1
|
Cheng Y, Lin S, Cao Z, Yu R, Fan Y, Chen J. The role of chronic low-grade inflammation in the development of sarcopenia: Advances in molecular mechanisms. Int Immunopharmacol 2025; 147:114056. [PMID: 39799736 DOI: 10.1016/j.intimp.2025.114056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 12/16/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
With the exacerbation of global population aging, sarcopenia has become an increasingly recognized public health issue. Sarcopenia, characterized by a progressive decline in skeletal muscle mass, strength, and function, significantly impacts the quality of life in the elderly. Herein, we explore the role of chroniclow-gradeinflammation in the development of sarcopenia and its underlying molecular mechanisms, including chronic inflammation-associated signaling pathways, immunosenescence, obesity and lipid infiltration, gut microbiota dysbiosis and intestinal barrier disruption, and the decline of satellite cells. The interplay and interaction of these molecular mechanisms provide new perspectives on the complexity of the pathogenesis of sarcopenia and offer a theoretical foundation for the development of future therapeutic strategies.
Collapse
Affiliation(s)
- Ying Cheng
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040 China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040 China
| | - Shangjin Lin
- Department of Orthopedics, Huadong Hospital Affiliated to Fudan University, Shanghai 200040 China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040 China
| | - Ziyi Cao
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040 China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040 China
| | - Runzhi Yu
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040 China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040 China
| | - Yongqian Fan
- Department of Orthopedics, Huadong Hospital Affiliated to Fudan University, Shanghai 200040 China.
| | - Jie Chen
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040 China.
| |
Collapse
|
2
|
Casey TM, Gouveia KM, Beckett LM, Markworth JF, Boerman JP. Molecular signatures of longissimus dorsi differ between dairy cattle based on prepartum muscle reserves and branched-chain volatile fatty acid supplementation. Physiol Genomics 2024; 56:597-608. [PMID: 38975796 DOI: 10.1152/physiolgenomics.00060.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024] Open
Abstract
Dairy cattle with high (HM) versus low muscle (LM) reserves as determined by longissimus dorsi muscle depth (LDD) in late gestation exhibit differential muscle mobilization related to subsequent milk production. Moreover, branched-chain volatile fatty acid (BCVFA) supplementation increased blood glucose levels. We hypothesized that differences in HM and LM reflect distinct muscle metabolism and that BCVFA supplementation altered metabolic pathways. At 42 days before expected calving (BEC), Holstein dairy cows were enrolled in a 2 × 2 factorial study of diet and muscle reserves, by assignment to control (CON)- or BCVFA-supplemented diets and LDD of HM (>4.6 cm) or LM (≤4.6 cm) groups: HM-CON (n = 13), HM-BCVFA (n = 10), LM-CON (n = 9), and LM-BCVFA (n = 9). Longisumus dorsi muscle was biopsied at 21 days BEC, total RNA was isolated, and protein-coding gene expression was measured with RNA sequencing. Between HM and LM, 713 genes were differentially expressed and 481 between BCVFA and CON (P < 0.05). Transcriptional signatures indicated differential distribution of type II fibers between groups, with MYH1 greater in LM cattle and MYH2 greater in HM cattle (P < 0.05). Signatures of LM cattle relative to HM cattle indicated greater activation of autophagy, ubiquitin-proteasome, and Ca2+-calpain pathways. HM cattle displayed greater expression of genes that encode extracellular matrix proteins and factors that regulate their proteolysis and turnover. BCVFA modified transcriptomes by increasing expression of genes that regulate fatty acid degradation and flux of carbons into the tricarboxylic acid cycle as acetyl CoA. Molecular signatures support distinct metabolic strategies between LM and HM cattle and that BCVFA supplementation increased substrates for energy generation.NEW & NOTEWORTHY Muscle biopsies of the longissimus dorsi of prepartum dairy cattle indicate that molecular signatures support distinct metabolic strategies between low- and high-muscle cattle and that branched-chain volatile fatty acid supplementation increased substrates for energy generation.
Collapse
Affiliation(s)
- Theresa M Casey
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, United States
| | - Kyrstin M Gouveia
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, United States
| | - Linda M Beckett
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, United States
| | - James F Markworth
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, United States
| | - Jacquelyn P Boerman
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, United States
| |
Collapse
|
3
|
Du J, Wu Q, Bae EJ. Epigenetics of Skeletal Muscle Atrophy. Int J Mol Sci 2024; 25:8362. [PMID: 39125931 PMCID: PMC11312722 DOI: 10.3390/ijms25158362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Skeletal muscle atrophy, characterized by diminished muscle strength and mass, arises from various causes, including malnutrition, aging, nerve damage, and disease-related secondary atrophy. Aging markedly escalates the prevalence of sarcopenia. Concurrently, the incidence of muscle atrophy significantly rises among patients with chronic ailments such as heart failure, diabetes, and chronic obstructive pulmonary disease (COPD). Epigenetics plays a pivotal role in skeletal muscle atrophy. Aging elevates methylation levels in the promoter regions of specific genes within muscle tissues. This aberrant methylation is similarly observed in conditions like diabetes, neurological disorders, and cardiovascular diseases. This study aims to explore the relationship between epigenetics and skeletal muscle atrophy, thereby enhancing the understanding of its pathogenesis and uncovering novel therapeutic strategies.
Collapse
Affiliation(s)
- Jiacheng Du
- Department of Biochemistry, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea
| | - Qian Wu
- Department of Biochemistry, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea
| | - Eun Ju Bae
- School of Pharmacy and Institute of New Drug Development, Jeonbuk National University, Jeonju 54896, Republic of Korea
| |
Collapse
|
4
|
Olthof MGL, Hasler A, Valdivieso P, Flück M, Gerber C, Gehrke R, Klein K, von Rechenberg B, Snedeker JG, Wieser K. Poly(ADP-Ribose) Polymerases-Inhibitor Talazoparib Inhibits Muscle Atrophy and Fatty Infiltration in a Tendon Release Infraspinatus Sheep Model: A Pilot Study. Metabolites 2024; 14:187. [PMID: 38668315 PMCID: PMC11051840 DOI: 10.3390/metabo14040187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Structural muscle changes, including muscle atrophy and fatty infiltration, follow rotator cuff tendon tear and are associated with a high repair failure rate. Despite extensive research efforts, no pharmacological therapy is available to successfully prevent both muscle atrophy and fatty infiltration after tenotomy of tendomuscular unit without surgical repair. Poly(ADP-ribose) polymerases (PARPs) are identified as a key transcription factors involved in the maintenance of cellular homeostasis. PARP inhibitors have been shown to influence muscle degeneration, including mitochondrial hemostasis, oxidative stress, inflammation and metabolic activity, and reduced degenerative changes in a knockout mouse model. Tenotomized infraspinatus were assessed for muscle degeneration for 16 weeks using a Swiss Alpine sheep model (n = 6). All sheep received daily oral administration of 0.5 mg Talazoparib. Due to animal ethics, the treatment group was compared with three different controls from prior studies of our institution. To mitigate potential batch heterogeneity, PARP-I was evaluated in comparison with three distinct control groups (n = 6 per control group) using the same protocol without treatment. The control sheep were treated with an identical study protocol without Talazoparib treatment. Muscle atrophy and fatty infiltration were evaluated at 0, 6 and 16 weeks post-tenotomy using DIXON-MRI. The controls and PARP-I showed a significant (control p < 0.001, PARP-I p = 0.01) decrease in muscle volume after 6 weeks. However, significantly less (p = 0.01) atrophy was observed in PARP-I after 6 weeks (control 1: 76.6 ± 8.7%; control 2: 80.3 ± 9.3%, control 3: 73.8 ± 6.7% vs. PARP-I: 90.8 ± 5.1% of the original volume) and 16 weeks (control 1: 75.7 ± 9.9; control 2: 74.2 ± 5.6%; control 3: 75.3 ± 7.4% vs. PARP-I 93.3 ± 10.6% of the original volume). All experimental groups exhibited a statistically significant (p < 0.001) augmentation in fatty infiltration following a 16-week period when compared to the initial timepoint. However, the PARP-I showed significantly less fatty infiltration (p < 0.003) compared to all controls (control 1: 55.6 ± 6.7%, control 2: 53.4 ± 9.4%, control 3: 52.0 ± 12.8% vs. PARP-I: 33.5 ± 8.4%). Finally, a significantly (p < 0.04) higher proportion and size of fast myosin heavy chain-II fiber type was observed in the treatment group. This study shows that PARP-inhibition with Talazoparib inhibits the progression of both muscle atrophy and fatty infiltration over 16 weeks in retracted sheep musculotendinous units.
Collapse
Affiliation(s)
- Maurits G. L. Olthof
- Department of Orthopaedics, Balgrist, University of Zurich, Forchstrasse 340, 8008 Zurich, Switzerland; (A.H.); (C.G.); (J.G.S.); (K.W.)
| | - Anita Hasler
- Department of Orthopaedics, Balgrist, University of Zurich, Forchstrasse 340, 8008 Zurich, Switzerland; (A.H.); (C.G.); (J.G.S.); (K.W.)
| | - Paola Valdivieso
- Laboratory for Muscle Plasticity, Department of Orthopedics, Balgrist Campus, University of Zurich, Forchstrasse 340, 8008 Zurich, Switzerland; (P.V.); (M.F.)
| | - Martin Flück
- Laboratory for Muscle Plasticity, Department of Orthopedics, Balgrist Campus, University of Zurich, Forchstrasse 340, 8008 Zurich, Switzerland; (P.V.); (M.F.)
| | - Christian Gerber
- Department of Orthopaedics, Balgrist, University of Zurich, Forchstrasse 340, 8008 Zurich, Switzerland; (A.H.); (C.G.); (J.G.S.); (K.W.)
| | - Rieke Gehrke
- Musculoskeletal Research Unit, Center for Applied Biotechnology and Molecular Medicine, Equine Department, Vetsuisse Faculty, Winterthurerstrasse 190, 8057 Zurich, Switzerland; (R.G.); (K.K.); (B.v.R.)
| | - Karina Klein
- Musculoskeletal Research Unit, Center for Applied Biotechnology and Molecular Medicine, Equine Department, Vetsuisse Faculty, Winterthurerstrasse 190, 8057 Zurich, Switzerland; (R.G.); (K.K.); (B.v.R.)
| | - Brigitte von Rechenberg
- Musculoskeletal Research Unit, Center for Applied Biotechnology and Molecular Medicine, Equine Department, Vetsuisse Faculty, Winterthurerstrasse 190, 8057 Zurich, Switzerland; (R.G.); (K.K.); (B.v.R.)
| | - Jess G. Snedeker
- Department of Orthopaedics, Balgrist, University of Zurich, Forchstrasse 340, 8008 Zurich, Switzerland; (A.H.); (C.G.); (J.G.S.); (K.W.)
- Institute for Biomechanics, ETH Zurich, Gloriastrasse 37/39, 8092 Zürich, Switzerland
| | - Karl Wieser
- Department of Orthopaedics, Balgrist, University of Zurich, Forchstrasse 340, 8008 Zurich, Switzerland; (A.H.); (C.G.); (J.G.S.); (K.W.)
| |
Collapse
|
5
|
Zhang HJ, Wang BH, Wang X, Huang CP, Xu SM, Wang JL, Huang TE, Xiao WL, Tian XL, Lan XQ, Wang QQ, Xiang Y. Handelin alleviates cachexia- and aging-induced skeletal muscle atrophy by improving protein homeostasis and inhibiting inflammation. J Cachexia Sarcopenia Muscle 2024; 15:173-188. [PMID: 38009816 PMCID: PMC10834327 DOI: 10.1002/jcsm.13381] [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: 10/13/2022] [Revised: 09/07/2023] [Accepted: 09/25/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Handelin is a bioactive compound from Chrysanthemum indicum L. that improves motor function and muscle integrity during aging in Caenorhabditis elegans. This study aimed to further evaluate the protective effects and molecular mechanisms of handelin in a mouse muscle atrophy model induced by cachexia and aging. METHODS A tumour necrosis factor (TNF)-α-induced atrophy model was used to examine handelin activity in cultured C2C12 myotubes in vitro. Lipopolysaccharide (LPS)-treated 8-week-old model mice and 23-month-old (aged) mice were used to examine the therapeutic effects of handelin on cachexia- and aging-induced muscle atrophy, respectively, in vivo. Protein and mRNA expressions were analysed by Western blotting, ELISA and quantitative PCR, respectively. Skeletal muscle mass was measured by histological analysis. RESULTS Handelin treatment resulted in an upregulation of protein levels of early (MyoD and myogenin) and late (myosin heavy chain, MyHC) differentiation markers in C2C12 myotubes (P < 0.05), and enhanced mitochondrial respiratory (P < 0.05). In TNF-α-induced myotube atrophy model, handelin maintained MyHC protein levels, increased insulin-like growth factor (Igf1) mRNA expression and phosphorylated protein kinase B protein levels (P < 0.05). Handelin also reduced atrogin-1 expression, inhibited nuclear factor-κB activation and reduced mRNA levels of interleukin (Il)6, Il1b and chemokine ligand 1 (Cxcl1) (P < 0.05). In LPS-treated mice, handelin increased body weight (P < 0.05), the weight (P < 0.01) and cross-sectional area (CSA) of the soleus muscle (P < 0.0001) and improved motor function (P < 0.05). In aged mice, handelin slightly increased the weight of the tibialis anterior muscle (P = 0.06) and CSA of the tibialis anterior and gastrocnemius muscles (P < 0.0001). In the tibialis anterior muscle of aged mice, handelin upregulated mRNA levels of Igf1 (P < 0.01), anti-inflammatory cytokine Il10 (P < 0.01), mitochondrial biogenesis genes (P < 0.05) and antioxidant-related enzymes (P < 0.05) and strengthened Sod and Cat enzyme activity (P < 0.05). Handelin also reduced lipid peroxidation and protein carbonylation, downregulated mRNA levels of Fbxo32, Mstn, Cxcl1, Il1b and Tnf (P < 0.05), and decreased IL-1β levels in serum (P < 0.05). Knockdown of Hsp70 or using an Hsp70 inhibitor abolished the ameliorating effects of handelin on myotube atrophy. CONCLUSIONS Handelin ameliorated cachexia- and aging-induced skeletal muscle atrophy in vitro and in vivo, by maintaining homeostasis of protein synthesis and degradation, possibly by inhibiting inflammation. Handelin is a potentially promising drug candidate for the treatment of muscle wasting.
