic CRF against metabolic, cardiac, and brain impairments in non-obese versus obese conditions remain incompletely understood. We aimed to identify the mechanisms through which high intrinsic CRF protects against metabolic, cardiac, and brain impairments in non-obese versus obese untrained rats. METHODS Seven-week-old male Wistar rats were divided into two groups (n=8 per group) to receive either a normal diet or a highfat diet (HFD). At weeks 12 and 28, CRF, carbohydrate and fatty acid oxidation, cardiac function, and metabolic parameters were evaluated. At week 28, behavior tests were performed. At the end of week 28, rats were euthanized to collect heart and brain samples for molecular studies. RESULTS The obese rats exhibited higher values for aging-related parameters than the non-obese rats, indicating that they experienced obesity-induced premature aging. High baseline CRF levels were positively correlated with several favorable metabolic, cardiac, and brain parameters at follow-up. Specifically, the protective effects of high CRF against metabolic, cardiac, and brain impairments were mediated by the modulation of body weight and composition, the lipid profile, substrate oxidation, mitochondrial function, insulin signaling, autophagy, apoptosis, inflammation, oxidative stress, cardiac function, neurogenesis, blood-brain barrier, synaptic function, accumulation of Alzheimer's disease-related proteins, and cognition. Interestingly, this effect was more obvious in HFD-fed rats. CONCLUSION The protective effect of high CRF is mediated by the modulation of several mechanisms. These effects exhibit greater efficacy under conditions of obesity-induced premature aging.
Collapse
Affiliation(s)
- Patcharapong Pantiya
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Chanisa Thonusin
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Natticha Sumneang
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Benjamin Ongnok
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Titikorn Chunchai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Sasiwan Kerdphoo
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Thidarat Jaiwongkam
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Busarin Arunsak
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Natthaphat Siri-Angkul
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Sirawit Sriwichaiin
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C. Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
- Corresponding author: Siriporn C. Chattipakorn. Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand Tel: +66-53-935329, Fax: +66-53-935368, E-mail:
| |
Collapse
|
31
|
Thonusin C, Pantiya P, Sumneang N, Chunchai T, Nawara W, Arunsak B, Siri-Angkul N, Sriwichaiin S, Chattipakorn SC, Chattipakorn N. Effectiveness of high cardiorespiratory fitness in cardiometabolic protection in prediabetic rats. Mol Med 2022; 28:31. [PMID: 35272616 PMCID: PMC8908596 DOI: 10.1186/s10020-022-00458-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/23/2022] [Indexed: 11/10/2022] Open
Abstract
Background Caloric restriction and exercise are lifestyle interventions that effectively attenuate cardiometabolic impairment. However, cardioprotective effects of long-term lifestyle interventions and short-term lifestyle interventions followed by weight maintenance in prediabetes have never been compared. High cardiorespiratory fitness (CRF) has been shown to provide protection against prediabetes and cardiovascular diseases, however, the interactions between CRF, prediabetes, caloric restriction, and exercise on cardiometabolic health has never been investigated. Methods Seven-week-old male Wistar rats were fed with either a normal diet (ND; n = 6) or a high-fat diet (HFD; n = 30) to induce prediabetes for 12 weeks. Baseline CRF and cardiometabolic parameters were determined at this timepoint. The ND-fed rats were fed continuously with a ND for 16 more weeks. The HFD-fed rats were divided into 5 groups (n = 6/group) to receive one of the following: (1) a HFD without any intervention for 16 weeks, (2) 40% caloric restriction for 6 weeks followed by an ad libitum ND for 10 weeks, (3) 40% caloric restriction for 16 weeks, (4) a HFD plus an exercise training program for 6 weeks followed by a ND without exercise for 10 weeks, or (5) a HFD plus an exercise training program for 16 weeks. At the end of the interventions, CRF and cardiometabolic parameters were re-assessed. Then, all rats were euthanized and heart tissues were collected. Results Either short-term caloric restriction or exercise followed by weight maintenance ameliorated cardiometabolic impairment in prediabetes, as indicated by increased insulin sensitivity, improved blood lipid profile, improved mitochondrial function and oxidative phosphorylation, reduced oxidative stress and inflammation, and improved cardiac function. However, these benefits were not as effective as those of either long-term caloric restriction or exercise. Interestingly, high-level baseline CRF was correlated with favorable cardiac and metabolic profiles at follow-up in prediabetic rats, both with and without lifestyle interventions. Conclusions Short-term lifestyle modification followed by weight maintenance improves cardiometabolic health in prediabetes. High CRF exerted protection against cardiometabolic impairment in prediabetes, both with and without lifestyle modification. These findings suggest that targeting the enhancement of CRF may contribute to the more effective treatment of prediabetes-induced cardiometabolic impairment. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00458-9.
Collapse
Affiliation(s)
- Chanisa Thonusin
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Patcharapong Pantiya
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Natticha Sumneang
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Titikorn Chunchai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Wichwara Nawara
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Busarin Arunsak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Natthaphat Siri-Angkul
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Sirawit Sriwichaiin
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand. .,Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand. .,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
32
|
Anhê FF, Zlitni S, Barra NG, Foley KP, Nilsson MI, Nederveen JP, Koch LG, Britton SL, Tarnopolsky MA, Schertzer JD. Life-long exercise training and inherited aerobic endurance capacity produce converging gut microbiome signatures in rodents. Physiol Rep 2022; 10:e15215. [PMID: 35246957 PMCID: PMC8897742 DOI: 10.14814/phy2.15215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 06/14/2023] Open
Abstract
High aerobic endurance capacity can be acquired by training and/or inherited. Aerobic exercise training (AET) and aging are linked to altered gut microbiome composition, but it is unknown if the environmental stress of exercise and host genetics that predispose for higher exercise capacity have similar effects on the gut microbiome during aging. We hypothesized that exercise training and host genetics would have conserved effects on the gut microbiome across different rodents. We studied young sedentary (Y-SED, 2-month-old) mice, old sedentary (O-SED, 26-month-old) mice, old mice with life-long AET (O-AET, 26-month-old), and aged rats selectively bred for high (HCR [High Capacity Runner], 21-month-old) and low (LCR [Low Capacity Runner], 21-month-old) aerobic capacity. Our results showed that O-SED mice had lower running capacity than Y-SED mice. The fecal microbiota of O-SED mice had a higher relative abundance of Lachnospiraceae, Ruminococcaceae, Turicibacteriaceae, and Allobaculum, but lower Bacteroidales, Alistipes, Akkermansia, and Anaeroplasma. O-AET mice had a higher running capacity than O-SED mice. O-AET mice had lower fecal levels of Lachnospiraceae, Turicibacteriaceae, and Allobaculum and higher Anaeroplasma than O-SED mice. Similar to O-AET mice, but despite no exercise training regime, aged HCR rats had lower Lachnospiraceae and Ruminococcaceae and expansion of certain Bacteroidales in the fecal microbiome compared to LCR rats. Our data show that environmental and genetic modifiers of high aerobic endurance capacity produce convergent gut microbiome signatures across different rodent species during aging. Therefore, we conclude that host genetics and life-long exercise influence the composition of the gut microbiome and can mitigate gut dysbiosis and functional decline during aging.
