1
|
Lee J, Lee SH, Kim H, Chung SW. Effect of electrical muscle stimulation on the improvement of deltoid muscle atrophy in a rat shoulder immobilization model. J Orthop Res 2024; 42:2634-2645. [PMID: 39097824 DOI: 10.1002/jor.25943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/24/2024] [Accepted: 07/09/2024] [Indexed: 08/05/2024]
Abstract
Immobilization following trauma or surgery induces skeletal muscle atrophy, and improvement in the muscle atrophy is critical for successful clinical outcomes. The purpose of this study is to evaluate the effect of electrical muscle stimulation (EMS) on muscle atrophy. The study design is a controlled laboratory study. Eighty rats (56 to establish the deltoid muscle atrophy [DMA] model and 24 to evaluate the effect of EMS on the model) were used. DMA was induced by completely immobilizing the right shoulder of each rat by placing sutures between the scapula and humeral shaft, with the left shoulder as a control. After establishing the DMA model, rats were randomly assigned into three groups: low-frequency EMS (L-EMS, 10 Hz frequency), medium-frequency EMS (M-EMS, 50 Hz frequency), and control (eight rats per group). After 3 weeks, the deltoid muscles of each rat were harvested, alterations in gene expression and muscle cell size were evaluated, and immunohistochemical analysis was performed. DMA was most prominent 3 weeks after shoulder immobilization. Murf1 and Atrogin were significantly induced at the initial phase and gradually decreased at approximately 3 weeks; however, MyoD expressed an inverse relationship with Murf1 and Atrogin. IL6 expression was prominent at 1 week. The time point for the EMS effect evaluation was selected at 3 weeks, when the DMA was the most prominent with a change in relevant gene expression. The M-EMS group cell size was significantly larger than that of L-EMS and control group in both the immobilized and intact shoulders (all p < 0.05), without significant differences between the L-EMS and control groups. The M-EMS group showed significantly lower mRNA expressions of Murf1 and Atrogin and higher expressions of MyoD and Col1A1 than that of the control group (all p < 0.05). In immunohistochemical analysis, similar results were observed with lower Atrogin staining and higher MyoD and Col1A1 staining in the M-EMS group. DMA model was established by complete shoulder immobilization, with the most prominent muscle atrophy observed at 3 weeks. M-EMS improved DMA with changes in the expression of relevant genes. M-EMS might be a solution for strengthening atrophied skeletal muscles and facilitating rehabilitation after trauma or surgery.
Collapse
Affiliation(s)
- Jeongkun Lee
- Department of Orthopaedic Surgery, School of Medicine, Konkuk University, Seoul, Korea
| | - Su Hyun Lee
- Department of Orthopaedic Surgery, School of Medicine, Konkuk University, Seoul, Korea
| | - Hyuntae Kim
- Department of Orthopaedic Surgery, School of Medicine, Konkuk University, Seoul, Korea
| | - Seok Won Chung
- Department of Orthopaedic Surgery, School of Medicine, Konkuk University, Seoul, Korea
| |
Collapse
|
2
|
Choi S, Lee MJ, Kim M, Bae Y, Park JU, Cho SW. Durable Muscle Extracellular Matrix Engineered with Adhesive Phenolic Moieties for Effective Skeletal Muscle Regeneration in Muscle Atrophy. Adv Healthc Mater 2024; 13:e2401826. [PMID: 39420690 DOI: 10.1002/adhm.202401826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/20/2024] [Indexed: 10/19/2024]
Abstract
Muscle atrophy detrimentally impacts health and exacerbates physical disability, leading to increased mortality. In particular, sarcopenia, aging-related degenerative muscle loss, necessitates urgent remedies. Current approaches for treating muscle atrophy include exercise and nutrition, while drug exploration remains in its early stages. Cell therapy, focusing on satellite cells, faces significant challenge due to poor engraftment, safety issue, and high cost. Cell-free approach using extracellular matrix (ECM) shows a regenerative potential, but a lack of mechanical and adhesive properties hinders prolonged efficacy of ECM therapy. Here, durable muscle ECM (MEM) hydrogels for muscle atrophy by fortifying MEM with adhesive phenolic moieties including catechol and pyrogallol are demonstrated. The resultant phenolic MEM hydrogels exhibit enhanced mechanical and adhesive properties and provide sustained muscle-like microenvironments to address muscle atrophy. No local and systemic toxicities are observed after phenolic MEM injection into tibialis anterior muscle. Notably, these engineered MEM hydrogels, devoid of cells or drugs, induce tissue rejuvenation by promoting muscle protein synthesis and facilitating functional muscle recovery in mouse models of disuse- and age-induced atrophy. This study introduces cell-free, ECM-based therapeutics with translational potential for muscle atrophy by reversing muscle loss and restoring function.
Collapse
Affiliation(s)
- Soojeong Choi
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- CellArtgen Inc., Seoul, 03722, Republic of Korea
| | - Mi Jeong Lee
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Moohyun Kim
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yunsu Bae
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jang-Ung Park
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- CellArtgen Inc., Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
| |
Collapse
|
3
|
Bellanti F, Lo Buglio A, Pannone G, Pedicillo MC, De Stefano IS, Pignataro A, Capurso C, Vendemiale G. An Amino Acid Mixture to Counteract Skeletal Muscle Atrophy: Impact on Mitochondrial Bioenergetics. Int J Mol Sci 2024; 25:6056. [PMID: 38892242 PMCID: PMC11173258 DOI: 10.3390/ijms25116056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/22/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Skeletal muscle atrophy (SMA) is caused by a rise in muscle breakdown and a decline in protein synthesis, with a consequent loss of mass and function. This study characterized the effect of an amino acid mixture (AA) in models of SMA, focusing on mitochondria. C57/Bl6 mice underwent immobilization of one hindlimb (I) or cardiotoxin-induced muscle injury (C) and were compared with controls (CTRL). Mice were then administered AA in drinking water for 10 days and compared to a placebo group. With respect to CTRL, I and C reduced running time and distance, along with grip strength; however, the reduction was prevented by AA. Tibialis anterior (TA) muscles were used for histology and mitochondria isolation. I and C resulted in TA atrophy, characterized by a reduction in both wet weight and TA/body weight ratio and smaller myofibers than those of CTRL. Interestingly, these alterations were lightly observed in mice treated with AA. The mitochondrial yield from the TA of I and C mice was lower than that of CTRL but not in AA-treated mice. AA also preserved mitochondrial bioenergetics in TA muscle from I and C mice. To conclude, this study demonstrates that AA prevents loss of muscle mass and function in SMA by protecting mitochondria.
Collapse
Affiliation(s)
- Francesco Bellanti
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (F.B.); (A.L.B.); (C.C.)
| | - Aurelio Lo Buglio
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (F.B.); (A.L.B.); (C.C.)
| | - Giuseppe Pannone
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (G.P.); (M.C.P.); (I.S.D.S.); (A.P.)
| | - Maria Carmela Pedicillo
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (G.P.); (M.C.P.); (I.S.D.S.); (A.P.)
| | - Ilenia Sara De Stefano
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (G.P.); (M.C.P.); (I.S.D.S.); (A.P.)
| | - Angela Pignataro
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (G.P.); (M.C.P.); (I.S.D.S.); (A.P.)
| | - Cristiano Capurso
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (F.B.); (A.L.B.); (C.C.)
| | - Gianluigi Vendemiale
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (F.B.); (A.L.B.); (C.C.)
| |
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
|
Sayed RKA, Hibbert JE, Jorgenson KW, Hornberger TA. The Structural Adaptations That Mediate Disuse-Induced Atrophy of Skeletal Muscle. Cells 2023; 12:2811. [PMID: 38132132 PMCID: PMC10741885 DOI: 10.3390/cells12242811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/07/2023] [Accepted: 12/09/2023] [Indexed: 12/23/2023] Open
Abstract
The maintenance of skeletal muscle mass plays a fundamental role in health and issues associated with quality of life. Mechanical signals are one of the most potent regulators of muscle mass, with a decrease in mechanical loading leading to a decrease in muscle mass. This concept has been supported by a plethora of human- and animal-based studies over the past 100 years and has resulted in the commonly used term of 'disuse atrophy'. These same studies have also provided a great deal of insight into the structural adaptations that mediate disuse-induced atrophy. For instance, disuse results in radial atrophy of fascicles, and this is driven, at least in part, by radial atrophy of the muscle fibers. However, the ultrastructural adaptations that mediate these changes remain far from defined. Indeed, even the most basic questions, such as whether the radial atrophy of muscle fibers is driven by the radial atrophy of myofibrils and/or myofibril hypoplasia, have yet to be answered. In this review, we thoroughly summarize what is known about the macroscopic, microscopic, and ultrastructural adaptations that mediated disuse-induced atrophy and highlight some of the major gaps in knowledge that need to be filled.
Collapse
Affiliation(s)
- Ramy K. A. Sayed
- Department of Comparative Biosciences, University of Wisconsin—Madison, Madison, WI 53706, USA; (R.K.A.S.); (J.E.H.); (K.W.J.)
- School of Veterinary Medicine, University of Wisconsin—Madison, Madison, WI 53706, USA
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt
| | - Jamie E. Hibbert
- Department of Comparative Biosciences, University of Wisconsin—Madison, Madison, WI 53706, USA; (R.K.A.S.); (J.E.H.); (K.W.J.)
- School of Veterinary Medicine, University of Wisconsin—Madison, Madison, WI 53706, USA
| | - Kent W. Jorgenson
- Department of Comparative Biosciences, University of Wisconsin—Madison, Madison, WI 53706, USA; (R.K.A.S.); (J.E.H.); (K.W.J.)
- School of Veterinary Medicine, University of Wisconsin—Madison, Madison, WI 53706, USA
| | - Troy A. Hornberger
- Department of Comparative Biosciences, University of Wisconsin—Madison, Madison, WI 53706, USA; (R.K.A.S.); (J.E.H.); (K.W.J.)
- School of Veterinary Medicine, University of Wisconsin—Madison, Madison, WI 53706, USA
| |
Collapse
|
6
|
Teixeira VON, Bartikoski BJ, do Espirito Santo RC, Alabarse PVG, Ghannan K, Silva JMS, Filippin LI, Visioli F, Martinez-Gamboa L, Feist E, Xavier RM. The role of proteasome in muscle wasting of experimental arthritis. Adv Rheumatol 2023; 63:14. [PMID: 36949513 DOI: 10.1186/s42358-023-00292-5] [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: 01/31/2022] [Accepted: 03/05/2023] [Indexed: 03/24/2023] Open
Abstract
BACKGROUND Rheumatoid arthritis is an autoimmune inflammatory disease that often leads patients to muscle impairment and physical disability. This study aimed to evaluate changes in the activity of proteasome system in skeletal muscles of mice with collagen-induced arthritis (CIA) and treated with etanercept or methotrexate. METHODS Male DBA1/J mice were divided into four groups (n = 8 each): CIA-Vehicle (treated with saline), CIA-ETN (treated with etanercept, 5.5 mg/kg), CIA-MTX (treated with methotrexate, 35 mg/kg) and CO (healthy control group). Mice were treated two times a week for 6 weeks. Clinical score and hind paw edema were measured. Muscles were weighted after euthanasia and used to quantify proteasome activity, gene (MuRF-1, PMSα4, PSMβ5, PMSβ6, PSMβ7, PSMβ8, PSMβ9, and PSMβ10), and protein (PSMβ1, PSMβ5, PSMβ1i, PSMβ5i) expression of proteasome subunits. RESULTS Both treatments slowed disease development, but only CIA-ETN maintained muscle weight compared to CIA-MTX and CIA-Vehicle groups. Etanercept treatment showed caspase-like activity of 26S proteasome similar to CO group, while CIA-Vehicle and CIA-MTX had higher activity compared to CO group (p: 0.0057). MuRF-1 mRNA expression was decreased after etanercept administration compared to CIA-Vehicle and CO groups (p: 0.002, p: 0.007, respectively). PSMβ8 and PSMβ9 mRNA levels were increased in CIA-Vehicle and CIA-MTX compared to CO group, while CIA-ETN presented no difference from CO. PMSβ6 mRNA expression was higher in CIA-Vehicle and CIA-MTX groups than in CO group. Protein levels of the PSMβ5 subunit were increased in CO group compared to CIA-Vehicle; after both etanercept and methotrexate treatments, PSMβ5 expression was higher than in CIA-Vehicle group and did not differ from CO group expression (p: 0.0025, p: 0.001, respectively). The inflammation-induced subunit β1 (LMP2) was enhanced after methotrexate treatment compared to CO group (p: 0.043). CONCLUSIONS The results of CIA-Vehicle show that arthritis increases muscle proteasome activation by enhanced caspase-like activity of 26S proteasome and increased PSMβ8 and PSMβ9 mRNA levels. Etanercept treatment was able to maintain the muscle weight and to modulate proteasome so that its activity and gene expression were compared to CO after TNF inhibition. The protein expression of inflammation-induced proteasome subunit was increased in muscle of CIA-MTX group but not following etanercept treatment. Thus, anti-TNF treatment may be an interesting approach to attenuate the arthritis-related muscle wasting.