Collapse
Affiliation(s)
- Hui-Jie Zhang
- Department of Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Ben-Hui Wang
- Department of Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Xiang Wang
- Department of Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Chun-Ping Huang
- Department of Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Si-Man Xu
- Department of Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Jia-Li Wang
- Department of Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Tian-E Huang
- Department of Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Wan-Li Xiao
- Department of Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Xiao-Li Tian
- Department of Aging and Vascular Diseases, Human Aging Research Institute and School of Life Science, Nanchang University and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Xin-Qiang Lan
- Department of Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Qi-Quan Wang
- Department of Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Yang Xiang
- Department of Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| |
Collapse
|
6
|
Shui XP, Ye F, Li CY, Zhang X, Wang MJ, Li B, Chen K, Liao YY. Effects of millimeter-wave for preventing joint stiffness in the immobilized knee rat model. Knee 2023; 42:236-245. [PMID: 37086540 DOI: 10.1016/j.knee.2023.03.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 03/14/2023] [Accepted: 03/30/2023] [Indexed: 04/24/2023]
Abstract
AIM To explore the effects and mechanism of millimeter-wave treatment on the development of joint stiffness in the immobilized knee rat model. METHODS Twenty-four Sprague-Dawley (SD) rats were randomly divided into the control group (O, n = 8), the surgical control group (OC, n = 8), and the millimeter-wave treatment group (MO, n = 8). After immobilized knee modeling, the knee mobility and quadriceps diameter was measured at the 6th week. Hematoxylin and eosin and Masson staining were performed to detect the pathology and fibrous lesions of the knee joint. Furthermore, the expression of TGF-β1 and Collagen I was quantified by immunohistochemical assay in the knee capsule, and Western blotting was performed to quantify the protein expression of NF-κB and MuRF1 in skeletal muscle. RESULTS Compared with the O group, knee mobility, and quadriceps diameter was decreased (P < 0.01), and articular capsule fibrosis and quadriceps atrophy occurred in all rats with fixed knee joints. Compared with the OC group, millimeter-wave treatment significantly increased articular mobility and the quadriceps diameter; and improved the fibrotic lesions of the joint capsule and quadriceps atrophy. Moreover, levels of TGF-β1, Collagen I, and MuRF1 were upregulated (P < 0.01) by knee immobilization, and collagen fiber content in the articular capsule was also increased (P < 0.01). However, millimeter-wave treatment reversed it. The most noteworthy result was that NF-κB expression was not significantly different in all groups. CONCLUSION Millimeter-wave treatment reversed joint contracture and quadriceps atrophy caused by joint fixation, inhibited TGF-β1 and Collagen I protein expression of the joint capsule and reduced MuRF1 expression of the quadriceps muscle, thereby inhibiting the development of joint stiffness.
Collapse
Affiliation(s)
- Xiao-Ping Shui
- Department of Acupuncture and Massage, Sichuan College of Traditional Chinese Medicine, Mian-yang 621000, Sichuan, China; Department of Rehabilitation, Mian-yang Orthopedic Hospital, Mian-yang 621000, Sichuan, China.
| | - Feng Ye
- Department of Acupuncture and Massage, Sichuan College of Traditional Chinese Medicine, Mian-yang 621000, Sichuan, China
| | - Chun-Ying Li
- Department of Geriatric Medicine, Mian-yang Hospital of Traditional Chinese Medicine, Mian-yang 621000, Sichuan, China
| | - Xin Zhang
- Department of Rehabilitation Medicine, Sichuan Provincial Orthopedic Hospital, Chengdu 610000, Sichuan, China
| | - Min-Jia Wang
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu 610000, Sichuan, China
| | - Bin Li
- Department of Rehabilitation, Mian-yang Orthopedic Hospital, Mian-yang 621000, Sichuan, China
| | - Ke Chen
- Department of Rehabilitation Medicine, Sichuan Provincial Orthopedic Hospital, Chengdu 610000, Sichuan, China
| | - Ying-Ying Liao
- Department of Rehabilitation Medicine, Sichuan Provincial Orthopedic Hospital, Chengdu 610000, Sichuan, China
| |
Collapse
|
7
|
The Inhibition of Autophagy and Pyroptosis by an Ethanol Extract of Nelumbo nucifera Leaf Contributes to the Amelioration of Dexamethasone-Induced Muscle Atrophy. Nutrients 2023; 15:nu15040804. [PMID: 36839161 PMCID: PMC9965294 DOI: 10.3390/nu15040804] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Muscle atrophy is characterized by a decline in muscle mass and function. Excessive glucocorticoids in the body due to aging or drug treatment can promote muscle wasting. In this study, we investigated the preventive effect of Nelumbo nucifera leaf (NNL) ethanolic extract on muscle atrophy induced by dexamethasone (DEX), a synthetic glucocorticoid, in mice and its underlying mechanisms. The administration of NNL extract increased weight, cross-sectional area, and grip strength of quadriceps (QD) and gastrocnemius (GA) muscles in DEX-induced muscle atrophy in mice. The NNL extract administration decreased the expression of muscle atrophic factors, such as muscle RING-finger protein-1 and atrogin-1, and autophagy factors, such as Beclin-1, microtubule-associated protein 1A/1B-light chain 3 (LC3-I/II), and sequestosome 1 (p62/SQSTM1) in DEX-injected mice. DEX injection increased the protein expression levels of NOD-like receptor pyrin domain-containing protein 3 (NLRP3), cleaved-caspase-1, interleukin-1beta (IL-1β), and cleaved-gasdermin D (GSDMD), which were significantly reduced by NNL extract administration (500 mg/kg/day). In vitro studies using C2C12 myotubes also revealed that NNL extract treatment inhibited the DEX-induced increase in autophagy factors, pyroptosis-related factors, and NF-κB. Overall, the NNL extract prevented DEX-induced muscle atrophy by downregulating the ubiquitin-proteasome system, autophagy pathway, and GSDMD-mediated pyroptosis pathway, which are involved in muscle degradation.
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
|
Hwang J, Thurmond DC. Exocytosis Proteins: Typical and Atypical Mechanisms of Action in Skeletal Muscle. Front Endocrinol (Lausanne) 2022; 13:915509. [PMID: 35774142 PMCID: PMC9238359 DOI: 10.3389/fendo.2022.915509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/11/2022] [Indexed: 11/18/2022] Open
Abstract
Insulin-stimulated glucose uptake in skeletal muscle is of fundamental importance to prevent postprandial hyperglycemia, and long-term deficits in insulin-stimulated glucose uptake underlie insulin resistance and type 2 diabetes. Skeletal muscle is responsible for ~80% of the peripheral glucose uptake from circulation via the insulin-responsive glucose transporter GLUT4. GLUT4 is mainly sequestered in intracellular GLUT4 storage vesicles in the basal state. In response to insulin, the GLUT4 storage vesicles rapidly translocate to the plasma membrane, where they undergo vesicle docking, priming, and fusion via the high-affinity interactions among the soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) exocytosis proteins and their regulators. Numerous studies have elucidated that GLUT4 translocation is defective in insulin resistance and type 2 diabetes. Emerging evidence also links defects in several SNAREs and SNARE regulatory proteins to insulin resistance and type 2 diabetes in rodents and humans. Therefore, we highlight the latest research on the role of SNAREs and their regulatory proteins in insulin-stimulated GLUT4 translocation in skeletal muscle. Subsequently, we discuss the novel emerging role of SNARE proteins as interaction partners in pathways not typically thought to involve SNAREs and how these atypical functions reveal novel therapeutic targets for combating peripheral insulin resistance and diabetes.
Collapse
Affiliation(s)
| | - Debbie C. Thurmond
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| |
Collapse
|
10
|
Dunn A, Haas G, Madsen J, Ziemkiewicz N, Au J, Johnson D, West C, Chauvin H, Gagyi SM, Garg K. Biomimetic sponges improve functional muscle recovery following composite trauma. J Orthop Res 2022; 40:1039-1052. [PMID: 34289186 PMCID: PMC8776909 DOI: 10.1002/jor.25143] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/02/2021] [Accepted: 06/24/2021] [Indexed: 02/04/2023]
Abstract
There is a dearth of therapies that are safe and effective for the treatment of volumetric muscle loss (VML), defined as the surgical or traumatic loss of muscle tissue, resulting in functional impairment. To address this gap in orthopedic care, we developed a porous sponge-like scaffold composed of extracellular matrix (ECM) proteins (e.g., gelatin, collagen, and laminin-111) and an immunosuppressant drug, FK-506. While the majority of VML injuries occur in orthopedic trauma cases, preclinical models typically study muscle injuries in isolation without a concomitant bone fracture. The goal of this study was to investigate the extent to which FK506 loaded biomimetic sponges support functional muscle regeneration and fracture healing in a composite trauma model involving VML injury to the tibialis anterior muscle and osteotomy (OST) to the tibia. In this model, implantation of the FK-506 loaded biomimetic sponges limited the extent of inflammation while increasing the total number of myofibers, mean myofiber cross-sectional area, myosin-to-collagen ratio, and peak isometric torque compared to untreated VML+OST muscles on Day 28. Although all tibia fractures were bridged by Day 28 post-injury, fracture healing was impaired in response to an adjacent VML injury. Sponge treatment increased bone callus volume, yet the bridged mineralized bone volume was not significantly different. Taken together, these results suggest that biomimetic sponges primarily benefitted muscle repair and may provide a promising therapy for traumatized muscle.
Collapse
Affiliation(s)
- Andrew Dunn
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University
| | - Gabriel Haas
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University
| | - Joshua Madsen
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University
| | - Natalia Ziemkiewicz
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University
| | - Jeffrey Au
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University
| | - David Johnson
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University
| | - Charles West
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University
| | - Hannah Chauvin
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University
| | | | - Koyal Garg
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University,Address correspondence to: Koyal Garg, PhD, Assistant Professor, Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, 3507 Lindell Blvd, St. Louis, MO 63103, Phone: 314.977.1434,
| |
Collapse
|
11
|
Yin D, Lin D, Xie Y, Gong A, Jiang P, Wu J. Neuregulin-1β Alleviates Sepsis-Induced Skeletal Muscle Atrophy by Inhibiting Autophagy via AKT/mTOR Signaling Pathway in Rats. Shock 2022; 57:397-407. [PMID: 34559744 DOI: 10.1097/shk.0000000000001860] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Several studies have shown that excessive protein degradation is a major cause of skeletal muscle atrophy induced by sepsis, and autophagy is the main pathway participating in protein degradation. However, the role of autophagy in sepsis is still controversial. Previously, we found that neuregulin-1β (NRG-1β) alleviated sepsis-induced diaphragm atrophy through the phosphatidylinositol-3 kinase signaling pathway. Akt/mechanistic target of rapamycin (mTOR) is a classic signaling pathway to regulate autophagy, which maintains intracellular homeostasis. This study aimed to investigate whether NRG-1β could alleviate sepsis-induced skeletal muscle atrophy by regulating autophagy. METHODS L6 rat myoblast cells were differentiated using 2% fetal bovine serum into myotubes, which were divided into four groups: Con group treated with normal serum; Sep group treated with septic serum to form a sepsis cell model; septic serum + NRG-1β (SN) group treated with septic serum for 24 h followed by injection with NRG-1β and incubation for another 48 h; and serum+NRG-1β+LY294002 group, in which the PI3K inhibitor LY294002 was added 30 min before NRG-1β, and other treatments were similar to those in SN group. Effects of NRG-1β were also evaluated in vivo using Sprague-Dawley (SD) rats, in which sepsis was induced by cecal ligation and puncture (CLP). RESULTS In L6 myotubes treated with septic serum, the expression of autophagy-related proteins UNC-51 like kinase 1, p-Beclin-1, and Beclin-1, and the ratio of LC3B II/I were highly increased, while protein p62 expression was decreased, indicating that autophagy was excessively activated. Moreover, NRG-1 expression was decreased, as detected by confocal immunofluorescence and western blotting. Upon exogenous addition of NRG-1β, autophagy was inhibited by the activation of Akt/mTOR signaling pathway, and cell viability was also increased. These effects disappeared in the presence of LY294002. In SD rats, sepsis was induced by CLP. NRG-1β was shown to inhibit autophagy in these rats via the Akt/mTOR pathway, leading to increased body weight of the septic SD rats and alleviation of atrophy of the tibialis anterior muscle. CONCLUSION NRG-1β could alleviate sepsis-induced skeletal muscle atrophy by inhibiting autophagy via the AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Dandan Yin
- Department of Anesthesiology, Affiliated Hospital of Jiangsu University, Zhenjiang City, Jiangsu Province, China
| | - Dawei Lin
- Department of Anesthesiology, Affiliated Hospital of Jiangsu University, Zhenjiang City, Jiangsu Province, China
| | - Yunbin Xie
- Department of Anesthesiology, The First People's Hospital of Changzhou, Changzhou City, Jiangsu Province, China
| | - Aihua Gong
- School of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, China
| | - Peng Jiang
- Department of Anesthesiology, Affiliated Hospital of Jiangsu University, Zhenjiang City, Jiangsu Province, China
| | - Jin Wu
- Department of Anesthesiology, Affiliated Hospital of Jiangsu University, Zhenjiang City, Jiangsu Province, China
| |
Collapse
|
12
|
Lee PHU, Chung M, Ren Z, Mair DB, Kim DH. Factors mediating spaceflight-induced skeletal muscle atrophy. Am J Physiol Cell Physiol 2022; 322:C567-C580. [PMID: 35171699 DOI: 10.1152/ajpcell.00203.2021] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Skeletal muscle atrophy is a well-known consequence of spaceflight. Because of the potential significant impact of muscle atrophy and muscle dysfunction on astronauts and to their mission, a thorough understanding of the mechanisms of this atrophy and the development of effective countermeasures is critical. Spaceflight-induced muscle atrophy is similar to atrophy seen in many terrestrial conditions, and therefore our understanding of this form of atrophy may also contribute to the treatment of atrophy in humans on Earth. The unique environmental features humans encounter in space include the weightlessness of microgravity, space radiation, and the distinctive aspects of living in a spacecraft. The disuse and unloading of muscles in microgravity are likely the most significant factors that mediate spaceflight-induced muscle atrophy, and have been extensively studied and reviewed. However, there are numerous other direct and indirect effects on skeletal muscle that may be contributing factors to the muscle atrophy and dysfunction seen as a result of spaceflight. This review offers a novel perspective on the issue of muscle atrophy in space by providing a comprehensive overview of the unique aspects of the spaceflight environment and the various ways in which they can lead to muscle atrophy. We systematically review the potential contributions of these different mechanisms of spaceflight-induced atrophy and include findings from both actual spaceflight and ground-based models of spaceflight in humans, animals, and in vitro studies.
