1
|
Wu S, Lu J. Liposome-Enabled Nanomaterials for Muscle Regeneration. SMALL METHODS 2025:e2402154. [PMID: 39967365 DOI: 10.1002/smtd.202402154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/08/2025] [Indexed: 02/20/2025]
Abstract
Muscle regeneration is a vital biological process that is crucial for maintaining muscle function and integrity, particularly for the treatment of muscle diseases such as sarcopenia and muscular dystrophy. Generally, muscular tissues can self-repair and regenerate under various conditions, including acute or chronic injuries, aging, and genetic mutation. However, regeneration becomes challenging beyond a certain threshold, particularly in severe muscle injuries or progressive diseases. In recent years, liposome-based nanotechnologies have shown potential as promising therapeutic strategies for muscle regeneration. Liposomes offer an adaptable platform for targeted drug delivery due to their cell membrane-like structure and excellent biocompatibility. They can enhance drug solubility, stability, and targeted delivery while minimizing systemic side effects by different mechanisms. This review summarizes recent advancements, discusses current applications and mechanisms, and highlights challenges and future directions for possible clinical translation of liposome-based nanomaterials in the treatment of muscle diseases. It is hoped this review offers new insights into the development of liposome-enabled nanomedicine to address current limitations.
Collapse
Affiliation(s)
- Shuang Wu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
- Clinical and Translational Oncology Program, The University of Arizona Cancer Center, Tucson, AZ, 85721, USA
- BIO5 Institute, The University of Arizona, Tucson, AZ, 85721, USA
- Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
2
|
Ruhee RT, Ma S, Suzuki K. Effects of Sulforaphane Treatment on Skeletal Muscle from Exhaustive Exercise-Induced Inflammation and Oxidative Stress Through the Nrf2/HO-1 Signaling Pathway. Antioxidants (Basel) 2025; 14:210. [PMID: 40002396 PMCID: PMC11851896 DOI: 10.3390/antiox14020210] [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: 11/29/2024] [Revised: 12/31/2024] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Skeletal muscle is primarily involved in exercise performance and health promotion. Sulforaphane (SFN) is a naturally occurring isothiocyanate that indirectly activates the transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2), thus inducing the expression of Nrf2 target genes, including antioxidant enzymes. This study aimed to identify the effects of a single dose of SFN administration on exhaustive exercise-induced inflammation and oxidative stress in skeletal muscle tissue and elucidate the underlying mechanisms. Thirty-six mice were divided into four groups: control, SFN, exercise (Ex), and SFN + Ex. The SFN group and SFN + Ex group received SFN orally (50 mg/kg body weight) 2 h before the running test. Exercise significantly reduced plasma glucose levels, while the SFN-treated group exhibited a smaller reduction. Acute exhaustive exercise increased the expression of pro-inflammatory cytokines in muscle tissue, while the SFN + Ex group exhibited significantly reduced expression of pro-inflammatory cytokines. The gene expression of Nrf2 and its target enzymes, including heme oxygenase (HO)-1, superoxide dismutase (SOD)-1, catalase (CAT), and glutathione peroxidase (GPx)-1, was measured in the gastrocnemius and soleus muscle tissue. Compared with the Ex group, the SFN + Ex group showed upregulated expression of all these parameters, including Nrf2. SFN treatment reduced acute exhaustive exercise-induced oxidative stress and inflammation via activation of the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Ruheea Taskin Ruhee
- Japan Society for the Promotion of Sciences, Chiyoda Ku 102-0083, Tokyo, Japan
| | - Sihui Ma
- Faculty of Human Sciences, Waseda University, Tokorozawa 359-1192, Japan;
| | - Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan
| |
Collapse
|
3
|
Soydan HE, Doğan A. Muscle Organoid and Assembloid Systems. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1474:1-12. [PMID: 38980551 DOI: 10.1007/5584_2024_816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Skeletal muscle is one of the most complex and largest tissues that perform important processes in the body, including performing voluntary movements and maintaining body temperature. Disruption of muscle homeostasis results in the development of several disorders, including diabetes and sarcopenia. To study the developmental and regenerative dynamics of skeletal muscle and the mechanism behind muscle diseases, it is important to model skeletal muscle and diseases in vitro. Since skeletal muscle has a complex structure and interaction with other tissues and cells that are required to perform their function, conventional 2D cultures are not sufficient to model the skeletal muscle with their interactions. Advances in the field of organoids and assembloids will enable the establishment of more complex and realistic tissue or disease models which cannot be fully recapitulated in conventional 2D culture systems for use in several areas, including disease research, regenerative, and tissue biology. To overcome these limitations, 3D organoid systems and assembloid systems are promising because of their success in recapitulating the complex structural organization, function, and cellular interactions of skeletal muscle.
Collapse
Affiliation(s)
- Hazar Eren Soydan
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, Turkey
| | - Ayşegül Doğan
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, Turkey.
| |
Collapse
|
4
|
Wu KC, Lin HW, Chu PC, Li CI, Kao HH, Lin CH, Cheng YJ. A non-invasive mouse model that recapitulates disuse-induced muscle atrophy in immobilized patients. Sci Rep 2023; 13:22201. [PMID: 38097709 PMCID: PMC10721881 DOI: 10.1038/s41598-023-49732-8] [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: 07/06/2023] [Accepted: 12/11/2023] [Indexed: 12/17/2023] Open
Abstract
Disuse muscle atrophy occurs consequent to prolonged limb immobility or bed rest, which represents an unmet medical need. As existing animal models of limb immobilization often cause skin erosion, edema, and other untoward effects, we here report an alternative method via thermoplastic immobilization of hindlimbs in mice. While significant decreases in the weight and fiber size were noted after 7 days of immobilization, no apparent skin erosion or edema was found. To shed light onto the molecular mechanism underlying this muscle wasting, we performed the next-generation sequencing analysis of gastrocnemius muscles from immobilized versus non-mobilized legs. Among a total of 55,487 genes analyzed, 787 genes were differentially expressed (> fourfold; 454 and 333 genes up- and down-regulated, respectively), which included genes associated with muscle tissue development, muscle system process, protein digestion and absorption, and inflammation-related signaling. From a clinical perspective, this model may help understand the molecular/cellular mechanism that drives muscle disuse and identify therapeutic strategies for this debilitating disease.
Collapse
Affiliation(s)
- Kun-Chang Wu
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan
| | - Hsiang-Wen Lin
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan
| | - Po-Chen Chu
- Department of Cosmeceutics and Graduate Institute of Cosmeceutics, China Medical University, Taichung, Taiwan
| | - Chia-Ing Li
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Hsiang-Han Kao
- Department of Family Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Hsueh Lin
- Department of Geriatric Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Yu-Jung Cheng
- Department of Physical Therapy and Graduate Institute of Rehabilitation Science, China Medical University, Dr. Yu-Jung Cheng, No. 100, Section 1, Jingmao Road, Beitun District, Taichung City, 406040, Taiwan.
- Department of Rehabilitation, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
5
|
Bieger P, Sangali TD, Ribeiro ÉCT, Schweigert Perry ID, Souza GC. Association of phase angle values and sarcopenia in older patients with heart failure. Nutr Clin Pract 2023; 38:672-685. [PMID: 36815519 DOI: 10.1002/ncp.10956] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/24/2022] [Accepted: 12/31/2022] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND Sarcopenia presents an accelerated and accentuated muscle loss in patients with heart failure (HF), leading to a worse prognosis for these patients. This study sought to assess the association of phase angle (PA) values with sarcopenia and its components, as well as to establish a PA cutoff point to predict outcomes such as hospitalization and mortality in older adult patients with HF. METHODS Sarcopenia diagnosis followed the European Working Group on Sarcopenia in Older People criteria. Anthropometric, bioelectrical impedance (PA and body composition), functional capacity and inflammatory markers were assessed. RESULTS Included patients were predominantly male (67%) and White, with a mean age of 69 ± 7 years, and a predominance of New York Heart Association I and II functional classes (82.1%) and reduced left ventricular ejection fraction (70.8%). Of the total sample, 23.6% were identified as sarcopenic. Mean PA values were lower in patients with sarcopenia (4.9 ± 0.9 and 6.0 ± 0.8°; P < 0.001). The cutoff point detected for sarcopenia in the receiver operating characteristic curve was 5.45°, which is an independent predictor for sarcopenia. PA values below this cutoff point were also associated with each of the sarcopenic components evaluated. PA proved to be an independent predictor for hospitalization (P = 0.042) in the entire sample studied. CONCLUSION PA is associated with diagnostic components of sarcopenia and the cutoff point 5.45° proved to be an independent predictor of sarcopenia and hospitalization >3 years in older adult patients with HF.
