1
|
Wang C, Liu X, Hu X, Wu T, Duan R. Therapeutic targeting of GDF11 in muscle atrophy: Insights and strategies. Int J Biol Macromol 2024; 279:135321. [PMID: 39236952 DOI: 10.1016/j.ijbiomac.2024.135321] [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: 02/19/2024] [Revised: 07/29/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
The exploration of novel therapeutic avenues for skeletal muscle atrophy is imperative due to its significant health impact. Recent studies have spotlighted growth differentiation factor 11 (GDF11), a TGFβ superfamily member, for its rejuvenating role in reversing age-related tissue dysfunction. This review synthesizes current findings on GDF11, elucidating its distinct biological functions and the ongoing debates regarding its efficacy in muscle homeostasis. By addressing discrepancies in current research outcomes and its ambiguous role due to its homological identity to myostatin, a negative regulator of muscle mass, this review aims to clarify the role of GDF11 in muscle homeostasis and its potential as a therapeutic target for muscle atrophy. Through a thorough examination of GDF11's mechanisms and effects, this review provides insights that could pave the way for innovative treatments for muscle atrophy, emphasizing the need and strategies to boost endogenous GDF11 levels for therapeutic potential.
Collapse
Affiliation(s)
- Chuanzhi Wang
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Xiaocao Liu
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Xilong Hu
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Tao Wu
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Rui Duan
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China.
| |
Collapse
|
2
|
Dagbasi A, Fuller A, Hanyaloglu AC, Carroll B, McLaughlin J, Frost G, Holliday A. The role of nutrient sensing dysregulation in anorexia of ageing: The little we know and the much we don't. Appetite 2024; 203:107718. [PMID: 39423861 DOI: 10.1016/j.appet.2024.107718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/01/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
The age-related decline in appetite and food intake - termed "anorexia of ageing" - is implicated in undernutrition in later life and hence provides a public health challenge for our ageing population. Eating behaviour is controlled, in part, by homeostatic mechanisms which sense nutrient status and provide feedback to appetite control regions of the brain. Such feedback signals, propagated by episodic gut hormones, are dysregulated in some older adults. The secretory responses of appetite-related gut hormones to feeding are amplified, inducing a more anorexigenic signal which is associated with reduced appetite and food intake. Such an augmented response would indicate an increase in gut sensitivity to nutrients. Consequently, this review explores the role of gastrointestinal tract nutrient sensing in age-related appetite dysregulation. We review and synthesise evidence for age-related alterations in nutrient sensing which may explain the observed hormonal dysregulation. Drawing on what is known regarding elements of nutrient sensing pathways in animal models, in other tissues of the body, and in certain models of disease, we identify potential causal mechanisms including alterations in enteroendocrine cell number and distribution, dysregulation of cell signalling pathways, and changes in the gut milieu. From identified gaps in evidence, we highlight interesting and important avenues for future research.
Collapse
Affiliation(s)
- Aygul Dagbasi
- Section of Nutrition, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London, W12 0NN, UK
| | - Amy Fuller
- Research Centre for Health and Life Sciences, Institute of Health and Wellbeing, Faculty of Health and Life Science, Coventry University, Coventry, CV1 5FB, UK
| | - Aylin C Hanyaloglu
- Institute of Reproductive and Developmental Biology (IRDB), Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Bernadette Carroll
- School of Biochemistry, University of Bristol, University Walk, Bristol, BS1 8TD, UK
| | - John McLaughlin
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester and Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK
| | - Gary Frost
- Section of Nutrition, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London, W12 0NN, UK
| | - Adrian Holliday
- School of Biomedical, Nutritional, and Sport Science, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK; Human Nutrition and Exercise Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK.
| |
Collapse
|
3
|
Fan X, Wang Y, Zhang Z, Yang R, Zhou Y, Gu J. Assessing the causal relationship between frailty and sex hormone-binding globulin or insulin-like growth factor-1 levels: A sex-stratified bidirectional Mendelian Randomization study. Exp Gerontol 2024; 195:112545. [PMID: 39154868 DOI: 10.1016/j.exger.2024.112545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 07/07/2024] [Accepted: 08/12/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND The association between frailty and sex hormone-binding globulin (SHBG) or insulin-like growth factor-1(IGF-1) levels demonstrates sex differences with inconsistent conclusions. This study aims to explore the causal relationship between frailty and SHBG or IGF-1 levels through bidirectional Mendelian randomization (MR). METHODS We conducted two-sample bidirectional sex-stratified MR analyses using summary-level data from genome-wide association studies (GWASs) to examine the causal relationship between frailty and IGF-1 or SHBG levels, as measured by frailty index (FI) and frailty phenotype (FP). We use the random-effects inverse-variance weighted (IVW), weighted median, MR-Egger, MR-Egger intercept, and leave-one-out approaches. RESULT The relationship between frailty and SHBG or IGF-1 levels is inversely related, with a significant decrease in SHBG levels in females. Specifically, SHBG levels significantly decrease with FI (β = -5.49; 95 % CI: -9.67 to -1.32; FDR = 0.02) and more pronounced with FP (β = -10.14; 95 % CI: -16.16 to -4.13; FDR = 0.01), as determined by the IVW approach. However, reverse analysis shows no significant effect of IGF-1 or SHBG levels on either FI or FP (p > 0.05). CONCLUSION Our study indicates a negative correlation between frailty and the levels of SHBG and IGF-1. It is suggested that further research is required to establish cut-off values for SHBG and IGF-1 levels in the frailty population. This is particularly important for females at higher risk, such as those undergoing menopause, to enable comprehensive assessment and early prevention efforts. While the findings imply that reduced IGF-1 and SHBG levels may not directly contribute to frailty, it is important not to overlook the underlying mechanisms through which they may indirectly influence frailty.
Collapse
Affiliation(s)
- Xinying Fan
- Department of General Practice, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Yuxin Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310003, Zhejiang, China
| | - Zhaoyu Zhang
- Department of General Practice, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Runjun Yang
- Department of Nuclear Medicine, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Yajing Zhou
- Department of Biostatistics, NHC Key Laboratory for Health Technology Assessment, Key Laboratory of Public Health Safety of Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Jie Gu
- Department of General Practice, Zhongshan Hospital of Fudan University, Shanghai 200032, China; International Medical Center, Zhongshan Hospital of Fudan University, Shanghai 200032, China.
| |
Collapse
|
4
|
Hsu WH, Wang SY, Chao YM, Chang KV, Han DS, Lin YL. Novel metabolic and lipidomic biomarkers of sarcopenia. J Cachexia Sarcopenia Muscle 2024; 15:2175-2186. [PMID: 39169398 PMCID: PMC11446726 DOI: 10.1002/jcsm.13567] [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: 08/23/2023] [Revised: 06/26/2024] [Accepted: 07/18/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND The pathophysiology of sarcopenia is complex and multifactorial and has not been fully elucidated. The impact of resistance training and nutritional support (RTNS) on metabolomics and lipodomics in older adults with sarcopenia remains uncertain. This study aimed to explore potential biomarkers of sarcopenia and clinical indicators of RTNS in older sarcopenic adults. METHODS Older individuals diagnosed with sarcopenia through routine health checkups at a community hospital were recruited for a 12-week randomized controlled trial focusing on RTNS. Plasma metabolomic and lipidomic profiles of 45 patients with sarcopenia and 47 matched controls were analysed using 1H-nuclear magnetic resonance (1H-NMR) and liquid chromatography-mass spectrometer (LC-MS). RESULTS At baseline, the patient and control groups had similar age, sex, and height distribution. The patient group had significantly lower weight, BMI, grip strength, gait speed, skeletal muscle index, lean mass of both the upper and lower limbs, and lower limb bone mass. There was a significant difference in 12 metabolites between the control and patient groups. They are isoleucine (patient/control fold change [FC] = 0.86 ± 0.04, P = 0.0005), carnitine (FC = 1.05 ± 0.01, P = 0.0110), 1-methylhistamine/3-methylhistamine (FC = 1.24 ± 0.14, P = 0.0039), creatinine (FC = 0.71 ± 0.04, P < 0.0001), carnosine (FC = 0.71 ± 0.04, P = 0.0007), ureidopropionic acid (FC = 0.61 ± 0.10, P = 0.0107), uric acid (FC = 0.88 ± 0.03, P = 0.0083), PC (18:2/20:0) (FC = 0.69 ± 0.03, P = 0.0010), PC (20:2/18:0) (FC = 0.70 ± 0.06, P = 0.0014), PC (18:1/20:1) (FC = 0.74 ± 0.05, P = 0.0015), PI 32:1 (FC = 4.72 ± 0.17, P = 0.0006), and PI 34:3 (FC = 1.88 ± 0.13, P = 0.0003). Among them, carnitine, 1-methylhistamine/3-methylhistamine, creatinine, ureidopropionic acid, uric acid, PI 32:1, and PI 34:3 were first identified. Notably, PI 32:1 had highest diagnostic accuracy (0.938) for sarcopenia. 1-Methylhistamine/3-methylhistamine, carnosine, PC (18:2/20:0), PI 32:1, and PI 34:3 levels were not different from the control group after RTNS. These metabolites are involved in amino acid metabolism, lipid metabolism, and the PI3K-AKT/mTOR signalling pathway through the ingenuity pathway analysis. CONCLUSIONS These findings provide information on metabolic changes, lipid perturbations, and the role of RTNS in patients with sarcopenia. They reveal new insights into its pathological mechanisms and potential therapies.
Collapse
Affiliation(s)
- Wei-Hsiang Hsu
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
- Institute of Biopharmaceutical Sciences, National Yang-Ming-Chiao-Tung University, Taipei, Taiwan
| | - San-Yuan Wang
- Master Program in Clinical Genomics and Proteomics, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Yen-Ming Chao
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Ke-Vin Chang
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei, Taiwan
- Department of Physical Medicine and Rehabilitation, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Der-Sheng Han
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei, Taiwan
- Department of Physical Medicine and Rehabilitation, National Taiwan University College of Medicine, Taipei, Taiwan
- Health Science and Wellness Center, National Taiwan University, Taipei, Taiwan
| | - Yun-Lian Lin
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
- Department of Pharmacy, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
5
|
Grima-Terrén M, Campanario S, Ramírez-Pardo I, Cisneros A, Hong X, Perdiguero E, Serrano AL, Isern J, Muñoz-Cánoves P. Muscle aging and sarcopenia: The pathology, etiology, and most promising therapeutic targets. Mol Aspects Med 2024; 100:101319. [PMID: 39312874 DOI: 10.1016/j.mam.2024.101319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/25/2024]
Abstract
Sarcopenia is a progressive muscle wasting disorder that severely impacts the quality of life of elderly individuals. Although the natural aging process primarily causes sarcopenia, it can develop in response to other conditions. Because muscle function is influenced by numerous changes that occur with age, the etiology of sarcopenia remains unclear. However, recent characterizations of the aging muscle transcriptional landscape, signaling pathway disruptions, fiber and extracellular matrix compositions, systemic metabolomic and inflammatory responses, mitochondrial function, and neurological inputs offer insights and hope for future treatments. This review will discuss age-related changes in healthy muscle and our current understanding of how this can deteriorate into sarcopenia. As our elderly population continues to grow, we must understand sarcopenia and find treatments that allow individuals to maintain independence and dignity throughout an extended lifespan.
Collapse
Affiliation(s)
- Mercedes Grima-Terrén
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain
| | - Silvia Campanario
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain
| | - Ignacio Ramírez-Pardo
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain
| | - Andrés Cisneros
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain
| | - Xiaotong Hong
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA
| | | | - Antonio L Serrano
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA
| | - Joan Isern
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA
| | - Pura Muñoz-Cánoves
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain.
| |
Collapse
|
6
|
Huang CY, Liu YH. Sex difference, proteostasis and mitochondrial function impact stroke-related sarcopenia-A systematic review and meta-analysis. Ageing Res Rev 2024; 101:102484. [PMID: 39218079 DOI: 10.1016/j.arr.2024.102484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 08/11/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The prevalence of stroke-related sarcopenia has been noted; however, epidemiological data and interventions that increase or reduce the incidence of stroke-related sarcopenia remain lacking. METHODS Studies on stroke-related sarcopenia were included in association or interventional analyses. All analyses were performed using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Two evaluators independently extracted the data. RESULTS Female stroke patients had a higher preference for sarcopenia than male patients (pooled odds ratio [OR] = 0.670, 95 % CI 0.533-0.842, p = 0.001). Although stroke patients without drug use have improved skeletal muscle mass index (SMI) (MD = 0.272, 95 % CI 0.087-0.457, p = 0.004), handgrip strength (HGS) was not significantly altered (MD = -0.068, 95 % CI -0.221-0.076, p = 0.354). Stroke patients with nutrient interventions have improved SMI (MD = -0.354, 95 % CI -0.635- -0.073, p = 0.014) and HGS (MD = -0.394, 95 % CI -0.678- -0.111, p = 0.006); the synergistic effect of rehabilitation exercise has not been ruled out. Whether a sex difference exists in these interventions remains to be investigated. The underlying pathological mechanisms and potential therapeutic strategies for this disease are discussed. CONCLUSION Sex difference, proteostasis, and mitochondrial function may impact the incidence of stroke-related sarcopenia. Understanding the underlying pathological mechanisms and potential therapeutic targets for this disease will provide new insights into disease treatment, prevention, and drug development.
Collapse
Affiliation(s)
- Chien-Yu Huang
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung 404328, Taiwan; Department of Chinese Medicine, China Medical University Hospital, Taichung 404333, Taiwan
| | - Yu-Huei Liu
- Graduate Institute of Integrated Medicine, China Medical University, Taichung 404333, Taiwan; Department of Medical Genetics and Medical Research, China Medical University Hospital, Taichung 404328, Taiwan; Drug Development Center, China Medical University, Taichung 404333, Taiwan.
| |
Collapse
|
7
|
Ataman M, Mittal N, Tintignac L, Schmidt A, Ham DJ, González A, Ruegg MA, Zavolan M. Calorie restriction and rapamycin distinctly mitigate aging-associated protein phosphorylation changes in mouse muscles. Commun Biol 2024; 7:974. [PMID: 39127848 DOI: 10.1038/s42003-024-06679-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
Calorie restriction (CR) and treatment with rapamycin (RM), an inhibitor of the mTORC1 growth-promoting signaling pathway, are known to slow aging and promote health from worms to humans. At the transcriptome and proteome levels, long-term CR and RM treatments have partially overlapping effects, while their impact on protein phosphorylation within cellular signaling pathways have not been compared. Here we measured the phosphoproteomes of soleus, tibialis anterior, triceps brachii and gastrocnemius muscles from adult (10 months) and 30-month-old (aged) mice receiving either a control, a calorie restricted or an RM containing diet from 15 months of age. We reproducibly detected and extensively analyzed a total of 6960 phosphosites, 1415 of which are not represented in standard repositories. We reveal the effect of these interventions on known mTORC1 pathway substrates, with CR displaying greater between-muscle variation than RM. Overall, CR and RM have largely consistent, but quantitatively distinct long-term effects on the phosphoproteome, mitigating age-related changes to different degrees. Our data expands the catalog of protein phosphorylation sites in the mouse, providing important information regarding their tissue-specificity, and revealing the impact of long-term nutrient-sensing pathway inhibition on mouse skeletal muscle.
