1
|
Kirk B, Lombardi G, Duque G. Bone and muscle crosstalk in ageing and disease. Nat Rev Endocrinol 2025; 21:375-390. [PMID: 40011751 DOI: 10.1038/s41574-025-01088-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/30/2025] [Indexed: 02/28/2025]
Abstract
Interorgan communication between bone and skeletal muscle is central to human health. A dysregulation of bone-muscle crosstalk is implicated in several age-related diseases. Ageing-associated changes in endocrine, inflammatory, nutritional and biomechanical stimuli can influence the differentiation capacity, function and survival of mesenchymal stem cells and bone-forming and muscle-forming cells. Consequently, the secretome phenotype of bone and muscle cells is altered, leading to impaired crosstalk and, ultimately, catabolism of both tissues. Adipose tissue acts as a third player in the bone-muscle interaction by secreting factors that affect bone and muscle cells. Physical exercise remains the key biological stimulus for bone-muscle crosstalk, either directly via the release of cytokines from bone, muscle or adipocytes, or indirectly through extracellular vesicles. Overall, bone-muscle crosstalk is considered an inherent process necessary to maintain the structure and function of both tissues across the life cycle. This Review summarizes the latest biomedical advances in bone-muscle crosstalk as it pertains to human ageing and disease. We also outline future research priorities to accommodate the understanding of this rapidly emerging field.
Collapse
Affiliation(s)
- Ben Kirk
- Department of Medicine, Western Health, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, Melbourne, Victoria, Australia
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry & Advanced Diagnostics, IRCCS Ospedale Galeazzi-Sant'Ambrogio, Milan, Italy
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| | - Gustavo Duque
- Department of Medicine, Western Health, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia.
- Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, Melbourne, Victoria, Australia.
- Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
- Dr. Joseph Kaufmann Chair in Geriatric Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
2
|
Chen G, Chen L, Li X, Mohammadi M. FGF-based drug discovery: advances and challenges. Nat Rev Drug Discov 2025; 24:335-357. [PMID: 39875570 DOI: 10.1038/s41573-024-01125-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2024] [Indexed: 01/30/2025]
Abstract
The fibroblast growth factor (FGF) family comprises 15 paracrine-acting and 3 endocrine-acting polypeptides, which govern a multitude of processes in human development, metabolism and tissue homeostasis. Therapeutic endocrine FGFs have recently advanced in clinical trials, with FGF19 and FGF21-based therapies on the cusp of approval for the treatment of primary sclerosing cholangitis and metabolic syndrome-associated steatohepatitis, respectively. By contrast, while paracrine FGFs were once thought to be promising drug candidates for wound healing, burns, tissue repair and ischaemic ailments based on their potent mitogenic and angiogenic properties, repeated failures in clinical trials have led to the widespread perception that the development of paracrine FGF-based drugs is not feasible. However, the observation that paracrine FGFs can exert FGF hormone-like metabolic activities has restored interest in these FGFs. The recent structural elucidation of the FGF cell surface signalling machinery and the formulation of a new threshold model for FGF signalling specificity have paved the way for therapeutically harnessing paracrine FGFs for the treatment of a range of metabolic diseases.
Collapse
Affiliation(s)
- Gaozhi Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lingfeng Chen
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Moosa Mohammadi
- Institute of Cell Growth Factor, Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health, Wenzhou, Zhejiang, China.
| |
Collapse
|
3
|
Huang ZL, Zhang SB, Xu SF, Gu XN, Wu ZQ, Zhang Y, Li J, Ji LL. TSG attenuated NAFLD and facilitated weight loss in HFD-fed mice via activating the RUNX1/FGF21 signaling axis. Acta Pharmacol Sin 2025:10.1038/s41401-025-01568-w. [PMID: 40307458 DOI: 10.1038/s41401-025-01568-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/14/2025] [Indexed: 05/02/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic liver disease characterized by steatosis in hepatocytes and is now becoming the major cause of liver-related mortality. Fibroblast growth factor 21 (FGF21) is an endocrine hormone mainly secreted by the liver, which can bind to its receptor (FGFR) and co-receptor beta klotho (KLB) to form a receptor complex, exerting its lipid-lowering function. 2,3,5,4'-Tetrahydroxy-stilbene-2-O-β-D-glucoside (TSG), a natural compound isolated from Polygonum multiflorum Thunb, has shown excellent activity in lowering lipid content and efficacy in improving NAFLD. In this study we investigated whether FGF21 was implicated in the therapeutic effect of TSG in NAFLD mice. NAFLD was induced in mice by feeding with a high-fat diet (HFD) for 12 weeks, and treated with TSG (20, 40 mg·kg-1·d-1, i.g.) during the last 4 weeks. We showed that TSG treatment significantly alleviated NAFLD in HFD-fed mice evidenced by reduced hepatic triglyceride (TG) and non-esterified fatty acids (NEFA), diminished lipid droplets and decreased NAFLD activity score (NAS) in liver tissues. We demonstrated that TSG treatment significantly increased the mRNA and protein levels of FGF21 in vitro and in vivo, and reduced lipid accumulation in both the liver and adipose tissues. Transcriptomics analysis revealed that TSG treatment significantly increased the nuclear translocation of a transcription factor RUNX1. Knockdown of Runx1 in HFD-fed mice eliminated the efficacy of TSG in alleviating NAFLD, reducing hepatic lipid accumulation and regulating FGF21 signaling pathway in liver and adipose tissues. In conclusion, TSG alleviates NAFLD by enhancing the FGF21-mediated lipid metabolism in a RUNX1-dependent manner.
Collapse
Affiliation(s)
- Zhen-Lin Huang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shao-Bo Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shang-Fu Xu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - Xin-Nan Gu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ze-Qi Wu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yue Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jian Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
- Technology Center of Jinling Pharmaceutical Co., Ltd, Nanjing, 210009, China
| | - Li-Li Ji
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
4
|
Scudese E, Marshall AG, Vue Z, Exil V, Rodriguez BI, Demirci M, Vang L, López EG, Neikirk K, Shao B, Le H, Stephens D, Hall DD, Rostami R, Rodman T, Kabugi K, Shao JQ, Mungai M, AshShareef ST, Hicsasmaz I, Manus S, Wanjalla CN, Whiteside A, Dasari R, Williams CR, Damo SM, Gaddy JA, Glancy B, Dantas EHM, Kinder A, Kadam A, Tomar D, Scartoni F, Baffi M, McReynolds MR, Phillips MA, Cooper A, Murray SA, Quintana AM, Wandira N, Ochayi OM, Ameka M, Kirabo A, Masenga SK, Harris C, Oliver A, Martin P, Gaye A, Korolkova O, Sharma V, Mobley BC, Katti P, Hinton A. 3D Mitochondrial Structure in Aging Human Skeletal Muscle: Insights Into MFN-2-Mediated Changes. Aging Cell 2025:e70054. [PMID: 40285369 DOI: 10.1111/acel.70054] [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: 12/14/2023] [Revised: 02/21/2025] [Accepted: 03/13/2025] [Indexed: 04/29/2025] Open
Abstract
Age-related skeletal muscle atrophy, known as sarcopenia, is characterized by loss of muscle mass, strength, endurance, and oxidative capacity. Although exercise has been shown to mitigate sarcopenia, the underlying governing mechanisms are poorly understood. Mitochondrial dysfunction is implicated in aging and sarcopenia; however, few studies explore how mitochondrial structure contributes to this dysfunction. In this study, we sought to understand how aging impacts mitochondrial three-dimensional (3D) structure and its regulators in skeletal muscle. We hypothesized that aging leads to remodeling of mitochondrial 3D architecture permissive to dysfunction and is ameliorated by exercise. Using serial block-face scanning electron microscopy (SBF-SEM) and Amira software, mitochondrial 3D reconstructions from patient biopsies were generated and analyzed. Across five human cohorts, we correlate differences in magnetic resonance imaging, mitochondria 3D structure, exercise parameters, and plasma immune markers between young (under 50 years) and old (over 50 years) individuals. We found that mitochondria are less spherical and more complex, indicating age-related declines in contact site capacity. Additionally, aged samples showed a larger volume phenotype in both female and male humans, indicating potential mitochondrial swelling. Concomitantly, muscle area, exercise capacity, and mitochondrial dynamic proteins showed age-related losses. Exercise stimulation restored mitofusin 2 (MFN2), one such of these mitochondrial dynamic proteins, which we show is required for the integrity of mitochondrial structure. Furthermore, we show that this pathway is evolutionarily conserved, as Marf, the MFN2 ortholog in Drosophila, knockdown alters mitochondrial morphology and leads to the downregulation of genes regulating mitochondrial processes. Our results define age-related structural changes in mitochondria and further suggest that exercise may mitigate age-related structural decline through modulation of mitofusin 2.