Collapse
Affiliation(s)
- Vivian Oliveira Nunes Teixeira
- Medical Sciences Program, Medicine Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratório de Doenças Autoimunes, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos Street, Santa Cecília, Porto Alegre, 2350, Brazil
| | - Bárbara Jonson Bartikoski
- Medical Sciences Program, Medicine Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratório de Doenças Autoimunes, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos Street, Santa Cecília, Porto Alegre, 2350, Brazil
| | - Rafaela Cavalheiro do Espirito Santo
- Medical Sciences Program, Medicine Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
- Laboratório de Doenças Autoimunes, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos Street, Santa Cecília, Porto Alegre, 2350, Brazil.
| | - Paulo Vinícius Gil Alabarse
- Medical Sciences Program, Medicine Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratório de Doenças Autoimunes, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos Street, Santa Cecília, Porto Alegre, 2350, Brazil
- University of California San Diego Medical Center Library, University of California San Diego School of Medicine, San Diego, USA
| | - Khetam Ghannan
- Schwerpunkt Rheumatologie und Klinische Immunologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jordana Miranda Souza Silva
- Medical Sciences Program, Medicine Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratório de Doenças Autoimunes, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos Street, Santa Cecília, Porto Alegre, 2350, Brazil
| | - Lidiane Isabel Filippin
- Laboratório de Doenças Autoimunes, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos Street, Santa Cecília, Porto Alegre, 2350, Brazil
- Health and Human Development Department, Universidade La Salle, Canoas, Brazil
| | - Fernanda Visioli
- Patology Department, Faculdade de Odontologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lorena Martinez-Gamboa
- Schwerpunkt Rheumatologie und Klinische Immunologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Eugen Feist
- Schwerpunkt Rheumatologie und Klinische Immunologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ricardo Machado Xavier
- Medical Sciences Program, Medicine Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratório de Doenças Autoimunes, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos Street, Santa Cecília, Porto Alegre, 2350, Brazil
| |
Collapse
|
7
|
Ghanemi A, Yoshioka M, St-Amand J. Secreted Protein Acidic and Rich in Cysteine ( SPARC)-Mediated Exercise Effects: Illustrative Molecular Pathways against Various Diseases. Diseases 2023; 11:diseases11010033. [PMID: 36810547 PMCID: PMC9944512 DOI: 10.3390/diseases11010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
The strong benefits of exercise, in addition to the development of both the therapeutic applications of physical activity and molecular biology tools, means that it has become very important to explore the underlying molecular patterns linking exercise and its induced phenotypic changes. Within this context, secreted protein acidic and rich in cysteine (SPARC) has been characterized as an exercise-induced protein that would mediate and induce some important effects of exercise. Herein, we suggest some underlying pathways to explain such SPARC-induced exercise-like effects. Such mechanistic mapping would not only allow us to understand the molecular processes of exercise and SPARC effects but would also highlight the potential to develop novel molecular therapies. These therapies would be based on mimicking the exercise benefits via either introducing SPARC or pharmacologically targeting the SPARC-related pathways to produce exercise-like effects. This is of a particular importance for those who do not have the ability to perform the required physical activity due to disabilities or diseases. The main objective of this work is to highlight selected potential therapeutic applications deriving from SPARC properties that have been reported in various publications.
Collapse
Affiliation(s)
- Abdelaziz Ghanemi
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Quebec, QC G1V 4G2, Canada
| | - Mayumi Yoshioka
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Quebec, QC G1V 4G2, Canada
| | - Jonny St-Amand
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Quebec, QC G1V 4G2, Canada
- Correspondence: ; Tel.: +1-(418)-654-2296; Fax: +1-(418)-654-2761
| |
Collapse
|
8
|
Alcohol Induces Zebrafish Skeletal Muscle Atrophy through HMGB1/TLR4/NF-κB Signaling. LIFE (BASEL, SWITZERLAND) 2022; 12:life12081211. [PMID: 36013390 PMCID: PMC9410481 DOI: 10.3390/life12081211] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/04/2022] [Accepted: 08/06/2022] [Indexed: 11/17/2022]
Abstract
Excessive alcohol consumption can cause alcoholic myopathy, but the molecular mechanism is still unclear. In this study, zebrafish were exposed to 0.5% alcohol for eight weeks to investigate the effect of alcohol on skeletal muscle and its molecular mechanism. The results showed that the body length, body weight, cross-sectional area of the skeletal muscle fibers, Ucrit, and MO2max of the zebrafish were significantly decreased after alcohol exposure. The expression of markers of skeletal muscle atrophy and autophagy was increased, and the expression of P62 was significantly reduced. The content of ROS, the mRNA expression of sod1 and sod2, and the protein expression of Nox2 were significantly increased. In addition, we found that the inflammatory factors Il1β and Tnfα were significantly enriched in skeletal muscle, and the expression of the HMGB1/TLR4/NF-κB signaling axis was also significantly increased. In summary, in this study, we established a zebrafish model of alcohol-induced skeletal muscle atrophy and further elucidated its pathogenesis.
Collapse
|
9
|
Ghanemi A, Yoshioka M, St-Amand J. Secreted Protein Acidic and Rich in Cysteine as an Exercise-Induced Gene: Towards Novel Molecular Therapies for Immobilization-Related Muscle Atrophy in Elderly Patients. Genes (Basel) 2022; 13:1014. [PMID: 35741776 PMCID: PMC9223229 DOI: 10.3390/genes13061014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/06/2022] [Accepted: 06/01/2022] [Indexed: 02/01/2023] Open
Abstract
Long periods of immobilization, among other etiologies, would result is muscle atrophy. Exercise is the best approach to reverse this atrophy. However, the limited or the non-ability to perform the required physical activity for such patients and the limited pharmacological options make developing novel therapeutic approaches a necessity. Within this context, secreted protein acidic and rich in cysteine (SPARC) has been characterized as an exercise-induced gene. Whereas the knock-out of this gene leads to a phenotype that mimics number of the ageing-induced and sarcopenia-related changes including muscle atrophy, overexpressing SPARC in mice or adding it to muscular cell culture produces similar effects as exercise including enhanced muscle mass, strength and metabolism. Therefore, this piece of writing aims to provide evidence supporting the potential use of SPARC/SPARC as a molecular therapy for muscle atrophy in the context of immobilization especially for elderly patients.
Collapse
Affiliation(s)
- Abdelaziz Ghanemi
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada;
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Quebec, QC G1V 4G2, Canada;
| | - Mayumi Yoshioka
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Quebec, QC G1V 4G2, Canada;
| | - Jonny St-Amand
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada;
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Quebec, QC G1V 4G2, Canada;
| |
Collapse
|
10
|
Parker E, Khayrullin A, Kent A, Mendhe B, Youssef El Baradie KB, Yu K, Pihkala J, Liu Y, McGee-Lawrence M, Johnson M, Chen J, Hamrick M. Hindlimb Immobilization Increases IL-1β and Cdkn2a Expression in Skeletal Muscle Fibro-Adipogenic Progenitor Cells: A Link Between Senescence and Muscle Disuse Atrophy. Front Cell Dev Biol 2022; 9:790437. [PMID: 35047502 PMCID: PMC8762295 DOI: 10.3389/fcell.2021.790437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/13/2021] [Indexed: 11/26/2022] Open
Abstract
Loss of muscle mass and strength contributes to decreased independence and an increased risk for morbidity and mortality. A better understanding of the cellular and molecular mechanisms underlying muscle atrophy therefore has significant clinical and therapeutic implications. Fibro-adipogenic progenitors (FAPs) are a skeletal muscle resident stem cell population that have recently been shown to play vital roles in muscle regeneration and muscle hypertrophy; however, the role that these cells play in muscle disuse atrophy is not well understood. We investigated the role of FAPs in disuse atrophy in vivo utilizing a 2-week single hindlimb immobilization model. RNA-seq was performed on FAPs isolated from the immobilized and non-immobilized limb. The RNAseq data show that IL-1β is significantly upregulated in FAPs following 2 weeks of immobilization, which we confirmed using droplet-digital PCR (ddPCR). We further validated the RNA-seq and ddPCR data from muscle in situ using RNAscope technology. IL-1β is recognized as a key component of the senescence-associated secretory phenotype, or SASP. We then tested the hypothesis that FAPs from the immobilized limb would show elevated senescence measured by cyclin-dependent kinase inhibitor 2A (Cdkn2a) expression as a senescence marker. The ddPCR and RNAscope data both revealed increased Cdkn2a expression in FAPs with immobilization. These data suggest that the gene expression profile of FAPs is significantly altered with disuse, and that disuse itself may drive senescence in FAPs further contributing to muscle atrophy.
Collapse
Affiliation(s)
- Emily Parker
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Andrew Khayrullin
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Andrew Kent
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Bharati Mendhe
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Khairat Bahgat Youssef El Baradie
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States.,Faculty of Science, Tanta University, Tanta, Egypt
| | - Kanglun Yu
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Jeanene Pihkala
- Flow Cytometry Core Facility Research Laboratory Director, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Meghan McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Maribeth Johnson
- Division of Biostatistics and Data Science, DPHS, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Jie Chen
- Division of Biostatistics and Data Science, DPHS, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Mark Hamrick
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
11
|
Yoon JH, Lee SM, Lee Y, Kim MJ, Yang JW, Choi JY, Kwak JY, Lee KP, Yang YR, Kwon KS. Alverine citrate promotes myogenic differentiation and ameliorates muscle atrophy. Biochem Biophys Res Commun 2022; 586:157-162. [PMID: 34847441 DOI: 10.1016/j.bbrc.2021.11.076] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/18/2021] [Indexed: 11/25/2022]
Abstract
Sarcopenia is the age-related loss of muscle mass and function and no pharmacological medication has been approved for its treatment. We established an atrogin-1/MAFbx promoter assay to find drug candidates that inhibit myotube atrophy. Alverine citrate (AC) was identified using high-throughput screening of an existing drug library. AC is an established medicine for stomach and intestinal spasms. AC treatment increased myotube diameter and inhibited atrophy signals induced by either C26-conditioned medium or dexamethasone in cultured C2C12 myoblasts. AC also enhanced myoblast fusion through the upregulation of fusion-related genes during C2C12 myoblast differentiation. Oral administration of AC improves muscle mass and physical performance in aged mice, as well as hindlimb-disused mice. Taken together, our data suggest that AC may be a novel therapeutic candidate for improving muscle weakness, including sarcopenia.
Collapse
Affiliation(s)
- Jong Hyeon Yoon
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Seung-Min Lee
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | | | - Min Ju Kim
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Jae Won Yang
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Jeong Yi Choi
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Ju Yeon Kwak
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Kwang-Pyo Lee
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Yong Ryoul Yang
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Ki-Sun Kwon
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea; Aventi Inc., Daejeon, Republic of Korea.
| |
Collapse
|
12
|
Molecular and Metabolic Mechanism of Low-Intensity Pulsed Ultrasound Improving Muscle Atrophy in Hindlimb Unloading Rats. Int J Mol Sci 2021; 22:ijms222212112. [PMID: 34829990 PMCID: PMC8625684 DOI: 10.3390/ijms222212112] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/31/2021] [Accepted: 11/02/2021] [Indexed: 12/16/2022] Open
Abstract
Low-intensity pulsed ultrasound (LIPUS) has been proved to promote the proliferation of myoblast C2C12. However, whether LIPUS can effectively prevent muscle atrophy has not been clarified, and if so, what is the possible mechanism. The aim of this study is to evaluate the effects of LIPUS on muscle atrophy in hindlimb unloading rats, and explore the mechanisms. The rats were randomly divided into four groups: normal control group (NC), hindlimb unloading group (UL), hindlimb unloading plus 30 mW/cm2 LIPUS irradiation group (UL + 30 mW/cm2), hindlimb unloading plus 80 mW/cm2 LIPUS irradiation group (UL + 80 mW/cm2). The tails of rats in hindlimb unloading group were suspended for 28 days. The rats in the LIPUS treated group were simultaneously irradiated with LIPUS on gastrocnemius muscle in both lower legs at the sound intensity of 30 mW/cm2 or 80 mW/cm2 for 20 min/d for 28 days. C2C12 cells were exposed to LIPUS at 30 or 80 mW/cm2 for 5 days. The results showed that LIPUS significantly promoted the proliferation and differentiation of myoblast C2C12, and prevented the decrease of cross-sectional area of muscle fiber and gastrocnemius mass in hindlimb unloading rats. LIPUS also significantly down regulated the expression of MSTN and its receptors ActRIIB, and up-regulated the expression of Akt and mTOR in gastrocnemius muscle of hindlimb unloading rats. In addition, three metabolic pathways (phenylalanine, tyrosine and tryptophan biosynthesis; alanine, aspartate and glutamate metabolism; glycine, serine and threonine metabolism) were selected as important metabolic pathways for hindlimb unloading effect. However, LIPUS promoted the stability of alanine, aspartate and glutamate metabolism pathway. These results suggest that the key mechanism of LIPUS in preventing muscle atrophy induced by hindlimb unloading may be related to promoting protein synthesis through MSTN/Akt/mTOR signaling pathway and stabilizing alanine, aspartate and glutamate metabolism.
Collapse
|
13
|
Singh A, Yadav A, Phogat J, Dabur R. Dynamics of autophagy and ubiquitin proteasome system coordination and interplay in skeletal muscle atrophy. Curr Mol Pharmacol 2021; 15:475-486. [PMID: 34365963 DOI: 10.2174/1874467214666210806163851] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/26/2021] [Accepted: 04/05/2021] [Indexed: 11/22/2022]
Abstract
Skeletal muscles are considered the largest reservoirs of the protein pool in the body and are critical for the maintenances of body homeostasis. Skeletal muscle atrophy is supported by various physiopathological conditions that lead to loss of muscle mass and contractile capacity of the skeletal muscle. Lysosomal mediated autophagy and ubiquitin-proteasomal system (UPS) concede the major intracellular systems of muscle protein degradation that result in the loss of mass and strength. Both systems recognize ubiquitination as a signal of degradation through different mechanisms, a sign of dynamic interplay between systems. Hence, growing shreds of evidence suggest the interdependency of autophagy and UPS in the progression of skeletal muscle atrophy under various pathological conditions. Therefore, understanding the molecular dynamics as well associated factors responsible for their interdependency is a necessity for the new therapeutic insights to counteract the muscle loss. Based on current literature, the present review summarizes the factors interplay in between the autophagy and UPS in favor of enhanced proteolysis of skeletal muscle and how they affect the anabolic signaling pathways under various conditions of skeletal muscle atrophy.
