1
|
Rathmacher JA, Pitchford LM, Stout JR, Townsend JR, Jäger R, Kreider RB, Campbell BI, Kerksick CM, Harty PS, Candow DG, Roberts BM, Arent SM, Kalman DS, Antonio J. International society of sports nutrition position stand: β-hydroxy-β-methylbutyrate (HMB). J Int Soc Sports Nutr 2025; 22:2434734. [PMID: 39699070 PMCID: PMC11740297 DOI: 10.1080/15502783.2024.2434734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024] Open
Abstract
Position Statement: The International Society of Sports Nutrition (ISSN) bases the following position stand on an analysis of the literature regarding the effects of β-Hydroxy-β-Methylbutyrate (HMB). The following 12 points have been approved by the Research Committee of the Society: 1. HMB is a metabolite of the amino acid leucine that is naturally produced in both humans and other animals. Two forms of HMB have been studied: Calcium HMB (HMB-Ca) and a free acid form of HMB (HMB-FA). HMB-FA appears to lead to increased appearance of HMB in the bloodstream when compared to HMB-Ca, though recent results are mixed. 2. The available safety/toxicity data suggest that chronic HMB-Ca and HMB-FA consumption are safe for oral HMB supplementation in humans up to at least one year. 3. There are no negative effects of HMB-Ca and HMB-FA on glucose tolerance and insulin sensitivity in humans. There may be improvements in glucose metabolism in younger adults. 4. The primary mode of action of HMB appears to be through its dual mechanism to enhance muscle protein synthesis and suppress muscle protein breakdown. HMB's activation of mTORC1 is independent of the leucine-sensing pathway (Sestrin2-GATOR2 complex). 5. HMB may help reduce muscle damage and promote muscle recovery, which can promote muscle growth/repair. HMB may also have anti-inflammatory effects, which could contribute to reducing muscle damage and soreness. 6. HMB consumption in close proximity to an exercise bout may be beneficial to increase muscle protein synthesis and attenuate the inflammatory response. HMB can provide a beneficial physiological effect when consumed both acutely and chronically in humans. 7. Daily HMB supplementation (38 mg/kg body weight) in combination with exercise training may improve body composition through increasing lean mass and/or decreasing fat mass with benefits in participants across age, sex, and training status. The most pronounced of these improvements in body composition with HMB have been observed in studies with robust resistance training programs and dietary control. 8. HMB may improve strength and power in untrained individuals, but its performance benefits in trained athletes are mixed and increase with an increase in study duration (>6 weeks). HMB's beneficial effects on athletic performance are thought to be driven by improved recovery. 9. HMB supplementation appears to potentially have a positive impact on aerobic performance, especially in trained athletes. The mechanisms of the effects are unknown. 10. HMB supplementation may be important in a non-exercising sedentary and aging population to improve muscle strength, functionality, and muscle quality. The effects of HMB supplementation with exercise are varied, but the combination may have a beneficial effect on the treatment of age-associated sarcopenia under select conditions. 11. HMB may be effective in countering muscle disuse atrophy during periods of inactivity due to illness or injury. The modulation of mitochondrial dynamics and lipid metabolism by HMB may be a potential mechanism for preventing disuse atrophy and aiding rehabilitation beyond HMB's effects on rates of muscle protein synthesis and degradation. 12. The efficacy of HMB in combination with certain nutrients may be enhanced under select conditions.
Collapse
Affiliation(s)
- John A. Rathmacher
- MTI Biotech Inc, Ames, IA, USA
- lowa State University, Department of Animal Science, Ames, IA, USA
| | - Lisa M. Pitchford
- MTI Biotech Inc, Ames, IA, USA
- Iowa State University, Department of Kinesiology, Ames, IA, USA
| | - Jeffrey R. Stout
- University of Central Florida, School of Kinesiology and Rehabilitation Sciences, Orlando, FL, USA
| | - Jeremy R. Townsend
- Research, Nutrition, and Innovation, AG1, Carson City, NV, USA
- Concordia University Chicago, Health & Human Performance, River Forest, IL, USA
| | | | - Richard B. Kreider
- Texas A&M University, Exercise & Sports Nutrition Lab, Department of Kinesiology and Sports Management, College Station, TX, USA
| | - Bill I. Campbell
- University of South Florida, Performance & Physique Enhancement Laboratory, Exercise Science Program, Tampa, FL, USA
| | - Chad M. Kerksick
- Lindenwood University, Exercise and Performance Nutrition Laboratory, College of Science, Technology, and Health, Saint Charles, MO, USA
| | - Patrick S. Harty
- Lindenwood University, Exercise and Performance Nutrition Laboratory, College of Science, Technology, and Health, Saint Charles, MO, USA
| | - Darren G. Candow
- University of Regina, Faculty of Kinesiology and Health Studies, Regina, SK, Canada
| | - Brandon M. Roberts
- 10 General Greene Ave, Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
| | - Shawn M. Arent
- University of South Carolina, Department of Exercise Science, Arnold School of Public Health, Columbia, SC, USA
| | - Douglas S. Kalman
- Dr. Kiran C Patel College of Osteopathic Medicine, Nova Southeastern University, Nutrition Department, Davie, FL, USA
| | - Jose Antonio
- Nova Southeastern University, Department of Health and Human Performance, Davie, FL, USA
| |
Collapse
|
2
|
Reifenberg P, Zimmer A. Branched-chain amino acids: physico-chemical properties, industrial synthesis and role in signaling, metabolism and energy production. Amino Acids 2024; 56:51. [PMID: 39198298 PMCID: PMC11358235 DOI: 10.1007/s00726-024-03417-2] [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: 06/13/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024]
Abstract
Branched-chain amino acids (BCAAs)-leucine (Leu), isoleucine (Ile), and valine (Val)-are essential nutrients with significant roles in protein synthesis, metabolic regulation, and energy production. This review paper offers a detailed examination of the physico-chemical properties of BCAAs, their industrial synthesis, and their critical functions in various biological processes. The unique isomerism of BCAAs is presented, focusing on analytical challenges in their separation and quantification as well as their solubility characteristics, which are crucial for formulation and purification applications. The industrial synthesis of BCAAs, particularly using bacterial strains like Corynebacterium glutamicum, is explored, alongside methods such as genetic engineering aimed at enhancing production, detailing the enzymatic processes and specific precursors. The dietary uptake, distribution, and catabolism of BCAAs are reviewed as fundamental components of their physiological functions. Ultimately, their multifaceted impact on signaling pathways, immune function, and disease progression is discussed, providing insights into their profound influence on muscle protein synthesis and metabolic health. This comprehensive analysis serves as a resource for understanding both the basic and complex roles of BCAAs in biological systems and their industrial application.
Collapse
Affiliation(s)
- Philipp Reifenberg
- Merck Life Science KGaA, Upstream R&D, Frankfurter Strasse 250, 64293, Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich‑Weiss‑Strasse 4, 64287, Darmstadt, Germany
| | - Aline Zimmer
- Merck Life Science KGaA, Upstream R&D, Frankfurter Strasse 250, 64293, Darmstadt, Germany.
| |
Collapse
|
3
|
Li G, Li Z, Liu J. Amino acids regulating skeletal muscle metabolism: mechanisms of action, physical training dosage recommendations and adverse effects. Nutr Metab (Lond) 2024; 21:41. [PMID: 38956658 PMCID: PMC11220999 DOI: 10.1186/s12986-024-00820-0] [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: 02/28/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024] Open
Abstract
Maintaining skeletal muscle mass is important for improving muscle strength and function. Hence, maximizing lean body mass (LBM) is the primary goal for both elite athletes and fitness enthusiasts. The use of amino acids as dietary supplements is widespread among athletes and physically active individuals. Extensive literature analysis reveals that branched-chain amino acids (BCAA), creatine, glutamine and β-alanine may be beneficial in regulating skeletal muscle metabolism, enhancing LBM and mitigating exercise-induced muscle damage. This review details the mechanisms of these amino acids, offering insights into their efficacy as supplements. Recommended dosage and potential side effects are then outlined to aid athletes in making informed choices and safeguard their health. Lastly, limitations within the current literature are addressed, highlighting opportunities for future research.
Collapse
Affiliation(s)
- Guangqi Li
- School of Physical Education, Northeast Normal university, No. 5268, Renmin Street, Changchun city, Jilin province, 130024, People's Republic of China
| | - Zhaojun Li
- Gaomi Municipal Center for Disease Control and Prevention, Gaomi city, Shandong, People's Republic of China
| | - Junyi Liu
- School of Physical Education, Northeast Normal university, No. 5268, Renmin Street, Changchun city, Jilin province, 130024, People's Republic of China.
| |
Collapse
|
4
|
Yadav A, Dabur R. Skeletal muscle atrophy after sciatic nerve damage: Mechanistic insights. Eur J Pharmacol 2024; 970:176506. [PMID: 38492879 DOI: 10.1016/j.ejphar.2024.176506] [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: 01/08/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 03/18/2024]
Abstract
Sciatic nerve injury leads to molecular events that cause muscular dysfunction advancement in atrophic conditions. Nerve damage renders muscles permanently relaxed which elevates intracellular resting Ca2+ levels. Increased Ca2+ levels are associated with several cellular signaling pathways including AMPK, cGMP, PLC-β, CERB, and calcineurin. Also, multiple enzymes involved in the tricarboxylic acid cycle and oxidative phosphorylation are activated by Ca2+ influx into mitochondria during muscle contraction, to meet increased ATP demand. Nerve damage induces mitophagy and skeletal muscle atrophy through increased sensitivity to Ca2+-induced opening of the permeability transition pore (PTP) in mitochondria attributed to Ca2+, ROS, and AMPK overload in muscle. Activated AMPK interacts negatively with Akt/mTOR is a highly prevalent and well-described central pathway for anabolic processes. Over the decade several reports indicate abnormal behavior of signaling machinery involved in denervation-induced muscle loss but end up with some controversial outcomes. Therefore, understanding how the synthesis and inhibitory stimuli interact with cellular signaling to control muscle mass and morphology may lead to new pharmacological insights toward understanding the underlying mechanism of muscle loss after sciatic nerve damage. Hence, the present review summarizes the existing literature on denervation-induced muscle atrophy to evaluate the regulation and expression of differential regulators during sciatic damage.
Collapse
Affiliation(s)
- Aarti Yadav
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Rajesh Dabur
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak, 124001, Haryana, India.
| |
Collapse
|
5
|
Cohen-Or M, Chapnik N, Froy O. β-Hydroxy-β-methylbutyrate (HMB) leads to phospholipase D2 (PLD2) activation and alters circadian rhythms in myotubes. Food Funct 2024; 15:4389-4398. [PMID: 38563085 DOI: 10.1039/d3fo04174c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
β-Hydroxy-β-methylbutyrate (HMB) is a breakdown product of leucine, which promotes muscle growth. Although some studies indicate that HMB activates AKT and mTOR, others show activation of the downstream effectors, P70S6K and S6, independent of mTOR. Our aim was to study the metabolic effect of HMB around the circadian clock in order to determine more accurately the signaling pathway involved. C2C12 myotubes were treated with HMB and clock, metabolic and myogenic markers were measured around the clock. HMB-treated C2C12 myotubes showed no activation of AKT and mTOR, but did show activation of P70S6K and S6. Activation of P70S6K and S6 was also found when myotubes were treated with HMB combined with metformin, an indirect mTOR inhibitor, or rapamycin, a direct mTOR inhibitor. The activation of the P70S6K and S6 independent of AKT and mTOR, was accompanied by increased activation of phospholipase D2 (PLD). In addition, HMB led to high amplitude and advanced circadian rhythms. In conclusion, HMB induces myogenesis in C2C12 by activating P70S6K and S6 via PLD2, rather than AKT and mTOR, leading to high amplitude advanced rhythms.
Collapse
Affiliation(s)
- Meytal Cohen-Or
- Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel.
| | - Nava Chapnik
- Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel.
| | - Oren Froy
- Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel.
| |
Collapse
|
6
|
Meza-Valderrama D, Sánchez-Rodríguez D, Messaggi-Sartor M, Muñoz-Redondo E, Morgado-Pérez A, Tejero-Sánchez M, De Jaime-Gil E, Leiva-Banuelos N, Marco E. Supplementation with β-hydroxy-β-methylbutyrate after resistance training in post-acute care patients with sarcopenia: A randomized, double-blind placebo-controlled trial. Arch Gerontol Geriatr 2024; 119:105323. [PMID: 38171034 DOI: 10.1016/j.archger.2023.105323] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2024]
Abstract
OBJECTIVES This study aimed to evaluate the efficacy of adding β-hydroxy-β- methylbutyrate (HMB) supplementation to a 12-week exercise-based rehabilitation program in older adults with sarcopenia after discharge from a post-acute geriatric rehabilitation unit. STUDY DESIGN A randomized, double-blind, placebo-controlled trial with two parallel groups. The intervention group received 3 g/day of Ca-HMB and participated in a 12- week resistance training program (3 sessions/week). The control group received a placebo and followed the same training program. MAIN OUTCOME MEASURES The primary outcomes were the improvements of handgrip strength and physical performance assessed through the Short Physical Performance Battery (SPPB) and 4-meter gait speed; and handgrip strength. All variables were assessed at baseline, post-intervention, and 1-year follow-up. RESULTS After completing the 12-week exercise program, the intervention group showed significant improvements in SPPB-Balance (1.3, 95 %CI 0.3 to 2.4) and total SPPB score (2.2, 95 %CI 0.4 to 4.0). Intra-group analysis demonstrated gains in the SPPB-Chair Stand (0.7 points, 95 %CI 0.0 to 1.4) and total SPPB score (2.1 points, 95 %CI 0.3 to 3.9) in the intervention group. Improvements in handgrip strength were observed in women (3.7 kg, 95 %CI: 0.2 to 7.3) at the end of the intervention, and persisted at the 1-year follow-up. CONCLUSIONS Our findings suggest that the supplementation of 3 g/day of Ca-HMB with resistance exercise may significantly enhance muscle strength and physical performance among older women with sarcopenia after recent hospitalization. Given this study's limitations, the intervention's effectiveness cannot be drawn, and further studies are needed.
