1
|
Smith ME, Wahl D, Cavalier AN, McWilliams GT, Rossman MJ, Giordano GR, Bryan AD, Seals DR, LaRocca TJ. Repetitive element transcript accumulation is associated with inflammaging in humans. GeroScience 2024; 46:5663-5679. [PMID: 38641753 PMCID: PMC11493880 DOI: 10.1007/s11357-024-01126-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 03/08/2024] [Indexed: 04/21/2024] Open
Abstract
Chronic, low-grade inflammation increases with aging, contributing to functional declines and diseases that reduce healthspan. Growing evidence suggests that transcripts from repetitive elements (RE) in the genome contribute to this "inflammaging" by stimulating innate immune activation, but evidence of RE-associated inflammation with aging in humans is limited. Here, we present transcriptomic and clinical data showing that RE transcript levels are positively related to gene expression of innate immune sensors, and to serum interleukin 6 (a marker of systemic inflammation), in a large group of middle-aged and older adults. We also: (1) use transcriptomics and whole-genome bisulfite (methylation) sequencing to show that many RE may be hypomethylated with aging, and that aerobic exercise, a healthspan-extending intervention, reduces RE transcript levels and increases RE methylation in older adults; and (2) extend our findings in a secondary dataset demonstrating age-related changes in RE chromatin accessibility. Collectively, our data support the idea that age-related RE transcript accumulation may play a role in inflammaging in humans, and that RE dysregulation with aging may be due in part to upstream epigenetic changes.
Collapse
Affiliation(s)
- Meghan E Smith
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Devin Wahl
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Alyssa N Cavalier
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Gabriella T McWilliams
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Matthew J Rossman
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Gregory R Giordano
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Angela D Bryan
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Thomas J LaRocca
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
2
|
Clemente-Suárez VJ, Rubio-Zarapuz A, Belinchón-deMiguel P, Beltrán-Velasco AI, Martín-Rodríguez A, Tornero-Aguilera JF. Impact of Physical Activity on Cellular Metabolism Across Both Neurodegenerative and General Neurological Conditions: A Narrative Review. Cells 2024; 13:1940. [PMID: 39682689 DOI: 10.3390/cells13231940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Regular physical activity plays a crucial role in modulating cellular metabolism and mitigating the progression of neurodegenerative diseases such as Alzheimer's, Parkinson's, and Multiple Sclerosis. OBJECTIVE The objective of this review is to evaluate the molecular mechanisms by which exercise influences cellular metabolism, with a focus on its potential as a therapeutic intervention for neurological disorders. METHODS A comprehensive literature review was conducted using peer-reviewed scientific articles, with a focus on the period between 2015 and 2024, to analyze the effects of exercise on mitochondrial function, oxidative stress, and metabolic health. RESULTS The findings indicate that exercise promotes mitochondrial biogenesis, enhances oxidative phosphorylation, and reduces reactive oxygen species, contributing to improved energy production and cellular resilience. These metabolic adaptations are associated with delayed disease progression and reduced symptoms in patients with neurodegenerative conditions. Additionally, integrating exercise with nutritional strategies may further enhance therapeutic outcomes by addressing metabolic disturbances comprehensively. CONCLUSIONS This review concludes that personalized exercise protocols should be developed to optimize metabolic benefits for patients with neurological diseases, while future research should focus on biomarker development for individualized treatment approaches. These findings highlight the importance of non-pharmacological interventions in managing neurodegenerative diseases.
Collapse
Affiliation(s)
- Vicente Javier Clemente-Suárez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain
- Grupo de Investigación en Cultura, Educación y Sociedad, Universidad de la Costa, Barranquilla 080002, Colombia
| | - Alejandro Rubio-Zarapuz
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain
| | - Pedro Belinchón-deMiguel
- Department of Nursing, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Villaviciosa de Odón, Spain
| | | | - Alexandra Martín-Rodríguez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain
- Faculty of Applied Social Sciences and Communications, Universidad Internacional de la Empresa (UNIE), 28015 Madrid, Spain
| | | |
Collapse
|
3
|
Day NJ, Kelly SS, Lui L, Mansfield TA, Gaffrey MJ, Trejo JB, Sagendorf TJ, Attah IK, Moore RJ, Douglas CM, Newman AB, Kritchevsky SB, Kramer PA, Marcinek DJ, Coen PM, Goodpaster BH, Hepple RT, Cawthon PM, Petyuk VA, Esser KA, Qian W, Cummings SR. Signatures of cysteine oxidation on muscle structural and contractile proteins are associated with physical performance and muscle function in older adults: Study of Muscle, Mobility and Aging (SOMMA). Aging Cell 2024; 23:e14094. [PMID: 38332629 PMCID: PMC11166363 DOI: 10.1111/acel.14094] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 02/10/2024] Open
Abstract
Oxidative stress is considered a contributor to declining muscle function and mobility during aging; however, the underlying molecular mechanisms remain poorly described. We hypothesized that greater levels of cysteine (Cys) oxidation on muscle proteins are associated with decreased measures of mobility. Herein, we applied a novel redox proteomics approach to measure reversible protein Cys oxidation in vastus lateralis muscle biopsies collected from 56 subjects in the Study of Muscle, Mobility and Aging (SOMMA), a community-based cohort study of individuals aged 70 years and older. We tested whether levels of Cys oxidation on key muscle proteins involved in muscle structure and contraction were associated with muscle function (leg power and strength), walking speed, and fitness (VO2 peak on cardiopulmonary exercise testing) using linear regression models adjusted for age, sex, and body weight. Higher oxidation levels of select nebulin Cys sites were associated with lower VO2 peak, while greater oxidation of myomesin-1, myomesin-2, and nebulin Cys sites was associated with slower walking speed. Higher oxidation of Cys sites in key proteins such as myomesin-2, alpha-actinin-2, and skeletal muscle alpha-actin were associated with lower leg power and strength. We also observed an unexpected correlation (R = 0.48) between a higher oxidation level of eight Cys sites in alpha-actinin-3 and stronger leg power. Despite this observation, the results generally support the hypothesis that Cys oxidation of muscle proteins impairs muscle power and strength, walking speed, and cardiopulmonary fitness with aging.
Collapse
Affiliation(s)
- Nicholas J. Day
- Biological Sciences DivisionPacific Northwest National LaboratoryRichlandWashingtonUSA
| | - Shane S. Kelly
- Biological Sciences DivisionPacific Northwest National LaboratoryRichlandWashingtonUSA
| | - Li‐Yung Lui
- San Francisco Coordinating CenterCalifornia Pacific Medical Center Research InstituteSan FranciscoCaliforniaUSA
| | - Tyler A. Mansfield
- San Francisco Coordinating CenterCalifornia Pacific Medical Center Research InstituteSan FranciscoCaliforniaUSA
| | - Matthew J. Gaffrey
- Biological Sciences DivisionPacific Northwest National LaboratoryRichlandWashingtonUSA
| | - Jesse B. Trejo
- Biological Sciences DivisionPacific Northwest National LaboratoryRichlandWashingtonUSA
| | - Tyler J. Sagendorf
- Biological Sciences DivisionPacific Northwest National LaboratoryRichlandWashingtonUSA
| | - Isaac K. Attah
- Biological Sciences DivisionPacific Northwest National LaboratoryRichlandWashingtonUSA
| | - Ronald J. Moore
- Biological Sciences DivisionPacific Northwest National LaboratoryRichlandWashingtonUSA
| | - Collin M. Douglas
- Department of Physiology and AgingUniversity of Florida College of MedicineGainesvilleFloridaUSA
| | - Anne B. Newman
- Department of EpidemiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Stephen B. Kritchevsky
- Department of Internal Medicine‐Gerontology and Geriatric MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Philip A. Kramer
- Department of Internal Medicine‐Gerontology and Geriatric MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | | | - Paul M. Coen
- Translational Research InstituteAdventHealthOrlandoFloridaUSA
| | | | - Russell T. Hepple
- Department of Physical TherapyUniversity of Florida College of MedicineGainesvilleFloridaUSA
| | - Peggy M. Cawthon
- San Francisco Coordinating CenterCalifornia Pacific Medical Center Research InstituteSan FranciscoCaliforniaUSA
- Department of Epidemiology and BiostatisticsUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Vladislav A. Petyuk
- Biological Sciences DivisionPacific Northwest National LaboratoryRichlandWashingtonUSA
| | - Karyn A. Esser
- Department of Physiology and AgingUniversity of Florida College of MedicineGainesvilleFloridaUSA
| | - Wei‐Jun Qian
- Biological Sciences DivisionPacific Northwest National LaboratoryRichlandWashingtonUSA
| | - Steven R. Cummings
- San Francisco Coordinating CenterCalifornia Pacific Medical Center Research InstituteSan FranciscoCaliforniaUSA
- Department of Epidemiology and BiostatisticsUniversity of CaliforniaSan FranciscoCaliforniaUSA
| |
Collapse
|
4
|
Da W, Chen Q, Shen B. The current insights of mitochondrial hormesis in the occurrence and treatment of bone and cartilage degeneration. Biol Res 2024; 57:37. [PMID: 38824571 PMCID: PMC11143644 DOI: 10.1186/s40659-024-00494-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 04/03/2024] [Indexed: 06/03/2024] Open
Abstract
It is widely acknowledged that aging, mitochondrial dysfunction, and cellular phenotypic abnormalities are intricately associated with the degeneration of bone and cartilage. Consequently, gaining a comprehensive understanding of the regulatory patterns governing mitochondrial function and its underlying mechanisms holds promise for mitigating the progression of osteoarthritis, intervertebral disc degeneration, and osteoporosis. Mitochondrial hormesis, referred to as mitohormesis, represents a cellular adaptive stress response mechanism wherein mitochondria restore homeostasis and augment resistance capabilities against stimuli by generating reactive oxygen species (ROS), orchestrating unfolded protein reactions (UPRmt), inducing mitochondrial-derived peptides (MDP), instigating mitochondrial dynamic changes, and activating mitophagy, all prompted by low doses of stressors. The varying nature, intensity, and duration of stimulus sources elicit divergent degrees of mitochondrial stress responses, subsequently activating one or more signaling pathways to initiate mitohormesis. This review focuses specifically on the effector molecules and regulatory networks associated with mitohormesis, while also scrutinizing extant mechanisms of mitochondrial dysfunction contributing to bone and cartilage degeneration through oxidative stress damage. Additionally, it underscores the potential of mechanical stimulation, intermittent dietary restrictions, hypoxic preconditioning, and low-dose toxic compounds to trigger mitohormesis, thereby alleviating bone and cartilage degeneration.
Collapse
Affiliation(s)
- Wacili Da
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Quan Chen
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Bin Shen
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
5
|
Cao G, Zuo J, Wu B, Wu Y. Polyphenol supplementation boosts aerobic endurance in athletes: systematic review. Front Physiol 2024; 15:1369174. [PMID: 38651044 PMCID: PMC11033476 DOI: 10.3389/fphys.2024.1369174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
In recent years, an increasing trend has been observed in the consumption of specific polyphenols, such as flavonoids and phenolic acids, derived from green tea, berries, and other similar sources. These compounds are believed to alleviate oxidative stress and inflammation resulting from exercise, potentially enhancing athletic performance. This systematic review critically examines the role of polyphenol supplementation in improving aerobic endurance among athletes and individuals with regular exercise habits. The review involved a thorough search of major literature databases, including PubMed, Web of Science, SCOPUS, SPORTDiscus, and Embase, covering re-search up to the year 2023. Out of 491 initially identified articles, 11 met the strict inclusion criteria for this review. These studies specifically focused on the incorporation of polyphenols or polyphenol-containing complexes in their experimental design, assessing their impact on aerobic endurance. The methodology adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, and the risk of bias was evaluated using the Cochrane bias risk assessment tool. While this review suggests that polyphenol supplementation might enhance certain aspects of aerobic endurance and promote fat oxidation, it is important to interpret these findings with caution, considering the limited number of studies available. Systematic Review Registration: https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42023453321.
