1
|
Hashemolhosseini S, Gessler L. Crosstalk among canonical Wnt and Hippo pathway members in skeletal muscle and at the neuromuscular junction. Neural Regen Res 2025; 20:2464-2479. [PMID: 39248171 DOI: 10.4103/nrr.nrr-d-24-00417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/05/2024] [Indexed: 09/10/2024] Open
Abstract
Skeletal muscles are essential for locomotion, posture, and metabolic regulation. To understand physiological processes, exercise adaptation, and muscle-related disorders, it is critical to understand the molecular pathways that underlie skeletal muscle function. The process of muscle contraction, orchestrated by a complex interplay of molecular events, is at the core of skeletal muscle function. Muscle contraction is initiated by an action potential and neuromuscular transmission requiring a neuromuscular junction. Within muscle fibers, calcium ions play a critical role in mediating the interaction between actin and myosin filaments that generate force. Regulation of calcium release from the sarcoplasmic reticulum plays a key role in excitation-contraction coupling. The development and growth of skeletal muscle are regulated by a network of molecular pathways collectively known as myogenesis. Myogenic regulators coordinate the differentiation of myoblasts into mature muscle fibers. Signaling pathways regulate muscle protein synthesis and hypertrophy in response to mechanical stimuli and nutrient availability. Several muscle-related diseases, including congenital myasthenic disorders, sarcopenia, muscular dystrophies, and metabolic myopathies, are underpinned by dysregulated molecular pathways in skeletal muscle. Therapeutic interventions aimed at preserving muscle mass and function, enhancing regeneration, and improving metabolic health hold promise by targeting specific molecular pathways. Other molecular signaling pathways in skeletal muscle include the canonical Wnt signaling pathway, a critical regulator of myogenesis, muscle regeneration, and metabolic function, and the Hippo signaling pathway. In recent years, more details have been uncovered about the role of these two pathways during myogenesis and in developing and adult skeletal muscle fibers, and at the neuromuscular junction. In fact, research in the last few years now suggests that these two signaling pathways are interconnected and that they jointly control physiological and pathophysiological processes in muscle fibers. In this review, we will summarize and discuss the data on these two pathways, focusing on their concerted action next to their contribution to skeletal muscle biology. However, an in-depth discussion of the non-canonical Wnt pathway, the fibro/adipogenic precursors, or the mechanosensory aspects of these pathways is not the focus of this review.
Collapse
Affiliation(s)
- Said Hashemolhosseini
- Institute of Biochemistry, Medical Faculty, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | | |
Collapse
|
2
|
Xu SH, Xu H, Xiao KW, Mao SJ. Exercise rehabilitation on patients with non-small cell lung cancer: A meta-analysis of randomized controlled trials. World J Clin Cases 2025; 13:100161. [DOI: 10.12998/wjcc.v13.i11.100161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/19/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024] Open
Abstract
BACKGROUND Lung cancer is one of the most common and deadly cancers worldwide. As the disease progresses and due to the side effects of treatment, patients’ physical activity significantly decreases.
AIM To systematically review and conduct a meta-analysis on the effects of exercise rehabilitation on the physical activity of lung cancer patients and determine the best implementation methods to provide clinical guidance.
METHODS Literature was searched through multiple electronic databases. A random effects model was used to combine effect sizes through standardized mean difference (SMD). The Cochrane risk of bias tool was used to assess the quality of the literature, sensitivity analysis was used to ensure the robustness of the results, and Egger’s test was used to detect publication bias and asymmetry.
RESULTS A total of 11 studies involving 541 patients were included in this study. The physical endurance, muscle function and cardiopulmonary function of non-small cell lung cancer (NSCLC) patients were evaluated. The overall effect size of the six-minute walk test (6MWT) was not statistically significant. However, subgroup analysis found that endurance significantly improved when exercise duration exceeded 0.5 hours (P ≤ 0.05). In terms of muscle function, the overall effect size was SMD = 0.619. Subgroup analysis showed that strength training, respiratory training, and cross-training (XT) significantly improved muscle function. Exercise rehabilitation significantly enhanced cardiopulmonary endurance (SMD = 0.856, P = 0.002), and the effect was better when the single exercise duration was more than 1 hour, age was over 65 years, and the intervention period was more than 3 months.
CONCLUSION Exercise rehabilitation effectively improved muscle function in NSCLC patients, especially strength training, respiratory training, and cross-training. Cardiopulmonary function also showed improvement, particularly when exercise duration exceeded 1 hour, age was ≥ 65 years, and the intervention period was more than 3 months. A single exercise duration of more than 0.5 hours can enhance patients’ physical endurance. Appropriately increasing exercise duration and selecting suitable exercise forms can effectively improve the physical activity of NSCLC patients.
Collapse
Affiliation(s)
- Sheng-Hui Xu
- Department of Sports, Gansu Vocational College of Communications, Lanzhou 730070, Gansu Province, China
| | - Hong Xu
- Department of Sports, Gansu Vocational College of Communications, Lanzhou 730070, Gansu Province, China
| | - Kai-Wen Xiao
- Department of Sports, Gansu Vocational College of Communications, Lanzhou 730070, Gansu Province, China
| | - Su-Jie Mao
- Department of Sports, Harbin Sports University, Harbin 150008, Heilongjiang Province, China
| |
Collapse
|
3
|
Spinelli S, Humma Z, Magnone M, Zocchi E, Sturla L. Role of Abscisic Acid in the Whole-Body Regulation of Glucose Uptake and Metabolism. Nutrients 2024; 17:13. [PMID: 39796447 DOI: 10.3390/nu17010013] [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: 11/16/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Abscisic acid (ABA) is a hormone with a long evolutionary history, dating back to the earliest living organisms, of which modern (ABA-producing) cyanobacteria are likely descendants, which existed long before the separation of the plant and animal kingdoms, with a conserved role as signals regulating cell responses to environmental challenges. In mammals, along with the anti-inflammatory and neuroprotective function of ABA, nanomolar ABA regulates the metabolic response to glucose availability by stimulating glucose uptake in skeletal muscle and adipose tissue via an insulin-independent mechanism and increasing metabolic energy production and also dissipation in brown and white adipocytes. Chronic ABA intake of micrograms per Kg body weight improves blood glucose, lipids, and morphometric parameters (waist circumference and body mass index) in borderline subjects for prediabetes and metabolic syndrome. This review summarizes the most recent in vitro and in vivo data obtained with nanomolar ABA, the involvement of the receptors LANCL1 and LANCL2 in the hormone's action, and the importance of mammals' endowment with two distinct hormones governing the metabolic response to glucose availability. Finally, unresolved issues and future directions for the clinical use of ABA in diabetes are discussed.
Collapse
Affiliation(s)
- Sonia Spinelli
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy
| | - Zelle Humma
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Mirko Magnone
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Elena Zocchi
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Laura Sturla
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| |
Collapse
|
4
|
Chuang YF, Cheng L, Chang WH, Yu SY, Hsu HT, An LM, Yen CH, Chang FR, Lo YC. Spatheliachromen mitigates methylglyoxal-induced myotube atrophy by activating Nrf2, inhibiting ubiquitin-mediated protein degradation, and restoring mitochondrial function. Eur J Pharmacol 2024; 984:177070. [PMID: 39442745 DOI: 10.1016/j.ejphar.2024.177070] [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: 08/06/2024] [Revised: 10/11/2024] [Accepted: 10/20/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Methylglyoxal (MGO) is a potent precursor of glycative stress that leads to oxidative stress and muscle atrophy in diabetes. Spatheliachromen (FPATM-20), derived from Ficus pumila var. awkeotsang, exhibited potential antioxidant activity. PURPOSE This study aimed to evaluate the potential impact and underlying mechanisms of FPATM-20 on MGO-induced myotube atrophy and mitochondrial dysfunction in mouse skeletal C2C12 myotubes. METHODS Atrophic and antioxidant factors were evaluated using immunofluorescence, enzyme-linked immunosorbent assay, and western blotting. Mitochondrial function was assessed using the ATP assay and Seahorse Cell Mito Stress Test. The glycogen content was determined using periodic acid-Schiff staining. Molecular docking was performed to determine the interaction between FPATM-20 and Keap1. RESULTS In myotubes treated with MGO, FPATM-20 activated the Nrf2 pathway, reduced ROS levels, enhanced antioxidant defense, and increased glycogen content. FPATM-20 improved myotube viability and size, upregulated myosin heavy chain (MyHC) expression, modulated ubiquitin-proteasome molecules (nuclear FoxO3a, atrogin-1, MuRF-1, and p62/SQSTM1), and inhibited apoptosis (Bax/Bcl-2 ratio and cleaved caspase 3). Moreover, FPATM-20 restored mitochondrial function, including mitochondrial membrane potential, mitochondrial oxygen consumption rate, and mitochondrial biogenesis pathway (nuclear PGC-1α/TFAM/FNDC5). The inhibition of Nrf2 with ML385 reversed the effects of FPATM-20 on MGO. Furthermore, molecular docking confirmed the binding of FPATM-20 to Keap1, a suppressor of Nrf2, showing the crucial role of Nrf2 in protective effects. CONCLUSIONS FPATM-20 protects myotubes from MGO toxicity by activating the Nrf2 antioxidant defense, reducing protein degradation and apoptosis, and enhancing mitochondrial function. Thus, FPATM-20 may be a novel agent for preventing skeletal muscle atrophy.
Collapse
Affiliation(s)
- Yu-Fan Chuang
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Lin Cheng
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wan-Hsuan Chang
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Szu-Yin Yu
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; Institute of Pharmacognosy, University of Szeged, Szeged, Hungary
| | - Hung-Te Hsu
- Department of Anesthesia, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung, Taiwan; Faculty of Anesthesiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Li-Mei An
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Hung Yen
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Ching Lo
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
5
|
Ali SR, Nkembo AT, Tipparaju SM, Ashraf M, Xuan W. Sarcopenia: recent advances for detection, progression, and metabolic alterations along with therapeutic targets. Can J Physiol Pharmacol 2024; 102:697-708. [PMID: 39186818 PMCID: PMC11663012 DOI: 10.1139/cjpp-2024-0201] [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] [Indexed: 08/28/2024]
Abstract
Sarcopenia, a disorder marked by muscle loss and dysfunction, is a global health concern, particularly in aging populations. Sarcopenia is intricately related to various health conditions, including obesity, dysphagia, and frailty, which underscores the complexity. Despite recent advances in metabolomics and other omics data for early detection and treatment, the precise characterization and diagnosis of sarcopenia remains challenging. In the present review we provide an overview of the complex metabolic mechanisms that underlie sarcopenia, with particular emphasis on protein, lipid, carbohydrate, and bone metabolism. The review highlights the importance of leucine and other amino acids in promoting muscle protein synthesis and clarifies the critical role played by amino acid metabolism in preserving muscular health. In addition, the review provides insights regarding lipid metabolism on sarcopenia, with an emphasis on the effects of inflammation and insulin resistance. The development of sarcopenia is largely influenced by insulin resistance, especially with regard to glucose metabolism. Overall, the review emphasizes the complex relationship between bone and muscle health by highlighting the interaction between sarcopenia and bone metabolism. Furthermore, the review outlines various therapeutic approaches and potential biomarkers for diagnosing sarcopenia. These include pharmacological strategies such as hormone replacement therapy and anabolic steroids as well as lifestyle modifications such as exercise, nutrition, and dietary changes.
Collapse
Affiliation(s)
- Syeda Roohina Ali
- Department of Pharmaceutical Sciences, USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, US
| | - Augustine T Nkembo
- Department of Pharmaceutical Sciences, USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, US
| | - Srinivas M Tipparaju
- Department of Pharmaceutical Sciences, USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, US
| | - Muhammad Ashraf
- Department of Pharmaceutical Sciences, USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, US
| | - Wanling Xuan
- Department of Pharmaceutical Sciences, USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, US
| |
Collapse
|
6
|
Lewandowski CG, Garofolo A, Leite HP. Sarcopenia in children and adolescents submitted to hematopoietic stem cell transplantation. Hematol Transfus Cell Ther 2024; 46 Suppl 6:S86-S92. [PMID: 38485546 DOI: 10.1016/j.htct.2024.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/04/2023] [Accepted: 01/18/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Sarcopenia, characterized by decreased muscle mass, strength, and function, is associated with adverse outcomes. The prevalence of sarcopenia and the effect of the inflammatory response on muscle strength loss in children undergoing hematopoietic stem cell transplantation (HSCT) are unknown. This study aimed to estimate the prevalence of sarcopenia on admission and its associated clinical factors in children and adolescents undergoing HSCT and to determine the extent to which the systemic inflammatory response during hospitalization affects muscle strength. METHOD This was a prospective study of patients aged 6-18 years old undergoing HSCT. The outcome variables were the prevalence of sarcopenia on admission (loss of strength and skeletal muscle mass) and the loss of handgrip strength during hospitalization. Potential explanatory variables included age, sarcopenia, nutritional status and systemic inflammation as measured by serum C-reactive protein, albumin and ferritin concentrations. RESULTS Eighty patients with a mean age of 11.5 (± 3.5) years were included. The main diagnoses were acute lymphocytic leukemia, acute myeloid leukemia and aplastic anemia. Eleven percent of patients had sarcopenia upon admission. Female sex, sarcopenia on admission (β coefficient: 0.70; 95% CI: -1.30 to -0.12; p-value = 0.018) and serum C-reactive protein concentration (β coefficient: -0.10; 95% CI: -0.16 to -0.04; p-value = 0.001) were associated with handgrip strength loss from admission to 14 days after HSCT. CONCLUSION Sarcopenia on admission and the degree of systemic inflammatory response are associated with decreased handgrip strength during hospitalization in pediatric patients undergoing HSCT. Further studies using the same criteria for diagnosing sarcopenia are required to enhance our understanding of these relationships.
