1
|
Izquierdo M, de Souto Barreto P, Arai H, Bischoff-Ferrari HA, Cadore EL, Cesari M, Chen LK, Coen PM, Courneya KS, Duque G, Ferrucci L, Fielding RA, García-Hermoso A, Gutiérrez-Robledo LM, Harridge SDR, Kirk B, Kritchevsky S, Landi F, Lazarus N, Liu-Ambrose T, Marzetti E, Merchant RA, Morley JE, Pitkälä KH, Ramírez-Vélez R, Rodriguez-Mañas L, Rolland Y, Ruiz JG, Sáez de Asteasu ML, Villareal DT, Waters DL, Won Won C, Vellas B, Fiatarone Singh MA. Global consensus on optimal exercise recommendations for enhancing healthy longevity in older adults (ICFSR). J Nutr Health Aging 2025; 29:100401. [PMID: 39743381 DOI: 10.1016/j.jnha.2024.100401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 01/04/2025]
Abstract
Aging, a universal and inevitable process, is characterized by a progressive accumulation of physiological alterations and functional decline over time, leading to increased vulnerability to diseases and ultimately mortality as age advances. Lifestyle factors, notably physical activity (PA) and exercise, significantly modulate aging phenotypes. Physical activity and exercise can prevent or ameliorate lifestyle-related diseases, extend health span, enhance physical function, and reduce the burden of non-communicable chronic diseases including cardiometabolic disease, cancer, musculoskeletal and neurological conditions, and chronic respiratory diseases as well as premature mortality. Physical activity influences the cellular and molecular drivers of biological aging, slowing aging rates-a foundational aspect of geroscience. Thus, PA serves both as preventive medicine and therapeutic agent in pathological states. Sub-optimal PA levels correlate with increased disease prevalence in aging populations. Structured exercise prescriptions should therefore be customized and monitored like any other medical treatment, considering the dose-response relationships and specific adaptations necessary for intended outcomes. Current guidelines recommend a multifaceted exercise regimen that includes aerobic, resistance, balance, and flexibility training through structured and incidental (integrated lifestyle) activities. Tailored exercise programs have proven effective in helping older adults maintain their functional capacities, extending their health span, and enhancing their quality of life. Particularly important are anabolic exercises, such as Progressive resistance training (PRT), which are indispensable for maintaining or improving functional capacity in older adults, particularly those with frailty, sarcopenia or osteoporosis, or those hospitalized or in residential aged care. Multicomponent exercise interventions that include cognitive tasks significantly enhance the hallmarks of frailty (low body mass, strength, mobility, PA level, and energy) and cognitive function, thus preventing falls and optimizing functional capacity during aging. Importantly, PA/exercise displays dose-response characteristics and varies between individuals, necessitating personalized modalities tailored to specific medical conditions. Precision in exercise prescriptions remains a significant area of further research, given the global impact of aging and broad effects of PA. Economic analyses underscore the cost benefits of exercise programs, justifying broader integration into health care for older adults. However, despite these benefits, exercise is far from fully integrated into medical practice for older people. Many healthcare professionals, including geriatricians, need more training to incorporate exercise directly into patient care, whether in settings including hospitals, outpatient clinics, or residential care. Education about the use of exercise as isolated or adjunctive treatment for geriatric syndromes and chronic diseases would do much to ease the problems of polypharmacy and widespread prescription of potentially inappropriate medications. This intersection of prescriptive practices and PA/exercise offers a promising approach to enhance the well-being of older adults. An integrated strategy that combines exercise prescriptions with pharmacotherapy would optimize the vitality and functional independence of older people whilst minimizing adverse drug reactions. This consensus provides the rationale for the integration of PA into health promotion, disease prevention, and management strategies for older adults. Guidelines are included for specific modalities and dosages of exercise with proven efficacy in randomized controlled trials. Descriptions of the beneficial physiological changes, attenuation of aging phenotypes, and role of exercise in chronic disease and disability management in older adults are provided. The use of exercise in cardiometabolic disease, cancer, musculoskeletal conditions, frailty, sarcopenia, and neuropsychological health is emphasized. Recommendations to bridge existing knowledge and implementation gaps and fully integrate PA into the mainstream of geriatric care are provided. Particular attention is paid to the need for personalized medicine as it applies to exercise and geroscience, given the inter-individual variability in adaptation to exercise demonstrated in older adult cohorts. Overall, this consensus provides a foundation for applying and extending the current knowledge base of exercise as medicine for an aging population to optimize health span and quality of life.
Collapse
Affiliation(s)
- Mikel Izquierdo
- Navarrabiomed, Hospital Universitario de Navarra (CHN)-Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain; CIBER of Frailty and Healthy Ageing (CIBERFES), Instituto de Salud Carlos III Madrid, Spain.
| | - Philipe de Souto Barreto
- IHU HealthAge, Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France; CERPOP, UPS/Inserm 1295, Toulouse, France
| | - Hidenori Arai
- National Center for Geriatrics and Gerontology, Obu, Japan
| | - Heike A Bischoff-Ferrari
- Department of Geriatrics and Aging Research, Research Centre on Aging and Mobility, University of Zurich, Zurich, Switzerland
| | - Eduardo L Cadore
- Exercise Research Laboratory, School of Physical Education, Physiotherapy and Dance, Universidade Federal do Rio Grande do Sul, Brazil
| | - Matteo Cesari
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Liang-Kung Chen
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Center for Geriatrics and Gerontology, Taipei Veterans General Hospital, Taipei Municipal Gab-Dau Hospital, Taipei, Taiwan
| | - Paul M Coen
- AdventHealth Orlando, Translational Research Institute, Orlando, Florida, United States
| | - Kerry S Courneya
- Faculty of Kinesiology, Sport, and Recreation, College of Health Sciences, University of Alberta, Edmonton, Alberta T6G 2H9, Canada
| | - Gustavo Duque
- Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Luigi Ferrucci
- National Institute on Aging, Baltimore, MD, United States
| | - Roger A Fielding
- Nutrition, Exercise Physiology, and Sarcopenia Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, United States
| | - Antonio García-Hermoso
- Navarrabiomed, Hospital Universitario de Navarra (CHN)-Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain; CIBER of Frailty and Healthy Ageing (CIBERFES), Instituto de Salud Carlos III Madrid, Spain
| | | | - Stephen D R Harridge
- Centre for Human and Applied Physiological Sciences, King's College London, United Kingdom
| | - Ben Kirk
- Department of Medicine-Western Health, Melbourne Medical School, University of Melbourne, St. Albans, Melbourne, VIC, Australia
| | - Stephen Kritchevsky
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Francesco Landi
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Norman Lazarus
- Centre for Human and Applied Physiological Sciences, King's College London, United Kingdom
| | - Teresa Liu-Ambrose
- Aging, Mobility, and Cognitive Health Laboratory, Department of Physical Therapy, Faculty of Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Centre for Aging SMART at Vancouver Coastal Health, Vancouver Coastal Health Research Institute,Vancouver, BC, Canada
| | - Emanuele Marzetti
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Reshma A Merchant
- Division of Geriatric Medicine, Department of Medicine, National University Hospital, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - John E Morley
- Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Kaisu H Pitkälä
- University of Helsinki and Helsinki University Hospital, PO Box 20, 00029 Helsinki, Finland
| | - Robinson Ramírez-Vélez
- Navarrabiomed, Hospital Universitario de Navarra (CHN)-Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain; CIBER of Frailty and Healthy Ageing (CIBERFES), Instituto de Salud Carlos III Madrid, Spain
| | - Leocadio Rodriguez-Mañas
- CIBER of Frailty and Healthy Ageing (CIBERFES), Instituto de Salud Carlos III Madrid, Spain; Geriatric Service, University Hospital of Getafe, Getafe, Spain
| | - Yves Rolland
- IHU HealthAge, Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France; CERPOP, UPS/Inserm 1295, Toulouse, France
| | - Jorge G Ruiz
- Memorial Healthcare System, Hollywood, Florida and Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, Florida, United States
| | - Mikel L Sáez de Asteasu
- Navarrabiomed, Hospital Universitario de Navarra (CHN)-Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain; CIBER of Frailty and Healthy Ageing (CIBERFES), Instituto de Salud Carlos III Madrid, Spain
| | - Dennis T Villareal
- Baylor College of Medicine, and Center for Translational Research on Inflammatory Diseases, Michael E DeBakey VA Medical Center, Houston, Texas, United States
| | - Debra L Waters
- Department of Medicine, School of Physiotherapy, University of Otago, Dunedin; Department of Internal Medicine/Geriatrics, University of New Mexico, Albuquerque, Mexico
| | - Chang Won Won
- Elderly Frailty Research Center, Department of Family Medicine, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bruno Vellas
- IHU HealthAge, Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France; CERPOP, UPS/Inserm 1295, Toulouse, France
| | - Maria A Fiatarone Singh
- Faculty of Medicine and Health, School of Health Sciences and Sydney Medical School, University of Sydney, New South Wales, Australia, and Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Roslindale, MA, United States
| |
Collapse
|
2
|
Lacombe A, Scorrano L. The interplay between mitochondrial dynamics and autophagy: From a key homeostatic mechanism to a driver of pathology. Semin Cell Dev Biol 2024; 161-162:1-19. [PMID: 38430721 DOI: 10.1016/j.semcdb.2024.02.001] [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: 11/06/2023] [Revised: 02/06/2024] [Accepted: 02/15/2024] [Indexed: 03/05/2024]
Abstract
The complex relationship between mitochondrial dynamics and autophagy illustrates how two cellular housekeeping processes are intimately linked, illuminating fundamental principles of cellular homeostasis and shedding light on disparate pathological conditions including several neurodegenerative disorders. Here we review the basic tenets of mitochondrial dynamics i.e., the concerted balance between fusion and fission of the organelle, and its interplay with macroautophagy and selective mitochondrial autophagy, also dubbed mitophagy, in the maintenance of mitochondrial quality control and ultimately in cell viability. We illustrate how conditions of altered mitochondrial dynamics reverberate on autophagy and vice versa. Finally, we illustrate how altered interplay between these two key cellular processes participates in the pathogenesis of human disorders affecting multiple organs and systems.