Collapse
Affiliation(s)
- Delky Meza-Valderrama
- Rehabilitation Research Group, Hospital del Mar Medical Research Institute, Dr. Aiguader 80, 08003 Barcelona, Catalonia, Spain; Physical Medicine and Rehabilitation Department, National Institute of Physical Medicine and Rehabilitation, Vía Centenario, 0819 Panamá City, Panamá; Sistema Nacional de Investigación- Secretaria Nacional de Ciencia e Investigación, Clayton Ciudad del Saber Edificio 205, 0819 Panamá City, Panamá.
| | - Dolores Sánchez-Rodríguez
- Rehabilitation Research Group, Hospital del Mar Medical Research Institute, Dr. Aiguader 80, 08003 Barcelona, Catalonia, Spain; Geriatrics Department, Brugmann University Hospital, Université Libre de Bruxelles, Place A.Van Gehuchten 4, 1020 Brussels, Belgium; WHO Collaborating Center for Public Health aspects of musculoskeletal health and ageing. Division of Public Health, Epidemiology and Health Economics, University of Liège, Pl. du Vingt Août 7, 4000, Liège, Belgium
| | - Monique Messaggi-Sartor
- Rehabilitation Research Group, Hospital del Mar Medical Research Institute, Dr. Aiguader 80, 08003 Barcelona, Catalonia, Spain; Physical Medicine and Rehabilitation Department. Hospital del Mar - Centre Esperança. Sant Josep de la Muntanya 12, 08024 Barcelona, Catalonia, Spain
| | - Elena Muñoz-Redondo
- Rehabilitation Research Group, Hospital del Mar Medical Research Institute, Dr. Aiguader 80, 08003 Barcelona, Catalonia, Spain; Physical Medicine and Rehabilitation Department. Hospital del Mar - Centre Esperança. Sant Josep de la Muntanya 12, 08024 Barcelona, Catalonia, Spain
| | - Andrea Morgado-Pérez
- Rehabilitation Research Group, Hospital del Mar Medical Research Institute, Dr. Aiguader 80, 08003 Barcelona, Catalonia, Spain; Physical Medicine and Rehabilitation Department. Hospital del Mar - Centre Esperança. Sant Josep de la Muntanya 12, 08024 Barcelona, Catalonia, Spain
| | - Marta Tejero-Sánchez
- Rehabilitation Research Group, Hospital del Mar Medical Research Institute, Dr. Aiguader 80, 08003 Barcelona, Catalonia, Spain; Physical Medicine and Rehabilitation Department. Hospital del Mar - Centre Esperança. Sant Josep de la Muntanya 12, 08024 Barcelona, Catalonia, Spain
| | - Elisabet De Jaime-Gil
- Geriatrics Department, Hospital del Mar - Centre Fòrum, Carrer de Llull 410, 08019, Barcelona, Catalonia, Spain
| | - Nuria Leiva-Banuelos
- Geriatrics Department, Hospital del Mar - Centre Fòrum, Carrer de Llull 410, 08019, Barcelona, Catalonia, Spain
| | - Ester Marco
- Rehabilitation Research Group, Hospital del Mar Medical Research Institute, Dr. Aiguader 80, 08003 Barcelona, Catalonia, Spain; Physical Medicine and Rehabilitation Department. Hospital del Mar - Centre Esperança. Sant Josep de la Muntanya 12, 08024 Barcelona, Catalonia, Spain; Faculty of Health and Life Sciences, Universitat Pompeu Fabra, Dr Aiguader Building (Mar Campus), Dr. Aiguader 80, 08003 Barcelona, Catalonia, Spain
| |
Collapse
|
7
|
Zamary KM, Bruno RS. Advances in nutrition approaches to support vascular and gut health: 2023 David Kritchevsky Award winners. Nutr Res 2024; 124:111-115. [PMID: 38574579 DOI: 10.1016/j.nutres.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024]
Affiliation(s)
- Kaitlyn M Zamary
- Human Nutrition Program, The Ohio State University, Columbus, Ohio 43210
| | - Richard S Bruno
- Human Nutrition Program, The Ohio State University, Columbus, Ohio 43210.
| |
Collapse
|
8
|
Bruno RS, Zamary K. Reemergence from the pandemic, annual awards, and editorial office updates: from the desk of the Editor-in-Chief, Nutrition Research. Nutr Res 2024; 123:131-136. [PMID: 37474411 DOI: 10.1016/j.nutres.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Affiliation(s)
- Richard S Bruno
- Human Nutrition Program, The Ohio State University, Columbus, Ohio 43210.
| | - Katie Zamary
- Human Nutrition Program, The Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
9
|
Maroto R, Graber TG, Romsdahl TB, Kudlicki A, Russell WK, Rasmussen BB. Metabolomic and Lipidomic Signature of Skeletal Muscle with Constitutively Active Mechanistic Target of Rapamycin Complex 1. J Nutr 2023; 153:3397-3405. [PMID: 37898335 PMCID: PMC10739780 DOI: 10.1016/j.tjnut.2023.10.016] [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: 07/13/2023] [Revised: 09/07/2023] [Accepted: 10/03/2023] [Indexed: 10/30/2023] Open
Abstract
BACKGROUND Regulation of mechanistic target of rapamycin complex 1 (mTORC1) plays an important role in aging and nutrition. For example, caloric restriction reduces mTORC1 signaling and extends lifespan, whereas nutrient abundance and obesity increase mTORC1 signaling and reduce lifespan. Skeletal muscle-specific knockout (KO) of DEP domain-containing 5 protein (DEPDC5) results in constitutively active mTORC1 signaling, muscle hypertrophy and an increase in mitochondrial respiratory capacity. The metabolic profile of skeletal muscle, in the setting of hyperactive mTORC1 signaling, is not well known. OBJECTIVES To determine the metabolomic and lipidomic signature in skeletal muscle from female and male wild-type (WT) and DEPDC5 KO mice. METHODS Tibialis anterior (TA) muscles from WT and transgenic (conditional skeletal muscle-specific DEPDC5 KO) were obtained from female and male adult mice. Polar metabolites and lipids were extracted using a Bligh-Dyer extraction from 5 samples per group and identified and quantified by LC-MS/MS. Resulting analyte peak areas were analyzed with t-test, analysis of variance, and Volcano plots for group comparisons (e.g., WT compared with KO) and multivariate statistical analysis for genotype and sex comparisons. RESULTS A total of 162 polar metabolites (organic acids, amino acids, and amines and acyl carnitines) and 1141 lipid metabolites were detected in TA samples by LC-MS/MS. Few polar metabolites showed significant differences in KO muscles compared with WT within the same sex group. P-aminobenzoic acid, β-alanine, and dopamine were significantly higher in KO male muscle whereas erythrose-4-phosphate and oxoglutaric acid were significantly reduced in KO females. The lipidomic profile of the KO groups revealed an increase of muscle phospholipids and reduced triacylglycerol and diacylglycerol compared with the WT groups. CONCLUSIONS Sex differences were detected in polar metabolome and lipids were dependent on genotype. The metabolomic profile of mice with hyperactive skeletal muscle mTORC1 is consistent with an upregulation of mitochondrial function and amino acid utilization for protein synthesis.
Collapse
Affiliation(s)
- Rosario Maroto
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States.
| | - Ted G Graber
- Department of Physical Therapy, East Carolina University, Greenville, NC, United States
| | - Trevor B Romsdahl
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States
| | - Andrzej Kudlicki
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States
| | - William K Russell
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States
| | - Blake B Rasmussen
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States; Department of Biochemistry & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.
| |
Collapse
|
10
|
Shefflette A, Patel N, Caruso J. Mitigating Sarcopenia with Diet and Exercise. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:6652. [PMID: 37681791 PMCID: PMC10487983 DOI: 10.3390/ijerph20176652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/22/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023]
Abstract
Sarcopenia is the loss of muscle mass and function from aging, inactivity, or disuse. It is a comorbidity to numerous conditions that exacerbates their severity and adversely impacts activities of daily living. While sarcopenia now receives more attention from the medical community, people with sarcopenia as a comorbidity nevertheless still sometimes receives less attention than other presenting diseases or conditions. Inevitable doctors' visits or hospital stays for those with sarcopenia as a comorbidity have far higher healthcare costs than those without this condition, which imposes a greater financial burden on the medical insurance and healthcare industries. This review offers information and guidance on this topic. Treatments for sarcopenia include dietary, exercise, and pharmacological interventions. Yet, the latter treatment is only recommended in extreme cases as it may evoke numerous side effects and has little support in the scientific literature. Currently, a more holistic approach, with an emphasis on lifestyle modification, to reduce the likelihood of sarcopenia is examined. The current review discusses dietary and exercise interventions to limit the occurrence and severity of sarcopenia. References cited in this review conformed to the Declaration of Helsinki requirements for the use of human research subjects. Most of this review's references (~97%) came from a PubMed search that spanned from 1997 to 2023. Search terms included "sarcopenia" OR "muscle wasting" OR "geriatrics"; OR "ageing"; and AND "diet" OR "exercise". In addition, papers relevant or supportive of the topic as well as those considered seminal were included in the review. Over 96% of the references were peer-reviewed articles.
Collapse
Affiliation(s)
| | | | - John Caruso
- Exercise Physiology Program, University of Louisville, Louisville, KY 40292, USA; (A.S.); (N.P.)
| |
Collapse
|
11
|
Wan Y, Piao L, Xu S, Meng X, Huang Z, Inoue A, Wang H, Yue X, Jin X, Nan Y, Shi GP, Murohara T, Umegaki H, Kuzuya M, Cheng XW. Cathepsin S activity controls chronic stress-induced muscle atrophy and dysfunction in mice. Cell Mol Life Sci 2023; 80:254. [PMID: 37589754 PMCID: PMC10435624 DOI: 10.1007/s00018-023-04888-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/06/2023] [Accepted: 07/17/2023] [Indexed: 08/18/2023]
Abstract
Exposure to chronic psychological stress (CPS) is an intractable risk factor for inflammatory and metabolic diseases. Lysosomal cysteinyl cathepsins play an important role in human pathobiology. Given that cathepsin S (CTSS) is upregulated in the stressed vascular and adipose tissues, we investigated whether CTSS participates in chronic stress-induced skeletal muscle mass loss and dysfunction, with a special focus on muscle protein metabolic imbalance and apoptosis. Eight-week-old male wildtype (CTSS+/+) and CTSS-knockout (CTSS-/-) mice were randomly assigned to non-stress and variable-stress groups. CTSS+/+ stressed mice showed significant losses of muscle mass, dysfunction, and fiber area, plus significant mitochondrial damage. In this setting, stressed muscle in CTSS+/+ mice presented harmful alterations in the levels of insulin receptor substrate 2 protein content (IRS-2), phospho-phosphatidylinositol 3-kinase, phospho-protein kinase B, and phospho-mammalian target of rapamycin, forkhead box-1, muscle RING-finger protein-1 protein, mitochondrial biogenesis-related peroxisome proliferator-activated receptor-γ coactivator-α, and apoptosis-related B-cell lymphoma 2 and cleaved caspase-3; these alterations were prevented by CTSS deletion. Pharmacological CTSS inhibition mimics its genetic deficiency-mediated muscle benefits. In C2C12 cells, CTSS silencing prevented stressed serum- and oxidative stress-induced IRS-2 protein reduction, loss of the myotube myosin heavy chain content, and apoptosis accompanied by a rectification of investigated molecular harmful changes; these changes were accelerated by CTSS overexpression. These findings demonstrated that CTSS plays a role in IRS-2-related protein anabolism and catabolism and cell apoptosis in stress-induced muscle wasting, suggesting a novel therapeutic strategy for the control of chronic stress-related muscle disease in mice under our experimental conditions by regulating CTSS activity.
Collapse
Affiliation(s)
- Ying Wan
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
| | - Limei Piao
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China.
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China.
| | - Shengnan Xu
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
| | - Xiangkun Meng
- Department of Vascular Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Zhe Huang
- Department of Neurology, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Aiko Inoue
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, 4660855, Japan
| | - Hailong Wang
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
| | - Xueling Yue
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
| | - Xueying Jin
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
| | - Yongshan Nan
- Department of Anesthesiology, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China.
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi-ken, 466-8550, Japan
| | - Hiroyuki Umegaki
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, 4660855, Japan
- Department of Community Healthcare and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi-ken, 466-8550, Japan
| | - Masafumi Kuzuya
- Department of Community Healthcare and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi-ken, 466-8550, Japan
- Meitetsu Hospital, Nagoya, Aichi, 451-8511, Japan
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China.
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China.
| |
Collapse
|
12
|
Wan Y, Piao L, Xu S, Inoue A, Meng X, Lei Y, Huang Z, Wang H, Yue X, Shi GP, Kuzuya M, Cheng XW. Cathepsin S deficiency improves muscle mass loss and dysfunction via the modulation of protein metabolism in mice under pathological stress conditions. FASEB J 2023; 37:e23086. [PMID: 37428652 DOI: 10.1096/fj.202300395rrr] [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: 03/02/2023] [Revised: 06/19/2023] [Accepted: 06/27/2023] [Indexed: 07/12/2023]
Abstract
Cathepsin S (CTSS) is a widely expressed cysteinyl protease that has garnered attention because of its enzymatic and non-enzymatic functions under inflammatory and metabolic pathological conditions. Here, we examined whether CTSS participates in stress-related skeletal muscle mass loss and dysfunction, focusing on protein metabolic imbalance. Eight-week-old male wildtype (CTSS+/+ ) and CTSS-knockout (CTSS-/- ) mice were randomly assigned to non-stress and variable-stress groups for 2 weeks, and then processed for morphological and biochemical studies. Compared with non-stressed mice, stressed CTSS+/+ mice showed significant losses of muscle mass, muscle function, and muscle fiber area. In this setting, the stress-induced harmful changes in the levels of oxidative stress-related (gp91phox and p22phox ,), inflammation-related (SDF-1, CXCR4, IL-1β, TNF-α, MCP-1, ICAM-1, and VCAM-1), mitochondrial biogenesis-related (PPAR-γ and PGC-1α) genes and/or proteins and protein metabolism-related (p-PI3K, p-Akt, p-FoxO3α, MuRF-1, and MAFbx1) proteins; and these alterations were rectified by CTSS deletion. Metabolomic analysis revealed that stressed CTSS-/- mice exhibited a significant improvement in the levels of glutamine metabolism pathway products. Thus, these findings indicated that CTSS can control chronic stress-related skeletal muscle atrophy and dysfunction by modulating protein metabolic imbalance, and thus CTSS was suggested to be a promising new therapeutic target for chronic stress-related muscular diseases.