Collapse
Affiliation(s)
- Estevão Scudese
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
- Sport Sciences and Exercise Laboratory (LaCEE), Catholic University of Petrópolis (UCP), Brazil
| | - Andrea G Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Vernat Exil
- Department of Pediatrics, Div. of Cardiology, St. Louis University School of Medicine, St. Louis, MO, USA
| | - Benjamin I Rodriguez
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Mert Demirci
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Edgar Garza López
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Bryanna Shao
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Han Le
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Dominique Stephens
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Duane D Hall
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Rahmati Rostami
- Department of Genetic Medicine, Joan & Sanford I. Weill Medical College of Cornell University, New York, NY, USA
| | - Taylor Rodman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Kinuthia Kabugi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | | | - Margaret Mungai
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | | | - Innes Hicsasmaz
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Sasha Manus
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Celestine N Wanjalla
- Division of Infection Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Aaron Whiteside
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, USA
| | - Revathi Dasari
- Department of Biology, Indian Institute of Science Education and Research (IISER), Tirupati, AP, India
| | - Clintoria R Williams
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, USA
| | - Steven M Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, TN, USA
| | - Jennifer A Gaddy
- Division of Infection Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Valley Healthcare Systems, U.S. Department of Veterans Affairs, Nashville, TN, USA
| | - Brian Glancy
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- NIAMS, NIH, Bethesda, MD, USA
| | - Estélio Henrique Martin Dantas
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
- Doctor's Degree Program in Nursing and Biosciences - PpgEnfBio, Federal University of the State of Rio de Janeiro - UNIRIO, Rio de Janeiro, RJ, Brazil
- Laboratory of Human Motricity Biosciences - LABIMH, Federal University of the State of Rio de Janeiro - UNIRIO, RJ, Brazil
- Brazilian Paralympic Academy - APB, Brazil
- Doctor's Degree Program in Health and Environment - PSA, Tiradentes University - UNIT, Aracaju, SE, Brazil
| | - André Kinder
- Artur Sá Earp Neto University Center - UNIFASE-FMP, Petrópolis Medical School, Brazil
| | - Ashlesha Kadam
- Department of Internal Medicine, Section of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Dhanendra Tomar
- Department of Internal Medicine, Section of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Fabiana Scartoni
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | - Matheus Baffi
- Sport Sciences and Exercise Laboratory (LaCEE), Catholic University of Petrópolis (UCP), Brazil
| | - Melanie R McReynolds
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA, USA
| | - Mark A Phillips
- Department of Integrative Biology, Oregon State University, Corvallis, OR, USA
| | - Anthonya Cooper
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sandra A Murray
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anita M Quintana
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, Texas, USA
| | - Nelson Wandira
- Institute of Health Sciences Busoga University, Iganga, Uganda
| | - Okwute M Ochayi
- Department of Human Physiology, Baze University, Abuja, Nigeria
| | - Magdalene Ameka
- KAVI Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
| | - Annet Kirabo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Sepiso K Masenga
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Physiological Sciences, School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia
| | - Chanel Harris
- Department of Biomedical Sciences, Meharry Medical College, Nashville, US
| | - Ashton Oliver
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Sciences, Meharry Medical College, Nashville, US
| | - Pamela Martin
- Department of Biomedical Sciences, Meharry Medical College, Nashville, US
| | - Amadou Gaye
- Department of Integrative Genomics and Epidemiology, Meharry Medical College, Nashville, TN, USA
| | - Olga Korolkova
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN, USA
| | - Vineeta Sharma
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN, USA
| | - Bret C Mobley
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Prasanna Katti
- Department of Biology, Indian Institute of Science Education and Research (IISER), Tirupati, AP, India
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
5
|
Iwasa M, Uemura K, Soufi M, Otake Y, Kinoshita T, Kutsuna T, Takashima K, Hamada H, Sato Y, Sugano N, Okada S, Takao M. Volume and quality of the gluteal muscles are associated with early physical function after total hip arthroplasty. Int J Comput Assist Radiol Surg 2025; 20:703-711. [PMID: 39836355 PMCID: PMC12034588 DOI: 10.1007/s11548-025-03321-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
PURPOSE Identifying muscles linked to postoperative physical function can guide protocols to enhance early recovery following total hip arthroplasty (THA). This study aimed to evaluate the association of preoperative pelvic and thigh muscle volume and quality with early physical function after THA in patients with unilateral hip osteoarthritis (HOA). METHODS Preoperative Computed tomography (CT) images of 61 patients (eight males and 53 females) with HOA were analyzed. Six muscle groups were segmented from CT images, and muscle volume and quality were calculated on the healthy and affected sides. Muscle quality was quantified using the mean CT values (Hounsfield units [HU]). Early postoperative physical function was evaluated using the Timed Up & Go test (TUG) at three weeks after THA. The effect of preoperative muscle volume and quality of both sides on early postoperative physical function was assessed. RESULTS On the healthy and affected sides, mean muscle mass was 9.7 cm3/kg and 8.1 cm3/kg, and mean muscle HU values were 46.0 HU and 39.1 HU, respectively. Significant differences in muscle volume and quality were observed between the affected and healthy sides. On analyzing the function of various muscle groups, the TUG score showed a significant association with the gluteus maximum volume and the gluteus medius/minimus quality on the affected side. CONCLUSION Patients with HOA showed significant muscle atrophy and fatty degeneration in the affected pelvic and thigh regions. The gluteus maximum volume and gluteus medius/minimus quality were associated with early postoperative physical function. Preoperative rehabilitation targeting the gluteal muscles on the affected side could potentially enhance recovery of physical function in the early postoperative period.
Collapse
Affiliation(s)
- Makoto Iwasa
- Department of Orthopaedic Medical Engineering, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Keisuke Uemura
- Department of Orthopaedic Medical Engineering, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Mazen Soufi
- Division of Information Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
| | - Yoshito Otake
- Division of Information Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
| | - Tomofumi Kinoshita
- Department of Orthopaedic Surgery, Ehime University Graduate School of Medicine, Matsuyama, Japan
| | - Tatsuhiko Kutsuna
- Department of Orthopaedic Surgery, Ehime University Graduate School of Medicine, Matsuyama, Japan
| | - Kazuma Takashima
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hidetoshi Hamada
- Department of Orthopaedic Medical Engineering, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshinobu Sato
- Division of Information Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
| | - Nobuhiko Sugano
- Department of Orthopaedic Medical Engineering, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Seiji Okada
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Masaki Takao
- Department of Orthopaedic Surgery, Ehime University Graduate School of Medicine, Matsuyama, Japan
| |
Collapse
|
6
|
Ramírez-Casas Y, Fernández-Martínez J, Martín-Estebané M, Aranda-Martínez P, López-Rodríguez A, Esquivel-Ruiz S, Yang Y, Escames G, Acuña-Castroviejo D. Melatonin and Exercise Restore Myogenesis and Mitochondrial Dynamics Deficits Associated With Sarcopenia in iMS-Bmal1 -/- Mice. J Pineal Res 2025; 77:e70049. [PMID: 40241474 DOI: 10.1111/jpi.70049] [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: 01/26/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025]
Abstract
Sarcopenia, a condition associated with aging, involves progressive loss of muscle mass, strength, and function, leading to impaired mobility, health, and increased mortality. The underlying mechanisms remain unclear, which limits the development of effective therapeutic interventions. Emerging evidence implicates chronodisruption as a key contributor to sarcopenia, emphasizing the role of Bmal1, a circadian clock gene critical for muscle integrity and mitochondrial function. In a skeletal muscle-specific and inducible Bmal1 knockout model (iMS-Bmal1-/-), we observed hallmark features of sarcopenia, including disrupted rhythms, impaired muscle function, and mitochondrial dysfunction. Exercise and melatonin treatment reversed these deficits independently of Bmal1. Building on these findings, the present study elucidates several mechanisms underlying these changes and the pathways by which melatonin and exercise exert their beneficial effects. Our findings indicate that iMS-Bmal1-/- mice exhibit reduced expression of satellite cell and muscle regulatory factors, indicating impaired muscle regeneration. While mitochondrial respiration remained unchanged, notable alterations in mitochondrial dynamics disrupted mitochondria in skeletal muscle. In addition, these mice showed alterations in muscle energy metabolism, compromised antioxidant defense, and inflammatory response. Remarkably, exercise and/or melatonin successfully mitigated these deficits, restoring muscle health in Bmal1-deficient mice. These findings position exercise and melatonin as promising therapeutic candidates for combating sarcopenia and emphasize the need to elucidate the molecular pathways underlying their protective effects.
Collapse
Affiliation(s)
- Yolanda Ramírez-Casas
- Centro de Investigación Biomédica, Facultad de Medicina, Departamento de Fisiología, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (Ibs. Granada), Hospital Universitario San Cecilio, Granada, Spain
| | - José Fernández-Martínez
- Centro de Investigación Biomédica, Facultad de Medicina, Departamento de Fisiología, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (Ibs. Granada), Hospital Universitario San Cecilio, Granada, Spain
| | - María Martín-Estebané
- Centro de Investigación Biomédica, Facultad de Medicina, Departamento de Fisiología, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (Ibs. Granada), Hospital Universitario San Cecilio, Granada, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Paula Aranda-Martínez
- Centro de Investigación Biomédica, Facultad de Medicina, Departamento de Fisiología, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (Ibs. Granada), Hospital Universitario San Cecilio, Granada, Spain
| | - Alba López-Rodríguez
- Centro de Investigación Biomédica, Facultad de Medicina, Departamento de Fisiología, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (Ibs. Granada), Hospital Universitario San Cecilio, Granada, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Sergio Esquivel-Ruiz
- Centro de Investigación Biomédica, Facultad de Medicina, Departamento de Fisiología, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (Ibs. Granada), Hospital Universitario San Cecilio, Granada, Spain
- Departamento de Farmacología, Facultad de Ciencias de la Salud de Melilla, Universidad de Granada, Granada, España
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Germaine Escames
- Centro de Investigación Biomédica, Facultad de Medicina, Departamento de Fisiología, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (Ibs. Granada), Hospital Universitario San Cecilio, Granada, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Darío Acuña-Castroviejo
- Centro de Investigación Biomédica, Facultad de Medicina, Departamento de Fisiología, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (Ibs. Granada), Hospital Universitario San Cecilio, Granada, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- UGC de Laboratorios Clínicos, Hospital Universitario San Cecilio, Granada, Spain
| |
Collapse
|
7
|
Orioli L, Thissen JP. Myokines as potential mediators of changes in glucose homeostasis and muscle mass after bariatric surgery. Front Endocrinol (Lausanne) 2025; 16:1554617. [PMID: 40171198 PMCID: PMC11958187 DOI: 10.3389/fendo.2025.1554617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Myokines are bioactive peptides released by skeletal muscle. Myokines exert auto-, para-, or endocrine effects, enabling them to regulate many aspects of metabolism in various tissues. However, the contribution of myokines to the dramatic changes in glucose homeostasis and muscle mass induced by bariatric surgery has not been established. Our review highlights that myokines such as brain-derived neurotrophic factor (BDNF), meteorin-like protein (Metrnl), secreted protein acidic and rich in cysteine (SPARC), apelin (APLN) and myostatin (MSTN) may mediate changes in glucose homeostasis and muscle mass after bariatric surgery. Our review also identifies myonectin as an interesting candidate for future studies, as this myokine may regulate lipid metabolism and muscle mass after bariatric surgery. These myokines may provide novel therapeutic targets and biomarkers for obesity, type 2 diabetes and sarcopenia.