Collapse
Affiliation(s)
- Cláudia G Lewandowski
- Discipline of Pediatric Nutrition, Department of Pediatrics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Adriana Garofolo
- Institute for Care, Rehabilitation and Assistance in Neuropelveology and Gynecology (INCREASING), São Paulo, Brazil
| | - Heitor P Leite
- Discipline of Pediatric Nutrition, Department of Pediatrics, Universidade Federal de São Paulo, São Paulo, Brazil.
| |
Collapse
|
7
|
Mahdabi M, Mehrgan MS, Rajabi Islami H. Deciphering the impact of stickwater hydrolysate on growth performance, immune response, and IGF-1/PI3K/AKT/mTOR signaling pathway in Siberian sturgeon (Acipenser baerii) fingerlings. FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:2605-2618. [PMID: 39373813 DOI: 10.1007/s10695-024-01407-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/24/2024] [Indexed: 10/08/2024]
Abstract
A feeding trial lasting 56 days was carried out to assess how the inclusion of stickwater hydrolysate (SWH) in the diet of Siberian sturgeon (Acipenser baerii) fingerlings affected their growth performance, immunity, digestive enzyme activity, and gene expression linked to the IGF-1/PI3K/AKT/mTOR signaling pathway. Siberian sturgeon fingerlings were acclimatized and fed isonitrogenous, isoenergetic diets with varying SWH concentrations (0%, 0.5%, 1.5%, and 2.5%). Growth parameters, serum proteins, immunological and digestive enzyme activities, and gene expression levels were assessed post-trial. Results demonstrated that 0.5%, and 1.5% SWH treatments significantly improved weight gain, specific growth rate, feed conversion ratio, and protein efficiency ratio. Notably, these diets also elevated serum protein and plasma globulin levels, reduced albumin-to-globulin ratios, and enhanced lysozyme, myeloperoxidase (MPO) activities, and immunoglobulin (Ig) M levels, indicating an immunostimulatory effect. Digestive enzyme activities were markedly increased in the SWH groups, particularly at 1.5%. Gene expression analyses revealed upregulation of mtorc1, s6K, akt, pi3k, and igf1, with concurrent downregulation of 4e-bp1 in the muscle of fish, signifying activation of the IGF-1/PI3K/AKT/mTOR pathway, which is central to protein synthesis and muscle growth. In conclusion, SWH at appropriate levels significantly enhances growth, digestive efficiency, and immune function in Siberian sturgeon fingerlings, while also activating key metabolic pathways.
Collapse
Affiliation(s)
- Mahdad Mahdabi
- Department of Fisheries, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mehdi Shamsaie Mehrgan
- Department of Fisheries, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Houman Rajabi Islami
- Department of Fisheries, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
8
|
DiLeo MR, Hall RE, Vellers HL, Daniels CL, Levitt DE. Alcohol Alters Skeletal Muscle Bioenergetic Function: A Scoping Review. Int J Mol Sci 2024; 25:12280. [PMID: 39596345 PMCID: PMC11594450 DOI: 10.3390/ijms252212280] [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: 09/30/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Bioenergetic pathways uniquely support sarcomere function which, in turn, helps to maintain functional skeletal muscle (SKM) mass. Emerging evidence supports alcohol (EtOH)-induced bioenergetic impairments in SKM and muscle precursor cells. We performed a scoping review to synthesize existing evidence regarding the effects of EtOH on SKM bioenergetics. Eligible articles from six databases were identified, and titles, abstracts, and full texts for potentially relevant articles were screened against inclusion criteria. Through the search, we identified 555 unique articles, and 21 met inclusion criteria. Three studies investigated EtOH effects on the adenosine triphosphate (ATP)-phosphocreatine (PCr) system, twelve investigated EtOH effects on glycolytic metabolism, and seventeen investigated EtOH effects on mitochondrial metabolism. Despite increased ATP-PCr system reliance, EtOH led to an overall decrease in bioenergetic function through decreased expression and activity of glycolytic and mitochondrial pathway components. However, effects varied depending on the EtOH dose and duration, model system, and sample type. The results detail the EtOH-induced shifts in energy metabolism, which may adversely affect sarcomere function and contribute to myopathy. These findings should be used to develop targeted interventions that improve SKM bioenergetic function, and thus sarcomere function, in people with Alcohol Use Disorder (AUD). Key areas in need of further investigation are also identified.
Collapse
Affiliation(s)
- Matthew R. DiLeo
- Metabolic Health and Muscle Physiology Laboratory, Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX 79409, USA; (M.R.D.); (R.E.H.); (C.L.D.)
| | - Rylea E. Hall
- Metabolic Health and Muscle Physiology Laboratory, Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX 79409, USA; (M.R.D.); (R.E.H.); (C.L.D.)
| | - Heather L. Vellers
- Mitochondrial Biology and Endurance Trainability Laboratory, Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX 79409, USA;
| | - Chelsea L. Daniels
- Metabolic Health and Muscle Physiology Laboratory, Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX 79409, USA; (M.R.D.); (R.E.H.); (C.L.D.)
| | - Danielle E. Levitt
- Metabolic Health and Muscle Physiology Laboratory, Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX 79409, USA; (M.R.D.); (R.E.H.); (C.L.D.)
| |
Collapse
|
9
|
Mayakrishnan V, Kannappan P, Balakarthikeyan J, Kim CY. Rodent model intervention for prevention and optimal management of sarcopenia: A systematic review on the beneficial effects of nutrients & non-nutrients and exercise to improve skeletal muscle health. Ageing Res Rev 2024; 102:102543. [PMID: 39427886 DOI: 10.1016/j.arr.2024.102543] [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: 12/28/2023] [Revised: 09/25/2024] [Accepted: 10/07/2024] [Indexed: 10/22/2024]
Abstract
Sarcopenia is a common musculoskeletal disorder characterized by degenerative processes and is strongly linked to an increased susceptibility to falls, fractures, physical limitations, and mortality. Several models have been used to explore therapeutic and preventative measures as well as to gain insight into the molecular mechanisms behind sarcopenia. With novel experimental methodologies emerging to design foods or novel versions of conventional foods, understanding the impact of nutrition on the prevention and management of sarcopenia has become important. This review provides a thorough assessment of the use of rodent models of sarcopenia for understanding the aging process, focusing the effects of nutrients, plant extracts, exercise, and combined interventions on skeletal muscle health. According to empirical research, nutraceuticals and functional foods have demonstrated potential benefits in enhancing physical performance. In preclinical investigations, the administration of herbal extracts and naturally occurring bioactive compounds yielded advantageous outcomes such as augmented muscle mass and strength generation. Furthermore, herbal treatments exhibited inhibitory effects on muscle atrophy and sarcopenia. A substantial body of information establishes a connection between diet and the muscle mass, strength, and functionality of older individuals. This suggests that nutrition has a major impact in both the prevention and treatment of sarcopenia.
Collapse
Affiliation(s)
- Vijayakumar Mayakrishnan
- Research Institute of Human Ecology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Priya Kannappan
- PSG College of Arts & Science, Civil Aerodrome, Coimbatore, Tamil Nadu 641014, India
| | | | - Choon Young Kim
- Research Institute of Human Ecology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea; Department of Food and Nutrition, Yeungnam University Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| |
Collapse
|
10
|
Dean E, Xu J, Jones AYM, Vongsirinavarat M, Lomi C, Kumar P, Ngeh E, Storz MA. An unbiased, sustainable, evidence-informed Universal Food Guide: a timely template for national food guides. Nutr J 2024; 23:126. [PMID: 39425106 PMCID: PMC11487974 DOI: 10.1186/s12937-024-01018-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 09/17/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Although national food guides are designed, ostensibly, to translate scientific evidence with respect to food, dietary patterns, and health, their development has increasingly become a corporate/political process as well as scientific one; often with corporate/political influences overriding science. Our aim was to construct an unbiased, sustainable, evidence-informed Universal Food Guide to serve as a template for countries to develop their unique guides, thereby, provide a valid resource for health professionals, health authorities, and the public. METHODS To address our aim, we conducted an integrative review of multiple evidence-informed sources (e.g., established databases, evidence syntheses, scholarly treatises, and policy documents) related to four areas: 1. Food guides' utility and conflicts of interest; 2. The evidence-based healthiest diet; 3. Constituents of the Universal Food Guide template; and 4. Implications for population health; regulation/governance; environment/climate/planetary health; and ethics. RESULTS The eating pattern that is healthiest for humans (i.e., most natural, and associated with maximal health across the life cycle; reduced non-communicable disease (NCD) risk; and minimal end-of-life illness) is whole food, low fat, plant-based, especially vegan, with the absence of ultra-processed food. Disparities in national food guide recommendations can be explained by factors other than science, specifically, corporate/political interests reflected in heavily government-subsidized, animal-sourced products; and trends toward dominance of daily consumption of processed/ultra-processed foods. Both trends have well-documented adverse consequences, i.e., NCDs and endangered environmental/planetary health. Commitment to an evidence-informed plant-based eating pattern, particularly vegan, will reduce risks/manifestations of NCDs; inform healthy food and nutrition policy regulation/governance; support sustainable environment/climate and planetary health; and is ethical with respect to 'best' evidence-based practice, and human and animal welfare. CONCLUSION The Universal Food Guide that serves as a template for national food guides is both urgent and timely given the well-documented health-harming influences that corporate stakeholders/politicians and advisory committees with conflicts of interest, exert on national food guides. Such influence contributes to the largely-preventable NCDs and environmental issues. Policy makers, health professionals, and the public need unbiased, scientific evidence as informed by the Universal Food Guide, to inform their recommendations and choices.
Collapse
Affiliation(s)
- Elizabeth Dean
- Faculty of Medicine, Department of Physical Therapy, University of British Columbia, Vancouver, BC, Canada.
| | - Jia Xu
- Healing Without Medicine, Shenzhen, China
- Physicians Committee for Responsible Medicine, Washington, USA
| | - Alice Yee-Men Jones
- School of Health & Rehabilitation Sciences, The University of Queensland, Brisbane, Australia
| | | | | | - Pintu Kumar
- All India Institute of Medical Sciences, New Delhi, India
| | - Etienne Ngeh
- Louis University Institute, Douala, Cameroon
- Research Organisation for Health Education and Rehabilitation, and Guideline International Network African Regional Community, Yaoundé, Cameroon
| | - Maximilian A Storz
- Department of Internal Medicine II, Centre for Complementary Medicine, Medical Center, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
11
|
Hazell G, McCallion E, Ahlskog N, Sutton ER, Okoh M, Shaqoura EIH, Hoolachan JM, Scaife T, Iqbal S, Bhomra A, Kordala AJ, Scamps F, Raoul C, Wood MJA, Bowerman M. Exercise, disease state and sex influence the beneficial effects of Fn14-depletion on survival and muscle pathology in the SOD1 G93A amyotrophic lateral sclerosis (ALS) mouse model. Skelet Muscle 2024; 14:23. [PMID: 39396990 PMCID: PMC11472643 DOI: 10.1186/s13395-024-00356-0] [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: 07/05/2024] [Accepted: 10/04/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a devastating and incurable neurodegenerative disease. Accumulating evidence strongly suggests that intrinsic muscle defects exist and contribute to disease progression, including imbalances in whole-body metabolic homeostasis. We have previously reported that tumour necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) and fibroblast growth factor inducible 14 (Fn14) are significantly upregulated in skeletal muscle of the SOD1G93A ALS mouse model. While antagonising TWEAK did not impact survival, we did observe positive effects in skeletal muscle. Given that Fn14 has been proposed as the main effector of the TWEAK/Fn14 activity and that Fn14 can act independently from TWEAK in muscle, we suggest that manipulating Fn14 instead of TWEAK in the SOD1G93A ALS mice could lead to differential and potentially improved benefits. METHODS We thus investigated the contribution of Fn14 to disease phenotypes in the SOD1G93A ALS mice. To do so, Fn14 knockout mice (Fn14-/-) were crossed onto the SOD1G93A background to generate SOD1G93A;Fn14-/- mice. Investigations were performed on both unexercised and exercised (rotarod and/or grid test) animals (wild type (WT), Fn14-/-, SOD1G93A and SOD1G93A;Fn14-/-). RESULTS Here, we firstly confirm that the TWEAK/Fn14 pathway is dysregulated in skeletal muscle of SOD1G93A mice. We then show that Fn14-depleted SOD1G93A mice display increased lifespan, myofiber size, neuromuscular junction endplate area as well as altered expression of known molecular effectors of the TWEAK/Fn14 pathway, without an impact on motor function. Importantly, we also observe a complex interaction between exercise (rotarod and grid test), genotype, disease state and sex that influences the overall effects of Fn14 deletion on survival, expression of known molecular effectors of the TWEAK/Fn14 pathway, expression of myosin heavy chain isoforms and myofiber size. CONCLUSIONS Our study provides further insights on the different roles of the TWEAK/Fn14 pathway in pathological skeletal muscle and how they can be influenced by age, disease, sex and exercise. This is particularly relevant in the ALS field, where combinatorial therapies that include exercise regimens are currently being explored. As such, a better understanding and consideration of the interactions between treatments, muscle metabolism, sex and exercise will be of importance in future studies.
Collapse
Affiliation(s)
- Gareth Hazell
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Eve McCallion
- School of Medicine, Keele University, Staffordshire, UK
| | - Nina Ahlskog
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Emma R Sutton
- School of Medicine, Keele University, Staffordshire, UK
| | - Magnus Okoh
- School of Medicine, Keele University, Staffordshire, UK
| | | | | | - Taylor Scaife
- School of Life Sciences, Keele University, Staffordshire, UK
| | - Sara Iqbal
- School of Life Sciences, Keele University, Staffordshire, UK
| | - Amarjit Bhomra
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Anna J Kordala
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | | | - Cedric Raoul
- INM, Univ Montpellier, INSERM, Montpellier, France
- ALS Reference Center, Univ Montpellier, CHU Montpellier, Montpellier, France
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Melissa Bowerman
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
- School of Medicine, Keele University, Staffordshire, UK.
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry, UK.
| |
Collapse
|
12
|
Lavalle S, Scapaticci R, Masiello E, Messina C, Aliprandi A, Mario Salerno V, Russo A, Pegreffi F. Advancements in sarcopenia diagnosis: from imaging techniques to non-radiation assessments. FRONTIERS IN MEDICAL TECHNOLOGY 2024; 6:1467155. [PMID: 39445171 PMCID: PMC11496100 DOI: 10.3389/fmedt.2024.1467155] [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: 07/19/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024] Open
Abstract
Sarcopenia is a prevalent condition with significant clinical implications, and it is expected to escalate globally, demanding for effective diagnostic strategies, possibly at an early stage of the disease. Imaging techniques play a pivotal role in comprehensively evaluating sarcopenia, offering insights into both muscle quantity and quality. Among all the imaging techniques currently used for the diagnosis and follow up of sarcopenia, it is possible to distinguish two classes: Rx based techniques, using ionizing radiations, and non-invasive techniques, which are based on the use of safe and low risk diagnostic procedures. Dual-energy x-ray Absorptiometry and Computed Tomography, while widely utilized, entail radiation exposure concerns. Ultrasound imaging offers portability, real-time imaging, and absence of ionizing radiation, making it a promising tool Magnetic Resonance Imaging, particularly T1-weighted and Dixon sequences, provides cross- sectional and high-resolution images and fat-water separation capabilities, facilitating precise sarcopenia quantification. Bioelectrical Impedance Analysis (BIA), a non-invasive technique, estimates body composition, including muscle mass, albeit influenced by hydration status. Standardized protocols, such as those proposed by the Sarcopenia through Ultrasound (SARCUS) Working Group, are imperative for ensuring consistency across assessments. Future research should focus on refining these techniques and harnessing the potential of radiomics and artificial intelligence to enhance diagnostic accuracy and prognostic capabilities in sarcopenia.