Collapse
Affiliation(s)
- Ajay Singh
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak-124001, Haryana. India
| | - Aarti Yadav
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak-124001, Haryana. India
| | - Jatin Phogat
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak-124001, Haryana. India
| | - Rajesh Dabur
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak-124001, Haryana. India
| |
Collapse
|
14
|
Marzuca-Nassr GN, Kuwabara WMT, Vitzel KF, Murata GM, Torres RP, Mancini-Filho J, Alba-Loureiro TC, Curi R. Endoplasmic Reticulum Stress and Autophagy Markers in Soleus Muscle Disuse-Induced Atrophy of Rats Treated with Fish Oil. Nutrients 2021; 13:nu13072298. [PMID: 34371808 PMCID: PMC8308346 DOI: 10.3390/nu13072298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/13/2022] Open
Abstract
Endoplasmic reticulum stress (ERS) and autophagy pathways are implicated in disuse muscle atrophy. The effects of high eicosapentaenoic (EPA) or high docosahexaenoic (DHA) fish oils on soleus muscle ERS and autophagy markers were investigated in a rat hindlimb suspension (HS) atrophy model. Adult Wistar male rats received daily by gavage supplementation (0.3 mL per 100 g b.w.) of mineral oil or high EPA or high DHA fish oils (FOs) for two weeks. Afterward, the rats were subjected to HS and the respective treatments concomitantly for an additional two-week period. After four weeks, we evaluated ERS and autophagy markers in the soleus muscle. Results were analyzed using two-way analysis of variance (ANOVA) and Bonferroni post hoc test. Gastrocnemius muscle ω-6/ω-3 fatty acids (FAs) ratio was decreased by both FOs indicating the tissue incorporation of omega-3 fatty acids. HS altered (p < 0.05) the protein content (decreasing total p38 and BiP and increasing p-JNK2/total JNK2 ratio, and caspase 3) and gene expressions (decreasing BiP and increasing IRE1 and PERK) of ERS and autophagy (decreasing Beclin and increasing LC3 and ATG14) markers in soleus. Both FOs attenuated (p < 0.05) the increase in PERK and ATG14 expressions induced by HS. Thus, both FOs could potentially attenuate ERS and autophagy in skeletal muscles undergoing atrophy.
Collapse
Affiliation(s)
- Gabriel Nasri Marzuca-Nassr
- Department of Internal Medicine, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (W.M.T.K.); (K.F.V.); (T.C.A.-L.); (R.C.)
- Correspondence: ; Tel.: +56-45-2596713
| | - Wilson Mitsuo Tatagiba Kuwabara
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (W.M.T.K.); (K.F.V.); (T.C.A.-L.); (R.C.)
| | - Kaio Fernando Vitzel
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (W.M.T.K.); (K.F.V.); (T.C.A.-L.); (R.C.)
- School of Health Sciences, College of Health, Massey University, Auckland 0745, New Zealand
| | - Gilson Masahiro Murata
- Nephrology Division, Medical Investigation Laboratory-29 (LIM-29), Medical School, University of São Paulo (FM-USP), São Paulo 01246-903, Brazil;
| | - Rosângela Pavan Torres
- Department of Lipids Laboratory, Food Science & Nutrition, Faculty of Pharmaceutical Science, University of São Paulo, São Paulo 05508-000, Brazil; (R.P.T.); (J.M.-F.)
| | - Jorge Mancini-Filho
- Department of Lipids Laboratory, Food Science & Nutrition, Faculty of Pharmaceutical Science, University of São Paulo, São Paulo 05508-000, Brazil; (R.P.T.); (J.M.-F.)
| | - Tatiana Carolina Alba-Loureiro
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (W.M.T.K.); (K.F.V.); (T.C.A.-L.); (R.C.)
| | - Rui Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (W.M.T.K.); (K.F.V.); (T.C.A.-L.); (R.C.)
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo 01506-000, Brazil
- Butantan Institute, São Paulo 05508-040, Brazil
| |
Collapse
|
15
|
Memme JM, Slavin M, Moradi N, Hood DA. Mitochondrial Bioenergetics and Turnover during Chronic Muscle Disuse. Int J Mol Sci 2021; 22:ijms22105179. [PMID: 34068411 PMCID: PMC8153634 DOI: 10.3390/ijms22105179] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022] Open
Abstract
Periods of muscle disuse promote marked mitochondrial alterations that contribute to the impaired metabolic health and degree of atrophy in the muscle. Thus, understanding the molecular underpinnings of muscle mitochondrial decline with prolonged inactivity is of considerable interest. There are translational applications to patients subjected to limb immobilization following injury, illness-induced bed rest, neuropathies, and even microgravity. Studies in these patients, as well as on various pre-clinical rodent models have elucidated the pathways involved in mitochondrial quality control, such as mitochondrial biogenesis, mitophagy, fission and fusion, and the corresponding mitochondrial derangements that underlie the muscle atrophy that ensues from inactivity. Defective organelles display altered respiratory function concurrent with increased accumulation of reactive oxygen species, which exacerbate myofiber atrophy via degradative pathways. The preservation of muscle quality and function is critical for maintaining mobility throughout the lifespan, and for the prevention of inactivity-related diseases. Exercise training is effective in preserving muscle mass by promoting favourable mitochondrial adaptations that offset the mitochondrial dysfunction, which contributes to the declines in muscle and whole-body metabolic health. This highlights the need for further investigation of the mechanisms in which mitochondria contribute to disuse-induced atrophy, as well as the specific molecular targets that can be exploited therapeutically.
Collapse
Affiliation(s)
| | | | | | - David A. Hood
- Correspondence: ; Tel.: +1-(416)-736-2100 (ext. 66640)
| |
Collapse
|
16
|
Lee EJ, Neppl RL. Influence of Age on Skeletal Muscle Hypertrophy and Atrophy Signaling: Established Paradigms and Unexpected Links. Genes (Basel) 2021; 12:genes12050688. [PMID: 34063658 PMCID: PMC8147613 DOI: 10.3390/genes12050688] [Citation(s) in RCA: 3] [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: 04/02/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/16/2022] Open
Abstract
Skeletal muscle atrophy in an inevitable occurrence with advancing age, and a consequence of disease including cancer. Muscle atrophy in the elderly is managed by a regimen of resistance exercise and increased protein intake. Understanding the signaling that regulates muscle mass may identify potential therapeutic targets for the prevention and reversal of muscle atrophy in metabolic and neuromuscular diseases. This review covers the major anabolic and catabolic pathways that regulate skeletal muscle mass, with a focus on recent progress and potential new players.
Collapse
|
17
|
Chaweewannakorn C, Harada T, Nyasha MR, Koide M, Shikama Y, Hagiwara Y, Sasaki K, Kanzaki M, Tsuchiya M. Imaging of muscle activity-induced morphometric changes in fibril network of myofascia by two-photon microscopy. J Anat 2021; 238:515-526. [PMID: 33078407 PMCID: PMC7855069 DOI: 10.1111/joa.13339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 08/10/2020] [Accepted: 09/28/2020] [Indexed: 01/15/2023] Open
Abstract
Myofascia, deep fascia enveloping skeletal muscles, consists of abundant collagen and elastin fibres that play a key role in the transmission of muscular forces. However, understanding of biomechanical dynamics in myofascia remains very limited due to less quantitative and relevant approaches for in vivo examination. The purpose of this study was to evaluate the myofascial fibril structure by means of a quantitative approach using two-photon microscopy (TPM) imaging in combination with intravital staining of Evans blue dye (EBD), a far-red fluorescence dye, which potentially labels elastin. With focus on myofascia of the tibial anterior (TA) muscle, the fibril structure intravitally stained with EBD was observed at the depth level of collagen fibrous membrane above the muscle belly. The EBD-labelled fibril structure and orientation in myofascia indicated biomechanical responses to muscle activity and ageing. The orientation histograms of EBD-labelled fibrils were significantly modified depending upon the intensity of muscle activity and ageing. Moreover, the density of EBD-labelled fibrils in myofascia decreased with habitual exercise but increased with muscle immobilization or ageing. In particular, the diameter of EBD-labelled fibrils in aged mice was significantly higher. The orientation histograms of EBD-labelled fibrils after habitual exercise, muscle immobilization and ageing showed significant differences compared to control. Indeed, the histograms in bilateral TA myofascia of exercise mice made simple waveforms without multiple sharp peaks, whilst muscular immobilization or ageing significantly shifted a histogram with sustaining multiple sharp peaks. Therefore, the dynamics of fibre network with EBD fluorescence in response to the biomechanical environment possibly indicate functional tissue adaptation in myofascia. Furthermore, on the basis of the knowledge that neutrophil recruitment occurs locally in working muscles, we suggested the unique reconstruction mechanism involving neutrophilic elastase in the myofascial fibril structure. In addition to the elastolytic susceptibility of EBD-labelled fibrils, distinct immunoreactivities and activities of neutrophil elastase in the myofascia were observed after electric pulse stimulation-induced muscle contraction for 15 min. Our findings of EBD-labelled fibril dynamics in myofascia through quantitative approach using TPM imaging and intravital fluorescence labelling potentially brings new insights to examine muscle physiology and pathology.
Collapse
Affiliation(s)
- Chayanit Chaweewannakorn
- Division of Advanced Prosthetic DentistryGraduate School of DentistryTohoku UniversitySendaiJapan
- Graduate School of Biomedical EngineeringTohoku UniversitySendaiJapan
| | - Takashi Harada
- Department of Orthopaedic SurgeryGraduate School of MedicineTohoku UniversitySendaiJapan
| | - Mazvita R. Nyasha
- Graduate School of Biomedical EngineeringTohoku UniversitySendaiJapan
| | - Masashi Koide
- Department of Orthopaedic SurgeryGraduate School of MedicineTohoku UniversitySendaiJapan
| | - Yosuke Shikama
- Department of Oral Disease ResearchNational Center for Geriatrics and GerontologyObuJapan
| | - Yoshihiro Hagiwara
- Department of Orthopaedic SurgeryGraduate School of MedicineTohoku UniversitySendaiJapan
| | - Keiichi Sasaki
- Division of Advanced Prosthetic DentistryGraduate School of DentistryTohoku UniversitySendaiJapan
| | - Makoto Kanzaki
- Graduate School of Biomedical EngineeringTohoku UniversitySendaiJapan
| | | |
Collapse
|
18
|
Pingel J, Pacolet A, Elfving B, Ledri LN. Intramuscular BoNT/A injections cause an inflammatory response in the muscle tissue of rats. EUR J INFLAMM 2021. [DOI: 10.1177/20587392211039942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Objectives The purpose of the present study was to investigate whether intramuscular BoNT/A injections cause an systemic inflammatory response and a local inflammatory response in the muscle tissue. Methods Thirty-two male Sprague Dawley rats treated with BoNT/A (i.m., 1IU) were divided in four groups, depending on the time of BoNT/A injection (2 days before, 1, 2, and 4 weeks before the experiment). Bio-Plex Pro Rat Cytokine 23-plex Multiplex Assay (Bio-Rad, USA). Results Systemic inflammation: 17 cytokines (IL1-α ( p = 0.005), IL-1β ( p = 0.01), IL-2 ( p = 0.04), IL-4 ( p = 0.03), IL-6 ( p = 0.03), IL-10 ( p = 0.02), IL12(p70) ( p = 0.03), IL-13 ( p = 0.04), IL-17 ( p = 0.03), GM-CSF ( p = 0.03), INF-γ ( p = 0.03), MIP-1α ( p = 0.03), MIP-3α ( p = 0.04), RANTES ( p = 0.001), TNF-α ( p = 0.04), vascular endothelial growth factor ( p = 0.03), and MCP-1 ( p = 0.02)) showed significantly higher expression levels 2 days after intramuscular BoNT/A injections compared to other time points (1, 2, and 4 weeks). Local inflammation: 12 cytokines (IL-1β ( p = 0.02), IL-6 ( p = 0.002), IL-10 ( p = 0.02), IL-13 ( p = 0.04), IL-17 ( p = 0.02), TNF-α ( p = 0.001), GM-CSF ( p = 0.01), M-CSF ( p = 0.04), MIP-1α ( p = 0.04), MIP-3α ( p = 0.002), RANTES ( p = 0.02), and MCP-1( p = 0.004)) showed higher expression levels 2 and/or 4 weeks after intramuscular BoNT/A injections compared to the other time points (2 days and 1 week). Conclusion Intramuscular BoNT/A injections result in a rapid systemic inflammatory response that only lasts a couple of days. At the same time, intramuscular BoNT/A injections cause an inflammatory response locally in the muscle with significantly higher cytokine levels 2 and/or 4 weeks after injections.
Collapse
Affiliation(s)
- Jessica Pingel
- Department of Neuroscience, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alexander Pacolet
- Department of Neuroscience, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Betina Elfving
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Litsa N Ledri
- Department of Neuroscience, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
19
|
FABP3-mediated membrane lipid saturation alters fluidity and induces ER stress in skeletal muscle with aging. Nat Commun 2020; 11:5661. [PMID: 33168829 PMCID: PMC7653047 DOI: 10.1038/s41467-020-19501-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 10/14/2020] [Indexed: 12/19/2022] Open
Abstract
Sarcopenia is characterized by decreased skeletal muscle mass and function with age. Aged muscles have altered lipid compositions; however, the role and regulation of lipids are unknown. Here we report that FABP3 is upregulated in aged skeletal muscles, disrupting homeostasis via lipid remodeling. Lipidomic analyses reveal that FABP3 overexpression in young muscles alters the membrane lipid composition to that of aged muscle by decreasing polyunsaturated phospholipid acyl chains, while increasing sphingomyelin and lysophosphatidylcholine. FABP3-dependent membrane lipid remodeling causes ER stress via the PERK-eIF2α pathway and inhibits protein synthesis, limiting muscle recovery after immobilization. FABP3 knockdown induces a young-like lipid composition in aged muscles, reduces ER stress, and improves protein synthesis and muscle recovery. Further, FABP3 reduces membrane fluidity and knockdown increases fluidity in vitro, potentially causing ER stress. Therefore, FABP3 drives membrane lipid composition-mediated ER stress to regulate muscle homeostasis during aging and is a valuable target for sarcopenia. Ageing leads to a loss of muscle mass and strength, called sarcopenia. Here, the authors show that fatty acid binding protein 3 (FABP3), a lipid chaperone, drives age-dependent lipidome remodeling in skeletal muscle and deteriorates muscle mass and contractility by modulating membrane fluidity and ER stress signaling.
