1
|
Von Ruff ZD, Miller MJ, Moro T, Reidy PT, Ebert SM, Volpi E, Adams CM, Rasmussen BB. Resistance exercise training in older men reduces ATF4-activated and senescence-associated mRNAs in skeletal muscle. GeroScience 2025:10.1007/s11357-025-01564-2. [PMID: 40011348 DOI: 10.1007/s11357-025-01564-2] [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: 10/22/2024] [Accepted: 02/10/2025] [Indexed: 02/28/2025] Open
Abstract
Sarcopenia increases the risk of frailty, morbidity, and mortality in older adults. Resistance exercise training improves muscle size and function; however, the response to exercise training is variable in older adults. The objective of our study was to determine both the age-independent and age-dependent changes to the transcriptome following progressive resistance exercise training. Skeletal muscle biopsies were obtained before and after 12 weeks of resistance exercise training in 8 young (24 ± 3.3 years) and 10 older (72 ± 4.9 years) men. RNA was extracted from each biopsy and prepared for analysis via RNA sequencing. We performed differential mRNA expression, gene ontology, and gene set enrichment analyses. We report that when comparing post-training vs pre-training 226 mRNAs and 959 mRNAs were differentially expressed in the skeletal muscle of young and older men, respectively. Additionally, 94 mRNAs increased, and 17 mRNAs decreased in both young and old, indicating limited overlap in response to resistance exercise training. Furthermore, the differential gene expression was larger in older skeletal muscle. Finally, we report three novel findings: 1) resistance exercise training decreased the abundance of ATF4-activated and senescence-associated skeletal muscle mRNAs in older men; 2) resistance exercise-induced increases in lean mass correlate with increased mRNAs encoding mitochondrial proteins; and 3) increases in muscle strength following resistance exercise positively correlate with increased mRNAs involved in translation, rRNA processing, and polyamine metabolism. We conclude that resistance exercise training elicits a differential gene expression response in young and old skeletal muscle, including reduced ATF-4 activated and senescence-associated gene expression.
Collapse
Affiliation(s)
| | - Matthew J Miller
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
- University of Iowa, Iowa City, IA, USA
| | - Tatiana Moro
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Paul T Reidy
- Department of Kinesiology, Nutrition, and Health, Miami University, Oxford, OH, USA
| | - Scott M Ebert
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
| | - Elena Volpi
- Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7756, San Antonio, TX, 78229, USA
| | - Christopher M Adams
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
| | - Blake B Rasmussen
- Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7756, San Antonio, TX, 78229, USA.
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7756, San Antonio, TX, 78229, USA.
| |
Collapse
|
2
|
Briand M, Raffin J, Gonzalez-Bautista E, Ritz P, Abellan Van Kan G, Pillard F, Faruch-Bilfeld M, Guyonnet S, Dray C, Vellas B, de Souto Barreto P, Rolland Y. Body composition and aging: cross-sectional results from the INSPIRE study in people 20 to 93 years old. GeroScience 2025; 47:863-875. [PMID: 39028455 PMCID: PMC11872965 DOI: 10.1007/s11357-024-01245-6] [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: 01/19/2024] [Accepted: 06/06/2024] [Indexed: 07/20/2024] Open
Abstract
Aging is characterized by several major changes, including altered body composition, which is associated with numerous negative clinical consequences such as sarcopenia, osteoporosis, and frailty. The study is to evaluate body composition parameters depending on age and sex in a population ranging from the young adult to the very old, and to identify break points in the association between body composition and age. In this cross-sectional study, we included the enrolment population of the French INSPIRE-T prospective cohort, accounting for 915 subjects (62% female). Age ranged from 20 to 93 years, median age (years) was 63 (IQR 27). Body composition (lean mass, fat mass, and bone mineral content) was assessed with dual-X-ray absorptiometry (DXA). Different break points in the relationship between age and body composition variables in males and females were identified using a segmented regression analysis adjusted on physical activity, nutritional status, educational level, and comorbidities. Lean mass decreased from the age of 55 years for males (CI 95% 44-66) and 31 years for females (CI 95% 23-39). For fat mass, we observed a trend towards an increase with age for males. For females, we observed an increase with age up to age 75 (CI 95% 62-86), followed by a decreasing trend. In this study, we described the relationship between body composition and age as a function of sex, establishing a foundation for further studies on predictive biomarkers of age-related body composition alteration.
Collapse
Affiliation(s)
- Marguerite Briand
- IHU HealthAge, Toulouse, France.
- Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France.
- Institut RESTORE, UMR 1301, University of Toulouse III, Inserm, UPS, CNRS, Toulouse, France.
| | - Jeremy Raffin
- IHU HealthAge, Toulouse, France
- Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France
| | - Emmanuel Gonzalez-Bautista
- IHU HealthAge, Toulouse, France
- Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France
| | - Patrick Ritz
- CERPOP UMR1295, University of Toulouse III, Inserm, UPS, Toulouse, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Toulouse University Hospital, Toulouse, France
| | - Gabor Abellan Van Kan
- IHU HealthAge, Toulouse, France
- Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France
- CERPOP UMR1295, University of Toulouse III, Inserm, UPS, Toulouse, France
| | - Fabien Pillard
- Department of Sport Medicine, Hospital and University of Toulouse, Toulouse, France
- Institut RESTORE, UMR 1301, University of Toulouse III, Inserm, UPS, CNRS, Toulouse, France
| | | | - Sophie Guyonnet
- IHU HealthAge, Toulouse, France
- Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France
- CERPOP UMR1295, University of Toulouse III, Inserm, UPS, Toulouse, France
| | - Cédric Dray
- Institut RESTORE, UMR 1301, University of Toulouse III, Inserm, UPS, CNRS, Toulouse, France
| | - Bruno Vellas
- IHU HealthAge, Toulouse, France
- Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France
- CERPOP UMR1295, University of Toulouse III, Inserm, UPS, Toulouse, France
| | - Philipe de Souto Barreto
- IHU HealthAge, Toulouse, France
- Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France
- CERPOP UMR1295, University of Toulouse III, Inserm, UPS, Toulouse, France
| | - Yves Rolland
- IHU HealthAge, Toulouse, France
- Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France
- CERPOP UMR1295, University of Toulouse III, Inserm, UPS, Toulouse, France
| |
Collapse
|
3
|
Torrens-Mas M, Navas-Enamorado C, Galmes-Panades A, Masmiquel L, Sanchez-Polo A, Capo X, Gonzalez-Freire M. GDF-15 as a proxy for epigenetic aging: associations with biological age markers, and physical function. Biogerontology 2024; 26:22. [PMID: 39644331 PMCID: PMC11625061 DOI: 10.1007/s10522-024-10165-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
Growth differentiation factor 15 (GDF-15) has emerged as a significant biomarker of aging, linked to various physiological and pathological processes. This study investigates circulating GDF-15 levels in a cohort of healthy individuals from the Balearic Islands, exploring its associations with biological age markers, including multiple DNA methylation (DNAm) clocks, physical performance, and other age-related biomarkers. Seventy-two participants were assessed for general health, body composition, and physical function, with GDF-15 levels quantified using ELISA. Our results indicate that GDF-15 levels significantly increase with age, particularly in individuals over 60. Strong positive correlations were observed between GDF-15 levels and DNAm GrimAge, DNAm PhenoAge, Hannum, and Zhang clocks, suggesting that GDF-15 could serve as a proxy for epigenetic aging. Additionally, GDF-15 levels were linked to markers of impaired glycemic control, systemic inflammation, and physical decline, including decreased lung function and grip strength, especially in men. These findings highlight the use of GDF-15 as a biomarker for aging and age-related functional decline. Given that GDF-15 is easier to measure than DNA methylation, it has the potential to be more readily implemented in clinical settings for broader health assessment and management.
Collapse
Affiliation(s)
- Margalida Torrens-Mas
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d´Investigació en Ciències de La Salut (IUNICS), University of the Balearic Islands, 07122, Palma, Spain
| | - Cayetano Navas-Enamorado
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain
| | - Aina Galmes-Panades
- Physical Activity and Sport Sciences Research Group (GICAFE), Institute for Educational Research and Innovation (IRIE), University of the Balearic Islands, 07122, Palma, Spain
- Consorcio CIBER, M.P. Fisiopatología de La Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Luis Masmiquel
- Vascular and Metabolic Pathologies Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain
| | - Andrés Sanchez-Polo
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain
| | - Xavier Capo
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain.
- Research Group in Community Nutrition and Oxidative Stress, University of the Balearic Islands-Institut Universitari d´Investigació en Ciències de La Salut (IUNICS), 07122, Palma, Spain.
| | - Marta Gonzalez-Freire
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain.
| |
Collapse
|
4
|
Feng L, Li B, Yong SS, Wu X, Tian Z. Exercise and nutrition benefit skeletal muscle: From influence factor and intervention strategy to molecular mechanism. SPORTS MEDICINE AND HEALTH SCIENCE 2024; 6:302-314. [PMID: 39309454 PMCID: PMC11411340 DOI: 10.1016/j.smhs.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/24/2024] [Accepted: 02/18/2024] [Indexed: 09/25/2024] Open
Abstract
Sarcopenia is a progressive systemic skeletal muscle disease induced by various physiological and pathological factors, including aging, malnutrition, denervation, and cardiovascular diseases, manifesting as the decline of skeletal muscle mass and function. Both exercise and nutrition produce beneficial effects on skeletal muscle growth and are viewed as feasible strategies to prevent sarcopenia. Mechanisms involve regulating blood flow, oxidative stress, inflammation, apoptosis, protein synthesis and degradation, and satellite cell activation through exerkines and gut microbiomes. In this review, we summarized and discussed the latest progress and future development of the above mechanisms for providing a theoretical basis and ideas for the prevention and treatment of sarcopenia.