Collapse
Affiliation(s)
- Ying Wan
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, P.R. China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, P.R. China
| | - Limei Piao
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, P.R. China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, P.R. China
| | - Shengnan Xu
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, P.R. China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, P.R. China
| | - Aiko Inoue
- Institute of Nano-Life-Systems, Innovation for Future Society, Nagoya University Institutes of Innovation for Future Society, Nagoya, Japan
| | - Xiangkun Meng
- Department of Vascular Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanna Lei
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, P.R. China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, P.R. China
| | - Zhe Huang
- Department of Neurology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hailong Wang
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, P.R. China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, P.R. China
| | - Xueling Yue
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, P.R. China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, P.R. China
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Masafumi Kuzuya
- Institute of Nano-Life-Systems, Innovation for Future Society, Nagoya University Institutes of Innovation for Future Society, Nagoya, Japan
- Meitetsu Hospital, Nagoya, Japan
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, P.R. China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, P.R. China
| |
Collapse
|
13
|
Barranco A, Garcia L, Gruart A, Delgado-Garcia JM, Rueda R, Ramirez M. Effects of β-Hydroxy β-Methylbutyrate Supplementation on Working Memory and Hippocampal Long-Term Potentiation in Rodents. Nutrients 2022; 14:nu14051090. [PMID: 35268065 PMCID: PMC8912805 DOI: 10.3390/nu14051090] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/05/2023] Open
Abstract
β-hydroxy β-methylbutyrate (HMB), a metabolite of the essential amino acid leucine, has been shown to preserve muscle mass and strength during aging. The signaling mechanism by which HMB elicits its favorable effects on protein metabolism in skeletal muscle is also preserved in the brain. However, there are only a few studies, all at relatively high doses, addressing the effect of HMB supplementation on cognition. This study evaluated the effects of different doses of HMB on the potentiation of hippocampal synapses following the experimental induction of long-term potentiation (LTP) in the hippocampus of behaving rats, as well as on working memory test (delayed matching-to-position, DMTP) in mice. HMB doses in rats were 225 (low), 450 (medium), and 900 (high) mg/kg body weight/day and were double in mice. Rats who received medium or high HMB doses improved LTP, suggesting that HMB administration enhances mechanisms related to neuronal plasticity. In the DMTP test, mice that received any of the tested doses of HMB performed better than the control group in the overall test with particularities depending on the dose and the task phase.
Collapse
Affiliation(s)
- Alejandro Barranco
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain;
| | | | - Agnes Gruart
- Division of Neurosciences, Pablo de Olavide University, 41001 Seville, Spain; (A.G.); (J.M.D.-G.)
| | | | - Ricardo Rueda
- Abbott Nutrition, Research and Development, 18004 Granada, Spain;
| | - Maria Ramirez
- Abbott Nutrition, Research and Development, 18004 Granada, Spain;
- Correspondence: ; Tel.: +34-669-127998
| |
Collapse
|
14
|
Trinity JD, Drummond MJ, Fermoyle CC, McKenzie AI, Supiano MA, Richardson RS. Cardiovasomobility: an integrative understanding of how disuse impacts cardiovascular and skeletal muscle health. J Appl Physiol (1985) 2022; 132:835-861. [PMID: 35112929 PMCID: PMC8934676 DOI: 10.1152/japplphysiol.00607.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cardiovasomobility is a novel concept that encompasses the integration of cardiovascular and skeletal muscle function in health and disease with critical modification by physical activity, or lack thereof. Compelling evidence indicates that physical activity improves health while a sedentary, or inactive, lifestyle accelerates cardiovascular and skeletal muscle dysfunction and hastens disease progression. Identifying causative factors for vascular and skeletal muscle dysfunction, especially in humans, has proven difficult due to the limitations associated with cross-sectional investigations. Therefore, experimental models of physical inactivity and disuse, which mimic hospitalization, injury, and illness, provide important insight into the mechanisms and consequences of vascular and skeletal muscle dysfunction. This review provides an overview of the experimental models of disuse and inactivity and focuses on the integrated responses of the vasculature and skeletal muscle in response to disuse/inactivity. The time course and magnitude of dysfunction evoked by various models of disuse/inactivity are discussed in detail, and evidence in support of the critical roles of mitochondrial function and oxidative stress are presented. Lastly, strategies aimed at preserving vascular and skeletal muscle dysfunction during disuse/inactivity are reviewed. Within the context of cardiovasomobility, experimental manipulation of physical activity provides valuable insight into the mechanisms responsible for vascular and skeletal muscle dysfunction that limit mobility, degrade quality of life, and hasten the onset of disease.
Collapse
Affiliation(s)
- Joel D Trinity
- Salt Lake City Veteran Affairs Medical Center Geriatric Research, Education, and Clinical Center, Salt Lake City, Utah.,Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| | - Micah J Drummond
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah.,Department of Physical Therapy, University of Utah, Salt Lake City, Utah
| | - Caitlin C Fermoyle
- Salt Lake City Veteran Affairs Medical Center Geriatric Research, Education, and Clinical Center, Salt Lake City, Utah.,Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Alec I McKenzie
- Salt Lake City Veteran Affairs Medical Center Geriatric Research, Education, and Clinical Center, Salt Lake City, Utah.,Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Mark A Supiano
- Salt Lake City Veteran Affairs Medical Center Geriatric Research, Education, and Clinical Center, Salt Lake City, Utah.,Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Russell S Richardson
- Salt Lake City Veteran Affairs Medical Center Geriatric Research, Education, and Clinical Center, Salt Lake City, Utah.,Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| |
Collapse
|
15
|
Aoki K, Konno M, Tokinoya K, Honda K, Abe T, Nagata T, Takehara M, Sugasawa T, Takekoshi K, Ohmori H. Long-Term Habitual Exercise and Combination of β-Hydroxy-β-Methylbutyrate plus Black Ginger Alter the Autophagy and Mitochondria Related Genes in SAMP8 Mice. J Nutr Sci Vitaminol (Tokyo) 2022; 68:39-46. [PMID: 35228494 DOI: 10.3177/jnsv.68.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Muscle mass and strength decrease with aging; however, habitual exercise can maintain muscle health. β-Hydroxy-β-methyl butyrate calcium (HMB) and black ginger (BG) improve muscle protein metabolism and energy production. Combining these two molecules, which have similar effects, may have a synergistic effect. Senescence-accelerated mouse-prone 8 (SAMP8) is a useful model of muscle aging. Therefore, we explored how the combination of habitual exercise, HMB, and BG affected muscle aging. We used 28-wk-old (28w) SAMP8 mice divided into six groups: 28 wk (28w), 44 wk (44w, Con), exercise (Ex), Ex+BG, Ex+HMB, and Ex+BG+HMB (Ex+Comb). Mice were required to run on a treadmill for 16 wk for 5 d per week. In 28w and 44w mice, grip strength tests and dissection were conducted. Muscle weight was measured, and qPCR and immunoblotting were conducted. Muscle mass and strength were declined in the 44w group. Exercise with HMB or BG alone had no effect, whereas muscle mass and strength were augmented in the Ex+Comb group. Similarly, levels of mitochondrial function- and biogenesis-related genes were increased. Autophagy-related protein (Atg3, 7, 16L1 and Beclin1) were altered in the Ex+Comb group. These results suggest that Ex+Comb affects autophagy. Overall, the combination of habitual exercise and HMB+BG may enhance muscle mass and strength by affecting the mitochondrial and autophagy systems in SAMP8.
Collapse
Affiliation(s)
- Kai Aoki
- Faculty of Medicine, University of Tsukuba
| | - Masaki Konno
- Graduate School of Comprehensive Human Sciences, University of Tsukuba
| | - Katsuyuki Tokinoya
- Japan Society for the Promotion of Science
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University
| | - Katsunari Honda
- School of Physical Education, Health and Sport Sciences, University of Tsukuba
| | - Takuya Abe
- Market and Product Development Department, Zenyaku Hanbai Co., Ltd
| | - Takeshi Nagata
- Pharmacology, Department of Drug Discovery, R&D Center. Zenyaku Kogyo Co., Ltd
| | | | | | | | - Hajime Ohmori
- Faculty of Health and Sport Sciences, University of Tsukuba
| |
Collapse
|
16
|
A Proton-Coupled Transport System for β-Hydroxy-β-Methylbutyrate (HMB) in Blood-Brain Barrier Endothelial Cell Line hCMEC/D3. Nutrients 2021; 13:nu13093220. [PMID: 34579098 DOI: 10.3390/nu13093220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 12/31/2022] Open
Abstract
β-Hydroxy-β-methylbutyrate (HMB), a leucine metabolite, is used as a nutritional ingredient to improve skeletal muscle health. Preclinical studies indicate that this supplement also elicits significant benefits in the brain; it promotes neurite outgrowth and prevents age-related reductions in neuronal dendrites and cognitive performance. As orally administered HMB elicits these effects in the brain, we infer that HMB crosses the blood-brain barrier (BBB). However, there have been no reports detailing the transport mechanism for HMB in BBB. Here we show that HMB is taken up in the human BBB endothelial cell line hCMEC/D3 via H+-coupled monocarboxylate transporters that also transport lactate and β-hydroxybutyrate. MCT1 (monocarboxylate transporter 1) and MCT4 (monocarboxylate transporter 4) belonging to the solute carrier gene family SLC16 (solute carrier, gene family 16) are involved, but additional transporters also contribute to the process. HMB uptake in BBB endothelial cells results in intracellular acidification, demonstrating cotransport with H+. Since HMB is known to activate mTOR with potential to elicit transcriptomic changes, we examined the influence of HMB on the expression of selective transporters. We found no change in MCT1 and MCT4 expression. Interestingly, the expression of LAT1 (system L amino acid transporter 1), a high-affinity transporter for branched-chain amino acids relevant to neurological disorders such as autism, is induced. This effect is dependent on mTOR (mechanistic target of rapamycine) activation by HMB with no involvement of histone deacetylases. These studies show that HMB in systemic circulation can cross the BBB via carrier-mediated processes, and that it also has a positive influence on the expression of LAT1, an important amino acid transporter in the BBB.
Collapse
|
17
|
Mann G, Mora S, Madu G, Adegoke OAJ. Branched-chain Amino Acids: Catabolism in Skeletal Muscle and Implications for Muscle and Whole-body Metabolism. Front Physiol 2021; 12:702826. [PMID: 34354601 PMCID: PMC8329528 DOI: 10.3389/fphys.2021.702826] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/24/2021] [Indexed: 12/20/2022] Open
Abstract
Branched-chain amino acids (BCAAs) are critical for skeletal muscle and whole-body anabolism and energy homeostasis. They also serve as signaling molecules, for example, being able to activate mammalian/mechanistic target of rapamycin complex 1 (mTORC1). This has implication for macronutrient metabolism. However, elevated circulating levels of BCAAs and of their ketoacids as well as impaired catabolism of these amino acids (AAs) are implicated in the development of insulin resistance and its sequelae, including type 2 diabetes, cardiovascular disease, and of some cancers, although other studies indicate supplements of these AAs may help in the management of some chronic diseases. Here, we first reviewed the catabolism of these AAs especially in skeletal muscle as this tissue contributes the most to whole body disposal of the BCAA. We then reviewed emerging mechanisms of control of enzymes involved in regulating BCAA catabolism. Such mechanisms include regulation of their abundance by microRNA and by post translational modifications such as phosphorylation, acetylation, and ubiquitination. We also reviewed implications of impaired metabolism of BCAA for muscle and whole-body metabolism. We comment on outstanding questions in the regulation of catabolism of these AAs, including regulation of the abundance and post-transcriptional/post-translational modification of enzymes that regulate BCAA catabolism, as well the impact of circadian rhythm, age and mTORC1 on these enzymes. Answers to such questions may facilitate emergence of treatment/management options that can help patients suffering from chronic diseases linked to impaired metabolism of the BCAAs.
Collapse
Affiliation(s)
| | | | | | - Olasunkanmi A. J. Adegoke
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| |
Collapse
|
18
|
Sumi K, Sakuda M, Munakata K, Nakamura K, Ashida K. α-Hydroxyisocaproic Acid Decreases Protein Synthesis but Attenuates TNFα/IFNγ Co-Exposure-Induced Protein Degradation and Myotube Atrophy via Suppression of iNOS and IL-6 in Murine C2C12 Myotube. Nutrients 2021; 13:nu13072391. [PMID: 34371902 PMCID: PMC8308709 DOI: 10.3390/nu13072391] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/25/2022] Open
Abstract
There is ongoing debate as to whether or not α-hydroxyisocaproic acid (HICA) positively regulates skeletal muscle protein synthesis resulting in the gain or maintenance of skeletal muscle. We investigated the effects of HICA on mouse C2C12 myotubes under normal conditions and during cachexia induced by co-exposure to TNFα and IFNγ. The phosphorylation of AMPK or ERK1/2 was significantly altered 30 min after HICA treatment under normal conditions. The basal protein synthesis rates measured by a deuterium-labeling method were significantly lowered by the HICA treatment under normal and cachexic conditions. Conversely, myotube atrophy induced by TNFα/IFNγ co-exposure was significantly improved by the HICA pretreatment, and this improvement was accompanied by the inhibition of iNOS expression and IL-6 production. Moreover, HICA also suppressed the TNFα/IFNγ co-exposure-induced secretion of 3-methylhistidine. These results demonstrated that HICA decreases basal protein synthesis under normal or cachexic conditions; however, HICA might attenuate skeletal muscle atrophy via maintaining a low level of protein degradation under cachexic conditions.