Collapse
Affiliation(s)
- Patrícia Bieger
- Graduate Program in Food, Nutrition and Health, Medical School, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Tamirys Delazeri Sangali
- Graduate Program in Food, Nutrition and Health, Medical School, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Édina Caroline Ternus Ribeiro
- Graduate Program in Health Sciences: Cardiology and Cardiovascular Sciences, Medical School, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Ingrid Dalira Schweigert Perry
- Food and Nutrition Research Center, Hospital de Clínicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Gabriela Corrêa Souza
- Graduate Program in Food, Nutrition and Health, Medical School, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Food and Nutrition Research Center, Hospital de Clínicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Heart Failure and Transplant Group, Nutrition Department, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
6
|
Kanazashi M, Tanaka M. Acute effect of electrical stimulation on muscle protein synthesis and break-down in the soleus muscle of hindlimb unloaded rats. Biomed Res 2023; 44:209-218. [PMID: 37779033 DOI: 10.2220/biomedres.44.209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Electrical stimulation (ES) is effective for disuse-induced muscle atrophy. However, the acute effect of ES on muscle protein synthesis (MPS) and muscle protein breakdown (MPB) remains unclear. We investigated the effect of a single-session ES treatment on mTORC1 signaling, MPS, and MPB in the soleus muscle of 2-week hindlimb unloaded rats. Sprague Dawley rats (n = 12 male) were randomly divided into control (CON) and hindlimb unloaded (HU) groups. After 2 weeks, the right soleus muscle was percutaneously stimulated and underwent supramaximal isometric contractions. The left soleus muscle served as an internal control. We collected soleus muscle samples 6 h after ES. Two weeks of HU decreased p70S6K and S6rp activation, downstream factors for mTORC1 signaling, and SUnSET method-assessed MPS, but increased the LC3-II/I ratio, an indicator of autophagy. ES on disused muscle successfully activated mTORC1 signaling but did not affect MPS. Contrary, ES decreased ubiquitinated proteins expression and LC3B-II/I ratio. HU might affect mTORC1 activation and MPS differently in response to acute ES possibly due to excessive ROS production caused by ES. Our findings suggest that ES applied to disused skeletal muscles may suppress MPB, but its effect on MPS appears to be attenuated.
Collapse
Affiliation(s)
- Miho Kanazashi
- Department of Physical Therapy, Faculty of Health and Welfare, Prefectural University of Hiroshima, 1-1 Gakuen-cho, Mihara-shi, Hiroshima 723-0053, Japan
| | - Masayuki Tanaka
- Department of Physical Therapy, Faculty of Health Sciences, Okayama Healthcare Professional Uni- versity, 3-2-18 Daiku, Kita-ku, Okayama-shi, Okayama 700-0913, Japan
| |
Collapse
|
7
|
Morales V, González A, Cabello-Verrugio C. Upregulation of CCL5/RANTES Gene Expression in the Diaphragm of Mice with Cholestatic Liver Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1408:201-218. [PMID: 37093429 DOI: 10.1007/978-3-031-26163-3_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Chronic liver diseases are a group of pathologies affecting the liver with high prevalence worldwide. Among them, cholestatic chronic liver diseases (CCLD) are characterized by alterations in liver function and increased plasma bile acids. Secondary to liver disease, under cholestasis, is developed sarcopenia, a skeletal muscle dysfunction with decreased muscle mass, strength, and physical function. CCL5/RANTES is a chemokine involved in the immune and inflammatory response. Indeed, CCL5 is a myokine because it is produced by skeletal muscle. Several studies show that bile acids induce CCL5/RANTES expression in liver cells. However, it is unknown if the expression of CCL5/RANTES is changed in the skeletal muscle of mice with cholestatic liver disease. We used a murine model of cholestasis-induced sarcopenia by intake of hepatotoxin 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC diet), in which we detected the mRNA levels for ccl5. We determined that mice fed the DDC diet presented high levels of serum bile acids and developed typical features of sarcopenia. Under these conditions, we detected the ccl5 gene expression in diaphragm muscle showing elevated mRNA levels compared to mice fed with a standard diet (chow diet). Our results collectively suggest an increased ccl5 gene expression in the diaphragm muscle concomitantly with elevated serum bile acids and the development of sarcopenia.
Collapse
Affiliation(s)
- Vania Morales
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Andrea González
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, 8370146, Chile.
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile.
| |
Collapse
|
8
|
Tacchi F, Orozco-Aguilar J, Valero-Breton M, Cabello-Verrugio C. Bile Acids Alter the Autophagy and Mitogenesis in Skeletal Muscle Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1408:183-199. [PMID: 37093428 DOI: 10.1007/978-3-031-26163-3_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Muscle atrophy decreases muscle mass with the subsequent loss of muscle function. Among the mechanisms that trigger sarcopenia is mitochondrial dysfunction. Mitochondria, whose primary function is to produce ATP, are dynamic organelles that present the process of formation (mitogenesis) and elimination (mitophagy). Failure of any of these processes contributes to mitochondrial malfunction. Mitogenesis is mainly controlled by Peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1α), a transcriptional coactivator that regulates the expression of TFAM, which participates in mitogenesis. Mitophagy is a process of selective autophagy. Autophagy corresponds to a degradative pathway of protein complexes and organelles. Liver disease caused sarcopenia and increased bile acids in the blood. We demonstrated that the treatment with cholic (CA) or deoxycholic (DCA) bile acids generates mitochondrial dysfunction and loss of biomass. This work assessed whether CA and DCA alter autophagy and mitogenesis. For this, western blot evaluated the autophagy process by determining the protein levels of the LC3II/LC3I ratio. In addition, we assessed mitogenesis using a luciferase-coupled plasmid reporter for the PGC-1α promoter and the protein levels of TFAM by western blot. Our results indicate that treatment with CA or DCA induces autophagy, represented by an increase in the LC3II/LC3I ratio. In addition, a decreased autophagic flux was observed. On the other hand, when treated with CA or DCA, a decrease in the activity of the PGC-1α promoter was observed. However, the levels of TFAM increased in myotubes incubated with CA and DCA. Our results demonstrate that CA and DCA modulate autophagy ad mitogenesis in C2C12 myotubes.
Collapse
Affiliation(s)
- Franco Tacchi
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Josué Orozco-Aguilar
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
- Laboratorio de Ensayos Biológicos (LEBi), Universidad de Costa Rica, San José, Costa Rica
- Facultad de Farmacia, Universidad de Costa Rica, San José, Costa Rica
| | - Mayalen Valero-Breton
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, 8370146, Chile.
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile.
| |
Collapse
|
9
|
Protective Effects of the Chalcone-Based Derivative AN07 on Inflammation-Associated Myotube Atrophy Induced by Lipopolysaccharide. Int J Mol Sci 2022; 23:ijms232112929. [PMID: 36361718 PMCID: PMC9655064 DOI: 10.3390/ijms232112929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/24/2022] [Accepted: 10/24/2022] [Indexed: 12/02/2022] Open
Abstract
Inflammation is a major cause of skeletal muscle atrophy in various diseases. 2-Hydroxy-4′-methoxychalcone (AN07) is a chalcone-based peroxisome-proliferator-activated receptor gamma (PPARγ) agonist with various effects, such as antiatherosclerosis, anti-inflammation, antioxidative stress, and neuroprotection. In this study, we examined the effects of AN07 on protein homeostasis pathway and mitochondrial function in inflammation-associated myotube atrophy induced by lipopolysaccharides (LPS). We found that AN07 significantly attenuated NF-κB activation, inflammatory factors (TNF-α, IL-1β, COX-2, and PGE2), Nox4 expression, and reactive oxygen species levels in LPS-treated C2C12 myotubes. Moreover, AN07 increased SOD2 expression and improved mitochondrial function, including mitochondrial membrane potential and mitochondrial oxygen consumption rate. We also demonstrated that AN07 attenuated LPS-induced reduction of myotube diameter, MyHC expression, and IGF-1/IGF-1R/p-Akt-mediated protein synthesis signaling. Additionally, AN07 downregulated LPS-induced autophagy–lysosomal protein degradation molecules (LC3-II/LC3-I and degraded p62) and ubiquitin–proteasome protein degradation molecules (n-FoxO1a/MuRF1/atrogin-1). However, the regulatory effects of AN07 on protein synthesis and degradation signaling were inhibited by the IGF-1R inhibitor AG1024 and the PI3K inhibitor wortmannin. In addition, the PPARγ antagonist GW9662 attenuated the effects of AN07 against LPS-induced inflammation, oxidation, and protein catabolism. In conclusion, our findings suggest that AN07 possesses protective effects on inflammation-induced myotube atrophy and mitochondrial dysfunction.
Collapse
|
10
|
Prado CM, Landi F, Chew STH, Atherton PJ, Molinger J, Ruck T, Gonzalez MC. Advances in Muscle Health and Nutrition: A Toolkit for Healthcare Professionals. Clin Nutr 2022; 41:2244-2263. [DOI: 10.1016/j.clnu.2022.07.041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 07/03/2022] [Accepted: 07/31/2022] [Indexed: 11/03/2022]
|
11
|
Chiappalupi S, Salvadori L, Mancuso F, Arato I, Calvitti M, Riuzzi F, Calafiore R, Luca G, Sorci G. Microencapsulated Sertoli cells sustain myoblast proliferation without affecting the myogenic potential. In vitro data. Data Brief 2022; 40:107744. [PMID: 35141363 PMCID: PMC8813587 DOI: 10.1016/j.dib.2021.107744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 11/25/2022] Open
Abstract
Sertoli cells (SeC) isolated from porcine testes have shown direct effects on muscle precursor cells sustaining C2C12 myoblasts proliferation and inhibiting oxidative stress and apoptosis in the early phase of the differentiation process, and stimulating myoblast fusion into myotubes and the expression of markers of myogenic differentiation in the late phase. This suggested that the cocktail of factors secreted by SeC stimulates proliferation in myoblasts without weakening their myogenic potential resulting in the formation of the critical myoblast amount necessary to rebuild the required muscle mass upon a damage. Here, we show that co-culturing C2C12 myoblasts with high doses of SeC microencapsulated in clinical grade alginate-based microcapsules (MC-SeC) for three days in differentiation medium (DM) translates into increased cell numbers and almost absence of myotube formation. However, after removal of MC-SeC, an intense fusion activity into myotubes was observed culminating in a fusion index similar to that of control after additional three days of culture in DM. These data definitely demonstrate that SeC-derived factors preserve the myogenic potential while sustaining cell proliferation in C2C12 myoblasts.