Collapse
Affiliation(s)
- Peter H U Lee
- Department of Cardiothoracic Surgery, Southcoast Health, Fall River, MA, United States.,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | | | - Zhanping Ren
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Devin B Mair
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
13
|
Ying L, Zhang Q, Yang YM, Zhou JY. A Combination of Serum Biomarkers in Elderly Patients with Sarcopenia: A Cross-Sectional Observational Study. Int J Endocrinol 2022; 2022:4026940. [PMID: 35237317 PMCID: PMC8885259 DOI: 10.1155/2022/4026940] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 01/28/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The pathogenesis of sarcopenia in the elderly has not yet been fully understood. This study aimed to explore the relationship between sarcopenia and several serum biomarkers in elderly population. METHODS It was an observational cross-sectional study of data collected from 70 patients. According to the criteria of the Asian Working Group for Sarcopenia (AWGS), subjects were divided into the sarcopenia group and nonsarcopenic group. We compared age, body mass index (BMI), biochemical indexes, smoking status, underlying disease, muscle mass, handgrip strength (HS), gait speed (GS), skinfold thickness, muscle thickness, and IL-6, IL-10, IL-17A, and TNF-α levels between these groups. RESULTS Of the 70 subjects, 35 patients were diagnosed with sarcopenia. The number of men was higher than that of women in both groups. The patients with sarcopenia were older and had lower BMI and muscle thickness but higher SARC-F questionnaire scores. However, the difference in smoking status and skinfold thickness between these two groups were not statistically significant. Higher IL-6, IL-17A, and TNF-α levels were observed in participants with sarcopenia (P < 0.05). Patients with sarcopenia had a lower IL-10 level. Positive associations were present between the severity of sarcopenia and IL-6, IL-17A, and TNF-α levels, while there was an inverse correlation between the presence of sarcopenia and IL-10 level. CONCLUSIONS Our research found that in sarcopenic elderly subjects, the serum levels of several biomarkers, such as IL-6, IL-17A, and TNF-α, were higher than those in nonsarcopenic elderly persons. Further studies are needed to explore the possible molecular mechanisms and discover new therapeutic targets.
Collapse
Affiliation(s)
- Lin Ying
- Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Qin Zhang
- Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- Key Laboratory of Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yun-mei Yang
- Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- Key Laboratory of Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jian-ying Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
14
|
Fu C, Huang AH, Galatz LM, Han WM. Cellular and molecular modulation of rotator cuff muscle pathophysiology. J Orthop Res 2021; 39:2310-2322. [PMID: 34553789 DOI: 10.1002/jor.25179] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/04/2021] [Accepted: 09/07/2021] [Indexed: 02/04/2023]
Abstract
Rotator cuff (RC) tendon tears are common shoulder injuries that result in irreversible and persistent degeneration of the associated muscles, which is characterized by severe inflammation, atrophy, fibrosis, and fatty infiltration. Although RC muscle degeneration strongly dictates the overall clinical outcomes, strategies to stimulate RC muscle regeneration have largely been overlooked to date. In this review, we highlight the current understanding of the cellular processes that coordinate muscle regeneration, and the roles of muscle resident cells, including immune cells, fibroadipogenic progenitors, and muscle satellite cells in the pathophysiologic regulation of RC muscles following injury. This review also provides perspectives for potential therapies to alleviate the hallmarks of RC muscle degeneration to address current limitations in postsurgical recovery.
Collapse
Affiliation(s)
- Chengcheng Fu
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Alice H Huang
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA.,Department of Orthopedic Surgery, Columbia University, New York City, New York, USA
| | - Leesa M Galatz
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Woojin M Han
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| |
Collapse
|
15
|
Haberecht-Müller S, Krüger E, Fielitz J. Out of Control: The Role of the Ubiquitin Proteasome System in Skeletal Muscle during Inflammation. Biomolecules 2021; 11:biom11091327. [PMID: 34572540 PMCID: PMC8468834 DOI: 10.3390/biom11091327] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 02/07/2023] Open
Abstract
The majority of critically ill intensive care unit (ICU) patients with severe sepsis develop ICU-acquired weakness (ICUAW) characterized by loss of muscle mass, reduction in myofiber size and decreased muscle strength leading to persisting physical impairment. This phenotype results from a dysregulated protein homeostasis with increased protein degradation and decreased protein synthesis, eventually causing a decrease in muscle structural proteins. The ubiquitin proteasome system (UPS) is the predominant protein-degrading system in muscle that is activated during diverse muscle atrophy conditions, e.g., inflammation. The specificity of UPS-mediated protein degradation is assured by E3 ubiquitin ligases, such as atrogin-1 and MuRF1, which target structural and contractile proteins, proteins involved in energy metabolism and transcription factors for UPS-dependent degradation. Although the regulation of activity and function of E3 ubiquitin ligases in inflammation-induced muscle atrophy is well perceived, the contribution of the proteasome to muscle atrophy during inflammation is still elusive. During inflammation, a shift from standard- to immunoproteasome was described; however, to which extent this contributes to muscle wasting and whether this changes targeting of specific muscular proteins is not well described. This review summarizes the function of the main proinflammatory cytokines and acute phase response proteins and their signaling pathways in inflammation-induced muscle atrophy with a focus on UPS-mediated protein degradation in muscle during sepsis. The regulation and target-specificity of the main E3 ubiquitin ligases in muscle atrophy and their mode of action on myofibrillar proteins will be reported. The function of the standard- and immunoproteasome in inflammation-induced muscle atrophy will be described and the effects of proteasome-inhibitors as treatment strategies will be discussed.
Collapse
Affiliation(s)
- Stefanie Haberecht-Müller
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Elke Krüger
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, 17475 Greifswald, Germany;
- Correspondence: (E.K.); (J.F.)
| | - Jens Fielitz
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, 17475 Greifswald, Germany
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, 17475 Greifswald, Germany
- Correspondence: (E.K.); (J.F.)
| |
Collapse
|
16
|
Redox Signaling and Sarcopenia: Searching for the Primary Suspect. Int J Mol Sci 2021; 22:ijms22169045. [PMID: 34445751 PMCID: PMC8396474 DOI: 10.3390/ijms22169045] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 12/16/2022] Open
Abstract
Sarcopenia, the age-related decline in muscle mass and function, derives from multiple etiological mechanisms. Accumulative research suggests that reactive oxygen species (ROS) generation plays a critical role in the development of this pathophysiological disorder. In this communication, we review the various signaling pathways that control muscle metabolic and functional integrity such as protein turnover, cell death and regeneration, inflammation, organismic damage, and metabolic functions. Although no single pathway can be identified as the most crucial factor that causes sarcopenia, age-associated dysregulation of redox signaling appears to underlie many deteriorations at physiological, subcellular, and molecular levels. Furthermore, discord of mitochondrial homeostasis with aging affects most observed problems and requires our attention. The search for the primary suspect of the fundamental mechanism for sarcopenia will likely take more intense research for the secret of this health hazard to the elderly to be unlocked.
Collapse
|
17
|
Role of NF-κB in Ageing and Age-Related Diseases: Lessons from Genetically Modified Mouse Models. Cells 2021; 10:cells10081906. [PMID: 34440675 PMCID: PMC8394846 DOI: 10.3390/cells10081906] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/17/2021] [Accepted: 07/23/2021] [Indexed: 12/21/2022] Open
Abstract
Ageing is a complex process, induced by multifaceted interaction of genetic, epigenetic, and environmental factors. It is manifested by a decline in the physiological functions of organisms and associated to the development of age-related chronic diseases and cancer development. It is considered that ageing follows a strictly-regulated program, in which some signaling pathways critically contribute to the establishment and maintenance of the aged state. Chronic inflammation is a major mechanism that promotes the biological ageing process and comorbidity, with the transcription factor NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) as a crucial mediator of inflammatory responses. This, together with the finding that the activation or inhibition of NF-κB can induce or reverse respectively the main features of aged organisms, has brought it under consideration as a key transcription factor that acts as a driver of ageing. In this review, we focused on the data obtained entirely through the generation of knockout and transgenic mouse models of either protein involved in the NF-κB signaling pathway that have provided relevant information about the intricate processes or molecular mechanisms that control ageing. We have reviewed the relationship of NF-κB and premature ageing; the development of cancer associated with ageing and the implication of NF-κB activation in the development of age-related diseases, some of which greatly increase the risk of developing cancer.
Collapse
|
18
|
Zhou Y, Cao F, Wu Q, Luo Y, Guo T, Han S, Huang M, Hu Z, Bai J, Luo F, Lin Q. Dietary Supplementation of Octacosanol Improves Exercise-Induced Fatigue and Its Molecular Mechanism. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:7603-7618. [PMID: 34223764 DOI: 10.1021/acs.jafc.1c01764] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Several publications report that octacosanol (OCT) has different biological functions. This study was designed to evaluate the antifatigue effect and molecular mechanism of octacosanol (200 mg/(kg day)) in forced exercise-induced fatigue models of trained male C57BL/6 mice. Results showed that octacosanol ameliorated the mice's autonomic activities, forelimb grip strength, and swimming endurance, and the levels of liver glycogen (LG), muscle glycogen (MG), blood lactic acid (BLA), lactate dehydrogenase (LDH), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) were also regulated. Gene analysis results showed that treatment with OCT upregulated 29 genes, while 38 genes were downregulated in gastrocnemius tissue. Gene ontology (GO) analyses indicated that these genes enriched functions in relation to myofibril, contractile fiber, and calcium-dependent adenosinetriphosphatase (ATPase) activity. Octacosanol supplementation significantly adjusted the messenger RNA (mRNA) and protein expression levels related to fatigue performance. Octacosanol has an observably mitigating effect in exercise-induced fatigue models, and its molecular mechanism may be related to the regulation of tripartite motif-containing 63 (Trim63), periaxin (Prx), calcium voltage-gated channel subunit α1 H (Cacna1h), and myosin-binding protein C (Mybpc3) expression.