Collapse
|
19
|
Gao R, Chilibeck PD. Nutritional interventions during bed rest and spaceflight: prevention of muscle mass and strength loss, bone resorption, glucose intolerance, and cardiovascular problems. Nutr Res 2020; 82:11-24. [PMID: 32890860 DOI: 10.1016/j.nutres.2020.07.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/02/2020] [Accepted: 07/01/2020] [Indexed: 12/22/2022]
Abstract
Bed rest is necessary for many medical conditions but also used as a ground-based model for space flight (along with head-down tilt to simulate fluid shifts in microgravity). The purpose of this review is to examine nutritional interventions during bed rest and spaceflight for prevention of muscle and strength loss, glucose intolerance, bone resorption, and cardiovascular problems. Increased dietary protein intake and supplementation with amino acids, β-hydroxy-β-methylbutyrate, or cofactors with antioxidant properties are effective for ameliorating bed rest-induced loss of muscle mass and strength. Previous literature involving bed rest with dietary protein/amino acid supplementation had mixed findings, likely due to differences in dosage. Although high protein intake in some studies prevents bed rest-induced muscle loss, it also increases bone resorption. High calcium intake and vitamin D supplementation are not beneficial for preventing bone degradation during bed rest or spaceflight. Very few studies investigated countermeasures to prevent glucose intolerance and cardiovascular risks during bed rest/spaceflight. Low-glycemic index diets might be beneficial for the prevention of bed rest-induced glucose intolerance and cardiovascular problems. The present evidence warrants additional studies on the exact threshold of protein/amino acid intake to prevent the loss of muscle mass and strength during bed rest/spaceflight specifically to maintain the beneficial effects of proteins on muscle mass and function without increasing bone resorption. Furthermore, it is suggested to study the effects of vitamin K supplementation on bone health during bed rest/spaceflight and determine the role of long-term low-glycemic index diets on glucose regulation and cardiovascular health during extended bed rest.
Collapse
Affiliation(s)
- Ruirui Gao
- College of Kinesiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada S7N 5B2
| | - Philip D Chilibeck
- College of Kinesiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada S7N 5B2.
| |
Collapse
|
20
|
Jakubowski JS, Nunes EA, Teixeira FJ, Vescio V, Morton RW, Banfield L, Phillips SM. Supplementation with the Leucine Metabolite β-hydroxy-β-methylbutyrate (HMB) does not Improve Resistance Exercise-Induced Changes in Body Composition or Strength in Young Subjects: A Systematic Review and Meta-Analysis. Nutrients 2020; 12:nu12051523. [PMID: 32456217 PMCID: PMC7285233 DOI: 10.3390/nu12051523] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 01/11/2023] Open
Abstract
β-hydroxy-β-methylbutyrate (HMB) is a leucine metabolite that is purported to increase fat-free mass (FFM) gain and performance in response to resistance exercise training (RET). The aim of this systematic review and meta-analysis was to determine the efficacy of HMB supplementation in augmenting FFM and strength gains during RET in young adults. Outcomes investigated were: total body mass (TBM), FFM, fat mass (FM), total single repetition maximum (1RM), bench press (BP) 1RM, and lower body (LwB) 1RM. Databases consulted were: Medical Literature Analysis and Retrieval System Online (Medline), Excerpta Medica database (Embase), The Cumulative Index to Nursing and Allied Health Literature (CINAHL), and SportDiscus. Fourteen studies fit the inclusion criteria; however, 11 were analyzed after data extraction and funnel plot analysis exclusion. A total of 302 participants (18-45 y) were included in body mass and composition analysis, and 248 were included in the strength analysis. A significant effect was found on TBM. However, there were no significant effects for FFM, FM, or strength outcomes. We conclude that HMB produces a small effect on TBM gain, but this effect does not translate into significantly greater increases in FFM, strength or decreases in FM during periods of RET. Our findings do not support the use of HMB aiming at improvement of body composition or strength with RET.
Collapse
Affiliation(s)
- Josephine S. Jakubowski
- Department of Kinesiology, McMaster University, Hamilton, ON L8S 4L8, Canada; (J.S.J.); (E.A.N.); (V.V.); (R.W.M.)
| | - Everson A. Nunes
- Department of Kinesiology, McMaster University, Hamilton, ON L8S 4L8, Canada; (J.S.J.); (E.A.N.); (V.V.); (R.W.M.)
- Department of Physiological Sciences, Federal University of Santa Catarina, Florianópolis, SC 88040-900, Brazil
| | - Filipe J. Teixeira
- CBIOS—Universidade Lusófona’s Research Center for Biosciences and Health Technologies, Campo Grande, 1749 024 Lisboa, Portugal;
| | - Victoria Vescio
- Department of Kinesiology, McMaster University, Hamilton, ON L8S 4L8, Canada; (J.S.J.); (E.A.N.); (V.V.); (R.W.M.)
| | - Robert W. Morton
- Department of Kinesiology, McMaster University, Hamilton, ON L8S 4L8, Canada; (J.S.J.); (E.A.N.); (V.V.); (R.W.M.)
| | - Laura Banfield
- Health Sciences Library, McMaster University, Hamilton, ON L8S 4L8, Canada;
| | - Stuart M. Phillips
- Department of Kinesiology, McMaster University, Hamilton, ON L8S 4L8, Canada; (J.S.J.); (E.A.N.); (V.V.); (R.W.M.)
- Correspondence: ; Tel.: +1-905-525-9140 (ext. 24465)
| |
Collapse
|
21
|
Regulation of Skeletal Muscle Function by Amino Acids. Nutrients 2020; 12:nu12010261. [PMID: 31963899 PMCID: PMC7019684 DOI: 10.3390/nu12010261] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/09/2020] [Accepted: 01/16/2020] [Indexed: 12/30/2022] Open
Abstract
Amino acids are components of proteins that also exist free-form in the body; their functions can be divided into (1) nutritional, (2) sensory, and (3) biological regulatory roles. The skeletal muscle, which is the largest organ in the human body, representing ~40% of the total body weight, plays important roles in exercise, energy expenditure, and glucose/amino acid usage—processes that are modulated by various amino acids and their metabolites. In this review, we address the metabolism and function of amino acids in the skeletal muscle. The expression of PGC1α, a transcriptional coactivator, is increased in the skeletal muscle during exercise. PGC1α activates branched-chain amino acid (BCAA) metabolism and is used for energy in the tricarboxylic acid (TCA) cycle. Leucine, a BCAA, and its metabolite, β-hydroxy-β-methylbutyrate (HMB), both activate mammalian target of rapamycin complex 1 (mTORC1) and increase protein synthesis, but the mechanisms of activation appear to be different. The metabolite of valine (another BCAA), β-aminoisobutyric acid (BAIBA), is increased by exercise, is secreted by the skeletal muscle, and acts on other tissues, such as white adipose tissue, to increase energy expenditure. In addition, several amino acid-related molecules reportedly activate skeletal muscle function. Oral 5-aminolevulinic acid (ALA) supplementation can protect against mild hyperglycemia and help prevent type 2 diabetes. β-alanine levels are decreased in the skeletal muscles of aged mice. β-alanine supplementation increased the physical performance and improved the executive function induced by endurance exercise in middle-aged individuals. Further studies focusing on the effects of amino acids and their metabolites on skeletal muscle function will provide data essential for the production of food supplements for older adults, athletes, and individuals with metabolic diseases.
Collapse
|
22
|
Suryawan A, Rudar M, Fiorotto ML, Davis TA. Differential regulation of mTORC1 activation by leucine and β-hydroxy-β-methylbutyrate in skeletal muscle of neonatal pigs. J Appl Physiol (1985) 2020; 128:286-295. [PMID: 31944890 DOI: 10.1152/japplphysiol.00332.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Leucine (Leu) and its metabolite β-hydroxy-β-methylbutyrate (HMB) stimulate mechanistic target of rapamycin (mTOR) complex 1 (mTORC1)-dependent protein synthesis in the skeletal muscle of neonatal pigs. This study aimed to determine whether HMB and Leu utilize common nutrient-sensing mechanisms to activate mTORC1. In study 1, neonatal pigs were fed one of five diets for 24 h: low protein (LP), high protein (HP), or LP supplemented with 4 (LP+HMB4), 40 (LP+HMB40), or 80 (LP+HMB80) μmol HMB·kg body wt-1·day-1. In study 2, neonatal pigs were fed for 24 h: LP, LP supplemented with Leu (LP+Leu), or HP diets delivering 9, 18, and 18 mmol Leu·kg body wt-1·day-1, respectively. The upstream signaling molecules that regulate mTORC1 activity were analyzed. mTOR phosphorylation on Ser2448 and Ser2481 was greater in LP+HMB40, LP+HMB80, and LP+Leu than in LP and greater in HP than in HMB-supplemented groups (P < 0.05), whereas HP and LP+Leu were similar. Rheb-mTOR complex formation was lower in LP than in HP (P < 0.05), with no enhancement by HMB or Leu supplementation. The Sestrin2-GATOR2 complex was more abundant in LP than in HP and was reduced by Leu (P < 0.05) but not HMB supplementation. RagA-mTOR and RagC-mTOR complexes were higher in LP+Leu and HP than in LP and HMB groups (P < 0.05). There were no treatment differences in RagB-SH3BP4, Vps34-LRS, and RagD-LRS complex abundances. Phosphorylation of Erk1/2 and TSC2, but not AMPK, was lower in LP than HP (P < 0.05) and unaffected by HMB or Leu supplementation. Our results demonstrate that HMB stimulates mTORC1 activation in neonatal muscle independent of the leucine-sensing pathway mediated by Sestrin2 and the Rag proteins.NEW & NOTEWORTHY Dietary supplementation with either leucine or its metabolite β-hydroxy-β-methylbutyrate (HMB) stimulates protein synthesis in skeletal muscle of the neonatal pig. Our results demonstrate that both leucine and HMB stimulate mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) phosphorylation in neonatal muscle. This leucine-stimulated process involves dissociation of the Sestrin2-GATOR2 complex and increased binding of Rag A/C to mTOR. However, HMB's activation of mTORC1 is independent of this leucine-sensing pathway.
Collapse
Affiliation(s)
- Agus Suryawan
- US Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Marko Rudar
- US Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Marta L Fiorotto
- US Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Teresa A Davis
- US Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
23
|
Effect of citrulline on muscle protein turnover in an in vitro model of muscle catabolism. Nutrition 2020; 71:110597. [PMID: 31896062 DOI: 10.1016/j.nut.2019.110597] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/12/2019] [Accepted: 09/11/2019] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Muscle net catabolism, as seen after severe trauma or sepsis or in postoperative situations, is mediated by hormones (e.g., cortisol) and proinflammatory cytokines (e.g., tumor necrosis factor alpha [TNF-α]). Specific amino acids may be able to limit this muscle mass loss. Citrulline (CIT) stimulates muscle protein synthesis in various situations, but little data exist on hypercatabolic situations and the effects on protein breakdown are unknown. Our aim was to assess the effect of CIT on protein turnover in an in vitro model of muscle hypercatabolism. METHODS Myotubes derived from C2C12 myoblasts were treated with 150 nM dexamethasone (DEX), 10 ng/mL TNF-α, or 0.006% ethanol (as control [CON]) for 24 h. Subsequently, myotubes were incubated with or without 5 mM CIT for 6 h. Muscle protein synthesis rate was evaluated by the surface sensing of translation method and by l-[3,5-3H]tyrosine (Tyr) incorporation. The muscle protein breakdown rate was evaluated from Tyr release into culture medium. CIT action was analyzed by non-parametric Kruskal-Wallis and Mann-Whitney tests. RESULTS CIT treatment significantly increased protein synthesis rates compared with the DEX or TNF-α group (surface sensing of translation method; DEX + CIT versus DEX; P = 0.03 and TNF-α+CIT versus TNF-α; P = 0.05) and significantly decreased protein breakdown rate in the CON and DEX groups (CON + CIT versus CON; P = 0.05 and DEX + CIT versus DEX; P = 0.05). CONCLUSIONS CIT treatment regulated muscle protein turnover in an in vitro model of muscle net catabolism. Exploring the underlying mechanisms would also be of interest.
Collapse
|
24
|
Duan Y, Zhong Y, Song B, Zheng C, Xu K, Kong X, Li F. Suppression of protein degradation by leucine requires its conversion to β-hydroxy-β-methyl butyrate in C2C12 myotubes. Aging (Albany NY) 2019; 11:11922-11936. [PMID: 31881014 PMCID: PMC6949090 DOI: 10.18632/aging.102509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 11/18/2019] [Indexed: 05/05/2023]
Abstract
The aims of this study were to investigate whether the inhibitory effect of Leucine (Leu) on starvation-induced protein degradation was mediated by its metabolite β-hydroxy-β-methyl butyrate (HMB), and to explore the mechanisms involved. The results showed that the beneficial effects of Leu on protein degradation and the oxygen consumption rate (OCR) of cells were observed at low levels (0.5 mM) rather than at high levels (10 mM). However, these effects were inferior to those of HMB. Moreover, HMB was able to increase/decrease the proportion of MyHC I/MyHC IIb protein expression, respectively. In these KICD-transfected cells, Leu was approximately as effective as HMB in inhibiting protein degradation and increasing the OCR as well as MyHC I protein expression of cells, and these effects of Leu were reverted to a normal state by mesotrione, a specific suppressor of KICD. In conclusion, HMB seems to be an active metabolite of Leu to suppress muscle protein degradation in a starvation model, and the mechanisms may be associated with improved mitochondrial oxidative capacity in muscle cells.