Collapse
Affiliation(s)
- Fernando F. Anhê
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonOntarioCanada
- Farncombe Family Digestive Health Research InstituteMcMaster UniversityHamiltonOntarioCanada
- Centre for Metabolism, Obesity and Diabetes ResearchMcMaster UniversityHamiltonOntarioCanada
| | - Soumaya Zlitni
- Departments of Genetics and MedicineStanford UniversityStanfordCaliforniaUSA
| | - Nicole G. Barra
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonOntarioCanada
- Farncombe Family Digestive Health Research InstituteMcMaster UniversityHamiltonOntarioCanada
- Centre for Metabolism, Obesity and Diabetes ResearchMcMaster UniversityHamiltonOntarioCanada
| | - Kevin P. Foley
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonOntarioCanada
- Farncombe Family Digestive Health Research InstituteMcMaster UniversityHamiltonOntarioCanada
- Centre for Metabolism, Obesity and Diabetes ResearchMcMaster UniversityHamiltonOntarioCanada
| | - Mats I. Nilsson
- Department of PediatricsMcMaster UniversityHamiltonOntarioCanada
| | | | - Lauren G. Koch
- Department of Physiology and PharmacologyThe University of ToledoCollege of Medicine and Life SciencesToledoOhioUSA
| | - Steven L. Britton
- Department of AnesthesiologyUniversity of MichiganAnn ArborMichiganUnited States
| | - Mark A. Tarnopolsky
- Department of PediatricsMcMaster UniversityHamiltonOntarioCanada
- Department of MedicineMcMaster UniversityHamiltonOntarioCanada
| | - Jonathan D. Schertzer
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonOntarioCanada
- Farncombe Family Digestive Health Research InstituteMcMaster UniversityHamiltonOntarioCanada
- Centre for Metabolism, Obesity and Diabetes ResearchMcMaster UniversityHamiltonOntarioCanada
| |
Collapse
|
33
|
Biro PA, Thomas F, Ujvari B, Beckmann C. A novel perspective suggesting high sustained energy expenditure may be net protective against cancer. Evol Med Public Health 2022; 10:170-176. [PMID: 35498120 PMCID: PMC9040660 DOI: 10.1093/emph/eoac012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 04/01/2022] [Indexed: 11/14/2022] Open
Abstract
Abstract
Energy expenditure (EE) is generally viewed as tumorigenic, due to production of reactive oxygen species (ROS) that can damage cells and DNA. On this basis, individuals within a species that sustain high EE should be more likely to develop cancer. Here, we argue the opposite, that high EE may be net protective effect against cancer, despite high ROS production. This is possible because individuals that sustain high EE have a greater energetic capacity (=greater energy acquisition, expenditure and ability to up-regulate output), and can therefore allocate energy to multiple cancer-fighting mechanisms with minimal energetic trade-offs. Our review finds that individuals sustaining high EE have greater antioxidant production, lower oxidative stress, greater immune function and lower cancer incidence. Our hypothesis and literature review suggest that EE may indeed be net protective against cancer, and that individual variation in energetic capacity may be a key mechanism to understand the highly individual nature of cancer risk in contemporary human populations and laboratory animals.
Lay summary The process of expending energy generates reactive oxygen species that can lead to oxidative stress, cell and DNA damage, and the accumulation of this damage is thought to be a major contributor to many ageing related diseases that include cancer. Here, we challenge this view, proposing how and why high energy expenditure (EE) may actually be net protective against cancer, and provide literature support for our hypothesis. We find individuals with high sustained EE have greater energetic capacity and thus can invest more in repair to counter oxidative stress, and more in immune function, both of which reduce cancer risk. Our hypothesis provides a novel mechanism to understand the highly individual nature of cancer, why taller individuals are more at risk, why physically active individuals have lower cancer risk, and why regular exercise can reduce cancer risk.