Collapse
Affiliation(s)
- Lili Feng
- College of Education, Physical Education Department, Zhejiang University, Hangzhou, 310058, China
| | - Bowen Li
- College of Education, Physical Education Department, Zhejiang University, Hangzhou, 310058, China
| | - Su Sean Yong
- College of Education, Physical Education Department, Zhejiang University, Hangzhou, 310058, China
| | - Xiaonan Wu
- The Information and Communication College, National University of Defense Technology, Xi'an, 710106, China
| | - Zhenjun Tian
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, 710119, China
| |
Collapse
|
5
|
Bathina S, Lopez VF, Prado M, Ballato E, Colleluori G, Tetlay M, Villareal DT, Mediwala S, Chen R, Qualls C, Armamento‐Villareal R. Health implications of racial differences in serum growth differentiation factor levels among men with obesity. Physiol Rep 2024; 12:e70124. [PMID: 39668628 PMCID: PMC11638490 DOI: 10.14814/phy2.70124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/04/2024] [Accepted: 11/04/2024] [Indexed: 12/14/2024] Open
Abstract
Growth differentiation factor (GDF15) has been considered a biomarker and recently a hormonal driver for diseases in different populations. However, the role of GDF15 as a biomarker of health outcomes in obese men from different racial/ethnic background has not been evaluated. The objective of this study was to investigate the racial/ethnic differences on the relationship between GDF15 and markers of glucometabolic status, hormonal profile, body composition and bone mineral density (BMD) in obese men. One hundred ninety-three obese men from diverse racial/ethnic backgrounds were enrolled. BMD and body composition were measured by dual energy X-ray absorptiometry. Serum GDF15, osteocalcin, C-terminal telopeptide, sclerostin, adiponectin, leptin, estradiol, testosterone, follicle-stimulating hormone, luteinizing hormone, 25-hydroxyvitamin D, lipid profile, and hemoglobin A1C (A1C) were measured. Non-African Americans (NAA) had significantly higher GDF15 level than African Americans (AA). Level was also higher in patients with type 2 diabetes (T2DM). In both the groups GDF15 correlated with A1C and lean mass. However. GDF15 correlated with body fat, LDL total cholesterol and femoral neck BMD only in NAA and with appendicular lean mass only in AA. Ethnicity, total cholesterol and T2DM were found to be independent predictors of GDF15. We conclude that GDF15 may influence glucometabolic status, body composition and bone parameters which may affect cardiovascular risk and osteoporosis between races.
Collapse
Affiliation(s)
- Siresha Bathina
- Division of Endocrinology Diabetes and Metabolism at Baylor College of MedicineHoustonTexasUSA
- Department of MedicineMichael E. De Bakey Veterans Affairs (VA) Medical CenterHoustonTexasUSA
| | - Virginia Fuenmayor Lopez
- Division of Endocrinology Diabetes and Metabolism at Baylor College of MedicineHoustonTexasUSA
- Department of MedicineMichael E. De Bakey Veterans Affairs (VA) Medical CenterHoustonTexasUSA
| | - Mia Prado
- Division of Endocrinology Diabetes and Metabolism at Baylor College of MedicineHoustonTexasUSA
- Department of MedicineMichael E. De Bakey Veterans Affairs (VA) Medical CenterHoustonTexasUSA
| | - Elliot Ballato
- Division of Endocrinology Diabetes and Metabolism at Baylor College of MedicineHoustonTexasUSA
| | - Georgia Colleluori
- Division of Endocrinology Diabetes and Metabolism at Baylor College of MedicineHoustonTexasUSA
| | - Maryam Tetlay
- Division of Endocrinology Diabetes and Metabolism at Baylor College of MedicineHoustonTexasUSA
| | - Dennis Tan Villareal
- Division of Endocrinology Diabetes and Metabolism at Baylor College of MedicineHoustonTexasUSA
- Department of MedicineMichael E. De Bakey Veterans Affairs (VA) Medical CenterHoustonTexasUSA
| | - Sanjay Mediwala
- Division of Endocrinology Diabetes and Metabolism at Baylor College of MedicineHoustonTexasUSA
- Department of MedicineMichael E. De Bakey Veterans Affairs (VA) Medical CenterHoustonTexasUSA
| | - Rui Chen
- Division of Endocrinology Diabetes and Metabolism at Baylor College of MedicineHoustonTexasUSA
- Department of MedicineMichael E. De Bakey Veterans Affairs (VA) Medical CenterHoustonTexasUSA
| | - Clifford Qualls
- Department of Mathematics and StatisticsUniversity of New MexicoAlbuquerqueNew MexicoUSA
| | - Reina Armamento‐Villareal
- Division of Endocrinology Diabetes and Metabolism at Baylor College of MedicineHoustonTexasUSA
- Department of MedicineMichael E. De Bakey Veterans Affairs (VA) Medical CenterHoustonTexasUSA
| |
Collapse
|
6
|
Kim S, Ayan B, Shayan M, Rando TA, Huang NF. Skeletal muscle-on-a-chip in microgravity as a platform for regeneration modeling and drug screening. Stem Cell Reports 2024; 19:1061-1073. [PMID: 39059375 PMCID: PMC11368695 DOI: 10.1016/j.stemcr.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 06/22/2024] [Accepted: 06/23/2024] [Indexed: 07/28/2024] Open
Abstract
Microgravity has been shown to lead to both muscle atrophy and impaired muscle regeneration. The purpose was to study the efficacy of microgravity to model impaired muscle regeneration in an engineered muscle platform and then to demonstrate the feasibility of performing drug screening in this model. Engineered human muscle was launched to the International Space Station National Laboratory, where the effect of microgravity exposure for 7 days was examined by transcriptomics and proteomics approaches. Gene set enrichment analysis of engineered muscle cultured in microgravity, compared to normal gravity conditions, highlighted a metabolic shift toward lipid and fatty acid metabolism, along with increased apoptotic gene expression. The addition of pro-regenerative drugs, insulin-like growth factor-1 (IGF-1) and a 15-hydroxyprostaglandin dehydrogenase inhibitor (15-PGDH-i), partially inhibited the effects of microgravity. In summary, microgravity mimics aspects of impaired myogenesis, and the addition of these drugs could partially inhibit the effects induced by microgravity.
Collapse
Affiliation(s)
- Soochi Kim
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Bugra Ayan
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Mahdis Shayan
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA; Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto, Health Care System, Palo Alto, CA 94304, USA.
| | - Ngan F Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA; Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto, Health Care System, Palo Alto, CA 94304, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA; Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
7
|
Enzer NA, Chiles J, Mason S, Shirahata T, Castro V, Regan E, Choi B, Yuan NF, Diaz AA, Washko GR, McDonald ML, Estépar RSJ, Ash SY. Proteomics and machine learning in the prediction and explanation of low pectoralis muscle area. Sci Rep 2024; 14:17981. [PMID: 39097658 PMCID: PMC11297919 DOI: 10.1038/s41598-024-68447-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 07/23/2024] [Indexed: 08/05/2024] Open
Abstract
Low muscle mass is associated with numerous adverse outcomes independent of other associated comorbid diseases. We aimed to predict and understand an individual's risk for developing low muscle mass using proteomics and machine learning. We identified eight biomarkers associated with low pectoralis muscle area (PMA). We built three random forest classification models that used either clinical measures, feature selected biomarkers, or both to predict development of low PMA. The area under the receiver operating characteristic curve for each model was: clinical-only = 0.646, biomarker-only = 0.740, and combined = 0.744. We displayed the heterogenetic nature of an individual's risk for developing low PMA and identified two distinct subtypes of participants who developed low PMA. While additional validation is required, our methods for identifying and understanding individual and group risk for low muscle mass could be used to enable developments in the personalized prevention of low muscle mass.
Collapse
Affiliation(s)
- Nicholas A Enzer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
| | - Joe Chiles
- Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
- COPDGene Study Consortium, Denver, CO, USA
| | - Stefanie Mason
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
| | - Toru Shirahata
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- Department of Respiratory Medicine, Saitama Medical University, Saitama, Japan
| | - Victor Castro
- Boston University School of Medicine, Boston, MA, USA
| | - Elizabeth Regan
- COPDGene Study Consortium, Denver, CO, USA
- Division of Rheumatology, Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Bina Choi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
| | - Nancy F Yuan
- Department of Biomedical Informatics, University of California at San Diego, San Diego, CA, USA
| | - Alejandro A Diaz
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- COPDGene Study Consortium, Denver, CO, USA
| | - George R Washko
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- COPDGene Study Consortium, Denver, CO, USA
| | - Merry-Lynn McDonald
- Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
- COPDGene Study Consortium, Denver, CO, USA
| | - Raúl San José Estépar
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Samuel Y Ash
- Department of Critical Care Medicine, South Shore Hospital, 55 Fogg Road, South Weymouth, MA, 02190, USA.
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
8
|
Shafqat A, Masters MC, Tripathi U, Tchkonia T, Kirkland JL, Hashmi SK. Long COVID as a disease of accelerated biological aging: An opportunity to translate geroscience interventions. Ageing Res Rev 2024; 99:102400. [PMID: 38945306 DOI: 10.1016/j.arr.2024.102400] [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: 04/21/2024] [Revised: 06/12/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
It has been four years since long COVID-the protracted consequences that survivors of COVID-19 face-was first described. Yet, this entity continues to devastate the quality of life of an increasing number of COVID-19 survivors without any approved therapy and a paucity of clinical trials addressing its biological root causes. Notably, many of the symptoms of long COVID are typically seen with advancing age. Leveraging this similarity, we posit that Geroscience-which aims to target the biological drivers of aging to prevent age-associated conditions as a group-could offer promising therapeutic avenues for long COVID. Bearing this in mind, this review presents a translational framework for studying long COVID as a state of effectively accelerated biological aging, identifying research gaps and offering recommendations for future preclinical and clinical studies.