Collapse
Affiliation(s)
- Salvatore Lavalle
- Department of Medicine and Surgery, Kore University of Enna, Enna, Italy
| | - Rosa Scapaticci
- Institute for the Electromagnetic Sensing of the Environment, National Research Council of Italy, Naples, Italy
| | - Edoardo Masiello
- Department of Radiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Carmelo Messina
- Department of Radiology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | | | | | - Arcangelo Russo
- Department of Medicine and Surgery, Kore University of Enna, Enna, Italy
| | - Francesco Pegreffi
- Department of Medicine and Surgery, Kore University of Enna, Enna, Italy
| |
Collapse
|
13
|
Silva WJ, Cruz A, Duque G. MicroRNAs and their Modulatory Effect on the Hallmarks of Osteosarcopenia. Curr Osteoporos Rep 2024; 22:458-470. [PMID: 39162945 DOI: 10.1007/s11914-024-00880-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/05/2024] [Indexed: 08/21/2024]
Abstract
PURPOSE OF THE REVIEW Osteosarcopenia is a geriatric syndrome associated with disability and mortality. This review summarizes the key microRNAs that regulate the hallmarks of sarcopenia and osteoporosis. Our objective was to identify components similarly regulated in the pathology and have therapeutic potential by influencing crucial cellular processes in both bone and skeletal muscle. RECENT FINDINGS The simultaneous decline in bone and muscle in osteosarcopenia involves a complex crosstalk between these tissues. Recent studies have uncovered several key mechanisms underlying this condition, including the disruption of cellular signaling pathways that regulate bone remodeling and muscle function and regeneration. Accordingly, emerging evidence reveals that dysregulation of microRNAs plays a significant role in the development of each of these hallmarks of osteosarcopenia. Although the recent recognition of osteosarcopenia as a single diagnosis of bone and muscle deterioration has provided new insights into the mechanisms of these underlying age-related diseases, several knowledge gaps have emerged, and a deeper understanding of the role of common microRNAs is still required. In this study, we summarize current evidence on the roles of microRNAs in the pathogenesis of osteosarcopenia and identify potential microRNA targets for treating this condition. Among these, microRNAs-29b and -128 are upregulated in the disease and exert adverse effects by inhibiting IGF-1 and SIRT1, making them potential targets for developing inhibitors of their activity. MicroRNA-21 is closely associated with the occurrence of muscle and bone loss. Conversely, microRNA-199b is downregulated in the disease, and its reduced activity may be related to increased myostatin and GSK3β activity, presenting it as a target for developing analogues that restore its function. Finally, microRNA-672 stands out for its ability to protect skeletal muscle and bone when expressed in the disease, highlighting its potential as a possible therapy for osteosarcopenia.
Collapse
Affiliation(s)
- William J Silva
- Department of Research and Development, Mirscience Therapeutics, São Paulo, Brazil
| | - André Cruz
- Department of Research and Development, Mirscience Therapeutics, São Paulo, Brazil
| | - Gustavo Duque
- Bone, Muscle & Geroscience Group. Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.
- Dr. Joseph Kaufmann Chair in Geriatric Medicine, Department of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
14
|
Guo A, Huang K, Lu Q, Tao B, Li K, Jiang D. TRIM16 facilitates SIRT-1-dependent regulation of antioxidant response to alleviate age-related sarcopenia. J Cachexia Sarcopenia Muscle 2024; 15:2056-2070. [PMID: 39192479 PMCID: PMC11446700 DOI: 10.1002/jcsm.13553] [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: 12/01/2023] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Age-related sarcopenia, characterized by reduced skeletal muscle mass and function, significantly affects the health of the elderly individuals. Oxidative stress plays a crucial role in the development of sarcopenia. Tripartite motif containing 16 (TRIM16) is implicated in orchestrating antioxidant responses to mitigate oxidative stress, yet its regulatory role in skeletal muscle remains unclear. This study aims to elucidate the impact of TRIM16 on enhancing antioxidant response through SIRT-1, consequently mitigating age-related oxidative stress, and ameliorating muscle atrophy. METHODS Aged mouse models were established utilizing male mice at 18 months with D-galactose (D-gal, 200 mg/kg) intervention and at 24 months with natural aging, while 3-month-old young mice served as controls. Muscle cell senescence was induced in C2C12 myoblasts using 30 g/L D-gal. TRIM16 was overexpressed in the skeletal muscle of aged mice and silenced/overexpressed in C2C12 myoblasts. The effects of TRIM16 on skeletal muscle mass, grip strength, morphological changes, myotube formation, myogenic differentiation, and muscle atrophy indicators were evaluated. Reactive oxygen species (ROS) levels and oxidative stress-related parameters were measured. The SIRT-1 inhibitor EX-527 was employed to elucidate the protective role of TRIM16 mediated through SIRT-1. RESULTS Aged mice displayed significant reductions in lean mass (-11.58%; -14.47% vs. young, P < 0.05), hindlimb lean mass (-17.38%; -15.95% vs. young, P < 0.05), and grip strength (-22.29%; -31.45% vs. young, P < 0.01). Skeletal muscle fibre cross-sectional area (CSA) decreased (-29.30%; -24.12% vs. young, P < 0.05). TRIM16 expression significantly decreased in aging skeletal muscle (-56.82%; -66.27% vs. young, P < 0.001) and senescent muscle cells (-46.53% vs. control, P < 0.001). ROS levels increased (+69.83% vs. control, P < 0.001), and myotube formation decreased in senescent muscle cells (-56.68% vs. control, P < 0.001). Expression of myogenic differentiation and antioxidant indicators decreased, while muscle atrophy markers increased in vivo and in vitro (all P < 0.05). Silencing TRIM16 in myoblasts induced oxidative stress and myotube atrophy, while TRIM16 overexpression partially mitigated aging effects on skeletal muscle. TRIM16 activation enhanced SIRT-1 expression (+75.38% vs. control, P < 0.001). SIRT-1 inhibitor EX-527 (100 μM) suppressed TRIM16's antioxidant response and mitigating muscle atrophy, offsetting the protective effect of TRIM16 on senescent muscle cells. CONCLUSIONS This study elucidates TRIM16's role in mitigating oxidative stress and ameliorating muscle atrophy through the activation of SIRT-1-dependent antioxidant effects. TRIM16 emerges as a potential therapeutic target for age-related sarcopenia.
Collapse
Affiliation(s)
- Ai Guo
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ke Huang
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Quanyi Lu
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bailong Tao
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kai Li
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dianming Jiang
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
15
|
Vantaggiato L, Landi C, Shaba E, Rossi D, Sorrentino V, Bini L. Protein Extraction Methods Suitable for Muscle Tissue Proteomic Analysis. Proteomes 2024; 12:27. [PMID: 39449499 PMCID: PMC11503273 DOI: 10.3390/proteomes12040027] [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: 07/05/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Muscle tissue is one of the most dynamic and plastic tissues of the mammalian body and covers different roles, such as force generation and metabolic control. Muscular proteomics provides an important opportunity to reveal the molecular mechanisms behind muscle pathophysiology. To ensure successful proteomic analysis, it is necessary to have an efficient and reproducible protein extraction method. This study aimed to evaluate the efficacy of two different extraction protocols of muscle samples for two-dimensional gel electrophoresis. In particular, mouse muscle proteins were extracted by an SDS-based buffer (Method A) and by a UREA/CHAPS/DTE/TRIS solution (Method B). The efficacies of the methods were assessed by performing an image analysis of the 2DE gels and by statistical and multivariate analyses. The 2DE gels in both preparations showed good resolution and good spot overlapping. Methods A and B produced 2DE gels with different means of total spots, higher for B. Image analysis showed different patterns of protein abundance between the protocols. The results showed that the two methods extract and solubilize proteins with different chemical-physical characteristics and different cellular localizations. These results attest the efficacy and reproducibility of both protein extraction methods, which can be parallelly applied for comprehensive proteomic profiling of muscle tissue.
Collapse
Affiliation(s)
- Lorenza Vantaggiato
- Functional Proteomics Lab., Department Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (L.V.); (E.S.); (L.B.)
| | - Claudia Landi
- Functional Proteomics Lab., Department Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (L.V.); (E.S.); (L.B.)
| | - Enxhi Shaba
- Functional Proteomics Lab., Department Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (L.V.); (E.S.); (L.B.)
| | - Daniela Rossi
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (D.R.); (V.S.)
| | - Vincenzo Sorrentino
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (D.R.); (V.S.)
| | - Luca Bini
- Functional Proteomics Lab., Department Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (L.V.); (E.S.); (L.B.)
| |
Collapse
|
16
|
Khalifeh DM, Czeglédi L, Gulyas G. Investigating the potential role of the pituitary adenylate cyclase-activating polypeptide (PACAP) in regulating the ubiquitin signaling pathway in poultry. Gen Comp Endocrinol 2024; 356:114577. [PMID: 38914296 DOI: 10.1016/j.ygcen.2024.114577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/13/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024]
Abstract
The physiological processes in animal production are regulated through biologically active molecules like peptides, proteins, and hormones identified through the development of the fundamental sciences and their application. One of the main polypeptides that plays an essential role in regulating physiological responses is the pituitary adenylate cyclase-activating polypeptide (PACAP). PACAP belongs to the glucagon/growth hormone-releasing hormone (GHRH)/vasoactive intestinal proteins (VIP) family and regulates feed intake, stress, and immune response in birds. Most of these regulations occur after PACAP stimulates the cAMP signaling pathway, which can regulate the expression of genes like MuRF1, FOXO1, Atrogin 1, and other ligases that are essential members of the ubiquitin system. On the other hand, PACAP stimulates the secretion of CRH in response to stress, activating the ubiquitin signaling pathway that plays a vital role in protein degradation and regulates oxidative stress and immune responses. Many studies conducted on rodents, mammals, and other models confirm the regulatory effects of PACAP, cAMP, and the ubiquitin pathway; however, there are no studies testing whether PACAP-induced cAMP signaling in poultry regulates the ubiquitin pathway. Besides, it would be interesting to investigate if PACAP can regulate ubiquitin signaling during stress response via CRH altered by HPA axis stimulation. Therefore, this review highlights a summary of research studies that indicate the potential interaction of the PACAP and ubiquitin signaling pathways on different molecular and physiological parameters in poultry species through the cAMP and stress signaling pathways.
Collapse
Affiliation(s)
- Doha Mohamad Khalifeh
- Department of Animal Science, Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Böszörményi Street 138, Debrecen 4032 Hungary; Doctoral School of Animal Science, University of Debrecen, Böszörményi Street 138, 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, Böszörményi Street 138, Debrecen 4032 Hungary
| | - Gabriella Gulyas
- Department of Animal Science, Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Böszörményi Street 138, Debrecen 4032 Hungary
| |
Collapse
|
17
|
Duranti E, Villa C. From Brain to Muscle: The Role of Muscle Tissue in Neurodegenerative Disorders. BIOLOGY 2024; 13:719. [PMID: 39336146 PMCID: PMC11428675 DOI: 10.3390/biology13090719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/02/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024]
Abstract
Neurodegenerative diseases (NDs), like amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), and Parkinson's disease (PD), primarily affect the central nervous system, leading to progressive neuronal loss and motor and cognitive dysfunction. However, recent studies have revealed that muscle tissue also plays a significant role in these diseases. ALS is characterized by severe muscle wasting as a result of motor neuron degeneration, as well as alterations in gene expression, protein aggregation, and oxidative stress. Muscle atrophy and mitochondrial dysfunction are also observed in AD, which may exacerbate cognitive decline due to systemic metabolic dysregulation. PD patients exhibit muscle fiber atrophy, altered muscle composition, and α-synuclein aggregation within muscle cells, contributing to motor symptoms and disease progression. Systemic inflammation and impaired protein degradation pathways are common among these disorders, highlighting muscle tissue as a key player in disease progression. Understanding these muscle-related changes offers potential therapeutic avenues, such as targeting mitochondrial function, reducing inflammation, and promoting muscle regeneration with exercise and pharmacological interventions. This review emphasizes the importance of considering an integrative approach to neurodegenerative disease research, considering both central and peripheral pathological mechanisms, in order to develop more effective treatments and improve patient outcomes.
Collapse
Affiliation(s)
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| |
Collapse
|
18
|
Zhang Q, Halle JL, Counts BR, Pi M, Carson JA. mTORC1 and BMP-Smad1/5 regulation of serum-stimulated myotube hypertrophy: a role for autophagy. Am J Physiol Cell Physiol 2024; 327:C124-C139. [PMID: 38766767 PMCID: PMC11371323 DOI: 10.1152/ajpcell.00237.2024] [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: 04/11/2024] [Revised: 05/14/2024] [Accepted: 05/14/2024] [Indexed: 05/22/2024]
Abstract
Protein synthesis regulation is critical for skeletal muscle hypertrophy, yet other established cellular processes are necessary for growth-related cellular remodeling. Autophagy has a well-acknowledged role in muscle quality control, but evidence for its role in myofiber hypertrophy remains equivocal. Both mammalian target of rapamycin complex I (mTORC1) and bone morphogenetic protein (BMP)-Smad1/5 (Sma and Mad proteins from Caenorhabditis elegans and Drosophila, respectively) signaling are reported regulators of myofiber hypertrophy; however, gaps remain in our understanding of how this regulation is integrated with growth processes and autophagy regulation. Therefore, we investigated the mTORC1 and Smad1/5 regulation of protein synthesis and autophagy flux during serum-stimulated myotube growth. Chronic serum stimulation experiments were performed on day 5 differentiated C2C12 myotubes incubated in differentiation medium [2% horse serum (HS)] or growth medium [5% fetal bovine serum (FBS)] for 48 h. Rapamycin or LDN193189 was dosed for 48 h to inhibit mTORC1 and BMP-Smad1/5 signaling, respectively. Acute serum stimulation was examined in day 7 differentiated myotubes. Protein synthesis was measured by puromycin incorporation. Bafilomycin A1 and immunoblotting for LC3B were used to assess autophagy flux. Chronic serum stimulation increased myotube diameter 22%, total protein 21%, total RNA 100%, and Smad1/5 phosphorylation 404% and suppressed autophagy flux. Rapamycin, but not LDN193189, blocked serum-induced myotube hypertrophy and the increase in total RNA. Acute serum stimulation increased protein synthesis 111%, Smad1/5 phosphorylation 559%, and rpS6 phosphorylation 117% and suppressed autophagy flux. Rapamycin increased autophagy flux during acute serum stimulation. These results provide evidence for mTORC1, but not BMP-Smad1/5, signaling being required for serum-induced myotube hypertrophy and autophagy flux by measuring LC3BII/I expression. Further investigation is warranted to examine the role of autophagy flux in myotube hypertrophy.NEW & NOTEWORTHY The present study demonstrates that myotube hypertrophy caused by chronic serum stimulation requires mammalian target of rapamycin complex 1 (mTORC1) signaling but not bone morphogenetic protein (BMP)-Smad1/5 signaling. The suppression of autophagy flux was associated with serum-induced myotube hypertrophy and mTORC1 regulation of autophagy flux by measuring LC3BII/I expression. Rapamycin is widely investigated for beneficial effects in aging skeletal muscle and sarcopenia; our results provide evidence that rapamycin can regulate autophagy-related signaling during myotube growth, which could benefit skeletal muscle functional and metabolic health.