Collapse
Affiliation(s)
- Peter A Biro
- Centre for Integrative Ecology, School of Life and Environmental Science, Deakin University, Geelong 3216, Australia
- Corresponding author. Centre for Integrative Ecology, School of Life and Environmental Science, Deakin University, Geelong 3216, Australia. Tel: +61 434 8569 921; E-mail:
| | - Frédéric Thomas
- CREEC, UMR IRD/CNRS/UM 5290, 911 Avenue Agropolis, BP 64501, 34394 Montpellier Cedex 5, France
| | - Beata Ujvari
- Centre for Integrative Ecology, School of Life and Environmental Science, Deakin University, Geelong 3216, Australia
| | - Christa Beckmann
- Centre for Integrative Ecology, School of Life and Environmental Science, Deakin University, Geelong 3216, Australia
- School of Science, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751, Australia
- Hawkesbury Institute for the Environment, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751, Australia
| |
Collapse
|
34
|
The Effect of Age and Intrinsic Aerobic Exercise Capacity on the Expression of Inflammation and Remodeling Markers in Rat Achilles Tendons. Int J Mol Sci 2021; 23:ijms23010079. [PMID: 35008516 PMCID: PMC8744822 DOI: 10.3390/ijms23010079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 12/19/2022] Open
Abstract
Old age, adiposity, and metabolic disorders are known as risk factors for chronic tendinopathy, which is a common problem in both athletes and the general population. However, the importance of these influencing factors has not yet been well understood. This study investigated alterations in gene expression and histology of Achilles tendons of young (10 weeks) and old (100 weeks) rats bred for low (low capacity runners, LCR) and high (high capacity runners, HCR) intrinsic aerobic exercise capacity. In this rat model, LCR displayed a phenotype of reduced exercise capacity, higher body weight, and metabolic dysfunctions compared to HCR. We hypothesized that the risk factors for tendinopathy in old LCR could lead to more pronounced impairments in Achilles tendon tissue. In quantitative real-time PCR (qPCR), age-related downregulation of tenocyte markers e.g., tenomodulin, genes related to matrix modeling and remodeling (e.g., collagens, elastin, biglycan, fibronectin, tenascin C) as well as transforming growth factor beta 3 (Tgfb3) have been detected. Inflammation marker cyclooxygenase 2 (Cox2) was downregulated in old rats, while microsomal prostaglandin E synthase 2 (Ptges2) was upregulated in old HCR and old LCR. In all groups, interleukin 6 (Il6), interleukin 1 beta (Il1b), and tumor necrosis factor alpha (Tnfa) showed no significant alteration. In histological evaluation, tendons of old rats had fewer and more elongated tenocyte nuclei than young rats. Even though a higher content of glycosaminoglycans, a sign of degeneration, was found in old HCR and LCR, no further signs of tendinopathy were detectable in tendons of old rats by histological evaluation. Low intrinsic aerobic exercise capacity and the associated phenotype did not show significant effects on gene expression and tendon histology. These findings indicate that aging seems to play a prominent role in molecular and structural alterations of Achilles tendon tissue and suggests that other risk factors associated with intrinsic aerobic exercise capacity are less influential in this rat model.
Collapse
|
35
|
Alsahly MB, Zakari MO, Koch LG, Britton S, Katwa LC, Lust RM. Influence of Intrinsic Aerobic Exercise Capacity and Sex on Cardiac Injury Following Acute Myocardial Ischemia and Reperfusion. Front Cardiovasc Med 2021; 8:751864. [PMID: 34901212 PMCID: PMC8661003 DOI: 10.3389/fcvm.2021.751864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Purpose: Previous reports have suggested that active exercise aside, intrinsic aerobic running capacity (Low = LCR, high = HCR) in otherwise sedentary animals may influence several cardiovascular health-related indicators. Relative to the HCR phenotype, the LCR phenotype is characterized by decreased endothelial reactivity, increased susceptibility to reperfusion-induced arrhythmias following short, non-infarction ischemia, and increased diet-induced insulin resistance. More broadly, the LCR phenotype has come to be characterized as a "disease prone" model, with the HCRs as "disease resistant." Whether these effects extend to injury outcomes in an overt infarction or whether the effects are gender specific is not known. This study was designed to determine whether HCR/LCR phenotypic differences would be evident in injury responses to acute myocardial ischemia-reperfusion injury (AIR), measured as infarct size and to determine whether sex differences in infarction size were preserved with phenotypic selection. Methods: Regional myocardial AIR was induced in vivo by either 15 or 30 min ligation of the left anterior descending coronary artery, followed by 2 h of reperfusion. Global ischemia was induced in isolated hearts ex vivo using a Langendorff perfusion system and cessation of perfusion for either 15 or 30 min followed by 2 h of reperfusion. Infarct size was determined using 2, 3, 5-triphenyltetrazolium chloride (TTC) staining, and normalized to area at risk in the regional model, or whole heart in the global model. Portions of the tissue were paraffin embedded for H&E staining and histology analysis. Results: Phenotype dependent differences in infarct size were seen with 15 min occlusion/2 h reperfusion (LCR > HCR, p < 0.05) in both regional and global models. In both models, longer occlusion times (30 min/2 h) produced significantly larger infarctions in both phenotypes, but phenotypic differences were no longer present (LCR vs. HCR, p = n.s.). Sex differences in infarct size were present in each phenotype (LCR male > LCR female, p < 0.05; HCR male > HCR female, p < 0.05 regardless of length of occlusion, or ischemia model. Conclusions: There is cardioprotection afforded by high intrinsic aerobic capacity, but it is not infinite/continuous, and may be overcome with sufficient injury burden. Phenotypic selection based on endurance running capacity preserved sex differences in response to both short and longer term coronary occlusive challenges. Outcomes could not be associated with differences in system characteristics such as circulating inflammatory mediators or autonomic nervous system influences, as similar phenotypic injury patterns were seen in vivo, and in isolated crystalloid perfused heart ex vivo.
Collapse
Affiliation(s)
- Musaad B Alsahly
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States.,Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Madaniah O Zakari
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States.,Department of Physiology, College of Medicine, Taibah University, Medina, Saudi Arabia
| | - Lauren G Koch
- Department of Physiology and Pharmacology, University of Toledo, Toledo, OH, United States
| | - Steven Britton
- Departments of Anesthesiology and Molecular and Integrative Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Laxmansa C Katwa
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Robert M Lust
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States.,East Carolina Diabetes and Obesity Center, East Carolina University, Greenville, NC, United States
| |
Collapse
|
36
|
Zhuang H, Karvinen S, Törmäkangas T, Zhang X, Ojanen X, Velagapudi V, Alen M, Britton SL, Koch LG, Kainulainen H, Cheng S, Wiklund P. Interactive effects of aging and aerobic capacity on energy metabolism-related metabolites of serum, skeletal muscle, and white adipose tissue. GeroScience 2021; 43:2679-2691. [PMID: 34089174 PMCID: PMC8602622 DOI: 10.1007/s11357-021-00387-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 05/17/2021] [Indexed: 12/25/2022] Open
Abstract
Aerobic capacity is a strong predictor of longevity. With aging, aerobic capacity decreases concomitantly with changes in whole body metabolism leading to increased disease risk. To address the role of aerobic capacity, aging, and their interaction on metabolism, we utilized rat models selectively bred for low and high intrinsic aerobic capacity (LCRs/HCRs) and compared the metabolomics of serum, muscle, and white adipose tissue (WAT) at two time points: Young rats were sacrificed at 9 months of age, and old rats were sacrificed at 21 months of age. Targeted and semi-quantitative metabolomics analysis was performed on the ultra-pressure liquid chromatography tandem mass spectrometry (UPLC-MS) platform. The effects of aerobic capacity, aging, and their interaction were studied via regression analysis. Our results showed that high aerobic capacity is associated with an accumulation of isovalerylcarnitine in muscle and serum at rest, which is likely due to more efficient leucine catabolism in muscle. With aging, several amino acids were downregulated in muscle, indicating more efficient amino acid metabolism, whereas in WAT less efficient amino acid metabolism and decreased mitochondrial β-oxidation were observed. Our results further revealed that high aerobic capacity and aging interactively affect lipid metabolism in muscle and WAT, possibly combating unfavorable aging-related changes in whole body metabolism. Our results highlight the significant role of WAT metabolism for healthy aging.