Collapse
|
20
|
Yang A, Lv Q, Chen F, Wang Y, Liu Y, Shi W, Liu Y, Wang D. The effect of vitamin D on sarcopenia depends on the level of physical activity in older adults. J Cachexia Sarcopenia Muscle 2020; 11:678-689. [PMID: 32020783 PMCID: PMC7296263 DOI: 10.1002/jcsm.12545] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 12/09/2019] [Accepted: 01/07/2020] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Sarcopenia in older adults is closely related to vitamin D deficiency and reduced levels of physical activity, but little has been reported on the interaction between physical activity and the positive effects of vitamin D. The purpose of this study was to explore the interactive effect of vitamin D and physical activity on muscle mass and function through animal experiments and population surveys. METHODS Male 4-week-old C57BL/6J mice were fed different purified diets: a vitamin D-deficient diet (with increased calcium and phosphorus to prevent the effects of abnormal mineral levels on muscle) or a 1,25-dihydroxyvitamin D3 (1,25D)-supplemented diet. After 24 weeks on the assigned diets, the mice were immobilized. The level of skeletal muscle atrophy in the mice was determined by grip strength, gastrocnemius (GA) muscle mass and muscle fiber cross-sectional area (CSA); additionally, the protein expression levels of FOXO3a and the E3 ubiquitin ligases MuRF1 and MAFbx were detected. A cross-sectional study included data from 4139 older adults (64.9% women, 67.9 ± 6.7 years) as part of a survey in Shenyang, Northeast China. The associations of serum 25(OH)D3 and physical activity with timed up and go test (TUG) performance, handgrip strength, calf circumference, and body muscle mass were assessed by a linear regression analysis that was adjusted for covariates. RESULTS In activity-limited mice, vitamin D deficiency accelerated the decrease in GA muscle weight, muscle fiber CSA, and grip strength and increased the protein expression of MuRF1, MAFbx, and FOXO3a (all P < 0.05). In addition, 1,25D supplementation may inhibit the grip-strength reduction induced by limited activity (P = 0.069). Serum 25(OH)D3 and physical activity were linearly related to TUG time (P < 0.001) and handgrip strength (P < 0.05) after adjustment for sex, age, body mass index (BMI), education level, smoking status, and serum calcium level. Serum 25(OH)D3 and physical activity had interactive effects on TUG (P < 0.001) and handgrip strength (P < 0.05) but not calf circumference or body muscle mass in older adults. CONCLUSIONS The effect of vitamin D on muscle strength and physical performance depends on physical activity level in the elderly. It is recommended that older adults strive to avoid both physical inactivity and vitamin D deficiency. Because physical inactivity and vitamin D deficiency may exacerbate muscle atrophy, the biological mechanism may involve synergistic effects of vitamin D and physical activity on the promotion of muscle protein ubiquitination and degradation.
Collapse
Affiliation(s)
- Aolin Yang
- Nutrition Department, The First Hospital of China Medical University, Shenyang, China
| | - Qingqing Lv
- Nutrition Department, The First Hospital of China Medical University, Shenyang, China
| | - Feng Chen
- Department of Geriatric Endocrinology, The First Hospital of China Medical University, Shenyang, China
| | - Yingfang Wang
- Department of Geriatric Endocrinology, The First Hospital of China Medical University, Shenyang, China
| | - Yixuan Liu
- Department of Geriatric Endocrinology, The First Hospital of China Medical University, Shenyang, China
| | - Wanying Shi
- Nutrition Department, The First Hospital of China Medical University, Shenyang, China
| | - Ying Liu
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang, China
| | - Difei Wang
- Department of Geriatric Endocrinology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
21
|
Hitachi K, Nakatani M, Funasaki S, Hijikata I, Maekawa M, Honda M, Tsuchida K. Expression Levels of Long Non-Coding RNAs Change in Models of Altered Muscle Activity and Muscle Mass. Int J Mol Sci 2020; 21:ijms21051628. [PMID: 32120896 PMCID: PMC7084395 DOI: 10.3390/ijms21051628] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/25/2020] [Accepted: 02/25/2020] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle is a highly plastic organ that is necessary for homeostasis and health of the human body. The size of skeletal muscle changes in response to intrinsic and extrinsic stimuli. Although protein-coding RNAs including myostatin, NF-κβ, and insulin-like growth factor-1 (IGF-1), have pivotal roles in determining the skeletal muscle mass, the role of long non-coding RNAs (lncRNAs) in the regulation of skeletal muscle mass remains to be elucidated. Here, we performed expression profiling of nine skeletal muscle differentiation-related lncRNAs (DRR, DUM1, linc-MD1, linc-YY1, LncMyod, Neat1, Myoparr, Malat1, and SRA) and three genomic imprinting-related lncRNAs (Gtl2, H19, and IG-DMR) in mouse skeletal muscle. The expression levels of these lncRNAs were examined by quantitative RT-PCR in six skeletal muscle atrophy models (denervation, casting, tail suspension, dexamethasone-administration, cancer cachexia, and fasting) and two skeletal muscle hypertrophy models (mechanical overload and deficiency of the myostatin gene). Cluster analyses of these lncRNA expression levels were successfully used to categorize the muscle atrophy models into two sub-groups. In addition, the expression of Gtl2, IG-DMR, and DUM1 was altered along with changes in the skeletal muscle size. The overview of the expression levels of lncRNAs in multiple muscle atrophy and hypertrophy models provides a novel insight into the role of lncRNAs in determining the skeletal muscle mass.
Collapse
Affiliation(s)
- Keisuke Hitachi
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science (ICMS), Fujita Health University, Toyoake 470-1192, Japan; (K.H.); (M.N.)
| | - Masashi Nakatani
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science (ICMS), Fujita Health University, Toyoake 470-1192, Japan; (K.H.); (M.N.)
| | - Shiori Funasaki
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science (ICMS), Fujita Health University, Toyoake 470-1192, Japan; (K.H.); (M.N.)
| | - Ikumi Hijikata
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science (ICMS), Fujita Health University, Toyoake 470-1192, Japan; (K.H.); (M.N.)
| | - Mizuki Maekawa
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science (ICMS), Fujita Health University, Toyoake 470-1192, Japan; (K.H.); (M.N.)
| | - Masahiko Honda
- Department of Biochemistry, Kindai University Faculty of Medicine, Osaka-Sayama 589-8511, Japan;
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center Research Institute, Suita 564-8565, Japan
| | - Kunihiro Tsuchida
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science (ICMS), Fujita Health University, Toyoake 470-1192, Japan; (K.H.); (M.N.)
- Correspondence: ; Tel.: +81-562-93-9384
| |
Collapse
|
22
|
Nguyen TTN, Choi H, Jun HS. Preventive Effects of Dulaglutide on Disuse Muscle Atrophy Through Inhibition of Inflammation and Apoptosis by Induction of Hsp72 Expression. Front Pharmacol 2020; 11:90. [PMID: 32153405 PMCID: PMC7046759 DOI: 10.3389/fphar.2020.00090] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/27/2020] [Indexed: 12/23/2022] Open
Abstract
Pathological conditions such as joint immobilization, long-time bed rest, or inactivity may result in disuse-induced muscle wasting and dysfunction. To investigate the effect of dulaglutide, a long-acting glucagon-like peptide-1 receptor agonist, on disuse muscle atrophy, disuse condition was induced by spiral wire immobilization in C57BL/6 mice and the mice were treated with dulaglutide. Dulaglutide treatment effectively improved muscle function and increased muscle mass compared with vehicle treatment. Dulaglutide inhibited the decrease of muscle fiber size and the expression of atrophic factors such as myostatin, atrogin-1/MAFbx, and muscle RING-finger protein-1 in immobilized mice. In addition, dulaglutide inhibited nuclear factor kappa B activation, leading to a decrease in the mRNA levels of proinflammatory cytokines, including tumor necrosis factor-α, interleukin (IL)-1β, and IL-6 in muscle of immobilized mice. Dulaglutide suppressed the expression of apoptotic markers such as caspase-3, cleaved poly-ADP ribose polymerase, and Bax under immobilization condition and increased the expression of heat shock protein 72 (Hsp72), which is related to the amelioration of inflammation and apoptosis during disuse time. Further study showed that dulaglutide could induce Hsp72 expression via the regulation of 5′-AMP-activated protein kinase signaling. Our data suggest that dulaglutide could exert beneficial effects against disuse-induced muscle atrophy.
Collapse
Affiliation(s)
- Tram Thi Ngoc Nguyen
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, South Korea
| | - Hojung Choi
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, South Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, South Korea
| | - Hee-Sook Jun
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, South Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, South Korea.,Gachon Medical Research Institute, Gil Hospital, Incheon, South Korea
| |
Collapse
|
23
|
Lee YJ, Kim GH, Park SI, Lim JH. Down-regulation of the mitochondrial i-AAA protease Yme1L induces muscle atrophy via FoxO3a and myostatin activation. J Cell Mol Med 2019; 24:899-909. [PMID: 31725201 PMCID: PMC6933342 DOI: 10.1111/jcmm.14799] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/25/2019] [Accepted: 10/13/2019] [Indexed: 12/16/2022] Open
Abstract
Muscle atrophy is closely associated with many diseases, including diabetes and cardiac failure. Growing evidence has shown that mitochondrial dysfunction is related to muscle atrophy; however, the underlying mechanisms are still unclear. To elucidate how mitochondrial dysfunction causes muscle atrophy, we used hindlimb-immobilized mice. Mitochondrial function is optimized by balancing mitochondrial dynamics, and we observed that this balance shifted towards mitochondrial fission and that MuRF1 and atrogin-1 expression levels were elevated in these mice. We also found that the expression of yeast mitochondrial escape 1-like ATPase (Yme1L), a mitochondrial AAA protease was significantly reduced both in hindlimb-immobilized mice and carbonyl cyanide m-chlorophenylhydrazone (CCCP)-treated C2C12 myotubes. When Yme1L was depleted in myotubes, the short form of optic atrophy 1 (Opa1) accumulated, leading to mitochondrial fragmentation. Moreover, a loss of Yme1L, but not of LonP1, activated AMPK and FoxO3a and concomitantly increased MuRF1 in C2C12 myotubes. Intriguingly, the expression of myostatin, a myokine responsible for muscle protein degradation, was significantly increased by the transient knock-down of Yme1L. Taken together, our results suggest that a deficiency in Yme1L and the consequential imbalance in mitochondrial dynamics result in the activation of FoxO3a and myostatin, which contribute to the pathological state of muscle atrophy.
Collapse
Affiliation(s)
- Yoo Jeong Lee
- Division of Endocrine and Metabolic Disease, Center for Biomedical Sciences, Korea National Institute of Health, Cheongju, Korea
| | - Gyu Hee Kim
- Division of Endocrine and Metabolic Disease, Center for Biomedical Sciences, Korea National Institute of Health, Cheongju, Korea
| | - Sang Ick Park
- Division of Endocrine and Metabolic Disease, Center for Biomedical Sciences, Korea National Institute of Health, Cheongju, Korea
| | - Joo Hyun Lim
- Division of Endocrine and Metabolic Disease, Center for Biomedical Sciences, Korea National Institute of Health, Cheongju, Korea
| |
Collapse
|
24
|
Cistanche tubulosa (Schenk) Wight Extract Enhances Hindlimb Performance and Attenuates Myosin Heavy Chain IId/IIx Expression in Cast-Immobilized Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:9283171. [PMID: 31885674 PMCID: PMC6925718 DOI: 10.1155/2019/9283171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/13/2019] [Accepted: 09/17/2019] [Indexed: 12/13/2022]
Abstract
Skeletal muscle atrophy is encountered in many clinical conditions, but a pharmacological treatment has not yet been established. Cistanche tubulosa (Schenk) Wight is an herbal medicine used in traditional Japanese and Chinese medicine. In the current study, we investigated the effect of C. tubulosa extract (CTE) on atrophied muscle in vivo. We also investigated hindlimb cast immobilization in mice and devised a novel type of hindlimb-immobilizing cast, consisting of sponge-like tape and a thin plastic tube. Using this method, 3 out of 4 groups of mice (n = 11 for each group) were cast-immobilized in the hindlimbs and administered CTE or vehicle for 13 days. A sham procedure was performed in the mice of the fourth group to which the vehicle was administered. Next, the triceps surae muscles (TS) were excised. To analyze the effect of the novel cast system and CTE administration on muscle atrophy, we evaluated TS wet weight and myofiber cross-sectional area (CSA). We also determined MyHC IId/IIx expression levels by western blotting, since their increase is a hallmark of disuse muscle atrophy, suggesting slow-to-fast myofiber type shift. Moreover, we performed two tests of hindlimb performance. The novel cast immobilization method significantly reduced TS wet weight and myofiber CSA. This was accompanied by deterioration of hindlimb function and an increase in MyHC IId/IIx expression. CTE administration did not alter TS wet weight or myofiber CSA; however, it showed a trend of amelioration of the loss of hindlimb function and of suppression of the increased MyHC IId/IIx expression in cast-immobilized mice. Our novel hindlimb cast immobilization method effectively induced muscle atrophy. CTE did not affect muscle mass, but suppressed the shift from slow to fast myofiber type in cast-immobilized mice, ameliorating hindlimb function deterioration.