Collapse
Affiliation(s)
- Meric Ataman
- Biozentrum, University of Basel, Basel, Switzerland.
- Swiss Institute of Bioinformatics, Basel, Switzerland.
| | | | - Lionel Tintignac
- Department of Neurology and Biomedicine, University of Basel; University Hospital Basel, Basel, Switzerland
| | | | - Daniel J Ham
- Biozentrum, University of Basel, Basel, Switzerland
| | - Asier González
- Biozentrum, University of Basel, Basel, Switzerland
- Departament de Bioquímica i Biologia Molecular and Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | | | - Mihaela Zavolan
- Biozentrum, University of Basel, Basel, Switzerland.
- Swiss Institute of Bioinformatics, Basel, Switzerland.
| |
Collapse
|
8
|
Benavente C, Padial P, Scott BR, Almeida F, Olcina G, Pérez-Regalado S, Feriche B. Strength and muscle mass development after a resistance-training period at terrestrial and normobaric intermittent hypoxia. Pflugers Arch 2024; 476:1221-1233. [PMID: 38916665 PMCID: PMC11271399 DOI: 10.1007/s00424-024-02978-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/20/2024] [Accepted: 06/03/2024] [Indexed: 06/26/2024]
Abstract
This study investigated the effect of a resistance training (RT) period at terrestrial (HH) and normobaric hypoxia (NH) on both muscle hypertrophy and maximal strength development with respect to the same training in normoxia (N). Thirty-three strength-trained males were assigned to N (FiO2 = 20.9%), HH (2,320 m asl) or NH (FiO2 = 15.9%). The participants completed an 8-week RT program (3 sessions/week) of a full body routine. Muscle thickness of the lower limb and 1RM in back squat were assessed before and after the training program. Blood markers of stress, inflammation (IL-6) and muscle growth (% active mTOR, myostatin and miRNA-206) were measured before and after the first and last session of the program. Findings revealed all groups improved 1RM, though this was most enhanced by RT in NH (p = 0.026). According to the moderate to large excess of the exercise-induced stress response (lactate and Ca2+) in HH and N, results only displayed increases in muscle thickness in these two conditions over NH (ES > 1.22). Compared with the rest of the environmental conditions, small to large increments in % active mTOR were only found in HH, and IL-6, myostatin and miR-206 in NH throughout the training period. In conclusion, the results do not support the expected additional benefit of RT under hypoxia compared to N on muscle growth, although it seems to favour gains in strength. The greater muscle growth achieved in HH over NH confirms the impact of the type of hypoxia on the outcomes.
Collapse
Affiliation(s)
- C Benavente
- Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, Granada, Spain
| | - P Padial
- Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, Granada, Spain
| | - B R Scott
- Centre for Healthy Ageing, Murdoch University, Perth, Australia
- PHysical Activity, Sport and Exercise (PHASE) Research Group, School of Allied Health (Exercise Science), Murdoch University, Perth, Australia
| | - F Almeida
- Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, Granada, Spain
| | - G Olcina
- Faculty of Sport Sciences, University of Extremadura, Cáceres, Spain
| | - S Pérez-Regalado
- Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, Granada, Spain
| | - B Feriche
- Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, Granada, Spain.
| |
Collapse
|
9
|
Kashani B, Zandi Z, Pourbagheri-Sigaroodi A, Yousefi AM, Ghaffari SH, Bashash D. The PI3K signaling pathway; from normal lymphopoiesis to lymphoid malignancies. Expert Rev Anticancer Ther 2024; 24:493-512. [PMID: 38690706 DOI: 10.1080/14737140.2024.2350629] [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: 12/29/2023] [Accepted: 04/29/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION As a vital mechanism of survival, lymphopoiesis requires the collaboration of different signaling molecules to orchestrate each step of cell development and maturation. The PI3K pathway is considerably involved in the maturation of lymphatic cells and therefore, its dysregulation can immensely affect human well-being and cause some of the most prevalent malignancies. As a result, studies that investigate this pathway could pave the way for a better understanding of the lymphopoiesis mechanisms, the undesired changes that lead to cancer progression, and how to design drugs to solve this issue. AREAS COVERED The present review addresses the aforementioned aspects of the PI3K pathway and helps pave the way for future therapeutic approaches. In order to access the articles, databases such as Medicine Medline/PubMed, Scopus, Google Scholar, and Science Direct were utilized. The search formula was established by identifying main keywords including PI3K/Akt/mTOR pathway, Lymphopoiesis, Lymphoid malignancies, and inhibitors. EXPERT OPINION The PI3K pathway is crucial for lymphocyte development and differentiation, making it a potential target for therapeutic intervention in lymphoid cancers. Studies are focused on developing PI3K inhibitors to impede the progression of hematologic malignancies, highlighting the pathway's significance in lymphoma and lymphoid leukemia.
Collapse
Affiliation(s)
- Bahareh Kashani
- Hematology, Oncology and Stem Cell Transplantation Research Center, School of Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Zandi
- Hematology, Oncology and Stem Cell Transplantation Research Center, School of Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir-Mohammad Yousefi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed H Ghaffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, School of Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Tsai SY. Lost in translation: challenges of current pharmacotherapy for sarcopenia. Trends Mol Med 2024:S1471-4914(24)00138-2. [PMID: 38880726 DOI: 10.1016/j.molmed.2024.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/18/2024]
Abstract
A healthy lifespan relies on independent living, in which active skeletal muscle is a critical element. The cost of not recognizing and acting earlier on unhealthy or aging muscle could be detrimental, since muscular weakness is inversely associated with all-cause mortality. Sarcopenia is characterized by a decline in skeletal muscle mass and strength and is associated with aging. Exercise is the only effective therapy to delay sarcopenia development and improve muscle health in older adults. Although numerous interventions have been proposed to reduce sarcopenia, none has yet succeeded in clinical trials. This review evaluates the biological gap between recent clinical trials targeting sarcopenia and the preclinical studies on which they are based, and suggests an alternative approach to bridge the discrepancy.
Collapse
Affiliation(s)
- Shih-Yin Tsai
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
11
|
Meyer NMT, Kabisch S, Dambeck U, Honsek C, Kemper M, Gerbracht C, Arafat AM, Birkenfeld AL, Schwarz PEH, Machann J, Osterhoff MA, Weickert MO, Pfeiffer AFH. IGF-1 and IGFBP-1 as Possible Predictors of Response to Lifestyle Intervention-Results from Randomized Controlled Trials. Int J Mol Sci 2024; 25:6400. [PMID: 38928106 PMCID: PMC11203659 DOI: 10.3390/ijms25126400] [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: 04/27/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Lifestyle interventions can prevent type 2 diabetes (T2DM). However, some individuals do not experience anticipated improvements despite weight loss. Biomarkers to identify such individuals at early stages are lacking. Insulin-like growth factor 1 (IGF- 1) and Insulin-like growth factor binding protein 1(IGFBP-1) were shown to predict T2DM onset in prediabetes. We assessed whether these markers also predict the success of lifestyle interventions, thereby possibly guiding personalized strategies. We analyzed the fasting serum levels of IGF-1, IGFBP-1, and Insulin-like growth factor binding protein 2 (IGFBP-2) in relation to changes in metabolic and anthropometric parameters, including intrahepatic lipids (IHLs) and visceral adipose tissue (VAT) volume, measured by magnetic resonance imaging (MRI), in 345 participants with a high risk for prediabetes (54% female; aged 36-80 years). Participants were enrolled in three randomized dietary intervention trials and assessed both at baseline and one year post-intervention. Statistical analyses were performed using IBM SPSS Statistics (version 28), and significance was set at p < 0.05. Within the 1-year intervention, overall significant improvements were observed. Stratifying individuals by baseline IGF-1 and IGFBP-1 percentiles revealed significant differences: higher IGF-1 levels were associated with more favorable changes compared to lower levels, especially in VAT and IHL. Lower baseline IGFBP-1 levels were associated with greater improvements, especially in IHL and 2 h glucose. Higher bioactive IGF-1 levels might predict better metabolic outcomes following lifestyle interventions in prediabetes, potentially serving as biomarkers for personalized interventions.
Collapse
Affiliation(s)
- Nina M. T. Meyer
- Department of Endocrinology and Metabolism (Diabetes and Nutritional Medicine), Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (N.M.T.M.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Department of Clinical Nutrition/DZD, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany
| | - Stefan Kabisch
- Department of Endocrinology and Metabolism (Diabetes and Nutritional Medicine), Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (N.M.T.M.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Department of Clinical Nutrition/DZD, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany
| | - Ulrike Dambeck
- Department of Clinical Nutrition/DZD, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany
| | - Caroline Honsek
- Department of Clinical Nutrition/DZD, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany
| | - Margrit Kemper
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Department of Clinical Nutrition/DZD, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany
| | - Christiana Gerbracht
- Department of Clinical Nutrition/DZD, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany
| | - Ayman M. Arafat
- Department of Endocrinology and Metabolism (Diabetes and Nutritional Medicine), Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (N.M.T.M.)
| | - Andreas L. Birkenfeld
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany
- Department of Internal Medicine IV—Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany
- Department of Diabetes, School of Life Course Science and Medicine, King’s College London, London WC2R 2LS, UK
| | - Peter E. H. Schwarz
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Department for Prevention and Care of Diabetes, Clinic of Medicine III, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Paul Langerhans Institute Dresden, Helmholtz Center Munich, Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Jürgen Machann
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany
- Section on Experimental Radiology, Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Martin A. Osterhoff
- Department of Clinical Nutrition/DZD, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany
| | - Martin O. Weickert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- The ARDEN NET Centre, ENETS CoE, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Centre of Applied Biological & Exercise Sciences (ABES), Faculty of Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
- Translational & Experimental Medicine, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology and Metabolism (Diabetes and Nutritional Medicine), Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (N.M.T.M.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Department of Clinical Nutrition/DZD, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany
| |
Collapse
|
12
|
Kumar A, Vaca-Dempere M, Mortimer T, Deryagin O, Smith JG, Petrus P, Koronowski KB, Greco CM, Segalés J, Andrés E, Lukesova V, Zinna VM, Welz PS, Serrano AL, Perdiguero E, Sassone-Corsi P, Benitah SA, Muñoz-Cánoves P. Brain-muscle communication prevents muscle aging by maintaining daily physiology. Science 2024; 384:563-572. [PMID: 38696572 DOI: 10.1126/science.adj8533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 03/26/2024] [Indexed: 05/04/2024]
Abstract
A molecular clock network is crucial for daily physiology and maintaining organismal health. We examined the interactions and importance of intratissue clock networks in muscle tissue maintenance. In arrhythmic mice showing premature aging, we created a basic clock module involving a central and a peripheral (muscle) clock. Reconstituting the brain-muscle clock network is sufficient to preserve fundamental daily homeostatic functions and prevent premature muscle aging. However, achieving whole muscle physiology requires contributions from other peripheral clocks. Mechanistically, the muscle peripheral clock acts as a gatekeeper, selectively suppressing detrimental signals from the central clock while integrating important muscle homeostatic functions. Our research reveals the interplay between the central and peripheral clocks in daily muscle function and underscores the impact of eating patterns on these interactions.
Collapse
Affiliation(s)
- Arun Kumar
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Mireia Vaca-Dempere
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Thomas Mortimer
- Institute for Research in Biomedicine (IRB), Barcelona, The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Oleg Deryagin
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Jacob G Smith
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
- Center for Epigenetics and Metabolism, U1233 INSERM, Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Paul Petrus
- Center for Epigenetics and Metabolism, U1233 INSERM, Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
- Department of Medicine (H7), Karolinska Institutet, Stockholm 141 86, Sweden
| | - Kevin B Koronowski
- Department of Biochemistry & Structural Biology, Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Carolina M Greco
- Center for Epigenetics and Metabolism, U1233 INSERM, Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
- Department of Biomedical Sciences, Humanitas University and Humanitas Research Hospital IRCCS, 20089, Rozzano (Milan), Italy
| | - Jessica Segalés
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Eva Andrés
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Vera Lukesova
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Valentina M Zinna
- Institute for Research in Biomedicine (IRB), Barcelona, The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Patrick-Simon Welz
- Cancer Research Programme, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Antonio L Serrano
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
- Altos Labs Inc., San Diego Institute of Science, San Diego, CA 92121, USA
| | - Eusebio Perdiguero
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
- Altos Labs Inc., San Diego Institute of Science, San Diego, CA 92121, USA
| | - Paolo Sassone-Corsi
- Center for Epigenetics and Metabolism, U1233 INSERM, Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
- Deceased
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB), Barcelona, The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
| | - Pura Muñoz-Cánoves
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
- Altos Labs Inc., San Diego Institute of Science, San Diego, CA 92121, USA
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
| |
Collapse
|
13
|
Lee HY, Lee J, Lim H, Kim HY, Koo YS, Lim JS, Yoon Y. Lactobacillus gasseri BNR17 Ameliorates Dexamethasone-Induced Muscle Loss in BALB/c Mice and C2C12 Myotubes. J Med Food 2024; 27:385-395. [PMID: 38574296 DOI: 10.1089/jmf.2023.k.0278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024] Open
Abstract
This study aimed to investigate the effects and mechanism of Lactobacillus gasseri BNR17, a probiotic strain isolated from human breast milk, on dexamethasone-induced muscle loss in mice and cultured myotubes. BALB/c mice were intraperitoneally injected with dexamethasone, and orally administered L. gasseri BNR17 for 21 days. L. gasseri BNR17 treatment ameliorated dexamethasone-induced decline in muscle function, as evidenced by an increase in forelimb grip strength, treadmill running time, and rotarod retention time in both female and male mice. In addition, L. gasseri BNR17 treatment significantly increased the mass of the gastrocnemius and quadriceps muscles. Dual-energy X-ray absorptiometry showed a significant increase in lean body mass and a decrease in fat mass in both whole body and hind limb after treatment with L. gasseri BNR17. It was found that L. gasseri BNR17 treatment downregulated serum myostatin level and the protein degradation pathway composed of muscle-specific ubiquitin E3 ligases, MuRF1 and MAFbx, and their transcription factor FoxO3. In contrast, L. gasseri BNR17 treatment upregulated serum insulin-like growth factor-1 level and Akt-mTOR-p70S6K signaling pathway involved in protein synthesis in muscle. As a result, L. gasseri BNR17 treatment significantly increased the levels of major muscular proteins such as myosin heavy chain and myoblast determination protein 1. Consistent with in vivo results, L. gasseri BNR17 culture supernatant significantly ameliorated dexamethasone-induced C2C12 myotube atrophy in vitro. In conclusion, L. gasseri BNR17 ameliorates muscle loss by downregulating the protein degradation pathway and upregulating the protein synthesis pathway.