Collapse
Affiliation(s)
- Yaping Zhou
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, Central South University of Forestry and Technology, No. 498, Shaoshan Road, Changsha 410004, Hunan, China
| | - Fuliang Cao
- Co-Innovation Center for the Sustainable Forestry in Southern China, College of Forestry, Nanjing Forestry University, Nanjing 210037, Jiangsu, China
| | - Qiang Wu
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, Central South University of Forestry and Technology, No. 498, Shaoshan Road, Changsha 410004, Hunan, China
| | - Yi Luo
- Department of Clinical Medicine, Medical College of Xiangya, Central South University, Changsha 410008, Hunan, China
| | - Tianyi Guo
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, Central South University of Forestry and Technology, No. 498, Shaoshan Road, Changsha 410004, Hunan, China
| | - Shuai Han
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, Central South University of Forestry and Technology, No. 498, Shaoshan Road, Changsha 410004, Hunan, China
| | - Mengzhen Huang
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, Central South University of Forestry and Technology, No. 498, Shaoshan Road, Changsha 410004, Hunan, China
| | - Zuomin Hu
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, Central South University of Forestry and Technology, No. 498, Shaoshan Road, Changsha 410004, Hunan, China
| | - Jie Bai
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, Central South University of Forestry and Technology, No. 498, Shaoshan Road, Changsha 410004, Hunan, China
| | - Feijun Luo
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, Central South University of Forestry and Technology, No. 498, Shaoshan Road, Changsha 410004, Hunan, China
| | - Qinlu Lin
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, Central South University of Forestry and Technology, No. 498, Shaoshan Road, Changsha 410004, Hunan, China
| |
Collapse
|
19
|
Kang YJ, Yoo JI, Baek KW. Differential gene expression profile by RNA sequencing study of elderly osteoporotic hip fracture patients with sarcopenia. J Orthop Translat 2021; 29:10-18. [PMID: 34036042 PMCID: PMC8138673 DOI: 10.1016/j.jot.2021.04.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/10/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
Background The purpose of this study was to report the RNA sequencing profile according to the presence or absence of sarcopenia in elderly patients with osteoporotic hip fracture. Therefore, an important genetic factor candidate for sarcopenia causing hip fracture in elderly with osteoporosis has been identified. Methods The patient group involved subjects over 65 years who had undergone hip fracture surgery. Among 323 hip fracture (HF) patients identified from May 2017 to December 2019, 162 HF patients (90 non-sarcopenia and 72 sarcopenia groups), excluding subjects with high energy trauma and non-osteoporosis, were finally included in the analysis. For RNA sequencing, each patient with hand grip strength (HGS) values in the top 10% were enrolled in the control group and with the bottom 10% in the patient group. After excluding patients with poor tissue quality, 6 patients and 5 patients were selected for sarcopenia and non-sarcopenia groups, respectively. For qPCR validation, each patient with HGS values in the top 20% and bottom 20% was enrolled in the control and patient groups, respectively. After excluding patients with poor tissue quality, 12 patients and 12 patients were enrolled in the sarcopenia and non-sarcopenia groups, respectively. Sarcopenia was defined according to the Asia Working Group for Sarcopenia (AWGS) criteria for low muscle strength (hand grip strength below 18 kg in women and 28 kg in men) and low muscle mass (SMI below 5.4 kg/m2 in women and 7.0 kg/m2 in men). The libraries were prepared for 100 bp paired-end sequencing using TruSeq Stranded mRNA Sample Preparation Kit (Illumina, CA, USA). The gene expression counts were supplied to Deseq2 to extract possible gene sets as differentially expressed genes (DEG) that discriminate between sarcopenia and non-sarcopenia groups that were carefully assigned by clinical observation. For the classification of the candidate genes from DEG analysis, we used the public databases; gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Quantitative real-time PCR was performed for validation. Results Samples collected were subjected to RNAseq using the Illumina platform. A total of 11 samples from both sarcopenia and non-sarcopenia groups were sequenced. Fifteen genes (RUNX 1, NGFR, CH3L1, BCL3, PLA2G2A, MYBPH, TEP1, SEMA6B, CSPG4, ACSL5, SLC25A3, NDUFB5, CYC1, ACAT1, and TCAP) were identified as differentially expressed genes (DEG) in both the groups. In the qPCR results, the expression levels of SLC25A3 and TCAP gene in the OS group were significantly lower than in the non-OS groups whereas an increase in RUNX1 mRNA level was observed in the OS samples (p < 0.05). Conclusions In summary, this study detected gene expression difference according to the presence or absence of sarcopenia in elderly osteoporosis female patients with hip fracture. We have also identified 15 important genes (RUNX 1, NGFR, CH3L1, BCL3, PLA2G2A, MYBPH, TEP1, SEMA6B, CSPG4, ACSL5, SLC25A3, NDUFB5, CYC1, ACAT1, TCAP), a few GO categories and biological pathways that may be associated with the osteosarcopenia. Our study may provide effective means for the prevention, diagnosis and treatment sarcopenia in elderly osteoporosis female patients. The Translational potential of this article These findings provide a novel insight into the effects of aging on the response in women with postmenopausal osteoporosis. Further studies are underway to identify the specific signalling pathways involved. These results reveal potential therapeutic targets that could aid the regenerative capacity of aging skeletal muscle.
Collapse
Affiliation(s)
- Yang-Jae Kang
- Division of Applied Life Science Department at Gyeongsang National University, PMBBRC, Jinju, Republic of Korea
- Division of Life Science Department at Gyeongsang National University, Jinju, Republic of Korea
| | - Jun-Il Yoo
- Department of Orthopaedic Surgery, Gyeongsang National University Hospital, Jinju, South Korea
- Corresponding author. Department of Orthopaedic Surgery, Gyeongsang National University Hospital, 90 Chilamdong, Jinju, Gyeongnamdo, 660-702, Republic of Korea.
| | - Kyung-Wan Baek
- Department of Orthopaedic Surgery, Gyeongsang National University Hospital, Jinju, South Korea
| |
Collapse
|
20
|
Peris-Moreno D, Cussonneau L, Combaret L, Polge C, Taillandier D. Ubiquitin Ligases at the Heart of Skeletal Muscle Atrophy Control. Molecules 2021; 26:molecules26020407. [PMID: 33466753 PMCID: PMC7829870 DOI: 10.3390/molecules26020407] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/08/2021] [Accepted: 01/10/2021] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle loss is a detrimental side-effect of numerous chronic diseases that dramatically increases mortality and morbidity. The alteration of protein homeostasis is generally due to increased protein breakdown while, protein synthesis may also be down-regulated. The ubiquitin proteasome system (UPS) is a master regulator of skeletal muscle that impacts muscle contractile properties and metabolism through multiple levers like signaling pathways, contractile apparatus degradation, etc. Among the different actors of the UPS, the E3 ubiquitin ligases specifically target key proteins for either degradation or activity modulation, thus controlling both pro-anabolic or pro-catabolic factors. The atrogenes MuRF1/TRIM63 and MAFbx/Atrogin-1 encode for key E3 ligases that target contractile proteins and key actors of protein synthesis respectively. However, several other E3 ligases are involved upstream in the atrophy program, from signal transduction control to modulation of energy balance. Controlling E3 ligases activity is thus a tempting approach for preserving muscle mass. While indirect modulation of E3 ligases may prove beneficial in some situations of muscle atrophy, some drugs directly inhibiting their activity have started to appear. This review summarizes the main signaling pathways involved in muscle atrophy and the E3 ligases implicated, but also the molecules potentially usable for future therapies.
Collapse
|
21
|
Gorza L, Sorge M, Seclì L, Brancaccio M. Master Regulators of Muscle Atrophy: Role of Costamere Components. Cells 2021; 10:cells10010061. [PMID: 33401549 PMCID: PMC7823551 DOI: 10.3390/cells10010061] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 12/11/2022] Open
Abstract
The loss of muscle mass and force characterizes muscle atrophy in several different conditions, which share the expression of atrogenes and the activation of their transcriptional regulators. However, attempts to antagonize muscle atrophy development in different experimental contexts by targeting contributors to the atrogene pathway showed partial effects in most cases. Other master regulators might independently contribute to muscle atrophy, as suggested by our recent evidence about the co-requirement of the muscle-specific chaperone protein melusin to inhibit unloading muscle atrophy development. Furthermore, melusin and other muscle mass regulators, such as nNOS, belong to costameres, the macromolecular complexes that connect sarcolemma to myofibrils and to the extracellular matrix, in correspondence with specific sarcomeric sites. Costameres sense a mechanical load and transduce it both as lateral force and biochemical signals. Recent evidence further broadens this classic view, by revealing the crucial participation of costameres in a sarcolemmal “signaling hub” integrating mechanical and humoral stimuli, where mechanical signals are coupled with insulin and/or insulin-like growth factor stimulation to regulate muscle mass. Therefore, this review aims to enucleate available evidence concerning the early involvement of costamere components and additional putative master regulators in the development of major types of muscle atrophy.
Collapse
Affiliation(s)
- Luisa Gorza
- Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy
- Correspondence:
| | - Matteo Sorge
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (M.S.); (L.S.); (M.B.)
| | - Laura Seclì
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (M.S.); (L.S.); (M.B.)
| | - Mara Brancaccio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (M.S.); (L.S.); (M.B.)
| |
Collapse
|
22
|
Sharanya A, Ciano M, Withana S, Kemp PR, Polkey MI, Sathyapala SA. Sex differences in COPD-related quadriceps muscle dysfunction and fibre abnormalities. Chron Respir Dis 2020; 16:1479973119843650. [PMID: 31131626 PMCID: PMC6537500 DOI: 10.1177/1479973119843650] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In chronic obstructive pulmonary disease (COPD), lower limb dysfunction is associated with reduced exercise capacity, increased hospitalizations and mortality. We investigated sex differences in the prevalence of quadriceps dysfunction and fibre abnormalities in a large COPD cohort, controlling for the normal sex differences in health. We compared existing data from 76 male and 38 female COPD patients where each variable was expressed as a function of gender-specific normal values (obtained from 16 male and 14 female controls). Female COPD patients had lower quadriceps muscle strength and peak workload on a maximal incremental cycle ergometry protocol compared to male patients. Female patients had a smaller type II fibre cross-sectional area (CSA) compared to male patients, suggesting a greater female preponderance to fibre atrophy, although this result was largely driven by a few male patients with a large type II fibre CSA. Female patients had significantly higher concentrations of a number of plasma pro-inflammatory cytokines including tumour necrosis factor alpha and interleukin 8 (IL8), but not lower levels of physical activity or arterial oxygenation, compared to males. Our data confirm results from a previous small study and suggest that female COPD patients have a greater prevalence of muscle wasting and weakness. Larger studies investigating sex differences in COPD-related muscle atrophy and weakness are needed, as the results will have implications for monitoring in clinical practice and for design of clinical trials evaluating novel muscle anabolic agents.
Collapse
Affiliation(s)
- Adithya Sharanya
- 1 Molecular Medicine, National Heart and Lung Institute, SAF Building, South Kensington Campus, Imperial College London, London, UK
| | - Margherita Ciano
- 1 Molecular Medicine, National Heart and Lung Institute, SAF Building, South Kensington Campus, Imperial College London, London, UK
| | - Shirmila Withana
- 2 Respiratory Medicine, Royal Brompton and Harefield NHS Foundation Trust, Harefield Hospital, Hill End Road, Harefield, Middlesex, UK
| | - Paul Richard Kemp
- 1 Molecular Medicine, National Heart and Lung Institute, SAF Building, South Kensington Campus, Imperial College London, London, UK
| | - Michael Iain Polkey
- 3 Respiratory Medicine, Royal Brompton and Harefield NHS Foundation Trust, Royal Brompton Hospital, First Floor, Fulham Road, London, UK
| | - Samantha Amanda Sathyapala
- 1 Molecular Medicine, National Heart and Lung Institute, SAF Building, South Kensington Campus, Imperial College London, London, UK
| |
Collapse
|
23
|
Ehmsen JT, Höke A. Cellular and molecular features of neurogenic skeletal muscle atrophy. Exp Neurol 2020; 331:113379. [PMID: 32533969 DOI: 10.1016/j.expneurol.2020.113379] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 05/26/2020] [Accepted: 06/08/2020] [Indexed: 12/28/2022]
Abstract
Neurogenic atrophy refers to the loss of muscle mass and function that results directly from injury or disease of the peripheral nervous system. Individuals with neurogenic atrophy may experience reduced functional status and quality of life and, in some circumstances, reduced survival. Distinct pathological findings on muscle histology can aid in diagnosis of a neurogenic cause for muscle dysfunction, and provide indicators for the chronicity of denervation. Denervation induces pleiotypic responses in skeletal muscle, and the molecular mechanisms underlying neurogenic muscle atrophy appear to share common features with other causes of muscle atrophy, including activation of FOXO transcription factors and corresponding induction of ubiquitin-proteasomal and lysosomal degradation. In this review, we provide an overview of histologic features of neurogenic atrophy and a summary of current understanding of underlying mechanisms.
Collapse
Affiliation(s)
- Jeffrey T Ehmsen
- Department of Neurology, Neuromuscular Division, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ahmet Höke
- Department of Neurology, Neuromuscular Division, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
24
|
Abstract
Significance: Regular contractile activity plays a critical role in maintaining skeletal muscle morphological integrity and physiological function. If the muscle is forced to stop contraction, such as during limb immobilization (IM), the IGF/Akt/mTOR signaling pathway that normally stimulates protein synthesis and inhibits proteolysis will be suppressed, whereas the FoxO-controlled catabolic pathways such as ubiquitin-proteolysis and autophagy/mitophagy will be activated and dominate, resulting in muscle fiber atrophy. Recent Advances: Mitochondria occupy a central position in the regulation of both protein synthesis and degradation through several redox-sensitive pathways, including peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), mitochondrial fusion and fission proteins, mitophagy, and sirtuins. Prolonged IM downregulates PGC-1α due to AMPK (5'-AMP-activated protein kinase) and FoxO activation, thus decreasing mitochondrial biogenesis and causing oxidative damage. Decrease of mitochondrial inner membrane potential and increase of mitochondrial fission can trigger cascades of mitophagy leading to loss of mitochondrial homeostasis (mitostasis), inflammation, and apoptosis. The phenotypic outcomes of these disorders are compromised muscle function and fiber atrophy. Critical Issues: Given the molecular mechanism of the pathogenesis, it is imperative that the integrity of intracellular signaling be restored to prevent the deterioration. So far, overexpression of PGC-1α via transgene and in vivo DNA transfection has been found to be effective in ameliorating mitostasis and reduces IM-induced muscle atrophy. Nutritional supplementation of select amino acids and phytochemicals also provides mechanistic and practical insights into the prevention of muscle disuse atrophy. Future Directions: In light of the importance of mitochondria in regulating the various critical signaling pathways, future work should focus on exploring new epigenetic strategies to restore mitostasis and redox balance.