Collapse
Affiliation(s)
- Quan Zhang
- Integrative Muscle Biology Laboratory, Division of Rehabilitation Sciences, College of Health ProfessionsUniversity of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Jessica L Halle
- Integrative Muscle Biology Laboratory, Division of Rehabilitation Sciences, College of Health ProfessionsUniversity of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Brittany R Counts
- Integrative Muscle Biology Laboratory, Division of Rehabilitation Sciences, College of Health ProfessionsUniversity of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Min Pi
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - James A Carson
- Huffines Institute for Sports Medicine & Human Performance, Department of Kinesiology & Sports Management , Texas A&M University, College Station, Texas, United States
| |
Collapse
|
19
|
Zhu Y, Hu Y, Pan Y, Li M, Niu Y, Zhang T, Sun H, Zhou S, Liu M, Zhang Y, Wu C, Ma Y, Guo Y, Wang L. Fatty infiltration in the musculoskeletal system: pathological mechanisms and clinical implications. Front Endocrinol (Lausanne) 2024; 15:1406046. [PMID: 39006365 PMCID: PMC11241459 DOI: 10.3389/fendo.2024.1406046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/10/2024] [Indexed: 07/16/2024] Open
Abstract
Fatty infiltration denotes the anomalous accrual of adipocytes in non-adipose tissue, thereby generating toxic substances with the capacity to impede the ordinary physiological functions of various organs. With aging, the musculoskeletal system undergoes pronounced degenerative alterations, prompting heightened scrutiny regarding the contributory role of fatty infiltration in its pathophysiology. Several studies have demonstrated that fatty infiltration affects the normal metabolism of the musculoskeletal system, leading to substantial tissue damage. Nevertheless, a definitive and universally accepted generalization concerning the comprehensive effects of fatty infiltration on the musculoskeletal system remains elusive. As a result, this review summarizes the characteristics of different types of adipose tissue, the pathological mechanisms associated with fatty infiltration in bone, muscle, and the entirety of the musculoskeletal system, examines relevant clinical diseases, and explores potential therapeutic modalities. This review is intended to give researchers a better understanding of fatty infiltration and to contribute new ideas to the prevention and treatment of clinical musculoskeletal diseases.
Collapse
Affiliation(s)
- Yihua Zhu
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yue Hu
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yalan Pan
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Traditional Chinese Medicine (TCM) Nursing Intervention Laboratory of Chronic Disease Key Laboratory, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Muzhe Li
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yuanyuan Niu
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Tianchi Zhang
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Haitao Sun
- Department of Orthopedic Surgery, Affiliated Huishan Hospital of Xinglin College of Nantong University, Wuxi, Jiangsu, China
| | - Shijie Zhou
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Mengmin Liu
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yili Zhang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Chengjie Wu
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yong Ma
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng TCM Hospital, Yancheng, Jiangsu, China
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, China
| | - Yang Guo
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, China
| | - Lining Wang
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Chinese Medicine Centre (International Collaboration between Western Sydney University and Beijing University of Chinese Medicine), Western Sydney University, Sydney, Australia
| |
Collapse
|
20
|
Sepúlveda-Lara A, Sepúlveda P, Marzuca-Nassr GN. Resistance Exercise Training as a New Trend in Alzheimer's Disease Research: From Molecular Mechanisms to Prevention. Int J Mol Sci 2024; 25:7084. [PMID: 39000191 PMCID: PMC11241132 DOI: 10.3390/ijms25137084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 07/16/2024] Open
Abstract
Alzheimer's disease is a pathology characterized by the progressive loss of neuronal connections, which leads to gray matter atrophy in the brain. Alzheimer's disease is the most prevalent type of dementia and has been classified into two types, early onset, which has been associated with genetic factors, and late onset, which has been associated with environmental factors. One of the greatest challenges regarding Alzheimer's disease is the high economic cost involved, which is why the number of studies aimed at prevention and treatment have increased. One possible approach is the use of resistance exercise training, given that it has been shown to have neuroprotective effects associated with Alzheimer's disease, such as increasing cortical and hippocampal volume, improving neuroplasticity, and promoting cognitive function throughout the life cycle. However, how resistance exercise training specifically prevents or ameliorates Alzheimer's disease has not been fully characterized. Therefore, the aim of this review was to identify the molecular basis by which resistance exercise training could prevent or treat Alzheimer's disease.
Collapse
Affiliation(s)
- Alexis Sepúlveda-Lara
- Doctorado en Ciencias mención Biología Celular y Molecular Aplicada, Facultad de Ciencias Agropecuarias, Universidad de La Frontera, Temuco 4811230, Chile;
| | - Paulina Sepúlveda
- Departamento de Ciencias Preclínicas, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile;
| | - Gabriel Nasri Marzuca-Nassr
- Departamento de Ciencias de la Rehabilitación, Facultad de Medicina, Universidad de la Frontera, Temuco 4811230, Chile
| |
Collapse
|
21
|
Artigas-Arias M, Curi R, Marzuca-Nassr GN. Myogenic microRNAs as Therapeutic Targets for Skeletal Muscle Mass Wasting in Breast Cancer Models. Int J Mol Sci 2024; 25:6714. [PMID: 38928418 PMCID: PMC11204047 DOI: 10.3390/ijms25126714] [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: 05/02/2024] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Breast cancer is the type of cancer with the highest prevalence in women worldwide. Skeletal muscle atrophy is an important prognostic factor in women diagnosed with breast cancer. This atrophy stems from disrupted skeletal muscle homeostasis, triggered by diminished anabolic signalling and heightened inflammatory conditions, culminating in an upregulation of skeletal muscle proteolysis gene expression. The importance of delving into research on modulators of skeletal muscle atrophy, such as microRNAs (miRNAs), which play a crucial role in regulating cellular signalling pathways involved in skeletal muscle protein synthesis and degradation, has been recognised. This holds true for conditions of homeostasis as well as pathologies like cancer. However, the determination of specific miRNAs that modulate skeletal muscle atrophy in breast cancer conditions has not yet been explored. In this narrative review, we aim to identify miRNAs that could directly or indirectly influence skeletal muscle atrophy in breast cancer models to gain an updated perspective on potential therapeutic targets that could be modulated through resistance exercise training, aiming to mitigate the loss of skeletal muscle mass in breast cancer patients.
Collapse
Affiliation(s)
- Macarena Artigas-Arias
- Programa de Doctorado en Ciencias Mención Biología Celular y Molecular Aplicada, Universidad de La Frontera, Temuco 4811230, Chile;
| | - Rui Curi
- Interdisciplinary Post-graduate Program in Health Sciences, Universidade Cruzeiro do Sul, São Paulo 01506-000, Brazil;
| | - Gabriel Nasri Marzuca-Nassr
- Departamento de Ciencias de la Rehabilitación, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
| |
Collapse
|
22
|
Vanhoutte D, Schips TG, Minerath RA, Huo J, Kavuri NSS, Prasad V, Lin SC, Bround MJ, Sargent MA, Adams CM, Molkentin JD. Thbs1 regulates skeletal muscle mass in a TGFβ-Smad2/3-ATF4-dependent manner. Cell Rep 2024; 43:114149. [PMID: 38678560 PMCID: PMC11217783 DOI: 10.1016/j.celrep.2024.114149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 03/22/2024] [Accepted: 04/09/2024] [Indexed: 05/01/2024] Open
Abstract
Loss of muscle mass is a feature of chronic illness and aging. Here, we report that skeletal muscle-specific thrombospondin-1 transgenic mice (Thbs1 Tg) have profound muscle atrophy with age-dependent decreases in exercise capacity and premature lethality. Mechanistically, Thbs1 activates transforming growth factor β (TGFβ)-Smad2/3 signaling, which also induces activating transcription factor 4 (ATF4) expression that together modulates the autophagy-lysosomal pathway (ALP) and ubiquitin-proteasome system (UPS) to facilitate muscle atrophy. Indeed, myofiber-specific inhibition of TGFβ-receptor signaling represses the induction of ATF4, normalizes ALP and UPS, and partially restores muscle mass in Thbs1 Tg mice. Similarly, myofiber-specific deletion of Smad2 and Smad3 or the Atf4 gene antagonizes Thbs1-induced muscle atrophy. More importantly, Thbs1-/- mice show significantly reduced levels of denervation- and caloric restriction-mediated muscle atrophy, along with blunted TGFβ-Smad3-ATF4 signaling. Thus, Thbs1-mediated TGFβ-Smad3-ATF4 signaling in skeletal muscle regulates tissue rarefaction, suggesting a target for atrophy-based muscle diseases and sarcopenia with aging.
Collapse
Affiliation(s)
- Davy Vanhoutte
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Tobias G Schips
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Rachel A Minerath
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jiuzhou Huo
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Naga Swathi Sree Kavuri
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Vikram Prasad
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Suh-Chin Lin
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michael J Bround
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michelle A Sargent
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Christopher M Adams
- Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jeffery D Molkentin
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
23
|
Xie S, Wu Q. Association between the systemic immune-inflammation index and sarcopenia: a systematic review and meta-analysis. J Orthop Surg Res 2024; 19:314. [PMID: 38802828 PMCID: PMC11131329 DOI: 10.1186/s13018-024-04808-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Sarcopenia is associated with increased morbidity and mortality. The systemic immune-inflammation index (SII) has been correlated to a variety of disorders. The present study conducted a systematic review and meta-analysis to investigate the relationship between SII and sarcopenia. METHODS A literature search was performed in Web of Science, PubMed, Embase, Cochrane Library, CINAHL, China National Knowledge Infrastructure, Chinese Biomedical Literature Database, Wanfang Database, and VIP Chinese Science and Technology Database, from inception to March 2024. Then, the literature quality was assessed. After the heterogeneity test, a random effects or fixed effects model was applied to establish the forest plot, and investigate the relationship between SII and sarcopenia. Then, the sensitivity analysis and publication bias were examined. RESULTS Nine articles, which included 18,634 adults, were analyzed. Sarcopenic adults had higher SII levels, when compared to non-sarcopenic adults (standardized mean difference [SMD] = 0.66, 95% confidence interval [CI] = 0.22 - 0.19, p = 0.003). The high SII level was associated to the increased risk of sarcopenia (odds ratio = 1.52, 95% CI = 1.09-2.13, p = 0.01). In addition, the subgroup analysis revealed that the SII levels were higher in the sarcopenic group, when compared to the non-sarcopenic group, in elderly adults, as well as in adults with or without gastrointestinal disorders. The analysis was robust with a low risk of publication bias. CONCLUSIONS SII is closely associated to sarcopenia. Sarcopenic adults had elevated SII levels. The high SII level increased the risk of sarcopenia. Large scale multi-center prospective studies are required to validate these study findings.
Collapse
Affiliation(s)
- Siye Xie
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou, 310000, China
| | - Qi Wu
- Department of Nursing, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, Zhejiang, 310003, China.
| |
Collapse
|
24
|
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
|
25
|
Salter D, Swamy S, Salis KM, Deep DK, Nadig P. A botanical extract blend of Mangifera indica and Sphaeranthus indicus combined with resistance exercise training improves muscle strength and endurance over exercise alone in young men: a randomized, blinded, placebo-controlled trial. Front Nutr 2024; 11:1393917. [PMID: 38765822 PMCID: PMC11099261 DOI: 10.3389/fnut.2024.1393917] [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: 02/29/2024] [Accepted: 04/19/2024] [Indexed: 05/22/2024] Open
Abstract
Resistance exercise training (RET) is used to improve muscular strength and function. This study tested the hypothesis that RET alongside daily supplementation of a Sphaeranthus indicus and Mangifera indica extract blend (SMI) would augment bench press (BP) and leg extension (LE) strength and repetitions to failure (RTF) compared to RET alone. Ninety-nine men (age 22 ± 3) completed the trial after randomization into one of four groups: (A1) 425 mg SMI plus one RET set; (A2) 850 mg SMI plus one RET set; (P1) placebo plus one RET set; and (P2) placebo plus two RET sets. RET sets were 6-8 BP and LE repetitions at 80% of a progressive one repetition maximum (1-RM), performed 3x/week for 8 weeks. Strength and RTF were evaluated at baseline and days 14, 28, and 56 while serum values of total testosterone (TT), free testosterone (FT), and cortisol (C) values were evaluated at baseline and day 56. RET significantly (p < 0.05) increased 1-RM, RTF, and T measures above baselines regardless of group assignment, but the increases were greater in the supplemented groups. At week 8, A1 bench pressed more than P1 (71.5.5 ± 17.5 kg vs. 62.0 ± 15.3 kg, p = 0.003), while A2 pressed 13.8 ± 3.0 kg more (95% CI 5.7-21.8, p < 0.001) than P1 and 9.9 ± 13.0 kg more (95% CI 1.7-18.2, p = 0.01) than P2. Also at week 8, the mean LE 1-RM of A1 (159.4 ± 22.6 kg) and A2 (162.2 ± 22.9 kg) was greater (p < 0.05) than that of P1 (142.2 ± 25.6 kg) and P2 (146.5 ± 19.7 kg). Supplementation improved RTF, TT, and FT values over those measured in exercise alone (p < 0.05), while C levels in A2 (9.3 ± 3.8 μg/dL) were lower than P2 (11.7 ± 3.8 μg/dL, p < 0.05). Daily supplementation with SMI was well tolerated and may help optimize muscle adaptive responses to RET in men.
Collapse
Affiliation(s)
- Dawna Salter
- Department of Clinical Research and Innovation, PLT Health Solutions, Inc., Morristown, NJ, United States
| | - Shubhatara Swamy
- Department of Pharmacology, Vydehi Institute of Medical Sciences and Research Centre, Bengaluru, India
| | - Kevin Manohar Salis
- Department of Pharmacology, Vydehi Institute of Medical Sciences and Research Centre, Bengaluru, India
| | | | - Pratibha Nadig
- Department of Pharmacology, Vydehi Institute of Medical Sciences and Research Centre, Bengaluru, India
| |
Collapse
|
26
|
Özen SD, Kir S. Ectodysplasin A2 receptor signaling in skeletal muscle pathophysiology. Trends Mol Med 2024; 30:471-483. [PMID: 38443222 DOI: 10.1016/j.molmed.2024.02.002] [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: 11/30/2023] [Revised: 02/05/2024] [Accepted: 02/09/2024] [Indexed: 03/07/2024]
Abstract
Skeletal muscle is essential in generating mechanical force and regulating energy metabolism and body temperature. Pathologies associated with muscle tissue often lead to impaired physical activity and imbalanced metabolism. Recently, ectodysplasin A2 receptor (EDA2R) signaling has been shown to promote muscle loss and glucose intolerance. Upregulated EDA2R expression in muscle tissue was associated with aging, denervation, cancer cachexia, and muscular dystrophies. Here, we describe the roles of EDA2R signaling in muscle pathophysiology, including muscle atrophy, insulin resistance, and aging-related sarcopenia. We also discuss the EDA2R pathway, which involves EDA-A2 as the ligand and nuclear factor (NF)κB-inducing kinase (NIK) as a downstream mediator, and the therapeutic potential of targeting these proteins in the treatment of muscle wasting and metabolic dysfunction.