Collapse
Affiliation(s)
- Hyeon-Yeong Lee
- Department of Microbiology, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Jongkyu Lee
- Department of Microbiology, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Hyemi Lim
- Department of Microbiology, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Hye-Young Kim
- Department of Microbiology, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Yeon-Su Koo
- Department of Microbiology, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Ji-Su Lim
- Department of Microbiology, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Yoosik Yoon
- Department of Microbiology, College of Medicine, Chung-Ang University, Seoul, Korea
| |
Collapse
|
14
|
Reda GK, Ndunguru SF, Csernus B, Gulyás G, Knop R, Szabó C, Czeglédi L, Lendvai ÁZ. Dietary restriction and life-history trade-offs: insights into mTOR pathway regulation and reproductive investment in Japanese quail. J Exp Biol 2024; 227:jeb247064. [PMID: 38563310 DOI: 10.1242/jeb.247064] [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: 11/26/2023] [Accepted: 03/18/2024] [Indexed: 04/04/2024]
Abstract
Resources are needed for growth, reproduction and survival, and organisms must trade off limited resources among competing processes. Nutritional availability in organisms is sensed and monitored by nutrient-sensing pathways that can trigger physiological changes or alter gene expression. Previous studies have proposed that one such signalling pathway, the mechanistic target of rapamycin (mTOR), underpins a form of adaptive plasticity when individuals encounter constraints in their energy budget. Despite the fundamental importance of this process in evolutionary biology, how nutritional limitation is regulated through the expression of genes governing this pathway and its consequential effects on fitness remain understudied, particularly in birds. We used dietary restriction to simulate resource depletion and examined its effects on body mass, reproduction and gene expression in Japanese quails (Coturnix japonica). Quails were subjected to feeding at 20%, 30% and 40% restriction levels or ad libitum for 2 weeks. All restricted groups exhibited reduced body mass, whereas reductions in the number and mass of eggs were observed only under more severe restrictions. Additionally, dietary restriction led to decreased expression of mTOR and insulin-like growth factor 1 (IGF1), whereas the ribosomal protein S6 kinase 1 (RPS6K1) and autophagy-related genes (ATG9A and ATG5) were upregulated. The pattern in which mTOR responded to restriction was similar to that for body mass. Regardless of the treatment, proportionally higher reproductive investment was associated with individual variation in mTOR expression. These findings reveal the connection between dietary intake and the expression of mTOR and related genes in this pathway.
Collapse
Affiliation(s)
- Gebrehaweria K Reda
- Department of Animal Science, Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Animal Science, University of Debrecen, 4032 Debrecen, Hungary
- Department of Evolutionary Zoology and Human Biology, Faculty of Life Science, University of Debrecen, 4032 Debrecen, Hungary
| | - Sawadi F Ndunguru
- Department of Animal Science, Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Animal Science, University of Debrecen, 4032 Debrecen, Hungary
- Department of Evolutionary Zoology and Human Biology, Faculty of Life Science, University of Debrecen, 4032 Debrecen, Hungary
| | - Brigitta Csernus
- Department of Animal Science, Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, 4032 Debrecen, Hungary
- Department of Evolutionary Zoology and Human Biology, Faculty of Life Science, University of Debrecen, 4032 Debrecen, Hungary
| | - Gabriella Gulyás
- Department of Animal Science, Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, 4032 Debrecen, Hungary
| | - Renáta Knop
- Department of Animal Science, Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, 4032 Debrecen, Hungary
| | - Csaba Szabó
- Department of Animal Nutrition and Physiology, Faculty of Agriculture and Food Sciences and Environmental Management, University of Debrecen, 4032 Debrecen, Hungary
| | - Levente Czeglédi
- Department of Animal Science, Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, 4032 Debrecen, Hungary
| | - Ádám Z Lendvai
- Department of Evolutionary Zoology and Human Biology, Faculty of Life Science, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
15
|
Reda GK, Ndunguru SF, Csernus B, Knop R, Lugata JK, Szabó C, Czeglédi L, Lendvai ÁZ. Dietary restriction reveals sex-specific expression of the mTOR pathway genes in Japanese quails. Sci Rep 2024; 14:8314. [PMID: 38594358 PMCID: PMC11004124 DOI: 10.1038/s41598-024-58487-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 03/29/2024] [Indexed: 04/11/2024] Open
Abstract
Limited resources affect an organism's physiology through the conserved metabolic pathway, the mechanistic target of rapamycin (mTOR). Males and females often react differently to nutritional limitation, but whether it leads to differential mTOR pathway expression remains unknown. Recently, we found that dietary restriction (DR) induced significant changes in the expression of mTOR pathway genes in female Japanese quails (Coturnix japonica). We simultaneously exposed 32 male and female Japanese quails to either 20%, 30%, 40% restriction or ad libitum feeding for 14 days and determined the expression of six key genes of the mTOR pathway in the liver to investigate sex differences in the expression patterns. We found that DR significantly reduced body mass, albeit the effect was milder in males compared to females. We observed sex-specific liver gene expression. DR downregulated mTOR expression more in females than in males. Under moderate DR, ATG9A and RPS6K1 expressions were increased more in males than in females. Like females, body mass in males was correlated positively with mTOR and IGF1, but negatively with ATG9A and RS6K1 expressions. Our findings highlight that sexes may cope with nutritional deficits differently and emphasise the importance of considering sexual differences in studies of dietary restriction.
Collapse
Affiliation(s)
- Gebrehaweria K Reda
- Department of Animal Science, Faculty of Agricultural and Food Sciences and Environmental Management, Institute of Animal Science, Biotechnology and Nature Conservation, University of Debrecen, 4032, Debrecen, Hungary.
- Doctoral School of Animal Science, University of Debrecen, 4032, Debrecen, Hungary.
- Department of Evolutionary Zoology and Human Biology, University of Debrecen, 4032, Debrecen, Hungary.
| | - Sawadi F Ndunguru
- Department of Animal Science, Faculty of Agricultural and Food Sciences and Environmental Management, Institute of Animal Science, Biotechnology and Nature Conservation, University of Debrecen, 4032, Debrecen, Hungary
- Doctoral School of Animal Science, University of Debrecen, 4032, Debrecen, Hungary
- Department of Evolutionary Zoology and Human Biology, University of Debrecen, 4032, Debrecen, Hungary
| | - Brigitta Csernus
- Department of Evolutionary Zoology and Human Biology, University of Debrecen, 4032, Debrecen, Hungary
| | - Renáta Knop
- Department of Animal Science, Faculty of Agricultural and Food Sciences and Environmental Management, Institute of Animal Science, Biotechnology and Nature Conservation, University of Debrecen, 4032, Debrecen, Hungary
| | - James K Lugata
- Doctoral School of Animal Science, University of Debrecen, 4032, Debrecen, Hungary
- Department of Animal Nutrition and Physiology, Faculty of Agriculture and Food Sciences and Environmental Management, University of Debrecen, 4032, Debrecen, Hungary
| | - Csaba Szabó
- Department of Animal Nutrition and Physiology, Faculty of Agriculture and Food Sciences and Environmental Management, University of Debrecen, 4032, Debrecen, Hungary
| | - Levente Czeglédi
- Department of Animal Science, Faculty of Agricultural and Food Sciences and Environmental Management, Institute of Animal Science, Biotechnology and Nature Conservation, University of Debrecen, 4032, Debrecen, Hungary
| | - Ádám Z Lendvai
- Department of Evolutionary Zoology and Human Biology, University of Debrecen, 4032, Debrecen, Hungary
| |
Collapse
|
16
|
Traoré M, Noviello C, Vergnol A, Gentil C, Halliez M, Saillard L, Gelin M, Forand A, Lemaitre M, Guesmia Z, Cadot B, Caldas E, Marty B, Mougenot N, Messéant J, Strochlic L, Sadoine J, Slimani L, Jolly A, De la Grange P, Hogrel JY, Pietri-Rouxel F, Falcone S. GDF5 as a rejuvenating treatment for age-related neuromuscular failure. Brain 2024:awae107. [PMID: 38584513 DOI: 10.1093/brain/awae107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/08/2024] [Accepted: 03/24/2024] [Indexed: 04/09/2024] Open
Abstract
Sarcopenia involves a progressive loss of skeletal muscle force, quality and mass during ageing, which results in increased inability and death; however, no cure has been established thus far. Growth differentiation factor 5 (GDF5) has been described to modulate muscle mass maintenance in various contexts. For our proof of concept, we overexpressed GDF5 by AAV vector injection in Tibialis Anterior (TA) muscle of adult aged (20 months) mice and performed molecular and functional analysis of skeletal muscle. We analysed human Vastus Lateralis muscle biopsies from adult young (21-42 years) and aged (77-80 years) donors, quantifying the molecular markers modified by GDF5 overexpression (OE) in mouse muscle. We validated the major effects of GDF5 overexpression using human immortalized myotubes and Schwann Cells (SCs). We established a pre-clinical study by treating chronically (for 4 months) aged mice using recombinant GDF5 protein (rGDF5) in systemic administration and evaluated the long-term effect of this treatment on muscle mass and function. Here, we demonstrated that GDF5 OE in the old TAs promoted an increase of 16.5% of muscle weight (P = 0.0471) associated with a higher percentage of 5000-6000 µm2 large fibres (P = 0.0211), without the induction of muscle regeneration. Muscle mass gain was associated with an amelioration of 26.8% of rate of force generation (P = 0.0330) and a better neuromuscular connectivity (P = 0.0098). Moreover, GDF5 OE preserved neuromuscular junction (NMJ) morphology (38.5% of nerve terminal area increase, P < 0.0001) and stimulated the expression of re-innervation-related genes, in particular markers of SCs (fold change 3.19 for S100b gene expression, P = 0.0101). To further characterize the molecular events induced by GDF5 OE during ageing, we performed a genome-wide transcriptomic analysis of treated muscles and showed that this factor leads to a "rejuvenating" transcriptomic signature in aged mice, as 42% of the transcripts dysregulated by ageing reverted to youthful expression levels upon GDF5 OE (P < 0.05). Towards a pre-clinical approach, we performed a long-term systemic treatment using rGDF5 and showed its effectiveness in counteracting age-related muscle wasting, improving muscle function (17,8% of absolute maximal force increase, P = 0.0079), ensuring neuromuscular connectivity and preventing NMJ degeneration (7,96% of AchR area increase, P = 0.0125). In addition, in human muscle biopsies, we found the same age-related alterations than those observed in mice and improved by GDF5 and reproduced its major effects on human cells, suggesting this treatment as efficient in humans. Overall, these data provide a foundation to examine the curative potential of GDF5 drug in clinical trials for sarcopenia and, eventually, other neuromuscular diseases.
Collapse
Affiliation(s)
- Massiré Traoré
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Chiara Noviello
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Amélie Vergnol
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Christel Gentil
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Marius Halliez
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Lucile Saillard
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Maxime Gelin
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Anne Forand
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Mégane Lemaitre
- Sorbonne Université, INSERM UMS28, Phénotypage du Petit Animal, Paris 75013, France
| | - Zoheir Guesmia
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Bruno Cadot
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Eriky Caldas
- Institut de Myologie, CEA, Laboratoire d'imagerie et de spectroscopie par RMN, F-75013 Paris, France
| | - Benjamin Marty
- Institut de Myologie, CEA, Laboratoire d'imagerie et de spectroscopie par RMN, F-75013 Paris, France
| | - Nathalie Mougenot
- Sorbonne Université, INSERM UMS28, Phénotypage du Petit Animal, Paris 75013, France
| | - Julien Messéant
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Laure Strochlic
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Jeremy Sadoine
- Université de Paris, Plateforme d'Imagerie du Vivant (PIV), Montrouge, France
| | - Lofti Slimani
- Université de Paris, Plateforme d'Imagerie du Vivant (PIV), Montrouge, France
| | | | | | - Jean-Yves Hogrel
- Institut de Myologie, Laboratoire de physiologie et d'évaluation neuromusculaire, Paris, F-75013 France
| | - France Pietri-Rouxel
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Sestina Falcone
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| |
Collapse
|
17
|
Bahat G, Ozkok S. The Current Landscape of Pharmacotherapies for Sarcopenia. Drugs Aging 2024; 41:83-112. [PMID: 38315328 DOI: 10.1007/s40266-023-01093-7] [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: 12/28/2023] [Indexed: 02/07/2024]
Abstract
Sarcopenia is a skeletal muscle disorder characterized by progressive and generalized decline in muscle mass and function. Although it is mostly known as an age-related disorder, it can also occur secondary to systemic diseases such as malignancy or organ failure. It has demonstrated a significant relationship with adverse outcomes, e.g., falls, disabilities, and even mortality. Several breakthroughs have been made to find a pharmaceutical therapy for sarcopenia over the years, and some have come up with promising findings. Yet still no drug has been approved for its treatment. The key factor that makes finding an effective pharmacotherapy so challenging is the general paradigm of standalone/single diseases, traditionally adopted in medicine. Today, it is well known that sarcopenia is a complex disorder caused by multiple factors, e.g., imbalance in protein turnover, satellite cell and mitochondrial dysfunction, hormonal changes, low-grade inflammation, senescence, anorexia of aging, and behavioral factors such as low physical activity. Therefore, pharmaceuticals, either alone or combined, that exhibit multiple actions on these factors simultaneously will likely be the drug of choice to manage sarcopenia. Among various drug options explored throughout the years, testosterone still has the most cumulated evidence regarding its effects on muscle health and its safety. A mas receptor agonist, BIO101, stands out as a recent promising pharmaceutical. In addition to the conventional strategies (i.e., nutritional support and physical exercise), therapeutics with multiple targets of action or combination of multiple therapeutics with different targets/modes of action appear to promise greater benefit for the prevention and treatment of sarcopenia.
Collapse
Affiliation(s)
- Gulistan Bahat
- Division of Geriatrics, Department of Internal Medicine, Istanbul Medical School, Istanbul University, Capa, 34390, Istanbul, Turkey.
| | - Serdar Ozkok
- Division of Geriatrics, Department of Internal Medicine, Hatay Training and Research Hospital, Hatay, 31040, Turkey
| |
Collapse
|
18
|
Shili CN, Kiyimba F, Hartsen S, Ramanathan R, Pezeshki A. Recombinant Phytase Modulates Blood Amino Acids and Proteomics Profiles in Pigs Fed with Low-Protein, -Calcium, and -Phosphorous Diets. Int J Mol Sci 2023; 25:341. [PMID: 38203511 PMCID: PMC10778770 DOI: 10.3390/ijms25010341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
A beneficial effect of corn-expressed phytase (CEP) on the growth performance of pigs fed with very low-protein (VLP) diets was previously shown. Little is known whether this improvement is related to alterations in the expression profiles of blood proteins and amino acids (AAs). The objective of this study was to investigate whether supplementation of VLP, low-calcium (Ca), and low-P diets with a CEP would alter the blood AAs and protein expression profiles in pigs. Forty-eight pigs were subjected to one of the following groups (n = 8/group) for 4 weeks: positive control (PC), negative control-reduced protein (NC), NC + low-dose CEP (LD), NC + high-dose CEP (HD), LD with reduced Ca/P (LDR), and HD with reduced Ca/P (HDR). Plasma leucine and phenylalanine concentrations were reduced in NC; however, the LD diet recovered the concentration of these AAs. Serum proteomics analysis revealed that proteins involved with growth regulation, such as selenoprotein P were upregulated while the IGF-binding proteins family proteins were differentially expressed in CEP-supplemented groups. Furthermore, a positive correlation was detected between growth and abundance of proteins involved in bone mineralization and muscle structure development. Taken together, CEP improved the blood profile of some essential AAs and affected the expression of proteins involved in the regulation of growth.