Collapse
Affiliation(s)
- Haihui Zhuang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Systems Biomedicine (Ministry of Education), and Exercise Translational Medicine Center, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Sira Karvinen
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland.
| | - Timo Törmäkangas
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Xiaobo Zhang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Systems Biomedicine (Ministry of Education), and Exercise Translational Medicine Center, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaowei Ojanen
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Vidya Velagapudi
- Metabolomics Unit, Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Markku Alen
- Department of Medical Rehabilitation, Oulu University Hospital, Oulu, Finland
| | - Steven L Britton
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Lauren G Koch
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Heikki Kainulainen
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Sulin Cheng
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Systems Biomedicine (Ministry of Education), and Exercise Translational Medicine Center, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Petri Wiklund
- Key Laboratory of Systems Biomedicine (Ministry of Education), and Exercise Translational Medicine Center, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
- Huawei Helsinki Research Center, Huawei Technologies Oy (Finland) Co. Ltd, Helsinki, Finland
| |
Collapse
|
37
|
Schwarzer M, Molis A, Schenkl C, Schrepper A, Britton SL, Koch LG, Doenst T. Genetically determined exercise capacity affects systemic glucose response to insulin in rats. Physiol Genomics 2021; 53:395-405. [PMID: 34297615 DOI: 10.1152/physiolgenomics.00014.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION Aerobic exercise capacity is inversely related to morbidity and mortality as well as to insulin resistance. However, exercising in patients has led to conflicting results, presumably because aerobic exercise capacity consists of intrinsic (genetically determined) and extrinsic (environmentally determined) parts. The contribution of both parts to insulin sensitivity is also not clear. We investigated sedentary and exercised (aerobic interval training) high (HCR) and low capacity runners (LCR) differing in their genetically determined aerobic exercise capacity to determine the contribution of both parts to insulin sensitivity. METHODS AND RESULTS LCR and HCR differed in their untrained exercise capacity and body weight. Sedentary LCR displayed a diabetic phenotype with higher random glucose, lower glucose infusion rate during hyperinsulinemic euglycemic clamping than HCR. Echocardiography showed equal morphological and functional parameters and no change with exercise. Four weeks of exercise caused significant improvements in aerobic exercise capacity, which was more pronounced in LCR. However, with respect to glucose use, exercise affected HCR only. In these animals, exercise increased 2-deoxyglucose uptake in gastrocnemius (+58.5 %, p= 0.1) and in epididymal fat (+106 %; p<0.05). Citrate synthase activity also increased in these tissues (gastrocnemius 69 % epididymal fat 63 %). CONCLUSION In our model of HCR and LCR, genetic predisposition for low exercise capacity is associated with impaired insulin sensitivity and impedes exercise-induced improvements in insulin response. Our results suggest that genetic predisposition for low aerobic exercise capacity impairs insulin response, which may not be overcome by exercise.
Collapse
Affiliation(s)
- Michael Schwarzer
- Department of Cardiothoracic Surgery, Jena University Hospital, Jena, Germany
| | - Annika Molis
- Department of Cardiothoracic Surgery, Jena University Hospital, Jena, Germany
| | - Christina Schenkl
- Department of Cardiothoracic Surgery, Jena University Hospital, Jena, Germany
| | - Andrea Schrepper
- Department of Cardiothoracic Surgery, Jena University Hospital, Jena, Germany
| | - Steven L Britton
- Department of Anesthesiology, Department of Molecular and Integrative Physiology, University of Michigan-Ann Arbor, Ann Arbor, Michigan, United States
| | - Lauren Gerard Koch
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, United States
| | - Torsten Doenst
- Department of Cardiothoracic Surgery, Jena University Hospital, Jena, Germany
| |
Collapse
|
38
|
Intrinsic exercise capacity induces divergent vascular plasticity via arachidonic acid-mediated inflammatory pathways in female rats. Vascul Pharmacol 2021; 140:106862. [PMID: 33872803 DOI: 10.1016/j.vph.2021.106862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/03/2021] [Accepted: 04/07/2021] [Indexed: 12/31/2022]
Abstract
Metabolic syndrome prevalence has increased among US adults, particularly among non-hispanic white and black women. Sedentary behavior often leads to chronic inflammation, a triggering factor of metabolic syndrome. Given that intrinsic exercise capacity is genetically inherited, we questioned if low-grade chronic inflammation would be present in a female rat model of low intrinsic exercise capacity-induced metabolic syndrome, while beneficial increase of resolution of inflammation would be present in a female rat model of high intrinsic exercise capacity. In the vascular system, two primary markers for inflammation and resolution of inflammation are cyclooxygenase (COX) and lipoxygenase (LOX), respectively. Our study focused on the novel hypothesis that untrained, inherited exercise capacity induces divergent vascular plasticity via changes in the delicate balance between COX and LOX inflammatory mediators. We used divergent rat strains with low (LCR) and high (HCR) aerobic running capacity. By using animals with contrasting intrinsic exercise capacities, it is possible to determine the exact triggers that lead to inherited vascular plasticity in female rats. We observed that female LCR displayed increased periovarian fat pad and body weight, which is congruent with their obesity-presenting phenotype. Furthermore, LCR presented with vascular hypocontractility and increased COX and LOX-derived pro-inflammatory factors. On the other hand, HCR presented with a "shutdown" of COX-induced vasoconstriction and enhanced resolution of inflammation to maintain vascular tone and homeostasis. In conclusion, LCR display low-grade chronic inflammation via increased COX activity. These results provide mechanistic clues as to why lower intrinsic aerobic capacity correlates with a predisposition to risk of vascular disease. Conversely, being born with higher intrinsic aerobic capacity is a significant factor for improved vascular physiology in female rats.