Collapse
Affiliation(s)
- Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.
| | - Mary Clare Masters
- Division of Infectious Diseases, Northwestern University, Chicago, IL, USA
| | - Utkarsh Tripathi
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA; Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Shahrukh K Hashmi
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA; Research and Innovation Center, Department of Health, Abu Dhabi, UAE; College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
9
|
León-González R, Ortolá R, Carballo-Casla A, Sotos-Prieto M, Buño-Soto A, Rodríguez-Sánchez I, Pastor-Barriuso R, Rodríguez-Artalejo F, García-Esquinas E. Growth Differentiation Factor 15 as a Biomarker of Cardiovascular Risk in Chronic Musculoskeletal Pain. J Gerontol A Biol Sci Med Sci 2024; 79:glae163. [PMID: 38975684 DOI: 10.1093/gerona/glae163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND It is unknown whether growth differentiation factor 15 (GDF-15) is associated with chronic musculoskeletal pain (CMP) and whether or not its association with incident cardiovascular disease (CVD) changes according to CMP status. METHODS In total, 1 957 randomly selected adults aged ≥65 years without prior CVD were followed up between 2015 and 2023. CMP was classified according to its intensity, frequency, and interference with daily activities. The association between GDF-15 levels and CMP was assessed using linear models with progressive inclusion of potential confounders, whereas the association between GDF-15 and CVD risk was evaluated with Cox proportional hazard models with similar adjustment and interaction terms between GDF-15 and CMP. The incremental predictive performance of GDF-15 over standard predictors was evaluated using discrimination and risk reclassification metrics. RESULTS GDF-15 concentrations were 6.90% (95% confidence interval [CI]: 2.56; 11.25) higher in individuals with CMP, and up to 8.89% (4.07; 15.71) and 15.79% (8.43; 23.16) higher in those with ≥3 CMP locations and interfering pain. These increased levels were influenced by a higher prevalence of cardiometabolic risk factors, functional impairments, depressive symptoms, and greater levels of inflammation in individuals with CMP. In fully adjusted models, a twofold increase in GDF-15 was associated with a 1.49 increased risk (95% CI: 1.08; 2.05) of a CVD event in individuals with CMP, but not among those without CMP (1.02 [0.77; 1.35]); p-interaction 0.041. Adding GDF-15 to models including the Framingham Risk Score improved predictive performance among individuals with CMP. CONCLUSIONS We provide evidence that GDF-15 could serve as a biomarker to assess CMP, as well as to predict CVD incidence in individuals with CMP.
Collapse
Affiliation(s)
- Rocío León-González
- Department of Preventive Medicine and Public Health, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rosario Ortolá
- Department of Preventive Medicine and Public Health, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Adrián Carballo-Casla
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Neurobiology, Aging Research Center, Care Sciences and Society Karolinska Institutet & Stockholm University, Stockholm, Sweden
| | - Mercedes Sotos-Prieto
- Department of Preventive Medicine and Public Health, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Environmental Health and Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Antonio Buño-Soto
- Department of Laboratory Medicine, La Paz University Hospital-IdiPaz, Madrid, Spain
| | | | - Roberto Pastor-Barriuso
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
- National Center for Epidemiology, Carlos III Health Institute, Madrid, Spain
| | - Fernando Rodríguez-Artalejo
- Department of Preventive Medicine and Public Health, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- IMDEA Food Institute, CEI UAM+CSIC, Madrid, Spain
| | - Esther García-Esquinas
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
- National Center for Epidemiology, Carlos III Health Institute, Madrid, Spain
| |
Collapse
|
10
|
Nicholson T, Dhaliwal A, Quinlan JI, Allen SL, Williams FR, Hazeldine J, McGee KC, Sullivan J, Breen L, Elsharkawy AM, Armstrong MJ, Jones SW, Greig CA, Lord JM. Accelerated aging of skeletal muscle and the immune system in patients with chronic liver disease. Exp Mol Med 2024; 56:1667-1681. [PMID: 39026032 PMCID: PMC11297261 DOI: 10.1038/s12276-024-01287-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/10/2024] [Accepted: 04/30/2024] [Indexed: 07/20/2024] Open
Abstract
Patients with chronic liver disease (CLD) often present with significant frailty, sarcopenia, and impaired immune function. However, the mechanisms driving the development of these age-related phenotypes are not fully understood. To determine whether accelerated biological aging may play a role in CLD, epigenetic, transcriptomic, and phenotypic assessments were performed on the skeletal muscle tissue and immune cells of CLD patients and age-matched healthy controls. Accelerated biological aging of the skeletal muscle tissue of CLD patients was detected, as evidenced by an increase in epigenetic age compared with chronological age (mean +2.2 ± 4.8 years compared with healthy controls at -3.0 ± 3.2 years, p = 0.0001). Considering disease etiology, age acceleration was significantly greater in both the alcohol-related (ArLD) (p = 0.01) and nonalcoholic fatty liver disease (NAFLD) (p = 0.0026) subgroups than in the healthy control subgroup, with no age acceleration observed in the immune-mediated subgroup or healthy control subgroup (p = 0.3). The skeletal muscle transcriptome was also enriched for genes associated with cellular senescence. Similarly, blood cell epigenetic age was significantly greater than that in control individuals, as calculated using the PhenoAge (p < 0.0001), DunedinPACE (p < 0.0001), or Hannum (p = 0.01) epigenetic clocks, with no difference using the Horvath clock. Analysis of the IMM-Age score indicated a prematurely aged immune phenotype in CLD patients that was 2-fold greater than that observed in age-matched healthy controls (p < 0.0001). These findings suggested that accelerated cellular aging may contribute to a phenotype associated with advanced age in CLD patients. Therefore, therapeutic interventions to reduce biological aging in CLD patients may improve health outcomes.
Collapse
Affiliation(s)
- Thomas Nicholson
- NIHR Birmingham Biomedical Research Centre, University Hospital Birmingham and University of Birmingham, Birmingham, UK
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Birmingham, UK
| | - Amritpal Dhaliwal
- NIHR Birmingham Biomedical Research Centre, University Hospital Birmingham and University of Birmingham, Birmingham, UK
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Jonathan I Quinlan
- NIHR Birmingham Biomedical Research Centre, University Hospital Birmingham and University of Birmingham, Birmingham, UK
- School of Sport, Exercise and Rehabilitation Science, University of Birmingham, Birmingham, UK
| | - Sophie L Allen
- NIHR Birmingham Biomedical Research Centre, University Hospital Birmingham and University of Birmingham, Birmingham, UK
- School of Sport, Exercise and Rehabilitation Science, University of Birmingham, Birmingham, UK
| | - Felicity R Williams
- NIHR Birmingham Biomedical Research Centre, University Hospital Birmingham and University of Birmingham, Birmingham, UK
- School of Sport, Exercise and Rehabilitation Science, University of Birmingham, Birmingham, UK
| | - Jon Hazeldine
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Birmingham, UK
| | - Kirsty C McGee
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Birmingham, UK
| | - Jack Sullivan
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Birmingham, UK
| | - Leigh Breen
- NIHR Birmingham Biomedical Research Centre, University Hospital Birmingham and University of Birmingham, Birmingham, UK
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Birmingham, UK
- School of Sport, Exercise and Rehabilitation Science, University of Birmingham, Birmingham, UK
| | - Ahmed M Elsharkawy
- NIHR Birmingham Biomedical Research Centre, University Hospital Birmingham and University of Birmingham, Birmingham, UK
- Liver Transplantation Unit, Queen Elizabeth Hospital, Birmingham, UK
| | - Matthew J Armstrong
- NIHR Birmingham Biomedical Research Centre, University Hospital Birmingham and University of Birmingham, Birmingham, UK
- Liver Transplantation Unit, Queen Elizabeth Hospital, Birmingham, UK
| | - Simon W Jones
- NIHR Birmingham Biomedical Research Centre, University Hospital Birmingham and University of Birmingham, Birmingham, UK
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Birmingham, UK
| | - Carolyn A Greig
- NIHR Birmingham Biomedical Research Centre, University Hospital Birmingham and University of Birmingham, Birmingham, UK
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Birmingham, UK
- School of Sport, Exercise and Rehabilitation Science, University of Birmingham, Birmingham, UK
| | - Janet M Lord
- NIHR Birmingham Biomedical Research Centre, University Hospital Birmingham and University of Birmingham, Birmingham, UK.
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Birmingham, UK.
| |
Collapse
|
11
|
Li J, Shen Z, Lin Y, Wang Z, Li M, Sun H, Wang Q, Zhao C, Xu J, Lu X, Gao W. DNA methylation of skeletal muscle function-related secretary factors identifies FGF2 as a potential biomarker for sarcopenia. J Cachexia Sarcopenia Muscle 2024; 15:1209-1217. [PMID: 38641928 PMCID: PMC11154778 DOI: 10.1002/jcsm.13472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 03/07/2024] [Accepted: 03/19/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND Sarcopenia is characterized by progressive loss of muscle mass and function due to aging. DNA methylation has been identified to play important roles in the dysfunction of skeletal muscle. The aim of our present study was to explore the whole blood sample-based methylation changes of skeletal muscle function-related factors in patients with sarcopenia. METHODS The overall DNA methylation levels were analysed by using MethlTarget™ DNA Methylation Analysis platform in a discovery set consistent of 50 sarcopenic older adults (aged ≥65 years) and 50 age- and sex-matched non-sarcopenic individuals. The candidate differentially methylated regions (DMRs) were further validated by Methylation-specific PCR (MSP) in another two independent larger sets and confirmed by pyrosequencing. Receiver operating characteristic (ROC) curve analysis was used to determine the optimum cut-off levels of fibroblast growth factor 2 (FGF2)_30 methylation best predicting sarcopenia and area under the ROC curve (AUC) was measured. The correlation between candidate DMRs and the risk of sarcopenia was investigated by univariate analysis and multivariate logistic regression analysis. RESULTS Among 1149 cytosine-phosphate-guanine (CpG) sites of 27 skeletal muscle function-related secretary factors, 17 differentially methylated CpG sites and 7 differentially methylated regions (DMRs) were detected between patients with sarcopenia and control subjects in the discovery set. Further methylation-specific PCR identified that methylation of fibroblast growth factor 2 (FGF2)_30 was lower in patients with sarcopenia and the level was decreased as the severity of sarcopenia increased, which was confirmed by pyrosequencing. Correlation analysis demonstrated that the methylation level of FGF2_30 was positively correlated to ASMI (r = 0.372, P < 0.001), grip strength (r = 0.334, P < 0.001), and gait speed (r = 0.411, P < 0.001). ROC curve analysis indicated that the optimal cut-off value of FGF2_30 methylation level that predicted sarcopenia was 0.15 with a sensitivity of 84.6% and a specificity of 70.1% (AUC = 0.807, 95% CI = 0.756-0.858, P < 0.001). Multivariate logistic regression analyses showed that lower FGF2_30 methylation level (<0.15) was significantly associated with increased risk of sarcopenia even after adjustment for potential confounders including age, sex, and BMI (adjusted OR = 9.223, 95% CI: 6.614-12.861, P < 0.001). CONCLUSIONS Our results suggest that lower FGF2_30 methylation is correlated with the risk and severity of sarcopenia in the older adults, indicating that FGF2 methylation serve as a surrogate biomarker for the screening and evaluation of sarcopenia.