Collapse
Affiliation(s)
- Gexin Cao
- Department of Exercise Physiology, School of Sports Science, Beijing Sports University, Beijing, China
- Laboratory of Sports Stress and Adaptation of General Administration of Sport, Beijing Sports University, Beijing, China
| | - Jing Zuo
- Laboratory of Sports Stress and Adaptation of General Administration of Sport, Beijing Sports University, Beijing, China
- Department of Anatomy Laboratory, School of Sports Science, Beijing Sports University, Beijing, China
| | - Baile Wu
- Department of Exercise Physiology, School of Sports Science, Beijing Sports University, Beijing, China
- Laboratory of Sports Stress and Adaptation of General Administration of Sport, Beijing Sports University, Beijing, China
| | - Ying Wu
- Department of Exercise Physiology, School of Sports Science, Beijing Sports University, Beijing, China
- Laboratory of Sports Stress and Adaptation of General Administration of Sport, Beijing Sports University, Beijing, China
| |
Collapse
|
6
|
Day NJ, Kelly SS, Lui LY, Mansfield TA, Gaffrey MJ, Trejo JB, Sagendorf TJ, Attah K, Moore RJ, Douglas CM, Newman AB, Kritchevsky SB, Kramer PA, Marcinek DJ, Coen PM, Goodpaster BH, Hepple RT, Cawthon PM, Petyuk VA, Esser KA, Qian WJ, Cummings SR. Signatures of Cysteine Oxidation on Muscle Structural and Contractile Proteins Are Associated with Physical Performance and Muscle Function in Older Adults: Study of Muscle, Mobility and Aging (SOMMA). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.07.23298224. [PMID: 37986748 PMCID: PMC10659491 DOI: 10.1101/2023.11.07.23298224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Oxidative stress is considered a contributor to declining muscle function and mobility during aging; however, the underlying molecular mechanisms remain poorly described. We hypothesized that greater levels of cysteine (Cys) oxidation on muscle proteins are associated with decreased measures of mobility. Herein, we applied a novel redox proteomics approach to measure reversible protein Cys oxidation in vastus lateralis muscle biopsies collected from 56 subjects in the Study of Muscle, Mobility and Aging (SOMMA), a community-based cohort study of individuals aged 70 years and older. We tested whether levels of Cys oxidation on key muscle proteins involved in muscle structure and contraction were associated with muscle function (leg power and strength), walking speed, and fitness (VO2 peak on cardiopulmonary exercise testing) using linear regression models adjusted for age, sex, and body weight. Higher oxidation levels of select nebulin Cys sites were associated with lower VO2 peak, while greater oxidation of myomesin-1, myomesin-2, and nebulin Cys sites was associated with slower walking speed. Higher oxidation of Cys sites in key proteins such as myomesin-2, alpha-actinin-2, and skeletal muscle alpha-actin were associated with lower leg power and strength. We also observed an unexpected correlation (r = 0.48) between a higher oxidation level of 8 Cys sites in alpha-actinin-3 and stronger leg power. Despite this observation, the results generally support the hypothesis that Cys oxidation of muscle proteins impair muscle power and strength, walking speed, and cardiopulmonary fitness with aging.
Collapse
Affiliation(s)
- Nicholas J. Day
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Shane S. Kelly
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Li-Yung Lui
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Tyler A. Mansfield
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Matthew J. Gaffrey
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Jesse B. Trejo
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Tyler J. Sagendorf
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Kwame Attah
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Ronald J. Moore
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Collin M. Douglas
- Department of Physiology, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Anne B. Newman
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Stephen B. Kritchevsky
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Philip A. Kramer
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - David J. Marcinek
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Paul M. Coen
- Translational Research Institute, AdventHealth, Orlando, Florida, USA
| | | | - Russell T. Hepple
- Department of Physical Therapy, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Peggy M. Cawthon
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, USA
| | - Vladislav A. Petyuk
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Karyn A. Esser
- Department of Physiology, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Steven R. Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, USA
| |
Collapse
|
7
|
Franco-Obregón A. Harmonizing Magnetic Mitohormetic Regenerative Strategies: Developmental Implications of a Calcium-Mitochondrial Axis Invoked by Magnetic Field Exposure. Bioengineering (Basel) 2023; 10:1176. [PMID: 37892906 PMCID: PMC10604793 DOI: 10.3390/bioengineering10101176] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/05/2023] [Accepted: 10/08/2023] [Indexed: 10/29/2023] Open
Abstract
Mitohormesis is a process whereby mitochondrial stress responses, mediated by reactive oxygen species (ROS), act cumulatively to either instill survival adaptations (low ROS levels) or to produce cell damage (high ROS levels). The mitohormetic nature of extremely low-frequency electromagnetic field (ELF-EMF) exposure thus makes it susceptible to extraneous influences that also impinge on mitochondrial ROS production and contribute to the collective response. Consequently, magnetic stimulation paradigms are prone to experimental variability depending on diverse circumstances. The failure, or inability, to control for these factors has contributed to the existing discrepancies between published reports and in the interpretations made from the results generated therein. Confounding environmental factors include ambient magnetic fields, temperature, the mechanical environment, and the conventional use of aminoglycoside antibiotics. Biological factors include cell type and seeding density as well as the developmental, inflammatory, or senescence statuses of cells that depend on the prior handling of the experimental sample. Technological aspects include magnetic field directionality, uniformity, amplitude, and duration of exposure. All these factors will exhibit manifestations at the level of ROS production that will culminate as a unified cellular response in conjunction with magnetic exposure. Fortunately, many of these factors are under the control of the experimenter. This review will focus on delineating areas requiring technical and biological harmonization to assist in the designing of therapeutic strategies with more clearly defined and better predicted outcomes and to improve the mechanistic interpretation of the generated data, rather than on precise applications. This review will also explore the underlying mechanistic similarities between magnetic field exposure and other forms of biophysical stimuli, such as mechanical stimuli, that mutually induce elevations in intracellular calcium and ROS as a prerequisite for biological outcome. These forms of biophysical stimuli commonly invoke the activity of transient receptor potential cation channel classes, such as TRPC1.
Collapse
Affiliation(s)
- Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; ; Tel.: +65-6777-8427 or +65-6601-6143
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117544, Singapore
| |
Collapse
|
8
|
Chirumbolo S, Bertossi D, Magistretti P. Insights on the role of L-lactate as a signaling molecule in skin aging. Biogerontology 2023; 24:709-726. [PMID: 36708434 PMCID: PMC9883612 DOI: 10.1007/s10522-023-10018-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/17/2023] [Indexed: 01/29/2023]
Abstract
L-lactate is a catabolite from the anaerobic metabolism of glucose, which plays a paramount role as a signaling molecule in various steps of the cell survival. Its activity, as a master tuner of many mechanisms underlying the aging process, for example in the skin, is still presumptive, however its crucial position in the complex cross-talk between mitochondria and the process of cell survival, should suggest that L-lactate may be not a simple waste product but a fine regulator of the aging/survival machinery, probably via mito-hormesis. Actually, emerging evidence is highlighting that ROS are crucial in the signaling of skin health, including mechanisms underlying wound repair, renewal and aging. The ROS, including superoxide anion, hydrogen peroxide, and nitric oxide, play both beneficial and detrimental roles depending upon their levels and cellular microenvironment. Physiological ROS levels are essential for cutaneous health and the wound repair process. Aberrant redox signaling activity drives chronic skin disease in elderly. On the contrary, impaired redox modulation, due to enhanced ROS generation and/or reduced levels of antioxidant defense, suppresses wound healing via promoting lymphatic/vascular endothelial cell apoptosis and death. This review tries to elucidate this issue.
Collapse
Affiliation(s)
- Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, Unit of Human Anatomy, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy.
| | - Dario Bertossi
- Department of Surgery, Dentistry, Paediatrics and Gynaecology-Unit of Maxillo-Facial Surgery, University of Verona, Verona, Italy
| | - Pierre Magistretti
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
| |
Collapse
|
9
|
Espinosa A, Casas M, Jaimovich E. Energy (and Reactive Oxygen Species Generation) Saving Distribution of Mitochondria for the Activation of ATP Production in Skeletal Muscle. Antioxidants (Basel) 2023; 12:1624. [PMID: 37627619 PMCID: PMC10451830 DOI: 10.3390/antiox12081624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/27/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Exercise produces oxidants from a variety of intracellular sources, including NADPH oxidases (NOX) and mitochondria. Exercise-derived reactive oxygen species (ROS) are beneficial, and the amount and location of these ROS is important to avoid muscle damage associated with oxidative stress. We discuss here some of the evidence that involves ROS production associated with skeletal muscle contraction and the potential oxidative stress associated with muscle contraction. We also discuss the potential role of H2O2 produced after NOX activation in the regulation of glucose transport in skeletal muscle. Finally, we propose a model based on evidence for the role of different populations of mitochondria in skeletal muscle in the regulation of ATP production upon exercise. The subsarcolemmal population of mitochondria has the enzymatic and metabolic components to establish a high mitochondrial membrane potential when fissioned at rest but lacks the capacity to produce ATP. Calcium entry into the mitochondria will further increase the metabolic input. Upon exercise, subsarcolemmal mitochondria will fuse to intermyofibrillar mitochondria and will transfer the mitochondria membrane potential to them. These mitochondria are rich in ATP synthase and will subsequentially produce the ATP needed for muscle contraction in long-term exercise. These events will optimize energy use and minimize mitochondria ROS production.
Collapse
Affiliation(s)
- Alejandra Espinosa
- Center for Studies of Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8320000, Chile; (A.E.)
- San Felipe Campus, School of Medicine, Faculty of Medicine, Universidad de Valparaiso, San Felipe 2172972, Chile
| | - Mariana Casas
- Center for Studies of Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8320000, Chile; (A.E.)
| | - Enrique Jaimovich
- Center for Studies of Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8320000, Chile; (A.E.)
| |
Collapse
|
10
|
Musci RV, Andrie KM, Walsh MA, Valenti ZJ, Linden MA, Afzali MF, Bork S, Campbell M, Johnson T, Kail TE, Martinez R, Nguyen T, Sanford J, Wist S, Murrell MD, McCord JM, Hybertson BM, Zhang Q, Javors MA, Santangelo KS, Hamilton KL. Phytochemical compound PB125 attenuates skeletal muscle mitochondrial dysfunction and impaired proteostasis in a model of musculoskeletal decline. J Physiol 2023; 601:2189-2216. [PMID: 35924591 PMCID: PMC9898472 DOI: 10.1113/jp282273] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 07/28/2022] [Indexed: 02/06/2023] Open
Abstract
Impaired mitochondrial function and disrupted proteostasis contribute to musculoskeletal dysfunction. However, few interventions simultaneously target these two drivers to prevent musculoskeletal decline. Nuclear factor erythroid 2-related factor 2 (Nrf2) activates a transcriptional programme promoting cytoprotection, metabolism, and proteostasis. We hypothesized daily treatment with a purported Nrf2 activator, PB125, in Hartley guinea pigs, a model of musculoskeletal decline, would attenuate the progression of skeletal muscle mitochondrial dysfunction and impaired proteostasis and preserve musculoskeletal function. We treated 2- and 5-month-old male and female Hartley guinea pigs for 3 and 10 months, respectively, with the phytochemical compound PB125. Longitudinal assessments of voluntary mobility were measured using Any-MazeTM open-field enclosure monitoring. Cumulative skeletal muscle protein synthesis rates were measured using deuterium oxide over the final 30 days of treatment. Mitochondrial oxygen consumption in soleus muscles was measured using high resolution respirometry. In both sexes, PB125 (1) increased electron transfer system capacity; (2) attenuated the disease/age-related decline in coupled and uncoupled mitochondrial respiration; and (3) attenuated declines in protein synthesis in the myofibrillar, mitochondrial and cytosolic subfractions of the soleus. These effects were not associated with statistically significant prolonged maintenance of voluntary mobility in guinea pigs. Collectively, treatment with PB125 contributed to maintenance of skeletal muscle mitochondrial respiration and proteostasis in a pre-clinical model of musculoskeletal decline. Further investigation is necessary to determine if these documented effects of PB125 are also accompanied by slowed progression of other aspects of musculoskeletal dysfunction. KEY POINTS: Aside from exercise, there are no effective interventions for musculoskeletal decline, which begins in the fifth decade of life and contributes to disability and cardiometabolic diseases. Targeting both mitochondrial dysfunction and impaired protein homeostasis (proteostasis), which contribute to the age and disease process, may mitigate the progressive decline in overall musculoskeletal function (e.g. gait, strength). A potential intervention to target disease drivers is to stimulate nuclear factor erythroid 2-related factor 2 (Nrf2) activation, which leads to the transcription of genes responsible for redox homeostasis, proteome maintenance and mitochondrial energetics. Here, we tested a purported phytochemical Nrf2 activator, PB125, to improve mitochondrial function and proteostasis in male and female Hartley guinea pigs, which are a model for musculoskeletal ageing. PB125 improved mitochondrial respiration and attenuated disease- and age-related declines in skeletal muscle protein synthesis, a component of proteostasis, in both male and female Hartley guinea pigs.