Collapse
Affiliation(s)
- Alice Lacombe
- Dept. of Biology, University of Padova, Padova, Italy
| | - Luca Scorrano
- Dept. of Biology, University of Padova, Padova, Italy; Veneto Institute of Molecular Medicine, Padova, Italy.
| |
Collapse
|
3
|
Huang YC, Hsu CC, Fu TC, Wang JS. Interval aerobic/resistance exercise training depresses adrenergic-induced apoptosis of lymphocytes in sedentary males. Eur J Appl Physiol 2024; 124:837-848. [PMID: 37712975 DOI: 10.1007/s00421-023-05311-3] [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: 05/05/2023] [Accepted: 08/30/2023] [Indexed: 09/16/2023]
Abstract
PURPOSE Adrenergic stimulation affects lymphocyte autophagy and apoptosis by activating β1-adrenergic receptor (β1-AR) and G protein-coupled receptor kinase 2 (GRK-2) downstream signaling. This study investigated how combined aerobic and resistance exercise training on the interval or continuous pattern influences aerobic/muscular fitness and β1-AR/GRK-2 signaling, and corresponding apoptosis/autophagy of lymphocytes in sedentary males. METHODS Thirty-four sedentary males were randomized into interval training (IT, age = 22.5 ± 0.6 years, fitness level = 47.5 ± 0.9 mL/min/kg, body mass index (BMI) = 22.4 ± 0.4 kg/m2, n = 17) and continuous training (CT, age = 21.6 ± 0.4 years, fitness level = 45.2 ± 1.0 mL/min/kg, BMI = 22.2 ± 0.3 kg/m2, n = 17) groups. These subjects performed IT (bicycle exercise at alternating 40% and 80%VO2 reserve (VO2R) and isokinetic exercise at alternating 60°/s and 180°/s) or CT (bicycle exercise at continuously 60%VO2R and isokinetic exercise at continuously 120°/s) for 30 min/day, 5 days/week for 6 weeks. Aerobic capacity and muscular strength/endurance were determined by the graded exercise test (GXT) and isokinetic strength test, respectively. Blood lymphocyte autophagy/apoptosis and β1-AR/GRK-2 signaling were analyzed using flow cytometry. RESULTS Both IT and CT groups increased isokinetic strengths at various angular velocities, whereas only IT significantly enhanced muscle endurance, indicated by lowered fatigue index from 47.0 ± 1.3% to 41.8 ± 1.6% (P < 0.05). Moreover, the IT group (143 ± 7%) revealed a higher improvement in VO2peak than CT group (132 ± 6%) (P < 0.05). Acute GXT augmented (i) GRK-2 and protein kinase A expressions, (ii) LAMP-2 upregulation and acridine orange staining, (iii) mitochondrial transmembrane potential diminishing, caspase-3 activation, and phosphatidylserine (PS) exposure caused by epinephrine in blood lymphocytes. However, the degree of epinephrine-induced lymphocyte PS exposure potentiated by GXT was suppressed from 65.2 ± 5.2% to 47.4 ± 6.5% following 6 weeks of the IT (P < 0.05). CONCLUSION The IT may be considered more beneficial than CT in terms of improving aerobic/muscular fitness and simultaneously ameliorating apoptosis of blood lymphocyte evoked by intense exercise or adrenergic stimulation in sedentary males.
Collapse
Affiliation(s)
- Yu-Chieh Huang
- Department of Physical Therapy, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Chih-Chin Hsu
- Heart Failure Center, Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Tieh-Cheng Fu
- Heart Failure Center, Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Jong-Shyan Wang
- Heart Failure Center, Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Keelung, Taiwan.
- Healthy Aging Research Center, Graduate Institute of Rehabilitation Science, Medical College, Chang Gung University, Tao-Yuan, Taiwan.
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Tao-Yuan, Taiwan.
- Graduate Institute of Rehabilitation Science, Chang Gung University, 259 Wen-Hwa 1St Road, Kwei-Shan, Tao-Yuan, 333, Taiwan.
| |
Collapse
|
4
|
Walter S, Jung T, Herpich C, Norman K, Pivovarova-Ramich O, Ott C. Determination of the autophagic flux in murine and human peripheral blood mononuclear cells. Front Cell Dev Biol 2023; 11:1122998. [PMID: 36994103 PMCID: PMC10040559 DOI: 10.3389/fcell.2023.1122998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/27/2023] [Indexed: 03/16/2023] Open
Abstract
The autophagy lysosomal system (ALS) is crucial for cellular homeostasis, contributing to maintain whole body health and alterations are associated with diseases like cancer or cardiovascular diseases. For determining the autophagic flux, inhibition of lysosomal degradation is mandatory, highly complicating autophagy measurement in vivo. To overcome this, herein blood cells were used as they are easy and routinely to isolate. Within this study we provide detailed protocols for determination of the autophagic flux in peripheral blood mononuclear cells (PBMCs) isolated from human and, to our knowledge the first time, also from murine whole blood, extensively discussing advantages and disadvantages of both methods. Isolation of PBMCs was performed using density gradient centrifugation. To minimize changes on the autophagic flux through experimental conditions, cells were directly treated with concanamycin A (ConA) for 2 h at 37°C in their serum or for murine cells in serum filled up with NaCl. ConA treatment decreased lysosomal cathepsins activity and increased Sequestosome 1 (SQSTM1) protein and LC3A/B-II:LC3A/B-I ratio in murine PBMCs, while transcription factor EB was not altered yet. Aging further enhanced ConA-associated increase in SQSTM1 protein in murine PBMCs but not in cardiomyocytes, indicating tissue-specific differences in autophagic flux. In human PBMCs, ConA treatment also decreased lysosomal activity and increased LC3A/B-II protein levels, demonstrating successful autophagic flux detection in human subjects. In summary, both protocols are suitable to determine the autophagic flux in murine and human samples and may facilitate a better mechanistic understanding of altered autophagy in aging and disease models and to further develop novel treatment strategies.
Collapse
Affiliation(s)
- Sophia Walter
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Tobias Jung
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Catrin Herpich
- Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Department of Geriatrics and Medical Gerontology, Charité Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, Berlin, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Kristina Norman
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Department of Geriatrics and Medical Gerontology, Charité Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, Berlin, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Olga Pivovarova-Ramich
- Department of Molecular Nutritional Medicine, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Department of Endocrinology, Diabetes and Nutrition, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Berlin, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Christiane Ott
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- *Correspondence: Christiane Ott,
| |
Collapse
|
5
|
Caponio D, Veverová K, Zhang SQ, Shi L, Wong G, Vyhnalek M, Fang EF. Compromised autophagy and mitophagy in brain ageing and Alzheimer's diseases. AGING BRAIN 2022; 2:100056. [PMID: 36908880 PMCID: PMC9997167 DOI: 10.1016/j.nbas.2022.100056] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/04/2022] [Accepted: 11/16/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer's disease (AD) is one of the most persistent and devastating neurodegenerative disorders of old age, and is characterized clinically by an insidious onset and a gradual, progressive deterioration of cognitive abilities, ranging from loss of memory to impairment of judgement and reasoning. Despite years of research, an effective cure is still not available. Autophagy is the cellular 'garbage' clearance system which plays fundamental roles in neurogenesis, neuronal development and activity, and brain health, including memory and learning. A selective sub-type of autophagy is mitophagy which recognizes and degrades damaged or superfluous mitochondria to maintain a healthy and necessary cellular mitochondrial pool. However, emerging evidence from animal models and human samples suggests an age-dependent reduction of autophagy and mitophagy, which are also compromised in AD. Upregulation of autophagy/mitophagy slows down memory loss and ameliorates clinical features in animal models of AD. In this review, we give an overview of autophagy and mitophagy and their link to the progression of AD. We also summarize approaches to upregulate autophagy/mitophagy. We hypothesize that age-dependent compromised autophagy/mitophagy is a cause of brain ageing and a risk factor for AD, while restoration of autophagy/mitophagy to more youthful levels could return the brain to health.
Collapse
Affiliation(s)
- Domenica Caponio
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway
| | - Kateřina Veverová
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic
| | - Shi-qi Zhang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway
| | - Liu Shi
- Department of Psychiatry, University of Oxford, Oxford, UK
- Novo Nordisk Research Centre Oxford (NNRCO)
| | - Garry Wong
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Martin Vyhnalek
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic
| | - Evandro F. Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway
- The Norwegian Centre on Healthy Ageing (NO-Age), Oslo, Norway
| |
Collapse
|
6
|
Coenzyme Q10 ameliorates aging-induced memory deficits via modulation of apoptosis, oxidative stress, and mitophagy in aged rats. Exp Gerontol 2022; 168:111950. [PMID: 36089173 DOI: 10.1016/j.exger.2022.111950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 08/11/2022] [Accepted: 09/03/2022] [Indexed: 11/21/2022]
Abstract
The behavioral effects and molecular signaling mechanisms of Coenzyme Q10 (Q10) in age-related memory impairment are poorly understood. This study aimed to investigate the effects of Q10 on memory impairment, oxidative stress, apoptosis, and mitophagy in aged rats. 40 aged (24 months old) and 10 young (3 months old) male Wistar rats were randomly divided into the following groups (n = 10/group): young + vehicle, aged + vehicle, and aged + Q10 (at 100, 200, 300 mg/kg/day doses). Treatments were administrated orally by gavage for 2 weeks. The novel object recognition test was used to assess episodic memory. Oxidative stress, apoptosis, and mitophagy-related protein expressions were measured in the hippocampus. We found that Q10 reversed aging-induced memory impairment at the dose of 300 mg/kg. Moreover, aging was associated with a reduction in ATP production, decrease in mitophagy-related proteins (PINK, Parkin, and P62 levels and LC3II/I ratio), excessive generation of reactive oxygen species and lipid peroxidation, and apoptosis in the hippocampus, which were partially reversed following oral administration of Q10. These findings indicate the therapeutic potential of Q10 in aging-induced memory decline.
Collapse
|
7
|
Cento AS, Leigheb M, Caretti G, Penna F. Exercise and Exercise Mimetics for the Treatment of Musculoskeletal Disorders. Curr Osteoporos Rep 2022; 20:249-259. [PMID: 35881303 PMCID: PMC9522759 DOI: 10.1007/s11914-022-00739-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/06/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The incidence of musculoskeletal disorders affecting bones, joints, and muscles is dramatically increasing in parallel with the increased longevity of the worldwide population, severely impacting on the individual's quality of life and on the healthcare costs. Inactivity and sedentary lifestyle are nowadays considered the main drivers of age-associated musculoskeletal disorders and exercise may counteract such alterations also in other bone- and muscle-centered disorders. This review aims at clarifying the potential use of exercise training to improve musculoskeletal health. RECENT FINDINGS Both the skeletal muscle and the bone are involved in a complex crosstalk determining, in part through tissue-specific and inflammatory/immune released factors, the occurrence of musculoskeletal disorders. Exercise is able to modulate the levels of those molecules and several associated molecular pathways. Evidence from preclinical and clinical trials supports the adoption of exercise and the future use of exercise mimicking drugs will optimize the care of individuals with musculoskeletal disorders.