Collapse
Affiliation(s)
- Cedrick N. Shili
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (C.N.S.); (F.K.); (R.R.)
| | - Frank Kiyimba
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (C.N.S.); (F.K.); (R.R.)
| | - Steve Hartsen
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Ranjith Ramanathan
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (C.N.S.); (F.K.); (R.R.)
| | - Adel Pezeshki
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (C.N.S.); (F.K.); (R.R.)
| |
Collapse
|
19
|
Gellhaus B, Böker KO, Schilling AF, Saul D. Therapeutic Consequences of Targeting the IGF-1/PI3K/AKT/FOXO3 Axis in Sarcopenia: A Narrative Review. Cells 2023; 12:2787. [PMID: 38132107 PMCID: PMC10741475 DOI: 10.3390/cells12242787] [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: 11/06/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
The high prevalence of sarcopenia in an aging population has an underestimated impact on quality of life by increasing the risk of falls and subsequent hospitalization. Unfortunately, the application of the major established key therapeutic-physical activity-is challenging in the immobile and injured sarcopenic patient. Consequently, novel therapeutic directions are needed. The transcription factor Forkhead-Box-Protein O3 (FOXO3) may be an option, as it and its targets have been observed to be more highly expressed in sarcopenic muscle. In such catabolic situations, Foxo3 induces the expression of two muscle specific ubiquitin ligases (Atrogin-1 and Murf-1) via the PI3K/AKT pathway. In this review, we particularly evaluate the potential of Foxo3-targeted gene therapy. Foxo3 knockdown has been shown to lead to increased muscle cross sectional area, through both the AKT-dependent and -independent pathways and the reduced impact on the two major downstream targets Atrogin-1 and Murf-1. Moreover, a Foxo3 reduction suppresses apoptosis, activates satellite cells, and initiates their differentiation into muscle cells. While this indicates a critical role in muscle regeneration, this mechanism might exhaust the stem cell pool, limiting its clinical applicability. As systemic Foxo3 knockdown has also been associated with risks of inflammation and cancer progression, a muscle-specific approach would be necessary. In this review, we summarize the current knowledge on Foxo3 and conceptualize a specific and targeted therapy that may circumvent the drawbacks of systemic Foxo3 knockdown. This approach presumably would limit the side effects and enable an activity-independent positive impact on skeletal muscle.
Collapse
Affiliation(s)
- Benjamin Gellhaus
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August University of Goettingen, 37075 Goettingen, Germany; (B.G.); (K.O.B.); (A.F.S.)
| | - Kai O. Böker
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August University of Goettingen, 37075 Goettingen, Germany; (B.G.); (K.O.B.); (A.F.S.)
| | - Arndt F. Schilling
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August University of Goettingen, 37075 Goettingen, Germany; (B.G.); (K.O.B.); (A.F.S.)
| | - Dominik Saul
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August University of Goettingen, 37075 Goettingen, Germany; (B.G.); (K.O.B.); (A.F.S.)
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72072 Tuebingen, Germany
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
20
|
Liu K, Qi A, Ru W, Jiang X, Cao H, Lan X, Huang Y, Lei C, Sun X, Chen H. Insertion/deletions within the bovine FoxO1 gene and their association analysis with growth traits in three Chinese cattle breeds. Anim Biotechnol 2023; 34:2051-2058. [PMID: 35491893 DOI: 10.1080/10495398.2022.2068024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
FOXO1 (FKHR) gene, as a transcription factor, plays a vital role in animal growth and development, participating in many biological processes. The aim of this study was to ascertain Insertion/deletions (Indels) polymorphism within bovine FoxO1 gene in 679 Chinese adult cows and associate them with stature traits. Two Indels (named as Indel-3 and Indel-4, recorded as rs383545622 and rs525318770 in NCBI, respectively) were successfully genotyped by the Once PCR method, which was reliable, rapid and cost effective for simultaneous detection of two or more Indels. Indel-3 and Indel-4 were located at the second intron. All four different haplotypes (H1: D3D4, H2: I3D4, H3: D3I4, H4: I3I4) could be identified, and the D (del-) allele, DD (del-/del-) genotype and D3D4 haplotype retained the highest frequency. However, individuals with DI (D3I3, D4I4 or H1H4/H2H3 genotype) showed significantly better phenotypic traits than those with the other genotypes in Nanyang cattle, showing a hybrid vigor. The results implied that this DI genotype can be applied to early selective breeding to improve the productivity of Nanyang cattle. Our results suggested that these two Indels within the bovine FoxO1 gene might be used as genetic markers for marker-assisted selection (MAS) in cattle breeding and genetics.
Collapse
Affiliation(s)
- Kunpeng Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Ao Qi
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Wenxiu Ru
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xiaojun Jiang
- Agriculture and Animal Husbandry Fine Seed Breeding Farm of Shaanxi Province, Fufeng, China
| | - Hui Cao
- Shaanxi Kingbull Livestock Co., LTD, Yangling, China
| | - Xianyong Lan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yongzhen Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xiuzhu Sun
- College of Grassland Agriculture, Northwest A&F University, Yangling, China
| | - Hong Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
21
|
Aslam MA, Ma EB, Huh JY. Pathophysiology of sarcopenia: Genetic factors and their interplay with environmental factors. Metabolism 2023; 149:155711. [PMID: 37871831 DOI: 10.1016/j.metabol.2023.155711] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/05/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023]
Abstract
Sarcopenia is a geriatric disorder characterized by a progressive decline in muscle mass and function. This disorder has been associated with a range of adverse health outcomes, including fractures, functional deterioration, and increased mortality. The pathophysiology of sarcopenia is highly complex and multifactorial, involving both genetic and environmental factors as key contributors. This review consolidates current knowledge on the genetic factors influencing the pathogenesis of sarcopenia, particularly focusing on the altered gene expression of structural and metabolic proteins, growth factors, hormones, and inflammatory cytokines. While the influence of environmental factors such as physical inactivity, chronic diseases, smoking, alcohol consumption, and sleep disturbances on sarcopenia is relatively well understood, there is a dearth of studies examining their mechanistic roles. Therefore, this review emphasizes the interplay between genetic and environmental factors, elucidating their cumulative role in exacerbating the progression of sarcopenia beyond their individual effects. The unique contribution of this review lies in synthesizing the latest evidence on the genetic factors and their interaction with environmental factors, aiming to inform the development of novel therapeutic or preventive interventions for sarcopenia.
Collapse
Affiliation(s)
- Muhammad Arif Aslam
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Eun Bi Ma
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Joo Young Huh
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea.
| |
Collapse
|
22
|
Mori Y, Ohara M, Terasaki M, Osaka N, Yashima H, Saito T, Otoyama-Kataoka Y, Omachi T, Higashimoto Y, Matsui T, Fukui T, Yamagishi SI. Subcutaneous Infusion of DNA-Aptamer Raised against Advanced Glycation End Products Prevents Loss of Skeletal Muscle Mass and Strength in Accelerated-Aging Mice. Biomedicines 2023; 11:3112. [PMID: 38137333 PMCID: PMC10740860 DOI: 10.3390/biomedicines11123112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/20/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
We have developed DNA aptamers that can inhibit the toxic effects of advanced glycation end products (AGE-Apts). We herein evaluated the effects of AGE-Apts on muscle mass and strength in senescence-accelerated mouse prone 8 (SAMP8) mice. Eight-month-old male SAMP8 mice received subcutaneous infusion of control DNA aptamers (CTR-Apts) or AGE-Apts. Mice in an age-matched senescence-accelerated mouse resistant strain 1 (SAMR1) group were treated with CTR-Apts as controls. The soleus muscles were collected after the 8-week intervention for weight measurement and histological, RT-PCR, and immunofluorescence analyses. Grip strength was measured before and after the 8-week intervention. AGE-Apt treatment inhibited the progressive decrease in the grip strength of SAMP8 mice. SAMP8 mice had lower soleus muscle weight and fiber size than SAMR1 mice, which was partly restored by AGE-Apt treatment. Furthermore, AGE-Apt-treated SAMP8 mice had a lower interstitial fibrosis area of the soleus muscle than CTR-Apt-treated SAMP8 mice. The soleus muscle levels of AGEs, oxidative stress, receptor for AGEs, and muscle ring-finger protein-1 were increased in the CTR-Apt-treated mice, all of which, except for AGEs, were inhibited by AGE-Apt treatment. Our present findings suggest that the subcutaneous delivery of AGE-Apts may be a novel therapeutic strategy for aging-related decrease in skeletal muscle mass and strength.
Collapse
Affiliation(s)
- Yusaku Mori
- Anti-Glycation Research Section, Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Shinagawa, Tokyo 142-8555, Japan
| | - Makoto Ohara
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Shinagawa, Tokyo 142-8555, Japan (M.T.); (N.O.); (Y.O.-K.); (T.O.)
| | - Michishige Terasaki
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Shinagawa, Tokyo 142-8555, Japan (M.T.); (N.O.); (Y.O.-K.); (T.O.)
| | - Naoya Osaka
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Shinagawa, Tokyo 142-8555, Japan (M.T.); (N.O.); (Y.O.-K.); (T.O.)
| | - Hironori Yashima
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Shinagawa, Tokyo 142-8555, Japan (M.T.); (N.O.); (Y.O.-K.); (T.O.)
| | - Tomomi Saito
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Shinagawa, Tokyo 142-8555, Japan (M.T.); (N.O.); (Y.O.-K.); (T.O.)
| | - Yurie Otoyama-Kataoka
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Shinagawa, Tokyo 142-8555, Japan (M.T.); (N.O.); (Y.O.-K.); (T.O.)
| | - Takemasa Omachi
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Shinagawa, Tokyo 142-8555, Japan (M.T.); (N.O.); (Y.O.-K.); (T.O.)
| | - Yuichiro Higashimoto
- Department of Chemistry, Kurume University School of Medicine, Kurume 830-0011, Fukuoka, Japan;
| | - Takanori Matsui
- Department of Bioscience and Biotechnology, Fukui Prefectural University, Eiheiji 910-1195, Fukui, Japan
| | - Tomoyasu Fukui
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Shinagawa, Tokyo 142-8555, Japan (M.T.); (N.O.); (Y.O.-K.); (T.O.)
| | - Sho-ichi Yamagishi
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Shinagawa, Tokyo 142-8555, Japan (M.T.); (N.O.); (Y.O.-K.); (T.O.)
| |
Collapse
|
23
|
Pang X, Zhang P, Chen X, Liu W. Ubiquitin-proteasome pathway in skeletal muscle atrophy. Front Physiol 2023; 14:1289537. [PMID: 38046952 PMCID: PMC10690626 DOI: 10.3389/fphys.2023.1289537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/06/2023] [Indexed: 12/05/2023] Open
Abstract
Skeletal muscles underpin myriad human activities, maintaining an intricate balance between protein synthesis and degradation crucial to muscle mass preservation. Historically, disruptions in this balance-where degradation overshadows synthesis-have marked the onset of muscle atrophy, a condition diminishing life quality and, in grave instances, imperiling life itself. While multiple protein degradation pathways exist-including the autophagy-lysosome, calcium-dependent calpain, and cysteine aspartate protease systems-the ubiquitin-proteasome pathway emerges as an especially cardinal avenue for intracellular protein degradation, wielding pronounced influence over the muscle atrophy trajectory. This paper ventures a panoramic view of predominant muscle atrophy types, accentuating the ubiquitin-proteasome pathway's role therein. Furthermore, by drawing from recent scholarly advancements, we draw associations between the ubiquitin-proteasome pathway and specific pathological conditions linked to muscle atrophy. Our exploration seeks to shed light on the ubiquitin-proteasome pathway's significance in skeletal muscle dynamics, aiming to pave the way for innovative therapeutic strategies against muscle atrophy and affiliated muscle disorders.
Collapse
Affiliation(s)
- XiangSheng Pang
- Department of Physical Education, College of Education, Zhejiang University, Hangzhou, Zhejiang, China
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| | - Peng Zhang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - XiaoPing Chen
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| | - WenMing Liu
- Department of Physical Education, College of Education, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Nevi L, Pöllänen N, Penna F, Caretti G. Targeting Epigenetic Regulators with HDAC and BET Inhibitors to Modulate Muscle Wasting. Int J Mol Sci 2023; 24:16404. [PMID: 38003594 PMCID: PMC10671811 DOI: 10.3390/ijms242216404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Epigenetic changes contribute to the profound alteration in the transcriptional program associated with the onset and progression of muscle wasting in several pathological conditions. Although HDACs and their inhibitors have been extensively studied in the field of muscular dystrophies, the potential of epigenetic inhibitors has only been marginally explored in other disorders associated with muscle atrophy, such as in cancer cachexia and sarcopenia. BET inhibitors represent a novel class of recently developed epigenetic drugs that display beneficial effects in a variety of diseases beyond malignancies. Based on the preliminary in vitro and preclinical data, HDACs and BET proteins contribute to the pathogenesis of cancer cachexia and sarcopenia, modulating processes related to skeletal muscle mass maintenance and/or metabolism. Thus, epigenetic drugs targeting HDACs and BET proteins may emerge as promising strategies to reverse the catabolic phenotype associated with cachexia and sarcopenia. Further preclinical studies are warranted to delve deeper into the molecular mechanisms associated with the functions of HDACs and BET proteins in muscle atrophy and to establish whether their epigenetic inhibitors represent a prospective therapeutic avenue to alleviate muscle wasting.
Collapse
Affiliation(s)
- Lorenzo Nevi
- Department of Biosciences, University of Milan, 20133 Milan, Italy;
| | - Noora Pöllänen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Fabio Penna
- Department of Clinical and Biological Sciences, University of Torino, 10125 Torino, Italy
| | | |
Collapse
|
25
|
Sakuma K, Hamada K, Yamaguchi A, Aoi W. Current Nutritional and Pharmacological Approaches for Attenuating Sarcopenia. Cells 2023; 12:2422. [PMID: 37830636 PMCID: PMC10572610 DOI: 10.3390/cells12192422] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/27/2023] [Accepted: 10/05/2023] [Indexed: 10/14/2023] Open
Abstract
Sarcopenia is characterized by a gradual slowing of movement due to loss of muscle mass and quality, decreased power and strength, increased risk of injury from falls, and often weakness. This review will focus on recent research trends in nutritional and pharmacological approaches to controlling sarcopenia. Because nutritional studies in humans are fairly limited, this paper includes many results from nutritional studies in mammals. The combination of resistance training with supplements containing amino acids is the gold standard for preventing sarcopenia. Amino acid (HMB) supplementation alone has no significant effect on muscle strength or muscle mass in sarcopenia, but the combination of HMB and exercise (whole body vibration stimulation) is likely to be effective. Tea catechins, soy isoflavones, and ursolic acid are interesting candidates for reducing sarcopenia, but both more detailed basic research on this treatment and clinical studies in humans are needed. Vitamin D supplementation has been shown not to improve sarcopenia in elderly individuals who are not vitamin D-deficient. Myostatin inhibitory drugs have been tried in many neuromuscular diseases, but increases in muscle mass and strength are less likely to be expected. Validation of myostatin inhibitory antibodies in patients with sarcopenia has been positive, but excessive expectations are not warranted.