Collapse
Affiliation(s)
- Jia‐Wen Li
- Department of Geriatrics, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| | - Zheng‐Kai Shen
- Jiangsu Province Center for Disease Control and PreventionNanjingChina
| | - Yu‐Shuang Lin
- Department of Geriatrics, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Zhi‐Yue Wang
- Department of Geriatrics, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Mei‐Lin Li
- Department of Geriatrics, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Hui‐Xian Sun
- Department of Geriatrics, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Quan Wang
- Department of Geriatrics, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Can Zhao
- Department of Geriatrics, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Jin‐Shui Xu
- Jiangsu Province Center for Disease Control and PreventionNanjingChina
| | - Xiang Lu
- Department of Geriatrics, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Wei Gao
- Department of Geriatrics, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| |
Collapse
|
12
|
Wang Y, Ding S. Extracellular vesicles in cancer cachexia: deciphering pathogenic roles and exploring therapeutic horizons. J Transl Med 2024; 22:506. [PMID: 38802952 PMCID: PMC11129506 DOI: 10.1186/s12967-024-05266-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Cancer cachexia (CC) is a debilitating syndrome that affects 50-80% of cancer patients, varying in incidence by cancer type and significantly diminishing their quality of life. This multifactorial syndrome is characterized by muscle and fat loss, systemic inflammation, and metabolic imbalance. Extracellular vesicles (EVs), including exosomes and microvesicles, play a crucial role in the progression of CC. These vesicles, produced by cancer cells and others within the tumor environment, facilitate intercellular communication by transferring proteins, lipids, and nucleic acids. A comprehensive review of the literature from databases such as PubMed, Scopus, and Web of Science reveals insights into the formation, release, and uptake of EVs in CC, underscoring their potential as diagnostic and prognostic biomarkers. The review also explores therapeutic strategies targeting EVs, which include modifying their release and content, utilizing them for drug delivery, genetically altering their contents, and inhibiting key cachexia pathways. Understanding the role of EVs in CC opens new avenues for diagnostic and therapeutic approaches, potentially mitigating the syndrome's impact on patient survival and quality of life.
Collapse
Affiliation(s)
- Yifeng Wang
- Department of Thoracic Surgery, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital, Nantong, 226001, P.R. China
- School of Medicine, Nantong University, Nantong, 226001, P.R. China
| | - Shengguang Ding
- Department of Thoracic Surgery, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital, Nantong, 226001, P.R. China.
| |
Collapse
|
13
|
Chiariello A, Conte G, Rossetti L, Trofarello L, Salvioli S, Conte M. Different roles of circulating and intramuscular GDF15 as markers of skeletal muscle health. Front Endocrinol (Lausanne) 2024; 15:1404047. [PMID: 38808117 PMCID: PMC11130406 DOI: 10.3389/fendo.2024.1404047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024] Open
Abstract
Introduction Growth Differentiation Factor 15 (GDF15) is a mitokine expressed in response to various stresses whose circulating levels increase with age and are associated with numerous pathological conditions, including muscle wasting and sarcopenia. However, the use of circulating GDF15 (c-GDF15) as a biomarker of sarcopenia is still debated. Moreover, the role of GDF15 intracellular precursor, pro-GDF15, in human skeletal muscle (SM-GDF15) is not totally understood. In order to clarify these points, the association of both forms of GDF15 with parameters of muscle strength, body composition, metabolism and inflammation was investigated. Methods the levels of c-GDF15 and SM-GDF15 were evaluated in plasma and muscle biopsies, respectively, of healthy subjects (HS) and patients with lower limb mobility impairment (LLMI), either young (<40 years-old) or old (>70 years-old). Other parameters included in the analysis were Isometric Quadriceps Strength (IQS), BMI, lean and fat mass percentage, Vastus lateralis thickness, as well as circulating levels of Adiponectin, Leptin, Resistin, IGF-1, Insulin, IL6, IL15 and c-PLIN2. Principal Component Analysis (PCA), Canonical Discriminant Analysis (CDA) and Receiving Operating Characteristics (ROC) analysis were performed. Results c-GDF15 but not SM-GDF15 levels resulted associated with decreased IQS and IGF-1 levels in both HS and LLMI, while only in LLMI associated with increased levels of Resistin. Moreover, in LLMI both c-GDF15 and SM-GDF15 levels were associated with IL-6 levels, but interestingly SM-GDF15 is lower in LLMI with respect to HS. Furthermore, a discrimination of the four groups of subjects based on these parameters was possible with PCA and CDA. In particular HS, LLMI over 70 years or under 40 years of age were discriminated based on SM-GDF15, c-GDF15 and Insulin levels, respectively. Conclusion our data support the idea that c-GDF15 level could be used as a biomarker of decreased muscle mass and strength. Moreover, it is suggested that c-GDF15 has a different diagnostic significance with respect to SM-GDF15, which is likely linked to a healthy and active state.
Collapse
Affiliation(s)
- Antonio Chiariello
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Giuseppe Conte
- Department of Agriculture, Food and Environment, University of Pisa, Pisa, Italy
| | - Luca Rossetti
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Interdepartmental Centre “Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate)”, University of Bologna, Bologna, Italy
| | - Lorenzo Trofarello
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Stefano Salvioli
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Maria Conte
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| |
Collapse
|
14
|
Pena-Leon V, Perez-Lois R, Villalon M, Folgueira C, Barja-Fernández S, Prida E, Baltar J, Santos F, Fernø J, García-Caballero T, Nogueiras R, Quiñones M, Al-Massadi O, Seoane LM. Gastric GDF15 levels are regulated by age, sex, and nutritional status in rodents and humans. J Endocrinol Invest 2024; 47:1139-1154. [PMID: 37955834 DOI: 10.1007/s40618-023-02232-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 10/21/2023] [Indexed: 11/14/2023]
Abstract
AIM Growth differentiation factor 15 (GDF15) is a stress response cytokine that has been proposed as a relevant metabolic hormone. Descriptive studies have shown that plasma GDF15 levels are regulated by short term changes in nutritional status, such as fasting, or in obesity. However, few data exist regarding how GDF15 levels are regulated in peripheral tissues. The aim of the present work was to study the variations on gastric levels of GDF15 and its precursor under different physiological conditions, such as short-term changes in nutritional status or overfeeding achieved by HFD. Moreover, we also address the sex- and age-dependent alterations in GDF15 physiology. METHODS The levels of gastric and plasma GDF15 and its precursor were measured in lean and obese mice, rats and humans by western blot, RT-PCR, ELISA, immunohistochemistry and by an in vitro organ culture system. RESULTS Our results show a robust regulation of gastric GDF15 production by fasting in rodents. In obesity an increase in GDF15 secretion from the stomach is reflected with an increase in circulating levels of GDF15 in rats and humans. Moreover, gastric GDF15 levels increase with age in both rats and humans. Finally, gastric GDF15 levels display sexual dimorphism, which could explain the difference in circulating GFD15 levels between males and females, observed in both humans and rodents. CONCLUSIONS Our results provide clear evidence that gastric GDF15 is a critical contributor of circulating GDF15 levels and can explain some of the metabolic effects induced by GDF15.
Collapse
Affiliation(s)
- V Pena-Leon
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - R Perez-Lois
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - M Villalon
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - C Folgueira
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - S Barja-Fernández
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - E Prida
- Translational Endocrinology Group, Endocrinology Section, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (IDIS/CHUS), Santiago de Compostela, Spain
| | - J Baltar
- Servicio de Cirugía General y del Aparato Digestivo, CHUS7SERGAS Santiago de Compostela, Rua R Baltar s/n, 15706, Santiago de Compostela, Spain
| | - F Santos
- Servicio de Cirugía General y del Aparato Digestivo, CHUS7SERGAS Santiago de Compostela, Rua R Baltar s/n, 15706, Santiago de Compostela, Spain
| | - J Fernø
- Hormone Laboratory, Department of Biochemistry and Pharmacology, Haukeland University Hospital, 5201, Bergen, Norway
| | - T García-Caballero
- Departamento de Ciencias Morfologicas, Facultad de Medicina, USC, Complejo Hospitalario de Santiago (CHUS/SERGAS), Santiago de Compostela, Spain
| | - R Nogueiras
- Departamento de Fisiología, Instituto de Investigación Sanitaria de Santiago de Compostela, CIMUS, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Spain, Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - M Quiñones
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Spain, Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - O Al-Massadi
- Translational Endocrinology Group, Endocrinology Section, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (IDIS/CHUS), Santiago de Compostela, Spain.
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Spain, Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain.
| | - L M Seoane
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain.
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Spain, Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain.
| |
Collapse
|
15
|
Oba K, Ishikawa J, Tamura Y, Fujita Y, Ito M, Iizuka A, Fujiwara Y, Kodera R, Toyoshima K, Chiba Y, Tanaka M, Araki A. Serum Growth Differentiation Factor 15 Levels Predict the Incidence of Frailty among Patients with Cardiometabolic Diseases. Gerontology 2024; 70:517-525. [PMID: 38286122 DOI: 10.1159/000536150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 01/02/2024] [Indexed: 01/31/2024] Open
Abstract
INTRODUCTION Frailty is a crucial health issue among older adults. Growth differentiation factor 15 (GDF15) is associated with inflammation, oxidative stress, insulin resistance, and mitochondrial dysfunction, which are possible pathogeneses of frailty. However, few longitudinal studies have investigated the association between GDF15 and the incidence of frailty. Therefore, we investigated whether high serum GDF15 levels are associated with the incidence of frailty. METHODS A total of 175 older adults (mean age: 77 ± 6 years; 63% women) with cardiometabolic diseases and no frailty out of the two criteria at baseline participated. Individuals with severe renal impairment or severe cognitive impairment were excluded. Serum GDF15 levels were measured at baseline. Patients were asked to assess frailty status at baseline and annually during follow-up using the modified version of the Cardiovascular Health Study (mCHS) and the Kihon Checklist (KCL). We examined the association between GDF15 tertiles and each frailty measure during follow-up (median 38-39 months). In the multivariate Cox regression analysis, with the GDF15 tertile groups as the explanatory variables, hazard ratios (HRs) and 95% confidence intervals (CIs) for incident frailty were calculated after adjusting for covariates and using the lowest tertile group as the reference. RESULTS During the follow-up period, 25.6% and 34.0% of patients developed frailty, as defined by the mCHS and KCL, respectively. The highest GDF15 tertile group had a significantly higher incidence of mCHS- or KCL-defined frailty than the lowest GDF15 tertile group. Multivariate Cox regression analysis revealed that the adjusted HRs for incident mCHS- and KCL-defined frailty in the highest GDF15 tertile group were 3.9 (95% CI: 1.3-12.0) and 2.7 (95% CI: 1.1-6.9), respectively. CONCLUSION High serum GDF15 levels predicted the incidence of frailty among older adults with cardiometabolic diseases and could be an effective marker of the risk for frailty in interventions aimed at preventing frailty, such as exercise and nutrition.