Collapse
Affiliation(s)
- Li Li Ji
- The Laboratory of Physiological Hygiene and Exercise Science, School of Kinesiology, University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - Dongwook Yeo
- The Laboratory of Physiological Hygiene and Exercise Science, School of Kinesiology, University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Chounghun Kang
- Departmet Physical Education, Inha University, Incheon, South Korea
| |
Collapse
|
25
|
Sharma B, Dabur R. Role of Pro-inflammatory Cytokines in Regulation of Skeletal Muscle Metabolism: A Systematic Review. Curr Med Chem 2020; 27:2161-2188. [DOI: 10.2174/0929867326666181129095309] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 11/15/2018] [Accepted: 11/19/2018] [Indexed: 12/18/2022]
Abstract
Background:
Metabolic pathways perturbations lead to skeletal muscular atrophy in the
cachexia and sarcopenia due to increased catabolism. Pro-inflammatory cytokines induce the catabolic
pathways that impair the muscle integrity and function. Hence, this review primarily concentrates
on the effects of pro-inflammatory cytokines in regulation of skeletal muscle metabolism.
Objective:
This review will discuss the role of pro-inflammatory cytokines in skeletal muscles during
muscle wasting conditions. Moreover, the coordination among the pro-inflammatory cytokines
and their regulated molecular signaling pathways which increase the protein degradation will be
discussed.
Results:
During normal conditions, pro-inflammatory cytokines are required to balance anabolism
and catabolism and to maintain normal myogenesis process. However, during muscle wasting their
enhanced expression leads to marked destructive metabolism in the skeletal muscles. Proinflammatory
cytokines primarily exert their effects by increasing the expression of calpains and E3
ligases as well as of Nf-κB, required for protein breakdown and local inflammation. Proinflammatory
cytokines also locally suppress the IGF-1and insulin functions, hence increase the
FoxO activation and decrease the Akt function, the central point of carbohydrates lipid and protein
metabolism.
Conclusion:
Current advancements have revealed that the muscle mass loss during skeletal muscular
atrophy is multifactorial. Despite great efforts, not even a single FDA approved drug is available
in the market. It indicates the well-organized coordination among the pro-inflammatory cytokines
that need to be further understood and explored.
Collapse
Affiliation(s)
- Bhawana Sharma
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana-124001, India
| | - Rajesh Dabur
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana-124001, India
| |
Collapse
|
26
|
Hahn A, Kny M, Pablo-Tortola C, Todiras M, Willenbrock M, Schmidt S, Schmoeckel K, Jorde I, Nowak M, Jarosch E, Sommer T, Bröker BM, Felix SB, Scheidereit C, Weber-Carstens S, Butter C, Luft FC, Fielitz J. Serum amyloid A1 mediates myotube atrophy via Toll-like receptors. J Cachexia Sarcopenia Muscle 2020; 11:103-119. [PMID: 31441598 PMCID: PMC7015249 DOI: 10.1002/jcsm.12491] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 06/28/2019] [Accepted: 07/22/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Critically ill patients frequently develop muscle atrophy and weakness in the intensive-care-unit setting [intensive care unit-acquired weakness (ICUAW)]. Sepsis, systemic inflammation, and acute-phase response are major risk factors. We reported earlier that the acute-phase protein serum amyloid A1 (SAA1) is increased and accumulates in muscle of ICUAW patients, but its relevance was unknown. Our objectives were to identify SAA1 receptors and their downstream signalling pathways in myocytes and skeletal muscle and to investigate the role of SAA1 in inflammation-induced muscle atrophy. METHODS We performed cell-based in vitro and animal in vivo experiments. The atrophic effect of SAA1 on differentiated C2C12 myotubes was investigated by analysing gene expression, protein content, and the atrophy phenotype. We used the cecal ligation and puncture model to induce polymicrobial sepsis in wild type mice, which were treated with the IкB kinase inhibitor Bristol-Myers Squibb (BMS)-345541 or vehicle. Morphological and molecular analyses were used to investigate the phenotype of inflammation-induced muscle atrophy and the effects of BMS-345541 treatment. RESULTS The SAA1 receptors Tlr2, Tlr4, Cd36, P2rx7, Vimp, and Scarb1 were all expressed in myocytes and skeletal muscle. Treatment of differentiated C2C12 myotubes with recombinant SAA1 caused myotube atrophy and increased interleukin 6 (Il6) gene expression. These effects were mediated by Toll-like receptors (TLR) 2 and 4. SAA1 increased the phosphorylation and activity of the transcription factor nuclear factor 'kappa-light-chain-enhancer' of activated B-cells (NF-κB) p65 via TLR2 and TLR4 leading to an increased binding of NF-κB to NF-κB response elements in the promoter region of its target genes resulting in an increased expression of NF-κB target genes. In polymicrobial sepsis, skeletal muscle mass, tissue morphology, gene expression, and protein content were associated with the atrophy response. Inhibition of NF-κB signalling by BMS-345541 increased survival (28.6% vs. 91.7%, P < 0.01). BMS-345541 diminished inflammation-induced atrophy as shown by a reduced weight loss of the gastrocnemius/plantaris (vehicle: -21.2% and BMS-345541: -10.4%; P < 0.05), tibialis anterior (vehicle: -22.7% and BMS-345541: -17.1%; P < 0.05) and soleus (vehicle: -21.1% and BMS-345541: -11.3%; P < 0.05) in septic mice. Analysis of the fiber type specific myocyte cross-sectional area showed that BMS-345541 reduced inflammation-induced atrophy of slow/type I and fast/type II myofibers compared with vehicle-treated septic mice. BMS-345541 reversed the inflammation-induced atrophy program as indicated by a reduced expression of the atrogenes Trim63/MuRF1, Fbxo32/Atrogin1, and Fbxo30/MuSA1. CONCLUSIONS SAA1 activates the TLR2/TLR4//NF-κB p65 signalling pathway to cause myocyte atrophy. Systemic inhibition of the NF-κB pathway reduced muscle atrophy and increased survival of septic mice. The SAA1/TLR2/TLR4//NF-κB p65 atrophy pathway could have utility in combatting ICUAW.
Collapse
Affiliation(s)
- Alexander Hahn
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Melanie Kny
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Cristina Pablo-Tortola
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Mihail Todiras
- Cardiovascular hormones, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Nicolae Testemiţanu State University of Medicine and Pharmacy, Chișinău, Moldova
| | - Michael Willenbrock
- Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Sibylle Schmidt
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Katrin Schmoeckel
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine, Greifswald, Germany
| | - Ilka Jorde
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine, Greifswald, Germany
| | - Marcel Nowak
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Intracellular Proteolysis, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Ernst Jarosch
- Intracellular Proteolysis, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Thomas Sommer
- Intracellular Proteolysis, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Institute of Biology, Humboldt-University Berlin, Berlin, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Barbara M Bröker
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine, Greifswald, Germany
| | - Stephan B Felix
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Claus Scheidereit
- Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Steffen Weber-Carstens
- Department of Anesthesiology and Intensive Care Medicine, Campus Virchow-Klinikum and Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Christian Butter
- Department of Cardiology, Heart Center Brandenburg and Medical University Brandenburg (MHB), Bernau, Germany
| | - Friedrich C Luft
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Jens Fielitz
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
27
|
Mechanism and Functions of Identified miRNAs in Poultry Skeletal Muscle Development – A Review. ANNALS OF ANIMAL SCIENCE 2019. [DOI: 10.2478/aoas-2019-0049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Abstract
Development of the skeletal muscle goes through several complex processes regulated by numerous genetic factors. Although much efforts have been made to understand the mechanisms involved in increased muscle yield, little work is done about the miRNAs and candidate genes that are involved in the skeletal muscle development in poultry. Comprehensive research of candidate genes and single nucleotide related to poultry muscle growth is yet to be experimentally unraveled. However, over a few periods, studies in miRNA have disclosed that they actively participate in muscle formation, differentiation, and determination in poultry. Specifically, miR-1, miR-133, and miR-206 influence tissue development, and they are highly expressed in the skeletal muscles. Candidate genes such as CEBPB, MUSTN1, MSTN, IGF1, FOXO3, mTOR, and NFKB1, have also been identified to express in the poultry skeletal muscles development. However, further researches, analysis, and comprehensive studies should be made on the various miRNAs and gene regulatory factors that influence the skeletal muscle development in poultry. The objective of this review is to summarize recent knowledge in miRNAs and their mode of action as well as transcription and candidate genes identified to regulate poultry skeletal muscle development.
Collapse
|
28
|
Song X, Zhu M, Li H, Liu B, Yan Z, Wang W, Li H, Sun J, Li S. USF1 promotes the development of knee osteoarthritis by activating the NF-κB signaling pathway. Exp Ther Med 2018; 16:3518-3524. [PMID: 30233704 PMCID: PMC6143875 DOI: 10.3892/etm.2018.6608] [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: 01/10/2018] [Accepted: 07/19/2018] [Indexed: 01/16/2023] Open
Abstract
The current study mainly aims to evaluate the expression pattern and underlying mechanism of upstream stimulating factor 1 (USF1) in the muscle tissues of knee osteoarthritis (KOA) patients. In accordance with previous findings, our data showed that muscle strength was significantly decreased in KOA patients compared with controls. Furthermore, several inflammatory factors, including tumor necrosis factor α (TNFα), IL-8, IL-6 and MCP-1, were associated with reduced muscle strength in KOA patients. Not surprisingly, NF-κB signaling was significantly activated in the muscle tissues of KOA patients compared with control individuals. Furthermore, we showed that USF1 was increased in the muscles of KOA patients compared with controls. More importantly, overexpression of USF1 in primary human skeletal muscle cells significantly increased the activation of NF-κB signaling as well as the levels of pro-inflammatory factors. In summary, we showed novel data that the upregulation of USF1 promoted NF-κB activation-induced inflammatory responses in muscle tissues of KOA patients.
Collapse
Affiliation(s)
- Xiandong Song
- Department of Orthopedics, Hongqi Hospital Affiliated with Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Min Zhu
- Department of Radiology, Hongqi Hospital Affiliated with Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Hao Li
- Department of Radiology, Hongqi Hospital Affiliated with Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Bo Liu
- Department of Orthopedics, Hongqi Hospital Affiliated with Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Zhaowei Yan
- Department of Orthopedics, Hongqi Hospital Affiliated with Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Weican Wang
- Department of Orthopedics, Hongqi Hospital Affiliated with Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Hongyi Li
- Department of Radiology, Hongqi Hospital Affiliated with Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Jiping Sun
- Department of Radiology, Hongqi Hospital Affiliated with Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Shixing Li
- Department of Radiology, Hongqi Hospital Affiliated with Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| |
Collapse
|
29
|
Wei Y, Gong L, Fu W, Xu S, Wang Z, Zhang J, Ning E, Chang H, Wang H, Gao Y. Unexpected regulation pattern of the IKKβ/NF‐κB/MuRF1 pathway with remarkable muscle plasticity in the Daurian ground squirrel (
Spermophilus dauricus
). J Cell Physiol 2018; 233:8711-8722. [DOI: 10.1002/jcp.26751] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 04/16/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Yanhong Wei
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaCollege of Life SciencesNorthwest University, Ministry of EducationXi'anChina
- School of Basic Medical SciencesNingxia Medical UniversityYinchuanChina
| | - Lingchen Gong
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaCollege of Life SciencesNorthwest University, Ministry of EducationXi'anChina
| | - Weiwei Fu
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaCollege of Life SciencesNorthwest University, Ministry of EducationXi'anChina
| | - Shenhui Xu
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaCollege of Life SciencesNorthwest University, Ministry of EducationXi'anChina
| | - Zhe Wang
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaCollege of Life SciencesNorthwest University, Ministry of EducationXi'anChina
| | - Jie Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaCollege of Life SciencesNorthwest University, Ministry of EducationXi'anChina
| | - Er Ning
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaCollege of Life SciencesNorthwest University, Ministry of EducationXi'anChina
| | - Hui Chang
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaCollege of Life SciencesNorthwest University, Ministry of EducationXi'anChina
| | - Huiping Wang
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaCollege of Life SciencesNorthwest University, Ministry of EducationXi'anChina
| | - Yunfang Gao
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaCollege of Life SciencesNorthwest University, Ministry of EducationXi'anChina
| |
Collapse
|
30
|
Belova SP, Shenkman BS, Kostrominova TY, Nemirovskaya TL. Paradoxical effect of IKKβ inhibition on the expression of E3 ubiquitin ligases and unloading-induced skeletal muscle atrophy. Physiol Rep 2018; 5:5/16/e13291. [PMID: 28839114 PMCID: PMC5582258 DOI: 10.14814/phy2.13291] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 04/19/2017] [Accepted: 04/26/2017] [Indexed: 12/13/2022] Open
Abstract
We tested whether NF‐κB pathway is indispensable for the increase in expression of E3‐ligases and unloading‐induced muscle atrophy using IKKβ inhibitor IMD‐0354. Three groups of rats were used: nontreated control (C), 3 days of unloading/hindlimb suspension with (HS+IMD) or without (HS) IMD‐0354. Levels of IκBα were higher in HS+IMD (1.16‐fold) and lower in HS (0.82‐fold) when compared with C group. IMD‐0354 treatment during unloading: had no effect on loss of muscle mass; increased mRNA levels of MuRF1 and MAFbx; increased levels of pFoxO3; and had no effect on levels of Bcl‐3, p105, and p50 proteins. Our study for the first time showed that inhibiting IKKβ in vivo during 3‐day unloading failed to diminish expression of ubiquitin ligases and prevent muscle atrophy.