Collapse
Affiliation(s)
- Alessia S Cento
- Department of Clinical and Biological Sciences, University of Torino, Corso Raffaello, 30, 10125, Torino, Italy
| | - Massimiliano Leigheb
- Orthopaedics and Traumatology Unit, "Maggiore della Carità" Hospital, Department of Health Sciences, University of Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| | - Giuseppina Caretti
- Department of Biosciences, University of Milan, Via Celoria 26, 20133, Milan, Italy
| | - Fabio Penna
- Department of Clinical and Biological Sciences, University of Torino, Corso Raffaello, 30, 10125, Torino, Italy.
| |
Collapse
|
8
|
Jewell S, Herath AM, Gordon R. Inflammasome Activation in Parkinson’s Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:S113-S128. [PMID: 35848038 PMCID: PMC9535572 DOI: 10.3233/jpd-223338] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Chronic sterile inflammation and persistent immune activation is a prominent pathological feature of Parkinson’s disease (PD). Inflammasomes are multi-protein intracellular signaling complexes which orchestrate inflammatory responses in immune cells to a diverse range of pathogens and host-derived signals. Widespread inflammasome activation is evident in PD patients at the sites of dopaminergic degeneration as well as in blood samples and mucosal biopsies. Inflammasome activation in the nigrostriatal system is also a common pathological feature in both neurotoxicant and α-synuclein models of PD where dopaminergic degeneration occurs through distinct mechanisms. The NLRP3 (NLR Family Pyrin Domain Containing 3) inflammasome has been shown to be the primary driver of inflammatory neurotoxicity in PD and other neurodegenerative diseases. Chronic NLRP3 inflammasome activation is triggered by pathogenic misfolded α-synuclein aggregates which accumulate and spread over the disease course in PD. Converging lines of evidence suggest that blocking inflammasome activation could be a promising therapeutic strategy for disease modification, with both NLRP3 knockout mice and CNS-permeable pharmacological inhibitors providing robust neuroprotection in multiple PD models. This review summarizes the current evidence and knowledge gaps around inflammasome activation in PD, the pathological mechanisms by which persistent inflammasome activation can drive dopaminergic degeneration and the therapeutic opportunities for disease modification using NLRP3 inhibitors.
Collapse
Affiliation(s)
- Shannon Jewell
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Ashane M. Herath
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Richard Gordon
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| |
Collapse
|
9
|
Qi JY, Yang LK, Wang XS, Wang M, Li XB, Feng B, Wu YM, Liu SB, Zhang K. Mechanism of CNS regulation by irisin, a multifunctional protein. Brain Res Bull 2022; 188:11-20. [PMID: 35850187 DOI: 10.1016/j.brainresbull.2022.07.007] [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: 03/13/2022] [Revised: 07/10/2022] [Accepted: 07/13/2022] [Indexed: 11/30/2022]
Abstract
Exercise not only builds up our body but also improves cognitive function. Skeletal muscle secretes myokine during exercise as a large reservoir of signaling molecules, which can be considered as a medium between exercise and brain health. Irisin is a circulating myokine derived from the Fibronectin type III domain-containing protein 5 (FNDC5). Irisin regulates energy metabolism because it can stimulate the "Browning" of white adipose tissue. It has been reported that irisin can cross the blood-brain barrier and increase the expression of a brain-derived neurotrophic factor (BDNF) in the hippocampus, which improves learning and memory. In addition, the neuroprotective effect of irisin has been verified in various disease models. Therefore, this review summarizes how irisin plays a neuroprotective role, including its signal pathway and mechanism. In addition, we will briefly discuss the therapeutic potential of irisin for neurological diseases.
Collapse
Affiliation(s)
- Jing-Yu Qi
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Liu-Kun Yang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Xin-Shang Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Min Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Xu-Bo Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Ban Feng
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Yu-Mei Wu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Shui-Bing Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.
| | - Kun Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
10
|
Versaci F, Valenti V, Forte M, Cammisotto V, Nocella C, Bartimoccia S, Schirone L, Schiavon S, Vecchio D, D’Ambrosio L, Spinosa G, D’Amico A, Chimenti I, Violi F, Frati G, Pignatelli P, Sciarretta S, Pastori D, Carnevale R. Aging-Related Decline of Autophagy in Patients with Atrial Fibrillation-A Post Hoc Analysis of the ATHERO-AF Study. Antioxidants (Basel) 2022; 11:antiox11040698. [PMID: 35453383 PMCID: PMC9030744 DOI: 10.3390/antiox11040698] [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: 02/01/2022] [Revised: 03/26/2022] [Accepted: 03/31/2022] [Indexed: 01/02/2023] Open
Abstract
Background: Aging is an independent risk factor for cardiovascular diseases. The autophagy process may play a role in delaying aging and improving cardiovascular function in aging. Data regarding autophagy in atrial fibrillation (AF) patients are lacking. Methods: A post hoc analysis of the prospective ATHERO-AF cohort study, including 150 AF patients and 150 sex- and age-matched control subjects (CS), was performed. For the analysis, the population was divided into three age groups: <50−60, 61−70, and >70 years. Oxidative stress (Nox2 activity and hydrogen peroxide, H2O2), platelet activation (PA) by sP-selectin and CD40L, endothelial dysfunction (nitric oxide, NO), and autophagy parameters (P62 and ATG5 levels) were assessed. Results: Nox2 activity and H2O2 production were higher in the AF patients than in the CS; conversely, antioxidant capacity was decreased in the AF patients compared to the CS, as was NO production. Moreover, sP-selectin and CD40L were higher in the AF patients than in the CS. The autophagy process was also significantly impaired in the AF patients. We found a significant difference in oxidative stress, PA, NO production, and autophagy across the age groups. Autophagy markers correlated with oxidative stress, PA, and endothelial dysfunction in both groups. Conclusions: This study provides evidence that the autophagy process may represent a mechanism for increased cardiovascular risk in the AF population.
Collapse
Affiliation(s)
- Francesco Versaci
- Department of Cardiology, Santa Maria Goretti Hospital, 04100 Latina, Italy; (F.V.); (V.V.)
| | - Valentina Valenti
- Department of Cardiology, Santa Maria Goretti Hospital, 04100 Latina, Italy; (F.V.); (V.V.)
| | - Maurizio Forte
- IRCCS Neuromed, 86077 Pozzilli, Italy; (M.F.); (G.F.); (S.S.)
| | - Vittoria Cammisotto
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (V.C.); (C.N.); (P.P.)
| | - Cristina Nocella
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (V.C.); (C.N.); (P.P.)
| | - Simona Bartimoccia
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
| | - Leonardo Schirone
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
| | - Sonia Schiavon
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
| | - Daniele Vecchio
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
| | - Luca D’Ambrosio
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
| | - Giulia Spinosa
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
| | - Alessandra D’Amico
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy;
| | - Isotta Chimenti
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
| | | | - Giacomo Frati
- IRCCS Neuromed, 86077 Pozzilli, Italy; (M.F.); (G.F.); (S.S.)
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (V.C.); (C.N.); (P.P.)
- Mediterranea Cardiocentro, 80122 Naples, Italy;
| | - Sebastiano Sciarretta
- IRCCS Neuromed, 86077 Pozzilli, Italy; (M.F.); (G.F.); (S.S.)
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
| | - Daniele Pastori
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (V.C.); (C.N.); (P.P.)
- Correspondence: (D.P.); (R.C.); Tel.: +39-0649970941 (D.P.); +39-07731757245 (R.C.); Fax: +39-0649972309 (D.P.); +39-07731757245 (R.C.)
| | - Roberto Carnevale
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
- Mediterranea Cardiocentro, 80122 Naples, Italy;
- Correspondence: (D.P.); (R.C.); Tel.: +39-0649970941 (D.P.); +39-07731757245 (R.C.); Fax: +39-0649972309 (D.P.); +39-07731757245 (R.C.)
| |
Collapse
|
11
|
Oliveira LDC, Morais GP, Ropelle ER, de Moura LP, Cintra DE, Pauli JR, de Freitas EC, Rorato R, da Silva ASR. Using Intermittent Fasting as a Non-pharmacological Strategy to Alleviate Obesity-Induced Hypothalamic Molecular Pathway Disruption. Front Nutr 2022; 9:858320. [PMID: 35445066 PMCID: PMC9014844 DOI: 10.3389/fnut.2022.858320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/25/2022] [Indexed: 12/18/2022] Open
Abstract
Intermittent fasting (IF) is a popular intervention used to fight overweight/obesity. This condition is accompanied by hypothalamic inflammation, limiting the proper signaling of molecular pathways, with consequent dysregulation of food intake and energy homeostasis. This mini-review explored the therapeutic modulation potential of IF regarding the disruption of these molecular pathways. IF seems to modulate inflammatory pathways in the brain, which may also be correlated with the brain-microbiota axis, improving hypothalamic signaling of leptin and insulin, and inducing the autophagic pathway in hypothalamic neurons, contributing to weight loss in obesity. Evidence also suggests that when an IF protocol is performed without respecting the circadian cycle, it can lead to dysregulation in the expression of circadian cycle regulatory genes, with potential health damage. In conclusion, IF may have the potential to be an adjuvant treatment to improve the reestablishment of hypothalamic responses in obesity.
Collapse
Affiliation(s)
- Luciana da Costa Oliveira
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Gustavo Paroschi Morais
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Eduardo R. Ropelle
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, São Paulo, Brazil
| | - Leandro P. de Moura
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, São Paulo, Brazil
| | - Dennys E. Cintra
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, São Paulo, Brazil
| | - José R. Pauli
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, São Paulo, Brazil
| | - Ellen C. de Freitas
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Rodrigo Rorato
- Postgraduate Program in Molecular Biology, Laboratory of Stress Neuroendocrinology, Department of Biophysics, Paulista Medical School, Federal University of São Paulo, São Paulo, Brazil
- Rodrigo Rorato,
| | - Adelino Sanchez R. da Silva
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
- *Correspondence: Adelino Sanchez R. da Silva,
| |
Collapse
|
12
|
Gomarasca M, Micielska K, Faraldi M, Flis M, Perego S, Banfi G, Ziemann E, Lombardi G. Impact of 12-Week Moderate-Intensity Aerobic Training on Inflammasome Complex Activation in Elderly Women. Front Physiol 2022; 13:792859. [PMID: 35273516 PMCID: PMC8902397 DOI: 10.3389/fphys.2022.792859] [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: 10/11/2021] [Accepted: 01/14/2022] [Indexed: 12/25/2022] Open
Abstract
Aging often associates with a chronic low-grade inflammatory status that can be consequent to the activation of Toll-like receptors (TLRs) and the downstream NLR family pyrin domain containing 3 (NLRP3) inflammasome and causes a chronic secretion of pro-inflammatory cytokines. Since exercise has known anti-inflammatory effects, we investigated the effect of Nordic walking training on inflammasome activation and downstream effectors in elderly women. A population of elderly women was divided into EXP (n = 29) that completed 12 weeks of the moderate-intensity aerobic training program and CTRL (n = 29), performing no activity. Blood samples were taken before and after the first (T1-pre and T1-post, respectively) and last (T2-pre and T2-post, respectively) exercise unit. Inflammasome activation status was assessed by whole blood NLRP3 and TLR4 expression by RT-qPCR. Serum levels of IL-1β, IL-6, TNFα, and IL-18 cytokines were assayed by multiplex fluorescent beads-based immunoassays or ELISA. NLRP3 and TLR4 levels were reduced 2 folds between T1-pre and T2-pre and induced at T2-post, compared to T2-pre, by 2.6- and 2.9-fold, respectively. A single exercise bout elicited a 1. 38-, 1. 5-, and 1.36-fold rise of IL-1β, TNFα, and IL-6 concentration, respectively, although not significant, at the beginning of the training (T1-pre vs. T1-post), a 1.4-fold decrease for IL-1β and TNFα at the end of the training (T1-pre vs. T2-pre), and a 2-, 1.8- and 1.26-fold increase after the last exercise session (T2-pre vs. T2-post) for the three cytokines. When stratifying the population based on BMI in normal weight (NW) and overweight (OW), NLRP3 and TLR4 expression was affected only in NW. As for inflammatory cytokines, IL-1β was modulated in NW at the beginning of the training, whereas in OW at the end of the training; for TNFα, this time-dependent modulation was significant only in OW. Applied aerobic training affected the resting expression of inflammasome constituents (NLRP3 and TLR4) and levels of downstream effectors (IL-1β, TNFα, and IL-6). However, at the end of the program, participants acquire an acute inflammatory response to exercise that was absent at baseline. Future studies would have to define the molecular mechanisms associated with, and how to potentiate, the exercise-associated inflammatory response.