Collapse
|
39
|
Pataky MW, Young WF, Nair KS. Hormonal and Metabolic Changes of Aging and the Influence of Lifestyle Modifications. Mayo Clin Proc 2021; 96:788-814. [PMID: 33673927 PMCID: PMC8020896 DOI: 10.1016/j.mayocp.2020.07.033] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/01/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023]
Abstract
Increased life expectancy combined with the aging baby boomer generation has resulted in an unprecedented global expansion of the elderly population. The growing population of older adults and increased rate of age-related chronic illness has caused a substantial socioeconomic burden. The gradual and progressive age-related decline in hormone production and action has a detrimental impact on human health by increasing risk for chronic disease and reducing life span. This article reviews the age-related decline in hormone production, as well as age-related biochemical and body composition changes that reduce the bioavailability and actions of some hormones. The impact of hormonal changes on various chronic conditions including frailty, diabetes, cardiovascular disease, and dementia are also discussed. Hormone replacement therapy has been attempted in many clinical trials to reverse and/or prevent the hormonal decline in aging to combat the progression of age-related diseases. Unfortunately, hormone replacement therapy is not a panacea, as it often results in various adverse events that outweigh its potential health benefits. Therefore, except in some specific individual cases, hormone replacement is not recommended. Rather, positive lifestyle modifications such as regular aerobic and resistance exercise programs and/or healthy calorically restricted diet can favorably affect endocrine and metabolic functions and act as countermeasures to various age-related diseases. We provide a critical review of the available data and offer recommendations that hopefully will form the groundwork for physicians/scientists to develop and optimize new endocrine-targeted therapies and lifestyle modifications that can better address age-related decline in heath.
Collapse
Affiliation(s)
- Mark W Pataky
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, MN
| | - William F Young
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, MN
| | - K Sreekumaran Nair
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, MN.
| |
Collapse
|
40
|
Hesketh SJ, Stansfield BN, Stead CA, Burniston JG. The application of proteomics in muscle exercise physiology. Expert Rev Proteomics 2021; 17:813-825. [PMID: 33470862 DOI: 10.1080/14789450.2020.1879647] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Exercise offers protection from non-communicable diseases and extends healthspan by offsetting natural physiological declines that occur in older age. Striated muscle is the largest bodily organ; it underpins the capacity for physical work, and the responses of muscle to exercise convey the health benefits of a physically active lifestyle. Proteomic surveys of muscle provide a means to study the protective effects of exercise and this review summaries some key findings from literature listed in PubMed during the last 10 years that have led to new insight in muscle exercise physiology. AREAS COVERED 'Bottom-up' analyses involving liquid-chromatography tandem mass spectrometry (LC-MS/MS) of peptide digests have become the mainstay of proteomic studies and have been applied to muscle mitochondrial fractions. Enrichment techniques for post-translational modifications, including phosphorylation, acetylation and ubiquitination, have evolved and the analysis of site-specific modifications has become a major area of interest in exercise proteomics. Finally, we consider emergent techniques for dynamic analysis of muscle proteomes that offer new insight to protein turnover and the contributions of synthesis and degradation to changes in protein abundance in response to exercise training. EXPERT OPINION Burgeoning methods for dynamic proteome profiling offer new opportunities to study the mechanisms of muscle adaptation.
Collapse
Affiliation(s)
- Stuart J Hesketh
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University , Liverpool, UK
| | - Ben N Stansfield
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University , Liverpool, UK
| | - Connor A Stead
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University , Liverpool, UK
| | - Jatin G Burniston
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University , Liverpool, UK
| |
Collapse
|
41
|
Mitochondrial health is enhanced in rats with higher vs. lower intrinsic exercise capacity and extended lifespan. NPJ Aging Mech Dis 2021; 7:1. [PMID: 33398019 PMCID: PMC7782588 DOI: 10.1038/s41514-020-00054-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 11/24/2020] [Indexed: 12/03/2022] Open
Abstract
The intrinsic aerobic capacity of an organism is thought to play a role in aging and longevity. Maximal respiratory rate capacity, a metabolic performance measure, is one of the best predictors of cardiovascular- and all-cause mortality. Rats selectively bred for high-(HCR) vs. low-(LCR) intrinsic running-endurance capacity have up to 31% longer lifespan. We found that positive changes in indices of mitochondrial health in cardiomyocytes (respiratory reserve, maximal respiratory capacity, resistance to mitochondrial permeability transition, autophagy/mitophagy, and higher lipids-over-glucose utilization) are uniformly associated with the extended longevity in HCR vs. LCR female rats. Cross-sectional heart metabolomics revealed pathways from lipid metabolism in the heart, which were significantly enriched by a select group of strain-dependent metabolites, consistent with enhanced lipids utilization by HCR cardiomyocytes. Heart–liver–serum metabolomics further revealed shunting of lipidic substrates between the liver and heart via serum during aging. Thus, mitochondrial health in cardiomyocytes is associated with extended longevity in rats with higher intrinsic exercise capacity and, probably, these findings can be translated to other populations as predictors of outcomes of health and survival.
Collapse
|
42
|
Roy S, Edwards JM, Tomcho JC, Schreckenberger Z, Bearss NR, Zhang Y, Morgan EE, Cheng X, Spegele AC, Vijay-Kumar M, McCarthy CG, Koch LG, Joe B, Wenceslau CF. Intrinsic Exercise Capacity and Mitochondrial DNA Lead to Opposing Vascular-Associated Risks. FUNCTION (OXFORD, ENGLAND) 2020; 2:zqaa029. [PMID: 33363281 PMCID: PMC7749784 DOI: 10.1093/function/zqaa029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 01/06/2023]
Abstract
Exercise capacity is a strong predictor of all-cause morbidity and mortality in humans. However, the associated hemodynamic traits that link this valuable indicator to its subsequent disease risks are numerable. Additionally, exercise capacity has a substantial heritable component and genome-wide screening indicates a vast amount of nuclear and mitochondrial DNA (mtDNA) markers are significantly associated with traits of physical performance. A long-term selection experiment in rats confirms a divide for cardiovascular risks between low- and high-capacity runners (LCR and HCR, respectively), equipping us with a preclinical animal model to uncover new mechanisms. Here, we evaluated the LCR and HCR rat model system for differences in vascular function at the arterial resistance level. Consistent with the known divide between health and disease, we observed that LCR rats present with resistance artery and perivascular adipose tissue dysfunction compared to HCR rats that mimic qualities important for health, including improved vascular relaxation. Uniquely, we show by generating conplastic strains, which LCR males with mtDNA of female HCR (LCR-mtHCR/Tol) present with improved vascular function. Conversely, HCR-mtLCR/Tol rats displayed indices for cardiac dysfunction. The outcome of this study suggests that the interplay between the nuclear genome and the maternally inherited mitochondrial genome with high intrinsic exercise capacity is a significant factor for improved vascular physiology, and animal models developed on an interaction between nuclear and mtDNA are valuable new tools for probing vascular risk factors in the offspring.