Collapse
Affiliation(s)
- Robert V. Musci
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Kendra M. Andrie
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Maureen A. Walsh
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Zackary J. Valenti
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Melissa A. Linden
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Maryam F. Afzali
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Sydney Bork
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Margaret Campbell
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Taylor Johnson
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Thomas E. Kail
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Richard Martinez
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Tessa Nguyen
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Joseph Sanford
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Sara Wist
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | | | - Joe M. McCord
- Pathways Bioscience, Aurora, CO
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Brooks M. Hybertson
- Pathways Bioscience, Aurora, CO
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Qian Zhang
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | | | - Kelly S. Santangelo
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Karyn L. Hamilton
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
- Columbine Health Systems Center for Healthy Aging, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
11
|
Costa-Machado LF, Garcia-Dominguez E, McIntyre RL, Lopez-Aceituno JL, Ballesteros-Gonzalez Á, Tapia-Gonzalez A, Fabregat-Safont D, Eisenberg T, Gomez J, Plaza A, Sierra-Ramirez A, Perez M, Villanueva-Bermejo D, Fornari T, Loza MI, Herradon G, Hofer SJ, Magnes C, Madeo F, Duerr JS, Pozo OJ, Galindo MI, Del Pino I, Houtkooper RH, Megias D, Viña J, Gomez-Cabrera MC, Fernandez-Marcos PJ. Peripheral modulation of antidepressant targets MAO-B and GABAAR by harmol induces mitohormesis and delays aging in preclinical models. Nat Commun 2023; 14:2779. [PMID: 37188705 PMCID: PMC10185515 DOI: 10.1038/s41467-023-38410-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 05/02/2023] [Indexed: 05/17/2023] Open
Abstract
Reversible and sub-lethal stresses to the mitochondria elicit a program of compensatory responses that ultimately improve mitochondrial function, a conserved anti-aging mechanism termed mitohormesis. Here, we show that harmol, a member of the beta-carbolines family with anti-depressant properties, improves mitochondrial function and metabolic parameters, and extends healthspan. Treatment with harmol induces a transient mitochondrial depolarization, a strong mitophagy response, and the AMPK compensatory pathway both in cultured C2C12 myotubes and in male mouse liver, brown adipose tissue and muscle, even though harmol crosses poorly the blood-brain barrier. Mechanistically, simultaneous modulation of the targets of harmol monoamine-oxidase B and GABA-A receptor reproduces harmol-induced mitochondrial improvements. Diet-induced pre-diabetic male mice improve their glucose tolerance, liver steatosis and insulin sensitivity after treatment with harmol. Harmol or a combination of monoamine oxidase B and GABA-A receptor modulators extend the lifespan of hermaphrodite Caenorhabditis elegans or female Drosophila melanogaster. Finally, two-year-old male and female mice treated with harmol exhibit delayed frailty onset with improved glycemia, exercise performance and strength. Our results reveal that peripheral targeting of monoamine oxidase B and GABA-A receptor, common antidepressant targets, extends healthspan through mitohormesis.
Collapse
Affiliation(s)
- Luis Filipe Costa-Machado
- Metabolic Syndrome Group - BIOPROMET. Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM + CSIC, E28049, Madrid, Spain
- Kaertor Foundation, EMPRENDIA Building, Floor 2, Office 4, Campus Vida, E-15706, Santiago de Compostela, Spain, E-15706, Santiago de Compostela, Spain
- BioFarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Esther Garcia-Dominguez
- Freshage Research Group, Department of Physiology, Faculty of Medicine, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, University of Valencia, Valencia, Spain
| | - Rebecca L McIntyre
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Jose Luis Lopez-Aceituno
- Metabolic Syndrome Group - BIOPROMET. Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM + CSIC, E28049, Madrid, Spain
| | - Álvaro Ballesteros-Gonzalez
- Developmental Biology and Disease Models Group, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
| | - Andrea Tapia-Gonzalez
- Neural Plasticity Group, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
| | - David Fabregat-Safont
- Applied Metabolomics Research Group, Hospital del Mar Medical Research Institute - (IMIM), Barcelona, Spain
- Environmental and Public Health Analytical Chemistry, Research Institute for Pesticides and Water, University Jaume I, 12006, Castelló de la Plana, Castellón, Spain
| | - Tobias Eisenberg
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Humboldtstraße 50, 8010, Graz, Austria
- BioTechMed Graz, 8010, Graz, Austria
- Field of Excellence BioHealth - University of Graz, Graz, Austria
| | - Jesús Gomez
- Confocal Microscopy Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Adrian Plaza
- Metabolic Syndrome Group - BIOPROMET. Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM + CSIC, E28049, Madrid, Spain
| | - Aranzazu Sierra-Ramirez
- Metabolic Syndrome Group - BIOPROMET. Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM + CSIC, E28049, Madrid, Spain
| | - Manuel Perez
- Confocal Microscopy Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - David Villanueva-Bermejo
- Department of Production and Characterization of Novel Foods, Institute of Food Science Research (CIAL UAM-CSIC), C/ Nicolás Cabrera, 9, P.O. Box. 28049, Madrid, Spain
| | - Tiziana Fornari
- Department of Production and Characterization of Novel Foods, Institute of Food Science Research (CIAL UAM-CSIC), C/ Nicolás Cabrera, 9, P.O. Box. 28049, Madrid, Spain
| | - María Isabel Loza
- Kaertor Foundation, EMPRENDIA Building, Floor 2, Office 4, Campus Vida, E-15706, Santiago de Compostela, Spain, E-15706, Santiago de Compostela, Spain
- BioFarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Gonzalo Herradon
- Lab. Pharmacology, Faculty of Pharmacy, Universidad CEU San Pablo, Urb. Montepríncipe, 28668, Boadilla del Monte, Madrid, Spain
| | - Sebastian J Hofer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Humboldtstraße 50, 8010, Graz, Austria
- BioTechMed Graz, 8010, Graz, Austria
- Field of Excellence BioHealth - University of Graz, Graz, Austria
| | - Christoph Magnes
- HEALTH-Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft mbH, 8010, Graz, Austria
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Humboldtstraße 50, 8010, Graz, Austria
- BioTechMed Graz, 8010, Graz, Austria
- Field of Excellence BioHealth - University of Graz, Graz, Austria
| | - Janet S Duerr
- Department of Biological Sciences, Ohio University, Athens, OH, 45701, USA
| | - Oscar J Pozo
- Applied Metabolomics Research Group, Hospital del Mar Medical Research Institute - (IMIM), Barcelona, Spain
| | - Maximo-Ibo Galindo
- Developmental Biology and Disease Models Group, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, 46022, Valencia, Spain
- UPV-CIPF Joint Research Unit "Disease Mechanisms and Nanomedicine". Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
| | - Isabel Del Pino
- Neural Plasticity Group, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, Campus de Sant Joan, 03550, Alicante, Spain
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Diego Megias
- Confocal Microscopy Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Jose Viña
- Freshage Research Group, Department of Physiology, Faculty of Medicine, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, University of Valencia, Valencia, Spain
| | - Mari Carmen Gomez-Cabrera
- Freshage Research Group, Department of Physiology, Faculty of Medicine, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, University of Valencia, Valencia, Spain
| | - Pablo J Fernandez-Marcos
- Metabolic Syndrome Group - BIOPROMET. Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM + CSIC, E28049, Madrid, Spain.
| |
Collapse
|
12
|
Schiavi A, Salveridou E, Brinkmann V, Shaik A, Menzel R, Kalyanasundaram S, Nygård S, Nilsen H, Ventura N. Mitochondria hormesis delays aging and associated diseases in Caenorhabditis elegans impacting on key ferroptosis players. iScience 2023; 26:106448. [PMID: 37020951 PMCID: PMC10067770 DOI: 10.1016/j.isci.2023.106448] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 11/28/2022] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
Excessive iron accumulation or deficiency leads to a variety of pathologies in humans and developmental arrest in the nematode Caenorhabditis elegans. Instead, sub-lethal iron depletion extends C. elegans lifespan. Hypoxia preconditioning protects against severe hypoxia-induced neuromuscular damage across species but it has low feasible application. In this study, we assessed the potential beneficial effects of genetic and chemical interventions acting via mild iron instead of oxygen depletion. We show that limiting iron availability in C. elegans through frataxin silencing or the iron chelator bipyridine, similar to hypoxia preconditioning, protects against hypoxia-, age-, and proteotoxicity-induced neuromuscular deficits. Mechanistically, our data suggest that the beneficial effects elicited by frataxin silencing are in part mediated by counteracting ferroptosis, a form of non-apoptotic cell death mediated by iron-induced lipid peroxidation. This is achieved by impacting on different key ferroptosis players and likely via gpx-independent redox systems. We thus point to ferroptosis inhibition as a novel potential strategy to promote healthy aging.
Collapse
Affiliation(s)
- Alfonso Schiavi
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | - Eva Salveridou
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | - Vanessa Brinkmann
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | - Anjumara Shaik
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | | | - Sumana Kalyanasundaram
- Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Ståle Nygård
- Bioinformatics Core Facility, Institute for Medical Informatics, Oslo University Hospital, Oslo, Norway
| | - Hilde Nilsen
- Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Natascia Ventura
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
13
|
Vargas-Mendoza N, Morales-González Á, Madrigal-Santillán EO, Angeles-Valencia M, Anguiano-Robledo L, González-López LL, Sosa-Gómez A, Fregoso-Aguilar T, Esquivel-Chirino C, Ruiz-Velazco-Benítez YA, Morales-González JA. Phytochemicals and modulation of exercise-induced oxidative stress: a novel overview of antioxidants. Am J Transl Res 2022; 14:8292-8314. [PMID: 36505319 PMCID: PMC9730074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/01/2022] [Indexed: 12/15/2022]
Abstract
The practice of physical exercise induces a series of physiological changes in the body at different levels, either acutely or chronically. During exercise, the increase in oxygen consumption promotes the production of reactive oxygen species (ROS) and reactive nitrogen species (RNS), which are necessary to maintain homeostasis. ROS/RNS activate cellular signaling pathways, such as the antioxidant cytoprotective systems, inflammation, and cell proliferation, which are crucial for cell survival. However, in exhaustive-extended physical exercise, workloads can exceed the endogenous antioxidant defenses, which may be related to impairment of muscle contraction, fatigue, and a decrease in athletic performance. This review addresses the role of some antioxidants from plant-derived extracts called phytochemicals that can mediate the response to oxidative stress induced by physical exercise by activating signaling pathways, such as Nrf2/Keap1/ARE, responsible for the endogenous antioxidant response and possibly having an impact on sports performance.
Collapse
Affiliation(s)
- Nancy Vargas-Mendoza
- Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico NacionalCiudad de Mexico, Mexico
| | | | | | - Marcelo Angeles-Valencia
- Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico NacionalCiudad de Mexico, Mexico
| | - Liliana Anguiano-Robledo
- Escuela Superior de Medicina, Laboratorio de Farmacología Molecular, Instituto Politécnico NacionalCiudad de Mexico, Mexico
| | - Laura Ligia González-López
- Centro Interdisciplinario de Ciencias de la Salud Unidad Santo Tomas, Instituto Politécnico NacionalCiudad de Mexico, Mexico
| | - Alejandra Sosa-Gómez
- Centro Interdisciplinario de Ciencias de la Salud Unidad Santo Tomas, Instituto Politécnico NacionalCiudad de Mexico, Mexico
| | - Tomás Fregoso-Aguilar
- Laboratorio de Hormonas y Conducta, Departamento de Fisiología, ENCB Campus Zacatenco, Instituto Politécnico NacionalCiudad de Mexico, Mexico
| | - Cesar Esquivel-Chirino
- Área de Básicas Médicas, División de Estudios Profesionales, Facultad de Odontología, Universidad Nacional Autónoma de MéxicoCiudad de Mexico, Mexico
| | | | - José A Morales-González
- Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico NacionalCiudad de Mexico, Mexico
| |
Collapse
|
14
|
Kanova M, Kohout P. Molecular Mechanisms Underlying Intensive Care Unit-Acquired Weakness and Sarcopenia. Int J Mol Sci 2022; 23:8396. [PMID: 35955530 PMCID: PMC9368893 DOI: 10.3390/ijms23158396] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/19/2022] [Accepted: 07/26/2022] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle is a highly adaptable organ, and its amount declines under catabolic conditions such as critical illness. Aging is accompanied by a gradual loss of muscle, especially when physical activity decreases. Intensive care unit-acquired weakness is a common and highly serious neuromuscular complication in critically ill patients. It is a consequence of critical illness and is characterized by a systemic inflammatory response, leading to metabolic stress, that causes the development of multiple organ dysfunction. Muscle dysfunction is an important component of this syndrome, and the degree of catabolism corresponds to the severity of the condition. The population of critically ill is aging; thus, we face another negative effect-sarcopenia-the age-related decline of skeletal muscle mass and function. Low-grade inflammation gradually accumulates over time, inhibits proteosynthesis, worsens anabolic resistance, and increases insulin resistance. The cumulative consequence is a gradual decline in muscle recovery and muscle mass. The clinical manifestation for both of the above conditions is skeletal muscle weakness, with macromolecular damage, and a common mechanism-mitochondrial dysfunction. In this review, we compare the molecular mechanisms underlying the two types of muscle atrophy, and address questions regarding possible shared molecular mechanisms, and whether critical illness accelerates the aging process.
