1
|
Yu Y, Jiang X, Yu T, Chen F, Huang R, Xun Z, Wang X, Liu X, Xie X, Sun C, Xu Y, Liu X, Sun H, Yuan X, Ma C, Li Y, Song X, Wang D, Shao D, Shi X, Cao L. Maintaining myoprotein and redox homeostasis via an orally recharged nanoparticulate supplement potentiates sarcopenia treatment. Biomaterials 2025; 314:122863. [PMID: 39366185 DOI: 10.1016/j.biomaterials.2024.122863] [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: 04/10/2024] [Revised: 09/10/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Sarcopenia is a progressive skeletal muscle disorder characterized by the accelerated loss of muscle mass and function, with no promising pharmacotherapies. Understanding the imbalance of myoprotein homeostasis within myotubes, which causes sarcopenia, may facilitate the development of novel treatments for clinical use. In this study, we found a strong correlation between low serum selenium levels and muscle function in elderly patients with sarcopenia. We hypothesized that supplementation with selenium might be beneficial for the management of sarcopenia. To verify this hypothesis, we developed diselenide-bridged mesoporous silica nanoparticles (Se-Se-MSNs) with ROS-responsive degradation and release to supplement selenium. Se-Se-MSNs outperformed free selenocysteine in alleviating sarcopenia in both dexamethasone (Dex)- and denervation-induced mouse models. Subsequently, Se-Se-MSNs were loaded with leucine (Leu@Se-Se-MSNs), another nutritional supplement used in sarcopenia management. Oral administration of Leu@Se-Se-MSNs restored myoprotein homeostasis by enhancing mTOR/S6K signaling and inactivating Akt/FoxO3a/MuRF1 signaling, thus exerting optimal therapeutic effects against sarcopenia and exhibiting a more favorable in vivo safety profile. This study provides a proof of concept for treating sarcopenia by maintaining myoprotein and redox homeostasis simultaneously and offers valuable insights into the development of multifunctional nanoparticle-based supplements for sarcopenia management.
Collapse
Affiliation(s)
- Yang Yu
- Health Sciences Institute, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Xuehan Jiang
- Health Sciences Institute, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China; Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shanxi, China
| | - Tianhao Yu
- The VIP Department, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110002, Liaoning, China
| | - Fangman Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, Guangzhou, China
| | - Runnian Huang
- Department of Gerontology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China; Department of Epidemiology and Health Statistics, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Zhe Xun
- Health Sciences Institute, China Medical University, Shenyang 110122, Liaoning, China
| | - Xiaoxun Wang
- Health Sciences Institute, China Medical University, Shenyang 110122, Liaoning, China
| | - Xu Liu
- Health Sciences Institute, China Medical University, Shenyang 110122, Liaoning, China
| | - Xiaochun Xie
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Chen Sun
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Yingxi Xu
- Health Sciences Institute, China Medical University, Shenyang 110122, Liaoning, China
| | - Xiyan Liu
- Health Sciences Institute, China Medical University, Shenyang 110122, Liaoning, China
| | - Huayi Sun
- Health Sciences Institute, China Medical University, Shenyang 110122, Liaoning, China
| | - Xiaoyue Yuan
- Department of Gerontology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China; Department of Epidemiology and Health Statistics, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Chunhua Ma
- Department of Gerontology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China; Department of Epidemiology and Health Statistics, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Yibai Li
- Health Sciences Institute, China Medical University, Shenyang 110122, Liaoning, China
| | - Xiaoyu Song
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China.
| | - Difei Wang
- Department of Gerontology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China.
| | - Dan Shao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, Guangzhou, China; School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Xuetao Shi
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, Guangzhou, China.
| | - Liu Cao
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
2
|
Yue H, Huan Y, Ren P, Yu X, Tang Q, Xue C, Xu J. Astaxanthin ameliorates dexamethasone-induced skeletal muscle atrophy and disorders of glucolipid metabolism. Biochem Biophys Res Commun 2025; 743:151138. [PMID: 39673970 DOI: 10.1016/j.bbrc.2024.151138] [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: 09/22/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/16/2024]
Abstract
Long-term use of glucocorticoids, such as dexamethasone, can lead to skeletal muscle atrophy and disturbances in glucolipid metabolism. Astaxanthin, a ketocarotenoid, has a variety of physiological activities. In this study, we investigated the effects of astaxanthin on dexamethasone-induced skeletal muscle atrophy and disorders of glycolipid metabolism. Male C57Bl/6J mice were administered dexamethasone alone or supplemented with different doses of astaxanthin (30 mg/kg/d, 60 mg/kg/d, 120 mg/kg/d) for 4 weeks. The results showed that astaxanthin improved locomotor performance and attenuated dexamethasone-induced reductions in skeletal muscle mass and cross-sectional area in mice. Further exploration of the mechanism revealed that astaxanthin was able to balance the catabolism and synthesis of skeletal muscle proteins and protect mitochondria by reducing the expression of the mitochondrial autophagy-associated proteins Lc3B and BNIP3. In addition, astaxanthin reduced the accumulation of fatty acid metabolites and visceral fat and improved the ability of skeletal muscle to utilize sugars, suggesting that astaxanthin may alleviate dexamethasone-induced disturbances in glycolipid metabolism. This study illustrates that astaxanthin can alleviate skeletal muscle atrophy and metabolic disturbances induced by long-term dexamethasone treatment. This suggests that astaxanthin may be a functional dietary supplement to reduce the side effects of glucocorticoid therapy.
Collapse
Affiliation(s)
- Han Yue
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266400, China
| | - Yuchen Huan
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266400, China
| | - Pengfei Ren
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266400, China
| | - Xinyue Yu
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266400, China
| | - Qingjuan Tang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266400, China.
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266400, China; Laboratory of Marine Drugs and Biological Products, Pilot National Laboratory for Marine Science and Technology, Qingdao, 266000, China
| | - Jie Xu
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266400, China
| |
Collapse
|
3
|
Kim A, Kim J, Kim BY, Seo CS, Kim YR, Song KH, Kim NS. Aquo-ethanolic extract of Lilii Bulbus attenuates dexamethasone-induced muscle loss and enhances muscle strength in experimental mice. Biomed Pharmacother 2024; 181:117658. [PMID: 39486367 DOI: 10.1016/j.biopha.2024.117658] [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: 07/25/2024] [Revised: 09/12/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
Traditionally, Lilium lancifolium bulb is known for its ability to nourish yin, nourish the lungs, clear the heart, soothe coughs, reduce irritability, and calm the mind. In Oriental Medicine, it is categorized as a tonic remedy for alleviating symptoms of fatigue and enhancing the strength of bones and muscles. In this study, we aimed to validate the effectiveness of the aquo-ethanolic extract of Lilli Bulbus (LBE) in a dexamethasone (DEX)-induced muscle atrophy model, both in vitro and in vivo, and elucidate its mechanism of action through muscle transcriptome analysis. The effects of LBE on the viability and myotube density of C2C12 myoblasts and differentiated C2C12 myotubes with and without DEX treatment were investigated. LBE pretreatment protected C2C12 myoblast cells and increased the muscle density of C2C12 myotubes in response to DEX. LBE showed potent free radical scavenging activities in cell-free biochemical assays as well as antioxidant activity in C2C12 myoblasts. LBE also exhibited protective effects in an experimental animal model of DEX-induced muscle atrophy, showing muscular function and motor coordination recovery. Transcriptomic analysis of three different muscle tissues from mice with DEX-induced muscle atrophy showed that the regulation of the extracellular matrix was perturbed by glucocorticoid treatment, and this perturbation was reversed by LBE treatment. Collectively, LBE alleviated skeletal muscle loss and maintained muscle function from the chronic toxicity of DEX by protecting muscle cells from various stressful conditions, as well as DEX itself, inhibiting muscle protein degradation, and preserving the muscle tissue microenvironment.