Collapse
Affiliation(s)
- Sevgi Döndü Özen
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Serkan Kir
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey.
| |
Collapse
|
27
|
Prajapati P, Kumar A, Mangrulkar S, Chaple DR, Saraf SA, Kushwaha S. Azilsartan prevents muscle loss and fast- to slow-twitch muscle fiber shift in natural ageing sarcopenic rats. Can J Physiol Pharmacol 2024; 102:342-360. [PMID: 38118126 DOI: 10.1139/cjpp-2023-0265] [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: 12/22/2023]
Abstract
Sarcopenia is a musculoskeletal disease that reduces muscle mass and strength in older individuals. The study investigates the effects of azilsartan (AZL) on skeletal muscle loss in natural sarcopenic rats. Male Sprague-Dawley rats aged 4-6 months and 18-21 months were selected as young-matched control and natural-aged (sarcopenic) rats, respectively. Rats were allocated into young and old control (YC and OC) and young and old AZL treatment (YT and OT) groups, which received vehicles and AZL (8 mg/kg, orally) for 6 weeks. Rats were then sacrificed after muscle function analysis. Serum and gastrocnemius (GN) muscles were isolated for further endpoints. AZL significantly improved muscle grip strength and antioxidant levels in sarcopenic rats. AZL also restored the levels of insulin, testosterone, and muscle biomarkers such as myostatin and creatinine kinase in sarcopenic rats. Furthermore, AZL treatment improved the cellular and ultrastructure of GN muscle and prevented the shift of type II (glycolytic) myofibers to type I (oxidative) myofibers. The results showed that AZL intervention restored protein synthesis in natural sarcopenic rats by increasing p-Akt-1 and decreasing muscle RING-finger protein-1 and tumor necrosis factor alpha immunoexpressions. In conclusion, the present findings showed that AZL could be an effective intervention in treating age-related muscle impairments.
Collapse
MESH Headings
- Animals
- Sarcopenia/prevention & control
- Sarcopenia/metabolism
- Sarcopenia/drug therapy
- Sarcopenia/pathology
- Male
- Oxadiazoles/pharmacology
- Oxadiazoles/therapeutic use
- Aging/drug effects
- Rats, Sprague-Dawley
- Rats
- Benzimidazoles/pharmacology
- Benzimidazoles/therapeutic use
- Muscle Fibers, Fast-Twitch/drug effects
- Muscle Fibers, Fast-Twitch/metabolism
- Muscle Fibers, Fast-Twitch/pathology
- Muscle Fibers, Slow-Twitch/drug effects
- Muscle Fibers, Slow-Twitch/metabolism
- Muscle Fibers, Slow-Twitch/pathology
- Muscle Strength/drug effects
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Proto-Oncogene Proteins c-akt/metabolism
- Myostatin/metabolism
- Antioxidants/pharmacology
Collapse
Affiliation(s)
- Priyanka Prajapati
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Anand Kumar
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Shubhada Mangrulkar
- Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur 441002, India
| | - D R Chaple
- Priyadarshini J.L. College of Pharmacy, Electronic Zone Building, MIDC Hingna Road, Nagpur 440016, India
| | - Shubhini A Saraf
- National Institute of Pharmaceutical Education & Research, Raebareli (NIPER-R), Near CRPF Base Camp, Sarojini Nagar, Lucknow 226002, India
| | - Sapana Kushwaha
- National Institute of Pharmaceutical Education & Research, Raebareli (NIPER-R), Near CRPF Base Camp, Sarojini Nagar, Lucknow 226002, India
| |
Collapse
|
28
|
Zheng Y, Feng J, Yu Y, Ling M, Wang X. Advances in sarcopenia: mechanisms, therapeutic targets, and intervention strategies. Arch Pharm Res 2024; 47:301-324. [PMID: 38592582 DOI: 10.1007/s12272-024-01493-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/25/2024] [Indexed: 04/10/2024]
Abstract
Sarcopenia is a multifactorial condition characterized by loss of muscle mass. It poses significant health risks in older adults worldwide. Both pharmacological and non-pharmacological approaches are reported to address this disease. Certain dietary patterns, such as adequate energy intake and essential amino acids, have shown positive outcomes in preserving muscle function. Various medications, including myostatin inhibitors, growth hormones, and activin type II receptor inhibitors, have been evaluated for their effectiveness in managing sarcopenia. However, it is important to consider the variable efficacy and potential side effects associated with these treatments. There are currently no drugs approved by the Food and Drug Administration for sarcopenia. The ongoing research aims to develop more effective strategies in the future. Our review of research on disease mechanisms and drug development will be a valuable contribution to future research endeavors.
Collapse
Affiliation(s)
- Youle Zheng
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Jin Feng
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Yixin Yu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Min Ling
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.
| |
Collapse
|
29
|
Takahashi A, Honda Y, Tanaka N, Miyake J, Maeda S, Kataoka H, Sakamoto J, Okita M. Skeletal Muscle Electrical Stimulation Prevents Progression of Disuse Muscle Atrophy via Forkhead Box O Dynamics Mediated by Phosphorylated Protein Kinase B and Peroxisome Proliferator-Activated Receptor gamma Coactivator-1alpha. Physiol Res 2024; 73:105-115. [PMID: 38466009 PMCID: PMC11019614 DOI: 10.33549/physiolres.935157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 10/12/2023] [Indexed: 04/26/2024] Open
Abstract
Although electrical muscle stimulation (EMS) of skeletal muscle effectively prevents muscle atrophy, its effect on the breakdown of muscle component proteins is unknown. In this study, we investigated the biological mechanisms by which EMS-induced muscle contraction inhibits disuse muscle atrophy progression. Experimental animals were divided into a control group and three experimental groups: immobilized (Im; immobilization treatment), low-frequency (LF; immobilization treatment and low-frequency muscle contraction exercise), and high-frequency (HF; immobilization treatment and high-frequency muscle contraction exercise). Following the experimental period, bilateral soleus muscles were collected and analyzed. Atrogin-1 and Muscle RING finger 1 (MuRF-1) mRNA expression levels were significantly higher for the experimental groups than for the control group but were significantly lower for the HF group than for the Im group. Peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1alpha) mRNA and protein expression levels in the HF group were significantly higher than those in the Im group, with no significant differences compared to the Con group. Both the Forkhead box O (FoxO)/phosphorylated FoxO and protein kinase B (AKT)/phosphorylated AKT ratios were significantly lower for the Im group than for the control group and significantly higher for the HF group than for the Im group. These results, the suppression of atrogin-1 and MuRF-1 expression for the HF group may be due to decreased nuclear expression of FoxO by AKT phosphorylation and suppression of FoxO transcriptional activity by PGC-1alpha. Furthermore, the number of muscle contractions might be important for effective EMS.
Collapse
Affiliation(s)
- A Takahashi
- Institute of Biomedical Sciences (Health Sciences), Nagasaki University, Nagasaki, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Zhao J, Liang L, Zhang W, Liu X, Huo G, Liu X, Lv X, Zhao J. Sea buckthorn oil regulates primary myoblasts proliferation and differentiation in vitro. In Vitro Cell Dev Biol Anim 2024; 60:139-150. [PMID: 38153639 DOI: 10.1007/s11626-023-00841-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/11/2023] [Indexed: 12/29/2023]
Abstract
Skeletal muscle is the main edible part of meat products, and its development directly affects the yield and palatability of meat. Sea buckthorn oil (SBO) contains plenty of bioactive substances and has been recognized as a potential functional food product. The study aimed to explore the effects and possible mechanisms of SBO on sheep primary myoblast proliferation and myogenic differentiation. The results implied that SBO exhibited a pro-proliferative effect on primary myoblasts, along with up-regulated proliferating cell nuclear antigen (PCNA) and Cyclin D1/cyclin-dependent kinase 4 (CDK4) abundances. And, SBO promoted myotube formation by increasing the expression of myogenin. Meanwhile, we found that SBO inhibited the expression of miRNA-292a. Moreover, the regulatory effect of SBO on myogenic differentiation of myoblasts was attenuated by miRNA-292a mimics. Of note, SBO activated protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathway and augmented glucose uptake and glucose transporter 4 (GLUT4) content, which might be attributed to AMP-activated protein kinase (AMPK) activation. Additionally, the results were shown that SBO increased the abundance of antioxidative enzymes, including glutathione peroxidase 4 (Gpx4) and catalase. In summary, these data suggested that SBO regulated the proliferation and myogenic differentiation of sheep primary myoblasts in vitro, which might potentiate the application of SBO in muscle growth.
Collapse
Affiliation(s)
- Jiamin Zhao
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, 030801, People's Republic of China
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong, China
| | - Lin Liang
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, 030801, People's Republic of China
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong, China
| | - Weipeng Zhang
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, 030801, People's Republic of China
| | - Xuan Liu
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, 030801, People's Republic of China
| | - Guoqiang Huo
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, 030801, People's Republic of China
| | - Xiangdong Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Xiaoyang Lv
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou University, Yangzhou, 22500, People's Republic of China
| | - Junxing Zhao
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, 030801, People's Republic of China.
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong, China.
| |
Collapse
|
31
|
Ma Y, Cai G, Chen J, Yang X, Hua G, Han D, Li X, Feng D, Deng X. Combined transcriptome and metabolome analysis reveals breed-specific regulatory mechanisms in Dorper and Tan sheep. BMC Genomics 2024; 25:70. [PMID: 38233814 PMCID: PMC10795462 DOI: 10.1186/s12864-023-09870-9] [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: 06/05/2023] [Accepted: 12/04/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Dorper and Tan sheep are renowned for their rapid growth and exceptional meat quality, respectively. Previous research has provided evidence of the impact of gut microbiota on breed characteristics. The precise correlation between the gastrointestinal tract and peripheral organs in each breed is still unclear. Investigating the metabolic network of the intestinal organ has the potential to improve animal growth performance and enhance economic benefits through the regulation of intestinal metabolites. RESULTS In this study, we identified the growth advantage of Dorper sheep and the high fat content of Tan sheep. A transcriptome study of the brain, liver, skeletal muscle, and intestinal tissues of both breeds revealed 3,750 differentially expressed genes (DEGs). The genes PPARGC1A, LPL, and PHGDH were found to be highly expressed in Doper, resulting in the up-regulation of pathways related to lipid oxidation, glycerophospholipid metabolism, and amino acid anabolism. Tan sheep highly express the BSEP, LDLR, and ACHE genes, which up-regulate the pathways involved in bile transport and cholesterol homeostasis. Hindgut content analysis identified 200 differentially accumulated metabolites (DAMs). Purines, pyrimidines, bile acids, and fatty acid substances were more abundant in Dorper sheep. Based on combined gene and metabolite analyses, we have identified glycine, serine, and threonine metabolism, tryptophan metabolism, bile secretion, cholesterol metabolism, and neuroactive ligand-receptor interaction as key factors contributing to the differences among the breeds. CONCLUSIONS This study indicates that different breeds of sheep exhibit unique breed characteristics through various physiological regulatory methods. Dorper sheep upregulate metabolic signals related to glycine, serine, and threonine, resulting in an increase in purine and pyrimidine substances. This, in turn, promotes the synthesis of amino acids and facilitates body development, resulting in a faster rate of weight gain. Tan sheep accelerate bile transport, reduce bile accumulation in the intestine, and upregulate cholesterol homeostasis signals in skeletal muscles. This promotes the accumulation of peripheral and intramuscular fat, resulting in improved meat quality. This work adopts a joint analysis method of multi-tissue transcriptome and gut metabolome, providing a successful case for analyzing the mechanisms underlying the formation of various traits.
Collapse
Affiliation(s)
- Yuhao Ma
- Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture and Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing, 100193, China
| | - Ganxian Cai
- Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture and Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing, 100193, China
| | - Jianfei Chen
- Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture and Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing, 100193, China
| | - Xue Yang
- Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture and Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing, 100193, China
| | - Guoying Hua
- Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture and Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing, 100193, China
| | - Deping Han
- Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture and Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing, 100193, China
| | - Xinhai Li
- Department of Animal Science and college of Agriculture, Ningxia University, Yinchuan, 750021, China
| | - Dengzhen Feng
- Department of Animal Science and college of Agriculture, Ningxia University, Yinchuan, 750021, China
| | - Xuemei Deng
- Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture and Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
32
|
Zaripova KA, Belova SP, Kostrominova TY, Shenkman BS, Nemirovskaya TL. P2Y1 and P2Y2 receptors differ in their role in the regulation of signaling pathways during unloading-induced rat soleus muscle atrophy. Arch Biochem Biophys 2024; 751:109844. [PMID: 38043889 DOI: 10.1016/j.abb.2023.109844] [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: 08/02/2023] [Revised: 11/02/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
The current study aimed to investigate the hypothesis that purinergic receptors P2Y1 and P2Y2 play a regulatory role in gene expression in unloaded muscle. ATP is released from cells through pannexin channels, and it interacts with P2Y1 and P2Y2 receptors, leading to the activation of markers of protein catabolism and a reduction in protein synthesis. To test this hypothesis thirty-two rats were randomly divided into four groups (8 per group): a non-treated control group (C), a group subjected to three days of hindlimb unloading with a placebo (HS), a group subjected to three days of hindlimb unloading treated with a P2Y1 receptor inhibitor, MRS2179 (HSM), and a group subjected to three days of hindlimb unloading treated with a P2Y2 receptor inhibitor, AR-C 118925XX (HSA). This study revealed several key findings following three days of soleus muscle unloading: 1: Inhibition of P2Y1 or P2Y2 receptors prevented the accumulation of ATP, the increase in IP3 receptor content, and the decrease in the phosphorylation of GSK-3beta. This inhibition also mitigated the reduction in the rate of protein synthesis. However, it had no significant effect on the markers of mTORC1-dependent signaling. 2: Blocking P2Y1 receptors prevented the unloading-induced upregulation of phosphorylated p38MAPK and partially reduced the increase in MuRF1mRNA expression. 3: Blocking P2Y2 receptors prevented muscle atrophy during unloading, partially maintained the levels of phosphorylated ERK1/2, reduced the increase in mRNA expression of MAFbx, ubiquitin, and IL-6 receptor, prevented the decrease in phosphorylated AMPK, and attenuated the increase in phosphorylated p70S6K. Taken together, these results suggest that the prevention of muscle atrophy during unloading, as achieved by the P2Y2 receptor inhibitor, is likely mediated through a reduction in catabolic processes and maintenance of energy homeostasis. In contrast, the P2Y1 receptor appears to play a relatively minor role in muscle atrophy during unloading.