Collapse
Affiliation(s)
- Kunihiro Sakuma
- Institute for Liberal Arts, Environment and Society, Tokyo Institute of Technology, Meguro-ku, Tokyo 152-8550, Japan;
| | - Kento Hamada
- Institute for Liberal Arts, Environment and Society, Tokyo Institute of Technology, Meguro-ku, Tokyo 152-8550, Japan;
| | - Akihiko Yamaguchi
- Department of Physical Therapy, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0293, Japan;
| | - Wataru Aoi
- Laboratory of Nutrition Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan;
| |
Collapse
|
26
|
Fuqua JD, Lawrence MM, Hettinger ZR, Borowik AK, Brecheen PL, Szczygiel MM, Abbott CB, Peelor FF, Confides AL, Kinter M, Bodine SC, Dupont‐Versteegden EE, Miller BF. Impaired proteostatic mechanisms other than decreased protein synthesis limit old skeletal muscle recovery after disuse atrophy. J Cachexia Sarcopenia Muscle 2023; 14:2076-2089. [PMID: 37448295 PMCID: PMC10570113 DOI: 10.1002/jcsm.13285] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/10/2023] [Accepted: 05/22/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Skeletal muscle mass and strength diminish during periods of disuse but recover upon return to weight bearing in healthy adults but are incomplete in old muscle. Efforts to improve muscle recovery in older individuals commonly aim at increasing myofibrillar protein synthesis via mammalian target of rapamycin (mTOR) stimulation despite evidence demonstrating that old muscle has chronically elevated levels of mammalian target of rapamycin complex 1 (mTORC1) activity. We hypothesized that protein synthesis is higher in old muscle than adult muscle, which contributes to a proteostatic stress that impairs recovery. METHODS We unloaded hindlimbs of adult (10-month) and old (28-month) F344BN rats for 14 days to induce atrophy, followed by reloading up to 60 days with deuterium oxide (D2 O) labelling to study muscle regrowth and proteostasis. RESULTS We found that old muscle has limited recovery of muscle mass during reloading despite having higher translational capacity and myofibrillar protein synthesis (0.029 k/day ± 0.002 vs. 0.039 k/day ± 0.002, P < 0.0001) than adult muscle. We showed that collagen protein synthesis was not different (0.005 k (1/day) ± 0.0005 vs. 0.004 k (1/day) ± 0.0005, P = 0.15) in old compared to adult, but old muscle had higher collagen concentration (4.5 μg/mg ± 1.2 vs. 9.8 μg/mg ± 0.96, P < 0.01), implying that collagen breakdown was slower in old muscle than adult muscle. This finding was supported by old muscle having more insoluble collagen (4.0 ± 1.1 vs. 9.2 ± 0.9, P < 0.01) and an accumulation of advanced glycation end products (1.0 ± 0.06 vs. 1.5 ± 0.08, P < 0.001) than adult muscle during reloading. Limited recovery of muscle mass during reloading is in part due to higher protein degradation (0.017 1/t ± 0.002 vs. 0.028 1/t ± 0.004, P < 0.05) and/or compromised proteostasis as evidenced by accumulation of ubiquitinated insoluble proteins (1.02 ± 0.06 vs. 1.22 ± 0.06, P < 0.05). Last, we showed that synthesis of individual proteins related to protein folding/refolding, protein degradation and neural-related biological processes was higher in old muscle during reloading than adult muscle. CONCLUSIONS Our data suggest that the failure of old muscle to recover after disuse is not due to limitations in the ability to synthesize myofibrillar proteins but because of other impaired proteostatic mechanisms (e.g., protein folding and degradation). These data provide novel information on individual proteins that accumulate in protein aggregates after disuse and certain biological processes such as protein folding and degradation that likely play a role in impaired recovery. Therefore, interventions to enhance regrowth of old muscle after disuse should be directed towards the identified impaired proteostatic mechanisms and not aimed at increasing protein synthesis.
Collapse
Affiliation(s)
- Jordan D. Fuqua
- Aging & Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
| | - Marcus M. Lawrence
- Aging & Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
- Department of Kinesiology and Outdoor RecreationSouthern Utah UniversityCedar CityUTUSA
| | - Zachary R. Hettinger
- Department of Physical Therapy, College of Health SciencesUniversity of KentuckyLexingtonKYUSA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKYUSA
| | - Agnieszka K. Borowik
- Aging & Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
| | - Parker L. Brecheen
- Aging & Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
| | - Marcelina M. Szczygiel
- Aging & Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
| | - Claire B. Abbott
- Aging & Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
| | - Frederick F. Peelor
- Aging & Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
| | - Amy L. Confides
- Department of Physical Therapy, College of Health SciencesUniversity of KentuckyLexingtonKYUSA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKYUSA
| | - Michael Kinter
- Aging & Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
| | - Sue C. Bodine
- Aging & Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
- Department of Internal MedicineUniversity of IowaIowa CityIAUSA
- Fraternal Order of Eagles Diabetes Research CenterUniversity of IowaIowa CityIAUSA
- Iowa City Veterans Affairs Medical CenterIowa CityIAUSA
| | - Esther E. Dupont‐Versteegden
- Department of Physical Therapy, College of Health SciencesUniversity of KentuckyLexingtonKYUSA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKYUSA
| | - Benjamin F. Miller
- Aging & Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
- Oklahoma City Veterans Affairs Medical CenterOklahoma CityOKUSA
| |
Collapse
|
27
|
Kim WS, Kim J. Exploring the impact of temporal heat stress on skeletal muscle hypertrophy in bovine myocytes. J Therm Biol 2023; 117:103684. [PMID: 37625343 DOI: 10.1016/j.jtherbio.2023.103684] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/19/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023]
Abstract
The primary aim of this investigation was to explore the impact of different temporal stress conditions on the regulators associated with skeletal muscle hypertrophy in bovine myocytes. Bovine satellite cells (BSCs) were extracted from three-month-old Holstein bull calves and subjected to myogenic differentiation under three thermal treatments: 38 °C (control; CON), 39.5 °C (moderate heat stress; MHS), and 41 °C (extreme heat stress; EHS) for a duration of 3 or 48 h. Exposure to EHS resulted in elevated (P < 0.01) expression levels of heat shock protein (HSP)20, HSP27, HSP70, and HSP90, along with increased (P < 0.01) protein levels. Moreover, cells exposed to MHS and EHS exhibited enhanced (P < 0.01) gene expression of myoblast determination protein 1 (MyoD), while myogenin (MyoG) was overexpressed (P < 0.01) in cells exposed to EHS. These findings suggest that heat exposure can potentially induce myogenic differentiation through the modulation of myogenic regulatory factors. Furthermore, our investigations revealed that exposure to EHS upregulated (P < 0.01) myosin heavy chain (MHC) I expression, whereas MHC IIA (P < 0.01) and IIX (P < 0.01) expression were increased; P < 0.01) under MHS conditions. These observations suggest that the temperature of the muscle may alter the proportion of muscle fiber types. Additionally, our data indicated that EHS activated (P < 0.01) the expression of insulin-like growth factor 1 (IGF-1) and triggered the activation of the Akt/mTOR/S6KB1 pathway, a known anabolic pathway associated with cellular protein synthesis. Consequently, these altered signaling pathways contributed to enhanced protein synthesis and increased myotube size. Overall, the results obtained from our current study revealed that extreme heat exposure (41 °C) may promote skeletal muscle hypertrophy by regulating myogenic regulatory factors and IGF-1-mediated mTOR pathway in bovine myocytes.
Collapse
Affiliation(s)
- Won Seob Kim
- Department of Animal Science, Michigan State University, East Lansing, MI, 48824, USA
| | - Jongkyoo Kim
- Animal Science and Food Science and Human Nutrition, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
28
|
Directo D, Lee SR. Cancer Cachexia: Underlying Mechanisms and Potential Therapeutic Interventions. Metabolites 2023; 13:1024. [PMID: 37755304 PMCID: PMC10538050 DOI: 10.3390/metabo13091024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/14/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023] Open
Abstract
Cancer cachexia, a multifactorial metabolic syndrome developed during malignant tumor growth, is characterized by an accelerated loss of body weight accompanied by the depletion of skeletal muscle mass. This debilitating condition is associated with muscle degradation, impaired immune function, reduced functional capacity, compromised quality of life, and diminished survival in cancer patients. Despite the lack of the known capability of fully reversing or ameliorating this condition, ongoing research is shedding light on promising preclinical approaches that target the disrupted mechanisms in the pathophysiology of cancer cachexia. This comprehensive review delves into critical aspects of cancer cachexia, including its underlying pathophysiological mechanisms, preclinical models for studying the progression of cancer cachexia, methods for clinical assessment, relevant biomarkers, and potential therapeutic strategies. These discussions collectively aim to contribute to the evolving foundation for effective, multifaceted counteractive strategies against this challenging condition.
Collapse
Affiliation(s)
| | - Sang-Rok Lee
- Department of Kinesiology, New Mexico State University, Las Cruces, NM 88003, USA;
| |
Collapse
|
29
|
Kubat GB, Bouhamida E, Ulger O, Turkel I, Pedriali G, Ramaccini D, Ekinci O, Ozerklig B, Atalay O, Patergnani S, Nur Sahin B, Morciano G, Tuncer M, Tremoli E, Pinton P. Mitochondrial dysfunction and skeletal muscle atrophy: Causes, mechanisms, and treatment strategies. Mitochondrion 2023; 72:33-58. [PMID: 37451353 DOI: 10.1016/j.mito.2023.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/02/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Skeletal muscle, which accounts for approximately 40% of total body weight, is one of the most dynamic and plastic tissues in the human body and plays a vital role in movement, posture and force production. More than just a component of the locomotor system, skeletal muscle functions as an endocrine organ capable of producing and secreting hundreds of bioactive molecules. Therefore, maintaining healthy skeletal muscles is crucial for supporting overall body health. Various pathological conditions, such as prolonged immobilization, cachexia, aging, drug-induced toxicity, and cardiovascular diseases (CVDs), can disrupt the balance between muscle protein synthesis and degradation, leading to skeletal muscle atrophy. Mitochondrial dysfunction is a major contributing mechanism to skeletal muscle atrophy, as it plays crucial roles in various biological processes, including energy production, metabolic flexibility, maintenance of redox homeostasis, and regulation of apoptosis. In this review, we critically examine recent knowledge regarding the causes of muscle atrophy (disuse, cachexia, aging, etc.) and its contribution to CVDs. Additionally, we highlight the mitochondrial signaling pathways involvement to skeletal muscle atrophy, such as the ubiquitin-proteasome system, autophagy and mitophagy, mitochondrial fission-fusion, and mitochondrial biogenesis. Furthermore, we discuss current strategies, including exercise, mitochondria-targeted antioxidants, in vivo transfection of PGC-1α, and the potential use of mitochondrial transplantation as a possible therapeutic approach.
Collapse
Affiliation(s)
- Gokhan Burcin Kubat
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey.
| | - Esmaa Bouhamida
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Oner Ulger
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey
| | - Ibrahim Turkel
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Gaia Pedriali
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Daniela Ramaccini
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Ozgur Ekinci
- Department of Pathology, Gazi University, 06500 Ankara, Turkey
| | - Berkay Ozerklig
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Ozbeyen Atalay
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Simone Patergnani
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Beyza Nur Sahin
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Giampaolo Morciano
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Meltem Tuncer
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Elena Tremoli
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Paolo Pinton
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
30
|
Zahran F, Nabil A, Nassr A, Barakat N. Amelioration of exosome and mesenchymal stem cells in rats infected with diabetic nephropathy by attenuating early markers and aquaporin-1 expression. BRAZ J BIOL 2023; 83:e271731. [PMID: 37466513 DOI: 10.1590/1519-6984.271731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/21/2023] [Indexed: 07/20/2023] Open
Abstract
Diabetic nephropathy (DN) is a prevalent diabetic microvascular condition. It is the leading cause of kidney disease in the advanced stages. There is no currently effective treatment available. This research aimed to investigate the curative potentials of exosomes isolated from mesenchymal stem cells affecting DN. This study was performed on 70 male adult albino rats. Adult rats were randomized into seven groups: Group I: Negative control group, Group II: DN group, Group III: Balanites treated group, Group IV: MSCs treated group, Group V: Exosome treated group, Group VI: Balanites + MSCs treated group and Group VII: Balanites + exosome treated group. Following the trial period, blood and renal tissues were subjected to biochemical, gene expression analyses, and histopathological examinations. Results showed that MDA was substantially increased, whereas TAC was significantly decreased in the kidney in the DN group compared to normal health rats. Undesired elevated values of MDA levels and a decrease in TAC were substantially ameliorated in groups co-administered Balanites aegyptiacae with MSCs or exosomes compared to the DN group. A substantial elevation in TNF-α and substantially diminished concentration of IGF-1 were noticed in DN rats compared to normal health rats. Compared to the DN group, the co-administration of Balanites aegyptiacae with MSCs or exosomes substantially improved the undesirable elevated values of TNF-α and IGF-1. Furthermore, in the DN group, the mRNA expression of Vanin-1, Nephrin, and collagen IV was significantly higher than in normal healthy rats. Compared with DN rats, Vanin-1, Nephrin, and collagen IV Upregulation were substantially reduced in groups co-administered Balanites aegyptiacae with MSCs or exosomes. In DN rats, AQP1 expression was significantly lower than in normal healthy rats. Furthermore, the groups co-administered Balanites aegyptiacae with MSCs or exosomes demonstrated a substantial increase in AQP1 mRNA expression compared to DN rats.