Collapse
Affiliation(s)
- Aeyung Kim
- KM Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea.
| | - Jinhee Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| | - Bu-Yeo Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| | - Chang-Seob Seo
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| | - Yu Ri Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| | - Kwang Hoon Song
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| | - No Soo Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| |
Collapse
|
4
|
Yoo J, Kim C, Lee H, Ko BS, Lee DW, Hwang JK. Fermented antler extract attenuates muscle atrophy by regulating the PI3K/Akt pathway and inflammatory response in immobilization-treated C57BL/6J mice. Food Sci Biotechnol 2024; 33:3617-3628. [PMID: 39493393 PMCID: PMC11525372 DOI: 10.1007/s10068-024-01606-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/04/2024] [Accepted: 05/16/2024] [Indexed: 11/05/2024] Open
Abstract
Muscle atrophy or muscle wasting, which is featured by reduced muscle function and mass, typically results from disuse, aging, and chronic diseases. The deer antler, which refers to the young and non-ossified antlers of various species of deer-related animals, is not fully calcified and comprises of densely growing hair. Here, we investigated whether Bacillus subtilis-fermented antler extract (FAE) inhibits immobilization-induced muscle atrophy in C57BL/6J mice. Oral administration of FAE increased grip strength, exercise performance, muscle mass, and volume in mice. FAE stimulated the phosphatidylinositol 3-kinase (PI3K)/Akt pathway, enhancing the mammalian target of rapamycin pathway for muscle synthesis. FAE phosphorylated Forkhead box O3 and downregulated muscle RING finger-1 and atrogin-1 for proteolysis. FAE inhibited the mRNA expression of tumor necrosis factor alpha and interleukin-6 through nuclear factor kappa B. Consequently, FAE attenuated muscle atrophy by regulating the PI3K/Akt pathway and inflammation.
Collapse
Affiliation(s)
- Jihee Yoo
- Department of Bioindustrial Engineering, Yonsei University, Seoul, Republic of Korea
| | - Changhee Kim
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Hyerin Lee
- Department of Bioindustrial Engineering, Yonsei University, Seoul, Republic of Korea
| | - Bong Soo Ko
- Research and Development Center, Biocare Corporation, Yangju, Republic of Korea
| | - Dong-Woo Lee
- Department of Bioindustrial Engineering, Yonsei University, Seoul, Republic of Korea
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jae-Kwan Hwang
- Department of Bioindustrial Engineering, Yonsei University, Seoul, Republic of Korea
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
5
|
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
|
6
|
Abate C, Giuffrè O, Amadeo A, Saija F, Cassone G, Foti C. Experimental and computational study on morin and its complexes with Mg 2+, Mn 2+, Zn 2+, and Al 3+: Coordination and antioxidant properties. J Inorg Biochem 2024; 258:112635. [PMID: 38852294 DOI: 10.1016/j.jinorgbio.2024.112635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/20/2024] [Accepted: 06/02/2024] [Indexed: 06/11/2024]
Abstract
Morin (MRN), an intriguing bioflavonol, has received increasing interest for its antioxidant properties, as have its metal complexes (Mz+-MRN). Understanding their antioxidant behavior is critical to assess their pharmaceutical, nutraceutical potential, and therapeutic impact in the design of advanced antioxidant drugs. To this end, knowing the speciation of different H+-MRN and Mz+-MRN is pivotal to understand and compare their antioxidant ability. In this work, the protonation constant values of MRN under physiological ionic strength and temperature conditions (I = 0.15 mol L-1 and t = 37 °C), determined by UV-vis spectrophotometric titrations, are introduced. Thus, a reliable speciation model on H+-MRN species in aqueous solution is presented, which exhibits five stable forms depending on pH, supplemented by quantum-mechanical calculations useful to determine the proton affinities of each functional group and corresponding deprotonation order. Furthermore, potentiometry and UV-vis spectrophotometry have been exploited to determine the thermodynamic interaction parameters of MRN with different metal cations (Mg2+, Mn2+, Zn2+, Al3+). The antioxidant ability of H+-MRN and Mz+-MRN has been evaluated by the 2,2'-diphenyl-1-benzopyran-4-one (DPPH) method, and the Zn2+-MRN system has proven to afford the most potent antioxidant effect. Ab initio molecular dynamics simulations of Mz+-MRN species at all possible chelation sites and under explicit water solvation allowed for the fine characterization not only of the metal chelation modalities of MRN in explicit water, but also of the role played by the local water environment around the metal cations. Those microscopic patterns reveal to be informative on the different antioxidant capabilities recorded experimentally.
Collapse
Affiliation(s)
- Chiara Abate
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, Università di Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy.
| | - Ottavia Giuffrè
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, Università di Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy; CNR-IPCF, Viale Ferdinando Stagno d'Alcontres 37, 98158 Messina, Italy
| | - Alessandro Amadeo
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, Università di Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy; Dipartimento di Chimica, Biologia e Biotecnologie, Università degli Studi di Perugia, 06123 Perugia, Italy
| | - Franz Saija
- CNR-IPCF, Viale Ferdinando Stagno d'Alcontres 37, 98158 Messina, Italy
| | - Giuseppe Cassone
- CNR-IPCF, Viale Ferdinando Stagno d'Alcontres 37, 98158 Messina, Italy.
| | - Claudia Foti
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, Università di Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| |
Collapse
|
7
|
Zhang Y, Li S, Huang Y, Song C, Chen W, Yang Y. Therapeutic Effect of Liquiritin Carbomer Gel on Topical Glucocorticoid-Induced Skin Inflammation in Mice. Pharmaceutics 2024; 16:1001. [PMID: 39204346 PMCID: PMC11359290 DOI: 10.3390/pharmaceutics16081001] [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: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 09/04/2024] Open
Abstract
Glucocorticoids are often used and highly effective anti-inflammatory medications, but prolonged topical application may alter the epidermis' normal structure and function, potentially resulting in a number of adverse effects. Topical glucocorticoid-induced skin inflammation is a dangerous condition that develops after topical glucocorticoid use. The patients become dependent on the medication and, even after the medication is stopped, the dermatitis symptoms recur, severely impairing their quality of life. Thus, the need to aggressively confront Topical glucocorticoid-induced skin inflammation is critical. Prior research has demonstrated that topical administration of licorice's flavonoid component liquiritin stimulates epidermal proliferation, which in turn enhances the creation of collagen and the healing of wounds. Therefore, the purpose of this work was to determine if topical use of liquiritin carbomer gel can treat glucocorticoid-induced changes in mice skin epidermal function, and the mechanisms involved. The findings demonstrated that, in the mice model of topical glucocorticoid-induced skin inflammation, liquiritin carbomer gel aided in the restoration of skin barrier function. These outcomes may have been caused by enhanced expression of the proteins Aquaporin 3, Keratin 10, and Claudin-1, as well as the restoration of epidermal hyaluronan content. In the meantime, liquiritin carbomer gel dramatically decreased the expression of TNF-α, IL-1β, IL-6, IFN-γ, and IgE in mice, according to ELISA tests. Furthermore, topical treatment of liquiritin carbomer gel boosted the expression of superoxide dismutase, catalase, and decreased malondialdehyde expression, potentially counteracting the detrimental effects of glucocorticoids on the epidermis. In summary, these findings imply that topical liquiritin carbomer gel can treat glucocorticoid-induced skin damage through various mechanisms of action.
Collapse
Affiliation(s)
- Yun Zhang
- School of Nursing, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Sijia Li
- School of Nursing, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yanfang Huang
- School of Nursing, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Congjing Song
- School of Nursing, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Weiqiang Chen
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yiling Yang
- School of Nursing, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
8
|
Salucci S, Burattini S, Versari I, Bavelloni A, Bavelloni F, Curzi D, Battistelli M, Gobbi P, Faenza I. Morpho-Functional Analyses Demonstrate That Tyrosol Rescues Dexamethasone-Induced Muscle Atrophy. J Funct Morphol Kinesiol 2024; 9:124. [PMID: 39051285 PMCID: PMC11270424 DOI: 10.3390/jfmk9030124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024] Open
Abstract
Prolonged exposure to high dosages of dexamethasone, which is a synthetic glucocorticoid and a well-known anti-inflammatory drug, may lead to an increase in reactive oxygen species production, contributing to muscle wasting. The prevention of muscle atrophy by ingestion of functional foods is an attractive issue. In the last decade, natural antioxidant compounds have been increasingly investigated as promising molecules able to counteract oxidative-stress-induced muscle atrophy. Recently, we have demonstrated the antioxidant properties of two main olive oil polyphenols also known for their anticancer and anti-inflammatory activities in different cell models. Here, the preventive effect of tyrosol on dexamethasone-induced muscle atrophy has been investigated by means of morpho-functional approaches in C2C12 myotubes. Dexamethasone-treated cells showed a reduced fiber size when compared to control ones. While long and confluent myotubes could be observed in control samples, those exposed to dexamethasone appeared as immature syncytia. Dysfunctional mitochondria and the accumulation of autophagic vacuoles contributed to myotube degeneration and death. Tyrosol administration before glucocorticoid treatment prevented muscle wasting and rescued mitochondrial and lysosomal functionality. These findings demonstrate that tyrosol attenuates dexamethasone-induced myotube damage, and encourage the use of this natural molecule in preclinical and clinical studies and in synergy with other functional foods or physical activity with the aim to prevent muscle atrophy.