Collapse
Affiliation(s)
- Marta Gomarasca
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Katarzyna Micielska
- Department of Physical Education and Lifelong Sports, Poznań University of Physical Education, Poznań, Poland.,Doctoral School, Gdańsk University of Physical Education and Sport, Gdańsk, Poland
| | - Martina Faraldi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Marta Flis
- Department of Physiology, Gdańsk University of Physical Education and Sport, Gdańsk, Poland
| | - Silvia Perego
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Giuseppe Banfi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Ewa Ziemann
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| |
Collapse
|
13
|
Huang CJ, Rodriguez AL, Visavadiya NP, Fico BG, Slusher AL, Ferrandi PJ, Whitehurst M. An exploratory investigation of apoptotic and autophagic responses in peripheral blood mononuclear cells following maximal aerobic exercise in obese individuals. Arch Physiol Biochem 2022; 128:209-216. [PMID: 31564171 DOI: 10.1080/13813455.2019.1671875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Autophagy is a critical molecular process in promoting cell survival against apoptosis. This study examined whether maximal aerobic exercise-mediated apoptosis in obesity might be underlying the involvement of autophagy in the peripheral blood mononuclear cells (PBMCs). Twelve healthy male subjects (6 obese and 6 normal-weight) were recruited to participate in a maximal graded exercise test on a treadmill. Obese subjects exhibited a significantly lower Bax, but a higher Bcl-2 protein level in conjunction with a reduced Bax/Bcl-2 AUCi compared to normal-weight subjects following exercise. Furthermore, a greater LC3-II/LC3-I ratio and LC3-II/LC3-I AUCi was observed in obese subjects compared to normal-weight subjects. LC3-II/LC3-I AUCi was also positively associated with obesity-associated parameters (BMI, waist/hip circumference, and fasting insulin level), but was negatively correlated with Bax/Bcl-2 AUCi. These findings demonstrate that maximal aerobic exercise differentially mediates the intrinsic apoptotic pathway and autophagic activity in human PBMCs isolated from obese compared to normal-weight individuals.
Collapse
Affiliation(s)
- Chun-Jung Huang
- Exercise Biochemistry Laboratory, Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL, USA
| | - Alexandra L Rodriguez
- Exercise Biochemistry Laboratory, Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL, USA
| | - Nishant P Visavadiya
- Exercise Biochemistry Laboratory, Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL, USA
| | - Brandon G Fico
- Department of Kinesiology and Health Education, University of Texas, Austin, TX, USA
| | - Aaron L Slusher
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Peter J Ferrandi
- Department of Health and Kinesiology, Purdue University, West Lafayette, IN, USA
| | - Michael Whitehurst
- Exercise Biochemistry Laboratory, Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL, USA
| |
Collapse
|
14
|
Shen W, He J, Hou T, Si J, Chen S. Common Pathogenetic Mechanisms Underlying Aging and Tumor and Means of Interventions. Aging Dis 2022; 13:1063-1091. [PMID: 35855334 PMCID: PMC9286910 DOI: 10.14336/ad.2021.1208] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/07/2021] [Indexed: 11/22/2022] Open
Abstract
Recently, there has been an increase in the incidence of malignant tumors among the older population. Moreover, there is an association between aging and cancer. During the process of senescence, the human body suffers from a series of imbalances, which have been shown to further accelerate aging, trigger tumorigenesis, and facilitate cancer progression. Therefore, exploring the junctions of aging and cancer and searching for novel methods to restore the junctions is of great importance to intervene against aging-related cancers. In this review, we have identified the underlying pathogenetic mechanisms of aging-related cancers by comparing alterations in the human body caused by aging and the factors that trigger cancers. We found that the common mechanisms of aging and cancer include cellular senescence, alterations in proteostasis, microbiota disorders (decreased probiotics and increased pernicious bacteria), persistent chronic inflammation, extensive immunosenescence, inordinate energy metabolism, altered material metabolism, endocrine disorders, altered genetic expression, and epigenetic modification. Furthermore, we have proposed that aging and cancer have common means of intervention, including novel uses of common medicine (metformin, resveratrol, and rapamycin), dietary restriction, and artificial microbiota intervention or selectively replenishing scarce metabolites. In addition, we have summarized the research progress of each intervention and revealed their bidirectional effects on cancer progression to compare their reliability and feasibility. Therefore, the study findings provide vital information for advanced research studies on age-related cancers. However, there is a need for further optimization of the described methods and more suitable methods for complicated clinical practices. In conclusion, targeting aging may have potential therapeutic effects on aging-related cancers.
Collapse
Affiliation(s)
- Weiyi Shen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China.
- Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang, China.
- Prevention and Treatment Research Center for Senescent Disease, Zhejiang University School of Medicine, Zhejiang, China
| | - Jiamin He
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China.
- Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang, China.
- Prevention and Treatment Research Center for Senescent Disease, Zhejiang University School of Medicine, Zhejiang, China
| | - Tongyao Hou
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China.
- Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang, China.
- Prevention and Treatment Research Center for Senescent Disease, Zhejiang University School of Medicine, Zhejiang, China
- Correspondence should be addressed to: Dr. Shujie Chen (), Dr. Jianmin Si () and Dr. Tongyao Hou (), Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang, China
| | - Jianmin Si
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China.
- Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang, China.
- Prevention and Treatment Research Center for Senescent Disease, Zhejiang University School of Medicine, Zhejiang, China
- Correspondence should be addressed to: Dr. Shujie Chen (), Dr. Jianmin Si () and Dr. Tongyao Hou (), Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang, China
| | - Shujie Chen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China.
- Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang, China.
- Prevention and Treatment Research Center for Senescent Disease, Zhejiang University School of Medicine, Zhejiang, China
- Correspondence should be addressed to: Dr. Shujie Chen (), Dr. Jianmin Si () and Dr. Tongyao Hou (), Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang, China
| |
Collapse
|
15
|
Marriott CFS, Petrella AFM, Marriott ECS, Boa Sorte Silva NC, Petrella RJ. High-Intensity Interval Training in Older Adults: a Scoping Review. SPORTS MEDICINE - OPEN 2021; 7:49. [PMID: 34279765 PMCID: PMC8289951 DOI: 10.1186/s40798-021-00344-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 07/04/2021] [Indexed: 12/12/2022]
Abstract
High-intensity interval training (HIIT) is an increasingly popular form of aerobic exercise which includes bouts of high-intensity exercise interspersed with periods of rest. The health benefits, risks, and optimal design of HIIT are still unclear. Further, most research on HIIT has been done in young and middle-aged adults, and as such, the tolerability and effects in senior populations are less well-known. The purpose of this scoping review was to characterize HIIT research that has been done in older adults including protocols, feasibility, and safety and to identify gaps in the current knowledge. Five databases were searched with variations of the terms, "high-intensity interval training" and "older adults" for experimental or quasi-experimental studies published in or after 2009. Studies were included if they had a treatment group with a mean age of 65 years or older who did HIIT, exclusively. Of 4644 papers identified, 69 met the inclusion criteria. The average duration of training was 7.9 (7.0) weeks (mean [SD]) and protocols ranged widely. The average sample size was 47.0 (65.2) subjects (mean [SD]). Healthy populations were the most studied group (n = 30), followed by subjects with cardiovascular (n = 12) or cardiac disease (n = 9), metabolic dysfunction (n = 8), and others (n = 10). The most common primary outcomes included changes in cardiorespiratory fitness (such as VO2peak) as well as feasibility and safety of the protocols as measured by the number of participant dropouts, adverse events, and compliance rate. HIIT protocols were diverse but were generally well-tolerated and may confer many health advantages to older adults. Larger studies and more research in clinical populations most representative of older adults are needed to further evaluate the clinical effects of HIIT in these groups.
Collapse
Affiliation(s)
- Catherine F. S. Marriott
- Centre for Studies in Family Medicine, Department of Family Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON Canada
| | - Andrea F. M. Petrella
- Centre for Studies in Family Medicine, Department of Family Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON Canada
| | - Emily C. S. Marriott
- Centre for Studies in Family Medicine, Department of Family Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON Canada
| | - Narlon C. Boa Sorte Silva
- Centre for Studies in Family Medicine, Department of Family Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON Canada
- Aging, Mobility, and Cognitive Neuroscience Lab, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC Canada
- Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Robert J. Petrella
- Centre for Studies in Family Medicine, Department of Family Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON Canada
- School of Kinesiology, Western University, London, ON Canada
- School of Kinesiology, Faculty of Education, University of British Columbia, Vancouver, BC Canada
- Department of Family Practice, Faculty of Medicine, University of British Columbia, 320 - 5950 University Boulevard, Vancouver, BC V6T 1Z3 Canada
| |
Collapse
|
16
|
Callahan MJ, Parr EB, Hawley JA, Camera DM. Can High-Intensity Interval Training Promote Skeletal Muscle Anabolism? Sports Med 2021; 51:405-421. [PMID: 33512698 DOI: 10.1007/s40279-020-01397-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Exercise training in combination with optimal nutritional support is an effective strategy to maintain or increase skeletal muscle mass. A single bout of resistance exercise undertaken with adequate protein availability increases rates of muscle protein synthesis and, when repeated over weeks and months, leads to increased muscle fiber size. While resistance-based training is considered the 'gold standard' for promoting muscle hypertrophy, other modes of exercise may be able to promote gains in muscle mass. High-intensity interval training (HIIT) comprises short bouts of exercise at or above the power output/speed that elicits individual maximal aerobic capacity, placing high tensile stress on skeletal muscle, and somewhat resembling the demands of resistance exercise. While HIIT induces rapid increases in skeletal muscle oxidative capacity, the anabolic potential of HIIT for promoting concurrent gains in muscle mass and cardiorespiratory fitness has received less scientific inquiry. In this review, we discuss studies that have determined muscle growth responses after HIIT, with a focus on molecular responses, that provide a rationale for HIIT to be implemented among populations who are susceptible to muscle loss (e.g. middle-aged or older adults) and/or in clinical settings (e.g. pre- or post-surgery).