Collapse
Affiliation(s)
- Laura Orioli
- Research Laboratory of Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jean-Paul Thissen
- Research Laboratory of Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
8
|
Lin Q, Li K, Li L, Guan L, Zeng Y, Cai D, Zhou J, Xu L. Plasma biomarkers in patients with age-related sarcopenia: a proteomic exploration and experimental validation. Aging Clin Exp Res 2024; 37:13. [PMID: 39725826 DOI: 10.1007/s40520-024-02903-7] [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: 06/29/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Various biomarkers associated with sarcopenia have been identified. However, there is a scarcity of studies exploring and validating biomarkers in individuals with age-related sarcopenia. AIMS This study aimed to investigate the proteome and identify potential biomarkers for age-related sarcopenia. METHODS Proteomic analysis and experimental validation were conducted using plasma from hospitalized older adults. Sarcopenia diagnosis was based on the Asian Working Group for Sarcopenia 2019 criteria. Data-independent acquisition-based proteomics was performed on plasma from 60 participants, with 30 diagnosed with sarcopenia and 30 without sarcopenia. Differentially expressed proteins (DEPs) were selected and evaluated by Receiver Operating Characteristic (ROC) analysis. Biomarker candidates were further quantitatively validated by enzyme-linked immunosorbent assay (ELISA) utilizing plasma from 6 participants with sarcopenia and 6 without sarcopenia. RESULTS A total of 39 DEPs were identified and 12 DEPs were selected for ROC analysis. 8 DEPs were included for ELISA validation based on their predictive performance. Paraoxonase-3 (PON3) consistently showed down-regulation in the sarcopenic group across both methodologies. Insulin-like growth factor-binding protein-2 (IGFBP2) showed inconsistency in the sarcopenic group, with up-regulation observed in proteomic analysis but down-regulation in ELISA. DISCUSSION Decline in PON3 may result in an overload of oxidative stress in skeletal muscles and contribute to sarcopenia. Protein modifications of IGFBP2 might exhibit during sarcopenia pathogenesis. CONCLUSIONS Plasma proteins are implicated in sarcopenia pathogenesis. PON3 is highlighted as a potential biomarker for patients with age-related sarcopenia. Further studies are imperative to gain an in-depth understanding of PON3 and IGFBP2.
Collapse
Affiliation(s)
- Qinqing Lin
- Department of Geriatric Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Kangyong Li
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liwei Li
- Department of Geriatric Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Lichang Guan
- Department of Geriatric Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yingtong Zeng
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Dake Cai
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jing Zhou
- Department of Geriatric Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Lishu Xu
- Department of Geriatric Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Guangdong Provincial Institute of Geriatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Timofte DV, Tudor RC, Mocanu V, Labusca L. Obesity, Osteoarthritis, and Myokines: Balancing Weight Management Strategies, Myokine Regulation, and Muscle Health. Nutrients 2024; 16:4231. [PMID: 39683624 PMCID: PMC11644804 DOI: 10.3390/nu16234231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Obesity and osteoarthritis (OA) are increasingly prevalent conditions that are intricately linked, with each exacerbating the other's pathogenesis and worsening patient outcomes. This review explores the dual impact of obesity on OA, highlighting the role of excessive weight in aggravating joint degeneration and the limitations OA imposes on physical activity, which further perpetuates obesity. The role of muscle tissue, particularly the release of myokines during physical activity, is examined in the context of OA and obesity. Myokines such as irisin, IL-6, and myostatin are discussed for their roles in metabolic regulation, inflammation, and tissue repair, offering insights into their potential therapeutic targets. This review emphasizes the importance of supervised weight management methods in parallel with muscle rehabilitation in improving joint health and metabolic balance. The potential for myokine modulation through targeted exercise and weight loss interventions to mitigate the adverse effects of obesity and OA is also discussed, suggesting avenues for future research and therapy development to reduce the burden of these chronic conditions.
Collapse
Affiliation(s)
- Daniel Vasile Timofte
- Department of Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (D.V.T.); (R.C.T.)
| | - Razvan Cosmin Tudor
- Department of Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (D.V.T.); (R.C.T.)
- Dr. Iacob Czihac Military Emergency Hospital Iasi, General Henri Mathias Berthelot Str. 7-9, 700483 Iași, Romania
| | - Veronica Mocanu
- Department of Morpho-Functional Sciences II (Pathophysiology), Center for Obesity BioBehavioral Experimental Research, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Luminita Labusca
- Department of Orthopedics and Traumatology, “Sf. Spiridon” Emergency Clinical Hospital, 700111 Iasi, Romania;
- National Institute of Research and Development in Technical Physics Iasi, 700050 Iasi, Romania
| |
Collapse
|
10
|
Zhou W, Du Z. Oleuropein mitigates non-alcoholic fatty liver disease (NAFLD) and modulates liver metabolites in high-fat diet-induced obese mice via activating PPARα. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:8634-8645. [PMID: 38952322 DOI: 10.1002/jsfa.13691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND This study aimed to elucidate the mechanism of oleuropein (OLE) ameliorates non-alcoholic fatty liver disease (NAFLD) and its underlying mechanisms. RESULTS Male C57BL/6J mice were fed either a low-fat diet (LFD), a high-fat diet (HFD), or a HFD supplemented with 0.03% (w/w) OLE for 16 weeks. OLE supplementation decreased body weight and liver weight, improved serum lipid profiles, and ameliorated HFD-induced hepatic dysfunction. Liver metabolomics analysis revealed that OLE increased the levels of nicotinamide, tauroursodeoxycholic acid, taurine, and docosahexaenoic acid, which were beneficial for lipid homeostasis and inflammation regulation. OLE exerted its protective effects by activating peroxisome proliferator-activated receptor alpha (PPARα), a key transcription factor that regulates fibroblast growth factor 21 (FGF21) expression and modulates lipid oxidation, lipogenesis and inflammation pathways. Importantly, OLE supplementation did not significantly affect body weight or liver weight in PPARα knockout (PPARα KO) mice, indicating that PPARα is essential for OLE-mediated NAFLD prevention. CONCLUSION Our results suggest that OLE alleviates NAFLD in mice by activating PPARα and modulating liver metabolites. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Wei Zhou
- Center for Prevention and Treatment of Cardiovascular Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zheng Du
- The First People's Hospital of Lianyungang Public Health Department, Lianyungang, China
| |
Collapse
|
11
|
Kim SJ, Jo Y, Park SJ, Ji E, Lee JY, Choi E, Baek JY, Jang IY, Jung HW, Kim K, Ryu D, Yoo HJ, Kim BJ. Metabolomic profiles of ovariectomized mice and their associations with body composition and frailty-related parameters in postmenopausal women. J Endocrinol Invest 2024; 47:2551-2563. [PMID: 38493245 DOI: 10.1007/s40618-024-02338-x] [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: 11/21/2023] [Accepted: 02/12/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Menopause, a dramatical estrogen-deficient condition, is considered the most significant milestone in women's health. PURPOSE To investigate the metabolite changes attributed to estrogen deficiency using random forest (RF)-based machine learning (ML) modeling strategy in ovariectomized (OVX) mice as well as determine the clinical relevance of selected metabolites in older women. METHODS AND RESULTS Untargeted and targeted metabolomic analyses revealed that metabolites related to TCA cycle, sphingolipids, phospholipids, fatty acids, and amino acids, were significantly changed in the plasma and/or muscle of OVX mice. Subsequent ML classifiers based on RF algorithm selected alpha-ketoglutarate (AKG), arginine, carnosine, ceramide C24, phosphatidylcholine (PC) aa C36:6, and PC ae C42:3 in plasma as well as PC aa 34:1, PC aa C34:3, PC aa C36:5, PC aa C32:1, PC aa C36:2, and sphingosine in muscle as top featured metabolites that differentiate the OVX mice from the sham-operated group. When circulating levels of AKG, arginine, and carnosine, which showed the most significant changes in OVX mice blood, were measured in postmenopausal women, higher plasma AKG levels were associated with lower bone mass, weak grip strength, poor physical performance, and increased frailty risk. CONCLUSIONS Metabolomics- and ML-based methods identified the key metabolites of blood and muscle that were significantly changed after ovariectomy in mice, and the clinical implication of several metabolites was investigated by looking at their correlation with body composition and frailty-related parameters in postmenopausal women. These findings provide crucial context for understanding the diverse physiological alterations caused by estrogen deficiency in women.
Collapse
Affiliation(s)
- S J Kim
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center,, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, South Korea
| | - Y Jo
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea
| | - S J Park
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - E Ji
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - J Y Lee
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - E Choi
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - J-Y Baek
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - I Y Jang
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - H-W Jung
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - K Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - D Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea.
| | - H J Yoo
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center,, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, South Korea.
| | - B-J Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, South Korea.
| |
Collapse
|
12
|
Nasso R, D'Errico A, Motti ML, Masullo M, Arcone R. Dietary Protein and Physical Exercise for the Treatment of Sarcopenia. Clin Pract 2024; 14:1451-1467. [PMID: 39194921 DOI: 10.3390/clinpract14040117] [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: 05/31/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
Sarcopenia is a multifactorial age-related disorder that causes a decrease in muscle mass, strength, and function, leading to alteration of movement, risk of falls, and hospitalization. This article aims to review recent findings on the factors underlying sarcopenia and the strategies required to delay and counteract its symptoms. We focus on molecular factors linked to ageing, on the role of low-grade chronic and acute inflammatory conditions such as cancer, which contributes to the onset of sarcopenia, and on the clinical criteria for its diagnosis. The use of drugs against sarcopenia is still subject to debate, and the suggested approaches to restore muscle health are based on adequate dietary protein intake and physical exercise. We also highlight the difference in the amount and quality of amino acids within animal- and plant-based diets, as studies have often shown varying results regarding their effect on sarcopenia in elderly people. In addition, many studies have reported that non-pharmacological approaches, such as an optimization of dietary protein intake and training programs based on resistance exercise, can be effective in preventing and delaying sarcopenia. These approaches not only improve the maintenance of skeletal muscle function, but also reduce health care costs and improve life expectancy and quality in elderly people.