Collapse
Affiliation(s)
- Marcela Kanova
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
- Institute of Physiology and Pathophysiology, Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic
| | - Pavel Kohout
- Department of Internal Medicine, 3rd Faculty of Medicine, Charles University Prague and Teaching Thomayer Hospital, 140 59 Prague, Czech Republic;
| |
Collapse
|
15
|
Touron J, Perrault H, Maisonnave L, Patrac V, Walrand S, Malpuech-Brugère C, Pereira B, Burelle Y, Costes F, Richard R. Effects of exercise-induced metabolic and mechanical loading on skeletal muscle mitochondrial function in male rats. J Appl Physiol (1985) 2022; 133:611-621. [PMID: 35900326 DOI: 10.1152/japplphysiol.00719.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Over the last decades, a growing interest in eccentric (ECC) exercise has emerged, but mitochondrial adaptations to ECC training remain poorly documented. Using an approach for manipulating mechanical and metabolic exercise power, we positioned that for same metabolic power, training using concentric (CON) or ECC contractions would induce similar skeletal muscle mitochondrial adaptations. Sixty adult rats were randomly assigned to a control (CTRL) or three treadmill training groups running at 15m·min-1 for 45min, 5days weekly for 8 weeks at targeted upward or downward slopes. Animals from the CON (+15%) and ECC30 (-30%) groups trained at iso-metabolic power while CON and ECC15 (-15%) exercised at iso-mechanical power. Assessments were made of Vastus Intermedius mitochondrial respiration (oxygraphy), enzymatic activities (spectrophotometry) and real-time qPCR for mRNA transcripts. Maximal rates of mitochondrial respiration was 14-15% higher in CON and ECC30 compared to CTRL and ECC15. Apparent Km for ADP for trained groups was 40-66% higher than CTRL, with statistical significance reached for CON and ECC30. Complex I and citrate synthase activities were 1.6 (ECC15) to 1.8 (ECC30 and CON) times values of CTRL. Complex IV activity was higher than CTRL (p<0.05) only for CON and ECC30. mRNA transcripts analyses showed higher TFAM, SLC25A4, CKMT2 and PPID in the ECC30 compared to CTRL. Findings confirm that training-induced skeletal muscle mitochondrial function adaptations are governed by the extent of metabolic overload irrespective of exercise modality. The distinctive ECC30 mRNA transcript pattern may reflect a cytoskeleton damage-repair or ECC adaptive cycle that differs from that of biogenesis.
Collapse
Affiliation(s)
- Julianne Touron
- UCA- INRAE UMR 1019, Human Nutrition Unit, ASMS team, Clermont-Ferrand, France
| | - Hélène Perrault
- Respiratory Division, McGill University Health Center, Montreal, Canada
| | - Laura Maisonnave
- UCA- INRAE UMR 1019, Human Nutrition Unit, ASMS team, Clermont-Ferrand, France
| | - Véronique Patrac
- UCA- INRAE UMR 1019, Human Nutrition Unit, ASMS team, Clermont-Ferrand, France
| | - Stephane Walrand
- UCA- INRAE UMR 1019, Human Nutrition Unit, ASMS team, Clermont-Ferrand, France
| | | | - Bruno Pereira
- Delegation to Clinical Research and Innovation, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France
| | - Yan Burelle
- Faculty of Health Sciences, University of Ottawa, Ottawa, Canada
| | - Frédéric Costes
- UCA- INRAE UMR 1019, Human Nutrition Unit, ASMS team, Clermont-Ferrand, France.,Department of Sports Medicine and Functional Explorations, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France
| | - Ruddy Richard
- UCA- INRAE UMR 1019, Human Nutrition Unit, ASMS team, Clermont-Ferrand, France.,Delegation to Clinical Research and Innovation, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France.,Department of Sports Medicine and Functional Explorations, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France
| |
Collapse
|
16
|
Eissenberg JC. Working Out: The Molecular Biology of Exercise. MISSOURI MEDICINE 2022; 119:379-384. [PMID: 36118818 PMCID: PMC9462916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The many health benefits of exercise are well-known. Conversely, the pathologies associated with a sedentary lifestyle are also well-documented. However, science and medicine have only recently begun to explain how exercise does what it does. Here, I discuss recent insight into the biochemical mechanisms underlying the benefits of exercise and the pathologies of inactivity.
Collapse
Affiliation(s)
- Joel C Eissenberg
- Professor of Biochemistry and Molecular Biology at Saint Louis School of Medicine, St. Louis, Missouri
| |
Collapse
|
17
|
Yoon TK, Lee CH, Kwon O, Kim MS. Exercise, Mitohormesis, and Mitochondrial ORF of the 12S rRNA Type-C (MOTS-c). Diabetes Metab J 2022; 46:402-413. [PMID: 35656563 PMCID: PMC9171157 DOI: 10.4093/dmj.2022.0092] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/27/2022] [Indexed: 12/03/2022] Open
Abstract
Low levels of mitochondrial stress are beneficial for organismal health and survival through a process known as mitohormesis. Mitohormetic responses occur during or after exercise and may mediate some salutary effects of exercise on metabolism. Exercise-related mitohormesis involves reactive oxygen species production, mitochondrial unfolded protein response (UPRmt), and release of mitochondria-derived peptides (MDPs). MDPs are a group of small peptides encoded by mitochondrial DNA with beneficial metabolic effects. Among MDPs, mitochondrial ORF of the 12S rRNA type-c (MOTS-c) is the most associated with exercise. MOTS-c expression levels increase in skeletal muscles, systemic circulation, and the hypothalamus upon exercise. Systemic MOTS-c administration increases exercise performance by boosting skeletal muscle stress responses and by enhancing metabolic adaptation to exercise. Exogenous MOTS-c also stimulates thermogenesis in subcutaneous white adipose tissues, thereby enhancing energy expenditure and contributing to the anti-obesity effects of exercise training. This review briefly summarizes the mitohormetic mechanisms of exercise with an emphasis on MOTS-c.
Collapse
Affiliation(s)
- Tae Kwan Yoon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, H+ Yangji Hospital, Seoul, Korea
| | - Chan Hee Lee
- Department of of Biomedical Science & Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Korea
| | - Obin Kwon
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Min-Seon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Burtscher J, Romani M, Bernardo G, Popa T, Ziviani E, Hummel FC, Sorrentino V, Millet GP. Boosting mitochondrial health to counteract neurodegeneration. Prog Neurobiol 2022; 215:102289. [DOI: 10.1016/j.pneurobio.2022.102289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/23/2022] [Accepted: 05/25/2022] [Indexed: 12/22/2022]
|
19
|
Exercise and Oxidative Stress Biomarkers among Adult with Cancer: A Systematic Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2097318. [PMID: 35222792 PMCID: PMC8881118 DOI: 10.1155/2022/2097318] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 10/07/2021] [Accepted: 01/20/2022] [Indexed: 01/07/2023]
Abstract
Evidence shows that exercise can have a favourable effect in cancer patients. The exercise’s clinical benefits are likely to concern multiple interrelated biological pathways, among which oxidative stress plays a key role. Regular training can induce an adaptive response that strengthens the antioxidative status of the body. To formulate public health recommendations regarding the optimal exercise prescription for cancer patients, a detailed understanding is needed regarding the effect of exercise on variables linked to oxidative stress and antioxidant status of patients. The goal of this systematic review, based on PRISMA, was to explore and critically analyse the evidence regarding the efficacy of exercise on oxidative stress biomarkers among people with cancer. Study search was conducted in the following databases: PubMed, Cochrane, CINAHL, Embase, PEDro, and SPORTDiscus. The studies’ quality was assessed with the Cochrane risk-of-bias tool and STROBE scale. After identification and screening steps, 10 articles were included. The findings provide an encouraging picture of exercise, including resistance training and aerobic activities, in people with cancer. The exercise improved the indicators of the total antioxidant capacity, increased the antioxidant enzymes’ activity, or reduced the biomarkers of oxidative damage in various forms of cancer such as breast, lung, head, and neck. Regarding oxidative DNA damage, the role of exercise intervention has been difficult to assess. The heterogeneity of study design and the plethora of biomarkers measured hampered the comparison of the articles. This limited the possibility of establishing a comprehensive conclusion on the sensitivity of biomarkers to estimate the exercise’s benefits. Further high-quality studies are required to provide data regarding oxidative stress biomarkers responding to exercise. This information will be useful to assess the efficacy of exercise in people with cancer and support the appropriate prescription of exercise in anticancer strategy.
Collapse
|
20
|
De Jong NP, Rudolph MC, Jackman MR, Sharp RR, Jones K, Houck J, Pan Z, Reusch JEB, MacLean PS, Bessesen DH, Bergouignan A. Short-Term Adaptations in Skeletal Muscle Mitochondrial Oxidative Capacity and Metabolic Pathways to Breaking up Sedentary Behaviors in Overweight or Obese Adults. Nutrients 2022; 14:nu14030454. [PMID: 35276813 PMCID: PMC8838620 DOI: 10.3390/nu14030454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 12/04/2022] Open
Abstract
Breaking up sedentary behavior with short-frequent bouts of physical activity (PA) differentially influences metabolic health compared with the performance of a single-continuous bout of PA matched for total active time. However, the underlying mechanisms are unknown. We compared skeletal muscle mitochondrial respiration (high-resolution respirometry) and molecular adaptations (RNA sequencing) following 4-day exposure to breaks vs. energy-matched single-continuous PA bout in inactive adults with overweight/obesity. Participants (9M/10F, 32.2 ± 6.4 years, 30.3 ± 3.0 kg/m2) completed three 4-day interventions of a randomized cross-over study: SED, sedentary control; MICRO, 5 min brisk walking each hour for 9 h; ONE: 45 min/d continuous brisk walking bout. Fasted muscle biopsies were collected on day 5. Mitochondrial coupling in the presence of lipid-associated substrates was higher after ONE (4.8 ± 2.5) compared to MICRO (3.1 ± 1.1, p = 0.02) and SED (2.3 ± 1.0, p = 0.001). Respiratory rates did not differ across groups with carbohydrate-associated substrates. In pathways associated with muscle contraction transcription signaling, ONE and MICRO similarly enhanced Oxidative Phosphorylation and Sirtuin Signaling expression (p < 0.0001, for both). However, ONE (p < 0.001, for all), but not MICRO, had greater pathway enrichment, including Ca++, mTOR, AMPK, and HIF1α signaling, than SED. Although breaking up sedentary behavior triggered skeletal muscle molecular adaptations favoring oxidative capacity, it did not improve mitochondrial function over the short term.
Collapse
Affiliation(s)
- Nathan P. De Jong
- Division of Endocrinology, Metabolism, Diabetes and Anschutz Health and Wellness Center, University of Colorado School of Medicine, Aurora, CO 80045, USA; (N.P.D.J.); (D.H.B.)
| | - Michael C. Rudolph
- Department of Physiology, Harold Hamm Diabetes Center, Oklahoma University Health Sciences Center, Oklahoma City, OK 73104, USA; (M.C.R.); (R.R.S.)
| | - Matthew R. Jackman
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA; (M.R.J.); (J.H.); (P.S.M.)
| | - Rachel R. Sharp
- Department of Physiology, Harold Hamm Diabetes Center, Oklahoma University Health Sciences Center, Oklahoma City, OK 73104, USA; (M.C.R.); (R.R.S.)
- Laboratory of Molecular Biology and Cytometric Research, Oklahoma University Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Ken Jones
- Laboratory of Molecular Biology and Cytometric Research, Oklahoma University Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Julie Houck
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA; (M.R.J.); (J.H.); (P.S.M.)
| | - Zhaoxing Pan
- Department of Biostatistics and Informatics, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA;
| | - Jane E. B. Reusch
- Center for Women’s Health Research, University of Colorado School of Medicine, Aurora, CO 80045, USA;
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO 80045, USA
| | - Paul S. MacLean
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA; (M.R.J.); (J.H.); (P.S.M.)
| | - Daniel H. Bessesen
- Division of Endocrinology, Metabolism, Diabetes and Anschutz Health and Wellness Center, University of Colorado School of Medicine, Aurora, CO 80045, USA; (N.P.D.J.); (D.H.B.)
- Denver Health Medical Center, Denver, CO 80204, USA
| | - Audrey Bergouignan
- Division of Endocrinology, Metabolism, Diabetes and Anschutz Health and Wellness Center, University of Colorado School of Medicine, Aurora, CO 80045, USA; (N.P.D.J.); (D.H.B.)