Collapse
|
25
|
Ma W, Zhang R, Huang Z, Zhang Q, Xie X, Yang X, Zhang Q, Liu H, Ding F, Zhu J, Sun H. PQQ ameliorates skeletal muscle atrophy, mitophagy and fiber type transition induced by denervation via inhibition of the inflammatory signaling pathways. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:440. [PMID: 31700876 DOI: 10.21037/atm.2019.08.101] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background Skeletal muscle atrophy involves and requires widespread changes in skeletal muscle gene expression and signaling pathway, resulting in excessive loss of muscle mass and strength, which is associated with poor prognosis and the decline of life quality in several diseases. However, the treatment of skeletal muscle atrophy remains an unresolved challenge to this day. The aim of the present study was to investigate the influence of pyrroloquinoline quinone (PQQ), a redox-active o-quinone found in various foods and mammalian tissues, on skeletal muscle atrophy, and to explore the underlying molecular mechanism. Methods After denervation, mice were injected intraperitoneally with saline plus PQQ (5 mg/kg/d) or saline only for 14 days. The level of inflammatory cytokines in tibialis anterior (TA) muscles was determined by quantitative real-time polymerase chain reaction (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA), and the level of signaling proteins of Janus kinase 2/signal transduction and activator of transcription 3 (Jak2/STAT3), TGF-β1/Smad3, JNK/p38 MAPK, and nuclear factor κB (NF-κB) signaling pathway were detected by Western blot. The skeletal muscle atrophy was evaluated by muscle wet weight ratio and cross-sectional areas (CSAs) of myofibers. The mitophagy was observed through transmission electron microscopy (TEM) analysis, and muscle fiber type transition was analyzed through fast myosin skeletal heavy chain antibody staining. Results The proinflammatory cytokines IL-6, IL-1β and TNF-α were largely induced in TA muscles after sciatic nerve transection. PQQ can significantly reverse this phenomenon, as evidenced by the decreased levels of proinflammatory cytokines IL-6, IL-1β and TNF-α. Moreover, PQQ could significantly attenuate the signal activation of Jak2/STAT3, TGF-β1/Smad3, JNK/p38 MAPK, and NF-κB in skeletal muscles after sciatic nerve transection. Furthermore, PQQ alleviated skeletal muscle atrophy, mitigated mitophagy and inhibited slow-to-fast muscle fiber type transition. Conclusions These results suggested that PQQ could attenuate denervation-induced skeletal muscle atrophy, mitophagy and fiber type transition through suppressing the Jak2/STAT3, TGF-β1/Smad3, JNK/p38 MAPK, and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Wenjing Ma
- 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 226001, China
| | - Ru Zhang
- Department of Imaging, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, China
| | - Ziwei Huang
- 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 226001, China
| | - Qiuyu Zhang
- 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 226001, China
| | - Xiaoying Xie
- 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 226001, China
| | - Xiaoming Yang
- 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 226001, China
| | - Qi Zhang
- 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 226001, China
| | - Hua Liu
- Department of Orthopedics, Haian Hospital of Traditional Chinese Medicine, Haian 226600, China
| | - Fei Ding
- 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 226001, China
| | - Jianwei Zhu
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong 226001, 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 226001, China
| |
Collapse
|
26
|
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
|
27
|
Munroe M, Dvoretskiy S, Lopez A, Leong J, Dyle MC, Kong H, Adams CM, Boppart MD. Pericyte transplantation improves skeletal muscle recovery following hindlimb immobilization. FASEB J 2019; 33:7694-7706. [PMID: 31021652 PMCID: PMC6529341 DOI: 10.1096/fj.201802580r] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 03/04/2019] [Indexed: 02/06/2023]
Abstract
Conditions of extended bed rest and limb immobilization can initiate rapid and significant loss of skeletal muscle mass and function. Physical rehabilitation is standard practice following a period of disuse, yet mobility may be severely compromised, and recovery is commonly delayed or incomplete in special populations. Thus, a novel approach toward recovery of muscle mass is highly desired. Pericytes [neuron-glial antigen 2 (NG2)+CD31-CD45- (Lineage- [Lin-]) and CD146+Lin-] demonstrate capacity to facilitate muscle repair, yet the ability to enhance myofiber growth following disuse is unknown. In the current study, 3-4-mo-old mice were unilaterally immobilized for 14 d (IM) or immobilized for 14 d followed by 14 d of remobilization (RE). Flow cytometry and targeted gene expression analyses were completed to assess pericyte quantity and function following IM and RE. In addition, a transplantation study was conducted to assess the impact of pericytes on recovery. Results from targeted analyses suggest minimal impact of disuse on pericyte gene expression, yet NG2+Lin- pericyte quantity is reduced following IM (P < 0.05). Remarkably, pericyte transplantation recovered losses in myofiber cross-sectional area and the capillary-to-fiber ratio following RE, whereas deficits remained with vehicle alone (P = 0.01). These findings provide the first evidence that pericytes effectively rehabilitate skeletal muscle mass following disuse atrophy.-Munroe, M., Dvoretskiy, S., Lopez, A., Leong, J., Dyle, M. C., Kong, H., Adams, C. M., Boppart, M. D. Pericyte transplantation improves skeletal muscle recovery following hindlimb immobilization.
Collapse
Affiliation(s)
- Michael Munroe
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Svyatoslav Dvoretskiy
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Amber Lopez
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Jiayu Leong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Michael C. Dyle
- Departments of Internal Medicine and University of Iowa, Iowa City, Iowa, USA
- Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois–Urbana-Champaign, Urbana, Illinois, USA
| | - Christopher M. Adams
- Departments of Internal Medicine and University of Iowa, Iowa City, Iowa, USA
- Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Marni D. Boppart
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois–Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
28
|
Granulocyte-colony stimulating factor enhances load-induced muscle hypertrophy in mice. Biochem Biophys Res Commun 2018; 506:944-949. [PMID: 30401566 DOI: 10.1016/j.bbrc.2018.10.196] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 10/30/2018] [Indexed: 11/23/2022]
Abstract
Granulocyte-colony stimulating factor (G-CSF) is a cytokine crucially involved in the regulation of granulopoiesis and the mobilization of hematopoietic stem cells from bone marrow. However, emerging data suggest that G-CSF exhibits more diverse functions than initially expected, such as conferring protection against apoptosis to neural cells and stimulating mitogenesis in cardiomyocytes and skeletal muscle stem cells after injury. In the present study, we sought to investigate the potential contribution of G-CSF to the regulation of muscle volume. We found that the administration of G-CSF significantly enhances muscle hypertrophy in two different muscle overload models. Interestingly, there was a significant increase in the transcripts of both G-CSF and G-CSF receptors in the muscles that were under overload stress. Using mutant mice lacking the G-CSF receptor, we confirmed that the anabolic effect is dependent on the G-CSF receptor signaling. Furthermore, we found that G-CSF increases the diameter of myotubes in vitro and induces the phosphorylation of AKT, mTOR, and ERK1/2 in the myoblast-like cell line C2C12 after differentiation induction. These findings indicate that G-CSF is involved in load-induced muscle hypertrophy and suggest that G-CSF is a potential agent for treating patients with muscle loss and sarcopenia.
Collapse
|
29
|
The Effects of Dokhwalgisaeng-tang against Disuse Muscle Atrophy in Gastrocnemius of Rats. JOURNAL OF ACUPUNCTURE RESEARCH 2018. [DOI: 10.13045/jar.2018.00227] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
30
|
Local cyclical compression modulates macrophage function in situ and alleviates immobilization-induced muscle atrophy. Clin Sci (Lond) 2018; 132:2147-2161. [PMID: 30209036 DOI: 10.1042/cs20180432] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 09/10/2018] [Accepted: 09/11/2018] [Indexed: 01/10/2023]
Abstract
Physical inactivity gives rise to numerous diseases and organismal dysfunctions, particularly those related to aging. Musculoskeletal disorders including muscle atrophy, which can result from a sedentary lifestyle, aggravate locomotive malfunction and evoke a vicious circle leading to severe functional disruptions of vital organs such as the brain and cardiovascular system. Although the significance of physical activity is evident, molecular mechanisms behind its beneficial effects are poorly understood. Here, we show that massage-like mechanical interventions modulate immobilization-induced pro-inflammatory responses of macrophages in situ and alleviate muscle atrophy. Local cyclical compression (LCC) on mouse calves, which generates intramuscular pressure waves with amplitude of 50 mmHg, partially restores the myofiber thickness and contracting forces of calf muscles that are decreased by hindlimb immobilization. LCC tempers the increase in the number of cells expressing pro-inflammatory proteins, tumor necrosis factor-α and monocyte chemoattractant protein-1 (MCP-1), including macrophages in situ The reversing effect of LCC on immobilization-induced thinning of myofibers is almost completely nullified when macrophages recruited from circulating blood are depleted by administration of clodronate liposomes. Furthermore, application of pulsatile fluid shear stress, but not hydrostatic pressure, reduces the expression of MCP-1 in macrophages in vitro Together with the LCC-induced movement of intramuscular interstitial fluid detected by µCT analysis, these results suggest that mechanical modulation of macrophage function is involved in physical inactivity-induced muscle atrophy and inflammation. Our findings uncover the implication of mechanosensory function of macrophages in disuse muscle atrophy, thereby opening a new path to develop a novel therapeutic strategy utilizing mechanical interventions.
Collapse
|
31
|
Dunham CL, Chamberlain AM, Meyer GA, Lake SP. Muscle does not drive persistent posttraumatic elbow contracture in a rat model. Muscle Nerve 2018; 58:843-851. [PMID: 30230560 DOI: 10.1002/mus.26344] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/19/2018] [Accepted: 09/11/2018] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Posttraumatic elbow contracture is clinically challenging because injury often disrupts multiple periarticular soft tissues. Tissue specific contribution to contracture, particularly muscle, remains poorly understood. METHODS In this study we used a previously developed animal model of elbow contracture. After surgically inducing a unilateral soft tissue injury, injured limbs were immobilized for 3, 7, 21, and 42 days (IM) or for 42 IM with 42 days of free mobilization (42/42 IM-FM). Biceps brachii active/passive mechanics and morphology were evaluated at 42 IM and 42/42 IM-FM, whereas biceps brachii and brachialis gene expression was evaluated at all time points. RESULTS Injured limb muscle exhibited significantly altered active/passive mechanics and decreased fiber area at 42 IM but returned to control levels by 42/42 IM-FM. Gene expression suggested muscle growth rather than a fibrotic response at 42/42 IM-FM. DISCUSSION Muscle is a transient contributor to motion loss in our rat model of posttraumatic elbow contracture. Muscle Nerve 58:843-851, 2018.
Collapse
Affiliation(s)
- Chelsey L Dunham
- Department of Biomedical Engineering, Washington University in St Louis, St Louis, Missouri, USA
| | - Aaron M Chamberlain
- Department of Orthopaedic Surgery, Washington University in St Louis, St Louis, Missouri, USA
| | - Gretchen A Meyer
- Program in Physical Therapy, Washington University in St Louis, St Louis, Missouri, USA
| | - Spencer P Lake
- Department of Mechanical Engineering and Materials Science, Washington University in St Louis, 1 Brookings Drive, Campus Box 1185, St. Louis, Missouri, 63130, USA
| |
Collapse
|
32
|
Kim YB, Yang HS, Kim HJ, Choi HR, In J, Yoon SY, Ro YJ. Effects of neuromuscular presynaptic muscarinic M 1 receptor blockade on rocuronium-induced neuromuscular blockade in immobilized tibialis anterior muscles. Clin Exp Pharmacol Physiol 2018; 45:1309-1316. [PMID: 30005130 PMCID: PMC6282578 DOI: 10.1111/1440-1681.13012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 11/29/2022]
Abstract
This in vivo study tested the hypothesis that the modulation of acetylcholine (ACh) release by the M1 muscarinic receptor (mAChR) in the neuromuscular junction of disused muscles may affect the tensions of the muscles during the neuromuscular monitoring of a rocuronium‐induced neuromuscular block and compared the results with those obtained from normal muscles. A total of 20 C57BL/6 (wild‐type) and 10 α7 knock out (α7KO) mice were used in this experiment. As a pre‐experimental procedure, knee and ankle joints of right hind limbs were fixed by needle pinning at the 90° flexed position. After 2 weeks, the main experiment was performed. Both tendons of the tibialis anterior (TA) muscles were obtained, and the muscle tensions were recorded while the dose‐responses of rocuronium were measured three times in the same mouse by the serial administration of pirenzepine (0, 0.001 and 0.01 μg/g). Weight losses were observed after 2 weeks of immobilization in both groups, and a decrease in the mass of TA muscles at the immobilized side was observed compared to those of the contralateral nonimmobilized side. Tension depression of the TA muscles at immobilized side of the α7KO group was faster than those of the wild‐type group, but these differences decreased after the administration of pirenzepine. The tension depressions were similar regardless of the pirenzepine doses at the same side in the group. Tension depression may become more rapid in the α7 AChR‐expressed disused muscles by the decreased release of ACh release upon neuronal firing by the blockade of facilitatory M1mAChR
Collapse
Affiliation(s)
- Yong Beom Kim
- Department of Anesthesiology and Pain Medicine, Gil Medical Center, College of Medicine, Gachon University, Incheon, Korea
| | - Hong-Seuk Yang
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, College of Medicine, Ulsan University, Seoul, Korea
| | - Ha Jung Kim
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, College of Medicine, Ulsan University, Seoul, Korea
| | - Hey-Ran Choi
- Department of Anaesthesiology and Pain Medicine, Seoul Paik Hospital, College of Medicine, Inje University, Seoul, Korea
| | - Junyong In
- Department of Anaesthesiology and Pain Medicine, College of Medicine, Ilsan Hospital, Dongguk University, Gyeonggido, Korea
| | - Soon-Young Yoon
- Department of Anesthesiology and Pain Medicine, Gil Medical Center, College of Medicine, Gachon University, Incheon, Korea
| | - Young Jin Ro
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, College of Medicine, Ulsan University, Seoul, Korea
| |
Collapse
|
33
|
Gao Y, Arfat Y, Wang H, Goswami N. Muscle Atrophy Induced by Mechanical Unloading: Mechanisms and Potential Countermeasures. Front Physiol 2018; 9:235. [PMID: 29615929 PMCID: PMC5869217 DOI: 10.3389/fphys.2018.00235] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/02/2018] [Indexed: 12/23/2022] Open
Abstract
Prolonged periods of skeletal muscle inactivity or mechanical unloading (bed rest, hindlimb unloading, immobilization, spaceflight and reduced step) can result in a significant loss of musculoskeletal mass, size and strength which ultimately lead to muscle atrophy. With advancement in understanding of the molecular and cellular mechanisms involved in disuse skeletal muscle atrophy, several different signaling pathways have been studied to understand their regulatory role in this process. However, substantial gaps exist in our understanding of the regulatory mechanisms involved, as well as their functional significance. This review aims to update the current state of knowledge and the underlying cellular mechanisms related to skeletal muscle loss during a variety of unloading conditions, both in humans and animals. Recent advancements in understanding of cellular and molecular mechanisms, including IGF1-Akt-mTOR, MuRF1/MAFbx, FOXO, and potential triggers of disuse atrophy, such as calcium overload and ROS overproduction, as well as their role in skeletal muscle protein adaptation to disuse is emphasized. We have also elaborated potential therapeutic countermeasures that have shown promising results in preventing and restoring disuse-induced muscle loss. Finally, identified are the key challenges in this field as well as some future prospectives.