Collapse
Affiliation(s)
- Ksenia A Zaripova
- Myology Laboratory, Institute of Biomedical Problems, RAS, Moscow, Russia
| | - Svetlana P Belova
- Myology Laboratory, Institute of Biomedical Problems, RAS, Moscow, Russia
| | - Tatiana Y Kostrominova
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine-Northwest, Gary, IN, USA
| | - Boris S Shenkman
- Myology Laboratory, Institute of Biomedical Problems, RAS, Moscow, Russia
| | | |
Collapse
|
33
|
Liu C, Zhang Q, Liu T, Zhang Q, Song M, Ruan G, Lin S, Wang Z, Zheng X, Chen Y, Zhang H, Ge Y, Xie H, Shi J, Deng L, Wu S, Shi H. Predicted lean body mass trajectories, and cancer risk and cancer-specific and all-cause mortality: A prospective cohort study. J Cachexia Sarcopenia Muscle 2023; 14:2916-2924. [PMID: 37969022 PMCID: PMC10751432 DOI: 10.1002/jcsm.13370] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 09/12/2023] [Accepted: 10/03/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND Although many studies have investigated the association between body composition, cancer risk and mortality, predicting these risks through a single body composition measurement undoubtedly increases the limitations of the study. Few studies have explored the association between the trajectory of changes in body composition and the risk of cancer and death. We aimed to explore the association of predicted lean mass trajectories with cancer risk, cancer-specific mortality and all-cause mortality. METHODS The participants in this study were all from the Kailuan cohort, a prospective, periodic, resurvey cohort study initiated in 2006. Latent mixture modelling was used to identify predicted lean mass trajectories for 2006-2010. The hazard ratios (HRs) and 95% confidence intervals (95% CIs) of the Cox proportional hazard models were used to describe the association between predicted lean mass trajectories and cancer risk and cancer-specific and all-cause mortality during follow-up (2010-2021). RESULTS A total of 44 374 participants (average age, 53.01 ± 11.41 years, 78.99% men and 21.01% women) were enrolled in this study. Five distinct trajectories were identified: low-stable (n = 12 060), low-increasing (n = 8027), moderately stable-decreasing (n = 4725), moderately stable-increasing (n = 8053) and high-stable (n = 11 509). During the 11-year follow-up period, 2183 cancer events were recorded. After adjusting for age, predicted fat mass in 2010, sex, BMI, sedentary, physical activity, smoke, alcohol use, salt consumption, high-fat diet, high-sensitivity C-reactive protein, serum creatinine, family history of tumour, hypertension, diabetes mellitus, compared with the low-stable group, participants in the low-increasing group (HR = 0.851, 95% CI, 0.748-0.969), moderately stable-increasing group (HR = 0.803, 95% CI, 0.697-0.925) and high-stable group (HR = 0.770, 95% CI, 0.659-0.901) had a lower cancer risk, but not in the moderately stable-decreasing group (HR = 0.864, 95% CI, 0.735-1.015). Compared with the low-stable group, the risk of cancer-specific mortality was reduced by 25.4% (8.8-38.9%), 36.5% (20.3-49.4%) and 35.4% (17.9-49.2%), and the risk of all-cause mortality was reduced by 24.2% (16.9-30.8%), 37.0% (30.0-43.2%) and 47.4% (41.0-53.1%) in the low-increasing, moderately stable-increasing group and high-stable groups, respectively. CONCLUSIONS Predicted lean mass trajectories may be closely associated with cancer risk and cancer-specific and all-cause mortality. Regular monitoring of body composition is necessary.
Collapse
Affiliation(s)
- Chenan Liu
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Geriatric Diseases, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Cancer FSMP for State Market RegulationBeijingChina
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijingChina
| | - Qingsong Zhang
- Department of General SurgeryKailuan General HospitalTangshanChina
| | - Tong Liu
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Geriatric Diseases, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Cancer FSMP for State Market RegulationBeijingChina
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijingChina
| | - Qi Zhang
- Department of GeneticsYale University School of MedicineNew HavenConnecticutUSA
| | - Mengmeng Song
- Cardiovascular Research InstituteUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Guotian Ruan
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Geriatric Diseases, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Cancer FSMP for State Market RegulationBeijingChina
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijingChina
| | - Shiqi Lin
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Geriatric Diseases, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Cancer FSMP for State Market RegulationBeijingChina
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijingChina
| | - Ziwen Wang
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Geriatric Diseases, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Cancer FSMP for State Market RegulationBeijingChina
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijingChina
| | - Xin Zheng
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Geriatric Diseases, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Cancer FSMP for State Market RegulationBeijingChina
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijingChina
| | - Yue Chen
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Geriatric Diseases, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Cancer FSMP for State Market RegulationBeijingChina
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijingChina
| | - Heyang Zhang
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Geriatric Diseases, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Cancer FSMP for State Market RegulationBeijingChina
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijingChina
| | - Yizhong Ge
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Geriatric Diseases, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Cancer FSMP for State Market RegulationBeijingChina
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijingChina
| | - Hailun Xie
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Geriatric Diseases, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Cancer FSMP for State Market RegulationBeijingChina
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijingChina
| | - Jinyu Shi
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Geriatric Diseases, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Cancer FSMP for State Market RegulationBeijingChina
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijingChina
| | - Li Deng
- Key Laboratory of Cancer FSMP for State Market RegulationBeijingChina
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijingChina
| | - Shouling Wu
- Department of CardiologyKailuan General HospitalTangshanChina
| | - Hanping Shi
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Geriatric Diseases, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Cancer FSMP for State Market RegulationBeijingChina
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijingChina
| |
Collapse
|
34
|
Fennel ZJ, Ducharme JB, Berkemeier QN, Specht JW, McKenna ZJ, Simpson SE, Nava RC, Escobar KA, Hafen PS, Deyhle MR, Amorim FT, Mermier CM. Effect of heat stress on heat shock protein expression and hypertrophy-related signaling in the skeletal muscle of trained individuals. Am J Physiol Regul Integr Comp Physiol 2023; 325:R735-R749. [PMID: 37842742 DOI: 10.1152/ajpregu.00031.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/17/2023]
Abstract
Muscle mass is balanced between hypertrophy and atrophy by cellular processes, including activation of the protein kinase B-mechanistic target of rapamycin (Akt-mTOR) signaling cascade. Stressors apart from exercise and nutrition, such as heat stress, can stimulate the heat shock protein A (HSPA) and C (HSPC) families alongside hypertrophic signaling factors and muscle growth. The effects of heat stress on HSP expression and Akt-mTOR activation in human skeletal muscle and their magnitude of activation compared with known hypertrophic stimuli are unclear. Here, we show a single session of whole body heat stress following resistance exercise increases the expression of HSPA and activation of the Akt-mTOR cascade in skeletal muscle compared with resistance exercise in a healthy, resistance-trained population. Heat stress alone may also exert similar effects, though the responses are notably variable and require further investigation. In addition, acute heat stress in C2C12 muscle cells enhanced myotube growth and myogenic fusion, albeit to a lesser degree than growth factor-mediated hypertrophy. Though the mechanisms by which heat stress stimulates hypertrophy-related signaling and the potential mechanistic role of HSPs remain unclear, these findings provide additional evidence implicating heat stress as a novel growth stimulus when combined with resistance exercise in human skeletal muscle and alone in isolated murine muscle cells. We believe these findings will help drive further applied and mechanistic investigation into how heat stress influences muscular hypertrophy and atrophy.NEW & NOTEWORTHY We show that acute resistance exercise followed by whole body heat stress increases the expression of HSPA and increases activation of the Akt-mTOR cascade in a physically active and resistance-trained population.
Collapse
Affiliation(s)
- Zachary J Fennel
- Department of Health, Exercise & Sports Sciences, University of New Mexico, Albuquerque, New Mexico, United States
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States
| | - Jeremy B Ducharme
- Department of Health, Exercise & Sports Sciences, University of New Mexico, Albuquerque, New Mexico, United States
| | - Quint N Berkemeier
- Department of Health, Exercise & Sports Sciences, University of New Mexico, Albuquerque, New Mexico, United States
| | - Jonathan W Specht
- Department of Health, Exercise & Sports Sciences, University of New Mexico, Albuquerque, New Mexico, United States
| | - Zachary J McKenna
- Department of Health, Exercise & Sports Sciences, University of New Mexico, Albuquerque, New Mexico, United States
- Institute for Exercise and Environmental Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Shandy E Simpson
- Department of Health, Exercise & Sports Sciences, University of New Mexico, Albuquerque, New Mexico, United States
| | - Roberto C Nava
- Fulcrum Therapeutics, Cambridge, Massachusetts, United States
| | - Kurt A Escobar
- Department of Kinesiology, California State University Long Beach, Long Beach, California, United States
| | - Paul S Hafen
- Division of Science, Indiana University Purdue University Columbus, Columbus, Indiana, United States
- Department of Anatomy, Cell Biology, and Physiology, Indiana Center for Musculoskeletal Health, Indiana University School of Medicine Indianapolis, Indianapolis, Indiana, United States
| | - Michael R Deyhle
- Department of Health, Exercise & Sports Sciences, University of New Mexico, Albuquerque, New Mexico, United States
- Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, New Mexico, United States
| | - Fabiano T Amorim
- Department of Health, Exercise & Sports Sciences, University of New Mexico, Albuquerque, New Mexico, United States
| | - Christine M Mermier
- Department of Health, Exercise & Sports Sciences, University of New Mexico, Albuquerque, New Mexico, United States
| |
Collapse
|
35
|
Zeng Q, Du ZQ. Advances in the discovery of genetic elements underlying longissimus dorsi muscle growth and development in the pig. Anim Genet 2023; 54:709-720. [PMID: 37796678 DOI: 10.1111/age.13365] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 07/08/2023] [Accepted: 07/08/2023] [Indexed: 10/07/2023]
Abstract
As a major source of protein in human diets, pig meat plays a crucial role in ensuring global food security. Key determinants of meat production refer to the chemical and physical compositions or characteristics of muscle fibers, such as the number, hypertrophy potential, fiber-type conversion and intramuscular fat deposition. However, the growth and formation of muscle fibers comprises a complex process under spatio-temporal regulation, that is, the intermingled and concomitant proliferation, differentiation, migration and fusion of myoblasts. Recently, with the fast and continuous development of next-generation sequencing technology, the integration of quantitative trait loci mapping with genome-wide association studies (GWAS) has greatly helped animal geneticists to discover and explore thousands of functional or causal genetic elements underlying muscle growth and development. However, owing to the underlying complex molecular mechanisms, challenges to in-depth understanding and utilization remain, and the cost of large-scale sequencing, which requires integrated analyses of high-throughput omics data, is high. In this review, we mainly elaborate on research advances in integrative analyses (e.g. GWAS, omics) for identifying functional genes or genomic elements for longissimus dorsi muscle growth and development for different pig breeds, describing several successful transcriptome analyses and functional genomics cases, in an attempt to provide some perspective on the future functional annotation of genetic elements for muscle growth and development in pigs.
Collapse
Affiliation(s)
- Qingjie Zeng
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Zhi-Qiang Du
- College of Animal Science, Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
36
|
Elowe CR, Babbitt C, Gerson AR. White-throated sparrow ( Zonotrichia albicollis) liver and pectoralis flight muscle transcriptomic changes in preparation for migration. Physiol Genomics 2023; 55:544-556. [PMID: 37694280 DOI: 10.1152/physiolgenomics.00018.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 08/03/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023] Open
Abstract
Migratory songbirds undertake challenging journeys to reach their breeding grounds each spring. They accomplish these nonstop flapping feats of endurance through a suite of physiological changes, including the development of substantial fat stores and flight muscle hypertrophy and an increased capacity for fat catabolism. In addition, migratory birds may show large reductions in organ masses during flight, including the flight muscle and liver, which they must rapidly rebuild during their migratory stopover before replenishing their fat stores. However, the molecular basis of this capacity for rapid tissue remodeling and energetic output has not been thoroughly investigated. We performed RNA-sequencing analysis of the liver and pectoralis flight muscle of captive white-throated sparrows in experimentally photostimulated migratory and nonmigratory condition to explore the mechanisms of seasonal change to metabolism and tissue mass regulation that may facilitate these migratory journeys. Based on transcriptional changes, we propose that tissue-specific adjustments in preparation for migration may alleviate the damaging effects of long-duration activity, including a potential increase to the inflammatory response in the muscle. Furthermore, we hypothesize that seasonal hypertrophy balances satellite cell recruitment and apoptosis, while little evidence appeared in the transcriptome to support myostatin-, insulin-like growth factor 1-, and mammalian target of rapamycin-mediated pathways for muscle growth. These findings can encourage more targeted molecular studies on the unique integration of pathways that we find in the development of the migratory endurance phenotype in songbirds.NEW & NOTEWORTHY Migratory songbirds undergo significant physiological changes to accomplish their impressive migratory journeys. However, we have a limited understanding of the regulatory mechanisms underlying these changes. Here, we explore the transcriptomic changes to the flight muscle and liver of white-throated sparrows as they develop the migratory condition. We use these patterns to develop hypotheses about metabolic flexibility and tissue restructuring in preparation for migration.
Collapse
Affiliation(s)
- Cory R Elowe
- Department of Biology, University of Massachusetts, Amherst, Massachusetts, United States
- Organismic and Evolutionary Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, United States
| | - Courtney Babbitt
- Department of Biology, University of Massachusetts, Amherst, Massachusetts, United States
- Organismic and Evolutionary Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, United States
| | - Alexander R Gerson
- Department of Biology, University of Massachusetts, Amherst, Massachusetts, United States
- Organismic and Evolutionary Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, United States
| |
Collapse
|
37
|
Duranti E, Villa C. Muscle Involvement in Amyotrophic Lateral Sclerosis: Understanding the Pathogenesis and Advancing Therapeutics. Biomolecules 2023; 13:1582. [PMID: 38002264 PMCID: PMC10669302 DOI: 10.3390/biom13111582] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal condition characterized by the selective loss of motor neurons in the motor cortex, brainstem, and spinal cord. Muscle involvement, muscle atrophy, and subsequent paralysis are among the main features of this disease, which is defined as a neuromuscular disorder. ALS is a persistently progressive disease, and as motor neurons continue to degenerate, individuals with ALS experience a gradual decline in their ability to perform daily activities. Ultimately, muscle function loss may result in paralysis, presenting significant challenges in mobility, communication, and self-care. While the majority of ALS research has traditionally focused on pathogenic pathways in the central nervous system, there has been a great interest in muscle research. These studies were carried out on patients and animal models in order to better understand the molecular mechanisms involved and to develop therapies aimed at improving muscle function. This review summarizes the features of ALS and discusses the role of muscle, as well as examines recent studies in the development of treatments.