- Institut Pluridisciplinaire Hubert Curien, Université de Strasbourg, Centre National de la Recherche Scientifique (CNRS), UMR7178, 67037 Strasbourg, France
- Correspondence: or
| |
Collapse
|
21
|
Yamamoto A, Sly PD, Henningham A, Begum N, Yeo AJ, Fantino E. Redox Homeostasis in Well-differentiated Primary Human Nasal Epithelial Cells. JOURNAL OF CELLULAR SIGNALING 2022; 3:193-206. [PMID: 36777036 PMCID: PMC9912202 DOI: 10.33696/signaling.3.083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Oxidative stress (OS) in the airway epithelium is associated with inflammation, cell damage, and mitochondrial dysfunction that may initiate or worsen respiratory disease. Redox regulation maintains the equilibrium of pro-oxidant/antioxidant reactions but can be disturbed by environmental exposures. The mechanism(s) underlying the induction and impact of OS on airway epithelium and how these influences on respiratory disease is poorly understood. The aim of this study was to develop a stress response model in primary human nasal epithelial cells (NECs) grown at the air-liquid interface (ALI) into a well-differentiated epithelium and to use this model to investigate the mechanisms underlying OS. Hydrogen peroxide (H2O2) was used to induce acute OS and the responses were measured with trans epithelial electrical resistance (TEER), membrane permeability, cell death (LDH release), mitochondrial reactive oxygen species (mtROS) generation, redox status (GSH/GSSG ratio), cellular ATP, and signaling pathways (SIRT1, FOXO3, p53, p21, PINK1, PARKIN, NRF2). Following 25 mM (sensitive) or 50mM (resistant) H2O2 exposure, cell integrity decreased (p<0.05), GSH/GSSG ratio reduced (p<0.05), and ATP production declined by 83% (p<0.05) in the sensitive and 55% (p<0.05) in the resistant group; mtROS production increased 3.4-fold (p<0.001). Significant inter-individual differences between healthy humans with regards to susceptibility to OS, and differential activation of various pathways (FOXO3, PARKIN) were observed. These intra-individual differences in susceptibility to OS may be attributed to resistant individuals having more mitochondria or greater mitochondrial function.
Collapse
Affiliation(s)
- Ayaho Yamamoto
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland 4101, Australia,Correspondence should be addressed to Ayaho Yamamoto,
| | - Peter D. Sly
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland 4101, Australia
| | - Anna Henningham
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland 4101, Australia
| | - Nelufa Begum
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland 4101, Australia
| | - Abrey J. Yeo
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland 4101, Australia,The University of Queensland Centre for Clinical Research, Herston, Queensland 4029, Australia
| | - Emmanuelle Fantino
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland 4101, Australia
| |
Collapse
|
22
|
Skaperda Z, Tekos F, Vardakas P, Nepka C, Kouretas D. Reconceptualization of Hormetic Responses in the Frame of Redox Toxicology. Int J Mol Sci 2021; 23:ijms23010049. [PMID: 35008472 PMCID: PMC8744777 DOI: 10.3390/ijms23010049] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/13/2021] [Accepted: 12/17/2021] [Indexed: 02/01/2023] Open
Abstract
Cellular adaptive mechanisms emerging after exposure to low levels of toxic agents or stressful stimuli comprise an important biological feature that has gained considerable scientific interest. Investigations of low-dose exposures to diverse chemical compounds signify the non-linear mode of action in the exposed cell or organism at such dose levels in contrast to the classic detrimental effects induced at higher ones, a phenomenon usually referred to as hormesis. The resulting phenotype is a beneficial effect that tests our physiology within the limits of our homeostatic adaptations. Therefore, doses below the region of adverse responses are of particular interest and are specified as the hormetic gain zone. The manifestation of redox adaptations aiming to prevent from disturbances of redox homeostasis represent an area of particular interest in hormetic responses, observed after exposure not only to stressors but also to compounds of natural origin, such as phytochemicals. Findings from previous studies on several agents demonstrate the heterogeneity of the specific zone in terms of the molecular events occurring. Major factors deeply involved in these biphasic phenomena are the bioactive compound per se, the dose level, the duration of exposure, the cell, tissue or even organ exposed to and, of course, the biomarker examined. In the end, the molecular fate is a complex toxicological event, based on beneficial and detrimental effects, which, however, are poorly understood to date.
Collapse
Affiliation(s)
- Zoi Skaperda
- Laboratory of Animal Physiology, Department of Biochemistry-Biotechnology, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece; (Z.S.); (F.T.); (P.V.)
| | - Fotios Tekos
- Laboratory of Animal Physiology, Department of Biochemistry-Biotechnology, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece; (Z.S.); (F.T.); (P.V.)
| | - Periklis Vardakas
- Laboratory of Animal Physiology, Department of Biochemistry-Biotechnology, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece; (Z.S.); (F.T.); (P.V.)
| | - Charitini Nepka
- Department of Pathology, University Hospital of Larissa, 41334 Larissa, Greece;
| | - Demetrios Kouretas
- Laboratory of Animal Physiology, Department of Biochemistry-Biotechnology, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece; (Z.S.); (F.T.); (P.V.)
- Correspondence: ; Tel.: +30-2410-565-277; Fax: +30-2410-565-293
| |
Collapse
|
23
|
Vargas-Mendoza N, Angeles-Valencia M, Morales-González Á, Madrigal-Santillán EO, Morales-Martínez M, Madrigal-Bujaidar E, Álvarez-González I, Gutiérrez-Salinas J, Esquivel-Chirino C, Chamorro-Cevallos G, Cristóbal-Luna JM, Morales-González JA. Oxidative Stress, Mitochondrial Function and Adaptation to Exercise: New Perspectives in Nutrition. Life (Basel) 2021; 11:life11111269. [PMID: 34833151 PMCID: PMC8624755 DOI: 10.3390/life11111269] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/10/2021] [Accepted: 11/13/2021] [Indexed: 02/07/2023] Open
Abstract
Cells have the ability to adapt to stressful environments as a part of their evolution. Physical exercise induces an increase of a demand for energy that must be met by mitochondria as the main (ATP) provider. However, this process leads to the increase of free radicals and the so-called reactive oxygen species (ROS), which are necessary for the maintenance of cell signaling and homeostasis. In addition, mitochondrial biogenesis is influenced by exercise in continuous crosstalk between the mitochondria and the nuclear genome. Excessive workloads may induce severe mitochondrial stress, resulting in oxidative damage. In this regard, the objective of this work was to provide a general overview of the molecular mechanisms involved in mitochondrial adaptation during exercise and to understand if some nutrients such as antioxidants may be implicated in blunt adaptation and/or an impact on the performance of exercise by different means.
Collapse
Affiliation(s)
- Nancy Vargas-Mendoza
- Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Col. Casco de Santo Tomás, Del. Miguel Hidalgo, Ciudad de México 11340, Mexico; (N.V.-M.); (M.A.-V.); (E.O.M.-S.)
| | - Marcelo Angeles-Valencia
- Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Col. Casco de Santo Tomás, Del. Miguel Hidalgo, Ciudad de México 11340, Mexico; (N.V.-M.); (M.A.-V.); (E.O.M.-S.)
| | - Ángel Morales-González
- Escuela Superior de Cómputo, Instituto Politécnico Nacional, Av. Juan de Dios Bátiz s/n Esquina Miguel Othón de Mendizabal, Unidad Profesional Adolfo López Mateos, Ciudad de México 07738, Mexico
- Correspondence: (Á.M.-G.); (J.A.M.-G.); Tel.: +52-55-5729-6300 (Á.M.-G. & J.A.M.-G.)
| | - Eduardo Osiris Madrigal-Santillán
- Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Col. Casco de Santo Tomás, Del. Miguel Hidalgo, Ciudad de México 11340, Mexico; (N.V.-M.); (M.A.-V.); (E.O.M.-S.)
| | - Mauricio Morales-Martínez
- Licenciatura en Nutrición, Universidad Intercontinental, Insurgentes Sur 4303, Santa Úrsula Xitla, Alcaldía Tlalpan, Ciudad de México 14420, Mexico;
| | - Eduardo Madrigal-Bujaidar
- Laboratorio de Genética, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional A. López Mateos, Av. Wilfrido Massieu, Col., Lindavista, Ciudad de México 07738, Mexico; (E.M.-B.); (I.Á.-G.)
| | - Isela Álvarez-González
- Laboratorio de Genética, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional A. López Mateos, Av. Wilfrido Massieu, Col., Lindavista, Ciudad de México 07738, Mexico; (E.M.-B.); (I.Á.-G.)
| | - José Gutiérrez-Salinas
- Laboratorio de Bioquímica y Medicina Experimental, Centro Médico Nacional “20 de Noviembre”, ISSSTE, Ciudad de México 03229, Mexico;
| | - César Esquivel-Chirino
- Área de Básicas Médicas, División de Estudios Profesionales, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Germán Chamorro-Cevallos
- Laboratorio de Toxicología Preclínica, Departamento de Farmacia, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Av. Wilfrido Massieu 399, Col. Nueva Industrial Vallejo, Del. Gustavo A. Madero, Ciudad de México 07738, Mexico; (G.C.-C.); (J.M.C.-L.)
| | - José Melesio Cristóbal-Luna
- Laboratorio de Toxicología Preclínica, Departamento de Farmacia, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Av. Wilfrido Massieu 399, Col. Nueva Industrial Vallejo, Del. Gustavo A. Madero, Ciudad de México 07738, Mexico; (G.C.-C.); (J.M.C.-L.)
| | - José A. Morales-González
- Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Col. Casco de Santo Tomás, Del. Miguel Hidalgo, Ciudad de México 11340, Mexico; (N.V.-M.); (M.A.-V.); (E.O.M.-S.)
- Correspondence: (Á.M.-G.); (J.A.M.-G.); Tel.: +52-55-5729-6300 (Á.M.-G. & J.A.M.-G.)
| |
Collapse
|
24
|
Harper C, Gopalan V, Goh J. Exercise rescues mitochondrial coupling in aged skeletal muscle: a comparison of different modalities in preventing sarcopenia. J Transl Med 2021; 19:71. [PMID: 33593349 PMCID: PMC7885447 DOI: 10.1186/s12967-021-02737-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/04/2021] [Indexed: 01/07/2023] Open
Abstract
Skeletal muscle aging is associated with a decline in motor function and loss of muscle mass- a condition known as sarcopenia. The underlying mechanisms that drive this pathology are associated with a failure in energy generation in skeletal muscle, either from age-related decline in mitochondrial function, or from disuse. To an extent, lifelong exercise is efficacious in preserving the energetic properties of skeletal muscle and thus may delay the onset of sarcopenia. This review discusses the cellular and molecular changes in skeletal muscle mitochondria during the aging process and how different exercise modalities work to reverse these changes. A key factor that will be described is the efficiency of mitochondrial coupling—ATP production relative to O2 uptake in myocytes and how that efficiency is a main driver for age-associated decline in skeletal muscle function. With that, we postulate the most effective exercise modality and protocol for reversing the molecular hallmarks of skeletal muscle aging and staving off sarcopenia. Two other concepts pertinent to mitochondrial efficiency in exercise-trained skeletal muscle will be integrated in this review, including- mitophagy, the removal of dysfunctional mitochondrial via autophagy, as well as the implications of muscle fiber type changes with sarcopenia on mitochondrial function.
Collapse
Affiliation(s)
- Colin Harper
- Clinical Translation Unit (CTU), Tulane University, New Orleans, USA
| | - Venkatesh Gopalan
- Agency for Science, Technology & Research (A*STAR), Singapore Bioimaging Consortium (SBIC), Singapore, Singapore
| | - Jorming Goh
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore. .,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Centre for Healthy Longevity, National University Health System (NUHS), Singapore, Singapore.
| |
Collapse
|
25
|
Direito R, Rocha J, Sepodes B, Eduardo-Figueira M. Phenolic Compounds Impact on Rheumatoid Arthritis, Inflammatory Bowel Disease and Microbiota Modulation. Pharmaceutics 2021; 13:pharmaceutics13020145. [PMID: 33499333 PMCID: PMC7912052 DOI: 10.3390/pharmaceutics13020145] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/30/2020] [Accepted: 01/18/2021] [Indexed: 12/21/2022] Open
Abstract
Non-communicable chronic diseases (NCDs) are nowadays the principal cause of death, especially in most industrialized nations. These illnesses have increased exponentially with the consumption of diets very high in fat and sugar, not to mention stress and physical inactivity among other factors. The potential impact of suboptimal diets on NCDs’ morbidity and mortality rates brings to the forefront the necessity for a new way of improving dietary habits. The literature provides extensive scientific work that presents evidence that phenolic compounds from diets have antioxidant, anti-inflammatory and antiproliferative activities that impact human health. Gut microbiota modulation by some phenolic compounds leads to favorable changes in abundance, diversity, and in the immune system. However, polyphenol’s limited bioavailability needs to be overcome, highlighting their application in new delivery systems and providing their health benefits in well-established ways such as health maintenance, treatment or adjuvant to conventional pharmacological treatments. In this context, novel dietary approaches, including new food supplements, have emerged to prevent diseases and preserve health.