Collapse
Affiliation(s)
- Yunfang Gao
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Ministry of Education, Northwest University, Xi'an, China
| | - Yasir Arfat
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Ministry of Education, Northwest University, Xi'an, China
| | - Huiping Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Ministry of Education, Northwest University, Xi'an, China
| | - Nandu Goswami
- Physiology Unit, Otto Loewi Center of Research for Vascular Biology, Immunity and Inflammation, Medical University of Graz, Graz, Austria
| |
Collapse
|
34
|
Kawanishi N, Nozaki R, Naito H, Machida S. TLR4-defective (C3H/HeJ) mice are not protected from cast immobilization-induced muscle atrophy. Physiol Rep 2018; 5:5/8/e13255. [PMID: 28432254 PMCID: PMC5408285 DOI: 10.14814/phy2.13255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/17/2017] [Accepted: 03/20/2017] [Indexed: 12/25/2022] Open
Abstract
Recent studies have shown that activation of Toll‐like receptor (TLR)4 signaling may be an important factor in muscle atrophy and excessive inflammatory response associated with immobilization. To examine the role of TLR4 signaling on cast immobilization‐induced skeletal muscle atrophy, we tested the hypothesis that muscle atrophy and inflammation after cast immobilization is reduced in TLR4‐defective mice. TLR4‐defective (C3H/HeJ) and wild type (C3H/HeN) mice were divided into control and cast‐immobilization groups. Cast immobilization was imposed for 14 days. Cast immobilization increased TLR4 mRNA expression in the gastrocnemius and decreased muscle mass and cross‐sectional area (CSA) of the gastrocnemius fibers. However, there was no difference in the gastrocnemius muscle mass and CSA between TLR4‐defective and wild type mice. Cast immobilization‐induced increase in ubiquitin E3 ligases (MAFbx/Atrogin‐1 and MuRF1), inflammatory cytokines, and macrophage/monocyte marker mRNAs were unaffected by defective TLR4. Our findings in C3H/HeJ mice suggested that TLR4 signaling might not play an essential role in immobilization‐induced muscle atrophy.
Collapse
Affiliation(s)
- Noriaki Kawanishi
- Institute of Health & Sports Science and Medicine, Juntendo University, Chiba, Japan.,Research Fellow of the Japan Society for the Promotion of Sciences, Tokyo, Japan
| | - Risa Nozaki
- Guraduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Hisashi Naito
- Institute of Health & Sports Science and Medicine, Juntendo University, Chiba, Japan.,Guraduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Shuichi Machida
- Institute of Health & Sports Science and Medicine, Juntendo University, Chiba, Japan .,Guraduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| |
Collapse
|
35
|
Cavey T, Pierre N, Nay K, Allain C, Ropert M, Loréal O, Derbré F. Simulated microgravity decreases circulating iron in rats: role of inflammation-induced hepcidin upregulation. Exp Physiol 2018; 102:291-298. [PMID: 28087888 DOI: 10.1113/ep086188] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 01/09/2017] [Indexed: 12/19/2022]
Abstract
NEW FINDINGS What is the central question of this study? Although microgravity is well known to reduce circulating iron in astronauts, the underlying mechanism is still unknown. We investigated whether hepcidin, a key hormone regulating iron metabolism, could be involved in this deleterious effect. What is the main finding and its importance? We show that hindlimb suspension, a model of microgravity, stimulates the production of hepcidin in liver of rats. In agreement with the biological role of hepcidin, we found a decrease of circulating iron and an increase of spleen iron content in hindlimb-unloaded rats. Consequently, our study supports the idea that hepcidin could play a role in the alteration of iron metabolism parameters observed during spaceflight. During spaceflight, humans exposed to microgravity exhibit an increase of iron storage and a reduction of circulating iron. Such perturbations could promote oxidative stress and anaemia in astronauts. The mechanism by which microgravity modulates iron metabolism is still unknown. Herein, we hypothesized that microgravity upregulates hepcidin, a hormone produced by the liver that is the main controller of iron homeostasis. To test this hypothesis, rats were submitted to hindlimb unloading (HU), the reference model to mimic the effects of microgravity in rodents. After 7 days, the mRNA level of hepcidin was increased in the liver of HU rats (+74%, P = 0.001). In agreement with the biological role of hepcidin, we found an increase of spleen iron content (+78%, P = 0.030) and a decrease of serum iron concentration (-35%, P = 0.002) and transferrin saturation (-25%, P = 0.011) in HU rats. These findings support a role of hepcidin in microgravity-induced iron metabolism alteration. Furthermore, among the signalling pathways inducing hepcidin mRNA expression, we found that only the interleukin-6/signal transducer and activator of transcription 3 (IL-6/STAT3) axis was activated by HU, as shown by the increase of phospho-STAT3 (+193%, P < 0.001) and of the hepatic mRNA level of haptoglobin (+167%, P < 0.001), a STAT3-inducible gene, in HU rats. Taken together, these data support the idea that microgravity may alter iron metabolism through an inflammatory process upregulating hepcidin.
Collapse
Affiliation(s)
- Thibault Cavey
- INSERM UMR 991 UMR, Rennes, France.,University of Rennes 1, Rennes, France.,Department of Biochemistry, CHU Rennes, France
| | - Nicolas Pierre
- Laboratory 'Movement, Sport and Health Sciences' (M2S), University Rennes 2-ENS Rennes, Bruz, France
| | - Kévin Nay
- Laboratory 'Movement, Sport and Health Sciences' (M2S), University Rennes 2-ENS Rennes, Bruz, France
| | - Coralie Allain
- INSERM UMR 991 UMR, Rennes, France.,University of Rennes 1, Rennes, France
| | - Martine Ropert
- INSERM UMR 991 UMR, Rennes, France.,Department of Biochemistry, CHU Rennes, France
| | - Olivier Loréal
- INSERM UMR 991 UMR, Rennes, France.,University of Rennes 1, Rennes, France
| | - Frédéric Derbré
- Laboratory 'Movement, Sport and Health Sciences' (M2S), University Rennes 2-ENS Rennes, Bruz, France
| |
Collapse
|
36
|
Jung HJ, Lee KP, Milholland B, Shin YJ, Kang JS, Kwon KS, Suh Y. Comprehensive miRNA Profiling of Skeletal Muscle and Serum in Induced and Normal Mouse Muscle Atrophy During Aging. J Gerontol A Biol Sci Med Sci 2017; 72:1483-1491. [PMID: 28329037 DOI: 10.1093/gerona/glx025] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 02/01/2017] [Indexed: 01/29/2023] Open
Abstract
Age-associated loss of muscle mass and function is a major cause of morbidity and mortality in the elderly adults. Muscular atrophy can also be induced by disuse associated with long-term bed rest or disease. Although miRNAs regulate muscle growth, regeneration, and aging, their potential role in acute muscle atrophy is poorly understood. Furthermore, alterations in circulating miRNA levels have been shown to occur during aging but their potential as noninvasive biomarkers for muscle atrophy remains largely unexplored. Here, we report comprehensive miRNA expression profiles by miRNA-seq analysis in tibialis anterior muscle and serum of a disuse-induced atrophy mouse model, mimicking the acute atrophy following long-term bed rest, as compared to those of young and old mice. Comparative analysis and validation studies have revealed that miR-455-3p was significantly decreased in muscle of both induced-atrophy model and old mice, whereas miR-434-3p was decreased in both serum and muscle of old mice, as compared to young mice. Furthermore, upregulation of miR-455-3p in fully differentiated C2C12 myoblasts induced a hypertrophic phenotype. These results suggest that deregulation of miR-455-3p may play a functional role in muscle atrophy and miR-434-3p could be a candidate serum biomarker of muscle aging.
Collapse
Affiliation(s)
- Hwa Jin Jung
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Kwang-Pyo Lee
- Aging Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon.,Department of Biomolecular Science
| | - Brandon Milholland
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Yeo Jin Shin
- Aging Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon
| | - Jae Sook Kang
- Aging Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon
| | - Ki-Sun Kwon
- Aging Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon
| | - Yousin Suh
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
37
|
Aihara M, Hirose N, Katsuta W, Saito F, Maruyama H, Hagiwara H. A new model of skeletal muscle atrophy induced by immobilization using a hook-and-loop fastener in mice. J Phys Ther Sci 2017; 29:1779-1783. [PMID: 29184288 PMCID: PMC5684009 DOI: 10.1589/jpts.29.1779] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 07/07/2017] [Indexed: 01/29/2023] Open
Abstract
[Purpose] To study muscle atrophy, the muscle atrophy model mice have been used
frequently. In particular, cast immobilization is the most common method to induce muscle
atrophy. However, it is time consuming and often causes adverse events including skin
injury, edema, and necrosis. The present study, we developed a hook-and-loop fastener
(Velcro) immobilization method as a new, simple, and less invasive approach to induce
muscle atrophy. [Subjects and Methods] Mice were bandaged in the knee joint extension and
ankle plantar extension position. Muscle atrophy was induced by either winding a cast or
Velcro around the limb. [Results] According to weight and fiber size, Velcro
immobilization induced equivalent muscle atrophy to cast immobilization. Velcro
immobilization reduced significantly the time for the procedure and the frequency of
adverse events. [Conclusion] Velcro immobilization can induce muscle atrophy comparable to
cast immobilization, but in a shorter time and with less complications. Velcro
immobilization may contribute to the study of disuse muscle atrophy in clinical practice
of physical therapy using a mouse model.
Collapse
Affiliation(s)
- Masahiro Aihara
- Department of Physical Therapy, Faculty of Medical Sciences, Teikyo University of Science: 2525 Yatsuzawa, Uenohara, Yamanashi 409-0193, Japan.,Division of Physical Therapy, Department of Health Science, International University of Health and Welfare Graduate School, Japan
| | - Noboru Hirose
- Department of Physical Therapy, Faculty of Medical Sciences, Teikyo University of Science: 2525 Yatsuzawa, Uenohara, Yamanashi 409-0193, Japan.,Department of Neurology, Teikyo University School of Medicine, Japan
| | - Wakana Katsuta
- Division of Biosciences, Graduate School of Science and Engineering, Teikyo University of Science, Japan.,Department of Rehabilitation, National Center of Neurology and Psychiatry, Japan
| | - Fumiaki Saito
- Department of Neurology, Teikyo University School of Medicine, Japan
| | - Hitoshi Maruyama
- Division of Physical Therapy, Department of Health Science, International University of Health and Welfare Graduate School, Japan
| | - Hiroki Hagiwara
- Department of Physical Therapy, Faculty of Medical Sciences, Teikyo University of Science: 2525 Yatsuzawa, Uenohara, Yamanashi 409-0193, Japan.,Department of Neurology, Teikyo University School of Medicine, Japan.,Division of Biosciences, Graduate School of Science and Engineering, Teikyo University of Science, Japan
| |
Collapse
|
38
|
Experimental Periodontitis in the Potentialization of the Effects of Immobilism in the Skeletal Striated Muscle. Inflammation 2017; 40:2000-2011. [PMID: 28822015 DOI: 10.1007/s10753-017-0640-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This study aims to evaluate if ligature-induced periodontitis can potentiates the deleterious effects of immobilization in the skeletal striated muscle, contributing to the development of muscle atrophy due to disuse. Forty Wistar rats were divided into four groups: (1) Control Group (CG), (2) Periodontal Disease (PDG), (3) Immobilized (IG), and (4) Immobilized with Periodontal Disease (IPDG). Periodontal disease was induced for 30 days, with ligature method, and the immobilization was performed with cast bandage for 15 days. Prior to euthanasia, nociceptive threshold and muscular grasping force were evaluated. Afterwards, the soleus muscle was dissected and processed for sarcomere counting and morphological/morphometric analysis. For data analysis, was used the one-way ANOVA and post-test Tukey (p < 0.05). The IG and IPDG presented lower muscle weight, lower muscular grip strength, and less number of sarcomeres compared to CG. The PDG showed reduction of muscle strength and nociceptive threshold after 15 days of periodontal disease and increased connective tissue compared to CG. The IPDG presented lower muscle length and nociceptive threshold. The IG presented reduction in cross-sectional area and smaller diameter, increase in the number of nuclei and a nucleus/fiber ratio, decrease in the number of capillaries and capillary/fiber ratio, with increase in connective tissue. The IPDG had increased nucleus/fiber ratio, decreased capillaries, and increased connective tissue when compared to the IG. The IPDG presented greater muscle tissue degeneration and increased inflammatory cells compared to the other groups. Ligature-induced periodontitis potentiated the deleterious effects of immobilization of the skeletal striated muscle.