Collapse
Affiliation(s)
- Yehui Duan
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, Hunan, P. R. China
| | - Yinzhao Zhong
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, Hunan, P. R. China
| | - Bo Song
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, Hunan, P. R. China
| | - Changbing Zheng
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou, Guangdong, China
| | - Kang Xu
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, Hunan, P. R. China
| | - Xiangfeng Kong
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, Hunan, P. R. China
| | - Fengna Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, Hunan, P. R. China
- Hunan Co-Innovation Center of Animal Production Safety, CICAPS, Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, Changsha, Hunan, China
| |
Collapse
|
25
|
Transport Mechanisms for the Nutritional Supplement β-Hydroxy-β-Methylbutyrate (HMB) in Mammalian Cells. Pharm Res 2019; 36:84. [PMID: 30997560 DOI: 10.1007/s11095-019-2626-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 04/08/2019] [Indexed: 10/27/2022]
Abstract
PURPOSE β-Hydroxy-β-methylbutyrate (HMB), a nutritional supplement, elicits anabolic activity in muscle. Here we investigated the mechanism of HMB uptake in muscle cells. METHODS Murine muscle cells (C2C12) and human mammary epithelial cells (MCF7) were used for uptake. As HMB is a monocarboxylate, focus was on monocarboxylate transporters, monitoring interaction of HMB with H+-coupled lactate uptake, and influence of H+ directly on HMB uptake. Involvement of MCT1-4 was studied using selective inhibitors and gene silencing. Involvement of human Na+/monocarboxylate transporter SMCT1 was also assessed using Xenopus oocytes. RESULTS H+-coupled lactate uptake was inhibited by HMB in both mammalian cells. HMB uptake was H+-coupled and inhibited by lactate. C2C12 cells expressed MCT1 and MCT4; MCF7 cells expressed MCT1-4; undifferentiated C2C12 cells expressed SMCT1. SMCT1 mediated Na+-coupled HMB transport. Inhibitors of MCT1/4, siRNA-mediated gene silencing, and expression pattern showed that MCT1-4 were responsible only for a small portion of HMB uptake in these cells. CONCLUSION HMB uptake in C2C12 and MCF7 cells is primarily H+-coupled and inhibited by lactate, but MCT1-4 are only partly responsible for HMB uptake. SMCT1 also transports HMB, but in a Na+-coupled manner. Other, yet unidentified, transporters mediate the major portion of HMB uptake in C2C12 and MCF7 cells.
Collapse
|
26
|
Gepner Y, Varanoske AN, Boffey D, Hoffman JR. Benefits of β-hydroxy-β-methylbutyrate supplementation in trained and untrained individuals. Res Sports Med 2018; 27:204-218. [PMID: 30348016 DOI: 10.1080/15438627.2018.1533470] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
β-Hydroxy-β-Methylbutyrate (HMB) is a metabolite of the branched-chain amino acid leucine and its ketoacid α-ketoisocaproate. HMB has been widely used as an ergogenic supplement to increase muscle strength, muscle hypertrophy and enhance recovery. The physiological mechanisms that underlie these benefits are related to HMB's ability to stimulate muscle protein synthesis and minimize muscle breakdown. Although evidence supporting the benefits of HMB supplementation is not conclusive, many of these studies have suffered from methodological flaws including different formulations, supplement duration and population studied. HMB in its free acid formulation is suggestive of having a greater potential for efficacy in both trained and untrained populations than its calcium-salt form. However, the evidence regarding HMB's role in limiting muscle degradation and increasing muscle protein synthesis has created an exciting interest in examining its efficacy among untrained individuals. Recent investigations examining intense training have demonstrated efficacy in maintaining muscle mass and attenuating the inflammatory response.
Collapse
Affiliation(s)
- Yftach Gepner
- a Institute of Exercise Physiology and Wellness, Sport and Exercise Science , University of Central Florida , Orlando , FL , USA
| | - Alyssa N Varanoske
- a Institute of Exercise Physiology and Wellness, Sport and Exercise Science , University of Central Florida , Orlando , FL , USA
| | - David Boffey
- a Institute of Exercise Physiology and Wellness, Sport and Exercise Science , University of Central Florida , Orlando , FL , USA
| | - Jay R Hoffman
- a Institute of Exercise Physiology and Wellness, Sport and Exercise Science , University of Central Florida , Orlando , FL , USA
| |
Collapse
|
27
|
Arazi H, Taati B, Suzuki K. A Review of the Effects of Leucine Metabolite (β-Hydroxy-β-methylbutyrate) Supplementation and Resistance Training on Inflammatory Markers: A New Approach to Oxidative Stress and Cardiovascular Risk Factors. Antioxidants (Basel) 2018; 7:antiox7100148. [PMID: 30347824 PMCID: PMC6210682 DOI: 10.3390/antiox7100148] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/03/2018] [Accepted: 10/18/2018] [Indexed: 01/03/2023] Open
Abstract
β-hydroxy β-methylbutyrate (HMB) is a bioactive metabolite formed from the breakdown of the branched-chain amino acid, leucine. Given the popularity of HMB supplements among different athletes, specifically, those who participate in regular resistance training, this review was performed to summarize current literature on some aspects of HMB supplementation that have received less attention. Because of the small number of published studies, it has not been possible to conclude the exact effects of HMB on cardiovascular parameters, oxidative stress, and inflammatory markers. Thus, the interpretation of outcomes should be taken cautiously. However, the data presented here suggest that acute HMB supplementation may attenuate the pro-inflammatory response following an intense bout of resistance exercise in athletes. Also, the available findings collectively indicate that chronic HMB consumption with resistance training does not improve cardiovascular risk factors and oxidative stress markers greater than resistance training alone. Taken together, there is clearly a need for further well-designed, long-term studies to support these findings and determine whether HMB supplementation affects the adaptations induced by resistance training associated with the body’s inflammatory condition, antioxidative defense system, and cardiovascular risk factors in humans.
Collapse
Affiliation(s)
- Hamid Arazi
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Guilan, Rasht 4199843653, Iran.
| | - Behzad Taati
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Guilan, Rasht 4199843653, Iran.
| | - Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan.
| |
Collapse
|
28
|
Antunes L, Yamada AK, Pertille A. EFFECTS OF BETA-HYDROXY-BETA-METHYL BUTYRATE IN MUSCLE REGENERATION OF RATS. REV BRAS MED ESPORTE 2018. [DOI: 10.1590/1517-869220182402181184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
ABSTRACT Introduction: Studies have shown that beta-hydroxy-beta-methyl butyrate (HMB) supplementation increases muscle strength and mass. Objective: To evaluate the effect of HMB supplementation on the muscle regeneration process in young and sedentary rats. Methods: Twenty-four male Wistar rats two months old were divided into two groups: lesion (LE) and supplemented (S), and evaluated in two moments - seven days (LE7; S7, n=6) and 21 days (LE21; S21, n=6). The right tibialis anterior muscle was subjected to cryolesion in all animals. After the injury, the LE group remained in the vivarium without any intervention. Group S received HMB calcium supplementation diluted in water by gavage (320 mg/kg/weight per day). The injury tibialis anterior (ITA), the tibialis anterior (TA), and the left soleus (SOL) muscles were removed, weighted and divided transversally into two parts, one for the analysis of the cross-sectional area (CSA) and the area of inflammation/regeneration and the other part to measure the muscular glycogen concentration. Data were evaluated using the SAS program considering mean and standard deviation. For analysis of variance the ANOVA test was used followed by the Tukey-HSD test (p≤0.05). Results: The ITA muscle weight was higher in S21 compared to S7 (p<0.05). The groups LE21 and S21 presented greater CSA of muscle fibers area and smaller ITA regeneration/inflammation area (p<0.05) when compared with the LE7 and S7 groups. There was an increase in muscle glycogen levels in S7 group compared to LE7 and S21 groups for TA and SOL (p<0.01), as well as in S21 group compared to LE21 for SOL (p<0.05). Conclusion: HMB did not influence the muscle regeneration process and did not favor anabolic activity in the different muscular fibers of young sedentary rats. Level of Evidence II; Therapeutic studies - Investigation of treatment results.
Collapse
|
29
|
Ogasawara S, Cheng XW, Inoue A, Hu L, Piao L, Yu C, Goto H, Xu W, Zhao G, Lei Y, Yang G, Kimura K, Umegaki H, Shi GP, Kuzuya M. Cathepsin K activity controls cardiotoxin-induced skeletal muscle repair in mice. J Cachexia Sarcopenia Muscle 2018; 9:160-175. [PMID: 29058826 PMCID: PMC5803616 DOI: 10.1002/jcsm.12248] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/30/2017] [Accepted: 09/05/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cathepsin K (CatK) is a widely expressed cysteine protease that has gained attention because of its enzymatic and non-enzymatic functions in signalling. Here, we examined whether CatK-deficiency (CatK-/- ) would mitigate injury-related skeletal muscle remodelling and fibrosis in mice, with a special focus on inflammation and muscle cell apoptosis. METHODS Cardiotoxin (CTX, 20 μM/200 μL) was injected into the left gastrocnemius muscle of male wild-type (CatK+/+ ) and CatK-/- mice, and the mice were processed for morphological and biochemical studies. RESULTS On post-injection Day 14, CatK deletion ameliorated muscle interstitial fibrosis and remodelling and performance. At an early time point (Day 3), CatK-/- reduced the lesion macrophage and leucocyte contents and cell apoptosis, the mRNA levels of monocyte chemoattractant protein-1, toll-like receptor-2 and toll-like receptor-4, and the gelatinolytic activity related to matrix metalloproteinase-2/-9. CatK deletion also restored the protein levels of caspase-3 and cleaved caspase-8 and the ratio of the BAX to the Bcl-2. Moreover, CatK deficiency protected muscle fibre laminin and desmin disorder in response to CTX injury. These beneficial muscle effects were mimicked by CatK-specific inhibitor treatment. In vitro experiments demonstrated that pharmacological CatK inhibition reduced the apoptosis of C2C12 mouse myoblasts and the levels of BAX and caspase-3 proteins induced by CTX. CONCLUSIONS These results demonstrate that CatK plays an essential role in skeletal muscle loss and fibrosis in response to CTX injury, possibly via a reduction of inflammation and cell apoptosis, suggesting a novel therapeutic strategy for the control of skeletal muscle diseases by regulating CatK activity.
Collapse
Affiliation(s)
- Shinyu Ogasawara
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan
| | - Xian Wu Cheng
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan.,Institute of Innovation for Future Society, Nagoya University, Nagoya, 464-0814, Aichiken, Japan.,Department of Cardiology and ICU, Yanbian University Hospital, Yanji, 133000, Jilin, China.,Department of Internal Medicine, Kyung Hee University, Seoul, 130-702, Korea
| | - Aiko Inoue
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan.,Institute of Innovation for Future Society, Nagoya University, Nagoya, 464-0814, Aichiken, Japan
| | - Lina Hu
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan.,Department of Public Health, Guilin Medical College, Guilin, 541004, Guangxi, China
| | - Limei Piao
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan.,Department of Cardiology and ICU, Yanbian University Hospital, Yanji, 133000, Jilin, China
| | - Chenglin Yu
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan.,Department of Cardiology and ICU, Yanbian University Hospital, Yanji, 133000, Jilin, China
| | - Hiroki Goto
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan
| | - Wenhu Xu
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan.,Department of Cardiology and ICU, Yanbian University Hospital, Yanji, 133000, Jilin, China
| | - Guangxian Zhao
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan.,Department of Cardiology and ICU, Yanbian University Hospital, Yanji, 133000, Jilin, China
| | - Yanna Lei
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan.,Department of Cardiology and ICU, Yanbian University Hospital, Yanji, 133000, Jilin, China
| | - Guang Yang
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan.,Department of Cardiology and ICU, Yanbian University Hospital, Yanji, 133000, Jilin, China
| | - Kaoru Kimura
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan
| | - Hiroyuki Umegaki
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 20115, USA
| | - Masafumi Kuzuya
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan.,Institute of Innovation for Future Society, Nagoya University, Nagoya, 464-0814, Aichiken, Japan
| |
Collapse
|
30
|
Duan Y, Li F, Guo Q, Wang W, Zhang L, Wen C, Chen X, Yin Y. β-Hydroxy-β-methyl Butyrate Is More Potent Than Leucine in Inhibiting Starvation-Induced Protein Degradation in C2C12 Myotubes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:170-176. [PMID: 29227681 DOI: 10.1021/acs.jafc.7b04841] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Leucine (Leu) and its metabolites α-ketoisocaproate (KIC) and β-hydroxy-β-methyl butyrate (HMB) are potent regulators of protein turnover. The aim of this study was to compare the inhibitory effects of Leu, KIC, and HMB on protein degradation and to investigate the mechanisms involved. The results showed that the inhibitory effect of HMB (0.38 ± 0.04) was more potent than that of Leu (0.76 ± 0.04) and KIC (0.56 ± 0.04, P < 0.01), and was significantly abolished in the presence of LY294002 (1.48 ± 0.02) and rapamycin (1.96 ± 0.02, P < 0.01). In the presence of insulin, the inhibitory effect of HMB (0.34 ± 0.03) was still more effective than that of Leu (0.60 ± 0.04) and KIC (0.57 ± 0.08, P < 0.05). Interestingly, LY294002 treatment markedly attenuated the effect of HMB, while rapamycin treatment failed to exert the same effect. Thus, HMB appears to be more potent than Leu and KIC in inhibiting protein degradation in the absence or presence of insulin, and this inhibitory effect may be dependent on PI3K/Akt signaling pathway regardless of insulin, and mTOR signaling was only involved in this effect of HMB in the absence of insulin.