Collapse
Affiliation(s)
- Rosa Direito
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; (J.R.); (B.S.); (M.E.-F.)
- Correspondence: ; Tel.: +351-96-3654-899
| | - João Rocha
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; (J.R.); (B.S.); (M.E.-F.)
- Department of Pharmacy, Pharmacology and Health Technologies, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Bruno Sepodes
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; (J.R.); (B.S.); (M.E.-F.)
- Department of Pharmacy, Pharmacology and Health Technologies, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Maria Eduardo-Figueira
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; (J.R.); (B.S.); (M.E.-F.)
- Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| |
Collapse
|
26
|
Vaiserman A, Cuttler JM, Socol Y. Low-dose ionizing radiation as a hormetin: experimental observations and therapeutic perspective for age-related disorders. Biogerontology 2021; 22:145-164. [PMID: 33420860 PMCID: PMC7794644 DOI: 10.1007/s10522-020-09908-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 11/24/2020] [Indexed: 01/31/2023]
Abstract
Hormesis is any kind of biphasic dose-response when low doses of some agents are beneficial while higher doses are detrimental. Radiation hormesis is the most thoroughly investigated among all hormesis-like phenomena, in particular in biogerontology. In this review, we aimed to summarize research evidence supporting hormesis through exposure to low-dose ionizing radiation (LDIR). Radiation-induced longevity hormesis has been repeatedly reported in invertebrate models such as C. elegans, Drosophila and flour beetles and in vertebrate models including guinea pigs, mice and rabbits. On the contrary, suppressing natural background radiation was repeatedly found to cause detrimental effects in protozoa, bacteria and flies. We also discussed here the possibility of clinical use of LDIR, predominantly for age-related disorders, e.g., Alzheimer's disease, for which no remedies are available. There is accumulating evidence that LDIR, such as those commonly used in X-ray imaging including computer tomography, might act as a hormetin. Of course, caution should be exercised when introducing new medical practices, and LDIR therapy is no exception. However, due to the low average residual life expectancy in old patients, the short-term benefits of such interventions (e.g., potential therapeutic effect against dementia) may outweigh their hypothetical delayed risks (e.g., cancer). We argue here that assessment and clinical trials of LDIR treatments should be given priority bearing in mind the enormous economic, social and ethical implications of potentially-treatable, age-related disorders.
Collapse
|
27
|
Vatner SF, Zhang J, Oydanich M, Berkman T, Naftalovich R, Vatner DE. Healthful aging mediated by inhibition of oxidative stress. Ageing Res Rev 2020; 64:101194. [PMID: 33091597 PMCID: PMC7710569 DOI: 10.1016/j.arr.2020.101194] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/29/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022]
Abstract
The progressive increase in lifespan over the past century carries with it some adversity related to the accompanying burden of debilitating diseases prevalent in the older population. This review focuses on oxidative stress as a major mechanism limiting longevity in general, and healthful aging, in particular. Accordingly, the first goal of this review is to discuss the role of oxidative stress in limiting longevity, and compare healthful aging and its mechanisms in different longevity models. Secondly, we discuss common signaling pathways involved in protection against oxidative stress in aging and in the associated diseases of aging, e.g., neurological, cardiovascular and metabolic diseases, and cancer. Much of the literature has focused on murine models of longevity, which will be discussed first, followed by a comparison with human models of longevity and their relationship to oxidative stress protection. Finally, we discuss the extent to which the different longevity models exhibit the healthful aging features through physiological protective mechanisms related to exercise tolerance and increased β-adrenergic signaling and also protection against diabetes and other metabolic diseases, obesity, cancer, neurological diseases, aging-induced cardiomyopathy, cardiac stress and osteoporosis.
Collapse
Affiliation(s)
- Stephen F Vatner
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Newark, New Jersey, USA.
| | - Jie Zhang
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Newark, New Jersey, USA
| | - Marko Oydanich
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Newark, New Jersey, USA
| | - Tolga Berkman
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Newark, New Jersey, USA
| | - Rotem Naftalovich
- Department of Anesthesiology, New Jersey Medical School, Newark, New Jersey, USA
| | - Dorothy E Vatner
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Newark, New Jersey, USA.
| |
Collapse
|
28
|
Aquila G, Re Cecconi AD, Brault JJ, Corli O, Piccirillo R. Nutraceuticals and Exercise against Muscle Wasting during Cancer Cachexia. Cells 2020; 9:E2536. [PMID: 33255345 PMCID: PMC7760926 DOI: 10.3390/cells9122536] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer cachexia (CC) is a debilitating multifactorial syndrome, involving progressive deterioration and functional impairment of skeletal muscles. It affects about 80% of patients with advanced cancer and causes premature death. No causal therapy is available against CC. In the last few decades, our understanding of the mechanisms contributing to muscle wasting during cancer has markedly increased. Both inflammation and oxidative stress (OS) alter anabolic and catabolic signaling pathways mostly culminating with muscle depletion. Several preclinical studies have emphasized the beneficial roles of several classes of nutraceuticals and modes of physical exercise, but their efficacy in CC patients remains scant. The route of nutraceutical administration is critical to increase its bioavailability and achieve the desired anti-cachexia effects. Accumulating evidence suggests that a single therapy may not be enough, and a bimodal intervention (nutraceuticals plus exercise) may be a more effective treatment for CC. This review focuses on the current state of the field on the role of inflammation and OS in the pathogenesis of muscle atrophy during CC, and how nutraceuticals and physical activity may act synergistically to limit muscle wasting and dysfunction.
Collapse
Affiliation(s)
- Giorgio Aquila
- Neuroscience Department, Mario Negri Institute for Pharmacological Research IRCCS, 20156 Milan, Italy; (G.A.); (A.D.R.C.)
- Italian Institute for Planetary Health, IIPH, 20156 Milan, Italy;
| | - Andrea David Re Cecconi
- Neuroscience Department, Mario Negri Institute for Pharmacological Research IRCCS, 20156 Milan, Italy; (G.A.); (A.D.R.C.)
- Italian Institute for Planetary Health, IIPH, 20156 Milan, Italy;
| | - Jeffrey J. Brault
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Oscar Corli
- Italian Institute for Planetary Health, IIPH, 20156 Milan, Italy;
- Oncology Department, Mario Negri Institute for Pharmacological Research IRCCS, 20156 Milan, Italy
| | - Rosanna Piccirillo
- Neuroscience Department, Mario Negri Institute for Pharmacological Research IRCCS, 20156 Milan, Italy; (G.A.); (A.D.R.C.)
- Italian Institute for Planetary Health, IIPH, 20156 Milan, Italy;
| |
Collapse
|
29
|
Ostrom EL, Traustadóttir T. Aerobic exercise training partially reverses the impairment of Nrf2 activation in older humans. Free Radic Biol Med 2020; 160:418-432. [PMID: 32866619 PMCID: PMC7704731 DOI: 10.1016/j.freeradbiomed.2020.08.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/15/2020] [Accepted: 08/19/2020] [Indexed: 12/30/2022]
Abstract
Nuclear factor erythroid-2-related factor 2 (Nrf2), is an inducible transcription factor that improves redox balance through stimulating antioxidant gene expression. In older humans the Nrf2 response to a single bout of acute exercise is blunted compared to young indicating impaired redox signaling. The purpose of this randomized controlled trial was to investigate if the signaling impairment could be reversed with exercise training in older men and women, while also comparing to young. Young (18-28y, n = 21) and older (≥60y, n = 19) men and women were randomized to 8-week aerobic exercise training (ET; 3 d/wk, 45 min/d) or a non-exercise control group (CON). Nrf2 nuclear localization, gene expression for NQO1, HO1, and GCLC, and GCLC protein were measured in PBMCs in response to acute exercise trial (AET; 30-min cycling at 70% VO2 peak pre- and post-intervention at 7 timepoints (Pre, +10 m, +30 m, +1 h, +4 h, +8 h, +24 h). Young had greater Nrf2 signaling response compared to older at pre-intervention (p = 0.05), whereas the older had significantly higher basal Nrf2 levels (p = 0.004). ET decreased basal Nrf2 expression compared to CON (p = 0.032) and improved the Nrf2 signaling response in both young and older (p < 0.05). The degree of restoration in Nrf2 signaling response was related to the degree of change in basal Nrf2 (p = 0.039), which was driven by older adults (p = 0.014). Lower basal nuclear Nrf2 levels were associated with changes seen in AET responses for Nrf2 and GCLC protein, as well as NQO1 and GCLC mRNA. Together these data demonstrate that exercise training improves Nrf2 signaling and downstream gene expression and that lower basal Nrf2 levels are associated with a more dynamic acute response. Our results provide evidence that the impaired Nrf2 signaling in sedentary older adults can be restored to a degree with moderate exercise training, albeit not to the level seen in young. CLINICALTRIALS.GOV ID: NCT03419988.
Collapse
Affiliation(s)
- Ethan L Ostrom
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, USA
| | - Tinna Traustadóttir
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, USA.
| |
Collapse
|
30
|
Ruggiero AD, Davis A, Sherrill C, Westwood B, Hawkins GA, Palmer ND, Chou JW, Reeves T, Cox LA, Kavanagh K. Skeletal muscle extracellular matrix remodeling with worsening glycemic control in nonhuman primates. Am J Physiol Regul Integr Comp Physiol 2020; 320:R226-R235. [PMID: 33206559 DOI: 10.1152/ajpregu.00240.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Type 2 diabetes (T2D) development may be mediated by skeletal muscle (SkM) function, which is responsible for >80% of circulating glucose uptake. The goals of this study were to assess changes in global- and location-level gene expression, remodeling proteins, fibrosis, and vascularity of SkM with worsening glycemic control, through RNA sequencing, immunoblotting, and immunostaining. We evaluated SkM samples from health-diverse African green monkeys (Cholorcebus aethiops sabaeus) to investigate these relationships. We assessed SkM remodeling at the molecular level by evaluating unbiased transcriptomics in age-, sex-, weight-, and waist circumference-matched metabolically healthy, prediabetic (PreT2D) and T2D monkeys (n = 13). Our analysis applied novel location-specific gene differences and shows that extracellular facing and cell membrane-associated genes and proteins are highly upregulated in metabolic disease. We verified transcript patterns using immunohistochemical staining and protein analyses of matrix metalloproteinase 16 (MMP16), tissue inhibitor of metalloproteinase 2 (TIMP2), and VEGF. Extracellular matrix (ECM) functions to support intercellular communications, including the coupling of capillaries to muscle cells, which was worsened with increasing blood glucose. Multiple regression modeling from age- and health-diverse monkeys (n = 33) revealed that capillary density was negatively predicted by only fasting blood glucose. The loss of vascularity in SkM co-occurred with reduced expression of hypoxia-sensing genes, which is indicative of a disconnect between altered ECM and reduced endothelial cells, and known perfusion deficiencies present in PreT2D and T2D. This report supports that rising blood glucose values incite ECM remodeling and reduce SkM capillarization, and that targeting ECM would be a rational approach to improve health with metabolic disease.
Collapse
Affiliation(s)
- Alistaire D Ruggiero
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Ashley Davis
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Chrissy Sherrill
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Brian Westwood
- Department of Hypertension, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Gregory A Hawkins
- Center for Precision Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina.,Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Nicholette D Palmer
- Center for Precision Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina.,Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Jeff W Chou
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Tony Reeves
- Center for Precision Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Laura A Cox
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina.,Center for Precision Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Kylie Kavanagh
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina.,College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
31
|
Nunes S, Viana SD, Preguiça I, Alves A, Fernandes R, Teodoro JS, Figueirinha A, Salgueiro L, Silva S, Jarak I, Carvalho RA, Cavadas C, Rolo AP, Palmeira CM, Pintado MM, Reis F. Blueberry Consumption Challenges Hepatic Mitochondrial Bioenergetics and Elicits Transcriptomics Reprogramming in Healthy Wistar Rats. Pharmaceutics 2020; 12:pharmaceutics12111094. [PMID: 33202669 PMCID: PMC7697217 DOI: 10.3390/pharmaceutics12111094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 12/17/2022] Open
Abstract
An emergent trend of blueberries’ (BB) “prophylactic” consumption, due to their phytochemicals’ richness and well-known health-promoting claims, is widely scaled-up. However, the benefits arising from BB indiscriminate intake remains puzzling based on incongruent preclinical and human data. To provide a more in-depth elucidation and support towards a healthier and safer consumption, we conducted a translation-minded experimental study in healthy Wistar rats that consumed BB in a juice form (25 g/kg body weight (BW)/day; 14 weeks’ protocol). Particular attention was paid to the physiological adaptations succeeding in the gut and liver tissues regarding the acknowledged BB-induced metabolic benefits. Systemically, BB boosted serum antioxidant activity and repressed the circulating levels of 3-hydroxybutyrate (3-HB) ketone bodies and 3-HB/acetoacetate ratio. Moreover, BB elicited increased fecal succinic acid levels without major changes on gut microbiota (GM) composition and gut ultra-structural organization. Remarkably, an accentuated hepatic mitochondrial bioenergetic challenge, ensuing metabolic transcriptomic reprogramming along with a concerted anti-inflammatory pre-conditioning, was clearly detected upon long-term consumption of BB phytochemicals. Altogether, the results disclosed herein portray a quiescent mitochondrial-related metabolomics and hint for a unified adaptive response to this nutritional challenge. The beneficial or noxious consequences arising from this dietary trend should be carefully interpreted and necessarily claims future research.