Collapse
|
12
|
Zheng YY, Wang Y, Chen X, Wei LS, Wang H, Tao T, Zhou YW, Jiang ZH, Qiu TT, Sun ZY, Sun J, Wang P, Zhao W, Li YQ, Chen HQ, Zhu MS, Zhang XN. The thymus regulates skeletal muscle regeneration by directly promoting satellite cell expansion. J Biol Chem 2021; 298:101516. [PMID: 34942145 PMCID: PMC8752954 DOI: 10.1016/j.jbc.2021.101516] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 01/22/2023] Open
Abstract
The thymus is the central immune organ, but it is known to progressively degenerate with age. As thymus degeneration is paralleled by the wasting of aging skeletal muscle, we speculated that the thymus may play a role in muscle wasting. Here, using thymectomized mice, we show that the thymus is necessary for skeletal muscle regeneration, a process tightly associated with muscle aging. Compared to control mice, the thymectomized mice displayed comparable growth of muscle mass, but decreased muscle regeneration in response to injury, as evidenced by small and sparse regenerative myofibers along with inhibited expression of regeneration-associated genes myh3, myod and myogenin. Using Pax7 immunofluorescence staining and BrdU incorporation assay, we determined that the decreased regeneration capacity was caused by a limited satellite cell pool. Interestingly, the conditioned culture medium of isolated thymocytes (TCMs) had a potent capacity to directly stimulate satellite cell expansion in vitro. These expanded cells were enriched in subpopulations of quiescent satellite cells (Pax7highMyoDlowEdUpos) and activated satellite cells (Pax7highMyoDhighEdUpos), which were efficiently incorporated into the regenerative myofibers. We thus propose that the thymus plays an essential role in muscle regeneration by directly promoting satellite cell expansion and may function profoundly in the muscle aging process.
Collapse
Affiliation(s)
- Yan-Yan Zheng
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School and Gulou Hospital affiliated Medical School, Nanjing University, Nanjing, 210061, China
| | - Ye Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School and Gulou Hospital affiliated Medical School, Nanjing University, Nanjing, 210061, China
| | - Xin Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School and Gulou Hospital affiliated Medical School, Nanjing University, Nanjing, 210061, China
| | - Li-Sha Wei
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School and Gulou Hospital affiliated Medical School, Nanjing University, Nanjing, 210061, China
| | - Han Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School and Gulou Hospital affiliated Medical School, Nanjing University, Nanjing, 210061, China
| | - Tao Tao
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School and Gulou Hospital affiliated Medical School, Nanjing University, Nanjing, 210061, China
| | - Yu-Wei Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School and Gulou Hospital affiliated Medical School, Nanjing University, Nanjing, 210061, China
| | - Zhi-Hui Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School and Gulou Hospital affiliated Medical School, Nanjing University, Nanjing, 210061, China
| | - Tian-Tian Qiu
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School and Gulou Hospital affiliated Medical School, Nanjing University, Nanjing, 210061, China
| | - Zhi-Yuan Sun
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210046, China
| | - Jie Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School and Gulou Hospital affiliated Medical School, Nanjing University, Nanjing, 210061, China
| | - Pei Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School and Gulou Hospital affiliated Medical School, Nanjing University, Nanjing, 210061, China
| | - Wei Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School and Gulou Hospital affiliated Medical School, Nanjing University, Nanjing, 210061, China
| | - Ye-Qiong Li
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School and Gulou Hospital affiliated Medical School, Nanjing University, Nanjing, 210061, China
| | - Hua-Qun Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210046, China.
| | - Min-Sheng Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School and Gulou Hospital affiliated Medical School, Nanjing University, Nanjing, 210061, China.
| | - Xue-Na Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School and Gulou Hospital affiliated Medical School, Nanjing University, Nanjing, 210061, China.
| |
Collapse
|
13
|
Swim training affects Akt signaling and ameliorates loss of skeletal muscle mass in a mouse model of amyotrophic lateral sclerosis. Sci Rep 2021; 11:20899. [PMID: 34686697 PMCID: PMC8536703 DOI: 10.1038/s41598-021-00319-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 10/11/2021] [Indexed: 12/01/2022] Open
Abstract
We tested the hypothesis that swim training reverses the impairment of Akt/FOXO3a signaling, ameliorating muscle atrophy in ALS mice. Transgenic male mice B6SJL-Tg (SOD1G93A) 1Gur/J were used as the ALS model (n = 35), with wild-type B6SJL (WT) mice as controls (n = 7). ALS mice were analyzed before ALS onset, at ALS onset, and at terminal ALS. Levels of insulin/Akt signaling pathway proteins were determined, and the body and tibialis anterior muscle mass and plasma creatine kinase. Significantly increased levels of FOXO3a in ALS groups (from about 13 to 21-fold) compared to WT mice were observed. MuRF1 levels in the ONSET untrained group (12.0 ± 1.7 AU) were significantly higher than in WT mice (1.12 ± 0.2 AU) and in the BEFORE ALS group (3.7 ± 0.9 AU). This was associated with body mass and skeletal muscle mass reduction. Swim training significantly ameliorated the reduction of skeletal muscle mass in both TERMINAL groups (p < 0.001) and partially reversed changes in the levels of Akt signaling pathway proteins. These findings shed light on the swimming-induced attenuation of skeletal muscle atrophy in ALS with possible practical implications for anti-cachexia approaches.
Collapse
|
14
|
Allosteric Modulation of GSK-3β as a New Therapeutic Approach in Limb Girdle Muscular Dystrophy R1 Calpain 3-Related. Int J Mol Sci 2021; 22:ijms22147367. [PMID: 34298987 PMCID: PMC8308041 DOI: 10.3390/ijms22147367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/28/2021] [Accepted: 07/06/2021] [Indexed: 12/28/2022] Open
Abstract
Limb-girdle muscular dystrophy R1 calpain 3-related (LGMDR1) is an autosomal recessive muscular dystrophy produced by mutations in the CAPN3 gene. It is a rare disease and there is no cure or treatment for the disease while the pathophysiological mechanism by which the absence of calpain 3 provokes the dystrophy in muscles is not clear. However, key proteins implicated in Wnt and mTOR signaling pathways, which regulate muscle homeostasis, showed a considerable reduction in their expression and in their phosphorylation in LGMDR1 patients' muscles. Finally, the administration of tideglusib and VP0.7, ATP non-competitive inhibitors of glycogen synthase kinase 3β (GSK-3β), restore the expression and phosphorylation of these proteins in LGMDR1 cells, opening the possibility of their use as therapeutic options.
Collapse
|
15
|
Jo K, Jang WY, Yun BS, Kim JS, Lee HS, Chang YB, Suh HJ. Effect of Deer Antler Extract on Muscle Differentiation and 5-Aminoimidazole-4-Carboxamide Ribonucleoside (AICAR)-Induced Muscle Atrophy in C2C12 Cells. Food Sci Anim Resour 2021; 41:623-635. [PMID: 34291211 PMCID: PMC8277185 DOI: 10.5851/kosfa.2021.e20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 01/13/2021] [Accepted: 04/08/2021] [Indexed: 11/06/2022] Open
Abstract
The effect of deer antler extract on muscle differentiation and muscle atrophy
were evaluated to minimize muscle loss following aging. Various deer antler
extracts (HWE, hot water extract of deer antler; FE, HWE of fermented deer
antler; ET, enzyme-assisted extract of deer antler; UE, extract prepared by
ultrasonication of deer antler) were evaluated for their effect on muscle
differentiation and inhibition of 5-aminoimidazole-4-carboxamide ribonucleoside
(AICAR)-induced muscle atrophy in C2C12 cells. Morphological changes according
to the effect of antler extracts on muscle differentiation were confirmed by
Jenner-Giemsa staining. In addition, the expression levels of genes related to
muscle differentiation and atrophy were confirmed through qRT-PCR. In the
presence of antler extracts, the length and thickness of myotubes and myogenin
differentiation 1 (MyoD1) and myogenic factor 5 (Myf5) gene expression were
increased compared to those in the control group (CON). Gene expression of
AMP-activated protein kinase (AMPK), MyoD1, and myogenin, along with the muscle
atrophy factors muscle RING finger-1 (MuRF-1) and forkhead box O3a (FoxO3a) upon
addition of deer antler extracts to muscle-atrophied C2C12 cells was determined
by qRT-PCR after treatment with AICAR. The expression of MuRF-1 and FoxO3a
decreased in the groups treated with antler extracts compared to that in the
group treated with AICAR alone. In addition, gene expression of MyoD1 and
myogenin in the muscle atrophy cell model was significantly increased compared
that into the CON. Therefore, our findings indicate that antler extract can
increase the expression of MyoD1, Myf5 and myogenin, inhibit muscle atrophy, and
promote muscle differentiation.