Collapse
Affiliation(s)
- Kazuhito Oba
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Joji Ishikawa
- Department of Cardiology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Yoshiaki Tamura
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Yasunori Fujita
- Research Team for Functional Biogerontology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Masafumi Ito
- Research Team for Functional Biogerontology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Ai Iizuka
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
- Research Team for Social Participation and Community, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Yoshinori Fujiwara
- Research Team for Social Participation and Community, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Remi Kodera
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Kenji Toyoshima
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Yuko Chiba
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Masashi Tanaka
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Atsushi Araki
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| |
Collapse
|
16
|
Enzer NA, Chiles J, Mason S, Shirahata T, Castro V, Regan E, Choi B, Yuan NF, Diaz AA, Washko GR, McDonald ML, Estépar RSJ, Ash SY. Proteomics and Machine Learning in the Prediction and Explanation of Low Pectoralis Muscle Area. RESEARCH SQUARE 2024:rs.3.rs-3957125. [PMID: 38496412 PMCID: PMC10942559 DOI: 10.21203/rs.3.rs-3957125/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Low muscle mass is associated with numerous adverse outcomes independent of other associated comorbid diseases. We aimed to predict and understand an individual's risk for developing low muscle mass using proteomics and machine learning. We identified 8 biomarkers associated with low pectoralis muscle area (PMA). We built 3 random forest classification models that used either clinical measures, feature selected biomarkers, or both to predict development of low PMA. The area under the receiver operating characteristic curve for each model was: clinical-only = 0.646, biomarker-only = 0.740, and combined = 0.744. We displayed the heterogenetic nature of an individual's risk for developing low PMA and identified 2 distinct subtypes of participants who developed low PMA. While additional validation is required, our methods for identifying and understanding individual and group risk for low muscle mass could be used to enable developments in the personalized prevention of low muscle mass.
Collapse
|
17
|
Rezaei S, Eslami R, Tartibian B. The effects of TRX suspension training on sarcopenic biomarkers and functional abilities in elderlies with sarcopenia: a controlled clinical trial. BMC Sports Sci Med Rehabil 2024; 16:58. [PMID: 38409184 PMCID: PMC10898163 DOI: 10.1186/s13102-024-00849-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 02/14/2024] [Indexed: 02/28/2024]
Abstract
BACKGROUND Sarcopenia is an age-related progressive loss of muscle mass and strength that can be modulated by resistance training. This study aimed to investigate the effects of TRX Suspension Training (TST) on serum levels of neuromuscular and growth factors and functional indices in elderly men with sarcopenia, an age-related condition characterized by progressive muscle mass and strength loss. METHODS Nineteen sarcopenic elderly men (age = 74.87 ± 4.58 years) were randomly assigned into two groups, the TST group (n = 10) and the control group (n = 9). Serum concentrations of regulatory muscle markers, anthropometric and body composition indices, and functional tests were evaluated at baseline and after 8 weeks. The training protocol consisted of eight weeks of TRX exercises, with three weekly sessions. RESULTS After 8 weeks of training, growth factors such as Follistatin (FST) (P = 0.001), 22 kDa C-terminal agrin fragment (CAF) (P = 0.031), and growth differentiation factor 15 (GDF15) (P = 0.049) increased significantly in the training group in comparison to the control group and Myostatin (MSTN) (P = 0.002) had a significant decrease. However, there was no significant difference in ASMM/m2 (P = 0.527), SMM/m2 (P = 0.621), or Body fat mass (P = 0.433) within or between groups. In addition, the TRX Suspension Training had a significant effect on the functional tests and improved gait speed (P = 0.037), chair stand (P = 0.016), and TUG (P = 0.016) as well as Handgrip strength (P = 0.035). CONCLUSION Our findings highlight the efficacy of TRX Suspension Training in enhancing the serum levels of muscle growth factors and functional capacities among elderly individuals with sarcopenia. Therefore, considering the ongoing COVID-19 pandemic, this protocol can prove beneficial for this demographic group. TRIAL REGISTRY Iranian Registry of Clinical Trials identifier: IRCT20230727058944N1, prospectively registered 20-09-2023, https://en.irct.ir/trial/71635.
Collapse
Affiliation(s)
- Sohrab Rezaei
- Exercise Physiology Department, Faculty of Sport Science, Allameh Tabataba'i University, Tehran, Iran
| | - Rasoul Eslami
- Exercise Physiology Department, Faculty of Sport Science, Allameh Tabataba'i University, Tehran, Iran.
| | - Bakhtyar Tartibian
- Exercise Physiology Department, Faculty of Sport Science, Allameh Tabataba'i University, Tehran, Iran
| |
Collapse
|
18
|
Han X, Ashraf M, Tipparaju SM, Xuan W. Muscle-Brain crosstalk in cognitive impairment. Front Aging Neurosci 2023; 15:1221653. [PMID: 37577356 PMCID: PMC10413125 DOI: 10.3389/fnagi.2023.1221653] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/07/2023] [Indexed: 08/15/2023] Open
Abstract
Sarcopenia is an age-related, involuntary loss of skeletal muscle mass and strength. Alzheimer's disease (AD) is the most common cause of dementia in elderly adults. To date, no effective cures for sarcopenia and AD are available. Physical and cognitive impairments are two major causes of disability in the elderly population, which severely decrease their quality of life and increase their economic burden. Clinically, sarcopenia is strongly associated with AD. However, the underlying factors for this association remain unknown. Mechanistic studies on muscle-brain crosstalk during cognitive impairment might shed light on new insights and novel therapeutic approaches for combating cognitive decline and AD. In this review, we summarize the latest studies emphasizing the association between sarcopenia and cognitive impairment. The underlying mechanisms involved in muscle-brain crosstalk and the potential implications of such crosstalk are discussed. Finally, future directions for drug development to improve age-related cognitive impairment and AD-related cognitive dysfunction are also explored.
Collapse
Affiliation(s)
| | | | | | - Wanling Xuan
- Department of Pharmaceutical Sciences, USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| |
Collapse
|
19
|
Wang BYH, Hsiao AWT, Wong N, Chen YF, Lee CW, Lee WYW. Is dexamethasone-induced muscle atrophy an alternative model for naturally aged sarcopenia model? J Orthop Translat 2023; 39:12-20. [PMID: 36605620 PMCID: PMC9793312 DOI: 10.1016/j.jot.2022.11.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 11/10/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022] Open
Abstract
Background Primary sarcopenia is usually known as age-related skeletal muscle loss; however, other factors like endocrine, lifestyle and inflammation can also cause muscle loss, known as secondary sarcopenia. Although many studies have used different sarcopenia animal models for exploring the underlying mechanism and therapeutic approaches for sarcopenia, limited study has provided evidence of the relevance of these animal models. This study aims to investigate the similarity and difference in muscle qualities between primary and secondary sarcopenia mice models, using naturally aged mice and dexamethasone-induced mice. Methods 21-month-old mice were used as naturally aged primary sarcopenia mice and 3-month-old mice received daily intraperitoneal injection of dexamethasone (20 mg/ kg body weight) for 10 days were used as secondary sarcopenia model. This study provided measurements for muscle mass and functions, including Dual-energy X-ray absorptiometry (DXA) scanning, handgrip strength test and treadmill running to exhaustion test. Besides, muscle contraction, muscle fibre type measurements and gene expression were also performed to provide additional information on muscle qualities. Results The results suggest two sarcopenia animal models shared a comparable decrease in forelimb lean mass, muscle fibre size, grip strength and muscle contraction ability. Besides, the upregulation of protein degradation genes was also observed in two sarcopenia animal models. However, only primary sarcopenia mice were identified with an early stage of mtDNA deletion. Conclusion Collectively, this study evaluated that the dexamethasone-induced mouse model could be served as an alternative model for primary sarcopenia, according to the comparable muscle mass and functional changes. However, whether dexamethasone-induced mice can be used as an animal model when studying the molecular mechanisms of sarcopenia needs to be carefully evaluated. The translational potential of this article The purpose of sarcopenia research is to investigate appropriate treatments for reversing the loss of skeletal muscle mass and functions. Using animal models for the preclinical study could predict the safety and efficacy of the treatments. This study compared the typical age-related sarcopenia mice model and dexamethasone-induced secondary sarcopenia mice to provide evidence of the pathological and functional changes in the mice models.
Collapse
Affiliation(s)
- Belle Yu-Hsuan Wang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
- Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science Park, Hong Kong
| | - Allen Wei-Ting Hsiao
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Nicodemus Wong
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
- Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science Park, Hong Kong
| | - Yi-Fan Chen
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11529, Taiwan
- Graduate Institute of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, 11031 Taipei, Taiwan
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, 11031 Taipei, Taiwan
| | - Chien-Wei Lee
- Center for Translational Genomics & Regenerative Medicine Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
| | - Wayne Yuk Wai Lee
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
- Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science Park, Hong Kong
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
20
|
The Contribution of Tumor Derived Exosomes to Cancer Cachexia. Cells 2023; 12:cells12020292. [PMID: 36672227 PMCID: PMC9856599 DOI: 10.3390/cells12020292] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 01/13/2023] Open
Abstract
Cancer cachexia is defined as unintentional weight loss secondary to neoplasia and is associated with poor prognosis and outcomes. Cancer cachexia associated weight loss affects both lean tissue (i.e., skeletal muscle) and adipose tissue. Exosomes are extracellular vesicles that originate from multivesicular bodies that contain intentionally loaded biomolecular cargo. Exosome cargo includes proteins, lipids, mitochondrial components, and nucleic acids. The cargo carried in exosomes is thought to alter cell signaling when it enters into recipient cells. Virtually every cell type secretes exosomes and exosomes are known to be present in nearly every biofluid. Exosomes alter muscle and adipose tissue metabolism and biological processes, including macrophage polarization and apoptosis which contribute to the development of the cachexia phenotype. This has led to an interest in the role of tumor cell derived exosomes and their potential role as biomarkers of cancer cell development as well as their contribution to cachexia and disease progression. In this review, we highlight published findings that have studied the effects of tumor derived exosomes (and extracellular vesicles) and their cargo on the progression of cancer cachexia. We will focus on the direct effects of tumor derived exosomes and their cellular cross talk on skeletal muscle and adipose tissue, the primary sites of weight loss due to cancer cachexia.