Collapse
Affiliation(s)
- F Zahran
- Zagazig University, Faculty of Science, Chemistry Department, Biochemistry Division, Zagazig, Egypt
| | - A Nabil
- Beni-Suef University, Faculty of Postgraduate Studies for Advanced Sciences - PSAS, Biotechnology and Life Sciences Department, Beni-Suef, Egypt
| | - A Nassr
- Zagazig University, Faculty of Science, Chemistry Department, Biochemistry Division, Zagazig, Egypt
| | - N Barakat
- Mansoura University, Urology and Nephrology Center, Mansoura, Egypt
| |
Collapse
|
31
|
Duan Y, Tao K, Fang Z, Lu Y. Possible-sarcopenic screening with disturbed plasma amino acid profile in the elderly. BMC Geriatr 2023; 23:427. [PMID: 37438737 DOI: 10.1186/s12877-023-04137-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 06/27/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND The mass and strength of skeletal muscle decline with age, leading to its progressive dysfunction. High-throughput metabolite profiling provides the opportunity to reveal metabolic mechanisms and the identification of biomarkers. However, the role of amino acid metabolism in possible sarcopenia remains unclear. OBJECTIVES The aim of this study included exploring variations in plasma amino acid concentrations in elderly individuals who have possible sarcopenia and further attempting to characterize a distinctive plasma amino acid profile through targeted metabolomics. METHODS A cross-sectional, correlational research design was used for this study. Thirty possible-sarcopenic elderly participants were recruited (n = 30), as determined by the Asian Working Group for Sarcopenia (AWGS). Meanwhile, a reference group of non-sarcopenic (sex-, age-, and Appendicular Skeletal muscle Mass Index (ASMI)-matched non-sarcopenic controls, n = 36) individuals was included to compare the potential differences in metabolic fingerprint of the plasma amino acids associated with sarcopenia. Both groups were conducted the body composition analysis, physical function examination, and plasma amino acid-targeted metabolomics. The amino acids in plasma were measured using ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS-MS). Also, orthogonal partial least-squares-discriminant analysis (OPLS-DA) was applied to characterize the plasma amino acid profile. RESULTS With respect to Handgrip Strength (HGS), the Five-Repetition Chair Stand Test (CS-5), the Six-Minute Walking Test (6MWT), the arm curl, the 30 s-Chair Stand Test (CST), the 2-Minute Step Test (2MST), the Timed Up-and-Go Test (TUGT), there was a decline in skeletal muscle function in the possible-sarcopenic group compared to the non-sarcopenic group. The mean plasma concentrations of arginine, asparagine, phenylalanine, serine, lysine, glutamine, and threonine were significantly lower in the possible sarcopenia group, whereas cirulline, proline, serine, and glutamic acid concentrations were higher. According to the multi-analysis, glutamine, serine, lysine, threonine, and proline were determined as the potential markers that indicated possible sarcopenia. CONCLUSIONS The findings characterize significantly altered plasma amino acid metabolisms in the elderly with possible sarcopenia, which aids to screening people who are at a high risk of developing condition, and motivating to design new preventive and therapeutic approaches.
Collapse
Affiliation(s)
- Yushuang Duan
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
- College of Rehabilitation, Weifang Medicine University, Weifang, China
| | - Kuan Tao
- School of Sports Engineering, Beijing Sport University, Beijing, China
| | - Zilong Fang
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Yifan Lu
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China.
| |
Collapse
|
32
|
Picca A, Lozanoska-Ochser B, Calvani R, Coelho-Júnior HJ, Leewenburgh C, Marzetti E. Inflammatory, mitochondrial, and senescence-related markers: Underlying biological pathways of muscle aging and new therapeutic targets. Exp Gerontol 2023; 178:112204. [PMID: 37169101 DOI: 10.1016/j.exger.2023.112204] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 05/13/2023]
Abstract
The maintenance of functional health is pivotal for achieving independent life in older age. The aged muscle is characterized by ultrastructural changes, including loss of type I and type II myofibers and a greater proportion of cytochrome c oxidase deficient and succinate dehydrogenase positive fibers. Both intrinsic (e.g., altered proteostasis, DNA damage, and mitochondrial dysfunction) and extrinsic factors (e.g., denervation, altered metabolic regulation, declines in satellite cells, and inflammation) contribute to muscle aging. Being a hub for several cellular activities, mitochondria are key to myocyte viability and mitochondrial dysfunction has been implicated in age-associated physical decline. The maintenance of functional organelles via mitochondrial quality control (MQC) processes is, therefore, crucial to skeletal myofiber viability and organismal health. The autophagy-lysosome pathway has emerged as a critical step of MQC in muscle by disposing organelles and proteins via their tagging for autophagosome incorporation and delivery to the lysosome for clearance. This pathway was found to be altered in muscle of physically inactive older adults. A relationship between this pathway and muscle tissue composition of the lower extremities as well as physical performance was also identified. Therefore, integrating muscle structure and myocyte quality control measures in the evaluation of muscle health may be a promising strategy for devising interventions fostering muscle health.
Collapse
Affiliation(s)
- Anna Picca
- Department of Medicine and Surgery, LUM University, Casamassima, 70100 Bari, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCSS, 00168 Rome, Italy
| | - Biliana Lozanoska-Ochser
- Department of Medicine and Surgery, LUM University, Casamassima, 70100 Bari, Italy; DAHFMO Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCSS, 00168 Rome, Italy; Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Hélio José Coelho-Júnior
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | - Emanuele Marzetti
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCSS, 00168 Rome, Italy; Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
33
|
MYTHO is a novel regulator of skeletal muscle autophagy and integrity. Nat Commun 2023; 14:1199. [PMID: 36864049 PMCID: PMC9981687 DOI: 10.1038/s41467-023-36817-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 02/17/2023] [Indexed: 03/04/2023] Open
Abstract
Autophagy is a critical process in the regulation of muscle mass, function and integrity. The molecular mechanisms regulating autophagy are complex and still partly understood. Here, we identify and characterize a novel FoxO-dependent gene, d230025d16rik which we named Mytho (Macroautophagy and YouTH Optimizer), as a regulator of autophagy and skeletal muscle integrity in vivo. Mytho is significantly up-regulated in various mouse models of skeletal muscle atrophy. Short term depletion of MYTHO in mice attenuates muscle atrophy caused by fasting, denervation, cancer cachexia and sepsis. While MYTHO overexpression is sufficient to trigger muscle atrophy, MYTHO knockdown results in a progressive increase in muscle mass associated with a sustained activation of the mTORC1 signaling pathway. Prolonged MYTHO knockdown is associated with severe myopathic features, including impaired autophagy, muscle weakness, myofiber degeneration, and extensive ultrastructural defects, such as accumulation of autophagic vacuoles and tubular aggregates. Inhibition of the mTORC1 signaling pathway in mice using rapamycin treatment attenuates the myopathic phenotype triggered by MYTHO knockdown. Skeletal muscles from human patients diagnosed with myotonic dystrophy type 1 (DM1) display reduced Mytho expression, activation of the mTORC1 signaling pathway and impaired autophagy, raising the possibility that low Mytho expression might contribute to the progression of the disease. We conclude that MYTHO is a key regulator of muscle autophagy and integrity.
Collapse
|
34
|
Nawaz AH, Lin S, Wang F, Zheng J, Sun J, Zhang W, Jiao Z, Zhu Z, An L, Zhang L. Investigating the heat tolerance and production performance in local chicken breed having normal and dwarf size. Animal 2023; 17:100707. [PMID: 36764018 DOI: 10.1016/j.animal.2023.100707] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
Heat stress significantly impairs the growth performance of broilers, which causes serious losses to the poultry industry every year. Thus, understanding the performance of indigenous chicken breeds under such environment is crucial to address heat stress problem. The purpose of this study was to investigate the effects of heat stress (HS) on production performance, tissue histology, heat shock response (HSP70, HSP90), and muscle growth-related genes (GHR, IGF-1, and IGF-1R) of Normal yellow chicken (NYC) and Dwarf yellow chicken (DYC). Seventy-two female birds from each strain were raised under normal environmental conditions up to 84 days, with birds from each strain being divided into two groups (HS and control). In the HS group, birds were subjected to high temperature at 35 ± 1 °C for 8 h daily and lasted for a week, while in the control group, birds were raised at 28 ± 1 °C. At 91 days old, bird's liver, hypothalamus, and breast muscle tissues were collected to evaluate the gene expression, histological changes, and the production performance. The Feed intake, weight gain ratio, total protein intake and protein efficiency ratio showed a significant reduction in the treatments (P < 0.01) and treatment × strain interaction (P < 0.05) with breast muscle rate significantly reducing among the treatments (P < 0.01) after 7 days of HS. Correspondingly, total abdominal fat showed significant change among treatment and strain (P < 0.01, P < 0.05), respectively. Besides, HS markedly upregulated the mRNA expression of HSP70 and HSP90 in the pectoralis major of both chicken strains, but no significant increase (P < 0.05) was found in mRNA expression of HSP90 in liver and hypothalamus tissues of both chicken strains. Moreover, HS significantly upregulated (P < 0.05) the expression of lipogenic genes (FASN, ACC) in liver tissues of NYC, while mRNA expression of these genes showed no variation in DYC. Similarly, HS downregulated the mRNA expression of muscle growth-related genes (GHR, IGF-1, and IGF-1R). Consequently, the histopathological analysis showed that histological changes were accompanied by inflammatory cell infiltration in liver tissues of both chicken strains; however, histopathological changes were more severe in NYC than dwarf chicken strain. Conclusively, this study depicted that the production performance and growth rate varied significantly between treatment and control group of NYC. However, heat treatment in DYC has not shown significant damaging consequences as compared to the control group that signifies the vital role of the dwarf trait in thermal tolerance.
Collapse
Affiliation(s)
- Ali Hassan Nawaz
- College of Coastal Agricultural Sciences, Guangdong Ocean University, 524088 Zhanjiang, Guangdong, PR China
| | - Shudai Lin
- College of Coastal Agricultural Sciences, Guangdong Ocean University, 524088 Zhanjiang, Guangdong, PR China
| | - Fujian Wang
- College of Coastal Agricultural Sciences, Guangdong Ocean University, 524088 Zhanjiang, Guangdong, PR China
| | - Jiahui Zheng
- College of Coastal Agricultural Sciences, Guangdong Ocean University, 524088 Zhanjiang, Guangdong, PR China
| | - Junli Sun
- Animal Husbandry Research Institute of Guangxi Zhuang Autonomous Region, 530214 Nanning, Guangxi, PR China
| | - Weilu Zhang
- College of Coastal Agricultural Sciences, Guangdong Ocean University, 524088 Zhanjiang, Guangdong, PR China
| | - Zhenhai Jiao
- College of Coastal Agricultural Sciences, Guangdong Ocean University, 524088 Zhanjiang, Guangdong, PR China
| | - Zijing Zhu
- College of Coastal Agricultural Sciences, Guangdong Ocean University, 524088 Zhanjiang, Guangdong, PR China
| | - Lilong An
- College of Coastal Agricultural Sciences, Guangdong Ocean University, 524088 Zhanjiang, Guangdong, PR China
| | - Li Zhang
- College of Coastal Agricultural Sciences, Guangdong Ocean University, 524088 Zhanjiang, Guangdong, PR China.
| |
Collapse
|
35
|
Branched-Chain Amino Acids and Di-Alanine Supplementation in Aged Mice: A Translational Study on Sarcopenia. Nutrients 2023; 15:nu15020330. [PMID: 36678201 PMCID: PMC9861351 DOI: 10.3390/nu15020330] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
In age-related sarcopenia, the gradual loss of skeletal muscle mass, function and strength is underpinned by an imbalanced rate of protein synthesis/breakdown. Hence, an adequate protein intake is considered a valuable strategy to mitigate sarcopenia. Here, we investigated the effects of a 12-week oral supplementation with branched-chain amino acids (BCAAs: leucine, isoleucine, and valine) with recognized anabolic properties, in 17-month-old (AGED) C57BL/6J male mice. BCAAs (2:1:1) were formulated in drinking water, alone or plus two L-Alanine equivalents (2ALA) or dipeptide L-Alanyl-L-Alanine (Di-ALA) to boost BCAAs bioavailability. Outcomes were evaluated on in/ex vivo readouts vs. 6-month-old (ADULT) mice. In vivo hind limb plantar flexor torque was improved in AGED mice treated with BCAAs + Di-ALA or 2ALA (recovery score, R.S., towards ADULT: ≥20%), and all mixtures significantly increased hind limb volume. Ex vivo, myofiber cross-sectional areas were higher in gastrocnemius (GC) and soleus (SOL) muscles from treated mice (R.S. ≥ 69%). Contractile indices of isolated muscles were improved by the mixtures, especially in SOL muscle (R.S. ≥ 20%). The latter displayed higher mTOR protein levels in mice supplemented with 2ALA/Di-ALA-enriched mixtures (R.S. ≥ 65%). Overall, these findings support the usefulness of BCAAs-based supplements in sarcopenia, particularly as innovative formulations potentiating BCAAs bioavailability and effects.
Collapse
|
36
|
Paez HG, Pitzer CR, Alway SE. Age-Related Dysfunction in Proteostasis and Cellular Quality Control in the Development of Sarcopenia. Cells 2023; 12:cells12020249. [PMID: 36672183 PMCID: PMC9856405 DOI: 10.3390/cells12020249] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Sarcopenia is a debilitating skeletal muscle disease that accelerates in the last decades of life and is characterized by marked deficits in muscle strength, mass, quality, and metabolic health. The multifactorial causes of sarcopenia have proven difficult to treat and involve a complex interplay between environmental factors and intrinsic age-associated changes. It is generally accepted that sarcopenia results in a progressive loss of skeletal muscle function that exceeds the loss of mass, indicating that while loss of muscle mass is important, loss of muscle quality is the primary defect with advanced age. Furthermore, preclinical models have suggested that aged skeletal muscle exhibits defects in cellular quality control such as the degradation of damaged mitochondria. Recent evidence suggests that a dysregulation of proteostasis, an important regulator of cellular quality control, is a significant contributor to the aging-associated declines in muscle quality, function, and mass. Although skeletal muscle mammalian target of rapamycin complex 1 (mTORC1) plays a critical role in cellular control, including skeletal muscle hypertrophy, paradoxically, sustained activation of mTORC1 recapitulates several characteristics of sarcopenia. Pharmaceutical inhibition of mTORC1 as well as caloric restriction significantly improves muscle quality in aged animals, however, the mechanisms controlling cellular proteostasis are not fully known. This information is important for developing effective therapeutic strategies that mitigate or prevent sarcopenia and associated disability. This review identifies recent and historical understanding of the molecular mechanisms of proteostasis driving age-associated muscle loss and suggests potential therapeutic interventions to slow or prevent sarcopenia.