Collapse
Affiliation(s)
- Kyungae Jo
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Korea
| | - Woo Young Jang
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Korea
| | - Beom Sik Yun
- R D Center, Kwangdong Pharm Co., Ltd, Seoul 08381, Korea
| | - Jin Soo Kim
- R D Center, Kwangdong Pharm Co., Ltd, Seoul 08381, Korea
| | - Hyun-Sun Lee
- Agency for Korea National Food Cluster, Iksan 54576, Korea
| | - Yeok Boo Chang
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Korea
| | - Hyung Joo Suh
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Korea
| |
Collapse
|
16
|
Falqueto H, Júnior JLR, Silvério MNO, Farias JCH, Schoenfeld BJ, Manfredi LH. Can conditions of skeletal muscle loss be improved by combining exercise with anabolic-androgenic steroids? A systematic review and meta-analysis of testosterone-based interventions. Rev Endocr Metab Disord 2021; 22:161-178. [PMID: 33783694 DOI: 10.1007/s11154-021-09634-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/02/2021] [Indexed: 12/11/2022]
Abstract
Sarcopenia, cachexia, and atrophy due to inactivity and disease states are characterized by a loss of skeletal muscle mass, often accompanied by reduced levels of anabolic hormones (e.g. testosterone). These conditions are associated with an increase in mortality, hospitalization and worsening in quality of life. Both physical exercise (EX) and anabolic-androgenic steroid (AAS) administration can improve the prognosis of patients as they increase physical functionality. However, there is a gap in the literature as to the impact of these therapies on the gains in strength and muscle mass and their implications for patient safety. Accordingly, we performed a random-effects meta-analysis to elucidate the effects of AAS and/or EX interventions on lean body mass (LBM) and muscle strength in conditions involving muscle loss. A systematic search for relevant clinical trials was conducted in MEDLINE, EMBASE, SCOPUS, Web of Science, and SPORTDiscus. Comparisons included AAS vs. Control, EX vs. Control, AAS vs. EX, AAS + EX vs. AAS and AAS + EX vs. EX. A total of 1114 individuals were analyzed. AAS increased LBM (effect size [ES]: 0.46; 95% CI: 0.25, 0.68, P = 0.00) and muscle strength (ES: 0.31; 95% CI: 0.08, 0.53, P = 0.01) when compared to a control group. EX promoted an increase in muscular strength (ES: 0.89; 95% CI: 0.53, 1.25, P = 0.00), with no effect on LBM when compared to the control group (ES: 0.15; 95% CI: -0.07, 0.38, P = 0.17). AAS did not demonstrate statistically significant differences when compared to EX for LBM and muscle strength. The combination of EX + AAS promoted a greater increase in LBM and muscular strength when compared to AAS or EX in isolation. Qualitatively, AAS administration had relatively few side effects. Significant heterogeneity was found in some analyses, which may be explained by the use of different AAS types and EX protocols. Our findings suggest that AAS administration in cachectic and sarcopenic conditions may be a viable interventional strategy to enhance muscle function when exercise is not a possible approach. Moreover, combining AAS with exercise may enhance positive outcomes in this population.
Collapse
Affiliation(s)
- Hugo Falqueto
- Medical School, Federal University of Fronteira Sul, SC 484 - Km 02, Chapecó, Santa Catarina, Brazil
- Graduate Program in Biomedical Sciences, UFFS, Chapecó, Santa Catarina, Brazil
| | - Jorge L R Júnior
- Laboratory of Sport Biomechanics, Sports Department, School of Physical Education, Physiotherapy and Occupational Therapy, Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Mauro N O Silvério
- Medical School, Federal University of Fronteira Sul, SC 484 - Km 02, Chapecó, Santa Catarina, Brazil
| | - Juliano C H Farias
- Medical School, Federal University of Fronteira Sul, SC 484 - Km 02, Chapecó, Santa Catarina, Brazil
| | | | - Leandro H Manfredi
- Medical School, Federal University of Fronteira Sul, SC 484 - Km 02, Chapecó, Santa Catarina, Brazil.
- Graduate Program in Biomedical Sciences, UFFS, Chapecó, Santa Catarina, Brazil.
| |
Collapse
|
17
|
Moon JY, Kim HS. α-Syntrophin alleviates ER stress to maintain protein homeostasis during myoblast differentiation. FEBS Lett 2021; 595:1656-1670. [PMID: 33834492 DOI: 10.1002/1873-3468.14088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/02/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022]
Abstract
We have previously shown evidence that α-syntrophin plays an important role in myoblast differentiation. In this study, we focused on abnormal myotube formation of the α-syntrophin knockdown C2 cell line (SNKD). The overall amount of intracellular protein and muscle-specific proteins in SNKD cells were significantly lower than those in the control. Akt-mTOR signaling, an important pathway for protein synthesis and muscle hypertrophy, was downregulated. In addition, the levels of endoplasmic reticulum (ER) stress markers increased in SNKD cells. The decrease in intracellular protein synthesis and reduction in the myotube diameter in SNKD cells were restored by 4-phenylbutyric acid, a chemical chaperone, or overexpression of α-syntrophin. These results suggest a novel role for α-syntrophin in protein homeostasis during myoblast differentiation.
Collapse
Affiliation(s)
- Jae Yun Moon
- Department of Biological Science, Ajou University, Suwon, Korea
| | - Hye Sun Kim
- Department of Biological Science, Ajou University, Suwon, Korea
| |
Collapse
|
18
|
Cui Q, Yang H, Gu Y, Zong C, Chen X, Lin Y, Sun H, Shen Y, Zhu J. RNA sequencing (RNA-seq) analysis of gene expression provides new insights into hindlimb unloading-induced skeletal muscle atrophy. ANNALS OF TRANSLATIONAL MEDICINE 2021; 8:1595. [PMID: 33437794 PMCID: PMC7791259 DOI: 10.21037/atm-20-7400] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Weightlessness-induced skeletal muscle atrophy, accompanied by complex biochemical and physiological changes, has potentially damaged consequences. However, there is still an insufficient effective strategy to treat skeletal muscle atrophy. Therefore, exploring the molecular mechanisms regulating skeletal muscle atrophy and effective protection is necessary. Methods RNA sequencing (RNA-seq) analysis was used to detect differentially expressed genes (DEGs) in the soleus muscle at 12, 24, 36 hours, three days, and seven days after hindlimb unloading in rats. Pearson correlation heatmaps and principal component analysis (PCA) were applied to analyze DEGs’ expression profiles. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were used for cluster analysis of DEGs. Ingenuity pathway analysis (IPA) was used to analyze specific biological processes further. Results At different time points (12, 24, 36 hours, three days, seven days) after hindlimb unloading, the expression levels of 712, 1,109, 1,433, 1,162, and 1,182 genes in rat soleus muscle were upregulated, respectively, whereas the expression levels of 1,186, 1,324, 1,632, 1,446, and 1,596 genes were downregulated, respectively. PCA revealed that rat soleus muscle showed three different transcriptional phases within seven days after hindlimb unloading. KEGG and GO annotation indicated that the first transcriptional phase primarily involved the activation of stress responses, including oxidative stress, and the inhibition of cell proliferation and angiogenesis; the second transcriptional phase primarily involved the activation of proteolytic systems and, to a certain degree, inflammatory responses; and the third transcriptional phase primarily involved extensive activation of the proteolytic system, significant inhibition of energy metabolism, and activation of the aging process and slow-to-fast muscle conversion. Conclusions Different physiological processes in rat skeletal muscles were activated sequentially after unloading. From these activated biological processes, the three transcriptional phases after skeletal muscle unloading can be successively defined as the stress response phase, the atrophic initiation phase, and the atrophic phase. Our study not only helps in the understanding of the molecular mechanisms underlying weightlessness-induced muscle atrophy but may also provide an important time window for the treatment and prevention of weightlessness-induced muscle atrophy.
Collapse
Affiliation(s)
- Qihao Cui
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, China
| | - Hua Yang
- Department of Neurosurgery, People's Hospital of Binhai County, Yancheng, China
| | - Yuming Gu
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, China
| | - Chenyu Zong
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, China
| | - Xin Chen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yinghao Lin
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, China
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Jianwei Zhu
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
19
|
Salvadori L, Mandrone M, Manenti T, Ercolani C, Cornioli L, Lianza M, Tomasi P, Chiappalupi S, Di Filippo ES, Fulle S, Poli F, Sorci G, Riuzzi F. Identification of Withania somnifera-Silybum marianum-Trigonella foenum-graecum Formulation as a Nutritional Supplement to Contrast Muscle Atrophy and Sarcopenia. Nutrients 2020; 13:E49. [PMID: 33375229 PMCID: PMC7824275 DOI: 10.3390/nu13010049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Muscle atrophy, i.e., the loss of skeletal muscle mass and function, is an unresolved problem associated with aging (sarcopenia) and several pathological conditions. The imbalance between myofibrillary protein breakdown (especially the adult isoforms of myosin heavy chain, MyHC) and synthesis, and the reduction of muscle regenerative potential are main causes of muscle atrophy. Methods: Starting from one-hundred dried hydroalcoholic extracts of medical plants, we identified those able to contrast the reduction of C2C12 myotube diameter in well-characterized in vitro models mimicking muscle atrophy associated to inflammatory states, glucocorticoid treatment or nutrient deprivation. Based on their ability to rescue type II MyHC (MyHC-II) expression in atrophying conditions, six extracts with different phytochemical profiles were selected, mixed in groups of three, and tested on atrophic myotubes. The molecular mechanism underpinning the effects of the most efficacious formulation, and its efficacy on myotubes obtained from muscle biopsies of young and sarcopenic subjects were also investigated. Results: We identified WST (Withania somnifera, Silybum marianum, Trigonella foenum-graecum) formulation as extremely efficacious in protecting C2C12 myotubes against MyHC-II degradation by stimulating Akt (protein kinase B)-dependent protein synthesis and p38 MAPK (p38 mitogen-activated protein kinase)/myogenin-dependent myoblast differentiation. WST sustains trophism in C2C12 and young myotubes, and rescues the size, developmental MyHC expression and myoblast fusion in sarcopenic myotubes. Conclusion: WST strongly counteracts muscle atrophy associated to different conditions in vitro. The future validation in vivo of our results might lead to the use of WST as a food supplement to sustain muscle mass in diffuse atrophying conditions, and to reverse the age-related functional decline of human muscles, thus improving people quality of life and reducing social and health-care costs.