Collapse
|
21
|
Chiariello A, Valente S, Pasquinelli G, Baracca A, Sgarbi G, Solaini G, Medici V, Fantini V, Poloni TE, Tognocchi M, Arcaro M, Galimberti D, Franceschi C, Capri M, Salvioli S, Conte M. The expression pattern of GDF15 in human brain changes during aging and in Alzheimer's disease. Front Aging Neurosci 2023; 14:1058665. [PMID: 36698863 PMCID: PMC9869280 DOI: 10.3389/fnagi.2022.1058665] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/08/2022] [Indexed: 01/11/2023] Open
Abstract
Introduction Growth Differentiation Factor 15 (GDF15) is a mitochondrial-stress-responsive molecule whose expression strongly increases with aging and age-related diseases. However, its role in neurodegenerative diseases, including Alzheimer's disease (AD), is still debated. Methods We have characterized the expression of GDF15 in brain samples from AD patients and non-demented subjects (controls) of different ages. Results Although no difference in CSF levels of GDF15 was found between AD patients and controls, GDF15 was expressed in different brain areas and seems to be predominantly localized in neurons. The ratio between its mature and precursor form was higher in the frontal cortex of AD patients compared to age-matched controls (p < 0.05). Moreover, this ratio was even higher for centenarians (p < 0.01), indicating that aging also affects GDF15 expression and maturation. A lower expression of OXPHOS complexes I, III, and V in AD patients compared to controls was also noticed, and a positive correlation between GDF15 and IL-6 mRNA levels was observed. Finally, when GDF15 was silenced in vitro in dermal fibroblasts, a decrease in OXPHOS complexes transcript levels and an increase in IL-6 levels were observed. Discussion Although GDF15 seems not to be a reliable CSF marker for AD, it is highly expressed in aging and AD brains, likely as a part of stress response aimed at counteracting mitochondrial dysfunction and neuroinflammation.
Collapse
Affiliation(s)
- Antonio Chiariello
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Sabrina Valente
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Gianandrea Pasquinelli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Alessandra Baracca
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, Bologna, Italy
| | - Gianluca Sgarbi
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, Bologna, Italy
| | - Giancarlo Solaini
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, Bologna, Italy
| | - Valentina Medici
- Department of Neurology and Neuropathology, Golgi-Cenci Foundation, Milan, Italy
| | - Valentina Fantini
- Department of Neurology and Neuropathology, Golgi-Cenci Foundation, Milan, Italy
| | - Tino Emanuele Poloni
- Department of Neurology and Neuropathology, Golgi-Cenci Foundation, Milan, Italy
| | - Monica Tognocchi
- Department of Agriculture, Food and Environment, University of Pisa, Pisa, Italy
| | - Marina Arcaro
- Fondazione Ca’ Granda IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Claudio Franceschi
- Department of Applied Mathematics of the Institute of ITMM, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Miriam Capri
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy,Interdepartmental Centre “Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate)”, University of Bologna, Bologna, Italy
| | - Stefano Salvioli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy,Interdepartmental Centre “Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate)”, University of Bologna, Bologna, Italy,*Correspondence: Stefano Salvioli, ✉
| | - Maria Conte
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy,Interdepartmental Centre “Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate)”, University of Bologna, Bologna, Italy
| |
Collapse
|
22
|
Chen Y, Hu Q, Wang C, Wang T. The crosstalk between BAT thermogenesis and skeletal muscle dysfunction. Front Physiol 2023; 14:1132830. [PMID: 37153220 PMCID: PMC10160478 DOI: 10.3389/fphys.2023.1132830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 04/11/2023] [Indexed: 05/09/2023] Open
Abstract
Metabolic defects increase the risk of skeletal muscle diseases, and muscle impairment might worsen metabolic disruption, leading to a vicious cycle. Both brown adipose tissue (BAT) and skeletal muscle play important roles in non-shivering thermogenesis to regulate energy homeostasis. BAT regulates body temperature, systemic metabolism, and seretion of batokines that have positive or negative impacts on skeletal muscle. Conversely, muscle can secrete myokines that regulate BAT function. This review explained the crosstalk between BAT and skeletal muscle, and then discussed the batokines and highlighted their impact on skeletal muscle under physiological conditions. BAT is now considered a potential therapeutic target for obesity and diabetes treatment. Moreover, manipulation of BAT may be an attractive approach for the treatment of muscle weakness by correcting metabolic deficits. Therefore, exploring BAT as a potential treatment for sarcopenia could be a promising avenue for future research.
Collapse
Affiliation(s)
- Yao Chen
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Hu
- Health Management Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Changyi Wang
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Changyi Wang, ; Tiantian Wang,
| | - Tiantian Wang
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Changyi Wang, ; Tiantian Wang,
| |
Collapse
|
23
|
He L, de Souto Barreto P, Sánchez Sánchez JL, Rolland Y, Guyonnet S, Parini A, Lucas A, Vellas B. Prospective Associations of Plasma Growth Differentiation Factor 15 With Physical Performance and Cognitive Functions in Older Adults. J Gerontol A Biol Sci Med Sci 2022; 77:2420-2428. [PMID: 35037034 DOI: 10.1093/gerona/glac020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Growth differentiation factor 15 (GDF15) has been associated with several age-related disorders, but its associations with functional abilities in community-dwelling older adults are not well studied. METHODS The study was a secondary analysis of 1 096 community-dwelling older adults (aged 69-94 years) recruited from the Multidomain Alzheimer's Preventive Trial. Plasma GDF15 was measured 1 year after participants' enrollment. Annual data of physical performance (grip strength and Short Physical Performance Battery [SPPB]) and global cognitive functions (Mini-Mental State Examination [MMSE] and a composite cognitive score) were measured for 4 years. Adjusted mixed-effects linear models were performed for cross-sectional and longitudinal association analyses. RESULTS A higher GDF15 was cross-sectionally associated with a weaker grip strength (β = -1.1E-03, 95% CI [-2.0E-03, -1.5E-04]), a lower SPPB score (β = -3.1E-04, 95% CI [-5.4E-04, -9.0E-05]), and worse cognitive functions (β = -2.4E-04, 95% CI [-3.3E-04, -1.6E-04] for composite cognitive score; β = -4.0E-04, 95% CI [-6.4E-04, -1.6E-04] for MMSE). Participants with higher GDF15 demonstrated greater longitudinal declines in SPPB (β = -1.0E-04, 95% CI [-1.7E-04, -2.0E-05]) and composite cognitive score (β = -2.0E-05, 95% CI [-4.0E-05, -3.6E-06]). The optimal initial GDF15 cutoff values for identifying participants with minimal clinically significant decline after 1 year were 2 189 pg/mL for SPPB (AUC: 0.580) and 2 330 pg/mL for composite cognitive score (AUC: 0.587). CONCLUSIONS Plasma GDF15 is cross-sectionally and longitudinally associated with lower-limb physical performance and global cognitive function in older adults. Circulating GDF15 alone has a limited capacity of discriminating older adults who will develop clinically significant functional declines. CLINICAL TRIAL REGISTRATION NCT00672685.
Collapse
Affiliation(s)
- Lingxiao He
- School of Public Health, Xiamen University, Xiamen, China.,Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalier-Universitaire de Toulouse, Toulouse, France
| | - Philipe de Souto Barreto
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalier-Universitaire de Toulouse, Toulouse, France.,CERPOP, INSERM 1295, Université de Toulouse, UPS, Toulouse, France
| | - Juan Luis Sánchez Sánchez
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalier-Universitaire de Toulouse, Toulouse, France.,Faculty of Sport Science, Universidad Europea de Madrid, Madrid, Spain
| | - Yves Rolland
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalier-Universitaire de Toulouse, Toulouse, France.,CERPOP, INSERM 1295, Université de Toulouse, UPS, Toulouse, France
| | - Sophie Guyonnet
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalier-Universitaire de Toulouse, Toulouse, France.,CERPOP, INSERM 1295, Université de Toulouse, UPS, Toulouse, France
| | - Angelo Parini
- Institute of Metabolic and Cardiovascular Diseases, UMR1297, Toulouse, France.,Paul Sabatier University, Toulouse, France
| | - Alexandre Lucas
- Institute of Metabolic and Cardiovascular Diseases, UMR1297, Toulouse, France.,Paul Sabatier University, Toulouse, France
| | - Bruno Vellas
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalier-Universitaire de Toulouse, Toulouse, France.,CERPOP, INSERM 1295, Université de Toulouse, UPS, Toulouse, France
| | | |
Collapse
|
24
|
Zha W, Sun Y, Gong W, Li L, Kim W, Li H. Ginseng and ginsenosides: Therapeutic potential for sarcopenia. Biomed Pharmacother 2022; 156:113876. [DOI: 10.1016/j.biopha.2022.113876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/05/2022] [Accepted: 10/13/2022] [Indexed: 11/02/2022] Open
|
25
|
Inflammaging: Implications in Sarcopenia. Int J Mol Sci 2022; 23:ijms232315039. [PMID: 36499366 PMCID: PMC9740553 DOI: 10.3390/ijms232315039] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
In a world in which life expectancy is increasing, understanding and promoting healthy aging becomes a contemporary demand. In the elderly, a sterile, chronic and low-grade systemic inflammation known as "inflammaging" is linked with many age-associated diseases. Considering sarcopenia as a loss of strength and mass of skeletal muscle related to aging, correlations between these two terms have been proposed. Better knowledge of the immune system players in skeletal muscle would help to elucidate their implications in sarcopenia. Characterizing the activators of damage sensors and the downstream effectors explains the inference with skeletal muscle performance. Sarcopenia has also been linked to chronic diseases such as diabetes, metabolic syndrome and obesity. Implications of inflammatory signals from these diseases negatively affect skeletal muscle. Autophagic mechanisms are closely related with the inflammasome, as autophagy eliminates stress signaling sent by damage organelles, but also acts with an immunomodulatory function affecting immune cells and cytokine release. The use of melatonin, an antioxidant, ROS scavenger and immune and autophagy modulator, or senotherapeutic compounds targeting senescent cells could represent strategies to counteract inflammation. This review aims to present the many factors regulating skeletal muscle inflammaging and their major implications in order to understand the molecular mechanisms involved in sarcopenia.