Collapse
Affiliation(s)
- Sara Nunes
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.N.); (S.D.V.); (I.P.); (A.A.); (R.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
| | - Sofia D. Viana
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.N.); (S.D.V.); (I.P.); (A.A.); (R.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
- Polytechnic Institute of Coimbra, ESTESC-Coimbra Health School, Pharmacy/Biomedical Laboratory Sciences, 3046-854 Coimbra, Portugal
| | - Inês Preguiça
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.N.); (S.D.V.); (I.P.); (A.A.); (R.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
| | - André Alves
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.N.); (S.D.V.); (I.P.); (A.A.); (R.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
| | - Rosa Fernandes
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.N.); (S.D.V.); (I.P.); (A.A.); (R.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
| | - João S. Teodoro
- Department of Life Sciences, Faculty of Science and Technology (FCTUC), University of Coimbra, 3000-456 Coimbra, Portugal; (J.S.T.); (R.A.C.); (A.P.R.); (C.M.P.)
- Center for Neurosciences and Cell Biology of Coimbra (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Artur Figueirinha
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.F.); (L.S.)
- LAQV, REQUIMTE, Faculty of Pharmacy, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Lígia Salgueiro
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.F.); (L.S.)
- CIEPQPF, Chemical Process Engineering and Forest Products Research Centre, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Sara Silva
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (S.S.); (M.M.P.)
| | - Ivana Jarak
- Department of Microscopy, Laboratory of Cell Biology and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal;
| | - Rui A. Carvalho
- Department of Life Sciences, Faculty of Science and Technology (FCTUC), University of Coimbra, 3000-456 Coimbra, Portugal; (J.S.T.); (R.A.C.); (A.P.R.); (C.M.P.)
- Associated Laboratory for Green Chemistry-Clean Technologies and Processes, REQUIMTE, Faculty of Sciences and Technology, University of Porto, 4050-313 Porto, Portugal
| | - Cláudia Cavadas
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
- Center for Neurosciences and Cell Biology of Coimbra (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.F.); (L.S.)
| | - Anabela P. Rolo
- Department of Life Sciences, Faculty of Science and Technology (FCTUC), University of Coimbra, 3000-456 Coimbra, Portugal; (J.S.T.); (R.A.C.); (A.P.R.); (C.M.P.)
- Center for Neurosciences and Cell Biology of Coimbra (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Carlos M. Palmeira
- Department of Life Sciences, Faculty of Science and Technology (FCTUC), University of Coimbra, 3000-456 Coimbra, Portugal; (J.S.T.); (R.A.C.); (A.P.R.); (C.M.P.)
- Center for Neurosciences and Cell Biology of Coimbra (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Maria M. Pintado
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (S.S.); (M.M.P.)
| | - Flávio Reis
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.N.); (S.D.V.); (I.P.); (A.A.); (R.F.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal;
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
- Correspondence: ; Tel.: +351-239-480-053
| |
Collapse
|
32
|
Epel ES. The geroscience agenda: Toxic stress, hormetic stress, and the rate of aging. Ageing Res Rev 2020; 63:101167. [PMID: 32979553 PMCID: PMC7520385 DOI: 10.1016/j.arr.2020.101167] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 08/20/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023]
Abstract
Geroscience offers a counterpoint to the challenged pursuit of curing diseases of aging, by focusing on slowing the biological aging process for extended healthspan earlier in life. Remarkable progress has led this field toward animal trials and the next challenge lies with translation to humans. There is an emerging number of small human trials that can take advantage of new models integrating behavioral and social factors. Understanding dynamic aging mechanisms, given the powerful social determinants of aging (Crimmins, 2020) and human variability and environmental contexts (Moffitt, 2020), will be critical. Behavioral and social factors are intrinsic to aging. Toxic stressors broadly defined can lead to stress-acceleration of aging, either directly impacting aging processes or by shaping poor behavioral health, and underlie the socioeconomic disparities of aging. In contrast, hormetic stressors, acute intermittent stressors of moderate intensity, can produce stress resilience, the ability for quick recovery and possibly rejuvenation of cells and tissues. Although health research usually examines static biomarkers, aging is reflected in dynamic ability to recover from challenges pointing to new interventions and targets for examining mechanisms. A fuller model incorporating stress resilience provides innovative biobehavioral interventions, both for bolstering response to challenges, such as COVID-19, and for improving healthspan.
Collapse
Affiliation(s)
- Elissa S Epel
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, & Center for Health and Community, University of California, 3333 California St, Ste 465, San Francisco, CA, 94122, United States.
| |
Collapse
|
33
|
Penna F, Ballarò R, Costelli P. The Redox Balance: A Target for Interventions Against Muscle Wasting in Cancer Cachexia? Antioxid Redox Signal 2020; 33:542-558. [PMID: 32037856 DOI: 10.1089/ars.2020.8041] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Significance: The management of cancer patients is frequently complicated by the occurrence of a complex syndrome known as cachexia. It is mainly characterized by muscle wasting, a condition that associates with enhanced protein breakdown and with negative energy balance. While the mechanisms underlying cachexia have been only partially elucidated, understanding the pathogenesis of muscle wasting in cancer hosts is mandatory to design new targeted therapeutic strategies. Indeed, most of cancer patients will experience cachexia during the course of their disease, and about 25% of cancer-related deaths are due to this syndrome, rather than to the tumor itself. Recent Advances: Compelling evidence suggests that an altered redox homeostasis likely contributes to cancer-induced muscle protein depletion, directly or indirectly activating the intracellular degradative pathways. In addition, oxidative stress impinges on both mitochondrial number and function; the other way round, altered mitochondria lead to enhanced redox imbalance, creating a vicious loop that eventually results in negative energy metabolism. Critical Issues: The present review focuses on the possibility that pharmacological and nonpharmacological strategies able to restore a physiologic redox balance could be useful components of treatment schedules aimed at counteracting cancer-induced muscle wasting. Future Directions: Exercise and the use of exercise mimetic drugs represent the most promising approaches capable of reinforcing the muscle antioxidant defenses of cancer patients. The results from ongoing and new clinical trials are needed to validate the preclinical studies and provide effective therapies for cancer cachexia. Antioxid. Redox Signal. 33, 542-558.
Collapse
Affiliation(s)
- Fabio Penna
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Riccardo Ballarò
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Paola Costelli
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| |
Collapse
|
34
|
Powers SK, Deminice R, Ozdemir M, Yoshihara T, Bomkamp MP, Hyatt H. Exercise-induced oxidative stress: Friend or foe? JOURNAL OF SPORT AND HEALTH SCIENCE 2020; 9:415-425. [PMID: 32380253 PMCID: PMC7498668 DOI: 10.1016/j.jshs.2020.04.001] [Citation(s) in RCA: 304] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/21/2020] [Accepted: 02/18/2020] [Indexed: 05/02/2023]
Abstract
The first report demonstrating that prolonged endurance exercise promotes oxidative stress in humans was published more than 4 decades ago. Since this discovery, many ensuing investigations have corroborated the fact that muscular exercise increases the production of reactive oxygen species (ROS) and results in oxidative stress in numerous tissues including blood and skeletal muscles. Although several tissues may contribute to exercise-induced ROS production, it is predicted that muscular contractions stimulate ROS production in active muscle fibers and that skeletal muscle is a primary source of ROS production during exercise. This contraction-induced ROS generation is associated with (1) oxidant damage in several tissues (e.g., increased protein oxidation and lipid peroxidation), (2) accelerated muscle fatigue, and (3) activation of biochemical signaling pathways that contribute to exercise-induced adaptation in the contracting muscle fibers. While our understanding of exercise and oxidative stress has advanced rapidly during the last decades, questions remain about whether exercise-induced increases in ROS production are beneficial or harmful to health. This review addresses this issue by discussing the site(s) of oxidant production during exercise and detailing the health consequences of exercise-induced ROS production.
Collapse
Affiliation(s)
- Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, 32608, USA
| | - Rafael Deminice
- Department of Physical Education, State University of Londrina, Londrina, 10011, Brazil
| | - Mustafa Ozdemir
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, 32608, USA; Department of Exercise and Sport Sciences, Hacettepe University, Ankara, 06800, Turkey.
| | - Toshinori Yoshihara
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, 32608, USA; Department of Exercise Physiology, Juntendo University, Tokyo, 270-1695, Japan
| | - Matthew P Bomkamp
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, 32608, USA
| | - Hayden Hyatt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, 32608, USA
| |
Collapse
|
35
|
Ferri E, Marzetti E, Calvani R, Picca A, Cesari M, Arosio B. Role of Age-Related Mitochondrial Dysfunction in Sarcopenia. Int J Mol Sci 2020; 21:ijms21155236. [PMID: 32718064 PMCID: PMC7432902 DOI: 10.3390/ijms21155236] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/15/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023] Open
Abstract
Skeletal muscle aging is associated with a significant loss of skeletal muscle strength and power (i.e., dynapenia), muscle mass and quality of life, a phenomenon known as sarcopenia. This condition affects nearly one-third of the older population and is one of the main factors leading to negative health outcomes in geriatric patients. Notwithstanding the exact mechanisms responsible for sarcopenia are not fully understood, mitochondria have emerged as one of the central regulators of sarcopenia. In fact, there is a wide consensus on the assumption that the loss of mitochondrial integrity in myocytes is the main factor leading to muscle degeneration. Mitochondria are also key players in senescence. It has been largely proven that the modulation of mitochondrial functions can induce the death of senescent cells and that removal of senescent cells improves musculoskeletal health, quality, and function. In this review, the crosstalk among mitochondria, cellular senescence, and sarcopenia will be discussed with the aim to elucidate the role that the musculoskeletal cellular senescence may play in the onset of sarcopenia through the mediation of mitochondria.
Collapse
Affiliation(s)
- Evelyn Ferri
- Geriatric Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.C.); (B.A.)
- Correspondence:
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (E.M.); (R.C.); (A.P.)
- Geriatric Unit, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (E.M.); (R.C.); (A.P.)
- Geriatric Unit, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Anna Picca
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (E.M.); (R.C.); (A.P.)
- Geriatric Unit, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Matteo Cesari
- Geriatric Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.C.); (B.A.)
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Beatrice Arosio
- Geriatric Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.C.); (B.A.)