Collapse
Affiliation(s)
- Sara Salucci
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, 40126 Bologna, Italy; (I.V.); (F.B.); (I.F.)
| | - Sabrina Burattini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (S.B.); (M.B.); (P.G.)
| | - Ilaria Versari
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, 40126 Bologna, Italy; (I.V.); (F.B.); (I.F.)
| | - Alberto Bavelloni
- Laboratory of Experimental Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Francesco Bavelloni
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, 40126 Bologna, Italy; (I.V.); (F.B.); (I.F.)
| | - Davide Curzi
- Department of Humanities, Movement, and Education Sciences, University “Niccolò Cusano”, 00166 Rome, Italy;
| | - Michela Battistelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (S.B.); (M.B.); (P.G.)
| | - Pietro Gobbi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (S.B.); (M.B.); (P.G.)
| | - Irene Faenza
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, 40126 Bologna, Italy; (I.V.); (F.B.); (I.F.)
| |
Collapse
|
9
|
Pan D, Yang L, Yang X, Xu D, Wang S, Gao H, Liu H, Xia H, Yang C, Lu Y, Sun J, Wang Y, Sun G. Potential nutritional strategies to prevent and reverse sarcopenia in aging process: Role of fish oil-derived ω-3 polyunsaturated fatty acids, wheat oligopeptide and their combined intervention. J Adv Res 2024; 57:77-91. [PMID: 37061218 PMCID: PMC10918331 DOI: 10.1016/j.jare.2023.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/24/2023] [Accepted: 04/10/2023] [Indexed: 04/17/2023] Open
Abstract
INTRODUCTION Nutritional support is potentially considered an essential step to prevent muscle loss and enhance physical function in older adults. OBJECTIVES This study aimed to assess the role of potential nutritional strategies, i.e., fish oil-derived ω-3 polyunsaturated fatty acids (PUFAs), wheat oligopeptide and their combined intervention, in preventing and reversing sarcopenia in aging process. METHODS One hundred 25-month-old Sprague-Dawley rats were randomly divided into 10 groups, and 10 newly purchased 6-month-old rats were included in young control group (n = 10). Fish oil (200, 400 or 800 mg/kg body weight), wheat oligopeptide (100, 200 or 400 mg/kg body weight), fish oil + wheat oligopeptide (800 + 100, 400 + 200 or 200 + 400 mg/kg body weight) or the equal volume of solvent were administered daily by gavage for 10 weeks. The effects of these interventions on natural aging rats were evaluated. RESULTS All intervention groups had a significant increase in muscle mass and grip strength and reduction in perirenal fat weight when compared to the aged control group (P < 0.05). The results of biochemical parameters, magnetic resonance imaging, proteomics and western blot suggested that the combination of wheat oligopeptide and fish oil-derived ω-3 PUFA, especially group WFM 2 (400 + 200 mg/kg body weight fish oil + wheat oligopeptide), was found to be more effective against aging-associated muscle loss than single intervention. Additionally, the interventions ameliorated fatty infiltration, muscle atrophy, and congestion in the intercellular matrix, and inflammatory cell infiltration in muscle tissue. The interventions also improved oxidative stress, anabolism, hormone levels, and inflammatory levels of skeletal muscle. CONCLUSIONS The combination of fish oil-derived ω-3 PUFA and wheat oligopeptide was found to be a promising nutritional support to prevent and reverse sarcopenia. The potential mechanism involved the promotion of protein synthesis and muscle regeneration, as well as the enhancement of muscle strength.
Collapse
Affiliation(s)
- Da Pan
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, 210009 Nanjing, PR China
| | - Ligang Yang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, 210009 Nanjing, PR China
| | - Xian Yang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, 210009 Nanjing, PR China
| | - Dengfeng Xu
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, 210009 Nanjing, PR China
| | - Shaokang Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, 210009 Nanjing, PR China; School of Medicine, Xizang Minzu University, 712082 Xianyang, PR China
| | - Han Gao
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Hechun Liu
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, 210009 Nanjing, PR China; Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, 210009 Nanjing, PR China
| | - Hui Xia
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, 210009 Nanjing, PR China
| | - Chao Yang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, 210009 Nanjing, PR China; Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, PR China
| | - Yifei Lu
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, 210009 Nanjing, PR China
| | - Jihan Sun
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, 210009 Nanjing, PR China
| | - Yuanyuan Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, 210009 Nanjing, PR China
| | - Guiju Sun
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, 210009 Nanjing, PR China.
| |
Collapse
|
10
|
Bahramzadeh A, Samavarchi Tehrani S, Goodarzi G, Seyyedebrahimi S, Meshkani R. Combination therapy of metformin and morin attenuates insulin resistance, inflammation, and oxidative stress in skeletal muscle of high-fat diet-fed mice. Phytother Res 2024; 38:912-924. [PMID: 38091524 DOI: 10.1002/ptr.8086] [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: 06/21/2023] [Revised: 10/25/2023] [Accepted: 11/26/2023] [Indexed: 02/15/2024]
Abstract
Lipid accumulation, inflammation, and oxidative stress are the most important causes of muscle insulin resistance. The aim of this study was to investigate the single and combined treatment effects of metformin (MET) and morin (MOR) on lipid accumulation, inflammation, and oxidative stress in the skeletal muscle of mice fed a high-fat diet. The mice were supplemented with MET (230 mg/kg diet), MOR (100 mg/kg diet), and MET + MOR for 9 weeks. Our results revealed that single treatment with MET or MOR, and with a stronger effect of MET + MOR combined treatment, reduced body weight gain, improved glucose intolerance and enhanced Akt phosphorylation in the muscle tissue. In addition, plasma and muscle triglyceride levels were decreased after treatment with MET and MOR. The expression of genes involved in macrophage infiltration and polarization and pro-inflammatory cytokines showed that MET + MOR combined treatment, significantly reduced inflammation in the muscle. Furthermore, combined treatment of MET + MOR with greater efficacy than the single treatment improved several oxidative stress markers in the muscle. Importantly, combined treatment of MET and MOR could increase the expression of nuclear factor erythroid 2-related factor 2, the master regulator of the antioxidant response. These findings suggest that combination of MET with MOR might ameliorate insulin resistance, inflammation, and oxidative stress in the skeletal muscle of mice fed high-fat diet.