Collapse
Affiliation(s)
- Rosarita Nasso
- Department of Medical, Movement and Well-Being Sciences (DiSMMeB), University of Naples "Parthenope", Via Medina 40, 80133 Napoli, Italy
| | - Antonio D'Errico
- Department of Medical, Movement and Well-Being Sciences (DiSMMeB), University of Naples "Parthenope", Via Medina 40, 80133 Napoli, Italy
| | - Maria Letizia Motti
- Department of Medical, Movement and Well-Being Sciences (DiSMMeB), University of Naples "Parthenope", Via Medina 40, 80133 Napoli, Italy
| | - Mariorosario Masullo
- Department of Medical, Movement and Well-Being Sciences (DiSMMeB), University of Naples "Parthenope", Via Medina 40, 80133 Napoli, Italy
| | - Rosaria Arcone
- Department of Medical, Movement and Well-Being Sciences (DiSMMeB), University of Naples "Parthenope", Via Medina 40, 80133 Napoli, Italy
| |
Collapse
|
13
|
Li HZ, Zhang JL, Yuan DL, Xie WQ, Ladel CH, Mobasheri A, Li YS. Role of signaling pathways in age-related orthopedic diseases: focus on the fibroblast growth factor family. Mil Med Res 2024; 11:40. [PMID: 38902808 PMCID: PMC11191355 DOI: 10.1186/s40779-024-00544-5] [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: 04/08/2023] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
Fibroblast growth factor (FGF) signaling encompasses a multitude of functions, including regulation of cell proliferation, differentiation, morphogenesis, and patterning. FGFs and their receptors (FGFR) are crucial for adult tissue repair processes. Aberrant FGF signal transduction is associated with various pathological conditions such as cartilage damage, bone loss, muscle reduction, and other core pathological changes observed in orthopedic degenerative diseases like osteoarthritis (OA), intervertebral disc degeneration (IVDD), osteoporosis (OP), and sarcopenia. In OA and IVDD pathologies specifically, FGF1, FGF2, FGF8, FGF9, FGF18, FGF21, and FGF23 regulate the synthesis, catabolism, and ossification of cartilage tissue. Additionally, the dysregulation of FGFR expression (FGFR1 and FGFR3) promotes the pathological process of cartilage degradation. In OP and sarcopenia, endocrine-derived FGFs (FGF19, FGF21, and FGF23) modulate bone mineral synthesis and decomposition as well as muscle tissues. FGF2 and other FGFs also exert regulatory roles. A growing body of research has focused on understanding the implications of FGF signaling in orthopedic degeneration. Moreover, an increasing number of potential targets within the FGF signaling have been identified, such as FGF9, FGF18, and FGF23. However, it should be noted that most of these discoveries are still in the experimental stage, and further studies are needed before clinical application can be considered. Presently, this review aims to document the association between the FGF signaling pathway and the development and progression of orthopedic diseases. Besides, current therapeutic strategies targeting the FGF signaling pathway to prevent and treat orthopedic degeneration will be evaluated.
Collapse
Affiliation(s)
- Heng-Zhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jing-Lve Zhang
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Medicine Central, South University, Changsha, 410083, China
| | - Dong-Liang Yuan
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Medicine Central, South University, Changsha, 410083, China
| | - Wen-Qing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | | | - Ali Mobasheri
- Faculty of Medicine, Research Unit of Health Sciences and Technology, University of Oulu, 90014, Oulu, Finland.
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406, Vilnius, Lithuania.
- Department of Rheumatology and Clinical Immunology, Universitair Medisch Centrum Utrecht, Utrecht, 3508, GA, the Netherlands.
- Department of Joint Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, B-4000, Liège, Belgium.
| | - Yu-Sheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
14
|
Scudese E, Vue Z, Katti P, Marshall AG, Demirci M, Vang L, López EG, Neikirk K, Shao B, Le H, Stephens D, Hall DD, Rostami R, Rodman T, Kabugi K, Harris C, Shao J, Mungai M, AshShareef ST, Hicsasmaz I, Manus S, Wanjalla C, Whiteside A, Dasari R, Williams C, Damo SM, Gaddy JA, Glancy B, Dantas EHM, Kinder A, Kadam A, Tomar D, Scartoni F, Baffi M, McReynolds MR, Phillips MA, Cooper A, Murray SA, Quintana AM, Exil V, Kirabo A, Mobley BC, Hinton A. 3D Mitochondrial Structure in Aging Human Skeletal Muscle: Insights into MFN-2 Mediated Changes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.13.566502. [PMID: 38168206 PMCID: PMC10760012 DOI: 10.1101/2023.11.13.566502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Age-related atrophy of skeletal muscle, is characterized by loss of mass, strength, endurance, and oxidative capacity during aging. Notably, bioenergetics and protein turnover studies have shown that mitochondria mediate this decline in function. Although exercise has been the only therapy to mitigate sarcopenia, the mechanisms that govern how exercise serves to promote healthy muscle aging are unclear. Mitochondrial aging is associated with decreased mitochondrial capacity, so we sought to investigate how aging affects mitochondrial structure and potential age-related regulators. Specifically, the three-dimensional (3D) mitochondrial structure associated with morphological changes in skeletal muscle during aging requires further elucidation. We hypothesized that aging causes structural remodeling of mitochondrial 3D architecture representative of dysfunction, and this effect is mitigated by exercise. We used serial block-face scanning electron microscopy to image human skeletal tissue samples, followed by manual contour tracing using Amira software for 3D reconstruction and subsequent analysis of mitochondria. We then applied a rigorous in vitro and in vivo exercise regimen during aging. Across 5 human cohorts, we correlate differences in magnetic resonance imaging, mitochondria 3D structure, exercise parameters, and plasma immune markers between young (under 50 years) and old (over 50 years) individuals. We found that mitochondria we less spherical and more complex, indicating age-related declines in contact site capacity. Additionally, aged samples showed a larger volume phenotype in both female and male humans, indicating potential mitochondrial swelling. Concomitantly, muscle area, exercise capacity, and mitochondrial dynamic proteins showed age-related losses. Exercise stimulation restored mitofusin 2 (MFN2), one such of these mitochondrial dynamic proteins, which we show is required for the integrity of mitochondrial structure. Furthermore, we show that this pathway is evolutionarily conserved as Marf, the MFN2 ortholog in Drosophila, knockdown alters mitochondrial morphology and leads to the downregulation of genes regulating mitochondrial processes. Our results define age-related structural changes in mitochondria and further suggest that exercise may mitigate age-related structural decline through modulation of mitofusin 2.
Collapse
Affiliation(s)
- Estevão Scudese
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
- Sport Sciences and Exercise Laboratory (LaCEE), Catholic University of Petrópolis (UCP), Brazil
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Prassana Katti
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, AP, 517619, India
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Andrea G. Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Mert Demirci
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Edgar Garza López
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Bryanna Shao
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Han Le
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Dominique Stephens
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Duane D. Hall
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Rahmati Rostami
- Department of Genetic Medicine, Joan & Sanford I. Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Taylor Rodman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kinuthia Kabugi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Chanel Harris
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Jianqiang Shao
- Central Microscopy Research Facility, Iowa City, IA 52242, USA
| | - Margaret Mungai
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Salma T. AshShareef
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Innes Hicsasmaz
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Sasha Manus
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Celestine Wanjalla
- Division of Infection Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Aaron Whiteside
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, 45435, USA
| | - Revathi Dasari
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, AP, 517619, India
| | - Clintoria Williams
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, 45435, USA
| | - Steven M. Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, TN, 37208, USA
| | - Jennifer A. Gaddy
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Tennessee Valley Healthcare Systems, U.S. Department of Veterans Affairs, Nashville, TN, 37212, USA
| | - Brian Glancy
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- NIAMS, NIH, Bethesda, MD, 20892, USA
| | - Estélio Henrique Martin Dantas
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
- Doctor’s Degree Program in Nursing and Biosciences - PpgEnfBio, Federal University of the State of Rio de Janeiro - UNIRIO, Rio de Janeiro, RJ, Brazil
- Laboratory of Human Motricity Biosciences - LABIMH, Federal University of the State of Rio de Janeiro - UNIRIO, RJ, Brazil
- Brazilian Paralympic Academy – APB
- Doctor’s Degree Program in Health and Environment - PSA, Tiradentes University - UNIT, Aracaju, SE, Brazil
| | - André Kinder
- Artur Sá Earp Neto University Center - UNIFASE-FMP, Petrópolis Medical School, Brazil
| | - Ashlesha Kadam
- Department of Internal Medicine, Section of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157 USA
| | - Dhanendra Tomar
- Department of Internal Medicine, Section of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157 USA
| | - Fabiana Scartoni
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | - Matheus Baffi
- Sport Sciences and Exercise Laboratory (LaCEE), Catholic University of Petrópolis (UCP), Brazil
| | - Melanie R. McReynolds
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA, 16801, USA
| | - Mark A. Phillips
- Department of Integrative Biology, Oregon State University, Corvallis, OR, 97331, USA
| | - Anthonya Cooper
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Sandra A. Murray
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Anita M. Quintana
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, Texas, USA
| | - Vernat Exil
- Department of Pediatrics, Div. of Cardiology, St. Louis University School of Medicine, St. Louis, MO, 63104, USA
| | - Annet Kirabo
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Bret C. Mobley
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| |
Collapse
|
15
|
Tian J, Song M, Cho KJ, Lee HY, Ju SH, Lim JR, Nga HT, Nguyen TL, Moon JS, Jang HJ, Hwang JM, Yi HS. Differences in Type 2 Fiber Composition in the Vastus Lateralis and Gluteus Maximus of Patients with Hip Fractures. Endocrinol Metab (Seoul) 2024; 39:521-530. [PMID: 38858821 PMCID: PMC11220211 DOI: 10.3803/enm.2024.1935] [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: 01/13/2024] [Revised: 03/15/2024] [Accepted: 03/21/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGRUOUND Aging leads to sarcopenia, which is characterized by reduced muscle mass and strength. Many factors, including altered muscle protein turnover, diminished neuromuscular function, hormonal changes, systemic inflammation, and the structure and composition of muscle fibers, play a crucial role in age-related muscle decline. This study explored differences in muscle fiber types contributing to overall muscle function decline in aging, focusing on individuals with hip fractures from falls. METHODS A pilot study at Chungnam National University Hospital collected muscle biopsies from hip fracture patients aged 20 to 80 undergoing surgical treatment. Muscle biopsies from the vastus lateralis and gluteus maximus were obtained during hip arthroplasty or internal fixation. Handgrip strength, calf and thigh circumference, and bone mineral density were evaluated in individuals with hip fractures from falls. We analyzed the relationships between each clinical characteristic and muscle fiber type. RESULTS In total, 26 participants (mean age 67.9 years, 69.2% male) were included in this study. The prevalence of sarcopenia was 53.8%, and that of femoral and lumbar osteoporosis was 19.2% and 11.5%, respectively. Vastus lateralis analysis revealed an age-related decrease in type IIx fibers, a higher proportion of type IIa fibers in women, and an association between handgrip strength and type IIx fibers in men. The gluteus maximus showed no significant correlations with clinical parameters. CONCLUSION This study identified complex associations between age, sex, handgrip strength, and muscle fiber composition in hip fracture patients, offering insights crucial for targeted interventions combating age-related muscle decline and improving musculoskeletal health.