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| |
Collapse
|
36
|
Tai YK, Ng C, Purnamawati K, Yap JLY, Yin JN, Wong C, Patel BK, Soong PL, Pelczar P, Fröhlich J, Beyer C, Fong CHH, Ramanan S, Casarosa M, Cerrato CP, Foo ZL, Pannir Selvan RM, Grishina E, Degirmenci U, Toh SJ, Richards PJ, Mirsaidi A, Wuertz‐Kozak K, Chong SY, Ferguson SJ, Aguzzi A, Monici M, Sun L, Drum CL, Wang J, Franco‐Obregón A. Magnetic fields modulate metabolism and gut microbiome in correlation with
Pgc‐1α
expression: Follow‐up to an in vitro magnetic mitohormetic study. FASEB J 2020; 34:11143-11167. [DOI: 10.1096/fj.201903005rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 06/07/2020] [Accepted: 06/15/2020] [Indexed: 01/07/2023]
Affiliation(s)
- Yee Kit Tai
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Charmaine Ng
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
| | - Kristy Purnamawati
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Jasmine Lye Yee Yap
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Jocelyn Naixin Yin
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Craig Wong
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Bharati Kadamb Patel
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
| | - Poh Loong Soong
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Pawel Pelczar
- Centre for Transgenic Models University of Basel Basel Switzerland
- Institute of Laboratory Animal Science University of Zürich Zürich Switzerland
| | | | - Christian Beyer
- Centre Suisse d'électronique et de microtechnique, CSEM SA Neuchatel Switzerland
| | - Charlene Hui Hua Fong
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Sharanya Ramanan
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Marco Casarosa
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio” University of Florence Florence Italy
- Institute for Biomechanics ETH Zürich Zürich Switzerland
| | | | - Zi Ling Foo
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Rina Malathi Pannir Selvan
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Elina Grishina
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Ufuk Degirmenci
- Institute of Molecular and Cell Biology, A*STAR Singapore Singapore
| | - Shi Jie Toh
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Pete J. Richards
- Competence Center for Applied Biotechnology and Molecular Medicine University of Zürich Zürich Switzerland
| | - Ali Mirsaidi
- Competence Center for Applied Biotechnology and Molecular Medicine University of Zürich Zürich Switzerland
| | - Karin Wuertz‐Kozak
- Competence Center for Applied Biotechnology and Molecular Medicine University of Zürich Zürich Switzerland
- Department of Biomedical Engineering Rochester Institute of Technology (RIT) Rochester NY USA
- Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS) Singapore Singapore
| | - Suet Yen Chong
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS) Singapore Singapore
| | - Stephen J. Ferguson
- Institute of Molecular and Cell Biology, A*STAR Singapore Singapore
- Competence Center for Applied Biotechnology and Molecular Medicine University of Zürich Zürich Switzerland
| | - Adriano Aguzzi
- Institut für Neuropathologie Universitätsspital Zürich Zürich Switzerland
| | - Monica Monici
- ASAcampus JL, ASA Res. Div. ‐ Dept. of Experimental and Clinical Biomedical Sciences “Mario Serio” University of Florence Florence Italy
| | - Lei Sun
- DUKE‐NUS Graduate Medical School Singapore Singapore Singapore
| | - Chester L. Drum
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS) Singapore Singapore
| | - Jiong‐Wei Wang
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS) Singapore Singapore
- Department of Physiology Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
| | - Alfredo Franco‐Obregón
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
- Institute of Molecular and Cell Biology, A*STAR Singapore Singapore
- Department of Physiology Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Institute for Health Innovation & Technology, iHealthtech National University of Singapore Singapore Singapore
| |
Collapse
|
37
|
Moore MN. Lysosomes, Autophagy, and Hormesis in Cell Physiology, Pathology, and Age-Related Disease. Dose Response 2020; 18:1559325820934227. [PMID: 32684871 PMCID: PMC7343375 DOI: 10.1177/1559325820934227] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/02/2020] [Accepted: 05/08/2020] [Indexed: 12/17/2022] Open
Abstract
Autophagy has been strongly linked with hormesis, however, it is only relatively recently that the mechanistic basis underlying this association has begun to emerge. Lysosomal autophagy is a group of processes that degrade proteins, protein aggregates, membranes, organelles, segregated regions of cytoplasm, and even parts of the nucleus in eukaryotic cells. These degradative processes are evolutionarily very ancient and provide a survival capability for cells that are stressed or injured. Autophagy and autophagic dysfunction have been linked with many aspects of cell physiology and pathology in disease processes; and there is now intense interest in identifying various therapeutic strategies involving its regulation. The main regulatory pathway for augmented autophagy is the mechanistic target of rapamycin (mTOR) cell signaling, although other pathways can be involved, such as 5'-adenosine monophosphate-activated protein kinase. Mechanistic target of rapamycin is a key player in the many highly interconnected intracellular signaling pathways and is responsible for the control of cell growth among other processes. Inhibition of mTOR (specifically dephosphorylation of mTOR complex 1) triggers augmented autophagy and the search is on the find inhibitors that can induce hormetic responses that may be suitable for treating many diseases, including many cancers, type 2 diabetes, and age-related neurodegenerative conditions.
Collapse
Affiliation(s)
- Michael N. Moore
- European Centre for Environment & Human Health (ECEHH), University of Exeter Medical School, Knowledge Spa, Royal Cornwall Hospital, Truro, United Kingdom
- Plymouth Marine Laboratory, Plymouth, Devon, United Kingdom
- School of Biological & Marine Sciences, University of Plymouth, Plymouth, United Kingdom
| |
Collapse
|
38
|
Kumar R, Saraswat K, Rizvi SI. 2 -Deoxy - d-glucose at chronic low dose acts as a caloric restriction mimetic through a mitohormetic induction of ROS in the brain of accelerated senescence model of rat. Arch Gerontol Geriatr 2020; 90:104133. [PMID: 32559563 DOI: 10.1016/j.archger.2020.104133] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Aging induces significant molecular alteration in brain morphology. Glycolytic inhibitor 2-Deoxy-d-glucose (2-DG) is considered to act as a caloric restriction mimetic (CRM) but it is correlated with elevated mortality risk in rats at persistent high dosage. MATERIALS AND METHODS In young and d-galactose induced accelerated senescent rat aging models, we tested a persistent low-dose dietary 2-DG administration and evaluated various aging biomarkers in brain tissue. RESULTS A significant increase in reactive oxygen species (ROS) was observed in 2-DG treated (both young and accelerated senescent rat model). Increased Ferric reducing antioxidant potential (FRAP) value, Superoxide Dismutase (SOD), Catalase (CAT), and activity of mitochondrial complexes I and IV was observed. There was also significant improvements in the autophagy expression of genes (Beclin-1 and Atg-3) after 2- DG treatment. CONCLUSION We propose that 2-DG induces a mitohormetic effect through elevation of ROS which reinforces defensive mechanism(s) through increased FRAP, SOD, CAT and autophagy gene expression. Our observations indicate that a consistently low dose 2-DG could be a valuable CRM.
Collapse
Affiliation(s)
- Raushan Kumar
- Department of Biochemistry, University of Allahabad, Allahabad, 211002, India
| | - Komal Saraswat
- Department of Biochemistry, University of Allahabad, Allahabad, 211002, India
| | - Syed Ibrahim Rizvi
- Department of Biochemistry, University of Allahabad, Allahabad, 211002, India.
| |
Collapse
|
39
|
Lee SA, Sypniewski C, Bensadon BA, McLaren C, Donahoo WT, Sibille KT, Anton S. Determinants of Adherence in Time-Restricted Feeding in Older Adults: Lessons from a Pilot Study. Nutrients 2020; 12:nu12030874. [PMID: 32213965 PMCID: PMC7146127 DOI: 10.3390/nu12030874] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 12/26/2022] Open
Abstract
Time-restricted feeding (TRF) is a type of intermittent fasting in which no calories are commonly consumed for approximately 12-18 hours on a daily basis. The health benefits of this eating pattern have been shown in overweight adults, with improvements in cardiometabolic risk factors as well as the preservation of lean mass during weight loss. Although TRF has been well studied in younger and middle-aged adults, few studies have evaluated the effects of TRF in older adults. Thus, the goal of this study was to evaluate older-adult perspectives regarding the real-world advantages, disadvantages, and challenges to adopting a TRF eating pattern among participants aged 65 and over. A four-week single-arm pre- and post-test design was used for this clinical pilot trial TRF intervention study. Participants were instructed to fast for approximately 16 h per day with the daily target range between 14 and 18 h. Participants were provided with the TRF protocol at a baseline visit, along with a pictorial guide that depicted food items and beverages that were allowed and not allowed during fasting windows to reinforce that calorie-containing items were to be avoided. The trial interventionist called each participant weekly to promote adherence, review the protocol, monitor for adverse events, and provide support and guidance for any challenges faced during the intervention. Participants were instructed to complete daily eating time logs by recording the times at which they first consumed calories and when they stopped consuming calories. At the end of the intervention, participants completed an exit interview and a study-specific Diet Satisfaction Survey (Table 1) to assess their satisfaction, feasibility, and overall experience with the study intervention. Of the 10 participants who commenced the study (mean age = 77.1 y; 6 women, 4 men), nine completed the entire protocol. Seven of the ten participants reported easy adjustment to a 16-hour fast and rated the difference from normal eating patterns as minimal. Eight participants reported no decrease in energy during fasting periods, with greater self-reported activity levels in yardwork and light exercise. Adverse events were rare, and included transient headaches, which dissipated with increased water intake, and dizziness in one participant, which subsided with a small snack. The findings of the current trial suggest that TRF is an eating approach that is well tolerated by most older adults. Six participants, however, did not fully understand the requirements of the fasting regimen, despite being provided with specific instructions and a pictorial guide at a baseline visit. This suggests that more instruction and/or participant contact is needed in the early stages of a TRF intervention to promote adherence.
Collapse
Affiliation(s)
- Stephanie A. Lee
- Department of Aging and Geriatric Research, Institute on Aging, University of Florida, Gainesville, FL 32610, USA; (C.S.); (B.A.B.); (C.M.); (K.T.S.)
- Correspondence: (S.A.L.); (S.A.); Tel.: +352-273-7514 (S.A.)
| | - Caroline Sypniewski
- Department of Aging and Geriatric Research, Institute on Aging, University of Florida, Gainesville, FL 32610, USA; (C.S.); (B.A.B.); (C.M.); (K.T.S.)
| | - Benjamin A. Bensadon
- Department of Aging and Geriatric Research, Institute on Aging, University of Florida, Gainesville, FL 32610, USA; (C.S.); (B.A.B.); (C.M.); (K.T.S.)
| | - Christian McLaren
- Department of Aging and Geriatric Research, Institute on Aging, University of Florida, Gainesville, FL 32610, USA; (C.S.); (B.A.B.); (C.M.); (K.T.S.)
| | - William T. Donahoo
- Department of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Kimberly T. Sibille
- Department of Aging and Geriatric Research, Institute on Aging, University of Florida, Gainesville, FL 32610, USA; (C.S.); (B.A.B.); (C.M.); (K.T.S.)
| | - Stephen Anton
- Department of Aging and Geriatric Research, Institute on Aging, University of Florida, Gainesville, FL 32610, USA; (C.S.); (B.A.B.); (C.M.); (K.T.S.)
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL 32610, USA
- Correspondence: (S.A.L.); (S.A.); Tel.: +352-273-7514 (S.A.)
| |
Collapse
|
40
|
Stone V, Maciel August P, Scortegagna Crestani M, Brum Saccomori A, Magro BM, Moura Maurmann R, Santos BG, Peres Klein C, Matté C. Adaptive effects of gestational caloric restriction in the mitochondria of Wistar rats' brain: A DOHaD approach. Int J Dev Neurosci 2019; 79:1-10. [DOI: 10.1016/j.ijdevneu.2019.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/14/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022] Open
Affiliation(s)
- Vinícius Stone
- Programa de Pós‐graduação em Ciências Biológicas: BioquímicaInstituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do SulRua Ramiro Barcelos2600‐Anexo (laboratório 23)CEP 90035‐003Porto AlegreRSBrazil
| | - Pauline Maciel August
- Programa de Pós‐graduação em Ciências Biológicas: BioquímicaInstituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do SulRua Ramiro Barcelos2600‐Anexo (laboratório 23)CEP 90035‐003Porto AlegreRSBrazil
| | - Mariana Scortegagna Crestani
- Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do SulRua Ramiro Barcelos2600‐Anexo (laboratório 23)CEP 90035‐003Porto AlegreRSBrazil
| | - André Brum Saccomori
- Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do SulRua Ramiro Barcelos2600‐Anexo (laboratório 23)CEP 90035‐003Porto AlegreRSBrazil
| | - Bárbara Mariño Magro
- Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do SulRua Ramiro Barcelos2600‐Anexo (laboratório 23)CEP 90035‐003Porto AlegreRSBrazil
| | - Rafael Moura Maurmann
- Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do SulRua Ramiro Barcelos2600‐Anexo (laboratório 23)CEP 90035‐003Porto AlegreRSBrazil
| | - Bernardo Gindri Santos
- Programa de Pós‐graduação em Ciências Biológicas: BioquímicaInstituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do SulRua Ramiro Barcelos2600‐Anexo (laboratório 23)CEP 90035‐003Porto AlegreRSBrazil
| | - Caroline Peres Klein
- Programa de Pós‐graduação em Ciências Biológicas: BioquímicaInstituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do SulRua Ramiro Barcelos2600‐Anexo (laboratório 23)CEP 90035‐003Porto AlegreRSBrazil
| | - Cristiane Matté
- Programa de Pós‐graduação em Ciências Biológicas: BioquímicaInstituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do SulRua Ramiro Barcelos2600‐Anexo (laboratório 23)CEP 90035‐003Porto AlegreRSBrazil
- Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do SulRua Ramiro Barcelos2600‐Anexo (laboratório 23)CEP 90035‐003Porto AlegreRSBrazil
- Programa de Pós‐graduação em Ciências Biológicas: FisiologiaInstituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do SulRua Sarmento Leite, 500CEP 90050‐170Porto AlegreRSBrazil
| |
Collapse
|