Collapse
Affiliation(s)
- Yehui Duan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture Chinese Academy of Sciences; Key Laboratory of Agro-ecological Processes in Subtropical Region; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture , Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Fengna Li
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture Chinese Academy of Sciences; Key Laboratory of Agro-ecological Processes in Subtropical Region; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture , Changsha 410125, China
- Hunan Co-Innovation Center of Animal Production Safety, CICAPS; Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, Changsha, Hunan 410128, China
| | - Qiuping Guo
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture Chinese Academy of Sciences; Key Laboratory of Agro-ecological Processes in Subtropical Region; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture , Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Wenlong Wang
- Laboratory of Animal Nutrition and Human Health, School of Biology, Hunan Normal University , Changsha, Hunan 410018, China
| | - Lingyu Zhang
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture Chinese Academy of Sciences; Key Laboratory of Agro-ecological Processes in Subtropical Region; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture , Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Chaoyue Wen
- Laboratory of Animal Nutrition and Human Health, School of Biology, Hunan Normal University , Changsha, Hunan 410018, China
| | - Xiao'an Chen
- Hunan Shengshi Fenghua Biological Technology Co., Ltd., Longhui 422200, China
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture Chinese Academy of Sciences; Key Laboratory of Agro-ecological Processes in Subtropical Region; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture , Changsha 410125, China
- Laboratory of Animal Nutrition and Human Health, School of Biology, Hunan Normal University , Changsha, Hunan 410018, China
| |
Collapse
|
31
|
[Molecular mechanism of sarcopenia]. Nihon Ronen Igakkai Zasshi 2018; 55:13-24. [PMID: 29503355 DOI: 10.3143/geriatrics.55.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
|
32
|
Bargi-Souza P, Goulart-Silva F, Nunes MT. Novel aspects of T 3 actions on GH and TSH synthesis and secretion: physiological implications. J Mol Endocrinol 2017; 59:R167-R178. [PMID: 28951438 DOI: 10.1530/jme-17-0068] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 09/26/2017] [Indexed: 12/27/2022]
Abstract
Thyroid hormones (THs) classically regulate the gene expression by transcriptional mechanisms. In pituitary, the encoding genes for growth hormone (GH) and thyroid-stimulating hormone (TSH) are examples of genes regulated by triiodothyronine (T3) in a positive and negative way, respectively. Recent studies have shown a rapid adjustment of GH and TSH synthesis/secretion induced by T3 posttranscriptional actions. In somatotrophs, T3 promotes an increase in Gh mRNA content, poly(A) tail length and binding to the ribosome, associated with a rearrangement of actin cytoskeleton. In thyrotrophs, T3 reduces Tshb mRNA content, poly(A) tail length and its association with the ribosome. In parallel, it promotes a redistribution of TSH secretory granules to more distal regions of the cell periphery, indicating a rapid effect of T3 inhibition of TSH secretion. T3 was shown to affect the content of tubulin and the polymerization of actin and tubulin cytoskeletons in the whole anterior pituitary gland, and to increase intracellular alpha (CGA) content. This review summarizes genomic and non-genomic/posttranscriptional actions of TH on the regulation of several steps of GH and TSH synthesis and secretion. These distinct mechanisms induced by T3 can occur simultaneously, even though non-genomic effects are promptly elicited and precede the genomic actions, coexisting in a functional network within the cells.
Collapse
Affiliation(s)
| | | | - M T Nunes
- Department of Physiology and Biophysics of the Institute of Biomedical SciencesUniversity of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
33
|
Fan J, Yang X, Li J, Shu Z, Dai J, Liu X, Li B, Jia S, Kou X, Yang Y, Chen N. Spermidine coupled with exercise rescues skeletal muscle atrophy from D-gal-induced aging rats through enhanced autophagy and reduced apoptosis via AMPK-FOXO3a signal pathway. Oncotarget 2017; 8:17475-17490. [PMID: 28407698 PMCID: PMC5392263 DOI: 10.18632/oncotarget.15728] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 02/06/2017] [Indexed: 12/20/2022] Open
Abstract
The quality control of skeletal muscle is a continuous requirement throughout the lifetime, although its functions and quality present as a declining trend during aging process. Dysfunctional or deficient autophagy and excessive apoptosis may contribute to the atrophy of senescent skeletal muscle. Spermidine, as a natural polyamine, can be involved in important cellular functions for lifespan extension and stress resistance in several model organisms through activating autophagy. Similarly, cellular autophagic responses to exercise have also been extensively investigated. In the present study, in order to confirm the mitigation or amelioration of skeletal muscle atrophy in aging rats through spermidine coupled with exercise intervention and explore corresponding mechanisms, the rat model with aging-related atrophy of skeletal muscle was established by intraperitoneal injection of D-galactose (D-gal) (200 mg/kgd), and model rats were subjected to the intervention with spermidine (5 mg/kgd) or swimming (60 min/d, 5 d/wk) or combination for 42 days. Spermidine coupled with exercise could attenuate D-gal-induced aging-related atrophy of skeletal muscle through induced autophagy and reduced apoptosis with characteristics of more autophagosomes, activated mitophagy, enhanced mitochondrial quality, alleviated cell shrinkage, and less swollen mitochondria under transmission scanning microscopic observation. Meanwhile, spermidine coupled with exercise could induce autophagy through activating AMPK-FOXO3a signal pathway with characterization of increased Beclin1 and LC3-II/LC3-I ratio, up-regulated anti-apoptotic Bcl-2, down-regulated pro-apoptotic Bax and caspase-3, as well as activated AMPK and FOXO3a. Therefore, spermidine combined with exercise can execute the prevention or treatment of D-gal-induced aging-related skeletal muscle atrophy through enhanced autophagy and reduced apoptosis mediated by AMPK-FOXO3a signal pathway.
Collapse
Affiliation(s)
- Jingjing Fan
- Hubei Key Laboratory of Exercise Training and Monitoring, Hubei Provincial Collaborative Innovation Center for Exercise and Health Promotion, College of Health Science, Wuhan Sports University, Wuhan, China
| | - Xiaoqi Yang
- Graduate School, Wuhan Sports University, Wuhan, China
| | - Jie Li
- Graduate School, Wuhan Sports University, Wuhan, China
| | - Ziyang Shu
- Graduate School, Wuhan Sports University, Wuhan, China
| | - Jun Dai
- Graduate School, Wuhan Sports University, Wuhan, China
| | - Xingran Liu
- Graduate School, Wuhan Sports University, Wuhan, China
| | - Biao Li
- Graduate School, Jilin Sport University, Changchun, China
| | - Shaohui Jia
- Hubei Key Laboratory of Exercise Training and Monitoring, Hubei Provincial Collaborative Innovation Center for Exercise and Health Promotion, College of Health Science, Wuhan Sports University, Wuhan, China
| | - Xianjuan Kou
- Hubei Key Laboratory of Exercise Training and Monitoring, Hubei Provincial Collaborative Innovation Center for Exercise and Health Promotion, College of Health Science, Wuhan Sports University, Wuhan, China
| | - Yi Yang
- Hubei Key Laboratory of Exercise Training and Monitoring, Hubei Provincial Collaborative Innovation Center for Exercise and Health Promotion, College of Health Science, Wuhan Sports University, Wuhan, China
| | - Ning Chen
- Hubei Key Laboratory of Exercise Training and Monitoring, Hubei Provincial Collaborative Innovation Center for Exercise and Health Promotion, College of Health Science, Wuhan Sports University, Wuhan, China
| |
Collapse
|
34
|
Holeček M. Beta-hydroxy-beta-methylbutyrate supplementation and skeletal muscle in healthy and muscle-wasting conditions. J Cachexia Sarcopenia Muscle 2017; 8:529-541. [PMID: 28493406 PMCID: PMC5566641 DOI: 10.1002/jcsm.12208] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/08/2017] [Accepted: 03/20/2017] [Indexed: 12/21/2022] Open
Abstract
Beta-hydroxy-beta-methylbutyrate (HMB) is a metabolite of the essential amino acid leucine that has been reported to have anabolic effects on protein metabolism. The aims of this article were to summarize the results of studies of the effects of HMB on skeletal muscle and to examine the evidence for the rationale to use HMB as a nutritional supplement to exert beneficial effects on muscle mass and function in various conditions of health and disease. The data presented here indicate that the beneficial effects of HMB have been well characterized in strength-power and endurance exercise. HMB attenuates exercise-induced muscle damage and enhances muscle hypertrophy and strength, aerobic performance, resistance to fatigue, and regenerative capacity. HMB is particularly effective in untrained individuals who are exposed to strenuous exercise and in trained individuals who are exposed to periods of high physical stress. The low effectiveness of HMB in strength-trained athletes could be due to the suppression of the proteolysis that is induced by the adaptation to training, which may blunt the effects of HMB. Studies performed with older people have demonstrated that HMB can attenuate the development of sarcopenia in elderly subjects and that the optimal effects of HMB on muscle growth and strength occur when it is combined with exercise. Studies performed under in vitro conditions and in various animal models suggest that HMB may be effective in treatment of muscle wasting in various forms of cachexia. However, there are few clinical reports of the effects of HMB on muscle wasting in cachexia; in addition, most of these studies evaluated the therapeutic potential of combinations of various agents. Therefore, it has not been possible to determine whether HMB was effective or if there was a synergistic effect. Although most of the endogenous HMB is produced in the liver, there are no reports regarding the levels and the effects of HMB supplementation in subjects with liver disease. Several studies have suggested that anabolic effects of HMB supplementation on skeletal muscle do not occur in healthy, non-exercising subjects. It is concluded that (i) HMB may be applied to enhance increases in the mass and strength of skeletal muscles in subjects who exercise and in the elderly and (ii) studies examining the effects of HMB administered alone are needed to obtain conclusions regarding the specific effectiveness in attenuating muscle wasting in various muscle-wasting disorders.
Collapse
Affiliation(s)
- Milan Holeček
- Department of Physiology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| |
Collapse
|
35
|
Standley RA, Distefano G, Pereira SL, Tian M, Kelly OJ, Coen PM, Deutz NEP, Wolfe RR, Goodpaster BH. Effects of β-hydroxy-β-methylbutyrate on skeletal muscle mitochondrial content and dynamics, and lipids after 10 days of bed rest in older adults. J Appl Physiol (1985) 2017; 123:1092-1100. [PMID: 28705993 DOI: 10.1152/japplphysiol.00192.2017] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 06/29/2017] [Accepted: 07/12/2017] [Indexed: 01/08/2023] Open
Abstract
Loss of muscle mass during periods of disuse likely has negative health consequences for older adults. We have previously shown that β-hydroxy-β-methylbutyrate (HMB) supplementation during 10 days of strict bed rest (BR) attenuates the loss of lean mass in older adults. To elucidate potential molecular mechanisms of HMB effects on muscle during BR and resistance training rehabilitation (RT), we examined mediators of skeletal muscle mitochondrial dynamics, autophagy and atrophy, and intramyocellular lipids. Nineteen older adults (60-76 yr) completed 10 days BR followed by 8-wk RT rehabilitation. Subjects were randomized to either HMB (3 g/day HMB; n = 11) or control (CON; n = 8) groups. Skeletal muscle cross-sectional area (CSA) was determined by histology from percutaneous vastus lateralis biopsies. We measured protein markers of mitochondrial content [oxidative phosphorylation (OXPHOS)], fusion and fission (MFN2, OPA1, FIS1, and DRP1), autophagy (Beclin1, LC3B, and BNIP3), and atrophy [poly-ubiquinated proteins (poly-ub)] by Western blot. Fatty acid composition of several lipid classes in skeletal muscle was measured by infusion-MS analysis. Poly-ub proteins and OXPHOS complex I increased in both groups following BR (P < 0.05, main effect for time), and muscle triglyceride content tended to increase following BR in the HMB group (P = 0.055). RT rehabilitation increased OXPHOS complex II protein (P < 0.05), and total OXPHOS content tended (P = 0.0504) to be higher in HMB group. In addition, higher levels of DRP1 and MFN2 were maintained in the HMB group after RT (P < 0.05). BNIP3 and poly-ub proteins were significantly reduced following rehabilitation in both groups (P < 0.05). Collectively, these data suggest that HMB influences mitochondrial dynamics and lipid metabolism during disuse atrophy and rehabilitation.NEW & NOTEWORTHY Mitochondrial content and dynamics remained unchanged over 10 days of BR in older adults. HMB stimulated intramuscular lipid storage as triacylglycerol following 10 days of bed rest (BR) and maintained higher mitochondrial OXPHOS content and dynamics during the 8-wk resistance exercise rehabilitation program.
Collapse
Affiliation(s)
- Robert A Standley
- Translational Research Institute for Metabolism and Diabetes-Florida Hospital, Orlando, Florida
| | - Giovanna Distefano
- Translational Research Institute for Metabolism and Diabetes-Florida Hospital, Orlando, Florida
| | | | - Min Tian
- Abbott Nutrition, Research, and Development, Columbus, Ohio
| | - Owen J Kelly
- Abbott Nutrition, Research, and Development, Columbus, Ohio
| | - Paul M Coen
- Translational Research Institute for Metabolism and Diabetes-Florida Hospital, Orlando, Florida
| | - Nicolaas E P Deutz
- Center for Translational Research in Aging and Longevity, Department of Health and Kinesiology, Texas A&M University, College Station, Teaxas
| | - Robert R Wolfe
- Center for Translational Research in Aging and Longevity, Donald W. Reynolds Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, Arkanas; and
| | - Bret H Goodpaster
- Translational Research Institute for Metabolism and Diabetes-Florida Hospital, Orlando, Florida;
| |
Collapse
|
36
|
Gepner Y, Hoffman JR, Shemesh E, Stout JR, Church DD, Varanoske AN, Zelicha H, Shelef I, Chen Y, Frankel H, Ostfeld I. Combined effect of Bacillus coagulans GBI-30, 6086 and HMB supplementation on muscle integrity and cytokine response during intense military training. J Appl Physiol (1985) 2017; 123:11-18. [DOI: 10.1152/japplphysiol.01116.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 04/03/2017] [Accepted: 04/06/2017] [Indexed: 12/16/2022] Open
Abstract
The purpose of this study was to compare the coadministration of the probiotic Bacillus coagulans GBI-30, 6086 (BC30) with β-hydroxy-β-methylbutyrate (HMB) calcium (CaHMB) to CaHMB alone on inflammatory response and muscle integrity during 40 days of intense military training. Soldiers were randomly assigned to one of two groups: CaHMB with BC30 (CaHMBBC30; n = 9) or CaHMB with placebo (CaHMBPL, n = 9). A third group of participants served as a control (CTL; n = 8). During the first 28 days soldiers were garrisoned on base and participated in the same training tasks. During the final 2 wk soldiers navigated 25–30 km per night in difficult terrain carrying ~35 kg of equipment. All assessments (blood draws and diffusion tensor imaging to assess muscle integrity) were conducted before and ~12 h after final supplement consumption. Analysis of covariance was used to analyze all blood and muscle measures. Significant attenuations were noted in IL-1β, IL-2, IL-6, CX3CL1, and TNF-α for both CaHMBBC30 and CaHMBPL compared with CTL. Plasma IL-10 concentrations were significantly attenuated for CaHMBBC30 compared with CTL only. A significant decrease in apparent diffusion coefficients was also observed for CaHMBBC30 compared with CaHMBPL. Results provide further evidence that HMB supplementation may attenuate the inflammatory response to intense training and that the combination of the probiotic BC30 with CaHMB may be more beneficial than CaHMB alone in maintaining muscle integrity during intense military training. NEW & NOTEWORTHY β-Hydroxy-β-methylbutyrate (HMB) in its free acid form was reported to attenuate inflammation and maintain muscle integrity during military training. However, this formulation was difficult to maintain in the field. In this investigation, soldiers ingested HMB calcium (CaHMB) with Bacillus coagulans (BC30) or CaHMB alone during 40 days of training. Results indicated that CaHMB attenuated the inflammatory response and that BC30 combined with CaHMB may be more beneficial than CaHMB alone in maintaining muscle integrity during intense military training.