Collapse
Affiliation(s)
- Laura Salvadori
- Department Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy;
- Interuniversity Institute of Myology (IIM), 06132 Perugia, Italy; (S.C.); (E.S.D.F.); (S.F.); (G.S.)
| | - Manuela Mandrone
- Department Pharmacy and Biotechnology (FaBiT), University of Bologna, 40126 Bologna, Italy; (M.M.); (M.L.); (P.T.); (F.P.)
| | - Tommaso Manenti
- Biokyma srl Laboratories, 52031 Anghiari, Italy; (T.M.); (C.E.); (L.C.)
| | - Catia Ercolani
- Biokyma srl Laboratories, 52031 Anghiari, Italy; (T.M.); (C.E.); (L.C.)
| | - Luca Cornioli
- Biokyma srl Laboratories, 52031 Anghiari, Italy; (T.M.); (C.E.); (L.C.)
| | - Mariacaterina Lianza
- Department Pharmacy and Biotechnology (FaBiT), University of Bologna, 40126 Bologna, Italy; (M.M.); (M.L.); (P.T.); (F.P.)
| | - Paola Tomasi
- Department Pharmacy and Biotechnology (FaBiT), University of Bologna, 40126 Bologna, Italy; (M.M.); (M.L.); (P.T.); (F.P.)
| | - Sara Chiappalupi
- Interuniversity Institute of Myology (IIM), 06132 Perugia, Italy; (S.C.); (E.S.D.F.); (S.F.); (G.S.)
- Department Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Ester Sara Di Filippo
- Interuniversity Institute of Myology (IIM), 06132 Perugia, Italy; (S.C.); (E.S.D.F.); (S.F.); (G.S.)
- Department Neuroscience, Imaging and Clinical Sciences, University G. D’Annunzio Chieti e Pescara, 66100 Chieti, Italy
| | - Stefania Fulle
- Interuniversity Institute of Myology (IIM), 06132 Perugia, Italy; (S.C.); (E.S.D.F.); (S.F.); (G.S.)
- Department Neuroscience, Imaging and Clinical Sciences, University G. D’Annunzio Chieti e Pescara, 66100 Chieti, Italy
| | - Ferruccio Poli
- Department Pharmacy and Biotechnology (FaBiT), University of Bologna, 40126 Bologna, Italy; (M.M.); (M.L.); (P.T.); (F.P.)
| | - Guglielmo Sorci
- Interuniversity Institute of Myology (IIM), 06132 Perugia, Italy; (S.C.); (E.S.D.F.); (S.F.); (G.S.)
- Department Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Francesca Riuzzi
- Interuniversity Institute of Myology (IIM), 06132 Perugia, Italy; (S.C.); (E.S.D.F.); (S.F.); (G.S.)
- Department Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| |
Collapse
|
20
|
Taurine Attenuates Catabolic Processes Related to the Onset of Sarcopenia. Int J Mol Sci 2020; 21:ijms21228865. [PMID: 33238549 PMCID: PMC7700215 DOI: 10.3390/ijms21228865] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/13/2020] [Accepted: 11/20/2020] [Indexed: 12/25/2022] Open
Abstract
Sarcopenia that occurs with advancing age is characterized by a gradual loss of muscle protein component due to the activation of catabolic pathways, increased level of inflammation, and mitochondrial dysfunction. Experimental evidence demonstrates that several physio-pathological processes involved in the onset of sarcopenia may be counteracted by the intake of specific amino acids or antioxidant molecules, suggesting that diet may represent an effective strategy for improving the anabolic response of muscle during aging. The non-essential amino acid taurine is highly expressed in several mammalian tissues, including skeletal muscle where it is involved in the ion channel regulation, in the modulation of intracellular calcium concentration, and where it plays an important role as an antioxidant and anti-inflammatory factor. Here, with the purpose to reproduce the chronic low-grade inflammation characteristics of senescent muscle in an in vitro system, we exploited the role of Tumor Necrosis Factor α (TNF) and we analyzed the effect of taurine in the modulation of different signaling pathways known to be dysregulated in sarcopenia. We demonstrated that the administration of high levels of taurine in myogenic L6 cells stimulates the differentiation process by downregulating the expression of molecules involved in inflammatory pathways and modulating processes such as autophagy and apoptosis. Although further studies are currently ongoing in our laboratory to better elucidate the molecular mechanisms responsible for the positive effect of taurine on myogenic differentiation, this study suggests that taurine supplementation may represent a strategy to delay the loss of mass and functionality characteristic of senescent muscles.
Collapse
|
21
|
Webster JM, Kempen LJAP, Hardy RS, Langen RCJ. Inflammation and Skeletal Muscle Wasting During Cachexia. Front Physiol 2020; 11:597675. [PMID: 33329046 PMCID: PMC7710765 DOI: 10.3389/fphys.2020.597675] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/19/2020] [Indexed: 12/16/2022] Open
Abstract
Cachexia is the involuntary loss of muscle and adipose tissue that strongly affects mortality and treatment efficacy in patients with cancer or chronic inflammatory disease. Currently, no specific treatments or interventions are available for patients developing this disorder. Given the well-documented involvement of pro-inflammatory cytokines in muscle and fat metabolism in physiological responses and in the pathophysiology of chronic inflammatory disease and cancer, considerable interest has revolved around their role in mediating cachexia. This has been supported by association studies that report increased levels of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) in some, but not all, cancers and in chronic inflammatory diseases such as chronic obstructive pulmonary disease (COPD) and rheumatoid arthritis (RA). In addition, preclinical studies including animal disease models have provided a substantial body of evidence implicating a causal contribution of systemic inflammation to cachexia. The presence of inflammatory cytokines can affect skeletal muscle through several direct mechanisms, relying on activation of the corresponding receptor expressed by muscle, and resulting in inhibition of muscle protein synthesis (MPS), elevation of catabolic activity through the ubiquitin-proteasomal system (UPS) and autophagy, and impairment of myogenesis. Additionally, systemic inflammatory mediators indirectly contribute to muscle wasting through dysregulation of tissue and organ systems, including GCs via the hypothalamus-pituitary-adrenal (HPA) axis, the digestive system leading to anorexia-cachexia, and alterations in liver and adipocyte behavior, which subsequently impact on muscle. Finally, myokines secreted by skeletal muscle itself in response to inflammation have been implicated as autocrine and endocrine mediators of cachexia, as well as potential modulators of this debilitating condition. While inflammation has been shown to play a pivotal role in cachexia development, further understanding how these cytokines contribute to disease progression is required to reveal biomarkers or diagnostic tools to help identify at risk patients, or enable the design of targeted therapies to prevent or delay the progression of cachexia.
Collapse
Affiliation(s)
- Justine M. Webster
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Laura J. A. P. Kempen
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Rowan S. Hardy
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Institute for Clinical Sciences, University of Birmingham, Birmingham, United Kingdom
- MRC Arthritis Research UK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, United Kingdom
| | - Ramon C. J. Langen
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
22
|
Conceição M, Forcina L, Wiklander OPB, Gupta D, Nordin JZ, Vrellaku B, McClorey G, Mäger I, Gӧrgens A, Lundin P, Musarò A, Wood MJA, Andaloussi SE, Roberts TC. Engineered extracellular vesicle decoy receptor-mediated modulation of the IL6 trans-signalling pathway in muscle. Biomaterials 2020; 266:120435. [PMID: 33049461 DOI: 10.1016/j.biomaterials.2020.120435] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/27/2020] [Accepted: 10/04/2020] [Indexed: 12/28/2022]
Abstract
The cytokine interleukin 6 (IL6) is a key mediator of inflammation that contributes to skeletal muscle pathophysiology. IL6 activates target cells by two main mechanisms, the classical and trans-signalling pathways. While classical signalling is associated with the anti-inflammatory activities of the cytokine, the IL6 trans-signalling pathway mediates chronic inflammation and is therefore a target for therapeutic intervention. Extracellular vesicles (EVs) are natural, lipid-bound nanoparticles, with potential as targeted delivery vehicles for therapeutic macromolecules. Here, we engineered EVs to express IL6 signal transducer (IL6ST) decoy receptors to selectively inhibit the IL6 trans-signalling pathway. The potency of the IL6ST decoy receptor EVs was optimized by inclusion of a GCN4 dimerization domain and a peptide sequence derived from syntenin-1 which targets the decoy receptor to EVs. The resulting engineered EVs were able to efficiently inhibit activation of the IL6 trans-signalling pathway in reporter cells, while having no effect on the IL6 classical signalling. IL6ST decoy receptor EVs, were also capable of blocking the IL6 trans-signalling pathway in C2C12 myoblasts and myotubes, thereby inhibiting the phosphorylation of STAT3 and partially reversing the anti-differentiation effects observed when treating cells with IL6/IL6R complexes. Treatment of a Duchenne muscular dystrophy mouse model with IL6ST decoy receptor EVs resulted in a reduction in STAT3 phosphorylation in the quadriceps and gastrocnemius muscles of these mice, thereby demonstrating in vivo activity of the decoy receptor EVs as a potential therapy. Taken together, this study reveals the IL6 trans-signalling pathway as a promising therapeutic target in DMD, and demonstrates the therapeutic potential of IL6ST decoy receptor EVs.