Collapse
Affiliation(s)
- Jingwen Tian
- Laboratory of Endocrinology and Immune System, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Minchul Song
- Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Kyu Jeong Cho
- Laboratory of Endocrinology and Immune System, Chungnam National University College of Medicine, Daejeon, Korea
| | - Ho Yeop Lee
- Laboratory of Endocrinology and Immune System, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Sang Hyeon Ju
- Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Jung Ryul Lim
- Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Ha Thi Nga
- Laboratory of Endocrinology and Immune System, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Thi Linh Nguyen
- Laboratory of Endocrinology and Immune System, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Ji Sun Moon
- Laboratory of Endocrinology and Immune System, Chungnam National University College of Medicine, Daejeon, Korea
| | - Hyo Ju Jang
- Laboratory of Endocrinology and Immune System, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Jung-Mo Hwang
- Department of Orthopedics, Chungnam National University Hospital, Daejeon, Korea
| | - Hyon-Seung Yi
- Laboratory of Endocrinology and Immune System, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| |
Collapse
|
16
|
Chen C, Xie L, Zhang M, Shama, Cheng KKY, Jia W. The interplay between the muscle and liver in the regulation of glucolipid metabolism. J Mol Cell Biol 2024; 15:mjad073. [PMID: 38095440 PMCID: PMC11078061 DOI: 10.1093/jmcb/mjad073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/24/2023] [Indexed: 05/09/2024] Open
Affiliation(s)
- Cheng Chen
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Liping Xie
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Mingliang Zhang
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Shama
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Kenneth King Yip Cheng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Weiping Jia
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| |
Collapse
|
17
|
Ozonoff A, Jayavelu ND, Liu S, Melamed E, Milliren CE, Qi J, Geng LN, McComsey GA, Cairns CB, Baden LR, Schaenman J, Shaw AC, Samaha H, Seyfert-Margolis V, Krammer F, Rosen LB, Steen H, Syphurs C, Dandekar R, Shannon CP, Sekaly RP, Ehrlich LIR, Corry DB, Kheradmand F, Atkinson MA, Brakenridge SC, Higuita NIA, Metcalf JP, Hough CL, Messer WB, Pulendran B, Nadeau KC, Davis MM, Sesma AF, Simon V, van Bakel H, Kim-Schulze S, Hafler DA, Levy O, Kraft M, Bime C, Haddad EK, Calfee CS, Erle DJ, Langelier CR, Eckalbar W, Bosinger SE, Peters B, Kleinstein SH, Reed EF, Augustine AD, Diray-Arce J, Maecker HT, Altman MC, Montgomery RR, Becker PM, Rouphael N. Features of acute COVID-19 associated with post-acute sequelae of SARS-CoV-2 phenotypes: results from the IMPACC study. Nat Commun 2024; 15:216. [PMID: 38172101 PMCID: PMC10764789 DOI: 10.1038/s41467-023-44090-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 11/29/2023] [Indexed: 01/05/2024] Open
Abstract
Post-acute sequelae of SARS-CoV-2 (PASC) is a significant public health concern. We describe Patient Reported Outcomes (PROs) on 590 participants prospectively assessed from hospital admission for COVID-19 through one year after discharge. Modeling identified 4 PRO clusters based on reported deficits (minimal, physical, mental/cognitive, and multidomain), supporting heterogenous clinical presentations in PASC, with sub-phenotypes associated with female sex and distinctive comorbidities. During the acute phase of disease, a higher respiratory SARS-CoV-2 viral burden and lower Receptor Binding Domain and Spike antibody titers were associated with both the physical predominant and the multidomain deficit clusters. A lower frequency of circulating B lymphocytes by mass cytometry (CyTOF) was observed in the multidomain deficit cluster. Circulating fibroblast growth factor 21 (FGF21) was significantly elevated in the mental/cognitive predominant and the multidomain clusters. Future efforts to link PASC to acute anti-viral host responses may help to better target treatment and prevention of PASC.
Collapse
Affiliation(s)
- Al Ozonoff
- Clinical & Data Coordinating Center (CDCC), Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
| | | | - Shanshan Liu
- Clinical & Data Coordinating Center (CDCC), Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
| | | | - Carly E Milliren
- Clinical & Data Coordinating Center (CDCC), Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
| | - Jingjing Qi
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Grace A McComsey
- Case Western Reserve University and University Hospitals of Cleveland, Cleveland, OH, USA
| | | | - Lindsey R Baden
- Boston Clinical Site: Precision Vaccines Program, Boston Children's Hospital, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
| | - Joanna Schaenman
- David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - Albert C Shaw
- Yale School of Medicine, and Yale School of Public Health, New Haven, CT, USA
| | | | | | | | - Lindsey B Rosen
- National Institute of Allergy and Infectious Diseases/National Institutes of Health, Bethesda, MD, USA
| | - Hanno Steen
- Boston Clinical Site: Precision Vaccines Program, Boston Children's Hospital, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
| | - Caitlin Syphurs
- Clinical & Data Coordinating Center (CDCC), Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
| | - Ravi Dandekar
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Casey P Shannon
- Centre for Heart Lung Innovation, Providence Research, St. Paul's Hospital, and the PROOF Centre of Excellence, Vancouver, BC, Canada
| | - Rafick P Sekaly
- Case Western Reserve University and University Hospitals of Cleveland, Cleveland, OH, USA
| | | | - David B Corry
- Baylor College of Medicine, and the Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Farrah Kheradmand
- Baylor College of Medicine, and the Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Mark A Atkinson
- University of Florida/University of South Florida, Tampa, FL, USA
| | | | | | - Jordan P Metcalf
- Oklahoma University Health Sciences Center, Oklahoma City, OK, USA
| | | | | | | | | | | | | | - Viviana Simon
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Harm van Bakel
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - David A Hafler
- Yale School of Medicine, and Yale School of Public Health, New Haven, CT, USA
| | - Ofer Levy
- Boston Clinical Site: Precision Vaccines Program, Boston Children's Hospital, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
| | | | | | - Elias K Haddad
- Drexel University/Tower Health Hospital, Philadelphia, PA, USA
| | - Carolyn S Calfee
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - David J Erle
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Charles R Langelier
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Walter Eckalbar
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | | | - Bjoern Peters
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Steven H Kleinstein
- Yale School of Medicine, and Yale School of Public Health, New Haven, CT, USA
| | - Elaine F Reed
- David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - Alison D Augustine
- National Institute of Allergy and Infectious Diseases/National Institutes of Health, Bethesda, MD, USA
| | - Joann Diray-Arce
- Clinical & Data Coordinating Center (CDCC), Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
| | | | | | - Ruth R Montgomery
- Yale School of Medicine, and Yale School of Public Health, New Haven, CT, USA
| | - Patrice M Becker
- National Institute of Allergy and Infectious Diseases/National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
18
|
Li S, Chen J, Wei P, Zou T, You J. Fibroblast Growth Factor 21: A Fascinating Perspective on the Regulation of Muscle Metabolism. Int J Mol Sci 2023; 24:16951. [PMID: 38069273 PMCID: PMC10707024 DOI: 10.3390/ijms242316951] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) plays a vital role in normal eukaryotic organism development and homeostatic metabolism under the influence of internal and external factors such as endogenous hormone changes and exogenous stimuli. Over the last few decades, comprehensive studies have revealed the key role of FGF21 in regulating many fundamental metabolic pathways, including the muscle stress response, insulin signaling transmission, and muscle development. By coordinating these metabolic pathways, FGF21 is thought to contribute to acclimating to a stressful environment and the subsequent recovery of cell and tissue homeostasis. With the emphasis on FGF21, we extensively reviewed the research findings on the production and regulation of FGF21 and its role in muscle metabolism. We also emphasize how the FGF21 metabolic networks mediate mitochondrial dysfunction, glycogen consumption, and myogenic development and investigate prospective directions for the functional exploitation of FGF21 and its downstream effectors, such as the mammalian target of rapamycin (mTOR).
Collapse
Affiliation(s)
| | | | | | - Tiande Zou
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang 330045, China; (S.L.); (J.C.); (P.W.)
| | - Jinming You
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang 330045, China; (S.L.); (J.C.); (P.W.)
| |
Collapse
|
19
|
Bondi D, Bevere M, Piccirillo R, Sorci G, Di Felice V, Re Cecconi AD, D'Amico D, Pietrangelo T, Fulle S. Integrated procedures for accelerating, deepening, and leading genetic inquiry: A first application on human muscle secretome. Mol Genet Metab 2023; 140:107705. [PMID: 37837864 DOI: 10.1016/j.ymgme.2023.107705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/15/2023] [Accepted: 10/01/2023] [Indexed: 10/16/2023]
Abstract
PURPOSE Beyond classical procedures, bioinformatic-assisted approaches and computational biology offer unprecedented opportunities for scholars. However, these amazing possibilities still need epistemological criticism, as well as standardized procedures. Especially those topics with a huge body of data may benefit from data science (DS)-assisted methods. Therefore, the current study dealt with the combined expert-assisted and DS-assisted approaches to address the broad field of muscle secretome. We aimed to apply DS tools to fix the literature research, suggest investigation targets with a data-driven approach, predict possible scenarios, and define a workflow. METHODS Recognized scholars with expertise on myokines were invited to provide a list of the most important myokines. GeneRecommender, GeneMANIA, HumanNet, and STRING were selected as DS tools. Networks were built on STRING and GeneMANIA. The outcomes of DS tools included the top 5 recommendations. Each expert-led discussion has been then integrated with an DS-led approach to provide further perspectives. RESULTS Among the results, 11 molecules had already been described as bona-fide myokines in literature, and 11 molecules were putative myokines. Most of the myokines and the putative myokines recommended by the DS tools were described as present in the cargo of extracellular vesicles. CONCLUSIONS Including both supervised and unsupervised learning methods, as well as encompassing algorithms focused on both protein interaction and gene represent a comprehensive approach to tackle complex biomedical topics. DS-assisted methods for reviewing existent evidence, recommending targets of interest, and predicting original scenarios are worth exploring as in silico recommendations to be integrated with experts' ideas for optimizing molecular studies.