Collapse
Affiliation(s)
- Shaunak Roy
- Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences
| | - Jonnelle M Edwards
- Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences
| | - Jeremy C Tomcho
- Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences
| | - Zachary Schreckenberger
- Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences
| | - Nicole R Bearss
- Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences
| | - Youjie Zhang
- Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences
| | - Eric E Morgan
- Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences,Department of Radiology Nationwide Children's Hospital, OH, USA
| | - Xi Cheng
- Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences
| | - Adam C Spegele
- Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences
| | - Matam Vijay-Kumar
- Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences
| | - Cameron G McCarthy
- Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences
| | - Lauren G Koch
- Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences
| | - Bina Joe
- Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences
| | - Camilla Ferreira Wenceslau
- Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences,Address correspondence to C.F.W. (e-mail: )
| |
Collapse
|
43
|
Koch LG, Britton SL. Aerobics - decades of data for future hypothesis-testing research. Nat Rev Endocrinol 2020; 16:627-628. [PMID: 32895502 DOI: 10.1038/s41574-020-00415-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Lauren Gerard Koch
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA.
| | - Steven Loyal Britton
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
44
|
Hjortbak MV, Grønnebæk TS, Jespersen NR, Lassen TR, Seefeldt JM, Tonnesen PT, Jensen RV, Koch LG, Britton SL, Pedersen M, Jessen N, Bøtker HE. Differences in intrinsic aerobic capacity alters sensitivity to ischemia-reperfusion injury but not cardioprotective capacity by ischemic preconditioning in rats. PLoS One 2020; 15:e0240866. [PMID: 33108389 PMCID: PMC7591019 DOI: 10.1371/journal.pone.0240866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/03/2020] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Aerobic capacity is a strong predictor of cardiovascular mortality. Whether aerobic capacity influences myocardial ischemia and reperfusion (IR) injury is unknown. PURPOSE To investigate the impact of intrinsic differences in aerobic capacity and the cardioprotective potential on IR injury. METHODS We studied hearts from rats developed by selective breeding for high (HCR) or low (LCR) capacity for treadmill running. The rats were randomized to: (1) control, (2) local ischemic preconditioning (IPC) or (3) remote ischemic preconditioning (RIC) followed by 30 minutes of ischemia and 120 minutes of reperfusion in an isolated perfused heart model. The primary endpoint was infarct size. Secondary endpoints included uptake of labelled glucose, content of selected mitochondrial proteins in skeletal and cardiac muscle, and activation of AMP-activated kinase (AMPK). RESULTS At baseline, running distance was 203±7 m in LCR vs 1905±51 m in HCR rats (p<0.01). Infarct size was significantly lower in LCR than in HCR controls (49±5% vs 68±5%, p = 0.04). IPC reduced infarct size by 47% in LCR (p<0.01) and by 31% in HCR rats (p = 0.01). RIC did not modulate infarct size (LCR: 52±5, p>0.99; HCR: 69±6%, p>0.99, respectively). Phosphorylaion of AMPK did not differ between LCR and HCR controls. IPC did not modulate cardiac phosphorylation of AMPK. Glucose uptake during reperfusion was similar in LCR and HCR rats. IPC increased glucose uptake during reperfusion in LCR animals (p = 0.02). Mitochondrial protein content in skeletal muscle was lower in LCR than in HCR (0.77±0.10 arbitrary units (AU) vs 1.09±0.07 AU, p = 0.02), but not in cardiac muscle. CONCLUSION Aerobic capacity is associated with altered myocardial sensitivity to IR injury, but the cardioprotective effect of IPC is not. Glucose uptake, AMPK activation immediately prior to ischemia and basal mitochondrial protein content in the heart seem to be of minor importance as underlying mechanisms for the cardioprotective effects.
Collapse
Affiliation(s)
- Marie Vognstoft Hjortbak
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- * E-mail:
| | | | - Nichlas Riise Jespersen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Thomas Ravn Lassen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jacob Marthinsen Seefeldt
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Pernille Tilma Tonnesen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Rebekka Vibjerg Jensen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Lauren Gerard Koch
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, United States of America
| | - Steven L. Britton
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Michael Pedersen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Niels Jessen
- Steno Diabetes Center Aarhus, Aahus University Hospital, Aarhus, Denmark
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
45
|
Mukherjee SD, Koch LG, Britton SL, Novak CM. Aerobic capacity modulates adaptive thermogenesis: Contribution of non-resting energy expenditure. Physiol Behav 2020; 225:113048. [PMID: 32628949 PMCID: PMC7594631 DOI: 10.1016/j.physbeh.2020.113048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/17/2020] [Accepted: 07/02/2020] [Indexed: 11/19/2022]
Abstract
Decreases in energy stores requires negative energy balance where caloric expenditure exceeds energy intake, which can induce adaptive thermogenesis-the reduction of energy expenditure (EE) beyond that accounted for by the weight lost. Adaptive thermogenesis varies between individuals. The component of total daily EE responsible for the interindividual variation in adaptive thermogenesis was investigated in this study, using a rat model that differs in obesity propensity and physical activity. Total daily EE and physical activity were examined before and after 21 days of 50% calorie restriction in male and female rats with lean and obesity-prone phenotypes-rats selectively bred for high and low intrinsic aerobic capacity (HCR and LCR, respectively). Calorie restriction significantly decreased EE more than was predicted by loss of weight and lean mass, demonstrating adaptive thermogenesis. Within sex, HCR and LCR did not significantly differ in resting EE. However, the calorie restriction-induced suppression in non-resting EE, which includes activity EE, was significantly greater in HCR than in LCR; this phenotypic difference was significant for both male and female rats. Calorie restriction also significantly suppressed physical activity levels more in HCR than LCR. When VO2max was assessed in male rats, calorie restriction significantly decreased O2 consumption without significantly affecting running performance (running time, distance), indicating increased energy efficiency. Percent weight loss did not significantly differ between groups. Altogether, these results suggest that individual differences in calorie restriction-induced adaptive thermogenesis may be accounted for by variation in aerobic capacity. Moreover, it is likely that activity EE, not resting or basal metabolism, may explain or predict the variation in individuals' adaptive thermogenesis.