Collapse
Affiliation(s)
- Yftach Gepner
- Sport and Exercise Science, Institute of Exercise Physiology and Wellness, University of Central Florida, Orlando, Florida
| | - Jay R. Hoffman
- Sport and Exercise Science, Institute of Exercise Physiology and Wellness, University of Central Florida, Orlando, Florida
| | - Elad Shemesh
- Israel Defense Force Medical Corps, Tel Hashomer, Israel
| | - Jeffrey R. Stout
- Sport and Exercise Science, Institute of Exercise Physiology and Wellness, University of Central Florida, Orlando, Florida
| | - David D. Church
- Sport and Exercise Science, Institute of Exercise Physiology and Wellness, University of Central Florida, Orlando, Florida
| | - Alyssa N. Varanoske
- Sport and Exercise Science, Institute of Exercise Physiology and Wellness, University of Central Florida, Orlando, Florida
| | - Hila Zelicha
- Department of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel; and
| | | | - Yacov Chen
- Israel Defense Force Medical Corps, Tel Hashomer, Israel
| | - Hagai Frankel
- Israel Defense Force Medical Corps, Tel Hashomer, Israel
| | - Ishay Ostfeld
- Israel Defense Force Medical Corps, Tel Hashomer, Israel
| |
Collapse
|
37
|
Li F, Li X, Peng X, Sun L, Jia S, Wang P, Ma S, Zhao H, Yu Q, Huo H. Ginsenoside Rg1 prevents starvation-induced muscle protein degradation via regulation of AKT/mTOR/FoxO signaling in C2C12 myotubes. Exp Ther Med 2017; 14:1241-1247. [PMID: 28781621 DOI: 10.3892/etm.2017.4615] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/19/2016] [Indexed: 01/07/2023] Open
Abstract
Skeletal muscle atrophy is often caused by catabolic conditions including fasting, disuse, aging and chronic diseases, such as chronic obstructive pulmonary disease. Atrophy occurs when the protein degradation rate exceeds the rate of protein synthesis. Therefore, maintaining a balance between the synthesis and degradation of protein in muscle cells is a major way to prevent skeletal muscle atrophy. Ginsenoside Rg1 (Rg1) is a primary active ingredient in Panax ginseng, which is considered to be one of the most valuable herbs in traditional Chinese medicine. In the current study, Rg1 was observed to inhibit the expression of MuRF-1 and atrogin-1 in C2C12 muscle cells in a starvation model. Rg1 also activated the phosphorylation of mammalian target of rapamycin (mTOR), protein kinase B (AKT), and forkhead transcription factor O, subtypes 1 and 3a. This phosphorylation was inhibited by LY294002, a phosphatidylinositol 3-kinase inhibitor. These data suggest that Rg1 may participate in the regulation of the balance between protein synthesis and degradation, and that the function of Rg1 is associated with the AKT/mTOR/FoxO signaling pathway.
Collapse
Affiliation(s)
- Fengyu Li
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Xiaoxue Li
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Xuewei Peng
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Lili Sun
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Shengnan Jia
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Ping Wang
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Shuang Ma
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Hongyan Zhao
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Qingmiao Yu
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Hongliang Huo
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| |
Collapse
|
38
|
Go GY, Lee SJ, Jo A, Lee J, Seo DW, Kang JS, Kim SK, Kim SN, Kim YK, Bae GU. Ginsenoside Rg1 from Panax ginseng enhances myoblast differentiation and myotube growth. J Ginseng Res 2017; 41:608-614. [PMID: 29021711 PMCID: PMC5628345 DOI: 10.1016/j.jgr.2017.05.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 05/23/2017] [Accepted: 05/25/2017] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Ginsenoside Rg1 belongs to protopanaxatriol-type ginsenosides and has diverse pharmacological activities. In this report, we investigated whether Rg1 could upregulate muscular stem cell differentiation and muscle growth. METHODS C2C12 myoblasts, MyoD-transfected 10T1/2 embryonic fibroblasts, and HEK293T cells were treated with Rg1 and differentiated for 2 d, subjected to immunoblotting, immunocytochemistry, or immunoprecipitation. RESULTS Rg1 activated promyogenic kinases, p38MAPK (mitogen-activated protein kinase) and Akt signaling, that in turn promote the heterodimerization with MyoD and E proteins, resulting in enhancing myogenic differentiation. Through the activation of Akt/mammalian target of rapamycin pathway, Rg1 induced myotube growth and prevented dexamethasone-induced myotube atrophy. Furthermore, Rg1 increased MyoD-dependent myogenic conversion of fibroblast. CONCLUSION Rg1 upregulates promyogenic kinases, especially Akt, resulting in improvement of myoblast differentiation and myotube growth.
Collapse
Affiliation(s)
- Ga-Yeon Go
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Sang-Jin Lee
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Ayoung Jo
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Jaecheol Lee
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, CA, USA
| | - Dong-Wan Seo
- College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon, Republic of Korea
| | - Si-Kwan Kim
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju, Republic of Korea
| | - Su-Nam Kim
- Natural Products Research Institute, Korea Institute of Science and Technology, Gangneung, Republic of Korea
| | - Yong Kee Kim
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
- Corresponding author. Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Cheongpa-ro 47-gil 100, Yongsan-Gu, Seoul 04310, Republic of Korea.Research Center for Cell Fate ControlCollege of PharmacySookmyung Women's UniversityCheongpa-ro 47-gil 100, Yongsan-GuSeoul04310Republic of Korea
| | - Gyu-Un Bae
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
- Corresponding author. Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Cheongpa-ro 47-gil 100, Yongsan-Gu, Seoul 04310, Republic of Korea.Research Center for Cell Fate ControlCollege of PharmacySookmyung Women's UniversityCheongpa-ro 47-gil 100, Yongsan-GuSeoul04310Republic of Korea
| |
Collapse
|
39
|
Inoue A, Cheng XW, Huang Z, Hu L, Kikuchi R, Jiang H, Piao L, Sasaki T, Itakura K, Wu H, Zhao G, Lei Y, Yang G, Zhu E, Li X, Sato K, Koike T, Kuzuya M. Exercise restores muscle stem cell mobilization, regenerative capacity and muscle metabolic alterations via adiponectin/AdipoR1 activation in SAMP10 mice. J Cachexia Sarcopenia Muscle 2017; 8:370-385. [PMID: 27897419 PMCID: PMC5476856 DOI: 10.1002/jcsm.12166] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 10/08/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Exercise train (ET) stimulates muscle response in pathological conditions, including aging. The molecular mechanisms by which exercise improves impaired adiponectin/adiponectin receptor 1 (AdipoR1)-related muscle actions associated with aging are poorly understood. Here we observed that in a senescence-accelerated mouse prone 10 (SAMP10) model, long-term ET modulated muscle-regenerative actions. METHODS 25-week-old male SAMP10 mice were randomly assigned to the control and the ET (45 min/time, 3/week) groups for 4 months. Mice that were maintained in a sedentary condition served controls. RESULTS ET ameliorated aging-related muscle changes in microstructure, mitochondria, and performance. The amounts of proteins or mRNAs for p-AMPKα, p-Akt, p-ERK1/2, p-mTOR, Bcl-XL, p-FoxO3, peroxisome proliferators-activated receptor-γ coactivator, adiponectin receptor1 (adpoR1), and cytochrome c oxidase-IV, and the numbers of CD34+ /integrin-α7+ muscle stem cells (MuSCs) and proliferating cells in the muscles and bone-marrow were enhanced by ET, whereas the levels of p-GSK-3α and gp91phox proteins and apoptotic cells were reduced by ET. The ET also resulted in increased levels of plasma adiponectin and the numbers of bone-marrow (BM)-derived circulating CD34+ /integrin-α7+ MuSCs and their functions. Integrin-α7+ MuSCs of exercised mice had improved changes of those beneficial molecules. These ET-mediated aged muscle benefits were diminished by adiponectin and AdipoR1 blocking as well as AMPK inhibition. Finally, recombinant mouse adiponectin enhanced AMPK and mTOR phosphorylations in BM-derived integrin-α7+ cells. CONCLUSIONS These findings suggest that ET can improve aging-related impairments of BM-derived MuSC regenerative capacity and muscle metabolic alterations via an AMPK-dependent mechanism that is mediated by an adiponectin/AdipoR1 axis in SAMP10 mice.
Collapse
Affiliation(s)
- Aiko Inoue
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan
| | - Xian Wu Cheng
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan.,Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan.,Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, 133000, Jilin PR., China.,Department of Cardiovascular of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Zhe Huang
- Department of Neurology, University of Occupational and Environmental Health, Kitakyushu, 807-8555, Fukuoka, Japan
| | - Lina Hu
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan
| | - Ryosuke Kikuchi
- Department of Medical Technique, Nagoya University Hospital, Nagoya, 466-8550, Aichiken, Japan
| | - Haiying Jiang
- Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, 133000, Jilin PR., China
| | - Limei Piao
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan.,Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, 133000, Jilin PR., China
| | - Takeshi Sasaki
- Department of Anatomy and Neuroscience, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Shizuokaken, Japan
| | - Kohji Itakura
- Division for Medical Research Engineering, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan
| | - Hongxian Wu
- Department of Sport Medicine, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan
| | - Guangxian Zhao
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, 133000, Jilin PR., China
| | - Yanna Lei
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, 133000, Jilin PR., China
| | - Guang Yang
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, 133000, Jilin PR., China
| | - Enbo Zhu
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, 133000, Jilin PR., China
| | - Xiang Li
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, 133000, Jilin PR., China
| | - Kohji Sato
- Department of Anatomy and Neuroscience, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Shizuokaken, Japan
| | - Teruhiko Koike
- Department of Sport Medicine, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan
| | - Masafumi Kuzuya
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan.,Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan
| |
Collapse
|
40
|
Cramer JT, Cruz-Jentoft AJ, Landi F, Hickson M, Zamboni M, Pereira SL, Hustead DS, Mustad VA. Impacts of High-Protein Oral Nutritional Supplements Among Malnourished Men and Women with Sarcopenia: A Multicenter, Randomized, Double-Blinded, Controlled Trial. J Am Med Dir Assoc 2016; 17:1044-1055. [DOI: 10.1016/j.jamda.2016.08.009] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 08/22/2016] [Indexed: 12/24/2022]
|
41
|
Schnuck JK, Johnson MA, Gould LM, Gannon NP, Vaughan RA. Acute β-Hydroxy-β-Methyl Butyrate Suppresses Regulators of Mitochondrial Biogenesis and Lipid Oxidation While Increasing Lipid Content in Myotubes. Lipids 2016; 51:1127-1136. [PMID: 27600148 DOI: 10.1007/s11745-016-4193-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 08/25/2016] [Indexed: 01/30/2023]
Abstract
Leucine modulates synthetic and degradative pathways in muscle, possibly providing metabolic benefits for both athletes and diseased populations. Leucine has become popular among athletes for improving performance and body composition, however little is known about the metabolic effects of the commonly consumed leucine-derived metabolite β-hydroxy-β-methyl butyrate (HMB). Our work measured the effects of HMB on metabolic protein expression, mitochondrial content and metabolism, as well as lipid content in skeletal muscle cells. Specifically, cultured C2C12 myotubes were treated with either a control or HMB ranging from 6.25 to 25 μM for 24 h and mRNA and/or protein expression, oxygen consumption, glucose uptake, and lipid content were measured. Contrary to leucine's stimulatory effect on metabolism, HMB-treated cells exhibited significantly reduced regulators of lipid oxidation including peroxisome proliferator-activated receptor alpha (PPARα) and PPARβ/δ, as well as downstream target carnitine palmitoyl transferase, without alterations in glucose or palmitate oxidation. Furthermore, HMB significantly inhibited activation of the master regulator of energetics, AMP-activated protein kinase. As a result, HMB-treated cells also displayed reduced total mitochondrial content compared with true control or cells equivocally treated with leucine. Additionally, HMB treatment amplified markers of lipid biosynthesis (PPARγ and fatty acid synthase) as well as consistently promoted elevated total lipid content versus control cells. Collectively, our results demonstrate that HMB did not improve mitochondrial metabolism or content, and may promote elevated cellular lipid content possibly through heightened PPARγ expression. These observations suggest that HMB may be most beneficial for populations interested in stimulating anabolic cellular processes.