Collapse
Affiliation(s)
- Arash Bahramzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sadra Samavarchi Tehrani
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Science, Tehran, Iran
| | - Golnaz Goodarzi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pathobiology and Laboratory Science, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - ShadiSadat Seyyedebrahimi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Kim YS, Han JH, Lim CH, Fang XQ, Jang HS, Lee SY, Yim WJ, Lim JH. Effects of Fermented Polygonum cuspidatum on the Skeletal Muscle Functions. Nutrients 2024; 16:305. [PMID: 38276543 PMCID: PMC10818974 DOI: 10.3390/nu16020305] [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: 12/06/2023] [Revised: 01/01/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
Plant extract fermentation is widely employed to enhance the nutritional and pharmaceutical value of functional foods. Polygonum cuspidatum (Pc) contains flavonoids, anthraquinones, and stilbenes, imparting protective effects against inflammatory diseases, cancer, diabetes, and cardiovascular diseases. However, the effects of fermented Pc on skeletal muscle strength remain unexplored. In this study, we generated fermented Pc using a complex of microorganisms containing Lactobacillus spp. (McPc) and assessed its effects on muscle strength and motor function in mice. Compared to unfermented Pc water extract, elevated levels of emodin and resveratrol were noted in McPc. This was identified and quantified using UPLC-QTOF/MS and HPLC techniques. Gene expression profiling through RNA-seq and quantitative RT-PCR revealed that McPc administration upregulated the expression of genes associated with antioxidants, glycolysis, oxidative phosphorylation, fatty acid oxidation, and mitochondrial biogenesis in cultured C2C12 myotubes and the gastrocnemius muscle in mice. McPc significantly improved skeletal muscle strength, motor coordination, and traction force in mice subjected to sciatic neurectomy and high-fat diet (HFD). McPc administration exhibited more pronounced improvement of obesity, hyperglycemia, fatty liver, and hyperlipidemia in HFD mice compared to control group. These findings support the notion that emodin and resveratrol-enriched McPc may offer health benefits for addressing skeletal muscle weakness.
Collapse
Affiliation(s)
- Young-Seon Kim
- Department of Medicinal Biosciences, College of Biomedical & Health Science, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea; (Y.-S.K.); (C.-H.L.); (X.-Q.F.)
- BK21 Program, Department of Applied Life Science, Graduate School, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
- Jung-Ang Microbe Research Institute (JM), 398, Jikji-daero, Heungdeok-gu, Cheongju 28576, Chungbuk, Republic of Korea; (J.-H.H.); (H.-S.J.); (S.-Y.L.); (W.-J.Y.)
| | - Ji-Hye Han
- Jung-Ang Microbe Research Institute (JM), 398, Jikji-daero, Heungdeok-gu, Cheongju 28576, Chungbuk, Republic of Korea; (J.-H.H.); (H.-S.J.); (S.-Y.L.); (W.-J.Y.)
| | - Chang-Hoon Lim
- Department of Medicinal Biosciences, College of Biomedical & Health Science, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea; (Y.-S.K.); (C.-H.L.); (X.-Q.F.)
- BK21 Program, Department of Applied Life Science, Graduate School, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
- Center for Metabolic Diseases, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
| | - Xue-Quan Fang
- Department of Medicinal Biosciences, College of Biomedical & Health Science, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea; (Y.-S.K.); (C.-H.L.); (X.-Q.F.)
- BK21 Program, Department of Applied Life Science, Graduate School, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
- Center for Metabolic Diseases, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
| | - Hyeock-Soon Jang
- Jung-Ang Microbe Research Institute (JM), 398, Jikji-daero, Heungdeok-gu, Cheongju 28576, Chungbuk, Republic of Korea; (J.-H.H.); (H.-S.J.); (S.-Y.L.); (W.-J.Y.)
| | - Sang-Yun Lee
- Jung-Ang Microbe Research Institute (JM), 398, Jikji-daero, Heungdeok-gu, Cheongju 28576, Chungbuk, Republic of Korea; (J.-H.H.); (H.-S.J.); (S.-Y.L.); (W.-J.Y.)
| | - Woo-Jong Yim
- Jung-Ang Microbe Research Institute (JM), 398, Jikji-daero, Heungdeok-gu, Cheongju 28576, Chungbuk, Republic of Korea; (J.-H.H.); (H.-S.J.); (S.-Y.L.); (W.-J.Y.)
| | - Ji-Hong Lim
- Department of Medicinal Biosciences, College of Biomedical & Health Science, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea; (Y.-S.K.); (C.-H.L.); (X.-Q.F.)
- BK21 Program, Department of Applied Life Science, Graduate School, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
- Center for Metabolic Diseases, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
| |
Collapse
|
12
|
Kim A, Kim YR, Park SM, Lee H, Park M, Yi JM, Cha S, Kim NS. Jakyak-gamcho-tang, a decoction of Paeoniae Radix and Glycyrrhizae Radix et Rhizoma, ameliorates dexamethasone-induced muscle atrophy and muscle dysfunction. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155057. [PMID: 37984121 DOI: 10.1016/j.phymed.2023.155057] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/17/2023] [Accepted: 08/29/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Although chronic treatment with glucocorticoids, such as dexamethasone, is frequently associated with muscle atrophy, effective and safe therapeutics for treating muscle atrophy remain elusive. Jakyak-gamcho-tang (JGT), a decoction of Paeoniae Radix and Glycyrrhizae Radix et Rhizoma, has long been used to relieve muscle tension and control muscle cramp-related pain. However, the effects of JGT on glucocorticoid-induced muscle atrophy are yet to be comprehensively clarified. PURPOSE The objective of the current study was to validate the protective effect of JGT in dexamethasone-induced muscle atrophy models and elucidate its underlying mechanism through integrated in silico - in vitro - in vivo studies. STUDY DESIGN AND METHODS Differential gene expression was preliminarily analyzed using the RNA-seq data to determine the effects of JGT on C2C12 myotubes. The protective effects of JGT were further validated in dexamethasone-treated C2C12 myotubes by assessing cell viability, myotube integrity, and mitochondrial function or in C57BL/6 N male mice with dexamethasone-induced muscle atrophy by evaluating muscle mass and physical performance. Transcriptomic pathway analysis was also performed to elucidate the underlying mechanism. RESULTS Based on preliminary gene set enrichment analysis using the RNA-seq data, JGT regulated various pathways related to muscle differentiation and regeneration. Dexamethasone-treated C2C12 myotubes and muscle tissues of atrophic mice displayed substantial muscle protein degradation and muscle loss, respectively, which was efficiently alleviated by JGT treatment. Importantly, JGT-mediated protective effects were associated with observations such as preservation of mitochondrial function, upregulation of myogenic signaling pathways, including protein kinase B/mammalian target of rapamycin/forkhead box O3, inhibition of ubiquitin-mediated muscle protein breakdown, and downregulation of inflammatory and apoptotic pathways induced by dexamethasone. CONCLUSION To the best of our knowledge, this is the first report to demonstrate that JGT could be a potential pharmaceutical candidate to prevent muscle atrophy induced by chronic glucocorticoid treatment, highlighting its known effects for relieving muscle spasms and pain. Moreover, transcriptomic pathway analysis can be employed as an efficient in silico tool to predict novel pharmacological candidates and elucidate molecular mechanisms underlying the effects of herbal medications comprising diverse biologically active ingredients.
Collapse
Affiliation(s)
- Aeyung Kim
- KM Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea
| | - Yu Ri Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Sang-Min Park
- KM Data Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea; College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Haeseung Lee
- KM Data Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea; College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Musun Park
- KM Data Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Jin-Mu Yi
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Seongwon Cha
- KM Data Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| | - No Soo Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| |
Collapse
|
13
|
Jiang Y, Qian Y, Hong H, Gao X, Liu W, Jin Q, Chen M, Jin Z, Liu Q, Wei Z. Morin protects chicks with T-2 toxin poisoning by decreasing heterophil extracellular traps, oxidative stress and inflammatory response. Br Poult Sci 2023; 64:614-624. [PMID: 37334824 DOI: 10.1080/00071668.2023.2226083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/16/2023] [Indexed: 06/21/2023]
Abstract
1. Fusarium tritici widely exists in a variety of grain feeds. The T-2 toxin is the main hazardous component produced by Fusarium tritici, making a serious hazard to poultry industry. Morin, belonging to the flavonoid family, can be extracted from mulberry plants and possesses anticancer, antioxidant and anti-inflammatory compounds, but whether morin protects chicks with T-2 toxin poisoning remains unclear. This experiment firstly established a chick model of T-2 toxin poisoning and then investigated the protective effects and mechanism of morin against T-2 toxin in chicks.2. The function of liver and kidney was measured by corresponding alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), blood urea nitrogen (BUN), creatinine (Cre) and uric acid (UA) kits. Histopathological changes were observed by haematoxylin-eosin staining. The status of oxidative stress was measured by MDA, SOD, CAT, GSH and GSH-PX kits. The mRNA levels of TNF-α, COX-2, IL-1β, IL-6, caspase-1, caspase-3 and caspase-11 were measured by quantitative real-time PCR. Heterophil extracellular trap (HET) release was analysed by immunofluorescence and fluorescence microplate.3. The model with T-2 toxin poisoning in chicks was successfully established. Morin significantly decreased T-2 toxin-induced ALT, AST, ALP, BUN, Cre and UA, and improved T-2 toxin-induced liver cell rupture, liver cord disorder and kidney interstitial oedema. Oxidative stress analysis showed that morin ameliorated T-2 toxin-induced damage by reducing malondialdehyde (MDA), increasing superoxide dismutase (SOD), catalase (CAT), glutathione (GSH) and glutathione peroxidase (GSH-PX). The qRT-PCR analysis showed that morin reduced T-2 toxin-induced mRNA expressions of TNF-α, COX-2, IL-1β, IL-6, caspase-1, caspase-3 and caspase-11. Moreover, morin significantly reduced the release of T-2 toxin-induced HET in vitro and in vivo.4. Morin can protect chicks from T-2 toxin poisoning by decreasing HETs, oxidative stress and inflammatory responses, which make it a useful compound against T-2 toxin poisoning in poultry feed.