Collapse
Affiliation(s)
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| |
Collapse
|
38
|
Pinto AJ, Bergouignan A, Dempsey PC, Roschel H, Owen N, Gualano B, Dunstan DW. Physiology of sedentary behavior. Physiol Rev 2023; 103:2561-2622. [PMID: 37326297 PMCID: PMC10625842 DOI: 10.1152/physrev.00022.2022] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 05/10/2023] [Accepted: 06/14/2023] [Indexed: 06/17/2023] Open
Abstract
Sedentary behaviors (SB) are characterized by low energy expenditure while in a sitting or reclining posture. Evidence relevant to understanding the physiology of SB can be derived from studies employing several experimental models: bed rest, immobilization, reduced step count, and reducing/interrupting prolonged SB. We examine the relevant physiological evidence relating to body weight and energy balance, intermediary metabolism, cardiovascular and respiratory systems, the musculoskeletal system, the central nervous system, and immunity and inflammatory responses. Excessive and prolonged SB can lead to insulin resistance, vascular dysfunction, shift in substrate use toward carbohydrate oxidation, shift in muscle fiber from oxidative to glycolytic type, reduced cardiorespiratory fitness, loss of muscle mass and strength and bone mass, and increased total body fat mass and visceral fat depot, blood lipid concentrations, and inflammation. Despite marked differences across individual studies, longer term interventions aimed at reducing/interrupting SB have resulted in small, albeit marginally clinically meaningful, benefits on body weight, waist circumference, percent body fat, fasting glucose, insulin, HbA1c and HDL concentrations, systolic blood pressure, and vascular function in adults and older adults. There is more limited evidence for other health-related outcomes and physiological systems and for children and adolescents. Future research should focus on the investigation of molecular and cellular mechanisms underpinning adaptations to increasing and reducing/interrupting SB and the necessary changes in SB and physical activity to impact physiological systems and overall health in diverse population groups.
Collapse
Affiliation(s)
- Ana J Pinto
- Division of Endocrinology, Metabolism, and Diabetes, Anschutz Health and Wellness Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Applied Physiology & Nutrition Research Group, Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Audrey Bergouignan
- Division of Endocrinology, Metabolism, and Diabetes, Anschutz Health and Wellness Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Institut Pluridisciplinaire Hubert Curien, Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France
| | - Paddy C Dempsey
- Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Diabetes Research Centre, College of Life Sciences, University of Leicester, Leicester, United Kingdom
| | - Hamilton Roschel
- Applied Physiology & Nutrition Research Group, Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Neville Owen
- Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Centre for Urban Transitions, Swinburne University of Technology, Melbourne, Victoria, Australia
| | - Bruno Gualano
- Applied Physiology & Nutrition Research Group, Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
- Food Research Center, University of Sao Paulo, Sao Paulo, Brazil
| | - David W Dunstan
- Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| |
Collapse
|
39
|
Guo M, Zhang J, Ma Y, Zhu Z, Zuo H, Yao J, Wu X, Wang D, Yu J, Meng M, Liu C, Zhang Y, Chen J, Lu J, Ding S, Hu C, Ma X, Xu L. AAV-Mediated nuclear localized PGC1α4 delivery in muscle ameliorates sarcopenia and aging-associated metabolic dysfunctions. Aging Cell 2023; 22:e13961. [PMID: 37584432 PMCID: PMC10577532 DOI: 10.1111/acel.13961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/17/2023] [Accepted: 07/31/2023] [Indexed: 08/17/2023] Open
Abstract
Sarcopenia is characterized of muscle mass loss and functional decline in elder individuals which severely affects human physical activity, metabolic homeostasis, and life quality. Physical exercise is considered effective in combating muscle atrophy and sarcopenia, yet it is not feasible to elders with limited mobility. PGC-1α4, a short isoform of PGC-1α, is strongly induced in muscle under resistance training, and promotes muscle hypertrophy. In the present study, we showed that the transcriptional levels and nuclear localization of PGC1α4 was reduced during aging, accompanied with muscle dystrophic morphology, and gene programs. We thus designed NLS-PGC1α4 and ectopically express it in myotubes to enhance PGC1α4 levels and maintain its location in nucleus. Indeed, NLS-PGC1α4 overexpression increased muscle sizes in myotubes. In addition, by utilizing AAV-NLS-PGC1α4 delivery into gastrocnemius muscle, we found that it could improve sarcopenia with grip strength, muscle weights, fiber size and molecular phenotypes, and alleviate age-associated adiposity, insulin resistance and hepatic steatosis, accompanied with altered gene signatures. Mechanistically, we demonstrated that NLS-PGC-1α4 improved insulin signaling and enhanced glucose uptake in skeletal muscle. Besides, via RNA-seq analysis, we identified myokines IGF1 and METRNL as potential targets of NLS-PGC-1α4 that possibly mediate the improvement of muscle and adipose tissue functionality and systemic energy metabolism in aged mice. Moreover, we found a negative correlation between PGC1α4 and age in human skeletal muscle. Together, our results revealed that NLS-PGC1α4 overexpression improves muscle physiology and systematic energy homeostasis during aging and suggested it as a potent therapeutic strategy against sarcopenia and aging-associated metabolic diseases.
Collapse
Affiliation(s)
- Mingwei Guo
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life Sciences, East China Normal UniversityShanghaiChina
| | - Jun Zhang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life Sciences, East China Normal UniversityShanghaiChina
| | - Ying Ma
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life Sciences, East China Normal UniversityShanghaiChina
| | - Zhenzhong Zhu
- Department of OrthopedicsSixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hui Zuo
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life Sciences, East China Normal UniversityShanghaiChina
| | - Jing Yao
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life Sciences, East China Normal UniversityShanghaiChina
| | - Xia Wu
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life Sciences, East China Normal UniversityShanghaiChina
| | - Dongmei Wang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life Sciences, East China Normal UniversityShanghaiChina
| | - Jian Yu
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life Sciences, East China Normal UniversityShanghaiChina
- Department of Endocrinology and MetabolismFengxian Central Hospital Affiliated to Southern Medical UniversityShanghaiChina
| | - Meiyao Meng
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life Sciences, East China Normal UniversityShanghaiChina
| | - Caizhi Liu
- Department of Endocrinology and MetabolismFengxian Central Hospital Affiliated to Southern Medical UniversityShanghaiChina
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes MellitusShanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Yi Zhang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes MellitusShanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Jiangrong Chen
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life Sciences, East China Normal UniversityShanghaiChina
| | - Jian Lu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and HealthEast China Normal UniversityShanghaiChina
| | - Shuzhe Ding
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and HealthEast China Normal UniversityShanghaiChina
| | - Cheng Hu
- Department of Endocrinology and MetabolismFengxian Central Hospital Affiliated to Southern Medical UniversityShanghaiChina
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes MellitusShanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Xinran Ma
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life Sciences, East China Normal UniversityShanghaiChina
- Department of Endocrinology and MetabolismFengxian Central Hospital Affiliated to Southern Medical UniversityShanghaiChina
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life SciencesEast China Normal UniversityShanghaiChina
- Chongqing Key Laboratory of Precision OpticsChongqing Institute of East China Normal UniversityChongqingChina
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life Sciences, East China Normal UniversityShanghaiChina
| |
Collapse
|
40
|
Roberts MD, McCarthy JJ, Hornberger TA, Phillips SM, Mackey AL, Nader GA, Boppart MD, Kavazis AN, Reidy PT, Ogasawara R, Libardi CA, Ugrinowitsch C, Booth FW, Esser KA. Mechanisms of mechanical overload-induced skeletal muscle hypertrophy: current understanding and future directions. Physiol Rev 2023; 103:2679-2757. [PMID: 37382939 PMCID: PMC10625844 DOI: 10.1152/physrev.00039.2022] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/12/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023] Open
Abstract
Mechanisms underlying mechanical overload-induced skeletal muscle hypertrophy have been extensively researched since the landmark report by Morpurgo (1897) of "work-induced hypertrophy" in dogs that were treadmill trained. Much of the preclinical rodent and human resistance training research to date supports that involved mechanisms include enhanced mammalian/mechanistic target of rapamycin complex 1 (mTORC1) signaling, an expansion in translational capacity through ribosome biogenesis, increased satellite cell abundance and myonuclear accretion, and postexercise elevations in muscle protein synthesis rates. However, several lines of past and emerging evidence suggest that additional mechanisms that feed into or are independent of these processes are also involved. This review first provides a historical account of how mechanistic research into skeletal muscle hypertrophy has progressed. A comprehensive list of mechanisms associated with skeletal muscle hypertrophy is then outlined, and areas of disagreement involving these mechanisms are presented. Finally, future research directions involving many of the discussed mechanisms are proposed.
Collapse
Affiliation(s)
- Michael D Roberts
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - John J McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States
| | - Troy A Hornberger
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Stuart M Phillips
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Abigail L Mackey
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital-Bispebjerg and Frederiksberg, and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gustavo A Nader
- Department of Kinesiology and Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States
| | - Marni D Boppart
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Andreas N Kavazis
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Paul T Reidy
- Department of Kinesiology, Nutrition and Health, Miami University, Oxford, Ohio, United States
| | - Riki Ogasawara
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Cleiton A Libardi
- MUSCULAB-Laboratory of Neuromuscular Adaptations to Resistance Training, Department of Physical Education, Federal University of São Carlos, São Carlos, Brazil
| | - Carlos Ugrinowitsch
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Karyn A Esser
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
41
|
Aljilani B, Tsintzas K, Jacques M, Radford S, Moran GW. Systematic review: Sarcopenia in paediatric inflammatory bowel disease. Clin Nutr ESPEN 2023; 57:647-654. [PMID: 37739718 DOI: 10.1016/j.clnesp.2023.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/19/2023] [Accepted: 08/07/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND Low skeletal muscle mass (MM) and deteriorated function (sarcopenia) can be a frequent complication in paediatric inflammatory bowel disease (IBD). AIM To conduct a systematic review of the paediatric IBD literature on skeletal muscle function and mass and identify interventions that could affect them. METHODS Systematic searches (EMBASE, Medline, Cochrane library central for registered control trials and Web of Science) were conducted using the terms 'lean body mass' (LM), 'fat free mass' (FFM) or 'MM' and 'IBD'. RESULTS Fourteenth studies were included, presenting data from 439 Crohn's disease (CD), 139 ulcerative colitis (UC) and 2 IBD-unclassified participants compared with healthy matched or unmatched groups or reference populations. Six out of 14 studies reported lower LM, whilst 7 studies observed lower MM and FFM in CD patients compared to healthy controls. Research in UC patients reported lower LM in 3 studies, lower MM in 3 studies and lower FFM in 4 studies. Three prospective studies measured the impact of enteral feeding and showed improvement on disease activity and LM or FFM, while one retrospective study did not show any impact on LM. CONCLUSION Despite the variety of experimental approaches and methods used to assess sarcopenia, most studies showed reduction in MM, LM and FFM in IBD. Nutritional intervention may have a positive effect on LM and FFM. Future research should focus on standardizing the terminology and methodologies used in assessing body composition and investigating sarcopenia in diseased and matched healthy control cohorts in adequately powered studies with a longitudinal design.
Collapse
Affiliation(s)
- Bayan Aljilani
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, King Abdulaziz University, P.O. Box 80215, Jeddah, 21589, Saudi Arabia; Translational Medical Sciences and National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham NG7 2UH, UK
| | - Kostas Tsintzas
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, The University of Nottingham Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Matthew Jacques
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, The University of Nottingham Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Shellie Radford
- National Institute of Health Research Nottingham Biomedical Research Centre, University of Nottingham and Nottingham University Hospitals, Nottingham, NG7 2UH, UK
| | - Gordon W Moran
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, King Abdulaziz University, P.O. Box 80215, Jeddah, 21589, Saudi Arabia; National Institute of Health Research Nottingham Biomedical Research Centre, University of Nottingham and Nottingham University Hospitals, Nottingham, NG7 2UH, UK.
| |
Collapse
|
42
|
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: 0.5] [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
|
43
|
Prajapati P, Kumar A, Singh J, Saraf SA, Kushwaha S. Azilsartan Ameliorates Skeletal Muscle Wasting in High Fat Diet (HFD)-induced Sarcopenic Obesity in Rats via Activating Akt Signalling Pathway. Arch Gerontol Geriatr 2023; 112:105025. [PMID: 37062187 DOI: 10.1016/j.archger.2023.105025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/26/2023] [Accepted: 04/10/2023] [Indexed: 04/18/2023]
Abstract
An association between the loss of skeletal muscle mass and obesity in the geriatric population has been identified as a disease known as sarcopenic obesity. Therefore, therapeutic/preventive interventions are needed to ameliorate sarcopenia. The present study investigates the effect of azilsartan (AZL) on skeletal muscle loss in High-Fat Diet (HFD)-induced sarcopenic obese (SO) rats. Four- and fourteen-months male Sprague Dawley rats were used and randomized in control and azilsartan treatment. 14 months animals were fed with HFD for four months and labeled as HFD-fed SO rats. Young & old rats received 0.5% carboxymethyl cellulose as a vehicle/AZL (8 mg/kg, per oral) treatment for six weeks. Grip strength and body composition analysis were performed after the last dose of AZL. Serum and gastrocnemius (GN)muscles were collected after animal sacrifice. AZL treatment significantly increased lean muscle mass, grip strength, myofibrillar protein, and antioxidant (superoxide dismutase & nitric oxide) levels in SO rats. AZL also restored the muscle biomarkers (creatine kinase, myostatin & testosterone), and insulin levels. AZL improves cellular, and ultracellular muscle structure and prevents type I to type II myofiber transitions in SO rats. Further, immunohistochemistry results showed increased expressions of pAkt and reduced expression of MuRF-1 and TNF-α exhibiting that AZL intervention could decrease protein degradation in SO rats. In conclusion, present results showed that AZL significantly increased lean mass, and restored muscle biomarkers, and muscle architecture. Taken together, the aforementioned findings suggest that azilsartan could be a possible therapeutic approach to reduce muscle wasting in sarcopenic obesity.