Collapse
|
26
|
Complementary combination of biomarkers for diagnosis of sarcopenia in C57BL/6J mice. Life Sci 2022; 312:121213. [PMID: 36423671 DOI: 10.1016/j.lfs.2022.121213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/14/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
AIMS The objective of this study is to provide a reliable strategy for the diagnosis of sarcopenia based on a complementary combination of biomarkers from various approaches. MATERIAL AND METHODS A total of 30 C57BL/6J mice were used for the experiment, in which 15 young mice (YM) at 24 weeks old and 15 aged mice (AM) at 88 weeks old. Extracted features-based digital biomarkers from the electromyography activity of tibialis anterior muscles were evaluated by using receiver operating characteristic analysis. Extracted tissular proteins and circulating hormones based chemical biomarkers were investigated by using immunoblotting and enzyme-linked immunosorbent assay. KEY FINDINGS In terms of digital biomarkers, the feature-based classification of mice groups showed good performance (Feature A: AUC = 0.986, accuracy = 0.928) and (Feature B: AUC = 0.999, accuracy = 0.990). On the other hand, muscle-specific protein levels based chemical biomarkers (e.g. MuRF1, FoxO1, and perilipin2) were observed significantly increase with age. Pro-inflammatory cytokines based biomarkers extracted from muscle tissue and circulating plasma (e.g. TNF-α, IL-6, and IL-8) were significantly higher in case of AM group compared to YM group. Circulating hormone-based chemical biomarkers (e.g. cortisol/DHEA ratio and cathepsin D) presented a significant increase in concentrations with age. Circulating neurotransmitter based biomarkers (e.g. acetylcholine, serotonin, and histamine) also increased significantly in concentrations from YM to AM. SIGNIFICANCE A complementary combination of digital and chemical biomarkers covers multiple domains of sarcopenia to provide an effective strategy for the early diagnosis of sarcopenia.
Collapse
|
27
|
De Paepe B. The Cytokine Growth Differentiation Factor-15 and Skeletal Muscle Health: Portrait of an Emerging Widely Applicable Disease Biomarker. Int J Mol Sci 2022; 23:ijms232113180. [PMID: 36361969 PMCID: PMC9654287 DOI: 10.3390/ijms232113180] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/21/2022] [Accepted: 10/26/2022] [Indexed: 12/04/2022] Open
Abstract
Growth differentiation factor 15 (GDF-15) is a stress-induced transforming growth factor-β superfamily cytokine with versatile functions in human health. Elevated GDF-15 blood levels associate with multiple pathological conditions, and are currently extensively explored for diagnosis, and as a means to monitor disease progression and evaluate therapeutic responses. This review analyzes GDF-15 in human conditions specifically focusing on its association with muscle manifestations of sarcopenia, mitochondrial myopathy, and autoimmune and viral myositis. The use of GDF-15 as a widely applicable health biomarker to monitor muscle disease is discussed, and its potential as a therapeutic target is explored.
Collapse
Affiliation(s)
- Boel De Paepe
- Neuromuscular Reference Center, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| |
Collapse
|
28
|
Wang T. Searching for the link between inflammaging and sarcopenia. Ageing Res Rev 2022; 77:101611. [PMID: 35307560 DOI: 10.1016/j.arr.2022.101611] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/14/2022] [Accepted: 03/15/2022] [Indexed: 12/17/2022]
Affiliation(s)
- Tiantian Wang
- Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
29
|
Zhang W, Sun W, Gu X, Miao C, Feng L, Shen Q, Liu X, Zhang X. GDF-15 in tumor-derived exosomes promotes muscle atrophy via Bcl-2/caspase-3 pathway. Cell Death Dis 2022; 8:162. [PMID: 35379793 PMCID: PMC8980041 DOI: 10.1038/s41420-022-00972-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 03/04/2022] [Accepted: 03/23/2022] [Indexed: 12/22/2022]
Abstract
Tumor-derived exosomes are emerging mediators of cancer cachexia, a kind of multifactorial syndrome characterized by serious loss of skeletal muscle mass and function. Our previous study had showed that microRNAs in exosomes of C26 colon tumor cells were involved in induction of muscle atrophy. Here, we focus on studying proteins in tumor-derived exosomes which might also contribute to the development of cancer cachexia. Results of comparing the protein profiles of cachexic C26 exosomes and non-cachexic MC38 exosomes suggested that growth differentiation factor 15 (GDF-15) was rich in C26 exosomes. Western blotting analysis confirmed the higher levels of GDF-15 in C26 cells and C26 exosomes, compared with that of MC38 cells. Results of animal study also showed that GDF-15 was rich in tumor tissues, serum exosomes, and gastrocnemius (GA) muscle tissues of C26 tumor-bearing mice. GDF-15 protein could directly induce muscle atrophy of cultured C2C12 myotubes via regulating Bcl-2/caspase-3 pathways. What’s more, overexpression of GDF-15 in MC38 cells could increase the potency of MC38 conditioned medium or exosomes in inducing muscle atrophy. Knockdown of GDF-15 in C26 cells decreased the potency of C26 conditioned medium or exosomes in inducing muscle atrophy. These results suggested that GDF-15 in tumor-derived exosomes could contribute to induction of muscle atrophy and also supported the possibility of targeting GDF-15 in treatment of cancer cachexia.
Collapse
Affiliation(s)
- Wanli Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Weikuan Sun
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Xiaofan Gu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Chunxiao Miao
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Lixing Feng
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Qiang Shen
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Xuan Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xiongwen Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China.
| |
Collapse
|
30
|
Conte M, Giuliani C, Chiariello A, Iannuzzi V, Franceschi C, Salvioli S. GDF15, an emerging key player in human aging. Ageing Res Rev 2022; 75:101569. [PMID: 35051643 DOI: 10.1016/j.arr.2022.101569] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/14/2022] [Indexed: 12/20/2022]
Abstract
Growth differentiation factor 15 (GDF15) is recently emerging not only as a stress-related mitokine, but also as a key player in the aging process, being one of the most up-regulated protein with age and associated with a variety of age-related diseases (ARDs). Many data indicate that GDF15 has protective roles in several tissues during different stress and aging, thus playing a beneficial role in apparent contrast with the observed association with many ARDs. A possible detrimental role for this protein is then hypothesized to emerge with age. Therefore, GDF15 can be considered as a pleiotropic factor with beneficial activities that can turn detrimental in old age possibly when it is chronically elevated. In this review, we summarize the current knowledge on the biology of GDF15 during aging. We also propose GDF15 as a part of a dormancy program, where it may play a role as a mediator of defense processes aimed to protect from inflammatory damage and other stresses, according to the life history theory.
Collapse
Affiliation(s)
- Maria Conte
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy; Interdepartmental Centre "Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate)", University of Bologna, Bologna, Italy.
| | - Cristina Giuliani
- Interdepartmental Centre "Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate)", University of Bologna, Bologna, Italy; Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Antonio Chiariello
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Vincenzo Iannuzzi
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Claudio Franceschi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy; Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky University, Nizhniy Novgorod, Russia
| | - Stefano Salvioli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy; Interdepartmental Centre "Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate)", University of Bologna, Bologna, Italy
| |
Collapse
|
31
|
Lee SH, Lee JY, Lim KH, Lee YS, Koh JM. Associations Between Plasma Growth and Differentiation Factor-15 with Aging Phenotypes in Muscle, Adipose Tissue, and Bone. Calcif Tissue Int 2022; 110:236-243. [PMID: 34499185 DOI: 10.1007/s00223-021-00912-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 08/27/2021] [Indexed: 02/04/2023]
Abstract
Growth and differentiation factor 15 (GDF-15) is associated with muscle, fat, and bone metabolism; however, this association has not been well characterized. Plasma GDF-15, appendicular skeletal muscle mass (ASM), fat mass (FM), and bone mineral density (BMD) were measured in 146 postmenopausal women. GDF-15 levels were higher in subjects with low Body Mass Index (BMI)-adjusted ASM than in those without (median [interquartile range] 831.3 [635.4-1011.4] vs. 583.8 [455.8-771.1] pg/mL, p = 0.018). The GDF-15 level was inversely correlated with BMI-adjusted ASM (r = - 0.377, p < 0.001) and BMD at femur neck (FN-BMD; r = - 0.201, p = 0.015), and positively correlated with percent FM (pFM; r = 0.328, p < 0.001). After adjusting for confounders, the GDF-15 level was inversely associated with BMI-adjusted ASM (β = -0.250, p = 0.006) and positively associated with pFM (β = 0.272, p = 0.004), and tended to be inversely associated with FN-BMD (β = - 0.176, p = 0.076). The area under the receiver-operating characteristic curve of GDF-15 level > 618.4 pg/mL for sarcopenia was 0.706 (95% confidence interval (CI) 0.625-0.779) with a sensitivity of 83.3% and a specificity of 54.5%. Using a GDF-15 level of 618.4 pg/mL as a cut-off, the GDF-15 level was associated with a significantly greater likelihood of sarcopenia (odds ratio [OR] 2.35; 95% CI 1.00-5.51; p = 0.049), obesity (OR 3.28; 95% CI 1.48-7.27; p = 0.001), osteopenic obesity (OR 3.10; 95% CI 1.31-7.30; p = 0.010), and sarcopenic or osteosarcopenic obesity (OR 4.84; 95% CI 0.88-26.69; p = 0.070). These findings support the potential of GDF-15 as a biomarker for age-related changes in muscle, fat, and bone.