Collapse
Affiliation(s)
- Y Jiang
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - Y Qian
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - H Hong
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - X Gao
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - W Liu
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - Q Jin
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - M Chen
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - Z Jin
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - Q Liu
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - Z Wei
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong Province, People's Republic of China
- College of Veterinary Medicine, Southwest University, Chongqing, China
| |
Collapse
|
14
|
Yoshioka Y, Oishi S, Onoda K, Shibata K, Miyoshi N. Diosgenin prevents dexamethasone-induced myotube atrophy in C2C12 cells. Arch Biochem Biophys 2023; 747:109759. [PMID: 37722527 DOI: 10.1016/j.abb.2023.109759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023]
Abstract
Several pathophysiological abnormalities, including a sedentary lifestyle, chronic diseases, and oxidative stress, can contribute to muscle atrophy triggered by an imbalance in muscle protein synthesis and degradation. Resolving muscle atrophy is a critical issue as it can reduce the quality of life. Here, one of the promising functional food factors, diosgenin (a steroidal sapogenin) showed strong preventive activities against dexamethasone (Dex)-induced muscle atrophy, as determined by the expression levels and morphology of the myosin heavy chain in C2C12 myotubes. Diosgenin inhibited protein expressions of Dex-induced skeletal muscle-specific ubiquitin ligase, including muscle RING finger 1 (MuRF1) and casitas B-lineage lymphoma protooncogene b (Cbl-b) but not atrogin-1. Diosgenin ameliorated Dex-induced declines of Akt phosphorylation at Ser473 and FoxO3a phosphorylation at Ser253, which probably at least partially contributed to the suppression of MuRF1, Cbl-b, and atrogin-1 gene expression. Additionally, diosgenin inhibited Dex-induced nuclear translocation of the glucocorticoid receptor (GR), diosgenin therefore may competitively inhibit the interaction between Dex and GR. These findings suggest that diosgenin is an effective functional food for preventing glucocorticoid-induced skeletal muscle atrophy.
Collapse
Affiliation(s)
- Yasukiyo Yoshioka
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Shiori Oishi
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Keita Onoda
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Katsumi Shibata
- Faculty of Clinical Nutrition and Dietetics, Konan Women's University, Kobe, Hyogo, 685-0001, Japan
| | - Noriyuki Miyoshi
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan.
| |
Collapse
|
15
|
Zhang H, Qi G, Wang K, Yang J, Shen Y, Yang X, Chen X, Yao X, Gu X, Qi L, Zhou C, Sun H. Oxidative stress: roles in skeletal muscle atrophy. Biochem Pharmacol 2023:115664. [PMID: 37331636 DOI: 10.1016/j.bcp.2023.115664] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/20/2023]
Abstract
Oxidative stress, inflammation, mitochondrial dysfunction, reduced protein synthesis, and increased proteolysis are all critical factors in the process of muscle atrophy. In particular, oxidative stress is the key factor that triggers skeletal muscle atrophy. It is activated in the early stages of muscle atrophy and can be regulated by various factors. The mechanisms of oxidative stress in the development of muscle atrophy have not been completely elucidated. This review provides an overview of the sources of oxidative stress in skeletal muscle and the correlation of oxidative stress with inflammation, mitochondrial dysfunction, autophagy, protein synthesis, proteolysis, and muscle regeneration in muscle atrophy. Additionally, the role of oxidative stress in skeletal muscle atrophy caused by several pathological conditions, including denervation, unloading, chronic inflammatory diseases (diabetes mellitus, chronic kidney disease, chronic heart failure, and chronic obstructive pulmonary disease), sarcopenia, hereditary neuromuscular diseases (spinal muscular atrophy, amyotrophic lateral sclerosis, and Duchenne muscular dystrophy), and cancer cachexia, have been discussed. Finally, this review proposes the alleviation oxidative stress using antioxidants, Chinese herbal extracts, stem cell and extracellular vesicles as a promising therapeutic strategy for muscle atrophy. This review will aid in the development of novel therapeutic strategies and drugs for muscle atrophy.
Collapse
Affiliation(s)
- Han Zhang
- 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, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, PR China
| | - Guangdong Qi
- Department of Endocrinology, Binhai County People's Hospital, Yancheng, Jiangsu Province, 224500, PR 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, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, PR China
| | - Jiawen Yang
- Department of Clinical Medicine, Medical College, Nantong University, Nantong 226001, 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, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, PR 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, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, PR China
| | - Xin Chen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, PR China
| | - Xinlei Yao
- 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, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, PR China
| | - Xiaosong Gu
- 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, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, PR China
| | - Lei Qi
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, PR China.
| | - Chun Zhou
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, PR 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, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, PR China; Research and Development Center for E-Learning, Ministry of Education, Beijing 100816, PR China.
| |
Collapse
|
16
|
Gatti M, Dittlau KS, Beretti F, Yedigaryan L, Zavatti M, Cortelli P, Palumbo C, Bertucci E, Van Den Bosch L, Sampaolesi M, Maraldi T. Human Neuromuscular Junction on a Chip: Impact of Amniotic Fluid Stem Cell Extracellular Vesicles on Muscle Atrophy and NMJ Integrity. Int J Mol Sci 2023; 24:ijms24054944. [PMID: 36902375 PMCID: PMC10003237 DOI: 10.3390/ijms24054944] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Neuromuscular junctions (NMJs) are specialized synapses, crucial for the communication between spinal motor neurons (MNs) and skeletal muscle. NMJs become vulnerable in degenerative diseases, such as muscle atrophy, where the crosstalk between the different cell populations fails, and the regenerative ability of the entire tissue is hampered. How skeletal muscle sends retrograde signals to MNs through NMJs represents an intriguing field of research, and the role of oxidative stress and its sources remain poorly understood. Recent works demonstrate the myofiber regeneration potential of stem cells, including amniotic fluid stem cells (AFSC), and secreted extracellular vesicles (EVs) as cell-free therapy. To study NMJ perturbations during muscle atrophy, we generated an MN/myotube co-culture system through XonaTM microfluidic devices, and muscle atrophy was induced in vitro by Dexamethasone (Dexa). After atrophy induction, we treated muscle and MN compartments with AFSC-derived EVs (AFSC-EVs) to investigate their regenerative and anti-oxidative potential in counteracting NMJ alterations. We found that the presence of EVs reduced morphological and functional in vitro defects induced by Dexa. Interestingly, oxidative stress, occurring in atrophic myotubes and thus involving neurites as well, was prevented by EV treatment. Here, we provided and validated a fluidically isolated system represented by microfluidic devices for studying human MN and myotube interactions in healthy and Dexa-induced atrophic conditions-allowing the isolation of subcellular compartments for region-specific analyses-and demonstrated the efficacy of AFSC-EVs in counteracting NMJ perturbations.