Collapse
Affiliation(s)
- Priyanka Prajapati
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Anand Kumar
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Jiten Singh
- Department of Pharmaceutical Sciences, Central University of Haryana, Jant-Pali, Mahendergarh, Haryana 123031, India
| | - Shubhini A Saraf
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Sapana Kushwaha
- National Institute of Pharmaceutical Education & Research, Raebareli (NIPER-R), New Transit campus, Bijnor-Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow 226002, India.
| |
Collapse
|
44
|
Liu SH, Lin WH, Tzeng HP, Chiang MT. Attenuation of diabetes-mediated muscle atrophy in rats by fish oil enriched omega-3 polyunsaturated fatty acids supplementation. J Food Drug Anal 2023; 31:458-472. [PMID: 39666277 PMCID: PMC10629917 DOI: 10.38212/2224-6614.3468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/12/2023] [Indexed: 12/13/2024] Open
Abstract
Diabetes is associated with an increased risk of muscle wasting/atrophy, which adversely affects quality of life. We hypothesized that long term supplementation of fish oil may have protective effects against sarcopenia or muscle atrophy in streptozotocin (STZ) and high-fat (HF) diet-induced diabetic rat model. Wistar rats at age of 7 weeks were injected with saline or STZ to induce hyperglycemia. After one week, they were fed on a normal control diet or HF diet with/without supplementation of fish oil for 18 weeks. Feeding diabetic rats with a fish oil-enriched diet alleviated body weight loss and the impaired glucose tolerance using OGTT test. Although fish oil did not improve the decreased muscle mass, the muscle atrophy induced by diabetes was attenuated by fish oil in gastrocnemius, soleus, tibialis anterior, and extensor digitorum longus muscles. Fish oil supplementation reversed the decreased expression of phospho (p)-AKT, pmTOR, and p-p70s6k, which are molecules related to protein synthesis. Besides, protein degradation-related signaling pathways were inhibited by fish oil, such as increasing p-FoxO1 and decreasing Atrogin-1 and MURF1 protein expression. Fish oil down-regulated the expression of autophagy-related molecules including ATG5, p62, and LC3B II/I ratio, which may result in less muscle atrophy. Inflammation-related signaling regulators including TNF-α, NF-κB, AGEs, and RAGE were suppressed by fish oil supplementation as well. Moreover, the down-regulated p-AMPKα, SIRT1, and PGC-1 in diabetic rats were counteracted by fish oil, which may improve mitochondrial function and further block FoxO action. These data suggest that long-term fish oil supplementation exerts protective effects against diabetes-induced muscle atrophy, which may in turn ameliorate insulin resistance and impaired glucose tolerance.
Collapse
Affiliation(s)
- Shing-Hwa Liu
- Institute of Toxicology, National Taiwan University, Taipei,
Taiwan
- Department of Pediatrics, College of Medicine and Hospital, National Taiwan University, Taipei,
Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung,
Taiwan
| | - Wei-Hsuan Lin
- Department of Food Science, National Taiwan Ocean University, Keelung,
Taiwan
| | - Huei-Ping Tzeng
- Institute of Toxicology, National Taiwan University, Taipei,
Taiwan
| | - Meng-Tsan Chiang
- Department of Food Science, National Taiwan Ocean University, Keelung,
Taiwan
| |
Collapse
|
45
|
Bermejo-Álvarez I, Pérez-Baos S, Gratal P, Medina JP, Largo R, Herrero-Beaumont G, Mediero A. Effects of Tofacitinib on Muscle Remodeling in Experimental Rheumatoid Sarcopenia. Int J Mol Sci 2023; 24:13181. [PMID: 37685986 PMCID: PMC10487422 DOI: 10.3390/ijms241713181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/22/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Sarcopenia is a frequent comorbidity of rheumatoid arthritis (RA). Clinical trials have shown that JAK inhibitors (JAKi) produce an asymptomatic increase in serum creatine kinase (CK) in RA, suggesting an impact on muscle. We evaluated the effect of JAKi in muscle remodeling in an experimental RA model. Antigen-induced arthritis (experimental RA, e-RA) was performed in 14 rabbits. Seven rabbits received tofacitinib (TOFA, orally 10 mg/kg/day). Animals were euthanized one day after the last ovalbumin injection, and muscles were prepared for histology, RT-PCR, and WB. C-reactive protein (CRP) and Myostatin (MSTN) serum concentration were determined by ELISA. Creatine and creatine kinase (CK) were analyzed. An increase in body weight as well as tibialis anterior cross-sectional area and diameter was observed in e-RA+TOFA vs. e-RA. e-RA decreased type II fibers and increased the myonuclei number, with all reverted by TOFA. TOFA did not modify CRP levels, neither did MSTN. TOFA significantly reduced IL-6, atrogin-1, and MuRF-1 compared with e-RA. e-RA+TOFA showed higher CK and lower creatine levels compared with e-RA. No differences in PAX-7 were found, while TOFA prevented the increase in MyoD1 in e-RA. Our model reflects the features of rheumatoid sarcopenia in RA. JAKi increased muscle mass through attenuating IL-6/JAK/STAT activation, decreasing atrogenes, and restoring muscle differentiation markers. These data together with an increase in CK support the role of CK as a valuable marker of muscle gain following JAKi treatment.
Collapse
Affiliation(s)
| | | | | | | | - Raquel Largo
- Bone and Joint Research Unit, Rheumatology Department, IIS-Fundación Jiménez Díaz UAM, 28040 Madrid, Spain
| | | | | |
Collapse
|
46
|
Zhou J, Zhao Y, Dai J, Zhang K. Environmentally relevant concentrations of antidepressant mirtazapine impair the neurodevelopment of zebrafish (Danio rerio). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 262:115335. [PMID: 37567106 DOI: 10.1016/j.ecoenv.2023.115335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/30/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
Mirtazapine is a commonly prescribed antidepressant and has been found widespread in aquatic environments. However, its toxicities to aquatic organisms has rarely been explored. Herein, we conducted a comprehensive study on the developmental effects of mirtazapine on early life stages of zebrafish at environmentally relevant concentrations (3.9 ng/L and 43.5 ng/L). Out of the endpoints measured, spontaneous contraction of embryos at 24 h post fertilization (hpf) and hatching rate and heart rate of embryos at 50 hpf and 56 hpf, respectively, were significantly affected. In light-dark transition behavior test, mirtazapine significantly reduced the swimming frequency and swimming speed of embryos at both concentrations of 3.9 ng/L and 43.5 ng/L. Furthermore, the total swimming distances in dark conditions were also significantly reduced. Transcriptomic analysis was further conducted. It demonstrated that the decreased neural activities in embryos may be associated with altered epinephrine and neuregulin signaling. The present results fill a data gap regarding the exposure of fish to mirtazapine at environmentally relevant concentrations and provide new insights into the neurotoxic mechanisms of mirtazapine exposure.
Collapse
Affiliation(s)
- Jie Zhou
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Yanbin Zhao
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Jiayin Dai
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Kun Zhang
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| |
Collapse
|
47
|
Chen X, Ji Y, Liu R, Zhu X, Wang K, Yang X, Liu B, Gao Z, Huang Y, Shen Y, Liu H, Sun H. Mitochondrial dysfunction: roles in skeletal muscle atrophy. J Transl Med 2023; 21:503. [PMID: 37495991 PMCID: PMC10373380 DOI: 10.1186/s12967-023-04369-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/18/2023] [Indexed: 07/28/2023] Open
Abstract
Mitochondria play important roles in maintaining cellular homeostasis and skeletal muscle health, and damage to mitochondria can lead to a series of pathophysiological changes. Mitochondrial dysfunction can lead to skeletal muscle atrophy, and its molecular mechanism leading to skeletal muscle atrophy is complex. Understanding the pathogenesis of mitochondrial dysfunction is useful for the prevention and treatment of skeletal muscle atrophy, and finding drugs and methods to target and modulate mitochondrial function are urgent tasks in the prevention and treatment of skeletal muscle atrophy. In this review, we first discussed the roles of normal mitochondria in skeletal muscle. Importantly, we described the effect of mitochondrial dysfunction on skeletal muscle atrophy and the molecular mechanisms involved. Furthermore, the regulatory roles of different signaling pathways (AMPK-SIRT1-PGC-1α, IGF-1-PI3K-Akt-mTOR, FoxOs, JAK-STAT3, TGF-β-Smad2/3 and NF-κB pathways, etc.) and the roles of mitochondrial factors were investigated in mitochondrial dysfunction. Next, we analyzed the manifestations of mitochondrial dysfunction in muscle atrophy caused by different diseases. Finally, we summarized the preventive and therapeutic effects of targeted regulation of mitochondrial function on skeletal muscle atrophy, including drug therapy, exercise and diet, gene therapy, stem cell therapy and physical therapy. This review is of great significance for the holistic understanding of the important role of mitochondria in skeletal muscle, which is helpful for researchers to further understanding the molecular regulatory mechanism of skeletal muscle atrophy, and has an important inspiring role for the development of therapeutic strategies for muscle atrophy targeting mitochondria in the future.
Collapse
Affiliation(s)
- Xin Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yanan Ji
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Ruiqi Liu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Xucheng Zhu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Kexin Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Xiaoming Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Boya Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Zihui Gao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yan Huang
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| | - Hua Liu
- Department of Orthopedics, Haian Hospital of Traditional Chinese Medicine, 55 Ninghai Middle Road, Nantong, Jiangsu, 226600, People's Republic of China.
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
48
|
Cao H, Du T, Li C, Wu L, Liu J, Guo Y, Li X, Yang G, Jin J, Shi X. MicroRNA-668-3p inhibits myoblast proliferation and differentiation by targeting Appl1. BMC Genomics 2023; 24:415. [PMID: 37488537 PMCID: PMC10364376 DOI: 10.1186/s12864-023-09431-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/06/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND Skeletal muscle is the largest tissue in the body, and it affects motion, metabolism and homeostasis. Skeletal muscle development comprises myoblast proliferation, fusion and differentiation to form myotubes, which subsequently form mature muscle fibres. This process is strictly regulated by a series of molecular networks. Increasing evidence has shown that noncoding RNAs, especially microRNAs (miRNAs), play vital roles in regulating skeletal muscle growth. Here, we showed that miR-668-3p is highly expressed in skeletal muscle. METHODS Proliferating and differentiated C2C12 cells were transfected with miR-668-3p mimics and/or inhibitor, and the mRNA and protein levels of its target gene were evaluated by RT‒qPCR and Western blotting analysis. The targeting of Appl1 by miR-668-3p was confirmed by dual luciferase assay. The interdependence of miR-668-3p and Appl1 was verified by cotransfection of C2C12 cells. RESULTS Our data reveal that miR-668-3p can inhibit myoblast proliferation and myogenic differentiation. Phosphotyrosine interacting with PH domain and leucine zipper 1 (Appl1) is a target gene of miR-668-3p, and it can promote myoblast proliferation and differentiation by activating the p38 MAPK pathway. Furthermore, the inhibitory effect of miR-668-3p on myoblast cell proliferation and myogenic differentiation could be rescued by Appl1. CONCLUSION Our results indicate a new mechanism by which the miR-668-3p/Appl1/p38 MAPK pathway regulates skeletal muscle development.
Collapse
Affiliation(s)
- Haigang Cao
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Tianning Du
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- Microbial Research Institute of Liaoning Province, Chaoyang, Liaoning, China
| | - Chenchen Li
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Lingling Wu
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Jieming Liu
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Yuan Guo
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiao Li
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Gongshe Yang
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Jianjun Jin
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.
| | - Xin'e Shi
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
49
|
Picca A, Guerra F, Calvani R, Romano R, Coelho-Junior HJ, Bucci C, Leeuwenburgh C, Marzetti E. Mitochondrial-derived vesicles in skeletal muscle remodeling and adaptation. Semin Cell Dev Biol 2023; 143:37-45. [PMID: 35367122 DOI: 10.1016/j.semcdb.2022.03.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/25/2022] [Accepted: 03/19/2022] [Indexed: 12/24/2022]
Abstract
Mitochondrial remodeling is crucial to meet the bioenergetic demand to support muscle contractile activity during daily tasks and muscle regeneration following injury. A set of mitochondrial quality control (MQC) processes, including mitochondrial biogenesis, dynamics, and mitophagy, are in place to maintain a well-functioning mitochondrial network and support muscle regeneration. Alterations in any of these pathways compromises mitochondrial quality and may potentially lead to impaired myogenesis, defective muscle regeneration, and ultimately loss of muscle function. Among MQC processes, mitophagy has gained special attention for its implication in the clearance of dysfunctional mitochondria via crosstalk with the endo-lysosomal system, a major cell degradative route. Along this pathway, additional opportunities for mitochondrial disposal have been identified that may also signal at the systemic level. This communication occurs via inclusion of mitochondrial components within membranous shuttles named mitochondrial-derived vesicles (MDVs). Here, we discuss MDV generation and release as a mitophagy-complementing route for the maintenance of mitochondrial homeostasis in skeletal myocytes. We also illustrate the possible role of muscle-derived MDVs in immune signaling during muscle remodeling and adaptation.
Collapse
Affiliation(s)
- Anna Picca
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies, Università del Salento, Lecce, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Roberta Romano
- Department of Biological and Environmental Sciences and Technologies, Università del Salento, Lecce, Italy
| | | | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies, Università del Salento, Lecce, Italy
| | - Christiaan Leeuwenburgh
- Department of Aging and Geriatric Research, Institute on Aging, Division of Biology of Aging, University of Florida, Gainesville, USA
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy; Università Cattolica del Sacro Cuore, Department of Geriatrics and Orthopedics, Rome, Italy.
| |
Collapse
|
50
|
Luo X, Zhang H, Cao X, Yang D, Yan Y, Lu J, Wang X, Wang H. Endurance Exercise-Induced Fgf21 Promotes Skeletal Muscle Fiber Conversion through TGF-β1 and p38 MAPK Signaling Pathway. Int J Mol Sci 2023; 24:11401. [PMID: 37511159 PMCID: PMC10379449 DOI: 10.3390/ijms241411401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Fgf21 has been identified as playing a regulatory role in muscle growth and function. Although the mechanisms through which endurance training regulates skeletal muscle have been widely studied, the contribution of Fgf21 remains poorly understood. Here, muscle size and function were measured, and markers of fiber type were evaluated using immunohistochemistry, immunoblots, or qPCR in endurance-exercise-trained wild-type and Fgf21 KO mice. We also investigated Fgf21-induced fiber conversion in C2C12 cells, which were incubated with lentivirus and/or pathway inhibitors. We found that endurance exercise training enhanced the Fgf21 levels of liver and GAS muscle and exercise capacity and decreased the distribution of skeletal muscle fiber size, and fast-twitch fibers were observed converting to slow-twitch fibers in the GAS muscle of mice. Fgf21 promoted the markers of fiber-type transition and eMyHC-positive myotubes by inhibiting the TGF-β1 signaling axis and activating the p38 MAPK signaling pathway without apparent crosstalk. Our findings suggest that the transformation and function of skeletal muscle fiber types in response to endurance training could be mediated by Fgf21 and its downstream signaling pathways. Our results illuminate the mechanisms of Fgf21 in endurance-exercise-induced fiber-type conversion and suggest a potential use of Fgf21 in improving muscle health and combating fatigue.
Collapse
Affiliation(s)
- Xiaomao Luo
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
| | - Huiling Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
| | - Xiaorui Cao
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
| | - Ding Yang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yi Yan
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
| | - Jiayin Lu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
| | - Xiaonan Wang
- Renal Division, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Haidong Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
| |
Collapse
|