Collapse
Affiliation(s)
- Danilo Bondi
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti - Pescara, Chieti, Italy; Interuniversity Institute of Myology (IIM), Perugia, Italy.
| | - Michele Bevere
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.
| | - Rosanna Piccirillo
- Department of Neurosciences, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy.
| | - Guglielmo Sorci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy; Interuniversity Institute of Myology (IIM), Perugia, Italy.
| | - Valentina Di Felice
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy.
| | - Andrea David Re Cecconi
- Department of Neurosciences, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy.
| | - Daniela D'Amico
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy.
| | - Tiziana Pietrangelo
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti - Pescara, Chieti, Italy; Interuniversity Institute of Myology (IIM), Perugia, Italy.
| | - Stefania Fulle
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti - Pescara, Chieti, Italy; Interuniversity Institute of Myology (IIM), Perugia, Italy.
| |
Collapse
|
20
|
Ji S, Park SJ, Lee JY, Baek JY, Jung HW, Kim K, Yoo HJ, Jang IY, Kim BJ. Lack of association between serum myonectin levels and sarcopenia in older Asian adults. Exp Gerontol 2023; 178:112229. [PMID: 37270069 DOI: 10.1016/j.exger.2023.112229] [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] [Revised: 05/22/2023] [Accepted: 05/31/2023] [Indexed: 06/05/2023]
Abstract
Myonectin is a muscle-secreted factor that helps maintain homeostasis in the body by regulating several functions, including lipid metabolism. Previous studies suggested that myonectin may play a role in muscle health in an autocrine manner, but its impact on human skeletal muscle is still unclear. We aimed to investigate the relationship of serum myonectin levels with sarcopenia and related muscle parameters. We conducted a cross-sectional study of 142 older adults whose muscle mass, grip strength, gait speed, chair stands, and short physical performance battery (SPPB) were evaluated in the geriatric clinic of a tertiary medical center. Sarcopenia was defined based on Asian-specific cutoff values, and circulating myonectin levels were measured using an enzyme immunoassay. Before and after adjusting for age, sex, and body mass index, the serum myonectin level was not significantly different when the patients were stratified by status of sarcopenia, muscle mass, muscle strength, and physical performance. Furthermore, whether given as a continuous variable or divided into quartile groups, the serum myonectin level had no association with the skeletal muscle mass, grip strength, gait speed, chair stand test, or SPPB score. Our findings did not confirm the potential role of myonectin in muscle metabolism observed in experimental research. Thus, serum myonectin levels cannot predict the risk of sarcopenia in older Asian adults.
Collapse
Affiliation(s)
- Sunghwan Ji
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - So Jeong Park
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Jin Young Lee
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Ji Yeon Baek
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Hee-Won Jung
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Kyunggon Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea; Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Hyun Ju Yoo
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Il-Young Jang
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea.
| | - Beom-Jun Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea.
| |
Collapse
|
21
|
Wei S, Nguyen TT, Zhang Y, Ryu D, Gariani K. Sarcopenic obesity: epidemiology, pathophysiology, cardiovascular disease, mortality, and management. Front Endocrinol (Lausanne) 2023; 14:1185221. [PMID: 37455897 PMCID: PMC10344359 DOI: 10.3389/fendo.2023.1185221] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023] Open
Abstract
Sarcopenic obesity is defined as the coexistence of sarcopenia and obesity in the same individual, characterized by of the co-presence of body fat accumulation and muscle loss. This condition is currently a major concern as it is associated with frailty and disabilities such as cardiovascular disease, fractures, dementia, cancer, and increased all-cause mortality. Particularly, older individuals remain at risk of sarcopenic obesity. Progress at several levels is needed to improve the global prognostic outlook for this condition, including the elaboration and implementation of a more uniform definition that may favor the identification and specification of prevalence by age group. Furthermore, improvements in the understanding of the pathogenesis of sarcopenic obesity may lead to the development of more specific therapeutic interventions to improve prognosis. We reviewed the knowledge on sarcopenic obesity and its associations with cardiovascular diseases and mortality.
Collapse
Affiliation(s)
- Shibo Wei
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Thanh T. Nguyen
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Yan Zhang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Karim Gariani
- Division of Endocrinology, Diabetes, Nutrition and Therapeutic Patient Education, Department of Medical Specialties, Geneva University Hospitals, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
22
|
Liu H, He X, Deng XY, Yan JL. Exploring the correlation between serum fibroblast growth factor-21 levels and Sarcopenia: a systematic review and meta-analysis. BMC Musculoskelet Disord 2023; 24:533. [PMID: 37386374 DOI: 10.1186/s12891-023-06641-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 06/15/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND Fibroblast growth factor 21 (FGF-21) plays an important role in the growth and metabolism of skeletal muscle cells. This study aims to systemically review the evidence regarding the relationship between FGF-21 levels and Sarcopenia, as well as the related influential factors. METHODS This review was conducted according to the PRISMA guidelines. We comprehensively searched PubMed, EMBASE, the Web of Science, Scopus, and Chinese Databases (CNKI, Wan Fang, VIP, and CBM) up to 1 May 2023. 3 investigators performed independent literature screening and data extraction of the included literature, and two investigators performed an independent quality assessment of case-control studies using the Joanna Briggs Institute (JBI) tool. Data analysis was performed using Review Manager 5.4 software. For continuous various outcomes, mean difference (MD) or standard mean difference (SMD) with 95% confidence intervals (CIs) was applied for assessment by fixed-effect or random-effect model analysis. The heterogeneity test was performed by the Q-statistic and quantified using I2, and publication bias was evaluated using a funnel plot. RESULTS Five studies with a total of 625 cases were included in the review. Meta-analysis showed lower BMI in the sarcopenia group [MD= -2.88 (95% CI, -3. 49, -2.27); P < 0.00001; I2 = 0%], significantly reduced grip strength in the sarcopenia group compared to the non-sarcopenia group [MD = -7.32(95% CI, -10.42,-4.23); P < 0.00001; I2 = 93%]. No statistically significant differences in serum FGF21 levels were found when comparing the two groups of subjects [SMD = 0.31(95% CI, -0.42, 1.04); P = 0.41; I2 = 94%], and no strong correlation was found between the onset of sarcopenia and serum FGF21 levels. CONCLUSION The diagnosis of sarcopenia is followed by a more significant decrease in muscle mass and strength, but there is a lack of strong evidence to support a direct relationship between elevated organismal FGF21 and sarcopenia, and it is not convincing to use FGF21 as a biological or diagnostic marker for sarcopenia. The currently used diagnostic criteria for sarcopenia and setting of cut-off values for each evaluation parameter no longer seem to match clinical practice.
Collapse
Affiliation(s)
- Hao Liu
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Xia He
- Affiliated Sichuan Provincial Rehabilitation Hospital of the Chengdu University of Traditional Chinese Medicine, Chengdu, 611135, China.
| | - Xiao-Yan Deng
- Tianhui Town Community Health Center, Chengdu, 610081, China
| | - Jing-Lu Yan
- Tianhui Town Community Health Center, Chengdu, 610081, China
| |
Collapse
|
23
|
Park MJ, Choi KM. Interplay of skeletal muscle and adipose tissue: sarcopenic obesity. Metabolism 2023; 144:155577. [PMID: 37127228 DOI: 10.1016/j.metabol.2023.155577] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/03/2023]
Abstract
Sarcopenic obesity is becoming a global health concern, owing to the rising older population, causing cardiometabolic morbidity and mortality. Loss of muscle exceeding normal age-related changes has been revealed to be associated with obesity, aggravating each other through complex interactions. Physiological regeneration and proliferation of muscle tissue are achieved through harmonious processes of regulated inflammation, autophagy, muscle satellite cell proliferation, and signaling molecule function. Adipokines and myokines are signaling molecules from adipose tissue and muscle, respectively, that exert autocrine, paracrine, and endocrine effects on fat and muscle tissues. These signaling molecules interact with each other to regulate metabolic homeostasis. However, excessive adiposity creates pro-inflammatory conditions, leading to metabolic disorders and the disorganization of systemic homeostasis. Therefore, obesity impedes muscle tissue regeneration and induces the loss of muscle mass and function. Numerous studies have attempted to demonstrate the pathophysiological interaction between sarcopenia and obesity, but the interwoven matrix of the relationship between myokines and adipokines has made it difficult for researchers to understand them. This review briefly describes updated information about the crosstalk between muscle and adipose tissue.
Collapse
Affiliation(s)
- Min Jeong Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyung Mook Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
24
|
Ladang A, Beaudart C, Reginster JY, Al-Daghri N, Bruyère O, Burlet N, Cesari M, Cherubini A, da Silva MC, Cooper C, Cruz-Jentoft AJ, Landi F, Laslop A, Maggi S, Mobasheri A, Ormarsdottir S, Radermecker R, Visser M, Yerro MCP, Rizzoli R, Cavalier E. Biochemical Markers of Musculoskeletal Health and Aging to be Assessed in Clinical Trials of Drugs Aiming at the Treatment of Sarcopenia: Consensus Paper from an Expert Group Meeting Organized by the European Society for Clinical and Economic Aspects of Osteoporosis, Osteoarthritis and Musculoskeletal Diseases (ESCEO) and the Centre Académique de Recherche et d'Expérimentation en Santé (CARES SPRL), Under the Auspices of the World Health Organization Collaborating Center for the Epidemiology of Musculoskeletal Conditions and Aging. Calcif Tissue Int 2023; 112:197-217. [PMID: 36633611 PMCID: PMC9859913 DOI: 10.1007/s00223-022-01054-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 12/18/2022] [Indexed: 01/13/2023]
Abstract
In clinical trials, biochemical markers provide useful information on the drug's mode of action, therapeutic response and side effect monitoring and can act as surrogate endpoints. In pharmacological intervention development for sarcopenia management, there is an urgent need to identify biomarkers to measure in clinical trials and that could be used in the future in clinical practice. The objective of the current consensus paper is to provide a clear list of biochemical markers of musculoskeletal health and aging that can be recommended to be measured in Phase II and Phase III clinical trials evaluating new chemical entities for sarcopenia treatment. A working group of the European Society for Clinical and Economic Aspects of Osteoporosis, Osteoarthritis and Musculoskeletal Diseases (ESCEO) proposed classifying biochemical markers into 2 series: biochemical markers evaluating musculoskeletal status and biochemical markers evaluating causal factors. For series 1, the group agreed on 4 biochemical markers that should be assessed in Phase II or Phase III trials (i.e., Myostatin-Follistatin, Brain Derived Neurotrophic Factor, N-terminal Type III Procollagen and Serum Creatinine to Serum Cystatin C Ratio - or the Sarcopenia Index). For series 2, the group agreed on 6 biochemical markers that should be assessed in Phase II trials (i.e., the hormones insulin-like growth factor-1 (IGF-I), dehydroepiandrosterone sulphate, and cortisol, and the inflammatory markers C-reactive protein (CRP), interleukin-6 and tumor necrosis factor-α), and 2 in Phase III trials (i.e., IGF-I and CRP). The group also proposed optional biochemical markers that may provide insights into the mode of action of pharmacological therapies. Further research and development of new methods for biochemical marker assays may lead to the evolution of these recommendations.