Collapse
Affiliation(s)
- Sromona Dudiki Mukherjee
- Department of Biological Sciences; Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio, United States.
| | - Lauren G Koch
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, United States
| | - Steven L Britton
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, United States; Department of Anesthesiology, and Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Colleen M Novak
- Department of Biological Sciences; School of Biomedical Sciences, Kent State University, Kent, Ohio, United States
| |
Collapse
|
46
|
Riddle NC. Variation in the response to exercise stimulation in Drosophila: marathon runner versus sprinter genotypes. J Exp Biol 2020; 223:jeb229997. [PMID: 32737212 DOI: 10.1242/jeb.229997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/27/2020] [Indexed: 12/13/2022]
Abstract
Animals' behaviors vary in response to their environment, both biotic and abiotic. These behavioral responses have significant impacts on animal survival and fitness, and thus, many behavioral responses are at least partially under genetic control. In Drosophila, for example, genes impacting aggression, courtship behavior, circadian rhythms and sleep have been identified. Animal activity also is influenced strongly by genetics. My lab previously has used the Drosophila melanogaster Genetics Reference Panel (DGRP) to investigate activity levels and identified over 100 genes linked to activity. Here, I re-examined these data to determine whether Drosophila strains differ in their response to rotational exercise stimulation, not simply in the amount of activity, but in activity patterns and timing of activity. Specifically, I asked whether there are fly strains exhibiting either a 'marathoner' pattern of activity, i.e. remaining active throughout the 2 h exercise period, or a 'sprinter' pattern, i.e. carrying out most of the activity early in the exercise period. The DGRP strains examined differ significantly in how much activity is carried out at the beginning of the exercise period, and this pattern is influenced by both sex and genotype. Interestingly, there was no clear link between the activity response pattern and lifespan of the animals. Using genome-wide association studies (GWAS), I identified 10 high confidence candidate genes that control the degree to which Drosophila exercise behaviors fit a marathoner or sprinter activity pattern. This finding suggests that, similar to other aspects of locomotor behavior, the timing of activity patterns in response to exercise stimulation is under genetic control.
Collapse
Affiliation(s)
- Nicole C Riddle
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
47
|
Hinds TD, Creeden JF, Gordon DM, Spegele AC, Britton SL, Koch LG, Stec DE. Rats Genetically Selected for High Aerobic Exercise Capacity Have Elevated Plasma Bilirubin by Upregulation of Hepatic Biliverdin Reductase-A (BVRA) and Suppression of UGT1A1. Antioxidants (Basel) 2020; 9:antiox9090889. [PMID: 32961782 PMCID: PMC7554716 DOI: 10.3390/antiox9090889] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/12/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022] Open
Abstract
Exercise in humans and animals increases plasma bilirubin levels, but the mechanism by which this occurs is unknown. In the present study, we utilized rats genetically selected for high capacity running (HCR) and low capacity running (LCR) to determine pathways in the liver that aerobic exercise modifies to control plasma bilirubin. The HCR rats, compared to the LCR, exhibited significantly higher levels of plasma bilirubin and the hepatic enzyme that produces it, biliverdin reductase-A (BVRA). The HCR also had reduced expression of the glucuronyl hepatic enzyme UGT1A1, which lowers plasma bilirubin. Recently, bilirubin has been shown to activate the peroxisome proliferator-activated receptor-α (PPARα), a ligand-induced transcription factor, and the higher bilirubin HCR rats had significantly increased PPARα-target genes Fgf21, Abcd3, and Gys2. These are known to promote liver function and glycogen storage, which we found by Periodic acid–Schiff (PAS) staining that hepatic glycogen content was higher in the HCR versus the LCR. Our results demonstrate that exercise stimulates pathways that raise plasma bilirubin through alterations in hepatic enzymes involved in bilirubin synthesis and metabolism, improving liver function, and glycogen content. These mechanisms may explain the beneficial effects of exercise on plasma bilirubin levels and health in humans.
Collapse
Affiliation(s)
- Terry D. Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40508, USA
- Correspondence: (T.D.H.J.); (D.E.S.)
| | - Justin F. Creeden
- Department of Neurosciences, University of Toledo College of Medicine, Toledo, OH 43614, USA; (J.F.C.); (D.M.G.)
| | - Darren M. Gordon
- Department of Neurosciences, University of Toledo College of Medicine, Toledo, OH 43614, USA; (J.F.C.); (D.M.G.)
| | - Adam C. Spegele
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA; (A.C.S.); (L.G.K.)
| | - Steven L. Britton
- Department of Anesthesiology, Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Lauren G. Koch
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA; (A.C.S.); (L.G.K.)
| | - David E. Stec
- Center for Excellence in Cardiovascular-Renal Research, Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 North State St, Jackson, MS 392161, USA
- Correspondence: (T.D.H.J.); (D.E.S.)
| |
Collapse
|
48
|
MacDonald TL, Pattamaprapanont P, Pathak P, Fernandez N, Freitas EC, Hafida S, Mitri J, Britton SL, Koch LG, Lessard SJ. Hyperglycaemia is associated with impaired muscle signalling and aerobic adaptation to exercise. Nat Metab 2020; 2:902-917. [PMID: 32694831 PMCID: PMC8278496 DOI: 10.1038/s42255-020-0240-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 06/12/2020] [Indexed: 12/11/2022]
Abstract
Increased aerobic exercise capacity, as a result of exercise training, has important health benefits. However, some individuals are resistant to improvements in exercise capacity, probably due to undetermined genetic and environmental factors. Here, we show that exercise-induced improvements in aerobic capacity are blunted and aerobic remodelling of skeletal muscle is impaired in several animal models associated with chronic hyperglycaemia. Our data point to chronic hyperglycaemia as a potential negative regulator of aerobic adaptation, in part, via glucose-mediated modifications of the extracellular matrix, impaired vascularization and aberrant mechanical signalling in muscle. We also observe low exercise capacity and enhanced c-Jun N-terminal kinase activation in response to exercise in humans with impaired glucose tolerance. Our work indicates that current shifts in dietary and metabolic health, associated with increasing incidence of hyperglycaemia, might impair muscular and organismal adaptations to exercise training, including aerobic capacity as one of its key health outcomes.