Collapse
Affiliation(s)
- Marcus J Callahan
- Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, 215 Spring street, Melbourne, VIC, 3000, Australia
| | - Evelyn B Parr
- Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, 215 Spring street, Melbourne, VIC, 3000, Australia
| | - John A Hawley
- Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, 215 Spring street, Melbourne, VIC, 3000, Australia.
| | - Donny M Camera
- Department of Health and Medical Sciences, Swinburne University of Technology, Melbourne, VIC, Australia
| |
Collapse
|
17
|
McCormick JJ, King KE, Côté MD, Meade RD, Akerman AP, Kenny GP. Impaired autophagy following ex vivo heating at physiologically relevant temperatures in peripheral blood mononuclear cells from elderly adults. J Therm Biol 2020; 95:102790. [PMID: 33454031 DOI: 10.1016/j.jtherbio.2020.102790] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 10/25/2022]
Abstract
With the increasing threat of climate change and the accompanying rise in the frequency and severity of extreme heat events, there are growing health concerns for heat-vulnerable elderly adults. Elderly adults are at increased risk of developing heat-related injuries, in part due to age-related declines in thermoregulatory and cellular function. Regarding the latter, the process of autophagy is activated as a cellular protective mechanism to counter heat-induced stress, but the extent that heat stress activates autophagy in elderly adults is not known. Further, the interplay between autophagy, the heat shock response (HSR), the acute inflammatory response, and apoptosis remains poorly understood in elderly adults. Therefore, the purpose of this study was to examine changes in autophagy, the HSR, inflammation, and apoptosis following increasing levels of ex vivo heat stress representative of physiologically relevant increases in body core temperatures (37-41 °C). Whole blood from 20 elderly adults (72 ± 4 years; 14 men, 6 women) was heated (via water immersion) to temperatures representative of normal resting conditions (normothermia; 37 °C), in addition to moderate and severe heat stress conditions (39, and 41 °C, respectively) for 90 min. Peripheral blood mononuclear cells (PBMC) were isolated and protein markers of autophagy, the HSR, acute inflammation, and apoptosis were examined. No significant increases in markers of autophagy or the HSR were observed following any temperature condition. However, an increase in acute inflammation was observed above baseline following moderate heat stress (39 °C), with further increases in inflammation and apoptosis observed during severe heat stress (41 °C). Our findings indicate that PBMCs from elderly adults do not exhibit increases in autophagy or the HSR following severe heat stress, potentially contributing to the elevated risk of cellular dysfunction seen in elderly adults during heat stress.
Collapse
Affiliation(s)
- James J McCormick
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Canada
| | - Kelli E King
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Canada
| | - Melissa D Côté
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Canada
| | - Robert D Meade
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Canada
| | - Ashley P Akerman
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Canada
| | - Glen P Kenny
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Canada; Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
18
|
Pesce M, Ballerini P, Paolucci T, Puca I, Farzaei MH, Patruno A. Irisin and Autophagy: First Update. Int J Mol Sci 2020; 21:ijms21207587. [PMID: 33066678 PMCID: PMC7588919 DOI: 10.3390/ijms21207587] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 02/08/2023] Open
Abstract
Aging and sedentary life style are considered independent risk factors for many disorders. Under these conditions, accumulation of dysfunctional and damaged cellular proteins and organelles occurs, resulting in a cellular degeneration and cell death. Autophagy is a conserved recycling pathway responsible for the degradation, then turnover of cellular proteins and organelles. This process is a part of the molecular underpinnings by which exercise promotes healthy aging and mitigate age-related pathologies. Irisin is a myokine released during physical activity and acts as a link between muscles and other tissues and organs. Its main beneficial function is the change of subcutaneous and visceral adipose tissue into brown adipose tissue, with a consequential increase in thermogenesis. Irisin modulates metabolic processes, acting on glucose homeostasis, reduces systemic inflammation, maintains the balance between resorption and bone formation, and regulates the functioning of the nervous system. Recently, some of its pleiotropic and favorable properties have been attributed to autophagy induction, posing irisin as an important regulator of autophagy by exercise. This review article proposes to bring together for the first time the "state of the art" knowledge regarding the effects of irisin and autophagy. Furthermore, treatments on relation between exercise/myokines and autophagy have been also achieved.
Collapse
Affiliation(s)
- Mirko Pesce
- Department of Medicine and Aging Sciences, University G. d’Annunzio, 66100 Chieti, Italy; (M.P.); (A.P.)
| | - Patrizia Ballerini
- Department of Neurosciences, Imaging and Clinical Sciences, University G. d’Annunzio, 66100 Chieti, Italy
- Correspondence:
| | - Teresa Paolucci
- Department of Oral, Medical and Biotechnological Sciences, University G. d’Annunzio, 66100 Chieti, Italy;
| | - Iris Puca
- Sport Academy SSD, 65010 Pescara, Italy;
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, 67146 Kermanshah, Iran;
| | - Antonia Patruno
- Department of Medicine and Aging Sciences, University G. d’Annunzio, 66100 Chieti, Italy; (M.P.); (A.P.)
| |
Collapse
|
19
|
Gritsenko A, Green JP, Brough D, Lopez-Castejon G. Mechanisms of NLRP3 priming in inflammaging and age related diseases. Cytokine Growth Factor Rev 2020; 55:15-25. [PMID: 32883606 PMCID: PMC7571497 DOI: 10.1016/j.cytogfr.2020.08.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023]
Abstract
The NLRP3 inflammasome is a vital part of the innate immune response, whilst its aberrant activation drives the progression of a number of non-communicable diseases. Thus, NLRP3 inflammasome assembly must be tightly controlled at several checkpoints. The priming step of NLRP3 inflammasome activation is associated with increased NLRP3 gene expression, as well as post-translational modifications that control NLRP3 levels and licence the NLRP3 protein for inflammasome assembly. Increasing life expectancy in modern society is accompanied by a growing percentage of elderly individuals. The process of aging is associated with chronic inflammation that drives and/or worsens a range of age related non-communicable conditions. The NLRP3 inflammasome is known to contribute to pathological inflammation in many settings, but the mechanisms that prime NLRP3 for activation throughout aging and related co-morbidities have not been extensively reviewed. Here we dissect the biochemical changes that occur during aging and the pathogenesis of age related diseases and analyse the mechanisms by which they prime the NLRP3 inflammasome, thus exacerbating inflammation.
Collapse
Affiliation(s)
- Anna Gritsenko
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Jack P Green
- Lydia Becker Institute of Immunology and Inflammation, Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - David Brough
- Lydia Becker Institute of Immunology and Inflammation, Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Gloria Lopez-Castejon
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
20
|
The Effect of Low-Volume High-Intensity Interval Training on Body Composition and Cardiorespiratory Fitness: A Systematic Review and Meta-Analysis. Sports Med 2020; 49:1687-1721. [PMID: 31401727 DOI: 10.1007/s40279-019-01167-w] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Evidence for the efficacy of low-volume high-intensity interval training (HIIT) for the modulation of body composition is unclear. OBJECTIVES We examined the effect of low-volume HIIT versus a non-exercising control and moderate-intensity continuous training (MICT) on body composition and cardiorespiratory fitness in normal weight, overweight and obese adults. We evaluated the impact of low-volume HIIT (HIIT interventions where the total amount of exercise performed during training was ≤ 500 metabolic equivalent minutes per week [MET-min/week]) compared to a non-exercising control and MICT. METHODS A database search was conducted in PubMed (MEDLINE), EMBASE, CINAHL, Web of Science, SPORTDiscus and Scopus from the earliest record to June 2019 for studies (randomised controlled trials and non-randomised controlled trials) with exercise training interventions with a minimum 4-week duration. Meta-analyses were conducted for between-group (low-volume HIIT vs. non-exercising control and low-volume HIIT vs. MICT) comparisons for change in total body fat mass (kg), body fat percentage (%), lean body mass (kg) and cardiorespiratory fitness. RESULTS From 11,485 relevant records, 47 studies were included. No difference was found between low-volume HIIT and a non-exercising control on total body fat mass (kg) (effect size [ES]: - 0.129, 95% confidence interval [CI] - 0.468 to 0.210; p = 0.455), body fat (%) (ES: - 0.063, 95% CI - 0.383 to 0.257; p = 0.700) and lean body mass (kg) (ES: 0.050, 95% CI - 0.250 to 0.351; p = 0.744), or between low-volume HIIT and MICT on total body fat mass (kg) (ES: - 0.021, 95% CI - 0.272 to 0.231; p = 0.872), body fat (%) (ES: 0.005, 95% CI - 0.294 to 0.304; p = 0.974) and lean body mass (kg) (ES: 0.030, 95% CI - 0.167 to 0.266; p = 0.768). However, low-volume HIIT significantly improved cardiorespiratory fitness compared with a non-exercising control (p < 0.001) and MICT (p = 0.017). CONCLUSION These data suggest that low-volume HIIT is inefficient for the modulation of total body fat mass or total body fat percentage in comparison with a non-exercise control and MICT. A novel finding of our meta-analysis was that there appears to be no significant effect of low-volume HIIT on lean body mass when compared with a non-exercising control, and while most studies tended to favour improvement in lean body mass with low-volume HIIT versus MICT, this was not significant. However, despite its lower training volume, low-volume HIIT induces greater improvements in cardiorespiratory fitness than a non-exercising control and MICT in normal weight, overweight and obese adults. Low-volume HIIT, therefore, appears to be a time-efficient treatment for increasing fitness, but not for the improvement of body composition.
Collapse
|
21
|
Wang L, Wang J, Cretoiu D, Li G, Xiao J. Exercise-mediated regulation of autophagy in the cardiovascular system. JOURNAL OF SPORT AND HEALTH SCIENCE 2020; 9:203-210. [PMID: 32444145 PMCID: PMC7242217 DOI: 10.1016/j.jshs.2019.10.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/07/2019] [Accepted: 09/04/2019] [Indexed: 05/02/2023]
Abstract
Cardiovascular disease is the leading cause of human death worldwide. Autophagy is an evolutionarily conserved degradation pathway, which is a highly conserved cellular degradation process in which lysosomes decompose their own organelles and recycle the resulting macromolecules. Autophagy is critical in maintaining cardiovascular homeostasis and function, and excessive or insufficient autophagy or autophagic flux can lead to cardiovascular disease. Enormous evidence indicates that exercise training plays a beneficial role in the prevention and treatment of cardiovascular diseases. The regulation of autophagy during exercise is a bidirectional process. For cardiovascular disease caused by either insufficient or excessive autophagy, exercise training restores normal autophagy function and delays the progression of cardiovascular disease. An in-depth exploration and discussion of exercise-mediated regulation of autophagy in the cardiovascular system can broaden our view about the prevention of various autophagy-related diseases through exercise training. In this article, we review autophagy and its related signaling pathways, as well as autophagy-dependent beneficial effects of exercise in cardiovascular system.