Collapse
Affiliation(s)
- Aurélie Ladang
- Department of Clinical Chemistry, CHU de Liège, University of Liège, Liège, Belgium.
| | - Charlotte Beaudart
- Division of Public Health, Epidemiology and Health Economics, WHO Collaborating Center for Public Health Aspects of Musculo-Skeletal Health and Ageing,, University of Liège, Liège, Belgium
| | - Jean-Yves Reginster
- Division of Public Health, Epidemiology and Health Economics, WHO Collaborating Center for Public Health Aspects of Musculo-Skeletal Health and Ageing,, University of Liège, Liège, Belgium
- Biochemistry Department, College of Science, Chair for Biomarkers of Chronic Diseases, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Nasser Al-Daghri
- Biochemistry Department, College of Science, Chair for Biomarkers of Chronic Diseases, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Olivier Bruyère
- Division of Public Health, Epidemiology and Health Economics, WHO Collaborating Center for Public Health Aspects of Musculo-Skeletal Health and Ageing,, University of Liège, Liège, Belgium
| | - Nansa Burlet
- Division of Public Health, Epidemiology and Health Economics, WHO Collaborating Center for Public Health Aspects of Musculo-Skeletal Health and Ageing,, University of Liège, Liège, Belgium
| | - Matteo Cesari
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Antonio Cherubini
- Geriatric Unit, IRCCS Istituti Clinici Scientifici Maugeri, Milan, Italy
| | | | - Cyrus Cooper
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | | | - Francesco Landi
- Department of Geriatrics, Neurosciences and Orthopedics, Catholic University of the Sacred Heart, Rome, Italy
| | - Andrea Laslop
- Scientific Office, Federal Office for Safety in Health Care, Vienna, Austria
| | | | - Ali Mobasheri
- Division of Public Health, Epidemiology and Health Economics, WHO Collaborating Center for Public Health Aspects of Musculo-Skeletal Health and Ageing,, University of Liège, Liège, Belgium
- State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | | | - Régis Radermecker
- Department of Diabetes, Nutrition and Metabolic Disorders, Clinical Pharmacology, University of Liege, CHU de Liège, Liège, Belgium
| | - Marjolein Visser
- Department of Health Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | - René Rizzoli
- Faculty of Medicine, Service of Bone Diseases, Geneva University Hospitals, Geneva, Switzerland
| | - Etienne Cavalier
- Department of Clinical Chemistry, CHU de Liège, University of Liège, Liège, Belgium
| |
Collapse
|
25
|
The Contribution of Diet Therapy and Probiotics in the Treatment of Sarcopenia Induced by Prolonged Immobilization Caused by the COVID-19 Pandemic. Nutrients 2022; 14:nu14214701. [PMID: 36364963 PMCID: PMC9654246 DOI: 10.3390/nu14214701] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 10/31/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
The prolonged immobilization associated with COVID-19 infection and the restrictions imposed by the pandemic have determined major changes in physical activity and eating habits, with a negative impact on physical performance. This study monitored non-pharmacological interventions (diet therapy and probiotics) in managing sarcopenia for patients with recent SARS-CoV-2 history (14 days). A prospective study was performed on 200 patients (between December 2020−December 2021), with SPPB score < 9, randomly divided into: Group K—DP (93 patients) with dietary therapy (protein 1.2−1.5 g/kg) and probiotics for two months; and Group K—non-DP (107 patients) without diet therapy and probiotics. All patients were included in a specific physical training program (40 min), three sessions per week. Skeletal muscle index (SMI), serum albumin, and hemoglobin were determined. The SMI was initially low for both groups without significant statistical differences (6.5 ± 0.52 kg/m2 for Group K—non-DP vs. 6.7 ± 0.57 Kg/m2 for Group K—DP, p = 0.135). After two months, significant difference between initial and final SMI values was determined for Group K—DP (6.92 ± 0.50 kg/m2 vs. 6.77 ± 0.56 kg/m2, p = 0.048). In Group K—DP, at end of study, were more patients with normal SMI (n = 32 → N = 70) values (p < 0.001) and fewer sarcopenia patients (p < 0.001). The initial serum albumin means values in the two groups (Group K—non-DP, 4.17 ± 1.04 g/dL, and Group K—DP, 3.95 ± 0.98 g/dL) were not statistically significantly different (p = 0.122). The hemoglobin level improved significantly following a hyper protein diet enriched with pro-biotics (p = 0.003). Diet therapy, consisting of increased protein intake and specific probiotics and specific physical therapy, demonstrated superiority in improving the functional status of patients with recent COVID-19 infection.
Collapse
|
26
|
Zhang Y, Jiang L, Su P, Ma Z, Kang W, Ye X, Liu Y, Yu J. Association between Plasma FGF21 Levels and Body Composition in Patients with Gastric Cancer. Nutr Cancer 2022; 75:349-356. [PMID: 36190321 DOI: 10.1080/01635581.2022.2118322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Accumulating evidence has suggested that Fibroblast growth factor 21 (FGF21) plays an important role in metabolic diseases. This study aimed to investigate the relationship between plasma FGF21 levels and body composition parameters in gastric cancer (GC) patients. METHODS This study was cross-sectional based on a prospective cohort of GC patients in a single center. Computer tomography (CT) and bioelectrical impedance analysis (BIA) were used to estimate skeletal muscle and adipose tissue mass. Blood samples were collected and plasma concentrations of FGF21 were measured by ELISA. Spearman's rank correlation test and logistic regression analysis were performed to assess associations between plasma FGF21 levels and these body composition parameters. RESULTS A total of 66 GC patients were enrolled in this study. Plasma FGF21 levels were significantly higher in women compared with men. The plasma FGF21 levels were positively correlated with fat mass index (FMI), fat mass percentage (FM%), and subcutaneous adipose tissue index (SATI). Furthermore, after adjustment for confounders, the lower plasma FGF21 levels were remain associated with increased odds for low SATI. CONCLUSIONS Plasma FGF21 levels were positively associated with FMI, FM%, and SATI in GC patients, suggesting a potential mechanistic link between FGF21 and subcutaneous adipose tissue in GC.
Collapse
Affiliation(s)
- Yingjing Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Jiang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pengfei Su
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiqiang Ma
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weiming Kang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Ye
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuqin Liu
- Department of Pathology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianchun Yu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
27
|
Acevedo‐Acevedo S, Stefkovich ML, Kang SWS, Cunningham RP, Cultraro CM, Porat‐Shliom N. LKB1 acts as a critical brake for the glucagon-mediated fasting response. Hepatol Commun 2022; 6:1949-1961. [PMID: 35357082 PMCID: PMC9315124 DOI: 10.1002/hep4.1942] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/23/2022] [Accepted: 03/06/2022] [Indexed: 11/08/2022] Open
Abstract
As important as the fasting response is for survival, an inability to shut it down once nutrients become available can lead to exacerbated disease and severe wasting. The liver is central to transitions between feeding and fasting states, with glucagon being a key initiator of the hepatic fasting response. However, the precise mechanisms controlling fasting are not well defined. One potential mediator of these transitions is liver kinase B1 (LKB1), given its role in nutrient sensing. Here, we show LKB1 knockout mice have a severe wasting and prolonged fasting phenotype despite increased food intake. By applying RNA sequencing and intravital microscopy, we show that loss of LKB1 leads to a dramatic reprogramming of the hepatic lobule through robust up-regulation of periportal genes and functions. This is likely mediated through the opposing effect that LKB1 has on glucagon pathways and gene expression. Conclusion: Our findings show that LKB1 acts as a brake to the glucagon-mediated fasting response, resulting in "periportalization" of the hepatic lobule and whole-body metabolic inefficiency. These findings reveal a mechanism by which hepatic metabolic compartmentalization is regulated by nutrient-sensing.
Collapse
Affiliation(s)
- Suehelay Acevedo‐Acevedo
- Thoracic and GI Malignancies BranchCenter for Cancer ResearchNational Cancer Institute, NIHBethesdaMarylandUSA
| | - Megan L. Stefkovich
- Thoracic and GI Malignancies BranchCenter for Cancer ResearchNational Cancer Institute, NIHBethesdaMarylandUSA
| | - Sun Woo Sophie Kang
- Thoracic and GI Malignancies BranchCenter for Cancer ResearchNational Cancer Institute, NIHBethesdaMarylandUSA
| | - Rory P. Cunningham
- Thoracic and GI Malignancies BranchCenter for Cancer ResearchNational Cancer Institute, NIHBethesdaMarylandUSA
| | - Constance M. Cultraro
- Thoracic and GI Malignancies BranchCenter for Cancer ResearchNational Cancer Institute, NIHBethesdaMarylandUSA
| | - Natalie Porat‐Shliom
- Thoracic and GI Malignancies BranchCenter for Cancer ResearchNational Cancer Institute, NIHBethesdaMarylandUSA
| |
Collapse
|
28
|
Huang Q, Wu M, Wu X, Zhang Y, Xia Y. Muscle-to-tumor crosstalk: The effect of exercise-induced myokine on cancer progression. Biochim Biophys Acta Rev Cancer 2022; 1877:188761. [PMID: 35850277 DOI: 10.1016/j.bbcan.2022.188761] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 07/11/2022] [Accepted: 07/11/2022] [Indexed: 02/07/2023]
Abstract
Physical exercise has gradually become a focus in cancer treatment due to its pronounced role in reducing cancer risk, enhancing therapeutic efficacy, and improving prognosis. In recent decades, skeletal muscles have been considered endocrine organs, exerting their biological functions via the endocrine, autocrine, and paracrine systems by secreting various types of myokines. The amount of myokines secreted varies depending on the intensity, type, and duration of exercise. Recent studies have shown that muscle-derived myokines are highly involved the effects of exercise on cancer. Multiple myokines, such as interleukin-6 (IL-6), oncostatin M (OSM), secreted protein acidic and rich in cysteine (SPARC), and irisin, directly mediate cancer progression by influencing the proliferation, apoptosis, stemness, drug resistance, metabolic reprogramming, and epithelial-mesenchymal transformation (EMT) of cancer cells. In addition, IL-6, interleukin-8 (IL-8), interleukin-15 (IL-15), brain-derived neurotrophic factor (BDNF), and irisin can improve obesity-induced inflammation by stimulating lipolysis of adipose tissues, promoting glucose uptake, and accelerating the browning of white fat. Furthermore, some myokines could regulate the tumor microenvironment, such as angiogenesis and the immune microenvironment. Cancer cachexia occurs in up to 80% of cancer patients and is responsible for 22%-30% of patient deaths. It is characterized by systemic inflammation and decreased muscle mass. Exercise-induced myokine production is important in regulating cancer cachexia. This review summarizes the roles and underlying mechanisms of myokines, such as IL-6, myostatin, IL-15, irisin, fibroblast growth factor 21 (FGF21) and musclin, in cancer cachexia. Through comprehensive analysis, we conclude that myokines are potential targets for inhibiting cancer progression and the associated cachexia.