Collapse
Affiliation(s)
- Mariana Conceição
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Laura Forcina
- DAHFMO-Unit of Histology and Medical Embryology, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, IMM, Sapienza University of Rome, Rome, Italy
| | - Oscar P B Wiklander
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Evox Therapeutics Limited, Oxford Science Park, Oxford, UK
| | - Dhanu Gupta
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Evox Therapeutics Limited, Oxford Science Park, Oxford, UK
| | - Joel Z Nordin
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Evox Therapeutics Limited, Oxford Science Park, Oxford, UK
| | | | - Graham McClorey
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Department of Paediatrics, University of Oxford, Oxford, UK
| | - Imre Mäger
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Department of Paediatrics, University of Oxford, Oxford, UK
| | - André Gӧrgens
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Evox Therapeutics Limited, Oxford Science Park, Oxford, UK; Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | - Per Lundin
- Evox Therapeutics Limited, Oxford Science Park, Oxford, UK
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, IMM, Sapienza University of Rome, Rome, Italy; Center for Life Nano Science @Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Department of Paediatrics, University of Oxford, Oxford, UK; MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK
| | - Samir El Andaloussi
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Evox Therapeutics Limited, Oxford Science Park, Oxford, UK
| | - Thomas C Roberts
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Department of Paediatrics, University of Oxford, Oxford, UK; MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK.
| |
Collapse
|
23
|
Nutrition and microRNAs: Novel Insights to Fight Sarcopenia. Antioxidants (Basel) 2020; 9:antiox9100951. [PMID: 33023202 PMCID: PMC7601022 DOI: 10.3390/antiox9100951] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/18/2022] Open
Abstract
Sarcopenia is a progressive age-related loss of skeletal muscle mass and strength, which may result in increased physical frailty and a higher risk of adverse events. Low-grade systemic inflammation, loss of muscle protein homeostasis, mitochondrial dysfunction, and reduced number and function of satellite cells seem to be the key points for the induction of muscle wasting, contributing to the pathophysiological mechanisms of sarcopenia. While a range of genetic, hormonal, and environmental factors has been reported to contribute to the onset of sarcopenia, dietary interventions targeting protein or antioxidant intake may have a positive effect in increasing muscle mass and strength, regulating protein homeostasis, oxidative reaction, and cell autophagy, thus providing a cellular lifespan extension. MicroRNAs (miRNAs) are endogenous small non-coding RNAs, which control gene expression in different tissues. In skeletal muscle, a range of miRNAs, named myomiRNAs, are involved in many physiological processes, such as growth, development, and maintenance of muscle mass and function. This review aims to present and to discuss some of the most relevant molecular mechanisms related to the pathophysiological effect of sarcopenia. Besides, we explored the role of nutrition as a possible way to counteract the loss of muscle mass and function associated with ageing, with special attention paid to nutrient-dependent miRNAs regulation. This review will provide important information to better understand sarcopenia and, thus, to facilitate research and therapeutic strategies to counteract the pathophysiological effect of ageing.
Collapse
|
24
|
Gabellini D, Musarò A. 16th Meeting of the Interuniversity Institute of Myology (IIM) - Assisi (Italy), October 17-20, 2019: Foreword, Program and Abstracts. Eur J Transl Myol 2020; 30:9345. [PMID: 33117514 PMCID: PMC7582450 DOI: 10.4081/ejtm.2020.9345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 09/11/2020] [Indexed: 11/24/2022] Open
Abstract
The 16th Meeting of the Interuniversity Institute of Myology (IIM), October 17-20, 2019, Assisi, Italy brought together scientists, pharma and patient organization representatives discussing new results on muscle research. Internationally renowned Keynote speakers presented advances on muscle development, homeostasis, metabolism, and disease. Speakers selected among submitted abstracts presented their new, unpublished data in seven scientific sessions. The remaining abstracts were showcased in two poster sessions. Young trainees where directly involved in the selection of keynote speakers, the organizing scientific sessions and roundtables discussions tailored to the interests of their peers. A broad Italian, European and North-American audience participated to the different initiatives. The meeting allowed muscle biology researchers to discuss ideas and scientific collaborations aimed at better understanding the mechanisms underlying muscle diseases in order to develop better therapeutic strategies. The active participation of young trainees was facilitated by the friendly and inclusive atmosphere, which fostered lively discussions identifying emerging areas of myology research and stimulated scientific cross-fertilization. The meeting was a success and the IIM community will continue to bring forward significant contributions to the understanding of muscle development and function, the pathogenesis of muscular diseases and the development of novel therapeutic approaches. Here, we report abstracts of the meeting illustrating novel results of basic, translational, and clinical research, which confirms that the Myology field is strong and healthy.
Collapse
Affiliation(s)
- Davide Gabellini
- Gene Expression and Muscular Dystrophy Group, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Laboratory Affiliated to “Istituto Pasteur Italia – Fondazione Cenci Bolognetti”, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
25
|
Docosahexaenoic Acid, a Potential Treatment for Sarcopenia, Modulates the Ubiquitin-Proteasome and the Autophagy-Lysosome Systems. Nutrients 2020; 12:nu12092597. [PMID: 32859116 PMCID: PMC7551806 DOI: 10.3390/nu12092597] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/22/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022] Open
Abstract
One of the characteristic features of aging is the progressive loss of muscle mass, a nosological syndrome called sarcopenia. It is also a pathologic risk factor for many clinically adverse outcomes in older adults. Therefore, delaying the loss of muscle mass, through either boosting muscle protein synthesis or slowing down muscle protein degradation using nutritional supplements could be a compelling strategy to address the needs of the world’s aging population. Here, we review the recently identified properties of docosahexaenoic acid (DHA). It was shown to delay muscle wasting by stimulating intermediate oxidative stress and inhibiting proteasomal degradation of muscle proteins. Both the ubiquitin–proteasome and the autophagy–lysosome systems are modulated by DHA. Collectively, growing evidence indicates that DHA is a potent pharmacological agent that could improve muscle homeostasis. Better understanding of cellular proteolytic systems associated with sarcopenia will allow us to identify novel therapeutic interventions, such as omega-3 polyunsaturated fatty acids, to treat this disease.
Collapse
|
26
|
Musarò A, Scicchitano BM. Counteracting sarcopenia: the role of IGF-1 isoforms. Aging (Albany NY) 2020; 11:3410-3411. [PMID: 31195371 PMCID: PMC6594807 DOI: 10.18632/aging.102027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 06/09/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome 00161, Italy
| | - Bianca Maria Scicchitano
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario "Agostino Gemelli", IRCCS, Roma 00168, Italy
| |
Collapse
|
27
|
Razzaque MS, Atfi A. Regulatory Role of the Transcription Factor Twist1 in Cancer-Associated Muscle Cachexia. Front Physiol 2020; 11:662. [PMID: 32655411 PMCID: PMC7324683 DOI: 10.3389/fphys.2020.00662] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/25/2020] [Indexed: 12/19/2022] Open
Abstract
Muscle cachexia is a catabolic response, usually takes place in various fatal diseases, such as sepsis, burn injury, and chronic kidney disease. Muscle cachexia is also a common co-morbidity seen in the vast majority of advanced cancer patients, often associated with low quality of life and death due to general organ dysfunction. The triggering events and underlying molecular mechanisms of muscle wasting are not yet clearly defined. Our recent study has shown that the ectopic expression of Twist1 in muscle progenitor cells is sufficient to drive muscle structural protein breakdown and attendant muscle atrophy, reminiscent of muscle cachexia. Intriguingly, muscle Twist1 expression is highly induced in cachectic muscles from several mouse models of pancreatic ductal adenocarcinoma (PDAC), raising the interesting possibility that Twist1 may mediate PDAC-driven muscle cachexia. Along these lines, both genetic and pharmacological inactivation of Twist1 function was highly significant at protecting against cancer cachexia, which translated into a significant survival benefit in the experimental PDAC animals. From a translational perspective, elevated expression of Twist1 is also detected in cancer patients with severe muscle wasting, implicating a role of Twist1 in cancer cachexia, and further providing a possible target for therapeutic attenuation of cachexia to improve cancer patient survival. In this article, we will briefly summarize how Twist1 acts as a master regulator of tumor-induced cachexia, and discuss the relevance of our findings to muscle wasting diseases in general. The mechanism of decreased muscle mass in various catabolic conditions is thought to rely on similar pathways, and, therefore, Twist1-induced cancer cachexia may benefit diverse groups of patients with clinical complications associated with loss of muscle mass and functions, beyond the expected benefits for cancer patients.
Collapse
Affiliation(s)
- Mohammed S Razzaque
- Department of Pathology, Lake Erie College of Osteopathic Medicine, Erie, PA, United States
| | - Azeddine Atfi
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
28
|
Forcina L, Cosentino M, Musarò A. Mechanisms Regulating Muscle Regeneration: Insights into the Interrelated and Time-Dependent Phases of Tissue Healing. Cells 2020; 9:E1297. [PMID: 32456017 PMCID: PMC7290814 DOI: 10.3390/cells9051297] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022] Open
Abstract
Despite a massive body of knowledge which has been produced related to the mechanisms guiding muscle regeneration, great interest still moves the scientific community toward the study of different aspects of skeletal muscle homeostasis, plasticity, and regeneration. Indeed, the lack of effective therapies for several physiopathologic conditions suggests that a comprehensive knowledge of the different aspects of cellular behavior and molecular pathways, regulating each regenerative stage, has to be still devised. Hence, it is important to perform even more focused studies, taking the advantage of robust markers, reliable techniques, and reproducible protocols. Here, we provide an overview about the general aspects of muscle regeneration and discuss the different approaches to study the interrelated and time-dependent phases of muscle healing.