Collapse
Affiliation(s)
| | | | - Tatiana Y Kostrominova
- Department of Anatomy and Cell Biology, Indiana University School of Medicine-Northwest, Gary, Indiana
| | - Tatiana L Nemirovskaya
- Institute of Biomedical Problems, RAS, Moscow, Russia .,Faculty of Basic Medicine, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
31
|
S-allyl cysteine inhibits TNFα-induced skeletal muscle wasting through suppressing proteolysis and expression of inflammatory molecules. Biochim Biophys Acta Gen Subj 2018; 1862:895-906. [DOI: 10.1016/j.bbagen.2017.12.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 12/15/2017] [Accepted: 12/26/2017] [Indexed: 12/20/2022]
|
32
|
Alaca N, Uslu S, Gulec Suyen G, Ince U, Serteser M, Kurtel H. Effects of different aerobic exercise frequencies on streptozotocin-nicotinamide-induced type 2 diabetic rats: Continuous versus short bouts and weekend warrior exercises. J Diabetes 2018; 10:73-84. [PMID: 28425181 DOI: 10.1111/1753-0407.12561] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/24/2017] [Accepted: 04/14/2017] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Exercise training is known to have multiple beneficial effects on type 2 diabetes mellitus (T2DM). The aim of this study was to explore the effects of aerobic exercise frequency on diabetic parameters, the histopathological structure of skeletal muscle, diabetic myopathy, and mitochondrial enzyme activity in an experimental model of T2DM. METHODS Sprague-Dawley rats (n = 35) were rendered diabetic by injection of nicotinamide (110 mg/kg) and streptozotocin (65 mg/kg). Rats with blood glucose concentrations between 7 and 17 mmol/L were used. Diabetic rats were randomly allocated to one of the following groups: (i) control sedentary; (ii) diabetic sedentary; (iii) diabetic with continuous exercise (30 min/day, 5 days/week); (iv) diabetic with short bouts of exercise (3 × 10 min/day, 5 days/week); and (v) diabetic rats as "weekend warriors" (35 + 40 min/day, 2 days/week). After 6 weeks swimming exercise (total duration 150 min/week), biochemical tests were performed to measure insulin, glucose, cytokines, serum and muscle myeloperoxidase (MPO), and malondialdehyde (MDA) levels. Histologic analysis (histomorphometric and mitochondrial enzyme analysis) was also performed. RESULTS Compared with diabetic sedentary rats, significant improvements were observed in all exercise groups in terms of glucose levels, weight loss, tissue MPO and MDA levels, muscular connective tissue, muscle atrophy, mitochondrial enzyme, and all histomorphometric analyses. CONCLUSIONS The results of the study emphasize the effects of training on inflammation, increased oxidative stress, myopathy, and mitochondrial damage in a rat model of T2DM, and demonstrate that there is no major difference between exercise modalities provided that the total duration of exercise remains the same.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/toxicity
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/therapy
- Diabetes Mellitus, Type 2/chemically induced
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/therapy
- Glucose/metabolism
- Male
- Muscle, Skeletal/physiology
- Niacinamide/toxicity
- Physical Conditioning, Animal
- Rats
- Rats, Sprague-Dawley
- Streptozocin/toxicity
- Swimming
- Vitamin B Complex/toxicity
Collapse
Affiliation(s)
- Nuray Alaca
- Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Acibadem University, Istanbul, Turkey
- Department of Physiology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Serap Uslu
- Department of Histology and Embryology, School of Medicine, Medeniyet University, Istanbul, Turkey
| | - Guldal Gulec Suyen
- Department of Physiology, School of Medicine, Acibadem University, Istanbul, Turkey
| | - Umit Ince
- Department of Pathology, School of Medicine, Acibadem University, Istanbul, Turkey
| | - Mustafa Serteser
- Department of Biochemistry, School of Medicine, Acibadem University, Istanbul, Turkey
| | - Hızır Kurtel
- Department of Physiology, School of Medicine, Marmara University, Istanbul, Turkey
| |
Collapse
|
33
|
Zhang N, Valentine JM, Zhou Y, Li ME, Zhang Y, Bhattacharya A, Walsh ME, Fischer KE, Austad SN, Osmulski P, Gaczynska M, Shoelson SE, Van Remmen H, Chen HI, Chen Y, Liang H, Musi N. Sustained NFκB inhibition improves insulin sensitivity but is detrimental to muscle health. Aging Cell 2017; 16:847-858. [PMID: 28556540 PMCID: PMC5506420 DOI: 10.1111/acel.12613] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2017] [Indexed: 01/06/2023] Open
Abstract
Older adults universally suffer from sarcopenia and approximately 60-70% are diabetic or prediabetic. Nonetheless, the mechanisms underlying these aging-related metabolic disorders are unknown. NFκB has been implicated in the pathogenesis of several aging-related pathologies including sarcopenia and type 2 diabetes and has been proposed as a target against them. NFκB also is thought to mediate muscle wasting seen with disuse, denervation, and some systemic diseases (e.g., cancer, sepsis). We tested the hypothesis that lifelong inhibition of the classical NFκB pathway would protect against aging-related sarcopenia and insulin resistance. Aged mice with muscle-specific overexpression of a super-repressor IκBα mutant (MISR) were protected from insulin resistance. However, MISR mice were not protected from sarcopenia; to the contrary, these mice had decreases in muscle mass and strength compared to wild-type mice. In MISR mice, NFκB suppression also led to an increase in proteasome activity and alterations in several genes and pathways involved in muscle growth and atrophy (e.g., myostatin). We conclude that the mechanism behind aging-induced sarcopenia is NFκB independent and differs from muscle wasting due to pathologic conditions. Our findings also indicate that, while suppressing NFκB improves insulin sensitivity in aged mice, this transcription factor is important for normal muscle mass maintenance and its sustained inhibition is detrimental to muscle function.
Collapse
Affiliation(s)
- Ning Zhang
- Barshop Institute for Longevity and Aging Studies; University of Texas Health Science Center at San Antonio; 15355 Lambda Drive San Antonio TX 78245 USA
| | - Joseph M. Valentine
- Barshop Institute for Longevity and Aging Studies; University of Texas Health Science Center at San Antonio; 15355 Lambda Drive San Antonio TX 78245 USA
| | - You Zhou
- Barshop Institute for Longevity and Aging Studies; University of Texas Health Science Center at San Antonio; 15355 Lambda Drive San Antonio TX 78245 USA
| | - Mengyao E. Li
- Barshop Institute for Longevity and Aging Studies; University of Texas Health Science Center at San Antonio; 15355 Lambda Drive San Antonio TX 78245 USA
- Joslin Diabetes Center; 1 Joslin Place Boston MA 02215 USA
| | - Yiqiang Zhang
- Barshop Institute for Longevity and Aging Studies; University of Texas Health Science Center at San Antonio; 15355 Lambda Drive San Antonio TX 78245 USA
| | - Arunabh Bhattacharya
- Barshop Institute for Longevity and Aging Studies; University of Texas Health Science Center at San Antonio; 15355 Lambda Drive San Antonio TX 78245 USA
| | - Michael E. Walsh
- Barshop Institute for Longevity and Aging Studies; University of Texas Health Science Center at San Antonio; 15355 Lambda Drive San Antonio TX 78245 USA
| | - Katherine E. Fischer
- Barshop Institute for Longevity and Aging Studies; University of Texas Health Science Center at San Antonio; 15355 Lambda Drive San Antonio TX 78245 USA
| | - Steven N. Austad
- Barshop Institute for Longevity and Aging Studies; University of Texas Health Science Center at San Antonio; 15355 Lambda Drive San Antonio TX 78245 USA
| | - Pawel Osmulski
- Barshop Institute for Longevity and Aging Studies; University of Texas Health Science Center at San Antonio; 15355 Lambda Drive San Antonio TX 78245 USA
| | - Maria Gaczynska
- Barshop Institute for Longevity and Aging Studies; University of Texas Health Science Center at San Antonio; 15355 Lambda Drive San Antonio TX 78245 USA
| | | | - Holly Van Remmen
- Barshop Institute for Longevity and Aging Studies; University of Texas Health Science Center at San Antonio; 15355 Lambda Drive San Antonio TX 78245 USA
| | - Hung I. Chen
- Greehey Children's Cancer Research Institute; 8403 Floyd Curl Dr San Antonio TX 78229 USA
- Department of Epidemiology and Biostatistics; University of Texas Health Science Center at San Antonio; 7703 Floyd Curl Dr San Antonio TX 78229 USA
| | - Yidong Chen
- Greehey Children's Cancer Research Institute; 8403 Floyd Curl Dr San Antonio TX 78229 USA
- Department of Epidemiology and Biostatistics; University of Texas Health Science Center at San Antonio; 7703 Floyd Curl Dr San Antonio TX 78229 USA
| | - Hanyu Liang
- Barshop Institute for Longevity and Aging Studies; University of Texas Health Science Center at San Antonio; 15355 Lambda Drive San Antonio TX 78245 USA
| | - Nicolas Musi
- Barshop Institute for Longevity and Aging Studies; University of Texas Health Science Center at San Antonio; 15355 Lambda Drive San Antonio TX 78245 USA
- San Antonio Geriatric Research, Education and Clinical Center; South Texas Veterans Health Care System; 7400 Merton Minter San Antonio TX 78229 USA
| |
Collapse
|
34
|
Gene expression profiling in Pekin duck embryonic breast muscle. PLoS One 2017; 12:e0174612. [PMID: 28472139 PMCID: PMC5417483 DOI: 10.1371/journal.pone.0174612] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 03/13/2017] [Indexed: 12/18/2022] Open
Abstract
Lean-type Pekin duck is a breed gained through long-term selection and great effort has been exerted to understand the mechanisms underlying increased muscle yields. However, the genes involved in Pekin duck embryonic breast muscle development have not been explored to date. In this study, we investigated gene expression profiles in Pekin Duck embryonic breast muscle at hatched day 13 (E13), E19, and E27 using RNA-seq. In total, we produced 519,312,178 raw reads resulting in 497,348,158 high-quality reads after filtering. The mapping, distribution of reads along annotated genes, and consistency across replicates demonstrates the high quality of the RNA-seq data used in this study, allowing us to continue with the downstream analysis. Significantly fewer differentially expressed genes (DEGs) were identified between E13 and E19 (203 DEGs) compared to E27 and E19 (2,797 DEGs). Many DEGs highly expressed in E19 are involved in metabolic processes and cell division. KEGG analysis showed many pathways associated with fat development were significantly enriched for DEGs highly expressed in E27. These results provide a basis for the further investigation of the mechanisms involved in Pekin duck embryonic breast muscle development.