Collapse
Affiliation(s)
- Martina Gatti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Katarina Stoklund Dittlau
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute, KU Leuven—University of Leuven, 3000 Leuven, Belgium
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Francesca Beretti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Correspondence:
| | - Laura Yedigaryan
- Department of Development and Regeneration, Stem Cell and Developmental Biology, KU Leuven—University of Leuven, 3000 Leuven, Belgium
| | - Manuela Zavatti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Pietro Cortelli
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, 40127 Bologna, Italy
| | - Carla Palumbo
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Emma Bertucci
- Department of Medical and Surgical Sciences for Mothers, Children and Adults, Azienda Ospedaliero Universitaria Policlinico, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute, KU Leuven—University of Leuven, 3000 Leuven, Belgium
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Maurilio Sampaolesi
- Department of Development and Regeneration, Stem Cell and Developmental Biology, KU Leuven—University of Leuven, 3000 Leuven, Belgium
- Histology and Medical Embryology Unit, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, 00185 Rome, Italy
| | - Tullia Maraldi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
17
|
Kim JY, Kim HM, Kim JH, Guo S, Lee DH, Lim GM, Kim W, Kim CY. Salvia plebeia R.Br. and Rosmarinic Acid Attenuate Dexamethasone-Induced Muscle Atrophy in C2C12 Myotubes. Int J Mol Sci 2023; 24:ijms24031876. [PMID: 36768200 PMCID: PMC9915874 DOI: 10.3390/ijms24031876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
Skeletal muscle atrophy occurs when protein degradation exceeds protein synthesis and is associated with increased circulating glucocorticoid levels. Salvia plebeia R.Br. (SPR) has been used as herbal remedy for a variety of inflammatory diseases and has various biological actions such as antioxidant and anti-inflammatory activities. However, there are no reports on the effects of SPR and its bioactive components on muscle atrophy. Herein, we investigated the anti-atrophic effect of SPR and rosmarinic acid (RosA), a major compound of SPR, on dexamethasone (DEX)-induced skeletal muscle atrophy in C2C12 myotubes. Myotubes were treated with 10 μM DEX in the presence or absence of SPR or RosA at different concentrations for 24 h and subjected to immunocytochemistry, western blot, and measurements of ROS and ATP levels. SPR and RosA increased viability and inhibited protein degradation in DEX-treated C2C12 myotubes. In addition, RosA promoted the Akt/p70S6K/mTOR pathway and reduced ROS production, and apoptosis. Furthermore, the treatment of RosA significantly recovered SOD activity, autophagy activity, mitochondrial contents, and APT levels in DEX-treated myotubes. These findings suggest that SPR and RosA may provide protective effects against DEX-induced muscle atrophy and have promising potential as a nutraceutical remedy for the treatment of muscle weakness and atrophy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wondong Kim
- Correspondence: (W.K.); (C.Y.K.); Tel.: +82-31-400-5817 (W.K.); +82-31-400-5809 (C.Y.K.); Fax: +82-31-400-5958 (C.Y.K.)
| | - Chul Young Kim
- Correspondence: (W.K.); (C.Y.K.); Tel.: +82-31-400-5817 (W.K.); +82-31-400-5809 (C.Y.K.); Fax: +82-31-400-5958 (C.Y.K.)
| |
Collapse
|
18
|
Edwards SJ, Carter S, Nicholson T, Allen SL, Morgan PT, Jones SW, Rendeiro C, Breen L. (-)-Epicatechin and its colonic metabolite hippuric acid protect against dexamethasone-induced atrophy in skeletal muscle cells. J Nutr Biochem 2022; 110:109150. [PMID: 36049668 DOI: 10.1016/j.jnutbio.2022.109150] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 05/13/2022] [Accepted: 08/10/2022] [Indexed: 01/13/2023]
Abstract
Cocoa flavanols have been shown to improve muscle function and may offer a novel approach to protect against muscle atrophy. Hippuric acid (HA) is a colonic metabolite of (-)-epicatechin (EPI), the primary bioactive compound of cocoa, and may be responsible for the associations between cocoa supplementation and muscle metabolic alterations. Accordingly, we investigated the effects of EPI and HA upon skeletal muscle morphology and metabolism within an in vitro model of muscle atrophy. Under atrophy-like conditions (24h 100μM dexamethasone (DEX)), C2C12 myotube diameter was significantly greater following co-incubation with either 25μM HA (11.19±0.39μm) or 25μM EPI (11.01±0.21μm) compared to the vehicle control (VC; 7.61±0.16μm, both P < .001). In basal and leucine-stimulated states, there was a significant reduction in myotube protein synthesis (MPS) rates following DEX treatment in VC (P = .024). Interestingly, co-incubation with EPI or HA abrogated the DEX-induced reductions in MPS rates, whereas no significant differences versus control treated myotubes (CTL) were noted. Furthermore, co-incubation with EPI or HA partially attenuated the increase in proteolysis seen in DEX-treated cells, preserving LC3 α/β II:I and caspase-3 protein expression in atrophy-like conditions. The protein content of PGC1α, ACC, and TFAM (regulators of mitochondrial function) were significantly lower in DEX-treated versus. CTL cells (all P < .050). However, co-incubation with EPI or HA was unable to prevent these DEX-induced alterations. For the first time we demonstrate that EPI and HA exert anti-atrophic effects on C2C12 myotubes, providing novel insight into the association between flavanol supplementation and favourable effects on muscle health.
Collapse
Affiliation(s)
- Sophie J Edwards
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - Steven Carter
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK; Department for Health, University of Bath, Bath, UK
| | - Thomas Nicholson
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Sophie Louise Allen
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK; National Institute for Health Research, Birmingham Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Paul T Morgan
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - Simon Wyn Jones
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Catarina Rendeiro
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - Leigh Breen
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK; National Institute for Health Research, Birmingham Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK; MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Birmingham, UK.
| |
Collapse
|
19
|
Kim JY, Kim HM, Kim JH, Lee JH, Zhang K, Guo S, Lee DH, Gao EM, Son RH, Kim SM, Kim CY. Preventive effects of the butanol fraction of Justicia procumbens L. against dexamethasone-induced muscle atrophy in C2C12 myotubes. Heliyon 2022; 8:e11597. [DOI: 10.1016/j.heliyon.2022.e11597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/06/2022] [Accepted: 11/10/2022] [Indexed: 11/23/2022] Open
|
20
|
Lee JA, Lee SH, Shin MR, Park HJ, Roh SS. Gardeniae Fructus Extract Alleviates Dexamethasone-Induced Muscle Atrophy in Mice. J Med Food 2022; 25:882-891. [PMID: 36084316 DOI: 10.1089/jmf.2022.k.0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Muscle atrophy (MA) is a case in which protein degeneration occurs excessively due to an imbalance between protein synthesis and breakdown, and is characterized by decreased muscle mass and weakened muscle strength. Despite mounting concern about MA, the number of patients with MA is increasing every year. The aim of the present study was to assess the impact of Gardeniae Fructus (GF) hot water extract on dexamethasone (DEX)-induced MA in mice. C57BL/6N mice were grouped (n = 8) as follows: Normal mice (Normal), MA mice were treated with distilled water (Control), MA mice were treated with GF 100 mg/kg (GF100), MA mice were treated with GF 200 mg/kg (GF200). For 10 days, DEX (25 mg/kg body weight, i.p.) injection was used to induce MA, and GF was administered. GF treatment restored the muscle weight decreased due to MA, and in particular, the weights of EDL+TA and Sol were significantly increased in the GF200 group. Also, it was confirmed that the swimming time was improved in the GF200 group. In addition, the expression of NADPH oxidase related to oxidative stress was significantly reduced, and protective (insulin-like growth factor I/phosphoinositide 3-kinase/protein kinase B pathway) and catabolic (AMP-activated kinase [AMPK]/sirtuin 1 [SIRT1]/proliferator-activated receptor-gamma coactivator-1α (PGC-1α)-forkhead box O (FOXO) pathway) pathways were significantly modulated. These results demonstrate that GF regulates muscle protein synthesis and catabolic pathways, and in particular, it is judged to improve MA by regulating the proteolytic AMPK/SIRT1/PGC-1α-FOXO pathway.