Collapse
Affiliation(s)
| | | | - Antonio Musarò
- Laboratory affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via Antonio Scarpa, 14, 00161 Rome, Italy; (L.F.); (M.C.)
| |
Collapse
|
29
|
Carraro U. Thirty years of translational research in Mobility Medicine: Collection of abstracts of the 2020 Padua Muscle Days. Eur J Transl Myol 2020; 30:8826. [PMID: 32499887 PMCID: PMC7254447 DOI: 10.4081/ejtm.2019.8826] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 01/22/2020] [Indexed: 12/11/2022] Open
Abstract
More than half a century of skeletal muscle research is continuing at Padua University (Italy) under the auspices of the Interdepartmental Research Centre of Myology (CIR-Myo), the European Journal of Translational Myology (EJTM) and recently also with the support of the A&CM-C Foundation for Translational Myology, Padova, Italy. The Volume 30(1), 2020 of the EJTM opens with the collection of abstracts for the conference "2020 Padua Muscle Days: Mobility Medicine 30 years of Translational Research". This is an international conference that will be held between March 18-21, 2020 in Euganei Hills and Padova in Italy. The abstracts are excellent examples of translational research and of the multidimensional approaches that are needed to classify and manage (in both the acute and chronic phases) diseases of Mobility that span from neurologic, metabolic and traumatic syndromes to the biological process of aging. One of the typical aim of Physical Medicine and Rehabilitation is indeed to reduce pain and increase mobility enough to enable impaired persons to walk freely, garden, and drive again. The excellent contents of this Collection of Abstracts reflect the high scientific caliber of researchers and clinicians who are eager to present their results at the PaduaMuscleDays. A series of EJTM Communications will also add to this preliminary evidence.
Collapse
Affiliation(s)
- Ugo Carraro
- Interdepartmental Research Centre of Myology (CIR-Myo), Department of Biomedical Sciences, University of Padova, Italy
- A&C M-C Foundation for Translational Myology, Padova, Italy
| |
Collapse
|
30
|
Zeb2 Regulates Myogenic Differentiation in Pluripotent Stem Cells. Int J Mol Sci 2020; 21:ijms21072525. [PMID: 32260521 PMCID: PMC7177401 DOI: 10.3390/ijms21072525] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 01/01/2023] Open
Abstract
Skeletal muscle differentiation is triggered by a unique family of myogenic basic helix-loop-helix transcription factors, including MyoD, MRF-4, Myf-5, and Myogenin. These transcription factors bind promoters and distant regulatory regions, including E-box elements, of genes whose expression is restricted to muscle cells. Other E-box binding zinc finger proteins target the same DNA response elements, however, their function in muscle development and regeneration is still unknown. Here, we show that the transcription factor zinc finger E-box-binding homeobox 2 (Zeb2, Sip-1, Zfhx1b) is present in skeletal muscle tissues. We investigate the role of Zeb2 in skeletal muscle differentiation using genetic tools and transgenic mouse embryonic stem cells, together with single-cell RNA-sequencing and in vivo muscle engraftment capability. We show that Zeb2 over-expression has a positive impact on skeletal muscle differentiation in pluripotent stem cells and adult myogenic progenitors. We therefore propose that Zeb2 is a novel myogenic regulator and a possible target for improving skeletal muscle regeneration. The non-neural roles of Zeb2 are poorly understood.
Collapse
|
31
|
De Spiegeleer A, Elewaut D, Van Den Noortgate N, Janssens Y, Debunne N, Van Langenhove S, Govindarajan S, De Spiegeleer B, Wynendaele E. Quorum sensing molecules as a novel microbial factor impacting muscle cells. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165646. [DOI: 10.1016/j.bbadis.2019.165646] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 12/15/2019] [Indexed: 02/07/2023]
|
32
|
Increased Circulating Levels of Interleukin-6 Affect the Redox Balance in Skeletal Muscle. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3018584. [PMID: 31827671 PMCID: PMC6881749 DOI: 10.1155/2019/3018584] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/01/2019] [Accepted: 09/26/2019] [Indexed: 12/25/2022]
Abstract
The extent of oxidative stress and chronic inflammation are closely related events which coexist in a muscle environment under pathologic conditions. It has been generally accepted that the inflammatory cells, as well as myofibers, are sources of reactive species which are, in turn, able to amplify the activation of proinflammatory pathways. However, the precise mechanism underlining the physiopathologic interplay between ROS generation and inflammatory response has to be fully clarified. Thus, the identification of key molecular players in the interconnected pathogenic network between the two processes might help to design more specific therapeutic approaches for degenerative diseases. Here, we investigated whether elevated circulating levels of the proinflammatory cytokine Interleukin-6 (IL-6) are sufficient to perturb the physiologic redox balance in skeletal muscle, independently of tissue damage and inflammatory response. We observed that the overexpression of circulating IL-6 enhances the generation and accumulation of free radicals in the diaphragm muscle of adult NSE/IL-6 mice, by deregulating redox-associated molecular circuits and impinging the nuclear factor erythroid 2-related factor 2- (Nrf2-) mediated antioxidant response. Our findings are coherent with a model in which uncontrolled levels of IL-6 in the bloodstream can influence the local redox homeostasis, inducing the establishment of prooxidative conditions in skeletal muscle tissue.
Collapse
|
33
|
De Spiegeleer A, Elewaut D, Van Den Noortgate N, Janssens Y, Debunne N, Van Langenhove S, Govindarajan S, De Spiegeleer B, Wynendaele E. WITHDRAWN: This article has been withdrawn. Biochim Biophys Acta Mol Basis Dis 2019:165585. [PMID: 31678164 DOI: 10.1016/j.bbadis.2019.165585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 09/11/2019] [Accepted: 09/22/2019] [Indexed: 11/21/2022]
Abstract
This article has been withdrawn at the request of the author for administrative reasons. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Anton De Spiegeleer
- Department of Geriatrics, Faculty of Medicine and Health Sciences, Ghent University Hospital, C. Heymanslaan 10, 9000 Ghent, Belgium; Drug Quality and Registration (DruQuaR) group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Unit for Molecular Immunology and Inflammation, VIB-Center for Inflammation Research, Technologiepark 71, 9052, Zwijnaarde, Ghent, Belgium
| | - Dirk Elewaut
- Unit for Molecular Immunology and Inflammation, VIB-Center for Inflammation Research, Technologiepark 71, 9052, Zwijnaarde, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University Hospital, C. Heymanslaan 10, 9000 Ghent, Belgium
| | - Nele Van Den Noortgate
- Department of Geriatrics, Faculty of Medicine and Health Sciences, Ghent University Hospital, C. Heymanslaan 10, 9000 Ghent, Belgium
| | - Yorick Janssens
- Drug Quality and Registration (DruQuaR) group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Nathan Debunne
- Drug Quality and Registration (DruQuaR) group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Selien Van Langenhove
- Drug Quality and Registration (DruQuaR) group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Srinath Govindarajan
- Unit for Molecular Immunology and Inflammation, VIB-Center for Inflammation Research, Technologiepark 71, 9052, Zwijnaarde, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University Hospital, C. Heymanslaan 10, 9000 Ghent, Belgium
| | - Bart De Spiegeleer
- Drug Quality and Registration (DruQuaR) group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Evelien Wynendaele
- Drug Quality and Registration (DruQuaR) group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
34
|
Kanazashi M, Tanaka M, Nakanishi R, Maeshige N, Fujino H. Effects of astaxanthin supplementation and electrical stimulation on muscle atrophy and decreased oxidative capacity in soleus muscle during hindlimb unloading in rats. J Physiol Sci 2019; 69:757-767. [PMID: 31273678 PMCID: PMC10717927 DOI: 10.1007/s12576-019-00692-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 06/24/2019] [Indexed: 12/15/2022]
Abstract
The effects of a combination of the antioxidant astaxanthin (AX) and electrical stimulation (ES) on muscle mass and mitochondrial oxidative capacity were investigated in the soleus muscle of hindlimb unloaded rats. Five groups of male Sprague-Dawley rats were used; control, 1-week hindlimb unloading (HU), HU + AX, HU + ES, and HU + AX + ES. Respective rats in the AX groups received 50-mg/kg AX twice daily during HU. Calf muscles of rats in the ES groups were electrically stimulated for 240 s/day during HU. One-week HU decreased muscle mass along with decreased FoxO3a phosphorylation and increased ubiquitinated proteins expressions, decreased oxidative enzymatic activity accompanied with decline in PGC-1α protein expression, and increased reactive oxygen species production. However, the combination treatment could synergistically attenuate/suppress all HU-related changes, suggesting protective effects on muscle atrophy and decreased muscle oxidative capacity due to chronic neuromuscular inactivity.