Collapse
Affiliation(s)
- Lijun Wang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Jiaqi Wang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Dragos Cretoiu
- Department of Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, Bucharest 050474, Romania; Alessandrescu-Rusescu National Institute of Mother and Child Health, Fetal Medicine Excellence Research Center, Bucharest 020395, Romania
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02215, USA
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
22
|
Aerobic Training Down-Regulates Pentraxin 3 and Pentraxin 3/Toll-Like Receptor 4 Ratio, Irrespective of Oxidative Stress Response, in Elderly Subjects. Antioxidants (Basel) 2020; 9:antiox9020110. [PMID: 32012711 PMCID: PMC7070734 DOI: 10.3390/antiox9020110] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 01/14/2023] Open
Abstract
Reactive oxygen and nitrogen species-mediated cellular aging has been linked to diseases such as atherothrombosis and cancer. Although pentraxin 3 (PTX3) is associated with aging-related diseases via TLR4-dependent anti-inflammatory effects, its relationship with oxidative stress in aging remains to be elucidated. Exercise is proposed as the key intervention for health maintenance in the elderly. This study aimed to examine the association of PTX3 levels with changes in oxidative stress in both plasma and peripheral blood mononuclear cells (PBMCs), following aerobic training in elderly adults. Nine trained and five controls participated in an eight-week aerobic training protocol. Enzyme-linked immunosorbent assay (ELISA) and Western blot analyses were used to determine PTX3, toll-like receptor 4 (TLR4), and levels of oxidative stress biomarkers [3-nitrotyrosine (3NT), 4-hydroxynonenal (4-HNE), glutathione (GSH), protein carbonyl (PC), reactive oxygen/ nitrogen species (ROS/RNS), and trolox equivalent antioxidant capacity (TEAC)] in plasma and/or PBMCs. Results showed a down-regulation of PTX3 expression in PBMCs following aerobic training, along with decreased PTX3/TLR4 ratios. Oxidative stress responses in PBMCs remained unchanged with the exercise protocol. Comparable levels of plasma PTX3 and oxidative stress biomarkers were observed in trained vs. control groups. No correlation was found between PTX3 and any oxidative stress biomarkers following training. These findings demonstrated the down-regulation of PTX3 and PTX3/TLR4 ratio, irrespective of oxidative stress response, in elderly adults following eight weeks of aerobic training.
Collapse
|
23
|
Abstract
Bone and skeletal muscle are integrated organs and their coupling has been considered mainly a mechanical one in which bone serves as attachment site to muscle while muscle applies load to bone and regulates bone metabolism. However, skeletal muscle can affect bone homeostasis also in a non-mechanical fashion, i.e., through its endocrine activity. Being recognized as an endocrine organ itself, skeletal muscle secretes a panel of cytokines and proteins named myokines, synthesized and secreted by myocytes in response to muscle contraction. Myokines exert an autocrine function in regulating muscle metabolism as well as a paracrine/endocrine regulatory function on distant organs and tissues, such as bone, adipose tissue, brain and liver. Physical activity is the primary physiological stimulus for bone anabolism (and/or catabolism) through the production and secretion of myokines, such as IL-6, irisin, IGF-1, FGF2, beside the direct effect of loading. Importantly, exercise-induced myokine can exert an anti-inflammatory action that is able to counteract not only acute inflammation due to an infection, but also a condition of chronic low-grade inflammation raised as consequence of physical inactivity, aging or metabolic disorders (i.e., obesity, type 2 diabetes mellitus). In this review article, we will discuss the effects that some of the most studied exercise-induced myokines exert on bone formation and bone resorption, as well as a brief overview of the anti-inflammatory effects of myokines during the onset pathological conditions characterized by the development a systemic low-grade inflammation, such as sarcopenia, obesity and aging.
Collapse
Affiliation(s)
- Marta Gomarasca
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Experimental Biochemistry & Molecular Biology, Milan, Italy
| | - Giuseppe Banfi
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Experimental Biochemistry & Molecular Biology, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Giovanni Lombardi
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Experimental Biochemistry & Molecular Biology, Milan, Italy; Gdańsk University of Physical Education & Sport, Gdańsk, Pomorskie, Poland.
| |
Collapse
|
24
|
Batatinha HAP, Diniz TA, de Souza Teixeira AA, Krüger K, Rosa-Neto JC. Regulation of autophagy as a therapy for immunosenescence-driven cancer and neurodegenerative diseases: The role of exercise. J Cell Physiol 2019; 234:14883-14895. [PMID: 30756377 DOI: 10.1002/jcp.28318] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/26/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
Aging is one of the risk factors for the development of low-grade inflammation morbidities, such as several types of cancer and neurodegenerative diseases, due to changes in the metabolism, hormonal secretion, and immunosenescence. The senescence of the immune system leads to improper control of infections and tissue damage increasing age-related diseases. One of the mechanisms that maintain cellular homeostasis is autophagy, a cell-survival mechanism, and it has been proposed as one of the most powerful antiaging therapies. Regular exercise can reestablish autophagy, probably through AMP-activated protein kinase activation, and help in reducing the age-related senescence diseases. Therefore, in this study, we discuss the effects of exercise training in immunosenescence and autophagy, preventing the two main age-related disease, cancer and neurodegeneration.
Collapse
Affiliation(s)
| | - Tiego Aparecido Diniz
- Department of Cell and Developmental Biology, University of São Paulo, São Paulo, São Paulo, Brazil
| | | | - Karsten Krüger
- Department Exercise and Health, Institute of Sports Science, Leibniz University Hannover, Hannover, Germany
| | - Jose Cesar Rosa-Neto
- Department of Cell and Developmental Biology, University of São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
25
|
de Lemos Muller CH, de Matos JR, Grigolo GB, Schroeder HT, Rodrigues-Krause J, Krause M. Exercise Training for the Elderly: Inflammaging and the Central Role for HSP70. ACTA ACUST UNITED AC 2019. [DOI: 10.1007/s42978-019-0015-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
26
|
Risk of Falls in Healthy Older Adults: Benefits of High-Intensity Interval Training Using Lower Body Suspension Exercises. J Aging Phys Act 2019; 27:325-333. [DOI: 10.1123/japa.2018-0190] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
27
|
The beneficial roles of exercise training via autophagy in neurological diseases and possible mechanisms. Life Sci 2019; 221:130-134. [PMID: 30769113 DOI: 10.1016/j.lfs.2019.02.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 02/04/2019] [Accepted: 02/11/2019] [Indexed: 02/06/2023]
Abstract
Autophagy is a conservative catabolism process, participating in delivering the cytosol and cytosolic components to the lysosome. Abnormal autophagy is related to human pathologies, for instance diabetes, neurodegeneration, cardiovascular, macular degeneration, pulmonary, and cancer. Enormous evidences indicate that autophagy may mediate the cellular pathological condition in the process of neurological diseases. Exercise as a form of physiological stress may cause an adaptation, and autophagy is a necessary process for adaptational response to exercise. Autophagy during exercise may improve neurological function, control tissue maintain tissue integrity, and activate different signals pathway for adaptation. In this review, we summarize the possible mechanisms of exercise training via autophagy in neurological diseases.
Collapse
|
28
|
Escobar KA, Cole NH, Mermier CM, VanDusseldorp TA. Autophagy and aging: Maintaining the proteome through exercise and caloric restriction. Aging Cell 2019; 18:e12876. [PMID: 30430746 PMCID: PMC6351830 DOI: 10.1111/acel.12876] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 08/31/2018] [Accepted: 09/28/2018] [Indexed: 12/16/2022] Open
Abstract
Accumulation of dysfunctional and damaged cellular proteins and organelles occurs during aging, resulting in a disruption of cellular homeostasis and progressive degeneration and increases the risk of cell death. Moderating the accrual of these defunct components is likely a key in the promotion of longevity. While exercise is known to promote healthy aging and mitigate age‐related pathologies, the molecular underpinnings of this phenomenon remain largely unclear. However, recent evidences suggest that exercise modulates the proteome. Similarly, caloric restriction (CR), a known promoter of lifespan, is understood to augment intracellular protein quality. Autophagy is an evolutionary conserved recycling pathway responsible for the degradation, then turnover of cellular proteins and organelles. This housekeeping system has been reliably linked to the aging process. Moreover, autophagic activity declines during aging. The target of rapamycin complex 1 (TORC1), a central kinase involved in protein translation, is a negative regulator of autophagy, and inhibition of TORC1 enhances lifespan. Inhibition of TORC1 may reduce the production of cellular proteins which may otherwise contribute to the deleterious accumulation observed in aging. TORC1 may also exert its effects in an autophagy‐dependent manner. Exercise and CR result in a concomitant downregulation of TORC1 activity and upregulation of autophagy in a number of tissues. Moreover, exercise‐induced TORC1 and autophagy signaling share common pathways with that of CR. Therefore, the longevity effects of exercise and CR may stem from the maintenance of the proteome by balancing the synthesis and recycling of intracellular proteins and thus may represent practical means to promote longevity.
Collapse
Affiliation(s)
- Kurt A. Escobar
- Department of Kinesiology; California State University, Long Beach; Long Beach California
| | - Nathan H. Cole
- Department of Health, Exercise, & Sports Sciences; University of New Mexico; Albuquerque New Mexico
| | - Christine M. Mermier
- Department of Health, Exercise, & Sports Sciences; University of New Mexico; Albuquerque New Mexico
| | - Trisha A. VanDusseldorp
- Department of Exercise Science & Sports Management; Kennesaw State University; Kennesaw Georgia
| |
Collapse
|
29
|
Arribat Y, Broskey NT, Greggio C, Boutant M, Conde Alonso S, Kulkarni SS, Lagarrigue S, Carnero EA, Besson C, Cantó C, Amati F. Distinct patterns of skeletal muscle mitochondria fusion, fission and mitophagy upon duration of exercise training. Acta Physiol (Oxf) 2019; 225:e13179. [PMID: 30144291 DOI: 10.1111/apha.13179] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 08/17/2018] [Accepted: 08/20/2018] [Indexed: 12/17/2022]
Abstract
AIM Healthy ageing interventions encompass regular exercise to prevent mitochondrial dysfunction, key player in sarcopenia pathogenesis. Mitochondrial biogenesis has been well documented, but mitochondrial remodelling in response to exercise training is poorly understood. Here we investigated fusion, fission and mitophagy before and after an exercise intervention in older adults. METHODS Skeletal muscle biopsies were collected from 22 healthy sedentary men and women before and after 4 months of supervised training. Eight lifelong trained age- and gender-matched volunteers served as positive controls. Transmission electron microscopy was used to estimate mitochondrial content. Western blotting and qRT-PCR were used to detect changes in specific proteins and transcripts. RESULTS After intervention, mitochondrial content increased to levels of controls. While enhancement of fusion was prevalent after intervention, inhibition of fission and increased mitophagy were dominant in controls. Similarly to PARKIN, BCL2L13 content was higher in controls. The observed molecular adaptations paralleled long-term effects of training on physical fitness, exercise efficiency and oxidative capacity. CONCLUSIONS This study describes distinct patterns of molecular adaptations in human skeletal muscle under chronic exercise training. After 16 weeks of exercise, the pattern was dominated by fusion to increase mitochondrial content to the metabolic demands of exercise. In lifelong exercise, the pattern was dominated by mitophagy synchronized with increased fusion and decreased fission, indicating an increased mitochondrial turnover. In addition to these temporally distinct adaptive mechanisms, this study suggests for the first time a specific role of BCL2L13 in chronic exercise that requires constant maintenance of mitochondrial quality.