Collapse
Affiliation(s)
- Jin A Lee
- Department of Herbology, College of Korean Medicine, Daegu Haany University, Daegu, Korea
| | - Se Hui Lee
- Department of Herbology, College of Korean Medicine, Daegu Haany University, Daegu, Korea
| | - Mi-Rae Shin
- Department of Herbology, College of Korean Medicine, Daegu Haany University, Daegu, Korea
| | - Hae-Jin Park
- DHU Bio Convergence Testing Center, Gyeongsan, Korea
| | - Seong-Soo Roh
- Department of Herbology, College of Korean Medicine, Daegu Haany University, Daegu, Korea
| |
Collapse
|
21
|
Ulla A, Ozaki K, Rahman MM, Nakao R, Uchida T, Maru I, Mawatari K, Fukawa T, Kanayama HO, Sakakibara I, Hirasaka K, Nikawa T. Morin improves dexamethasone-induced muscle atrophy by modulating atrophy-related genes and oxidative stress in female mice. Biosci Biotechnol Biochem 2022; 86:1448-1458. [PMID: 35977398 DOI: 10.1093/bbb/zbac140] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/08/2022] [Indexed: 11/12/2022]
Abstract
This study investigated the effect of morin, a flavonoid, on dexamethasone-induced muscle atrophy in C57BL/6J female mice. Dexamethasone (10 mg/kg body weight) for 10 days significantly reduced body weight, gastrocnemius and tibialis anterior muscle mass, and muscle protein in mice. Dexamethasone significantly upregulated muscle atrophy-associated ubiquitin ligases, including atrogin-1 and MuRF-1, and the upstream transcription factors FoxO3a and Klf15. Additionally, dexamethasone significantly induced the expression of oxidative stress-sensitive ubiquitin ligase Cbl-b and the accumulation of the oxidative stress markers malondialdehyde and advanced protein oxidation products in both the plasma and skeletal muscle samples. Intriguingly, morin treatment (20 mg/kg body weight) for 17 days effectively attenuated the loss of muscle mass and muscle protein and suppressed the expression of ubiquitin ligases while reducing the expression of upstream transcriptional factors. Therefore, morin might act as a potential therapeutic agent to attenuate muscle atrophy by modulating atrophy inducing genes and preventing oxidative stress.
Collapse
Affiliation(s)
- Anayt Ulla
- Department of Nutritional Physiology, Tokushima University Graduate School, Tokushima, Japan
| | - Kanae Ozaki
- Bizen Chemical Co. Ltd., Okayama, 709-0716, Japan
| | - Md Mizanur Rahman
- Department of Nutritional Physiology, Tokushima University Graduate School, Tokushima, Japan
| | - Reiko Nakao
- Department of Nutritional Physiology, Tokushima University Graduate School, Tokushima, Japan
| | - Takayuki Uchida
- Department of Nutritional Physiology, Tokushima University Graduate School, Tokushima, Japan
| | - Isafumi Maru
- Bizen Chemical Co. Ltd., Okayama, 709-0716, Japan
| | - Kazuaki Mawatari
- Department of Preventive Environment and Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Tomoya Fukawa
- Department of Urology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Hiro-Omi Kanayama
- Department of Urology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Iori Sakakibara
- Department of Nutritional Physiology, Tokushima University Graduate School, Tokushima, Japan
| | - Katsuya Hirasaka
- Organization for Marine Science and Technology, Nagasaki University, Nagasaki, Japan
| | - Takeshi Nikawa
- Department of Nutritional Physiology, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
22
|
Adel M, Elsayed HRH, El-Nablaway M, Hamed S, Eladl A, Fouad S, El Nashar EM, Al-Otaibi ML, Rabei MR. Targeting Hydrogen Sulfide Modulates Dexamethasone-Induced Muscle Atrophy and Microvascular Rarefaction, through Inhibition of NOX4 and Induction of MGF, M2 Macrophages and Endothelial Progenitors. Cells 2022; 11:cells11162500. [PMID: 36010575 PMCID: PMC9406793 DOI: 10.3390/cells11162500] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/06/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Long-term use of Glucocorticoids produces skeletal muscle atrophy and microvascular rarefaction. Hydrogen sulfide (H2S) has a potential role in skeletal muscle regeneration. However, the mechanisms still need to be elucidated. This is the first study to explore the effect of Sodium hydrosulfide (NaHS) H2S donor, against Dexamethasone (Dex)-induced soleus muscle atrophy and microvascular rarefaction and on muscle endothelial progenitors and M2 macrophages. Rats received either; saline, Dex (0.6 mg/Kg/day), Dex + NaHS (5 mg/Kg/day), or Dex + Aminooxyacetic acid (AOAA), a blocker of H2S (10 mg/Kg/day) for two weeks. The soleus muscle was examined for contractile properties. mRNA expression for Myostatin, Mechano-growth factor (MGF) and NADPH oxidase (NOX4), HE staining, and immunohistochemical staining for caspase-3, CD34 (Endothelial progenitor marker), vascular endothelial growth factor (VEGF), CD31 (endothelial marker), and CD163 (M2 macrophage marker) was performed. NaHS could improve the contractile properties and decrease oxidative stress, muscle atrophy, and the expression of NOX4, caspase-3, Myostatin, VEGF, and CD31 and could increase the capillary density and expression of MGF with a significant increase in expression of CD34 and CD163 as compared to Dex group. However, AOAA worsened the studied parameters. Therefore, H2S can be a promising target to attenuate muscle atrophy and microvascular rarefaction.
Collapse
Affiliation(s)
- Mohamed Adel
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Hassan Reda Hassan Elsayed
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
- Department of Anatomy, Faculty of Physical therapy, Horus University, New Damietta 34517, Egypt
- Correspondence: ; Tel.: +20-122-9310-701
| | - Mohammad El-Nablaway
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
- Department of Medical Biochemistry, College of Medicine, Almaarefa University, Riyad 71666, Saudi Arabia
| | - Shereen Hamed
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Amira Eladl
- Department of Pharmacology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Samah Fouad
- Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Eman Mohamad El Nashar
- Department of Anatomy, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha 13511, Egypt
| | - Mohammed Lafi Al-Otaibi
- Department of Orthopedics, College Medicine, King Khalid University, Abha 61421, Saudi Arabia
| | - Mohammed R. Rabei
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
- Department of Physiology, Faculty of Medicine, King Salman International University, El Tor 46511, Egypt
| |
Collapse
|
23
|
Leonurine Protects Bone Mesenchymal Stem Cells from Oxidative Stress by Activating Mitophagy through PI3K/Akt/mTOR Pathway. Cells 2022; 11:cells11111724. [PMID: 35681421 PMCID: PMC9179429 DOI: 10.3390/cells11111724] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 05/08/2022] [Accepted: 05/20/2022] [Indexed: 02/06/2023] Open
Abstract
Osteoporosis bears an imbalance between bone formation and resorption, which is strongly related to oxidative stress. The function of leonurine on bone marrow-derived mesenchymal stem cells (BMSCs) under oxidative stress is still unclear. Therefore, this study was aimed at identifying the protective effect of leonurine on H2O2 stimulated rat BMSCs. We found that leonurine can alleviate cell apoptosis and promote the differentiation ability of rat BMSCs induced by oxidative stress at an appropriate concentration at 10 μM. Meanwhile, the intracellular ROS level and the level of the COX2 and NOX4 mRNA decreased after leonurine treatment in vitro. The ATP level and mitochondrial membrane potential were upregulated after leonurine treatment. The protein level of PINK1 and Parkin showed the same trend. The mitophage in rat BMSCs blocked by 3-MA was partially rescued by leonurine. Bioinformatics analysis and leonurine-protein coupling provides a strong direct combination between leonurine and the PI3K protein at the position of Asp841, Glu880, Val882. In conclusion, leonurine protects the proliferation and differentiation of BMSCs from oxidative stress by activating mitophagy, which depends on the PI3K/Akt/mTOR pathway. The results showed that leonurine may have potential usage in osteoporosis and bone defect repair in osteoporosis patients.