Collapse
Affiliation(s)
- Miho Kanazashi
- Department of Physical Therapy, Faculty of Health and Welfare, Prefectural University of Hiroshima, 1-1 Gakuen-cho, Mihara-shi, Hiroshima, 723-0053, Japan
| | - Masayuki Tanaka
- Department of Physical Therapy, Faculty of Human Sciences, Osaka University of Human Sciences, 1-4-1 Shojaku, Settsu-shi, Osaka, 566-8501, Japan
| | - Ryosuke Nakanishi
- Department of Physical Therapy, Faculty of Rehabilitation, Kobe International University, 9-1-6 Koyocho-naka, Higashinada-ku, Kobe-shi, Hyogo, 658-0032, Japan
| | - Noriaki Maeshige
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe-shi, Hyogo, 654-0142, Japan
| | - Hidemi Fujino
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe-shi, Hyogo, 654-0142, Japan.
| |
Collapse
|
35
|
Sorrentino S, Barbiera A, Proietti G, Sica G, Adamo S, Scicchitano BM. Inhibition of Phosphoinositide 3-Kinase/Protein Kinase B Signaling Hampers the Vasopressin-dependent Stimulation of Myogenic Differentiation. Int J Mol Sci 2019; 20:ijms20174188. [PMID: 31461843 PMCID: PMC6747374 DOI: 10.3390/ijms20174188] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/18/2019] [Accepted: 08/24/2019] [Indexed: 01/09/2023] Open
Abstract
Arginine-vasopressin (AVP) promotes muscle differentiation, hypertrophy, and regeneration through the combined activation of the calcineurin and Calcium/Calmodulin-dependent Protein Kinase (CaMK) pathways. The AVP system is impaired in several neuromuscular diseases, suggesting that AVP may act as a physiological factor in skeletal muscle. Since the Phosphoinositide 3-kinases/Protein Kinase B/mammalian Target Of Rapamycin (PI3K/Akt/mTOR) signaling plays a significant role in regulating muscle mass, we evaluated its role in the AVP myogenic effect. In L6 cells AKT1 expression was knocked down, and the AVP-dependent expression of mTOR and Forkhead box O3 (FoxO) was analyzed by Western blotting. The effect of the PI3K inhibitor LY294002 was evaluated by cellular and molecular techniques. Akt knockdown hampered the AVP-dependent mTOR expression while increased the levels of FoxO transcription factor. LY294002 treatment inhibited the AVP-dependent expression of Myocyte Enhancer Factor-2 (MEF2) and myogenin and prevented the nuclear translocation of MEF2. LY294002 also repressed the AVP-dependent nuclear export of histone deacetylase 4 (HDAC4) interfering with the formation of multifactorial complexes on the myogenin promoter. We demonstrate that the PI3K/Akt pathway is essential for the full myogenic effect of AVP and that, by targeting this pathway, one may highlight novel strategies to counteract muscle wasting in aging or neuromuscular disorders.
Collapse
Affiliation(s)
- Silvia Sorrentino
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| | - Alessandra Barbiera
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| | - Gabriella Proietti
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| | - Gigliola Sica
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| | - Sergio Adamo
- Dipartimento di Scienze Anatomiche, Istologiche, Medico-legali e dell'Apparato Locomotore (SAIMLAL), Sezione di Istologia ed Embriologia Medica, Sapienza Università, via A. Scarpa 16, 00161 Roma, Italy.
| | - Bianca Maria Scicchitano
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| |
Collapse
|
36
|
Neuromuscular Junction as an Entity of Nerve-Muscle Communication. Cells 2019; 8:cells8080906. [PMID: 31426366 PMCID: PMC6721719 DOI: 10.3390/cells8080906] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 12/11/2022] Open
Abstract
One of the crucial systems severely affected in several neuromuscular diseases is the loss of effective connection between muscle and nerve, leading to a pathological non-communication between the two tissues. The neuromuscular junction (NMJ) represents the critical region at the level of which muscle and nerve communicate. Defects in signal transmission between terminal nerve endings and muscle membrane is a common feature of several physio-pathologic conditions including aging and Amyotrophic Lateral Sclerosis (ALS). Nevertheless, controversy exists on whether pathological events beginning at the NMJ precede or follow loss of motor units. In this review, the role of NMJ in the physio-pathologic interplay between muscle and nerve is discussed.
Collapse
|
37
|
Cui W, Liu CX, Zhang YC, Shen Q, Feng ZH, Wang J, Lu SF, Wu J, Li JX. A novel oleanolic acid derivative HA-19 ameliorates muscle atrophy via promoting protein synthesis and preventing protein degradation. Toxicol Appl Pharmacol 2019; 378:114625. [PMID: 31201822 DOI: 10.1016/j.taap.2019.114625] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/18/2019] [Accepted: 06/11/2019] [Indexed: 12/13/2022]
Abstract
Muscle atrophy refers to a decrease in the size of muscles in the body, occurs in certain muscles with inactivity in many diseases and lacks effective therapies up to date. Natural products still play an important role in drug discovery. In the present study, derivatives of a natural product, oleanolic acid, were screened with myoblast differentiation and myotube atrophy assays, respectively. Results revealed that one of the derivatives, HA-19 showed the most potent anti-muscle atrophy activity, and was used for further studies. We demonstrated that HA-19 led to the increase of the protein synthesis by activating mechanistic target of rapamycin complex 1 (mTORC1)/p70 S6K pathways, and also enhanced myoblast proliferation and terminal differentiation via up-regulating of the myogenic transcription factors Pax7, MyoD and Myogenin. The interesting thing was that HA-19 also suppressed protein degradation to prevent myotube atrophy by down-regulating negative growth factors, FoxO1, MuRF1 and Atrogin-1. The results were also supported by puromycin labelling and protein ubiquitination assays. These data revealed that HA-19 possessed a "dual effect" on inhibition of muscle atrophy. In disuse-induced muscle atrophy mice model, HA-19 treatment significantly increased the weights of bilateral tibialis anterior (TA), gastrocnemius (Gastroc.), quadriceps (Quad.), suggesting the effectiveness of HA-19 to remit disuse-induced muscle atrophy. Our finding demonstrated that HA-19 has a great potential as an inhibitor or lead compound for the anti-muscle atrophy drug discovery.
Collapse
Affiliation(s)
- Wei Cui
- State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Chen-Xi Liu
- State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yu-Chao Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Qi Shen
- State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Zhen-Hua Feng
- The Center of Diagnosis and Treatment for Joint Disease, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing 210008, China
| | - Jie Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Sheng-Feng Lu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jing Wu
- State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Jian-Xin Li
- State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
38
|
Forcina L, Miano C, Scicchitano BM, Musarò A. Signals from the Niche: Insights into the Role of IGF-1 and IL-6 in Modulating Skeletal Muscle Fibrosis. Cells 2019; 8:E232. [PMID: 30862132 PMCID: PMC6468756 DOI: 10.3390/cells8030232] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/06/2019] [Accepted: 03/06/2019] [Indexed: 12/16/2022] Open
Abstract
Muscle regeneration, characterized by the activation and proliferation of satellite cells and other precursors, is accompanied by an inflammatory response and the remodeling of the extracellular matrix (ECM), necessary to remove cellular debris and to mechanically support newly generated myofibers and activated satellite cells. Muscle repair can be considered concluded when the tissue architecture, vascularization, and innervation have been restored. Alterations in these connected mechanisms can impair muscle regeneration, leading to the replacement of functional muscle tissue with a fibrotic scar. In the present review, we will discuss the cellular mediators of fibrosis and how the altered expression and secretion of soluble mediators, such as IL-6 and IGF-1, can modulate regulatory networks involved in the altered regeneration and fibrosis during aging and diseases.
Collapse
Affiliation(s)
- Laura Forcina
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Via A. Scarpa, 14, 00161 Rome, Italy.
| | - Carmen Miano
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Via A. Scarpa, 14, 00161 Rome, Italy.
| | - Bianca Maria Scicchitano
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy.
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Via A. Scarpa, 14, 00161 Rome, Italy.
| |
Collapse
|
39
|
Forcina L, Miano C, Pelosi L, Musarò A. An Overview about the Biology of Skeletal Muscle Satellite Cells. Curr Genomics 2019; 20:24-37. [PMID: 31015789 PMCID: PMC6446479 DOI: 10.2174/1389202920666190116094736] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/19/2018] [Accepted: 01/08/2019] [Indexed: 12/14/2022] Open
Abstract
The peculiar ability of skeletal muscle tissue to operate adaptive changes during post-natal de-velopment and adulthood has been associated with the existence of adult somatic stem cells. Satellite cells, occupying an exclusive niche within the adult muscle tissue, are considered bona fide stem cells with both stem-like properties and myogenic activities. Indeed, satellite cells retain the capability to both maintain the quiescence in uninjured muscles and to be promptly activated in response to growth or re-generative signals, re-engaging the cell cycle. Activated cells can undergo myogenic differentiation or self-renewal moving back to the quiescent state. Satellite cells behavior and their fate decision are finely controlled by mechanisms involving both cell-autonomous and external stimuli. Alterations in these regu-latory networks profoundly affect muscle homeostasis and the dynamic response to tissue damage, con-tributing to the decline of skeletal muscle that occurs under physio-pathologic conditions. Although the clear myogenic activity of satellite cells has been described and their pivotal role in muscle growth and regeneration has been reported, a comprehensive picture of inter-related mechanisms guiding muscle stem cell activity has still to be defined. Here, we reviewed the main regulatory networks determining satellite cell behavior. In particular, we focused on genetic and epigenetic mechanisms underlining satel-lite cell maintenance and commitment. Besides intrinsic regulations, we reported current evidences about the influence of environmental stimuli, derived from other cell populations within muscle tissue, on satel-lite cell biology.
Collapse
Affiliation(s)
- Laura Forcina
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14 Rome 00161, Italy
| | - Carmen Miano
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14 Rome 00161, Italy
| | - Laura Pelosi
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14 Rome 00161, Italy
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14 Rome 00161, Italy
| |
Collapse
|