Collapse
Affiliation(s)
- Seung Hun Lee
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| | - Jee Yang Lee
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Kyeong-Hye Lim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Young-Sun Lee
- Asan Institute for Life Sciences, Seoul, 05505, Republic of Korea
| | - Jung-Min Koh
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| |
Collapse
|
32
|
Sanchez-Sanchez JL, He L, Virecoulon Giudici K, Guyonnet S, Parini A, Dray C, Valet P, Pereira O, Vellas B, Rolland Y, de Souto Barreto P. Circulating Levels of Apelin, GDF-15 and Sarcopenia: Lack of Association in the MAPT Study. J Nutr Health Aging 2022; 26:564-570. [PMID: 35718864 DOI: 10.1007/s12603-022-1800-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVES Apelin and GDF-15 have been proposed as biomarkers of age-related sarcopenia but evidence in human models is scarce. This study aimed to explore the associations between blood apelin and GDF-15 with sarcopenia incidence and the evolution of sarcopenia components over two years in older adults >70 years. DESIGN Secondary longitudinal analysis of the Multidomain Alzheimer Preventive Trial. PARTICIPANTS Older adults (>70 years) attending primary care centers in France and Monaco. SETTING Community. MEASUREMENTS Serum Apelin (pg/mL) and plasma GDF-15 (pg/mL) were measured. Outcomes included sarcopenia defined by the European Working Group on Sarcopenia in Older People (EWGSOP) and its determinants (appendicular lean mass [ALM] evaluated through a Dual-energy X-ray Absorptiometry (DXA) scan, handgrip strength (HGS) and the 4-meter gait speed) measured over 2 years. Linear mixed models and logistic regression were used to explore the longitudinal associations. RESULTS We included 168 subjects from MAPT (median age=76y, IQR=73-79; 78% women). Serum apelin was not significantly associated with sarcopenia incidence (OR=1.001;95%CI=1.000,1.001;p-value>0.05 in full-adjusted models) nor with ALM (β=-5.8E-05;95%CI=-1.0E-04,2.12E-04;p>0.05), HGS (β=-1.1E-04;95%CI=-5.0E-04,2.8E-04;p>0.05), and GS (β=-5.1E-06;95%CI=-1.0E-05,2.0E-05;p>0.05) in fully adjusted models. Similarly, plasma GDF-15 was not associated with both the incidence of sarcopenia (OR=1.001,95%CI=1.000,1.002,p>0.05) and the evolution of its determinants ([ALM, β=2.1E-05;95%CI=-2.6E-04,3.03E-04;p>0.05], HGS [β=-5.9E-04;95%CI=-1.26E-03,8.1E-05; p>0.05] nor GS [β=-2.6E-06;95%CI=-3.0E-05, 2.3E-05;p>0.05]) in fully adjusted models. CONCLUSIONS Blood apelin and GDF-15 were not associated with sarcopenia incidence or with the evolution of sarcopenia components over a 2-year follow-up in community-dwelling older adults. Well-powered longitudinal studies are needed to confirm or refute our findings.
Collapse
Affiliation(s)
- J L Sanchez-Sanchez
- Juan Luis Sánchez, Gérontopôle de Toulouse, Institut du Vieillissement, 37 Allées Jules Guesde, 31000 Toulouse, France, +34662309412,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Hong SW, Kang JH. Growth differentiation factor-15 as a modulator of bone and muscle metabolism. Front Endocrinol (Lausanne) 2022; 13:948176. [PMID: 36325442 PMCID: PMC9618662 DOI: 10.3389/fendo.2022.948176] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
This study aims to clarify the potential role of growth differentiation factor-15 (GDF-15) as a myokine in bone metabolism and muscle function in females with osteoporosis. In total, 45 female participants (71.0 ± 8.5 years) with distal radius fractures were recruited. Participants were classified as healthy/osteopenic (n = 28) (CON) or osteoporotic (n = 17) (OP) according to their T-score from the areal bone mineral density (aBMD) of the femoral neck. Body mass index, upper arm and calf circumferences, and handgrip strength were assessed. Total hip, femoral neck, and lumbar spine aBMD was measured via dual-energy x-ray absorptiometry. The focal bone quality of the distal radius was evaluated via 3D reconstructed computed tomographic images. Serum levels of GDF-15, insulin-like growth factor-1, and inflammatory markers such as tumor necrosis factor-α (TNF-α), interleukin-6, and interleukin-1β (IL-1β), as well as the corresponding mRNA levels in the pronator quadratus muscle were determined. Participants in the OP group had higher serum GDF-15 levels than those in the CON group. The mRNA levels of GDF-15, IL-1β, and TNF-α in the pronator quadratus muscle were significantly higher in the OP group than in the CON one. Levels of both serum GDF-15 and GDF-15 mRNA in muscle were positively correlated with age and negatively associated with the aBMD of the total hip and focal bone quality of the distal radius. Handgrip power was not correlated with circulating GDF-15 levels but was correlated with circumferences of the upper arm and calf, and levels of GDF-15 mRNA in muscle specimens. The mRNA levels of GDF-15 were correlated with those of inflammatory cytokines such as TNF-α and IL-1β. The mRNA levels of TNF-α were associated with circumferences of the upper arm and calf and with the aBMD of the total hip. The mRNA levels of GDF-15 in muscle were correlated with serum levels of GDF-15 and TNF-α. GDF-15 may have associations with bone metabolism in humans via paracrinological and endocrinological mechanisms. Maintenance of muscle mass and function would be influenced more by GDF-15 in muscle than by circulating GDF-15. The role of GDF-15 in bone metabolism and muscle homeostasis could be related to inflammatory responses.
Collapse
Affiliation(s)
- Seok Woo Hong
- Department of Orthopedic Surgery, Kangbuk Samsung Hospital, School of Medicine, Sungkyunkwan University, Seoul, South Korea
| | - Jeong-Hyun Kang
- Clinic of Oral Medicine and Orofacial Pain, Institute of Oral Health Science, School of Medicine, Ajou University, Suwon, South Korea
- *Correspondence: Jeong-Hyun Kang,
| |
Collapse
|
34
|
Liu D, Fan YB, Tao XH, Pan WL, Wu YX, Wang XH, He YQ, Xiao WF, Li YS. Mitochondrial Quality Control in Sarcopenia: Updated Overview of Mechanisms and Interventions. Aging Dis 2021; 12:2016-2030. [PMID: 34881083 PMCID: PMC8612607 DOI: 10.14336/ad.2021.0427] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/27/2021] [Indexed: 12/22/2022] Open
Abstract
Sarcopenia is a common geriatric disorder characterized by decreased muscle strength, low muscle mass and poor physical performance. This aging-related skeletal muscle deterioration leads to adverse outcomes and severely impairs the quality of life of patients. The accumulation of dysfunctional mitochondria with aging is an important factor in the occurrence and progression of sarcopenia. Mitochondrial quality control (MQC) fundamentally ensures the normal mitochondrial functions and is comprised of four main parts: proteostasis, biogenesis, dynamics and autophagy. Therefore, any pathophysiologic factors compromising the quality control of homeostasis in the skeletal muscle may lead to sarcopenia. However, the specific theoretical aspects of these processes have not been fully elucidated. Current therapeutic interventions using nutritional and pharmaceutical treatments show a modest therapeutic efficacy; however, only physical exercise is recommended as the first-line therapy for sarcopenia, which can ameliorate skeletal muscle deficiency by maintaining the homeostatic MQC. In this review, we summarized the known mechanisms that contribute to the pathogenesis of sarcopenia by impairing normal mitochondrial functions and described potential interventions that mitigate sarcopenia through improving MQC.
Collapse
Affiliation(s)
- Di Liu
- 1Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yi-Bin Fan
- 2Department of Dermatology, Zhejiang provincial people's hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Xiao-Hua Tao
- 2Department of Dermatology, Zhejiang provincial people's hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Wei-Li Pan
- 2Department of Dermatology, Zhejiang provincial people's hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Yu-Xiang Wu
- 3School of Kinesiology, Jianghan University, Wuhan 430056, China
| | - Xiu-Hua Wang
- 4Xiang Ya Nursing School, The Central South University, Changsha 410013, China
| | - Yu-Qiong He
- 1Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Wen-Feng Xiao
- 1Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.,5National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yu-Sheng Li
- 1Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.,5National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| |
Collapse
|
35
|
Distinct roles of UVRAG and EGFR signaling in skeletal muscle homeostasis. Mol Metab 2021; 47:101185. [PMID: 33561544 PMCID: PMC7921879 DOI: 10.1016/j.molmet.2021.101185] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 11/20/2022] Open
Abstract
Objective Autophagy is a physiological self-eating process that can promote cell survival or activate cell death in eukaryotic cells. In skeletal muscle, it is important for maintaining muscle mass and function that is critical to sustain mobility and regulate metabolism. The UV radiation resistance-associated gene (UVRAG) regulates the early stages of autophagy and autophagosome maturation and plays a key role in endosomal trafficking. This study investigated the essential in vivo role of UVRAG in skeletal muscle biology. Methods To determine the role of UVRAG in skeletal muscle in vivo, we generated muscle-specific UVRAG knockout mice using the Cre-loxP system driven by Myf6 promoter that is exclusively expressed in skeletal muscle. Myf6-Cre+ UVRAGfl/fl (M-UVRAG−/−) mice were compared to littermate Myf6-Cre+ UVRAG+/+ (M-UVRAG+/+) controls under basal conditions on a normal chow diet. Body composition, muscle function, and mitochondria morphology were assessed in muscles of the WT and KO mice at 24 weeks of age. Results M-UVRAG−/− mice developed accelerated sarcopenia and impaired muscle function compared to M-UVRAG+/+ littermates at 24 weeks of age. Interestingly, these mice displayed improved glucose tolerance and increased energy expenditure likely related to upregulated Fgf21, a marker of muscle dysfunction. Skeletal muscle of the M-UVRAG−/− mice showed altered mitochondrial morphology with increased mitochondrial fission and EGFR accumulation reflecting defects in endosomal trafficking. To determine whether increased EGFR signaling had a causal role in muscle dysfunction, the mice were treated with an EGFR inhibitor, gefitinib, which partially restored markers of muscle and mitochondrial deregulation. Conversely, constitutively active EGFR transgenic expression in UVRAG-deficient muscle led to further detrimental effects with non-overlapping distinct defects in muscle function, with EGFR activation affecting the muscle fiber type whereas UVRAG deficiency impaired mitochondrial homeostasis. Conclusions Our results show that both UVRAG and EGFR signaling are critical for maintaining muscle mass and function with distinct mechanisms in the differentiation pathway. Deletion of UVRAG in skeletal muscle accelerates muscle wasting with aging. UVRAG in skeletal muscle regulates mitochondrial dynamics and function. UVRAG deletion leads to EGFR accumulation in skeletal muscle. Constitutively active EGFR contributes to muscle fiber type determination.
Collapse
|