Collapse
Affiliation(s)
- Jamie K Schnuck
- Department of Exercise Science, High Point University, One University Parkway, High Point, NC, 27268, USA
| | - Michele A Johnson
- Department of Exercise Science, High Point University, One University Parkway, High Point, NC, 27268, USA
| | - Lacey M Gould
- Department of Exercise Science, High Point University, One University Parkway, High Point, NC, 27268, USA
| | - Nicholas P Gannon
- School of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Roger A Vaughan
- Department of Exercise Science, High Point University, One University Parkway, High Point, NC, 27268, USA.
| |
Collapse
|
42
|
Kim DH, Park MH, Chung KW, Kim MJ, Park D, Lee B, Lee EK, Choi YJ, Kim ND, Yu BP, Chung HY. Suppression of FoxO6 by lipopolysaccharide in aged rat liver. Oncotarget 2016; 6:34143-57. [PMID: 26506521 PMCID: PMC4741442 DOI: 10.18632/oncotarget.6219] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 09/15/2015] [Indexed: 12/19/2022] Open
Abstract
The beneficial role of FoxO during aging has been proposed for its promotion of resistance to oxidative stress and inhibition of pro-inflammatory mediators. On the other hand, NF-κB is a pro-inflammatory transcription factor which is a key mediator of inflammatory cytokine generation. However, the correlation between FoxO6 and NF-κB during aging has not fully been explored. The main purpose of the present study was to elucidate mechanisms underlying the protective role of FoxO6 in the maintenance of cellular homeostasis under potent pro-inflammatory conditions induced by LPS. Initial experimentation revealed that reduced FoxO6 activity during aging was caused by its phosphorylation, which suppressed its transcriptional activity in aged livers. Transfection with FoxO6-wt virus and FoxO6-siRNA in HepG2 cells revealed that FoxO6 phosphorylation by LPS leads to NF-κB activation via Akt and Pak1 pathways. Furthermore, Pak1 activity was increased in a phosphatidylinositol 3-kinase independent manner, and LPS-induced FoxO6 phosphorylation and FoxO6 inactivation were Pak1-dependent in nuclear fractions of cells. Further revealed Pak1 phosphorylation by LPS permitted interaction between FoxO6 and Akt. Current study suggests FoxO6 phosphorylation facilitates the nuclear translocation of NF-κB via Akt and Pak1 pathways induced by LPS in aged rats.
Collapse
Affiliation(s)
- Dae Hyun Kim
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Gumjung-gu, Busan, Korea
| | - Min Hi Park
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Gumjung-gu, Busan, Korea
| | - Ki Wung Chung
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Gumjung-gu, Busan, Korea
| | - Min Jo Kim
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Gumjung-gu, Busan, Korea
| | - Daeui Park
- In silico Toxicology Research Center, Korea Institute of Toxicology, Daejeon, Korea
| | - Bonggi Lee
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Gumjung-gu, Busan, Korea
| | - Eun Kyeong Lee
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Gumjung-gu, Busan, Korea
| | - Yeon Ja Choi
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Gumjung-gu, Busan, Korea
| | - Nam Deuk Kim
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Gumjung-gu, Busan, Korea
| | - Byung Pal Yu
- Department of Physiology, The University of Texas Health Science Center at San Antonio, TX, USA
| | - Hae Young Chung
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Gumjung-gu, Busan, Korea
| |
Collapse
|
43
|
β-Hydroxy-β-methylbutyrate attenuates cytokine response during sustained military training. Nutr Res 2016; 36:553-63. [DOI: 10.1016/j.nutres.2016.02.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 02/16/2016] [Accepted: 02/18/2016] [Indexed: 11/19/2022]
|
44
|
Brioche T, Pagano AF, Py G, Chopard A. Muscle wasting and aging: Experimental models, fatty infiltrations, and prevention. Mol Aspects Med 2016; 50:56-87. [PMID: 27106402 DOI: 10.1016/j.mam.2016.04.006] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 04/13/2016] [Accepted: 04/13/2016] [Indexed: 12/21/2022]
Abstract
Identification of cost-effective interventions to maintain muscle mass, muscle strength, and physical performance during muscle wasting and aging is an important public health challenge. It requires understanding of the cellular and molecular mechanisms involved. Muscle-deconditioning processes have been deciphered by means of several experimental models, bringing together the opportunities to devise comprehensive analysis of muscle wasting. Studies have increasingly recognized the importance of fatty infiltrations or intermuscular adipose tissue for the age-mediated loss of skeletal-muscle function and emphasized that this new important factor is closely linked to inactivity. The present review aims to address three main points. We first mainly focus on available experimental models involving cell, animal, or human experiments on muscle wasting. We next point out the role of intermuscular adipose tissue in muscle wasting and aging and try to highlight new findings concerning aging and muscle-resident mesenchymal stem cells called fibro/adipogenic progenitors by linking some cellular players implicated in both FAP fate modulation and advancing age. In the last part, we review the main data on the efficiency and molecular and cellular mechanisms by which exercise, replacement hormone therapies, and β-hydroxy-β-methylbutyrate prevent muscle wasting and sarcopenia. Finally, we will discuss a potential therapeutic target of sarcopenia: glucose 6-phosphate dehydrogenase.
Collapse
Affiliation(s)
- Thomas Brioche
- Université de Montpellier, INRA, UMR 866 Dynamique Musculaire et Métabolisme, Montpellier F-34060, France.
| | - Allan F Pagano
- Université de Montpellier, INRA, UMR 866 Dynamique Musculaire et Métabolisme, Montpellier F-34060, France
| | - Guillaume Py
- Université de Montpellier, INRA, UMR 866 Dynamique Musculaire et Métabolisme, Montpellier F-34060, France
| | - Angèle Chopard
- Université de Montpellier, INRA, UMR 866 Dynamique Musculaire et Métabolisme, Montpellier F-34060, France
| |
Collapse
|
45
|
Girón MD, Vílchez JD, Salto R, Manzano M, Sevillano N, Campos N, Argilés JM, Rueda R, López-Pedrosa JM. Conversion of leucine to β-hydroxy-β-methylbutyrate by α-keto isocaproate dioxygenase is required for a potent stimulation of protein synthesis in L6 rat myotubes. J Cachexia Sarcopenia Muscle 2016; 7:68-78. [PMID: 27065075 PMCID: PMC4799859 DOI: 10.1002/jcsm.12032] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 03/03/2015] [Accepted: 03/10/2015] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND L-Leu and its metabolite β-hydroxy-β-methylbutyrate (HMB) stimulate muscle protein synthesis enhancing the phosphorylation of proteins that regulate anabolic signalling pathways. Alterations in these pathways are observed in many catabolic diseases, and HMB and L-Leu have proven their anabolic effects in in vivo and in vitro models. The aim of this study was to compare the anabolic effects of L-Leu and HMB in myotubes grown in the absence of any catabolic stimuli. METHODS Studies were conducted in vitro using rat L6 myotubes under normal growth conditions (non-involving L-Leu-deprived conditions). Protein synthesis and mechanistic target of rapamycin signalling pathway were determined. RESULTS Only HMB was able to increase protein synthesis through a mechanism that involves the phosphorylation of the mechanistic target of rapamycin as well as its downstream elements, pS6 kinase, 4E binding protein-1, and eIF4E. HMB was significantly more effective than L-Leu in promoting these effects through an activation of protein kinase B/Akt. Because the conversion of L-Leu to HMB is limited in muscle, L6 cells were transfected with a plasmid that codes for α-keto isocaproate dioxygenase, the key enzyme involved in the catabolic conversion of α-keto isocaproate into HMB. In these transfected cells, L-Leu was able to promote protein synthesis and mechanistic target of rapamycin regulated pathway activation equally to HMB. Additionally, these effects of leucine were reverted to a normal state by mesotrione, a specific inhibitor of α-keto isocaproate dioxygenase. CONCLUSION Our results suggest that HMB is an active L-Leu metabolite able to maximize protein synthesis in skeletal muscle under conditions, in which no amino acid deprivation occurred. It may be proposed that supplementation with HMB may be very useful to stimulate protein synthesis in wasting conditions associated with chronic diseases, such as cancer or chronic heart failure.
Collapse
Affiliation(s)
- María D Girón
- Department of Biochemistry and Molecular Biology II School of Pharmacy, University of Granada Granada Spain
| | - José D Vílchez
- Department of Biochemistry and Molecular Biology II School of Pharmacy, University of Granada Granada Spain
| | - Rafael Salto
- Department of Biochemistry and Molecular Biology II School of Pharmacy, University of Granada Granada Spain
| | | | - Natalia Sevillano
- Department of Biochemistry and Molecular Biology II School of Pharmacy, University of Granada Granada Spain
| | | | - Josep M Argilés
- Cancer Research Group, Department de Bioquimica I Biologia Molecular Facultat de Biologia, Universitat de Barcelona Barcelona Spain
| | | | | |
Collapse
|
46
|
Bond P. Regulation of mTORC1 by growth factors, energy status, amino acids and mechanical stimuli at a glance. J Int Soc Sports Nutr 2016; 13:8. [PMID: 26937223 PMCID: PMC4774173 DOI: 10.1186/s12970-016-0118-y] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 02/18/2016] [Indexed: 12/05/2022] Open
Abstract
The mechanistic/mammalian target of rapamycin complex 1 (mTORC1) plays a pivotal role in the regulation of skeletal muscle protein synthesis. Activation of the complex leads to phosphorylation of two important sets of substrates, namely eIF4E binding proteins and ribosomal S6 kinases. Phosphorylation of these substrates then leads to an increase in protein synthesis, mainly by enhancing translation initiation. mTORC1 activity is regulated by several inputs, such as growth factors, energy status, amino acids and mechanical stimuli. Research in this field is rapidly evolving and unraveling how these inputs regulate the complex. Therefore this review attempts to provide a brief and up-to-date narrative on the regulation of this marvelous protein complex. Additionally, some sports supplements which have been shown to regulate mTORC1 activity are discussed.
Collapse
Affiliation(s)
- Peter Bond
- PeterBond.nl, Waterhoenlaan 25, Zeist, Netherlands
| |
Collapse
|
47
|
Activated Integrin-Linked Kinase Negatively Regulates Muscle Cell Enhancement Factor 2C in C2C12 Cells. BIOMED RESEARCH INTERNATIONAL 2016; 2015:748470. [PMID: 26788505 PMCID: PMC4695646 DOI: 10.1155/2015/748470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 11/25/2015] [Accepted: 11/29/2015] [Indexed: 01/28/2023]
Abstract
Our previous study reported that muscle cell enhancement factor 2C (MEF2C) was fully activated after inhibition of the phosphorylation activity of integrin-linked kinase (ILK) in the skeletal muscle cells of goats. It enhanced the binding of promoter or enhancer of transcription factor related to proliferation of muscle cells and then regulated the expression of these genes. In the present investigation, we explored whether ILK activation depended on PI3K to regulate the phosphorylation and transcriptional activity of MEF2C during C2C12 cell proliferation. We inhibited PI3K activity in C2C12 with LY294002 and then found that ILK phosphorylation levels and MEF2C phosphorylation were decreased and that MCK mRNA expression was suppressed significantly. After inhibiting ILK phosphorylation activity with Cpd22 and ILK-shRNA, we found MEF2C phosphorylation activity and MCK mRNA expression were increased extremely significantly. In the presence of Cpd22, PI3K activity inhibition increased MEF2C phosphorylation and MCK mRNA expression indistinctively. We conclude that ILK negatively and independently of PI3K regulated MEF2C phosphorylation activity and MCK mRNA expression in C2C12 cells. The results provide new ideas for the study of classical signaling pathway of PI3K-ILK-related proteins and transcription factors.
Collapse
|
48
|
Girón MD, Vílchez JD, Shreeram S, Salto R, Manzano M, Cabrera E, Campos N, Edens NK, Rueda R, López-Pedrosa JM. β-Hydroxy-β-methylbutyrate (HMB) normalizes dexamethasone-induced autophagy-lysosomal pathway in skeletal muscle. PLoS One 2015; 10:e0117520. [PMID: 25658432 PMCID: PMC4319954 DOI: 10.1371/journal.pone.0117520] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 12/27/2014] [Indexed: 01/07/2023] Open
Abstract
Dexamethasone-induced muscle atrophy is due to an increase in protein breakdown and a decrease in protein synthesis, associated with an over-stimulation of the autophagy-lysosomal pathway. These effects are mediated by alterations in IGF-1 and PI3K/Akt signaling. In this study, we have investigated the effects of β-Hydroxy-β-methylbutyrate (HMB) on the regulation of autophagy and proteosomal systems. Rats were treated during 21 days with dexamethasone as a model of muscle atrophy. Co-administration of HMB attenuated the effects promoted by dexamethasone. HMB ameliorated the loss in body weight, lean mass and the reduction of the muscle fiber cross-sectional area (shrinkage) in gastrocnemius muscle. Consequently, HMB produced an improvement in muscle strength in the dexamethasone-treated rats. To elucidate the molecular mechanisms responsible for these effects, rat L6 myotubes were used. In these cells, HMB significantly attenuated lysosomal proteolysis induced by dexamethasone by normalizing the changes observed in autophagosome formation, LC3 II, p62 and Bnip3 expression after dexamethasone treatment. HMB effects were mediated by an increase in FoxO3a phosphorylation and concomitant decrease in FoxO transcriptional activity. The HMB effect was due to the restoration of Akt signaling diminished by dexamethasone treatment. Moreover, HMB was also involved in the regulation of the activity of ubiquitin and expression of MurF1 and Atrogin-1, components of the proteasome system that are activated or up-regulated by dexamethasone. In conclusion, in vivo and in vitro studies suggest that HMB exerts protective effects against dexamethasone-induced muscle atrophy by normalizing the Akt/FoxO axis that controls autophagy and ubiquitin proteolysis.
Collapse
Affiliation(s)
- María D. Girón
- Department of Biochemistry and Molecular Biology II, University of Granada, Granada, Spain
- * E-mail:
| | - Jose D. Vílchez
- Department of Biochemistry and Molecular Biology II, University of Granada, Granada, Spain
| | | | - Rafael Salto
- Department of Biochemistry and Molecular Biology II, University of Granada, Granada, Spain
| | | | - Elena Cabrera
- Department of Biochemistry and Molecular Biology II, University of Granada, Granada, Spain
| | | | - Neile K. Edens
- Abbott Nutrition R&D, Columbus, Ohio, United States of America
| | | | | |
Collapse
|