Collapse
|
35
|
Shi LL, Zhang N, Xie XM, Chen YJ, Wang R, Shen L, Zhou JS, Hu JG, Lü HZ. Transcriptome profile of rat genes in injured spinal cord at different stages by RNA-sequencing. BMC Genomics 2017; 18:173. [PMID: 28201982 PMCID: PMC5312572 DOI: 10.1186/s12864-017-3532-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 02/01/2017] [Indexed: 12/15/2022] Open
Abstract
Background Spinal cord injury (SCI) results in fatal damage and currently has no effective treatment. The pathological mechanisms of SCI remain unclear. In this study, genome-wide transcriptional profiling of spinal cord samples from injured rats at different time points after SCI was performed by RNA-Sequencing (RNA-Seq). The transcriptomes were systematically characterized to identify the critical genes and pathways that are involved in SCI pathology. Results RNA-Seq results were obtained from total RNA harvested from the spinal cords of sham control rats and rats in the acute, subacute, and chronic phases of SCI (1 day, 6 days and 28 days after injury, respectively; n = 3 in every group). Compared with the sham-control group, the number of differentially expressed genes was 1797 in the acute phase (1223 upregulated and 574 downregulated), 6590 in the subacute phase (3460 upregulated and 3130 downregulated), and 3499 in the chronic phase (1866 upregulated and 1633 downregulated), with an adjusted P-value <0.05 by DESeq. Gene ontology (GO) enrichment analysis showed that differentially expressed genes were most enriched in immune response, MHC protein complex, antigen processing and presentation, translation-related genes, structural constituent of ribosome, ion gated channel activity, small GTPase mediated signal transduction and cytokine and/or chemokine activity. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that the most enriched pathways included ribosome, antigen processing and presentation, retrograde endocannabinoid signaling, axon guidance, dopaminergic synapses, glutamatergic synapses, GABAergic synapses, TNF, HIF-1, Toll-like receptor, NF-kappa B, NOD-like receptor, cAMP, calcium, oxytocin, Rap1, B cell receptor and chemokine signaling pathway. Conclusions This study has not only characterized changes in global gene expression through various stages of SCI progression in rats, but has also systematically identified the critical genes and signaling pathways in SCI pathology. These results will expand our understanding of the complex molecular mechanisms involved in SCI and provide a foundation for future studies of spinal cord tissue damage and repair. The sequence data from this study have been deposited into Sequence Read Archive (http://www.ncbi.nlm.nih.gov/sra; accession number PRJNA318311). Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3532-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ling-Ling Shi
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, People's Republic of China.,Department of Immunology, Bengbu Medical College, Anhui, 233030, People's Republic of China
| | - Nan Zhang
- Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Anhui, 233004, People's Republic of China
| | - Xiu-Mei Xie
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, People's Republic of China.,Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Anhui, 233004, People's Republic of China
| | - Yue-Juan Chen
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, People's Republic of China.,Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Anhui, 233004, People's Republic of China
| | - Rui Wang
- Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Anhui, 233004, People's Republic of China
| | - Lin Shen
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, People's Republic of China.,Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Anhui, 233004, People's Republic of China
| | - Jian-Sheng Zhou
- Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Anhui, 233004, People's Republic of China
| | - Jian-Guo Hu
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, People's Republic of China. .,Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Anhui, 233004, People's Republic of China.
| | - He-Zuo Lü
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, People's Republic of China. .,Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Anhui, 233004, People's Republic of China. .,Department of Immunology, Bengbu Medical College, Anhui, 233030, People's Republic of China.
| |
Collapse
|
36
|
Sakuma K, Aoi W, Yamaguchi A. Molecular mechanism of sarcopenia and cachexia: recent research advances. Pflugers Arch 2017; 469:573-591. [PMID: 28101649 DOI: 10.1007/s00424-016-1933-3] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 12/28/2016] [Accepted: 12/29/2016] [Indexed: 12/17/2022]
Abstract
Skeletal muscle provides a fundamental basis for human function, enabling locomotion and respiration. Muscle loss occurs as a consequence of several chronic diseases (cachexia) and normal aging (sarcopenia). Although many negative regulators (atrogin-1, muscle ring finger-1, nuclear factor-kappaB (NF-κB), myostatin, etc.) have been proposed to enhance protein degradation during both sarcopenia and cachexia, the adaptation of these mediators markedly differs within both conditions. Sarcopenia and cachectic muscles have been demonstrated to be abundant in myostatin-linked molecules. The ubiquitin-proteasome system (UPS) is activated during rapid atrophy model (cancer cachexia), but few mediators of the UPS change during sarcopenia. NF-κB signaling is activated in cachectic, but not in sarcopenic, muscle. Recent studies have indicated the age-related defect of autophagy signaling in skeletal muscle, whereas autophagic activation occurs in cachectic muscle. This review provides recent research advances dealing with molecular mediators modulating muscle mass in both sarcopenia and cachexia.
Collapse
Affiliation(s)
- Kunihiro Sakuma
- Institute for Liberal Arts, Environment and Society, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro-ku, Tokyo, 152-8550, Japan.
| | - Wataru Aoi
- Laboratory of Health Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-cho Shimogamo, Sakyo-ku, Kyoto, 606-8522, Japan
| | - Akihiko Yamaguchi
- Department of Physical Therapy, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Hokkaido, 061-0293, Japan
| |
Collapse
|
37
|
Ogilvie H, Cacciani N, Akkad H, Larsson L. Targeting Heat Shock Proteins Mitigates Ventilator Induced Diaphragm Muscle Dysfunction in an Age-Dependent Manner. Front Physiol 2016; 7:417. [PMID: 27729867 PMCID: PMC5037190 DOI: 10.3389/fphys.2016.00417] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 09/05/2016] [Indexed: 01/25/2023] Open
Abstract
Intensive care unit (ICU) patients are often overtly subjected to mechanical ventilation and immobilization, which leads to impaired limb and respiratory muscle function. The latter, termed ventilator-induced diaphragm dysfunction (VIDD) has recently been related to compromised heat shock protein (Hsp) activation. The administration of a pharmacological drug BGP-15 acting as a Hsp chaperone co-inducer has been found to partially alleviate VIDD in young rats. Considering that the mean age in the ICU is increasing, we aimed to explore whether the beneficial functional effects are also present in old rats. For that, we exposed young (7–8 months) and old (28–32 months) rats to 5-day controlled mechanical ventilation and immobilization with or without systemic BGP-15 administration. We then dissected diaphragm muscles, membrane–permeabilized bundles and evaluated the contractile function at single fiber level. Results confirmed that administration of BGP-15 restored the force-generating capacity of isolated muscle cells from young rats in conjunction with an increased expression of Hsp72. On the other hand, our results highlighted that old rats did not positively respond to the BGP-15 treatment. Therefore, it is of crucial importance to comprehend in more depth the effect of VIDD on diaphragm function and ascertain any further age-related differences.
Collapse
Affiliation(s)
- Hannah Ogilvie
- Basic and Clinical Muscle Biology, Department of Physiology and Pharmacology, Karolinska Institutet Stockholm, Sweden
| | - Nicola Cacciani
- Basic and Clinical Muscle Biology, Department of Physiology and Pharmacology, Karolinska Institutet Stockholm, Sweden
| | - Hazem Akkad
- Basic and Clinical Muscle Biology, Department of Physiology and Pharmacology, Karolinska Institutet Stockholm, Sweden
| | - Lars Larsson
- Basic and Clinical Muscle Biology, Department of Physiology and Pharmacology, Karolinska InstitutetStockholm, Sweden; Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska InstitutetStockholm, Sweden
| |
Collapse
|
38
|
Pierre N, Appriou Z, Gratas-Delamarche A, Derbré F. From physical inactivity to immobilization: Dissecting the role of oxidative stress in skeletal muscle insulin resistance and atrophy. Free Radic Biol Med 2016; 98:197-207. [PMID: 26744239 DOI: 10.1016/j.freeradbiomed.2015.12.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 12/23/2015] [Accepted: 12/24/2015] [Indexed: 12/16/2022]
Abstract
In the literature, the terms physical inactivity and immobilization are largely used as synonyms. The present review emphasizes the need to establish a clear distinction between these two situations. Physical inactivity is a behavior characterized by a lack of physical activity, whereas immobilization is a deprivation of movement for medical purpose. In agreement with these definitions, appropriate models exist to study either physical inactivity or immobilization, leading thereby to distinct conclusions. In this review, we examine the involvement of oxidative stress in skeletal muscle insulin resistance and atrophy induced by, respectively, physical inactivity and immobilization. A large body of evidence demonstrates that immobilization-induced atrophy depends on the chronic overproduction of reactive oxygen and nitrogen species (RONS). On the other hand, the involvement of RONS in physical inactivity-induced insulin resistance has not been investigated. This observation outlines the need to elucidate the mechanism by which physical inactivity promotes insulin resistance.
Collapse
Affiliation(s)
- Nicolas Pierre
- EA1274 Laboratory "Movement, Sport and Health Sciences" M2S, Rennes 2 University - ENS Rennes, Bruz, France
| | - Zephyra Appriou
- EA1274 Laboratory "Movement, Sport and Health Sciences" M2S, Rennes 2 University - ENS Rennes, Bruz, France
| | - Arlette Gratas-Delamarche
- EA1274 Laboratory "Movement, Sport and Health Sciences" M2S, Rennes 2 University - ENS Rennes, Bruz, France
| | - Frédéric Derbré
- EA1274 Laboratory "Movement, Sport and Health Sciences" M2S, Rennes 2 University - ENS Rennes, Bruz, France.
| |
Collapse
|
39
|
Yokoyama S, Ohno Y, Egawa T, Yasuhara K, Nakai A, Sugiura T, Ohira Y, Yoshioka T, Okita M, Origuchi T, Goto K. Heat shock transcription factor 1-associated expression of slow myosin heavy chain in mouse soleus muscle in response to unloading with or without reloading. Acta Physiol (Oxf) 2016; 217:325-37. [PMID: 27084024 DOI: 10.1111/apha.12692] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 11/28/2015] [Accepted: 04/11/2016] [Indexed: 12/28/2022]
Abstract
AIM The effects of heat shock transcription factor 1 (HSF1) deficiency on the fibre type composition and the expression level of nuclear factor of activated T cells (NFAT) family members (NFATc1, NFATc2, NFATc3 and NFATc4), phosphorylated glycogen synthase kinase 3α (p-GSK3α) and p-GSK3β, microRNA-208b (miR-208b), miR-499 and slow myosin heavy chain (MyHC) mRNAs (Myh7 and Myh7b) of antigravitational soleus muscle in response to unloading with or without reloading were investigated. METHODS HSF1-null and wild-type mice were subjected to continuous 2-week hindlimb suspension followed by 2- or 4-week ambulation recovery. RESULTS In wild-type mice, the relative population of slow type I fibres, the expression level of NFATc2, p-GSK3 (α and β), miR-208b, miR-499 and slow MyHC mRNAs (Myh7 and Myh7b) were all decreased with hindlimb suspension, but recovered after it. Significant interactions between train and time (the relative population of slow type I fibres; P = 0.01, the expression level of NFATc2; P = 0.001, p-GSKβ; P = 0.009, miR-208b; P = 0.002, miR-499; P = 0.04) suggested that these responses were suppressed in HSF1-null mice. CONCLUSION HSF1 may be a molecule in the regulation of the expression of slow MyHC as well as miR-208b, miR-499, NFATc2 and p-GSK3 (α and β) in mouse soleus muscle.
Collapse
Affiliation(s)
- S. Yokoyama
- Department of Locomotive Rehabilitation Science; Unit of Rehabilitation Sciences; Nagasaki University Graduate School of Biomedical Sciences; Nagasaki Japan
- Laboratory of Physiology; School of Health Science; Toyohashi SOZO University; Toyohashi Japan
| | - Y. Ohno
- Laboratory of Physiology; School of Health Science; Toyohashi SOZO University; Toyohashi Japan
| | - T. Egawa
- Department of Physiology; Graduate School of Health Science; Toyohashi SOZO University; Toyohashi Japan
| | - K. Yasuhara
- Department of Orthopaedic Surgery; St. Marianna University School of Medicine; Kawasaki Japan
| | - A. Nakai
- Department of Molecular Biology; Graduate School of Medicine; Yamaguchi University; Ube Japan
| | - T. Sugiura
- Faculty of Education; Yamaguchi University; Yamaguchi Japan
| | - Y. Ohira
- Faculty and Graduate School of Health and Sports Sciences; Doshisha University; Kyotanabe Japan
| | | | - M. Okita
- Department of Locomotive Rehabilitation Science; Unit of Rehabilitation Sciences; Nagasaki University Graduate School of Biomedical Sciences; Nagasaki Japan
| | - T. Origuchi
- Department of Locomotive Rehabilitation Science; Unit of Rehabilitation Sciences; Nagasaki University Graduate School of Biomedical Sciences; Nagasaki Japan
| | - K. Goto
- Laboratory of Physiology; School of Health Science; Toyohashi SOZO University; Toyohashi Japan
- Department of Physiology; Graduate School of Health Science; Toyohashi SOZO University; Toyohashi Japan
| |
Collapse
|
40
|
Kizilarslanoglu MC, Kuyumcu ME, Yesil Y, Halil M. Sarcopenia in critically ill patients. J Anesth 2016; 30:884-90. [PMID: 27376823 DOI: 10.1007/s00540-016-2211-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 06/26/2016] [Indexed: 12/25/2022]
Abstract
Sarcopenia occurring as a primary consequence of aging and secondary due to certain medical problems including chronic disease, malnutrition and inactivity is a progressive generalized loss of skeletal muscle mass, strength and function. The prevalence of sarcopenia increases with aging (approximately 5-13 % in the sixth and seventh decades). However, data showing the prevalence and clinical outcomes of sarcopenia in intensive care units (ICUs) are limited. A similar condition to sarcopenia in the ICU, called ICU-acquired weakness (ICU-AW), has been reported more frequently. Here, we aim to examine the importance of sarcopenia, especially ICU-AW, in ICU patients via related articles in Medline.