Collapse
Affiliation(s)
- Hector G. Paez
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Center for Muscle, Metabolism and Neuropathology, Division of Regenerative and Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Christopher R. Pitzer
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Center for Muscle, Metabolism and Neuropathology, Division of Regenerative and Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Stephen E. Alway
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Center for Muscle, Metabolism and Neuropathology, Division of Regenerative and Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- The Tennessee Institute of Regenerative Medicine, Memphis, TN 38163, USA
- Correspondence:
| |
Collapse
|
37
|
Picca A, Triolo M, Wohlgemuth SE, Martenson MS, Mankowski RT, Anton SD, Marzetti E, Leeuwenburgh C, Hood DA. Relationship between Mitochondrial Quality Control Markers, Lower Extremity Tissue Composition, and Physical Performance in Physically Inactive Older Adults. Cells 2023; 12:183. [PMID: 36611976 PMCID: PMC9818256 DOI: 10.3390/cells12010183] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023] Open
Abstract
Altered mitochondrial quality and function in muscle may be involved in age-related physical function decline. The role played by the autophagy-lysosome system, a major component of mitochondrial quality control (MQC), is incompletely understood. This study was undertaken to obtain initial indications on the relationship between autophagy, mitophagy, and lysosomal markers in muscle and measures of physical performance and lower extremity tissue composition in young and older adults. Twenty-three participants were enrolled, nine young (mean age: 24.3 ± 4.3 years) and 14 older adults (mean age: 77.9 ± 6.3 years). Lower extremity tissue composition was quantified volumetrically by magnetic resonance imaging and a tissue composition index was calculated as the ratio between muscle and intermuscular adipose tissue volume. Physical performance in older participants was assessed via the Short Physical Performance Battery (SPPB). Protein levels of the autophagy marker p62, the mitophagy mediator BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3), the lysosomal markers transcription factor EB, vacuolar-type ATPase, and lysosomal-associated membrane protein 1 were measured by Western immunoblotting in vastus lateralis muscle biopsies. Older adults had smaller muscle volume and lower tissue composition index than young participants. The protein content of p62 and BNIP3 was higher in older adults. A negative correlation was detected between p62 and BNIP3 and the tissue composition index. p62 and BNIP3 were also related to the performance on the 5-time sit-to-stand test of the SPPB. Our results suggest that an altered expression of markers of the autophagy/mitophagy-lysosomal system is related to deterioration of lower extremity tissue composition and muscle dysfunction. Additional studies are needed to clarify the role of defective MQC in human muscle aging and identify novel biological targets for drug development.
Collapse
Affiliation(s)
- Anna Picca
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy
- Department of Medicine and Surgery, LUM University, 70100 Casamassima, Italy
| | - Matthew Triolo
- Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada
- School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada
| | | | - Matthew S. Martenson
- Department of Physiology and Aging, University of Florida, Gainesville, FL 32610, USA
| | - Robert T. Mankowski
- Department of Physiology and Aging, University of Florida, Gainesville, FL 32610, USA
| | - Stephen D. Anton
- Department of Physiology and Aging, University of Florida, Gainesville, FL 32610, USA
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | - David A. Hood
- Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada
- School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
38
|
Pharmacological inhibition of HDAC6 improves muscle phenotypes in dystrophin-deficient mice by downregulating TGF-β via Smad3 acetylation. Nat Commun 2022; 13:7108. [PMID: 36402791 PMCID: PMC9675748 DOI: 10.1038/s41467-022-34831-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 11/01/2022] [Indexed: 11/21/2022] Open
Abstract
The absence of dystrophin in Duchenne muscular dystrophy disrupts the dystrophin-associated glycoprotein complex resulting in skeletal muscle fiber fragility and atrophy, associated with fibrosis as well as microtubule and neuromuscular junction disorganization. The specific, non-conventional cytoplasmic histone deacetylase 6 (HDAC6) was recently shown to regulate acetylcholine receptor distribution and muscle atrophy. Here, we report that administration of the HDAC6 selective inhibitor tubastatin A to the Duchenne muscular dystrophy, mdx mouse model increases muscle strength, improves microtubule, neuromuscular junction, and dystrophin-associated glycoprotein complex organization, and reduces muscle atrophy and fibrosis. Interestingly, we found that the beneficial effects of HDAC6 inhibition involve the downregulation of transforming growth factor beta signaling. By increasing Smad3 acetylation in the cytoplasm, HDAC6 inhibition reduces Smad2/3 phosphorylation, nuclear translocation, and transcriptional activity. These findings provide in vivo evidence that Smad3 is a new target of HDAC6 and implicate HDAC6 as a potential therapeutic target in Duchenne muscular dystrophy.
Collapse
|
39
|
Mammalian Target of Rapamycin (mTOR) Signaling at the Crossroad of Muscle Fiber Fate in Sarcopenia. Int J Mol Sci 2022; 23:ijms232213823. [PMID: 36430301 PMCID: PMC9696247 DOI: 10.3390/ijms232213823] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/02/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) is a major regulator of skeletal myocyte viability. The signaling pathways triggered by mTOR vary according to the type of endogenous and exogenous factors (e.g., redox balance, nutrient availability, physical activity) as well as organismal age. Here, we provide an overview of mTOR signaling in skeletal muscle, with a special focus on the role played by mTOR in the development of sarcopenia. Intervention strategies targeting mTOR in sarcopenia (e.g., supplementation of plant extracts, hormones, inorganic ions, calorie restriction, and exercise) have also been discussed.
Collapse
|
40
|
Meyer NMT, Kabisch S, Dambeck U, Honsek C, Kemper M, Gerbracht C, Arafat AM, Birkenfeld AL, Schwarz PEH, Machann J, Osterhoff MA, Weickert MO, Pfeiffer AFH. Low IGF1 and high IGFBP1 predict diabetes onset in prediabetic patients. Eur J Endocrinol 2022; 187:555-565. [PMID: 36005859 DOI: 10.1530/eje-22-0034] [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] [Received: 01/26/2022] [Accepted: 08/25/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVES Some individuals develop type 2 diabetes mellitus (T2DM) despite significant metabolic improvements through lifestyle intervention. We tested the hypotheses that insulin growth factor 1 (IGF1) and its binding proteins 1 and 2 predict the onset of T2DM in prediabetes patients and determine the capacity for metabolic regeneration. DESIGN We measured fasting serum IGF1, insulin growth factor-binding protein 1 (IGFBP1) and IGFBP2 in three randomized controlled lifestyle intervention trials, covering at least 1 year of intervention period and 1 year of additional follow-up. METHODS Within a sample of 414 high-risk prediabetes patients (58% women; 28-80 years), we analyzed fasting serum concentrations of IGF1, IGFBP1 and IGFBP2 in relation to diabetes incidence and metabolic parameters over 2 years. Three hundred and forty-five subjects finished the first year of intervention. RESULTS The interventions significantly improved body weight (BMI: -3.24%, P < 0.001), liver fat (-36.8%, P < 0.001), insulin sensitivity (IS) (homeostatic model assessment-insulin resistance: -6.3%, P < 0.001) and insulin secretion (disposition index: +35%, P < 0.001) in the cohort. Fourteen percent developed T2DM within 2 years. Mean IGFBP1 levels at baseline were lower in prediabetes compared to a healthy population. Also, prediabetes patients with obesity and nonalcoholic fatty liver disease had lower IGFBP1. Those with impaired glucose tolerance had higher IGFBP1 compared to those with only impaired fasting glucose. Baseline IGF1 was lower (122.5 vs 146.6 µg/L) and IGFBP1 was higher (3.32 vs 2.09 µg/L) in subjects who developed T2DM (n = 57), resulting in a significant prediction of diabetes incidence (hazard ratio (HR) IGF1: 0.991 µg/L, P = 0.003; HR IGFBP1: 1.061 µg/L, P = 0.002). This translates into a 20% and 9% difference in T2DM incidence for IGF1 and IGFBP1, respectively. Despite reduced weight, visceral fat and hepatic fat in response to 1 year of lifestyle intervention, those who developed T2DM had not improved insulin sensitivity, glucose tolerance or IGFBP1. CONCLUSIONS Lower IGF1 and higher IGFBP1 in prediabetes predicted the incidence of T2DM, indicating an impairment of beta-cell function, which explains the unresponsiveness to lifestyle intervention.
Collapse
Affiliation(s)
- Nina M T Meyer
- Department of Endocrinology and Metabolism (Diabetes and Nutritional Medicine), Charité Universitätsmedizin Berlin, Berlin, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Research Group Clinical Nutrition/DZD, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Stefan Kabisch
- Department of Endocrinology and Metabolism (Diabetes and Nutritional Medicine), Charité Universitätsmedizin Berlin, Berlin, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Research Group Clinical Nutrition/DZD, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Ulrike Dambeck
- Research Group Clinical Nutrition/DZD, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Caroline Honsek
- Research Group Clinical Nutrition/DZD, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Margrit Kemper
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Research Group Clinical Nutrition/DZD, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Christiana Gerbracht
- Research Group Clinical Nutrition/DZD, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Ayman M Arafat
- Department of Endocrinology and Metabolism (Diabetes and Nutritional Medicine), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas L Birkenfeld
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the Eberhard Karls University of Tübingen, Tübingen, Germany
- Department of Internal Medicine IV - Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany
- Department of Diabetes, School of Life Course Science and Medicine, King's College London, London, UK
| | - Peter E H Schwarz
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Departments for Prevention and Care of Diabetes and Medicine III, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Jürgen Machann
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the Eberhard Karls University of Tübingen, Tübingen, Germany
- Section on Experimental Radiology, Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, Tübingen, Germany
| | - Martin A Osterhoff
- Research Group Clinical Nutrition/DZD, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Martin O Weickert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, The ARDEN NET Centre, ENETS CoE, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
- Centre of Applied Biological & Exercise Sciences (ABES), Faculty of Health & Life Sciences, Coventry University, Coventry, UK
- Translational & Experimental Medicine, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Andreas F H Pfeiffer
- Department of Endocrinology and Metabolism (Diabetes and Nutritional Medicine), Charité Universitätsmedizin Berlin, Berlin, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Research Group Clinical Nutrition/DZD, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| |
Collapse
|
41
|
Jaitovich A. Impaired regenerative capacity contributes to skeletal muscle dysfunction in chronic obstructive pulmonary disease. Am J Physiol Cell Physiol 2022; 323:C974-C989. [PMID: 35993519 PMCID: PMC9484993 DOI: 10.1152/ajpcell.00292.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/16/2022] [Accepted: 08/16/2022] [Indexed: 01/18/2023]
Abstract
Locomotor skeletal muscle dysfunction is a relevant comorbidity of chronic obstructive pulmonary disease (COPD) and is strongly associated with worse clinical outcomes including higher mortality. Over the last decades, a large body of literature helped characterize the process, defining the disruptive muscle phenotype caused by COPD that involves reduction in muscle mass, force-generation capacity, fatigue-tolerance, and regenerative potential following injury. A major limitation in the field has been the scarcity of well-calibrated animal models to conduct mechanistic research based on loss- and gain-of-function studies. This article provides an overall description of the process, the tools available to mechanistically investigate it, and the potential role of mitochondrially driven metabolic signals on the regulation muscle regeneration after injury in COPD. Finally, a description of future avenues to further expand on the area is proposed based on very recent evidence involving mitochondrial metabolic cues affecting myogenesis.
Collapse
Affiliation(s)
- Ariel Jaitovich
- Division of Pulmonary and Critical Care Medicine, Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| |
Collapse
|
42
|
Transcriptional regulation of autophagy in aging. CURRENT OPINION IN PHYSIOLOGY 2022. [DOI: 10.1016/j.cophys.2022.100591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
43
|
Greene MA, Udoka ANS, Powell RR, Noorai RE, Bruce T, Duckett SK. Impact of fetal exposure to mycotoxins on longissimus muscle fiber hypertrophy and miRNA profile. BMC Genomics 2022; 23:595. [PMID: 35971074 PMCID: PMC9380335 DOI: 10.1186/s12864-022-08794-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 07/25/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Longissimus muscle samples were collected from lambs exposed in utero to mycotoxins [E-, endophyte-free tall fescue seed without ergot alkaloids (negative control) or E + , endophyte-infected tall fescue seed containing ergot alkaloids] during mid-gestation (MID; E + /E-) or late-gestation (LATE; E-/E +) harvested at two developmental stages (FETAL, gestational d133) or (MAT, near maturity, 250 d of age; n = 3/treatment/developmental stage). Muscle samples were examined to determine the impact of in utero mycotoxin exposure on skeletal muscle fiber hypertrophy and the miRNA profile at FETAL and MAT. RESULTS Longissimus weight was greater (P < 0.05) in E + /E- lambs compared to E-/E + lambs at MAT; however, FETAL longissimus weight did not differ (P > 0.10) between fescue treatments. Type I fiber cross sectional area was larger (P < 0.10) for E + /E- than E-/E + at MAT but did not differ (P > 0.10) between fescue treatments at FETAL. Type II fiber area was larger (P < 0.05) at MAT in E + /E- compared to E-/E + but did not differ (P < 0.05) between fescue treatments at FETAL. Cross-sectional Type I and Type II longissimus muscle fiber area increased (P < 0.05) from FETAL to MAT by 6.86-fold and 10.83-fold, respectively. The ratio of Type II:Type I muscle fibers was lower (P = 0.04) at MAT compared to FETAL. There were 120 miRNA differentially expressed (q < 0.05) between FETAL and MAT. Maternal fescue treatment did not alter (q > 0.05) expression of miRNAs in the longissimus muscle. miR-133, -29a, -22-3p, and -410-3p were identified as highly significant with a log2 fold change > 4. In vitro satellite cell cultures showed that selected miRNAs (miR-22-3p, 29a, 27a, and 133a) are differentially regulated during proliferation and differentiation indicating a role of miRNA in muscle hypertrophy. CONCLUSIONS Exposure to mycotoxins did not alter fiber type but had long-term impacts on postnatal muscle hypertrophy and cross-sectional area. The miRNA profile of the longissimus was not altered by Maternal mycotoxin exposure at FETAL or MAT. Developmental age altered the miRNA transcriptome and mRNA expression of known genes related to muscle growth. These results indicate that Maternal exposure to E + fescue seed during LATE gestation can alter postnatal muscle hypertrophy in sheep; however, these changes are not regulated by the miRNA transcriptome of the longissimus muscle.
Collapse
Affiliation(s)
- M A Greene
- Department of Animal and Veterinary Sciences, Clemson, USA
| | - A N S Udoka
- Department of Animal and Veterinary Sciences, Clemson, USA
| | - R R Powell
- Clemson Light Imaging Facility, Clemson, USA
| | - R E Noorai
- Genomics and Bioinformatics Facility, Clemson University, Clemson, USA
| | - T Bruce
- Clemson Light Imaging Facility, Clemson, USA.,Department of Bioengineering, Clemson University, Clemson, SC, 29634, USA
| | - S K Duckett
- Department of Animal and Veterinary Sciences, Clemson, USA.
| |
Collapse
|
44
|
Hyun J, Ryu B, Oh S, Chung DM, Seo M, Park SJ, Byun K, Jeon YJ. Reversibility of sarcopenia by Ishige okamurae and its active derivative diphloroethohydroxycarmalol in female aging mice. Biomed Pharmacother 2022; 152:113210. [PMID: 35689860 DOI: 10.1016/j.biopha.2022.113210] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 11/21/2022] Open
Abstract
With the rapid increase in the elderly population worldwide, the number of people with sarcopenia has also increased significantly, and this disease is emerging as a medical and social issue. The development of pharmaceutics targeting sarcopenia is limited owing to the occurrence of side effects, and exercise therapy also has a limited scope of application. Therefore, it is necessary to develop safe and biocompatible agents to treat age-related sarcopenia. Ishige okamurae (IO), an edible brown alga, and its active substance, diphloroethohydroxycarmalol (DPHC), have been reported to have various physiological functions, including skeletal muscle regeneration ability. However, this effect has not been verified in an in vivo aging model. As an aging model, the oral IO extracts and DPHC supplemented 14-month-old female C57BL/6J mice were compared to the young group in this study; the mice model showed a substantial restoration of physical exercise ability with the imbalance of famine hormone and senescence-associated secretary phenotypes compared with those in young mice. Regarding the lean mass increase in aging mice following IO extract and DPHC administration, the muscular characteristics and molecular alterations in the gastrocnemius and soleus muscles, which are sensitive to the damage that occurs during the aging process, were significantly improved. Collectively, the current study reveals that the natural agent IO extract and its derivative DPHC can reverse sarcopenia that occurs during the process of aging by improving the imbalance of muscle regeneration in vivo.