Collapse
Affiliation(s)
- Qianrui Huang
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mengling Wu
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuyi Wu
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu 610041, China
| | - Yiwen Zhang
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yong Xia
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu 610041, China.
| |
Collapse
|
29
|
Research Progress of Fibroblast Growth Factor 21 in Fibrotic Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5042762. [PMID: 35677107 PMCID: PMC9168133 DOI: 10.1155/2022/5042762] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/04/2022] [Accepted: 05/10/2022] [Indexed: 11/24/2022]
Abstract
Fibrosis is a common pathological outcome of chronic injuries, characterized by excessive deposition of extracellular matrix components in organs, as seen in most chronic inflammatory diseases. At present, there is an increasing tendency of the morbidity and mortality of diseases caused by fibrosis, but the treatment measures for fibrosis are still limited. Fibroblast growth factor 21 (FGF21) belongs to the FGF19 subfamily, which also has the name endocrine FGFs because of their endocrine manner. In recent years, it has been found that plasma FGF21 level is significantly correlated with fibrosis progression. Furthermore, there is evidence that FGF21 has a pronounced antifibrotic effect in a variety of fibrotic diseases. This review summarizes the biological effects of FGF21 and discusses what is currently known about this factor and fibrosis disease, highlighting emerging insights that warrant further research.
Collapse
|
30
|
Chiu LT, Wang CH, Lin YL, Hsu BG. Association of serum fibroblast growth factor 21 levels with skeletal muscle mass and mortality in chronic hemodialysis patients. J Formos Med Assoc 2022; 121:2481-2489. [DOI: 10.1016/j.jfma.2022.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 01/11/2023] Open
|
31
|
Alizadeh Pahlavani H. Exercise Therapy for People With Sarcopenic Obesity: Myokines and Adipokines as Effective Actors. Front Endocrinol (Lausanne) 2022; 13:811751. [PMID: 35250869 PMCID: PMC8892203 DOI: 10.3389/fendo.2022.811751] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/24/2022] [Indexed: 12/15/2022] Open
Abstract
Sarcopenic obesity is defined as a multifactorial disease in aging with decreased body muscle, decreased muscle strength, decreased independence, increased fat mass, due to decreased physical activity, changes in adipokines and myokines, and decreased satellite cells. People with sarcopenic obesity cause harmful changes in myokines and adipokines. These changes are due to a decrease interleukin-10 (IL-10), interleukin-15 (IL-15), insulin-like growth factor hormone (IGF-1), irisin, leukemia inhibitory factor (LIF), fibroblast growth factor-21 (FGF-21), adiponectin, and apelin. While factors such as myostatin, leptin, interleukin-6 (IL-6), interleukin-8 (IL-8), and resistin increase. The consequences of these changes are an increase in inflammatory factors, increased degradation of muscle proteins, increased fat mass, and decreased muscle tissue, which exacerbates sarcopenia obesity. In contrast, exercise, especially strength training, reverses this process, which includes increasing muscle protein synthesis, increasing myogenesis, increasing mitochondrial biogenesis, increasing brown fat, reducing white fat, reducing inflammatory factors, and reducing muscle atrophy. Since some people with chronic diseases are not able to do high-intensity strength training, exercises with blood flow restriction (BFR) are newly recommended. Numerous studies have shown that low-intensity BFR training produces the same increase in hypertrophy and muscle strength such as high-intensity strength training. Therefore, it seems that exercise interventions with BFR can be an effective way to prevent the exacerbation of sarcopenia obesity. However, due to limited studies on adipokines and exercises with BFR in people with sarcopenic obesity, more research is needed.
Collapse
|
32
|
Wu CT, Chaffin AT, Ryan KK. Fibroblast Growth Factor 21 Facilitates the Homeostatic Control of Feeding Behavior. J Clin Med 2022; 11:580. [PMID: 35160033 PMCID: PMC8836936 DOI: 10.3390/jcm11030580] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/01/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) is a stress hormone that is released from the liver in response to nutritional and metabolic challenges. In addition to its well-described effects on systemic metabolism, a growing body of literature now supports the notion that FGF21 also acts via the central nervous system to control feeding behavior. Here we review the current understanding of FGF21 as a hormone regulating feeding behavior in rodents, non-human primates, and humans. First, we examine the nutritional contexts that induce FGF21 secretion. Initial reports describing FGF21 as a 'starvation hormone' have now been further refined. FGF21 is now better understood as an endocrine mediator of the intracellular stress response to various nutritional manipulations, including excess sugars and alcohol, caloric deficits, a ketogenic diet, and amino acid restriction. We discuss FGF21's effects on energy intake and macronutrient choice, together with our current understanding of the underlying neural mechanisms. We argue that the behavioral effects of FGF21 function primarily to maintain systemic macronutrient homeostasis, and in particular to maintain an adequate supply of protein and amino acids for use by the cells.
Collapse
Affiliation(s)
| | | | - Karen K. Ryan
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA 95616, USA; (C.-T.W.); (A.T.C.)
| |
Collapse
|
33
|
Bag Soytas R, Suzan V, Arman P, Emiroglu Gedik T, Unal D, Cengiz M, Bolayirli IM, Suna Erdincler D, Doventas A, Yavuzer H. Association of FGF-19 and FGF-21 levels with primary sarcopenia. Geriatr Gerontol Int 2021; 21:959-962. [PMID: 34405516 DOI: 10.1111/ggi.14263] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/12/2021] [Accepted: 07/31/2021] [Indexed: 01/26/2023]
Abstract
AIM Serum fibroblast growth factor (FGF)-19 and FGF-21 levels have been reported to be associated with muscle hemostasis. This study aims to explore the relationship between the levels of these markers and sarcopenia. METHODS In our single-center, cross-sectional study, patients over 65 years old presenting to the geriatric outpatient clinic were included. Patients with secondary sarcopenia were excluded. The Strength-Assistance with walking-Rising from a chair-Climbing stairs and Falls (SARC-F) questionnaire was applied to all patients. Sarcopenia was determined by handgrip strength (HGS), bioelectrical impedance analysis and 6-m walk test. Serum samples were stored at -80°C until measurement. The ELISA method was used to assess FGF-19 and FGF-21 levels. RESULTS In total, 88 patients (54 women) were included. There were 43 patients in the sarcopenia group and 45 patients without sarcopenia in the control group. In those with sarcopenia, FGF-19 was lower (P = 0.04) and FGF-21 was higher (P = 0.021). There was a direct correlation between FGF-19 with SARC-F and HGS (P = 0.04, B = 0.178, P = 0.006, B = 0.447) while FGF-21 was inversely correlated with HGS and positively correlated with 6-m walking time (P = 0.016, B = -0.428, P = 0.004, B = 0.506). CONCLUSIONS Our results reveal that low FGF-19 and high FGF-21 may be associated with sarcopenia and this finding could be explained by the impacted muscle strength. Geriatr Gerontol Int 2021; 21: 959-962.
Collapse
Affiliation(s)
- Rabia Bag Soytas
- Division of Geriatrics, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Veysel Suzan
- Division of Geriatrics, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Pinar Arman
- Division of Geriatrics, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Tugce Emiroglu Gedik
- Division of Geriatrics, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Damla Unal
- Division of Geriatrics, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Mahir Cengiz
- Department of Internal Medicine, Biruni University Medical Faculty, Istanbul, Turkey
| | - Ibrahim Murat Bolayirli
- Department of Biochemistry, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Deniz Suna Erdincler
- Division of Geriatrics, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Alper Doventas
- Division of Geriatrics, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Hakan Yavuzer
- Division of Geriatrics, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
34
|
Cao Z, Zhao K, Jose I, Hoogenraad NJ, Osellame LD. Biomarkers for Cancer Cachexia: A Mini Review. Int J Mol Sci 2021; 22:4501. [PMID: 33925872 PMCID: PMC8123431 DOI: 10.3390/ijms22094501] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/14/2021] [Accepted: 04/23/2021] [Indexed: 01/08/2023] Open
Abstract
Cancer cachexia is a common condition in many cancer patients, particularly those with advanced disease. Cancer cachexia patients are generally less tolerant to chemotherapies and radiotherapies, largely limiting their treatment options. While the search for treatments of this condition are ongoing, standards for the efficacy of treatments have yet to be developed. Current diagnostic criteria for cancer cachexia are primarily based on loss of body mass and muscle function. However, these criteria are rather limiting, and in time, when weight loss is noticeable, it may be too late for treatment. Consequently, biomarkers for cancer cachexia would be valuable adjuncts to current diagnostic criteria, and for assessing potential treatments. Using high throughput methods such as "omics approaches", a plethora of potential biomarkers have been identified. This article reviews and summarizes current studies of biomarkers for cancer cachexia.
Collapse
Affiliation(s)
- Zhipeng Cao
- Department of Biochemistry and Genetics, La Trobe University, Bundoora, VIC 3086, Australia; (K.Z.); (I.J.); (N.J.H.)
| | - Kening Zhao
- Department of Biochemistry and Genetics, La Trobe University, Bundoora, VIC 3086, Australia; (K.Z.); (I.J.); (N.J.H.)
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Irvin Jose
- Department of Biochemistry and Genetics, La Trobe University, Bundoora, VIC 3086, Australia; (K.Z.); (I.J.); (N.J.H.)
| | - Nick J. Hoogenraad
- Department of Biochemistry and Genetics, La Trobe University, Bundoora, VIC 3086, Australia; (K.Z.); (I.J.); (N.J.H.)
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Laura D. Osellame
- Department of Biochemistry and Genetics, La Trobe University, Bundoora, VIC 3086, Australia; (K.Z.); (I.J.); (N.J.H.)
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| |
Collapse
|