Collapse
|
39
|
Yang J, Min S, Xie F, Chen J, Hao X, Ren L. Electroacupuncture alleviates neuromuscular dysfunction in an experimental rat model of immobilization. Oncotarget 2017; 8:85537-85548. [PMID: 29156739 PMCID: PMC5689629 DOI: 10.18632/oncotarget.20246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 07/25/2017] [Indexed: 02/06/2023] Open
Abstract
Immobilization-related skeletal muscle atrophy is a major concern to patients in Intensive Care Units and it has a profound effect on the quality of life. However, the underlying molecular events for the therapeutic effect of electroacupuncture to treat muscle atrophy have not been fully elucidated. Here we developed an immobilization mouse model and tested the hypothesis that skeletal muscle weakness may be caused by the increased expression of γ and α7 nicotinic acetylcholine receptors (nAChRs) on muscle cell membranes, while electroacupuncture could decrease the expression of γ and α7 nicotinic acetylcholine receptors. Compared with the rats in control, those treated with immobilization for 14 days showed a significant reduction of tibialis anterior muscle weight, muscle atrophy and dysfunction, which was associated with a significant decrease expression of neuregulin-1 and increased expression of γ- and α7-nAChR in tibialis anterior muscle. Electroacupuncture significantly enhanced the expression of neuregulin-1 and alleviated the muscle loss, while diminished the expression of γ- and α7-nAChR. Taken together, the beneficial effect of electroacupuncture may be attributed to suppressing γ- and α7-nAChR production, enhancing neuromuscular function and neuregulin-1 protein synthesis. These results suggest that electroacupuncture is a potential therapy for preventing muscle atrophy during immobilization.
Collapse
Affiliation(s)
- Jun Yang
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Su Min
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fei Xie
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jingyuan Chen
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xuechao Hao
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Li Ren
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
40
|
Little RD, Prieto-Potin I, Pérez-Baos S, Villalvilla A, Gratal P, Cicuttini F, Largo R, Herrero-Beaumont G. Compensatory anabolic signaling in the sarcopenia of experimental chronic arthritis. Sci Rep 2017; 7:6311. [PMID: 28740214 PMCID: PMC5524910 DOI: 10.1038/s41598-017-06581-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 06/14/2017] [Indexed: 01/06/2023] Open
Abstract
Inflammatory activity in rheumatoid arthritis may alter the regulation of muscle mass leading to a secondary sarcopenia, commonly termed rheumatoid cachexia (RC). We characterized alterations to muscle structure and various pro-inflammatory, catabolic and regenerative markers in an animal model of RC. Antigen induced arthritis (AiA) was performed in 20 male adult rabbits. AiA animals exhibited significantly less weight gain, a markedly elevated serum C-reactive protein (CRP), lighter muscles with shorter cross-sectional diameter and increased myonuclei when compared to controls. Atrogin-1 and MuRF-1 were up-regulated alongside an increase in IL-1β, active NF-κB and a higher ratio of phosphorylated to inactive p38 MAPK. CCL-2 and TNF levels were reduced and IL-6 was unchanged between groups. We observed decreased pSTAT3, unchanged pSTAT1 and Myf5, but increased Pax7, MyoD and myogenin. AiA rabbits had a reduction in myostatin from gastrocnemii and synovium with a congruent decrease in serum myostatin compared to controls. Chronic arthritis induced an RC-like secondary sarcopenia with increased muscle protein breakdown. Elevated IL-1β may trigger proteolysis via elevated NF-κB and p38 MAPK signaling with a compensatory anabolic response suggested by myonuclear expansion, increased Pax7, MyoD and myogenin, reduced pSTAT3 as well as reduced serum, synovial and muscular myostatin.
Collapse
Affiliation(s)
- Robert D Little
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Alfred Hospital, Melbourne, VIC 3004, Australia
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Iván Prieto-Potin
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
- Red Temática de Investigación Cooperativa de Envejecimiento y Fragilidad (RETICEF)-Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Pérez-Baos
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Amanda Villalvilla
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Paula Gratal
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Flavia Cicuttini
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Raquel Largo
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain.
- Red Temática de Investigación Cooperativa de Envejecimiento y Fragilidad (RETICEF)-Instituto de Salud Carlos III, Madrid, Spain.
| | - Gabriel Herrero-Beaumont
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
- Red Temática de Investigación Cooperativa de Envejecimiento y Fragilidad (RETICEF)-Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
41
|
OKAMOTO T, MACHIDA S. Changes in FOXO and proinflammatory cytokines in the late stage of immobilized fast and slow muscle atrophy . Biomed Res 2017; 38:331-342. [DOI: 10.2220/biomedres.38.331] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
| | - Shuichi MACHIDA
- Graduate School of Health and Sports Science, Juntendo University
| |
Collapse
|
42
|
Atherton PJ, Greenhaff PL, Phillips SM, Bodine SC, Adams CM, Lang CH. Control of skeletal muscle atrophy in response to disuse: clinical/preclinical contentions and fallacies of evidence. Am J Physiol Endocrinol Metab 2016; 311:E594-604. [PMID: 27382036 PMCID: PMC5142005 DOI: 10.1152/ajpendo.00257.2016] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 07/05/2016] [Indexed: 01/08/2023]
Abstract
Muscle wasting resulting wholly or in part from disuse represents a serious medical complication that, when prolonged, can increase morbidity and mortality. Although much knowledge has been gained over the past half century, the underlying etiology by which disuse alters muscle proteostasis remains enigmatic. Multidisciplinary and novel methodologies are needed to fill gaps and overcome barriers to improved patient care. The present review highlights seminal concepts from a symposium at Experimental Biology 2016. These proceedings focus on 1) the role of insulin resistance in mediating disuse-induced changes in muscle protein synthesis (MPS) and breakdown (MPB), as well as cross-talk between carbohydrate and protein metabolism; 2) the relative importance of MPS/MPB in mediating involuntary muscle loss in humans and animals; 3) interpretative limitations associated with MPS/MPB "markers," e.g., MuRF1/MAFbx mRNA; and finally, 4) how OMIC technologies can be leveraged to identify molecular pathways (e.g., ATF4, p53, p21) mediating disuse atrophy. This perspective deals primarily with "simple atrophy" due to unloading. Nonetheless, it is likely that disuse is a pervasive contributor to muscle wasting associated with catabolic disease-related atrophy (i.e., due to associated sedentary behaviour of disease burden). Key knowledge gaps and challenges are identified to stimulate discussion and identify opportunities for translational research. Data from animal and human studies highlight both similarities and differences. Integrated preclinical and clinical research is encouraged to better understand the metabolic and molecular underpinnings and translational relevance,for disuse atrophy. These approaches are crucial to clinically prevent or reverse muscle atrophy, thereby reestablishing homeostasis and recovery.
Collapse
Affiliation(s)
- Philip J Atherton
- Royal Derby Medical School, Royal Derby Hospital, Derby, United Kingdom;
| | - Paul L Greenhaff
- MRC-ARUK Centre for Musculoskeletal Ageing Research, ARUK Centre for Sport, Exercise, and Osteoarthritis, School of Life Sciences, The Medical School, University of Nottingham, Nottingham, United Kingdom
| | - Stuart M Phillips
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Sue C Bodine
- Departments of Neurobiology, Physiology and Behavior, and Physiology and Membrane Biology, University of California Davis, Veterans Affairs Northern California Health Care System, Mather, California
| | - Christopher M Adams
- Departments of Internal Mediciane and Molecular Physiology and Biophysics and Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa; and
| | - Charles H Lang
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
43
|
Chrzanowski SM, Batra A, Lee-McMullen B, Vohra RS, Forbes SC, Jiang H, Vandenborne K, Walter GA. Near-Infrared Optical Imaging Noninvasively Detects Acutely Damaged Muscle. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2692-700. [PMID: 27565039 DOI: 10.1016/j.ajpath.2016.06.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 05/28/2016] [Accepted: 06/08/2016] [Indexed: 12/01/2022]
Abstract
Muscle damage is currently assessed through methods such as muscle biopsy, serum biomarkers, functional testing, and imaging procedures, each with its own inherent limitations, and a pressing need for a safe, repeatable, inexpensive, and noninvasive modality to assess the state of muscle health remains. Our aim was to develop and assess near-infrared (NIR) optical imaging as a novel noninvasive method of detecting and quantifying muscle damage. An immobilization-reambulation model was used for inducing muscle damage and recovery in the lower hindlimbs in mice. Confirmation of muscle damage was obtained using in vivo indocyanine green-enhanced NIR optical imaging, magnetic resonance imaging, and ex vivo tissue analysis. The soleus of the immobilized-reambulated hindlimb was found to have a greater amount of muscle damage compared to that in the contralateral nonimmobilized limb, confirmed by in vivo indocyanine green-enhanced NIR optical imaging (3.86-fold increase in radiant efficiency), magnetic resonance imaging (1.41-fold increase in T2), and an ex vivo spectrophotometric assay of indocyanine green uptake (1.87-fold increase in normalized absorbance). Contrast-enhanced NIR optical imaging provides a sensitive, rapid, and noninvasive screening method that can be used for imaging and quantifying muscle damage and recovery in vivo.
Collapse
Affiliation(s)
- Stephen M Chrzanowski
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Abhinandan Batra
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | | | - Ravneet S Vohra
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Sean C Forbes
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | - Huabei Jiang
- Department of Biomedical Engineering, University of Florida, Gainesville, Florida
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | - Glenn A Walter
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida.
| |
Collapse
|
44
|
Onda A, Kono H, Jiao Q, Akimoto T, Miyamoto T, Sawada Y, Suzuki K, Kusakari Y, Minamisawa S, Fukubayashi T. New mouse model of skeletal muscle atrophy using spiral wire immobilization. Muscle Nerve 2016; 54:788-91. [PMID: 27227343 DOI: 10.1002/mus.25202] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2016] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Disuse-induced skeletal muscle atrophy is a serious concern; however, there is not an effective mouse model to elucidate the molecular mechanisms. We developed a noninvasive atrophy model in mice. METHODS After the ankle joints of mice were bandaged into a bilateral plantar flexed position, either bilateral or unilateral hindlimbs were immobilized by wrapping in bonsai steel wire. RESULTS After 3, 5, or 10 days of immobilization of the hip, knee, and ankle, the weight of the soleus and plantaris muscles decreased significantly in both bilateral and unilateral immobilization. MAFbx/atrogin-1 and MuRF1 mRNA was found to have significantly increased in both muscles, consistent with disuse-induced atrophy. Notably, the procedure did not result in either edema or necrosis in the fixed hindlimbs. CONCLUSIONS This method allows repeated, direct access to the immobilized muscle, making it a useful procedure for concurrent application and assessment of various therapeutic interventions. Muscle Nerve 54: 788-791, 2016.
Collapse
Affiliation(s)
- Akiko Onda
- Department of Internal Medicine, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Hajime Kono
- Department of Internal Medicine, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan.
| | - Qibin Jiao
- Department of Cardiology, Hangzhou Cardiovascular Disease Institute, Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| | - Takayuki Akimoto
- Center for Disease Biology and Integrative Medicine, The University of Tokyo, Japan
| | - Toshikazu Miyamoto
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Japan
| | - Yasuhiro Sawada
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Saitama, Japan
| | | | - Yoichiro Kusakari
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| | - Susumu Minamisawa
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| | | |
Collapse
|
45
|
Derbré F, Droguet M, Léon K, Troadec S, Pennec JP, Giroux-Metges MA, Rannou F. Single Muscle Immobilization Decreases Single-Fibre Myosin Heavy Chain Polymorphism: Possible Involvement of p38 and JNK MAP Kinases. PLoS One 2016; 11:e0158630. [PMID: 27383612 PMCID: PMC4934689 DOI: 10.1371/journal.pone.0158630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 06/20/2016] [Indexed: 11/29/2022] Open
Abstract
PURPOSE Muscle contractile phenotype is affected during immobilization. Myosin heavy chain (MHC) isoforms are the major determinant of the muscle contractile phenotype. We therefore sought to evaluate the effects of muscle immobilization on both the MHC composition at single-fibre level and the mitogen-activated protein kinases (MAPK), a family of intracellular signaling pathways involved in the stress-induced muscle plasticity. METHODS The distal tendon of female Wistar rat Peroneus Longus (PL) was cut and fixed to the adjacent bone at neutral muscle length. Four weeks after the surgery, immobilized and contralateral PL were dissociated and the isolated fibres were sampled to determine MHC composition. Protein kinase 38 (p38), extracellular signal-regulated kinases (ERK1/2), and c-Jun- NH2-terminal kinase (JNK) phosphorylations were measured in 6- and 15-day immobilized and contralateral PL. RESULTS MHC distribution in immobilized PL was as follows: I = 0%, IIa = 11.8 ± 2.8%, IIx = 53.0 ± 6.1%, IIb = 35.3 ± 7.3% and I = 6.1 ± 3.9%, IIa = 22.1 ± 3.4%, IIx = 46.6 ± 4.5%, IIb = 25.2 ± 6.6% in contralateral muscle. The MHC composition in immobilized muscle is consistent with a faster contractile phenotype according to the Hill's model of the force-velocity relationship. Immobilized and contralateral muscles displayed a polymorphism index of 31.1% (95% CI 26.1-36.0) and 39.3% (95% CI 37.0-41.5), respectively. Significant increases in p38 and JNK phosphorylation were observed following 6 and 15 days of immobilization. CONCLUSIONS Single muscle immobilization at neutral length induces a shift of MHC composition toward a faster contractile phenotype and decreases the polymorphic profile of single fibres. Activation of p38 and JNK could be a potential mechanism involved in these contractile phenotype modifications during muscle immobilization.