Collapse
Affiliation(s)
- Jimin Hyun
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, the Republic of Korea
| | - Bomi Ryu
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, the Republic of Korea.
| | - Seyeon Oh
- Functional Cellular Networks Laboratory, Department of Medicine, Graduate School, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, the Republic of Korea
| | - Dong-Min Chung
- Shinwoo corperation. Ltd. Jinju 52839, the Republic of Korea
| | - Minyoung Seo
- Shinwoo corperation. Ltd. Jinju 52839, the Republic of Korea
| | - Shin Jae Park
- Shinwoo corperation. Ltd. Jinju 52839, the Republic of Korea
| | - Kyunghee Byun
- Functional Cellular Networks Laboratory, Department of Medicine, Graduate School, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, the Republic of Korea; Department of Anatomy & Cell Biology, Gachon University College of Medicine, Incheon 21936, the Republic of Korea.
| | - You-Jin Jeon
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, the Republic of Korea; Marine Science Institute, Jeju National University, Jeju 63333, the Republic of Korea.
| |
Collapse
|
45
|
Abstract
Obesity remains a serious relevant public health concern throughout the world despite related countermeasures being well understood (i.e. mainly physical activity and an adjusted diet). Among different nutritional approaches, there is a growing interest in ketogenic diets (KD) to manipulate body mass (BM) and to enhance fat mass loss. KD reduce the daily amount of carbohydrate intake drastically. This results in increased fatty acid utilisation, leading to an increase in blood ketone bodies (acetoacetate, 3-β-hydroxybutyrate and acetone) and therefore metabolic ketosis. For many years, nutritional intervention studies have focused on reducing dietary fat with little or conflicting positive results over the long term. Moreover, current nutritional guidelines for athletes propose carbohydrate-based diets to augment muscular adaptations. This review discusses the physiological basis of KD and their effects on BM reduction and body composition improvements in sedentary individuals combined with different types of exercise (resistance training or endurance training) in individuals with obesity and athletes. Ultimately, we discuss the strengths and the weaknesses of these nutritional interventions together with precautionary measures that should be observed in both individuals with obesity and athletic populations. A literature search from 1921 to April 2021 using Medline, Google Scholar, PubMed, Web of Science, Scopus and Sportdiscus Databases was used to identify relevant studies. In summary, based on the current evidence, KD are an efficient method to reduce BM and body fat in both individuals with obesity and athletes. However, these positive impacts are mainly because of the appetite suppressive effects of KD, which can decrease daily energy intake. Therefore, KD do not have any superior benefits to non-KD in BM and body fat loss in individuals with obesity and athletic populations in an isoenergetic situation. In sedentary individuals with obesity, it seems that fat-free mass (FFM) changes appear to be as great, if not greater, than decreases following a low-fat diet. In terms of lean mass, it seems that following a KD can cause FFM loss in resistance-trained individuals. In contrast, the FFM-preserving effects of KD are more efficient in endurance-trained compared with resistance-trained individuals.
Collapse
|
46
|
Anderson B, Ordaz A, Zlomislic V, Allen RT, Garfin SR, Schuepbach R, Farshad M, Schenk S, Ward SR, Shahidi B. Paraspinal Muscle Health is Related to Fibrogenic, Adipogenic, and Myogenic Gene Expression in Patients with Lumbar Spine Pathology. BMC Musculoskelet Disord 2022; 23:608. [PMID: 35739523 PMCID: PMC9229083 DOI: 10.1186/s12891-022-05572-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/14/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Lumbar spine pathology is a common feature of lower back and/or lower extremity pain and is associated with observable degenerative changes in the lumbar paraspinal muscles that are associated with poor clinical prognosis. Despite the commonly observed phenotype of muscle degeneration in this patient population, its underlying molecular mechanisms are not well understood. The aim of this study was to investigate the relationships between groups of genes within the atrophic, myogenic, fibrogenic, adipogenic, and inflammatory pathways and multifidus muscle health in individuals undergoing surgery for lumbar spine pathology. METHODS Multifidus muscle biopsies were obtained from patients (n = 59) undergoing surgery for lumbar spine pathology to analyze 42 genes from relevant adipogenic/metabolic, atrophic, fibrogenic, inflammatory, and myogenic gene pathways using quantitative polymerase chain reaction. Multifidus muscle morphology was examined preoperatively in these patients at the level and side of biopsy using T2-weighted magnetic resonance imaging to determine whole muscle compartment area, lean muscle area, fat cross-sectional areas, and proportion of fat within the muscle compartment. These measures were used to investigate the relationships between gene expression patterns and muscle size and quality. RESULTS Relationships between gene expression and imaging revealed significant associations between decreased expression of adipogenic/metabolic gene (PPARD), increased expression of fibrogenic gene (COL3A1), and lower fat fraction on MRI (r = -0.346, p = 0.018, and r = 0.386, p = 0.047 respectively). Decreased expression of myogenic gene (mTOR) was related to greater lean muscle cross-sectional area (r = 0.388, p = 0.045). CONCLUSION Fibrogenic and adipogenic/metabolic genes were related to pre-operative muscle quality, and myogenic genes were related to pre-operative muscle size. These findings provide insight into molecular pathways associated with muscle health in the presence of lumbar spine pathology, establishing a foundation for future research that addresses how these changes impact outcomes in this patient population.
Collapse
Affiliation(s)
- Brad Anderson
- Department of Orthopaedic Surgery, University of California San Diego, 350 Dickinson Street, Suite 121, Mail Code 8894, San Diego, CA, 92103-8894, USA
| | - Angel Ordaz
- Department of Orthopaedic Surgery, University of California San Diego, 350 Dickinson Street, Suite 121, Mail Code 8894, San Diego, CA, 92103-8894, USA.
| | - Vinko Zlomislic
- Department of Orthopaedic Surgery, University of California San Diego, 350 Dickinson Street, Suite 121, Mail Code 8894, San Diego, CA, 92103-8894, USA
| | - R Todd Allen
- Department of Orthopaedic Surgery, University of California San Diego, 350 Dickinson Street, Suite 121, Mail Code 8894, San Diego, CA, 92103-8894, USA
| | - Steven R Garfin
- Department of Orthopaedic Surgery, University of California San Diego, 350 Dickinson Street, Suite 121, Mail Code 8894, San Diego, CA, 92103-8894, USA
| | - Regula Schuepbach
- Department of Orthopaedics, Balgrist University Hospital, University of Zurich, Forchstrasse 340, 8008, Zurich, Switzerland
| | - Mazda Farshad
- Department of Orthopaedics, Balgrist University Hospital, University of Zurich, Forchstrasse 340, 8008, Zurich, Switzerland
| | - Simon Schenk
- Department of Orthopaedic Surgery, University of California San Diego, 350 Dickinson Street, Suite 121, Mail Code 8894, San Diego, CA, 92103-8894, USA
| | - Samuel R Ward
- Department of Orthopaedic Surgery, University of California San Diego, 350 Dickinson Street, Suite 121, Mail Code 8894, San Diego, CA, 92103-8894, USA
| | - Bahar Shahidi
- Department of Orthopaedic Surgery, University of California San Diego, 350 Dickinson Street, Suite 121, Mail Code 8894, San Diego, CA, 92103-8894, USA
| |
Collapse
|
47
|
Saneyasu T, Ogasawara K, Fujiwara Y, Honda K, Kamisoyama H. Atrogin-1 knockdown inhibits the autophagy-lysosome system in mammalian and avian myotubes. Comp Biochem Physiol A Mol Integr Physiol 2022; 271:111262. [PMID: 35750158 DOI: 10.1016/j.cbpa.2022.111262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/07/2022] [Accepted: 06/17/2022] [Indexed: 10/17/2022]
Abstract
Atrogin-1 plays an important role in ubiquitin-proteasome proteolysis in vertebrate skeletal muscles. Recently, atrogin-1 has been shown to be involved in the autophagy-lysosome system, another proteolytic system, in the murine and fish hearts and skeletal muscles. With the aim to elucidate the effect of atrogin-1 on the autophagy-lysosome system in mammalian and avian skeletal muscles, this study has examined the effects of atrogin-1 knockdown on autophagy-lysosome-related proteins in C2C12 and chicken embryonic myotubes. Using the levels of microtubule-associated protein light chain 3 (LC3)-II protein, it was confirmed that atrogin-1 knockdown blocked the autophagic flux in both the myotubes. In addition, atrogin-1 knockdown in C2C12 myotubes significantly decreased the level of autophagy-related gene (ATG)12-ATG5 conjugate, which is supposedly necessary for the fusion of autophagosomes and lysosomes. Atrogin-1 knockdown also resulted in downregulation of forkhead box O3, a transcription factor for ATG12. These data suggest that atrogin-1 is essential for the normal autophagy-lysosome system in the striated muscles of vertebrates.
Collapse
Affiliation(s)
- Takaoki Saneyasu
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan.
| | - Kazuki Ogasawara
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Yuki Fujiwara
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Kazuhisa Honda
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Hiroshi Kamisoyama
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| |
Collapse
|
48
|
Guerrero-Navarro L, Jansen-Dürr P, Cavinato M. Age-Related Lysosomal Dysfunctions. Cells 2022; 11:cells11121977. [PMID: 35741106 PMCID: PMC9221958 DOI: 10.3390/cells11121977] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/10/2022] [Accepted: 06/16/2022] [Indexed: 12/10/2022] Open
Abstract
Organismal aging is normally accompanied by an increase in the number of senescent cells, growth-arrested metabolic active cells that affect normal tissue function. These cells present a series of characteristics that have been studied over the last few decades. The damage in cellular organelles disbalances the cellular homeostatic processes, altering the behavior of these cells. Lysosomal dysfunction is emerging as an important factor that could regulate the production of inflammatory molecules, metabolic cellular state, or mitochondrial function.
Collapse
Affiliation(s)
- Lena Guerrero-Navarro
- Institute for Biomedical Aging Research, Universität Innsbruck, 6020 Innsbruck, Austria; (L.G.-N.); (P.J.-D.)
- Center for Molecular Biosciences Innsbruck, Innrain 58, 6020 Innsbruck, Austria
| | - Pidder Jansen-Dürr
- Institute for Biomedical Aging Research, Universität Innsbruck, 6020 Innsbruck, Austria; (L.G.-N.); (P.J.-D.)
- Center for Molecular Biosciences Innsbruck, Innrain 58, 6020 Innsbruck, Austria
| | - Maria Cavinato
- Institute for Biomedical Aging Research, Universität Innsbruck, 6020 Innsbruck, Austria; (L.G.-N.); (P.J.-D.)
- Center for Molecular Biosciences Innsbruck, Innrain 58, 6020 Innsbruck, Austria
- Correspondence:
| |
Collapse
|
49
|
Iturin A Rescued STb-R-Induced Pork Skeletal Muscle Growth Restriction through the Hypothalamic-Pituitary-mTORC1 Growth Axis. Animals (Basel) 2022; 12:ani12121568. [PMID: 35739903 PMCID: PMC9219473 DOI: 10.3390/ani12121568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/29/2022] [Accepted: 06/16/2022] [Indexed: 11/17/2022] Open
Abstract
The engineered STb-Rosetta Escherichia coli (STb-R) was designed to investigate the effects of Iturin A on the skeletal muscle growth of weaned piglets. A total of 28 piglets were randomly divided into 4 groups (7 piglets per group): the control group (100 mL PBS), the Iturin A group (100 mL 320 mg/kg body weight (BW) Iturin A), the STb-R group (100 mL 1 × 1010 CFU/mL STb-R), and the Iturin A + STb-R group (100 mL 320 mg/kg BW Iturin A + 1 × 1010 CFU/mL STb-R). Compared with the control, STb-R-reduced body weight gain were rescued by Iturin A. The semimembranosus muscle weight recovered to normal level in the Iturin A + STb-R group. The level of relevant genes of the growth axis were elevated by Iturin A, including GHRH in the hypothalamus, GHRHR and GH in the pituitary, and GHR, IGF-1 and IGF-1R in the semimembranosus muscle. Moreover, Iturin A increased the mean fiber area and the number of proliferating cells in the semimembranosus muscle, which were decreased by STb-R. Additionally, the mTORC1 pathway was reactivated by Iturin A to relieve the suppression of STb-R. Collectively, the hypothalamic-pituitary growth axis-mediated Iturin A reactivated the mTORC1 pathway to rescue STb-R-restricted pork skeletal muscle growth.
Collapse
|
50
|
McIntyre RL, Liu YJ, Hu M, Morris BJ, Willcox BJ, Donlon TA, Houtkooper RH, Janssens GE. Pharmaceutical and nutraceutical activation of FOXO3 for healthy longevity. Ageing Res Rev 2022; 78:101621. [PMID: 35421606 DOI: 10.1016/j.arr.2022.101621] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/10/2022] [Accepted: 04/07/2022] [Indexed: 12/12/2022]
Abstract
Life expectancy has increased substantially over the last 150 years. Yet this means that now most people also spend a greater length of time suffering from various age-associated diseases. As such, delaying age-related functional decline and extending healthspan, the period of active older years free from disease and disability, is an overarching objective of current aging research. Geroprotectors, compounds that target pathways that causally influence aging, are increasingly recognized as a means to extend healthspan in the aging population. Meanwhile, FOXO3 has emerged as a geroprotective gene intricately involved in aging and healthspan. FOXO3 genetic variants are linked to human longevity, reduced disease risks, and even self-reported health. Therefore, identification of FOXO3-activating compounds represents one of the most direct candidate approaches to extending healthspan in aging humans. In this work, we review compounds that activate FOXO3, or influence healthspan or lifespan in a FOXO3-dependent manner. These compounds can be classified as pharmaceuticals, including PI3K/AKT inhibitors and AMPK activators, antidepressants and antipsychotics, muscle relaxants, and HDAC inhibitors, or as nutraceuticals, including primary metabolites involved in cell growth and sustenance, and secondary metabolites including extracts, polyphenols, terpenoids, and other purified natural compounds. The compounds documented here provide a basis and resource for further research and development, with the ultimate goal of promoting healthy longevity in humans.
Collapse
Affiliation(s)
- Rebecca L McIntyre
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Yasmine J Liu
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Man Hu
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Brian J Morris
- School of Medical Sciences, University of Sydney, Sydney, NSW, Australia; Department of Research, Kuakini Medical Center, Honolulu, HI, USA; Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Bradley J Willcox
- Department of Research, Kuakini Medical Center, Honolulu, HI, USA; Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Timothy A Donlon
- Department of Research, Kuakini Medical Center, Honolulu, HI, USA; Department of Cell and Molecular Biology and Department of Pathology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Georges E Janssens
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|