Collapse
Affiliation(s)
- Yoan Arribat
- Aging and Muscle Metabolism Lab; Department of Physiology; University of Lausanne; Lausanne Switzerland
| | - Nicholas T. Broskey
- Aging and Muscle Metabolism Lab; Department of Physiology; University of Lausanne; Lausanne Switzerland
| | - Chiara Greggio
- Aging and Muscle Metabolism Lab; Department of Physiology; University of Lausanne; Lausanne Switzerland
| | - Marie Boutant
- Nestlé Institute of Health Sciences; Lausanne Switzerland
| | - Sonia Conde Alonso
- Aging and Muscle Metabolism Lab; Department of Physiology; University of Lausanne; Lausanne Switzerland
| | | | - Sylviane Lagarrigue
- Aging and Muscle Metabolism Lab; Department of Physiology; University of Lausanne; Lausanne Switzerland
| | - Elvis A. Carnero
- Aging and Muscle Metabolism Lab; Department of Physiology; University of Lausanne; Lausanne Switzerland
| | - Cyril Besson
- Sport Medicine Unit; University Hospital (CHUV); Lausanne Switzerland
| | - Carles Cantó
- Nestlé Institute of Health Sciences; Lausanne Switzerland
| | - Francesca Amati
- Aging and Muscle Metabolism Lab; Department of Physiology; University of Lausanne; Lausanne Switzerland
- Sport Medicine Unit; University Hospital (CHUV); Lausanne Switzerland
- Institute of Sports Sciences (ISSUL); University of Lausanne; Lausanne Switzerland
- Department of Medicine; Service of Endocrinology, Diabetology and Metabolism; University Hospital (CHUV); Lausanne Switzerland
| |
Collapse
|
30
|
Affiliation(s)
- Andrea del Campo
- Escuela de Química y Farmacia, Facultad de Ingeniería; Ciencia y Tecnología, Universidad Bernardo O´Higgins; Santiago Chile
| |
Collapse
|
31
|
Estébanez B, Moreira OC, Almar M, de Paz JA, Gonzalez-Gallego J, Cuevas MJ. Effects of a resistance-training programme on endoplasmic reticulum unfolded protein response and mitochondrial functions in PBMCs from elderly subjects. Eur J Sport Sci 2019; 19:931-940. [PMID: 30614406 DOI: 10.1080/17461391.2018.1561950] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Aging has been related with a decline in the ability to handle protein folding, which leads to endoplasmic reticulum stress and alterations in unfolded protein response (UPR). Importantly, physical activity could activate the UPR and attenuate or prevent age-induced endoplasmic reticulum (ER) dysfunction. The current study evaluated the effects of a resistance exercise on UPR and mitochondrial functions in peripheral blood mononuclear cells (PBMCs) from elderly subjects. Thirty healthy women and men (age, 72.8, sx- = 2.2 years) were randomized to a training group, which performed an 8-week resistance training programme, or a control group, which followed their daily routines. The phosphorylation of PERK and IRE1, as well as ATF4, and XBP1 protein expression, significantly increased following the training, while expression of BiP, AFT6 and CHOP remain without changes. Additionally, the intervention also induced an increase in PGC-1α and Mfn1 protein levels, while no changes were found in Drp1 expression. Finally, the resistance protocol was not able to activate PINK1/Parkin and Bnip3/Nix pathways. The results obtained seem to indicate that 8-week resistance exercise activates the UPR, stimulates mitochondrial biogenesis, maintains mitochondrial dynamics and prevents mitophagy activation by unfolded proteins in PBMCs from elderly subjects.
Collapse
Affiliation(s)
- Brisamar Estébanez
- a Institute of Biomedicine (IBIOMED) , University of León , León , Spain
| | - Osvaldo C Moreira
- a Institute of Biomedicine (IBIOMED) , University of León , León , Spain.,b Institute of Biological Sciences and Health , Federal University of Viçosa - Campus Florestal , Florestal , Brazil
| | - Mar Almar
- a Institute of Biomedicine (IBIOMED) , University of León , León , Spain
| | - José A de Paz
- a Institute of Biomedicine (IBIOMED) , University of León , León , Spain
| | | | - María J Cuevas
- a Institute of Biomedicine (IBIOMED) , University of León , León , Spain
| |
Collapse
|
32
|
Estébanez B, de Paz JA, Cuevas MJ, González-Gallego J. Endoplasmic Reticulum Unfolded Protein Response, Aging and Exercise: An Update. Front Physiol 2018; 9:1744. [PMID: 30568599 PMCID: PMC6290262 DOI: 10.3389/fphys.2018.01744] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 11/20/2018] [Indexed: 12/14/2022] Open
Abstract
The endoplasmic reticulum (ER) is a dynamic and multifunctional organelle responsible for protein biosynthesis, folding, assembly and modifications. Loss of protein folding regulation, which leads to unfolded or misfolded proteins accumulation inside the ER lumen, drives ER stress (ERS) and unfolded protein response (UPR) activation. During aging, there is a decline in the ability of the cell to handle protein folding, accumulation and aggregation, and the function of UPR is compromised. There is a progressive failure of the chaperoning systems and a decline in many of its components, so that the UPR activation cannot rescue the ERS. Physical activity has been proposed as a powerful tool against aged-related diseases, which are linked to ERS. Interventional studies have demonstrated that regular exercise is able to decrease oxidative stress and inflammation and reverse mitochondrial and ER dysfunctions. Exercise-induced metabolic stress could activate the UPR since muscle contraction is directly involved in its activation, mediating exercise-induced adaptation responses. In fact, regular moderate-intensity exercise-induced ERS acts as a protective mechanism against current and future stressors. However, biological responses vary according to exercise intensity and therefore induce different degrees of ERS and UPR activation. This article reviews the effects of aging and exercise on ERS and UPR, also analyzing possible changes induced by different types of exercise in elderly subjects.
Collapse
Affiliation(s)
| | - José A de Paz
- Institute of Biomedicine (IBIOMED), University of León, León, Spain
| | - María J Cuevas
- Institute of Biomedicine (IBIOMED), University of León, León, Spain
| | - Javier González-Gallego
- Institute of Biomedicine (IBIOMED), University of León, León, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| |
Collapse
|
33
|
Carter HN, Kim Y, Erlich AT, Zarrin‐khat D, Hood DA. Autophagy and mitophagy flux in young and aged skeletal muscle following chronic contractile activity. J Physiol 2018; 596:3567-3584. [PMID: 29781176 PMCID: PMC6092298 DOI: 10.1113/jp275998] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/02/2018] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS A healthy mitochondrial pool is dependent on the removal of dysfunctional organelles via mitophagy, but little is known about how mitophagy is altered with ageing and chronic exercise. Chronic contractile activity (CCA) is a standardized exercise model that can elicit mitochondrial adaptations in both young and aged muscle, albeit to a lesser degree in the aged group. Assessment of mitophagy flux revealed enhanced targeting of mitochondria for degradation in aged muscle, in contrast to previous theories. Mitophagy flux was significantly reduced as an adaptation to CCA suggesting that an improvement in organelle quality reduces the need for mitochondrial turnover. CCA enhances lysosomal capacity and may ameliorate lysosomal dysfunction in aged muscle. ABSTRACT Skeletal muscle exhibits deficits in mitochondrial quality with age. Central to the maintenance of a healthy mitochondrial pool is the removal of dysfunctional organelles via mitophagy. Little is known on how mitophagy is altered with ageing and chronic exercise. We assessed mitophagy flux using colchicine treatment in vivo following chronic contractile activity (CCA) of muscle in young and aged rats. CCA evoked mitochondrial biogenesis in young muscle, with an attenuated response in aged muscle. Mitophagy flux was higher in aged muscle and was correlated with the enhanced expression of mitophagy receptors and upstream transcriptional regulators. CCA decreased mitophagy flux in both age groups, suggesting an improvement in organelle quality. CCA also reduced the exaggerated expression of TFEB evident in aged muscle, which may be promoting the age-induced increase in lysosomal markers. Thus, aged muscle possesses an elevated drive for autophagy and mitophagy which may contribute to the decline in organelle content observed with age, but which may serve to maintain mitochondrial quality. CCA improves organelle integrity and reduces mitophagy, illustrating that chronic exercise is a modality to improve muscle quality in aged populations.