Collapse
Affiliation(s)
- Tara L MacDonald
- Research Division, Joslin Diabetes Center, Boston, MA, USA
- Harvard Medical School, Harvard University, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Pattarawan Pattamaprapanont
- Research Division, Joslin Diabetes Center, Boston, MA, USA
- Harvard Medical School, Harvard University, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Prerana Pathak
- Research Division, Joslin Diabetes Center, Boston, MA, USA
| | | | - Ellen C Freitas
- School of Physical Education and Sport, University of São Paulo, Ribeirão Preto, Brazil
| | - Samar Hafida
- Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Joanna Mitri
- Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Steven L Britton
- Department of Molecular and Integrative Physiology, and Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - Lauren G Koch
- Department of Physiology and Pharmacology, The University of Toledo, Toledo, OH, USA
| | - Sarah J Lessard
- Research Division, Joslin Diabetes Center, Boston, MA, USA.
- Harvard Medical School, Harvard University, Boston, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
49
|
Biro PA, Thomas F, Ujvari B, Beckmann C. Can Energetic Capacity Help Explain Why Physical Activity Reduces Cancer Risk? Trends Cancer 2020; 6:829-837. [PMID: 32601046 DOI: 10.1016/j.trecan.2020.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/14/2020] [Accepted: 06/03/2020] [Indexed: 12/22/2022]
Abstract
Increased physical activity reduces cancer risk in humans, but why this whole-organism attribute reduces cancer remains unclear. Active individuals tend to have high capacity to generate energy on a sustained basis, which in turn can permit greater immune responses crucial for fighting emerging neoplasia. Thus, we suggest energetic capacity as a potential mechanism to explain the activity-cancer link, given that humans are intrinsically (not externally) energy limited. Human and rodent studies show that individuals with high energetic capacity mount greater immune responses and have lower cancer incidence; these trends persist after controlling for actual physical activity, supporting a direct role of energetic capacity. If true, exercise efforts might best target those that increase one's energetic capacity, which may be both individual and exercise specific.
Collapse
Affiliation(s)
- Peter A Biro
- Centre for Integrative Ecology, School of Life and Environmental Science, Deakin University, Geelong, VIC 3216, Australia.
| | - Frédéric Thomas
- CREEC, UMR IRD/CNRS/UM 5290, 911 Avenue Agropolis, BP 64501, 34394 Montpellier Cedex 5, France
| | - Beata Ujvari
- Centre for Integrative Ecology, School of Life and Environmental Science, Deakin University, Geelong, VIC 3216, Australia
| | - Christa Beckmann
- Centre for Integrative Ecology, School of Life and Environmental Science, Deakin University, Geelong, VIC 3216, Australia; School of Science, Western Sydney University, Parramatta, NSW 2116, Australia
| |
Collapse
|
50
|
Contrepois K, Wu S, Moneghetti KJ, Hornburg D, Ahadi S, Tsai MS, Metwally AA, Wei E, Lee-McMullen B, Quijada JV, Chen S, Christle JW, Ellenberger M, Balliu B, Taylor S, Durrant MG, Knowles DA, Choudhry H, Ashland M, Bahmani A, Enslen B, Amsallem M, Kobayashi Y, Avina M, Perelman D, Schüssler-Fiorenza Rose SM, Zhou W, Ashley EA, Montgomery SB, Chaib H, Haddad F, Snyder MP. Molecular Choreography of Acute Exercise. Cell 2020; 181:1112-1130.e16. [PMID: 32470399 PMCID: PMC7299174 DOI: 10.1016/j.cell.2020.04.043] [Citation(s) in RCA: 279] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 12/10/2019] [Accepted: 04/21/2020] [Indexed: 02/07/2023]
Abstract
Acute physical activity leads to several changes in metabolic, cardiovascular, and immune pathways. Although studies have examined selected changes in these pathways, the system-wide molecular response to an acute bout of exercise has not been fully characterized. We performed longitudinal multi-omic profiling of plasma and peripheral blood mononuclear cells including metabolome, lipidome, immunome, proteome, and transcriptome from 36 well-characterized volunteers, before and after a controlled bout of symptom-limited exercise. Time-series analysis revealed thousands of molecular changes and an orchestrated choreography of biological processes involving energy metabolism, oxidative stress, inflammation, tissue repair, and growth factor response, as well as regulatory pathways. Most of these processes were dampened and some were reversed in insulin-resistant participants. Finally, we discovered biological pathways involved in cardiopulmonary exercise response and developed prediction models revealing potential resting blood-based biomarkers of peak oxygen consumption.
Collapse
Affiliation(s)
- Kévin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Si Wu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Kegan J Moneghetti
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Melbourne, VIC, Australia; Stanford Sports Cardiology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Daniel Hornburg
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Sara Ahadi
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Ming-Shian Tsai
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Ahmed A Metwally
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Eric Wei
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Jeniffer V Quijada
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Songjie Chen
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeffrey W Christle
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; Stanford Sports Cardiology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Mathew Ellenberger
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Brunilda Balliu
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Shalina Taylor
- Pediatrics Department, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthew G Durrant
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - David A Knowles
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Department of Radiology, Stanford University, Stanford, CA, USA
| | - Hani Choudhry
- Department of Biochemistry, Faculty of Science, Cancer and Mutagenesis Unit, King Fahd Center for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Melanie Ashland
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Amir Bahmani
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Brooke Enslen
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Myriam Amsallem
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Yukari Kobayashi
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Monika Avina
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Dalia Perelman
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Wenyu Zhou
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Euan A Ashley
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; Center for Undiagnosed Diseases, Stanford University, Stanford, CA, USA
| | - Stephen B Montgomery
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Department of Pathology, Stanford University, Stanford, CA, USA
| | - Hassan Chaib
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Francois Haddad
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA.
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA.
| |
Collapse
|