Collapse
|
24
|
Ahn J, Kim J, Jeon JS, Jang YJ. A Microfluidic Stretch System Upregulates Resistance Exercise-Related Pathway. BIOCHIP JOURNAL 2022. [DOI: 10.1007/s13206-022-00051-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
25
|
Mahmoud MF, Ali N, Mostafa I, Hasan RA, Sobeh M. Coriander Oil Reverses Dexamethasone-Induced Insulin Resistance in Rats. Antioxidants (Basel) 2022; 11:441. [PMID: 35326092 PMCID: PMC8944706 DOI: 10.3390/antiox11030441] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/16/2022] [Accepted: 02/19/2022] [Indexed: 02/04/2023] Open
Abstract
In the present study, we aimed to investigate the effect of coriander oil on dexamethasone-induced insulin resistance in rats and characterize its chemical composition using gas chromatography-mass spectrometry (GC-MS). Rats were divided into five groups (n = 6): Normal control, insulin resistance (IR) control, IR + metformin (50 mg/kg/day, PO, Per Oral), IR + coriander oil low dose (0.5 mL/kg, PO), and IR + coriander oil high dose (1 mL/kg, PO). IR groups were injected with a dose of 10 mg/kg dexamethasone subcutaneously for four consecutive days. All groups received either vehicle or drugs daily for four days. Animal weights and pancreatic weights were measured, and oral glucose tolerance test was performed at the end of study. Fasting glucose, triglycerides (TG), total cholesterol (TC), HDL and insulin levels in serum, MDA, and GSH levels in pancreatic tissue were measured and HOMA-IR was calculated. Immunoexpression of apoptosis markers BAX, and BCL2 was measured in pancreatic tissues and BAX/BCL2 ratio was calculated. Histopathological examination of pancreatic tissues was also performed. Pancreatic weight, serum HDL, pancreatic GSH, and BCL2 were decreased while serum glucose, insulin, TG, TC levels, AUC of OGGT, HOMA-IR, pancreatic MDA, BAX, and BAX/BCL2 ratio were increased in IR rats. Histopathological examination showed congestion, vacuolation and hemorrhage in pancreatic islets. These changes were reversed by metformin and the high dose of coriander oil treatments. The obtained activities could be attributed to the presence of 21 volatile compounds, identified by GC-MS. Our study indicates that coriander oil can be used as an adjuvant antihyperglycemic agent in type 2 diabetes. Further experiments are needed to determine the therapeutic dose and the treatment time.
Collapse
Affiliation(s)
- Mona F. Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt;
| | - Noura Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt;
| | - Islam Mostafa
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt;
| | - Rehab A. Hasan
- Department of Histology, Faculty of Medicine for Girls, Al Azhar University, Cairo 11751, Egypt;
| | - Mansour Sobeh
- AgroBioSciences, Mohammed VI Polytechnic University, Lot 660, Hay MoulayRachid, Ben-Guerir 43150, Morocco
| |
Collapse
|
26
|
Oh S, Choi CH, Lee BJ, Park JH, Son KH, Byun K. Fermented Oyster Extract Attenuated Dexamethasone-Induced Muscle Atrophy by Decreasing Oxidative Stress. Molecules 2021; 26:molecules26237128. [PMID: 34885708 PMCID: PMC8658907 DOI: 10.3390/molecules26237128] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/12/2021] [Accepted: 11/22/2021] [Indexed: 12/25/2022] Open
Abstract
It is well known that oxidative stress induces muscle atrophy, which decreases with the activation of Nrf2/HO-1. Fermented oyster extracts (FO), rich in γ-aminobutyric acid (GABA) and lactate, have shown antioxidative effects. We evaluated whether FO decreased oxidative stress by upregulating Nrf2/HO-1 and whether it decreased NF-κB, leading to decreased IL-6 and TNF-α. Decreased oxidative stress led to the downregulation of Cbl-b ubiquitin ligase, which increased IGF-1 and decreased FoxO3, atrogin1, and Murf1, and eventually decreased muscle atrophy in dexamethasone (Dexa)-induced muscle atrophy animal model. For four weeks, mice were orally administered with FO, GABA, lactate, or GABA+Lactate, and then Dexa was subcutaneously injected for ten days. During Dexa injection period, FO, GABA, lactate, or GABA+Lactate were also administered, and grip strength test and muscle harvesting were performed on the day of the last Dexa injection. We compared the attenuation effect of FO with GABA, lactate, and GABA+lactate treatment. Nrf2 and HO-1 expressions were increased by Dexa but decreased by FO; SOD activity and glutathione levels were decreased by Dexa but increased by FO; NADPH oxidase activity was increased by Dexa but decreased by FO; NF-κB, IL-6, and TNF-α activities were increased by Dexa were decreased by FO; Cbl-b expression was increased by Dexa but restored by FO; IGF-1 expression was decreased by Dexa but increased by FO; FoxO3, Atrogin-1, and MuRF1 expressions were increased by Dexa but decreased by FO. The gastrocnemius thickness and weight were decreased by Dexa but increased by FO. The cross-sectional area of muscle fiber and grip strength were decreased by Dexa but increased by FO. In conclusion, FO decreased Dexa-induced oxidative stress through the upregulation of Nrf2/HO-1. Decreased oxidative stress led to decreased Cbl-b, FoxO3, atrogin1, and MuRF1, which attenuated muscle atrophy.
Collapse
Affiliation(s)
- Seyeon Oh
- Functional Cellular Networks Laboratory, Department of Medicine, Graduate School and Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon 21999, Korea;
| | - Chang Hu Choi
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon University, Incheon 21565, Korea;
| | - Bae-Jin Lee
- Marine Bioprocess Co., Ltd., Smart Marine BioCenter, Busan 46048, Korea; (B.-J.L.); (J.-H.P.)
| | - Joung-Hyun Park
- Marine Bioprocess Co., Ltd., Smart Marine BioCenter, Busan 46048, Korea; (B.-J.L.); (J.-H.P.)
| | - Kuk-Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon University, Incheon 21565, Korea;
- Correspondence: (K.-H.S.); (K.B.); Tel.: +82-32-460-3666 (K.-H.S.); +82-32-899-6511 (K.B.)
| | - Kyunghee Byun
- Functional Cellular Networks Laboratory, Department of Medicine, Graduate School and Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon 21999, Korea;
- Department of Anatomy and Cell Biology, Gachon University College of Medicine, Incheon 21936, Korea
- Correspondence: (K.-H.S.); (K.B.); Tel.: +82-32-460-3666 (K.-H.S.); +82-32-899-6511 (K.B.)
| |
Collapse
|
27
|
Polyphenols and Their Effects on Muscle Atrophy and Muscle Health. Molecules 2021; 26:molecules26164887. [PMID: 34443483 PMCID: PMC8398525 DOI: 10.3390/molecules26164887] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 12/17/2022] Open
Abstract
Skeletal muscle atrophy is the decrease in muscle mass and strength caused by reduced protein synthesis/accelerated protein degradation. Various conditions, such as denervation, disuse, aging, chronic diseases, heart disease, obstructive lung disease, diabetes, renal failure, AIDS, sepsis, cancer, and steroidal medications, can cause muscle atrophy. Mechanistically, inflammation, oxidative stress, and mitochondrial dysfunction are among the major contributors to muscle atrophy, by modulating signaling pathways that regulate muscle homeostasis. To prevent muscle catabolism and enhance muscle anabolism, several natural and synthetic compounds have been investigated. Recently, polyphenols (i.e., natural phytochemicals) have received extensive attention regarding their effect on muscle atrophy because of their potent antioxidant and anti-inflammatory properties. Numerous in vitro and in vivo studies have reported polyphenols as strongly effective bioactive molecules that attenuate muscle atrophy and enhance muscle health. This review describes polyphenols as promising bioactive molecules that impede muscle atrophy induced by various proatrophic factors. The effects of each class/subclass of polyphenolic compounds regarding protection against the muscle disorders induced by various pathological/physiological factors are summarized in tabular form and discussed. Although considerable variations in antiatrophic potencies and mechanisms were observed among structurally diverse polyphenolic compounds, they are vital factors to be considered in muscle atrophy prevention strategies.
Collapse
|