Collapse
Affiliation(s)
- Heather N. Carter
- Muscle Health Research Centre, School of Kinesiology and Health ScienceYork UniversityTorontoOntarioM3J 1P3Canada
| | - Yuho Kim
- Muscle Health Research Centre, School of Kinesiology and Health ScienceYork UniversityTorontoOntarioM3J 1P3Canada
| | - Avigail T. Erlich
- Muscle Health Research Centre, School of Kinesiology and Health ScienceYork UniversityTorontoOntarioM3J 1P3Canada
| | - Dorrin Zarrin‐khat
- Muscle Health Research Centre, School of Kinesiology and Health ScienceYork UniversityTorontoOntarioM3J 1P3Canada
- Department of BiologyYork UniversityTorontoOntarioM3J 1P3Canada
| | - David A. Hood
- Muscle Health Research Centre, School of Kinesiology and Health ScienceYork UniversityTorontoOntarioM3J 1P3Canada
| |
Collapse
|
34
|
Cardinale DA, Lilja M, Mandić M, Gustafsson T, Larsen FJ, Lundberg TR. Resistance Training with Co-ingestion of Anti-inflammatory Drugs Attenuates Mitochondrial Function. Front Physiol 2017; 8:1074. [PMID: 29311990 PMCID: PMC5742251 DOI: 10.3389/fphys.2017.01074] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/06/2017] [Indexed: 01/09/2023] Open
Abstract
Aim: The current study aimed to examine the effects of resistance exercise with concomitant consumption of high vs. low daily doses of non-steroidal anti-inflammatory drugs (NSAIDs) on mitochondrial oxidative phosphorylation in skeletal muscle. As a secondary aim, we compared the effects of eccentric overload with conventional training. Methods: Twenty participants were randomized to either a group taking high doses (3 × 400 mg/day) of ibuprofen (IBU; 27 ± 5 year; n = 11) or a group ingesting a low dose (1 × 75 mg/day) of acetylsalicylic acid (ASA; 26 ± 4 year; n = 9) during 8 weeks of supervised knee extensor resistance training. Each of the subject's legs were randomized to complete the training program using either a flywheel (FW) device emphasizing eccentric overload, or a traditional weight stack machine (WS). Maximal mitochondrial oxidative phosphorylation (CI+IIP) from permeabilized skeletal muscle bundles was assessed using high-resolution respirometry. Citrate synthase (CS) activity was assessed using spectrophotometric techniques and mitochondrial protein content using western blotting. Results: After training, CI+IIP decreased (P < 0.05) in both IBU (23%) and ASA (29%) with no difference across medical treatments. Although CI+IIP decreased in both legs, the decrease was greater (interaction p = 0.015) in WS (33%, p = 0.001) compared with FW (19%, p = 0.078). CS activity increased (p = 0.027) with resistance training, with no interactions with medical treatment or training modality. Protein expression of ULK1 increased with training in both groups (p < 0.001). The increase in quadriceps muscle volume was not correlated with changes in CI+IIP (R = 0.16). Conclusion: These results suggest that 8 weeks of resistance training with co-ingestion of anti-inflammatory drugs reduces mitochondrial function but increases mitochondrial content. The observed changes were not affected by higher doses of NSAIDs consumption, suggesting that the resistance training intervention was the prime mediator of the decreased mitochondrial phosphorylation. Finally, we noted that flywheel resistance training, emphasizing eccentric overload, rescued some of the reduction in mitochondrial function seen with conventional resistance training.
Collapse
Affiliation(s)
- Daniele A Cardinale
- Åstrand Laboratory, The Swedish School of Sport and Health Sciences, Stockholm, Sweden.,Elite Performance Centre, Bosön-Swedish Sports Confederation, Lidingö, Sweden
| | - Mats Lilja
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, and Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | - Mirko Mandić
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, and Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas Gustafsson
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, and Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | - Filip J Larsen
- Åstrand Laboratory, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Tommy R Lundberg
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, and Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
35
|
Mejías-Peña Y, Estébanez B, Rodriguez-Miguelez P, Fernandez-Gonzalo R, Almar M, de Paz JA, González-Gallego J, Cuevas MJ. Impact of resistance training on the autophagy-inflammation-apoptosis crosstalk in elderly subjects. Aging (Albany NY) 2017; 9:408-418. [PMID: 28160545 PMCID: PMC5361672 DOI: 10.18632/aging.101167] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/25/2017] [Indexed: 12/13/2022]
Abstract
Aging is associated with a decline in autophagy and a state of low-grade inflammation which further affects apoptosis and autophagy. Importantly, these alterations could reverse with regular physical activity. This study assessed the effects of a resistance exercise training program on autophagy, NLRP3 inflammasome, and apoptosis in peripheral blood mononuclear cells (PBMCs) from old subjects. Twenty-six healthy women and men (age, 69.6±1.5 yr) were randomized to a training (TG) or a control (CG) group. TG performed an 8-week resistance training program, while CG followed their daily routines. Protein expression of beclin-1, Atg12, Atg16 and LAMP-2 increased following the training program, while expression of p62/SQSTM1 and phosphorylation of ULK-1 at Ser757 were significantly lower. Resistance exercise also induced a decrease in NLRP3 expression and in the caspase-1/procaspase-1 ratio. Expression of Bcl-2 and Bcl-xL, as well as the Bad/BcL-2 ratio were reduced, and there was a significant decrease in the protein content of caspase-3. The results obtained seem to indicate that 8-week resistance training stimulates autophagy, prevents NLRP3 inflammasome activation, and reduces apoptosis in PBMCs from elderly subjects. These data could have a significant impact in prevention and rehabilitation programs currently employed in elderly population.
Collapse
Affiliation(s)
| | | | - Paula Rodriguez-Miguelez
- Division of Clinical Translational Science, Georgia Prevention Institute, Department of Pediatrics, Augusta University, Augusta, GA, USA
| | - Rodrigo Fernandez-Gonzalo
- Radiobiology Unit, Laboratory of Molecular and Cellular Biology, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK•CEN), Mol, Belgium
| | - Mar Almar
- Institute of Biomedicine (IBIOMED), University of León, León, Spain
| | - José A de Paz
- Institute of Biomedicine (IBIOMED), University of León, León, Spain
| | | | - María J Cuevas
- Institute of Biomedicine (IBIOMED), University of León, León, Spain
| |
Collapse
|
36
|
Freitas HR, Ferreira GDC, Trevenzoli IH, Oliveira KDJ, de Melo Reis RA. Fatty Acids, Antioxidants and Physical Activity in Brain Aging. Nutrients 2017; 9:nu9111263. [PMID: 29156608 PMCID: PMC5707735 DOI: 10.3390/nu9111263] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 11/11/2017] [Accepted: 11/14/2017] [Indexed: 12/25/2022] Open
Abstract
Polyunsaturated fatty acids and antioxidants are important mediators in the central nervous system. Lipid derivatives may control the production of proinflammatory agents and regulate NF-κB activity, microglial activation, and fatty acid oxidation; on the other hand, antioxidants, such as glutathione and ascorbate, have been shown to signal through transmitter receptors and protect against acute and chronic oxidative stress, modulating the activity of different signaling pathways. Several authors have investigated the role of these nutrients in the brains of the young and the aged in degenerative diseases such as Alzheimer’s and Parkinson’s, and during brain aging due to adiposity- and physical inactivity-mediated metabolic disturbances, chronic inflammation, and oxidative stress. Through a literature review, we aimed to highlight recent data on the role of adiposity, fatty acids, antioxidants, and physical inactivity in the pathophysiology of the brain and in the molecular mechanisms of senescence. Data indicate the complexity and necessity of endogenous/dietary antioxidants for the maintenance of redox status and the control of neuroglial signaling under stress. Recent studies also indicate that omega-3 and -6 fatty acids act in a competitive manner to generate mediators for energy metabolism, influencing feeding behavior, neural plasticity, and memory during aging. Finding pharmacological or dietary resources that mitigate or prevent neurodegenerative affections continues to be a great challenge and requires additional effort from researchers, clinicians, and nutritionists in the field.
Collapse
Affiliation(s)
- Hércules Rezende Freitas
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
| | - Gustavo da Costa Ferreira
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
- Laboratory of Neuroenergetics and Inborn Errors of Metabolism, Institute of Medical Biochemistry Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
| | - Isis Hara Trevenzoli
- Laboratory of Molecular Endocrinology, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
| | - Karen de Jesus Oliveira
- Laboratory of Endocrine Physiology and Metabology, Biomedical Institute, Universidade Federal Fluminense, Niterói 24210-130, Brazil.
| | - Ricardo Augusto de Melo Reis
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
| |
Collapse
|
37
|
Mitochondrial Function and Mitophagy in the Elderly: Effects of Exercise. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:2012798. [PMID: 28900532 PMCID: PMC5576425 DOI: 10.1155/2017/2012798] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/04/2017] [Accepted: 07/06/2017] [Indexed: 12/22/2022]
Abstract
Aging is a natural, multifactorial and multiorganic phenomenon wherein there are gradual physiological and pathological changes over time. Aging has been associated with a decrease of autophagy capacity and mitochondrial functions, such as biogenesis, dynamics, and mitophagy. These processes are essential for the maintenance of mitochondrial structural integrity and, therefore, for cell life, since mitochondrial dysfunction leads to an impairment of energy metabolism and increased production of reactive oxygen species, which consequently trigger mechanisms of cellular senescence and apoptotic cell death. Moreover, reduced mitochondrial function can contribute to age-associated disease phenotypes in model organisms and humans. Literature data show beneficial effects of exercise on the impairment of mitochondrial biogenesis and dynamics and on the decrease in the mitophagic capacity associated to aging. Thus, exercise could have effects on the major cell signaling pathways that are involved in the mitochondria quality and quantity control in the elderly. Although it is known that several exercise protocols are able to modify the activity and turnover of mitochondria, further studies are necessary in order to better identify the mechanisms of interaction between mitochondrial functions, aging, and physical activity, as well as to analyze possible factors influencing these processes.
Collapse
|
38
|
Abstract
We present the hypothesis that an accumulation of dysfunctional mitochondria initiates a signaling cascade leading to motor neuron and muscle fiber death and culminating in sarcopenia. Interactions between neural and muscle cells that contain dysfunctional mitochondria exacerbate sarcopenia. Preventing sarcopenia will require identifying mitochondrial sources of dysfunction that are reversible.
Collapse
Affiliation(s)
- Stephen E Alway
- 1Division of Exercise Physiology; 2Center for Cardiovascular and Respiratory Sciences, and Mitochondria, Metabolism, and Bioenergetics; and 3Centers for Neuroscience, West Virginia University School of Medicine, Morgantown, WV
| | | | | |
Collapse
|
39
|
Zhang C, Li W, Wen J, Yang Z. Autophagy is involved in mouse kidney development and podocyte differentiation regulated by Notch signalling. J Cell Mol Med 2017; 21:1315-1328. [PMID: 28158917 PMCID: PMC5487928 DOI: 10.1111/jcmm.13061] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 11/18/2016] [Indexed: 01/19/2023] Open
Abstract
Podocyte dysfunction results in glomerular diseases accounted for 90% of end-stage kidney disease. The evolutionarily conserved Notch signalling makes a crucial contribution in podocyte development and function. However, the underlying mechanism of Notch pathway modulating podocyte differentiation remains less obvious. Autophagy, reported to be related with Notch signalling pathways in different animal models, is regarded as a possible participant during podocyte differentiation. Here, we found the dynamic changes of Notch1 were coincided with autophagy: they both increased during kidney development and podocyte differentiation. Intriguingly, when Notch signalling was down-regulated by DAPT, autophagy was greatly diminished, and differentiation was also impaired. Further, to better understand the relationship between Notch signalling and autophagy in podocyte differentiation, rapamycin was added to enhance autophagy levels in DAPT-treated cells, and as a result, nephrin was recovered and DAPT-induced injury was ameliorated. Therefore, we put forward that autophagy is involved in kidney development and podocyte differentiation regulated by Notch signalling.
Collapse
Affiliation(s)
- Chuyue Zhang
- School of MedicineState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive Materials Ministry of EducationNankai UniversityTianjinChina
| | - Wen Li
- School of MedicineState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive Materials Ministry of EducationNankai UniversityTianjinChina
| | - Junkai Wen
- School of MedicineState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive Materials Ministry of EducationNankai UniversityTianjinChina
| | - Zhuo Yang
- School of MedicineState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive Materials Ministry of EducationNankai UniversityTianjinChina
| |
Collapse
|