Collapse
Affiliation(s)
- Frédéric Derbré
- Laboratory “Movement Sport and health Sciences”(M2S) -EA1274, University Rennes 2-ENS Rennes, Rennes, France
| | - Mickaël Droguet
- Physiology Department-EA1274 M2S, School of Medicine, Brest, France
| | - Karelle Léon
- Physiology Department-EA1274 M2S, School of Medicine, Brest, France
| | - Samuel Troadec
- Physiology Department-EA1274 M2S, School of Medicine, Brest, France
| | | | | | - Fabrice Rannou
- Physiology Department-EA1274 M2S, School of Medicine, Brest, France
| |
Collapse
|
46
|
Cho SH, Kim JH, Song W. In Vivo Rodent Models of Skeletal Muscle Adaptation to Decreased Use. Endocrinol Metab (Seoul) 2016; 31:31-7. [PMID: 26996420 PMCID: PMC4803558 DOI: 10.3803/enm.2016.31.1.31] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 01/17/2016] [Accepted: 01/25/2016] [Indexed: 12/25/2022] Open
Abstract
Skeletal muscle possesses plasticity and adaptability to external and internal physiological changes. Due to these characteristics, skeletal muscle shows dramatic changes depending on its response to stimuli such as physical activity, nutritional changes, disease status, and environmental changes. Modulation of the rate of protein synthesis/degradation plays an important role in atrophic responses. The purpose of this review is to describe different features of skeletal muscle adaptation with various models of deceased use. In this review, four models were addressed: immobilization, spinal cord transection, hindlimb unloading, and aging. Immobilization is a form of decreased use in which skeletal muscle shows electrical activity, tension development, and motion. These results differ by muscle group. Spinal cord transection was selected to simulate spinal cord injury. Similar to the immobilization model, dramatic atrophy occurs in addition to fiber type conversion in this model. Despite the fact that electromyography shows unremarkable changes in muscle after hindlimb unloading, decreased muscle mass and contractile force are observed. Lastly, aging significantly decreases the numbers of muscle fibers and motor units. Skeletal muscle responses to decreased use include decreased strength, decreased fiber numbers, and fiber type transformation. These four models demonstrated different changes in the skeletal muscle. This review elucidates the different skeletal muscle adaptations in these four decreased use animal models and encourages further studies.
Collapse
Affiliation(s)
- Su Han Cho
- Health and Exercise Science Laboratory, Institute of Sport Science, Seoul National University, Seoul, Korea
| | - Jang Hoe Kim
- Health and Exercise Science Laboratory, Institute of Sport Science, Seoul National University, Seoul, Korea
| | - Wook Song
- Health and Exercise Science Laboratory, Institute of Sport Science, Seoul National University, Seoul, Korea
- Institute on Aging, Seoul National University, Seoul, Korea.
| |
Collapse
|
47
|
Sun S, Henriksen K, Karsdal MA, Byrjalsen I, Rittweger J, Armbrecht G, Belavy DL, Felsenberg D, Nedergaard AF. Collagen Type III and VI Turnover in Response to Long-Term Immobilization. PLoS One 2015; 10:e0144525. [PMID: 26641456 PMCID: PMC4671681 DOI: 10.1371/journal.pone.0144525] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 11/19/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Muscle mass and function are perturbed by immobilization and remobilization. When muscle mass changes, the quality and quantity of the extracellular matrix protein, particularly the collagens, change with it. In this study, we investigated the temporal profile of three peptide biomarkers derived from turnover of collagen type III and type VI in a long-term immobilization and remobilization study. We also compared individual biomarker levels with Lean body Mass (LBM) and changes therein, hypothesizing that these biomarkers would be biomarkers of the remodeling processes associated with immobilization and/or remobilization. METHODS In the Berlin bed rest study, 20 young men were recruited and randomly assigned to 8-week's strict bed rest with or without resistive vibration exercise countermeasure. We measured three neo-epitope ELISA kits in the serum samples of this study: Pro-C3, measured the synthesis of collagen type III; Pro-C6, measured the synthesis of collagen type VI; and C6M measured the degradation of collagen type VI induced by MMP-2 and MMP-9 cleavage. RESULTS Pro-C3 and Pro-C6 biomarkers are up-regulated with both immobilization and remobilization, whereas C6M is hardly affected at all. We found that Pro-C3 and C6M levels are related to LBM at baseline and that high levels of Pro-C6 are associated with smaller changes in muscle mass during both immobilization and remobilization. CONCLUSION The Pro-C3 and-C6 biomarkers change likely reflect remodeling changes in response to unloading or reloading, whereas C6M does not appear to respond to unloading. Pro-C3 and C6M levels correlate with LBM at baseline, while Pro-C6 is related to the anabolic and catabolic responses to unloading and reloading.
Collapse
Affiliation(s)
- Shu Sun
- Nordic Bioscience Biomarkers and Research, Herlev, Denmark
- * E-mail:
| | - Kim Henriksen
- Nordic Bioscience Biomarkers and Research, Herlev, Denmark
| | | | | | - Jörn Rittweger
- Institute of Aerospace Medicine, German Aerospace Center, Köln, Germany
| | - Gabriele Armbrecht
- Center for Muscle and Bone Research, Charité Universitätsmedizin Berlin and Free and Humboldt Universities, Berlin, Germany
| | - Daniel L. Belavy
- Center for Muscle and Bone Research, Charité Universitätsmedizin Berlin and Free and Humboldt Universities, Berlin, Germany
- Deakin University, Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Burwood, Victoria, 3125, Australia
| | - Dieter Felsenberg
- Center for Muscle and Bone Research, Charité Universitätsmedizin Berlin and Free and Humboldt Universities, Berlin, Germany
| | - Anders F. Nedergaard
- Nordic Bioscience Biomarkers and Research, Herlev, Denmark
- Institute of Sports Medicine Copenhagen, Bispebjerg Hospital, Copenhagen NW, Denmark
| |
Collapse
|
48
|
Kang C, Goodman CA, Hornberger TA, Ji LL. PGC-1α overexpression by in vivo transfection attenuates mitochondrial deterioration of skeletal muscle caused by immobilization. FASEB J 2015; 29:4092-106. [PMID: 26178167 DOI: 10.1096/fj.14-266619] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 01/20/2015] [Indexed: 12/29/2022]
Abstract
Prolonged immobilization (IM) causes skeletal muscle atrophy characterized by mitochondrial deterioration and proteolysis. Muscle remobilization (RM) increases reactive oxygen species generation, proinflammatory cytokine expression, and oxidative stress, preventing muscle from quick recovery. Thus, we hypothesized that overexpression of peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) via in vivo transfection would promote mitochondrial biogenesis and antioxidant defense, thus ameliorating the aforementioned deteriorations in a mouse model with 14-d IM followed by 5-d RM. PGC-1α transfection in tibialis anterior muscle resulted in a 7.2- and 4-fold increase in PGC-1α content in cytosol and nucleus, respectively. Mitochondrial biogenic (cytochrome c, mitochondrial transcription factor A), morphologic (mitochondrial density, mDNA/nDNA ratio), and functional (cytochrome c oxidase activity, ATP synthesis rate) markers, as well as fiber cross-sectional area, significantly increased in IM-RM muscle by PGC-1α overexpression. These effects were accompanied by an 18% decrease in H2O2, 30% decrease in nuclear factor-κB-DNA binding, and 25% reduction of IL-1β and-6 production in IM-RM muscle. There was a 34% increase in superoxide dismutase-2 activity, along with a 3.5-fold increase in NAD-dependent deacetylase sirtuin-3 expression caused by enhanced PGC-1α-estrogen-related receptor α binding. Our findings highlighted the importance of PGC-1α in protecting muscle from metabolic and redox disturbances caused by IM.
Collapse
Affiliation(s)
- Chounghun Kang
- *Laboratory of Physiological Hygiene and Exercise Science, School of Kinesiology, University of Minnesota at Twin Cities, Minneapolis, Minnesota, USA; and Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Craig A Goodman
- *Laboratory of Physiological Hygiene and Exercise Science, School of Kinesiology, University of Minnesota at Twin Cities, Minneapolis, Minnesota, USA; and Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Troy A Hornberger
- *Laboratory of Physiological Hygiene and Exercise Science, School of Kinesiology, University of Minnesota at Twin Cities, Minneapolis, Minnesota, USA; and Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Li Li Ji
- *Laboratory of Physiological Hygiene and Exercise Science, School of Kinesiology, University of Minnesota at Twin Cities, Minneapolis, Minnesota, USA; and Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
49
|
Files DC, Sanchez MA, Morris PE. A conceptual framework: the early and late phases of skeletal muscle dysfunction in the acute respiratory distress syndrome. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2015; 19:266. [PMID: 26134116 PMCID: PMC4488983 DOI: 10.1186/s13054-015-0979-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Patients with acute respiratory distress syndrome (ARDS) often develop severe diaphragmatic and limb skeletal muscle dysfunction. Impaired muscle function in ARDS is associated with increased mortality, increased duration of mechanical ventilation, and functional disability in survivors. In this review, we propose that muscle dysfunction in ARDS can be categorized into an early and a late phase. These early and late phases are based on the timing in relationship to lung injury and the underlying mechanisms. The early phase occurs temporally with the onset of lung injury, is driven by inflammation and disuse, and is marked predominantly by muscle atrophy from increased protein degradation. The ubiquitin-proteasome, autophagy, and calpain-caspase pathways have all been implicated in early-phase muscle dysfunction. Late-phase muscle weakness persists in many patients despite resolution of lung injury and cessation of ongoing acute inflammation-driven muscle atrophy. The clinical characteristics and mechanisms underlying late-phase muscle dysfunction do not involve the massive protein degradation and atrophy of the early phase and may reflect a failure of the musculoskeletal system to regain homeostatic balance. Owing to these underlying mechanistic differences, therapeutic interventions for treating muscle dysfunction in ARDS may differ during the early and late phases. Here, we review clinical and translational investigations of muscle dysfunction in ARDS, placing them in the conceptual framework of the early and late phases. We hypothesize that this conceptual model will aid in the design of future mechanistic and clinical investigations of the skeletal muscle system in ARDS and other critical illnesses.
Collapse
Affiliation(s)
- D Clark Files
- Section on Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA. .,Critical Illness Injury and Recovery Research Center Chadwick Miller MD Department of Emergency Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
| | - Michael A Sanchez
- Section on Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Peter E Morris
- Section on Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA.,Critical Illness Injury and Recovery Research Center Chadwick Miller MD Department of Emergency Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| |
Collapse
|
50
|
Files DC, Liu C, Pereyra A, Wang ZM, Aggarwal NR, D'Alessio FR, Garibaldi BT, Mock JR, Singer BD, Feng X, Yammani RR, Zhang T, Lee AL, Philpott S, Lussier S, Purcell L, Chou J, Seeds M, King LS, Morris PE, Delbono O. Therapeutic exercise attenuates neutrophilic lung injury and skeletal muscle wasting. Sci Transl Med 2015; 7:278ra32. [PMID: 25761888 PMCID: PMC4820823 DOI: 10.1126/scitranslmed.3010283] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Early mobilization of critically ill patients with the acute respiratory distress syndrome (ARDS) has emerged as a therapeutic strategy that improves patient outcomes, such as the duration of mechanical ventilation and muscle strength. Despite the apparent efficacy of early mobility programs, their use in clinical practice is limited outside of specialized centers and clinical trials. To evaluate the mechanisms underlying mobility therapy, we exercised acute lung injury (ALI) mice for 2 days after the instillation of lipopolysaccharides into their lungs. We found that a short duration of moderate intensity exercise in ALI mice attenuated muscle ring finger 1 (MuRF1)-mediated atrophy of the limb and respiratory muscles and improved limb muscle force generation. Exercise also limited the influx of neutrophils into the alveolar space through modulation of a coordinated systemic neutrophil chemokine response. Granulocyte colony-stimulating factor (G-CSF) concentrations were systemically reduced by exercise in ALI mice, and in vivo blockade of the G-CSF receptor recapitulated the lung exercise phenotype in ALI mice. Additionally, plasma G-CSF concentrations in humans with acute respiratory failure (ARF) undergoing early mobility therapy showed greater decrements over time compared to control ARF patients. Together, these data provide a mechanism whereby early mobility therapy attenuates muscle wasting and limits ongoing alveolar neutrophilia through modulation of systemic neutrophil chemokines in lung-injured mice and humans.
Collapse
Affiliation(s)
- D Clark Files
- Department of Internal Medicine-Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA. Wake Forest Critical Illness, Injury and Recovery Research Center, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| | - Chun Liu
- Department of Internal Medicine-Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Andrea Pereyra
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA. National Scientific and Technical Research Council (CONICET) and School of Medicine, National University of La Plata, 1900 La Plata, Argentina
| | - Zhong-Min Wang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Neil R Aggarwal
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Asthma and Allergy Center, Baltimore, MD 21205, USA
| | - Franco R D'Alessio
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Asthma and Allergy Center, Baltimore, MD 21205, USA
| | - Brian T Garibaldi
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Asthma and Allergy Center, Baltimore, MD 21205, USA
| | - Jason R Mock
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Asthma and Allergy Center, Baltimore, MD 21205, USA
| | - Benjamin D Singer
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Asthma and Allergy Center, Baltimore, MD 21205, USA
| | - Xin Feng
- Department of Otolaryngology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Raghunatha R Yammani
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Tan Zhang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Amy L Lee
- Department of Internal Medicine-Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Sydney Philpott
- Department of Internal Medicine-Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Stephanie Lussier
- Department of Internal Medicine-Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Lina Purcell
- Department of Internal Medicine-Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Jeff Chou
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Michael Seeds
- Department of Internal Medicine-Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA. Wake Forest Critical Illness, Injury and Recovery Research Center, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Landon S King
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Asthma and Allergy Center, Baltimore, MD 21205, USA
| | - Peter E Morris
- Department of Internal Medicine-Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA. Wake Forest Critical Illness, Injury and Recovery Research Center, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|