Collapse
Affiliation(s)
- Muhammet C Kizilarslanoglu
- Division of Geriatrics, Department of Internal Medicine, Hacettepe University School of Medicine, 06100, Ankara, Turkey.
| | - Mehmet E Kuyumcu
- Division of Geriatrics, Department of Internal Medicine, Hacettepe University School of Medicine, 06100, Ankara, Turkey
| | - Yusuf Yesil
- Division of Geriatrics, Department of Internal Medicine, Hacettepe University School of Medicine, 06100, Ankara, Turkey
| | - Meltem Halil
- Division of Geriatrics, Department of Internal Medicine, Hacettepe University School of Medicine, 06100, Ankara, Turkey
| |
Collapse
|
41
|
Notch Signaling Mediates Skeletal Muscle Atrophy in Cancer Cachexia Caused by Osteosarcoma. Sarcoma 2016; 2016:3758162. [PMID: 27378829 PMCID: PMC4917717 DOI: 10.1155/2016/3758162] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 04/05/2016] [Accepted: 04/28/2016] [Indexed: 11/17/2022] Open
Abstract
Skeletal muscle atrophy in cancer cachexia is mediated by the interaction between muscle stem cells and various tumor factors. Although Notch signaling has been known as a key regulator of both cancer development and muscle stem cell activity, the potential involvement of Notch signaling in cancer cachexia and concomitant muscle atrophy has yet to be elucidated. The murine K7M2 osteosarcoma cell line was used to generate an orthotopic model of sarcoma-associated cachexia, and the role of Notch signaling was evaluated. Skeletal muscle atrophy was observed in the sarcoma-bearing mice, and Notch signaling was highly active in both tumor tissues and the atrophic skeletal muscles. Systemic inhibition of Notch signaling reduced muscle atrophy. In vitro coculture of osteosarcoma cells with muscle-derived stem cells (MDSCs) isolated from normal mice resulted in decreased myogenic potential of MDSCs, while the application of Notch inhibitor was able to rescue this repressed myogenic potential. We further observed that Notch-activating factors reside in the exosomes of osteosarcoma cells, which activate Notch signaling in MDSCs and subsequently repress myogenesis. Our results revealed that signaling between tumor and muscle via the Notch pathway may play an important role in mediating the skeletal muscle atrophy seen in cancer cachexia.
Collapse
|
42
|
Fielitz J. Cancer cachexia-when proteasomal inhibition is not enough. J Cachexia Sarcopenia Muscle 2016; 7:239-45. [PMID: 27386167 PMCID: PMC4929817 DOI: 10.1002/jcsm.12124] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 04/29/2016] [Indexed: 01/06/2023] Open
Affiliation(s)
- Jens Fielitz
- Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC) Charité--Universitätsmedizin Berlin, Max Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association Berlin Germany; Department of Cardiology Heart Center Brandenburg and Medical School Brandenburg (MHB) Bernau Germany
| |
Collapse
|
43
|
Peddada KV, Peddada KV, Shukla SK, Mishra A, Verma V. Role of Curcumin in Common Musculoskeletal Disorders: a Review of Current Laboratory, Translational, and Clinical Data. Orthop Surg 2016; 7:222-31. [PMID: 26311096 DOI: 10.1111/os.12183] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 06/17/2015] [Indexed: 12/19/2022] Open
Abstract
The Indian spice turmeric, in which the active and dominant biomolecule is curcumin, has been demonstrated to have significant medicinal properties, including anti-inflammatory and anti-neoplastic effects. This promise is potentially very applicable to musculoskeletal disorders, which are common causes of physician visits worldwide. Research at the laboratory, translational and clinical levels that supports the use of curcumin for various musculoskeletal disorders, such as osteoarthritis, osteoporosis, musculocartilaginous disorders, and sarcoma is here in comprehensively summarized. Though more phase I-III trials are clearly needed, thus far the existing data show that curcumin can indeed potentially be useful in treatment of the hundreds of millions worldwide who are afflicted by these musculoskeletal disorders.
Collapse
Affiliation(s)
| | | | - Surendra K Shukla
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Anusha Mishra
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Vivek Verma
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
44
|
RNA-Seq Profiling of Intact and Enucleated Oocyte SCNT Embryos Reveals the Role of Pig Oocyte Nucleus in Somatic Reprogramming. PLoS One 2016; 11:e0153093. [PMID: 27070804 PMCID: PMC4829232 DOI: 10.1371/journal.pone.0153093] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/20/2016] [Indexed: 12/28/2022] Open
Abstract
The specific molecular mechanisms involved in somatic reprogramming remain unidentified. Removal of the oocyte genome is one of the primary causes of developmental failure in cloned embryos, whereas intact oocyte shows stronger reprogramming capability than enucleated oocyte. To identify the reason for the low efficiency of cloning and elucidate the mechanisms involved in somatic reprogramming by the oocyte nucleus, we injected pig cumulus cells into 539 intact MII oocytes and 461 enucleated MII oocytes. Following activation, 260 polyploidy embryos developed to the blastocyst stage whereas only 93 traditionally cloned embryos (48.2% vs. 20.2%, P < 0.01) reached blastocyst stage. Blastocysts generated from intact oocytes also had more cells than those generated from enucleated oocytes (60.70 vs. 46.65, P < 0.01). To identify the genes that contribute to this phenomenon, two early embryos in 2-cell and 4-cell stages were collected for single-cell RNA sequencing. The two kinds of embryos were found to have dramatically different transcriptome profiles. Intact oocyte nuclear transfer embryos showed 1,738 transcripts that were up-regulated relative to enucleated cloned embryos at the 2-cell stage and 728 transcripts that were down-regulated (|log2Ratio| ≥ 5). They showed 2,941 transcripts that were up-regulated during the 4-cell stage and 1,682 that were down-regulated (|log2Ratio| ≥ 5). The most significantly enriched gene ontology categories were those involved in the regulation of binding, catalytic activity, and molecular transducer activity. Other genes that were notably up-regulated and expressed in intact oocyte nuclear transfer embryos were metabolic process. This study provides a comprehensive profile of the differences in gene expression between intact oocyte nuclear transfer embryos and traditional nuclear transfer embryos. This work thus paves the way for further research on the mechanisms underlying somatic reprogramming by oocytes.
Collapse
|
45
|
The Systemic Effect of Burn Injury and Trauma on Muscle and Bone Mass and Composition. Plast Reconstr Surg 2016; 136:612e-623e. [PMID: 26505718 DOI: 10.1097/prs.0000000000001723] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND By understanding the global inflammatory effects on distant myopathies, surgeons can better guide the rehabilitative process for burn patients. The authors tested the systemic effect of burn injury on distant injured muscle and native bone using immunohistochemistry and validated a new morphometric analytic modality to reproducibly quantify muscle atrophy using computed tomographic imaging. METHODS In vivo studies were performed on C57/BL6 mice using an Achilles tenotomy with concurrent burn injury model. Total muscle and bone (tibia and fibula) volume/density were quantified near the site of Achilles tenotomy using micro-computed tomography at 5, 7, and 9 to 12 weeks after surgery. The impact of burn injury on the inflammatory cascade [nuclear factor (NF)-κB, p-NF-κB] and the interconnected protein catabolism signaling pathway (Atrogin-1) was assessed by immunohistochemistry. RESULTS Muscle volume and density at the site of Achilles tenotomy in burned mice were significantly diminished compared with nonburned mice at 5 weeks and 9 to 12 weeks. Similar decreases in muscle volume and density were observed when comparing tenotomy to no tenotomy. Cortical bone health remained stable in burn/tenotomy mice compared with tenotomy. Muscle atrophy was associated with up-regulation of p-NF-κB, NF-κB, and Atrogin-1 assessed by immunohistochemistry. CONCLUSIONS Burn injury significantly decreases muscle volume and density. Increased muscle atrophy using our computed tomographic morphometric analysis correlated with a significant increase in intramuscular inflammatory markers and proteolysis enzymes. This study demonstrates a unique characterization of how burn injuries may worsen local myopathy. Moreover, it provides a novel approach for quantifying muscle atrophy over an expanded period.
Collapse
|
46
|
Tryon LD, Vainshtein A, Memme J, Crilly MJ, Hood DA. WITHDRAWN: Relationship between the regulation of muscle atrophy and mitochondrial turnover during chronic disuse. Integr Med Res 2016. [DOI: 10.1016/j.imr.2014.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
47
|
Malavaki CJ, Sakkas GK, Mitrou GI, Kalyva A, Stefanidis I, Myburgh KH, Karatzaferi C. Skeletal muscle atrophy: disease-induced mechanisms may mask disuse atrophy. J Muscle Res Cell Motil 2016; 36:405-21. [DOI: 10.1007/s10974-015-9439-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 12/08/2015] [Indexed: 01/07/2023]
|
48
|
Lee D, Goldberg AL. Muscle Wasting in Fasting Requires Activation of NF-κB and Inhibition of AKT/Mechanistic Target of Rapamycin (mTOR) by the Protein Acetylase, GCN5. J Biol Chem 2015; 290:30269-79. [PMID: 26515065 DOI: 10.1074/jbc.m115.685164] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Indexed: 12/25/2022] Open
Abstract
NF-κB is best known for its pro-inflammatory and anti-apoptotic actions, but in skeletal muscle, NF-κB activation is important for atrophy upon denervation or cancer. Here, we show that also upon fasting, NF-κB becomes activated in muscle and is critical for the subsequent atrophy. Following food deprivation, the expression and acetylation of the p65 of NF-κB on lysine 310 increase markedly in muscles. NF-κB inhibition in mouse muscles by overexpression of the IκBα superrepressor (IκBα-SR) or of p65 mutated at Lys-310 prevented atrophy. Knockdown of GCN5 with shRNA or a dominant-negative GCN5 or overexpression of SIRT1 decreased p65K310 acetylation and muscle wasting upon starvation. In addition to reducing atrogene expression, surprisingly inhibiting NF-κB with IκBα-SR or by GCN5 knockdown in these muscles also enhanced AKT and mechanistic target of rapamycin (mTOR) activities, which also contributed to the reduction in atrophy. These new roles of NF-κB and GCN5 in regulating muscle proteolysis and AKT/mTOR signaling suggest novel approaches to combat muscle wasting.
Collapse
Affiliation(s)
- Donghoon Lee
- From the Department of Cell Biology, Harvard Medical School, Boston, Masachusetts 02115
| | - Alfred L Goldberg
- From the Department of Cell Biology, Harvard Medical School, Boston, Masachusetts 02115
| |
Collapse
|
49
|
Egawa T, Goto A, Ohno Y, Yokoyama S, Ikuta A, Suzuki M, Sugiura T, Ohira Y, Yoshioka T, Hayashi T, Goto K. Involvement of AMPK in regulating slow-twitch muscle atrophy during hindlimb unloading in mice. Am J Physiol Endocrinol Metab 2015; 309:E651-62. [PMID: 26244519 DOI: 10.1152/ajpendo.00165.2015] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 08/03/2015] [Indexed: 01/08/2023]
Abstract
AMPK is considered to have a role in regulating skeletal muscle mass. However, there are no studies investigating the function of AMPK in modulating skeletal muscle mass during atrophic conditions. In the present study, we investigated the difference in unloading-associated muscle atrophy and molecular functions in response to 2-wk hindlimb suspension between transgenic mice overexpressing the dominant-negative mutant of AMPK (AMPK-DN) and their wild-type (WT) littermates. Male WT (n = 24) and AMPK-DN (n = 24) mice were randomly divided into two groups: an untreated preexperimental control group (n = 12 in each group) and an unloading (n = 12 in each group) group. The relative soleus muscle weight and fiber cross-sectional area to body weight were decreased by ∼30% in WT mice by hindlimb unloading and by ∼20% in AMPK-DN mice. There were no changes in puromycin-labeled protein or Akt/70-kDa ribosomal S6 kinase signaling, the indicators of protein synthesis. The expressions of ubiquitinated proteins and muscle RING finger 1 mRNA and protein, markers of the ubiquitin-proteasome system, were increased by hindlimb unloading in WT mice but not in AMPK-DN mice. The expressions of molecules related to the protein degradation system, phosphorylated forkhead box class O3a, inhibitor of κBα, microRNA (miR)-1, and miR-23a, were decreased only in WT mice in response to hindlimb unloading, and 72-kDa heat shock protein expression was higher in AMPK-DN mice than in WT mice. These results imply that AMPK partially regulates unloading-induced atrophy of slow-twitch muscle possibly through modulation of the protein degradation system, especially the ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Tatsuro Egawa
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University, Toyohashi, Aichi, Japan
| | - Ayumi Goto
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University, Toyohashi, Aichi, Japan; Laboratory of Sports and Exercise Medicine, Graduate School of Human and Environmental Studies, Kyoto University, Kyoto, Japan
| | - Yoshitaka Ohno
- Laboratory of Physiology, School of Health Sciences, Toyohashi SOZO University, Toyohashi, Aichi, Japan
| | - Shingo Yokoyama
- Laboratory of Physiology, School of Health Sciences, Toyohashi SOZO University, Toyohashi, Aichi, Japan
| | - Akihiro Ikuta
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University, Toyohashi, Aichi, Japan
| | - Miho Suzuki
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University, Toyohashi, Aichi, Japan
| | - Takao Sugiura
- Department of Exercise and Sports Physiology, Faculty of Education, Yamaguchi University, Yamaguchi, Japan
| | - Yoshinobu Ohira
- Graduate School of Health and Sports Science, Doshisha University, Kyotanabe, Kyoto, Japan; and
| | | | - Tatsuya Hayashi
- Laboratory of Sports and Exercise Medicine, Graduate School of Human and Environmental Studies, Kyoto University, Kyoto, Japan
| | - Katsumasa Goto
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University, Toyohashi, Aichi, Japan;
| |
Collapse
|
50
|
Minegishi Y, Haramizu S, Misawa K, Shimotoyodome A, Hase T, Murase T. Deletion of nuclear factor-κB p50 upregulates fatty acid utilization and contributes to an anti-obesity and high-endurance phenotype in mice. Am J Physiol Endocrinol Metab 2015; 309:E523-33. [PMID: 26173458 DOI: 10.1152/ajpendo.00071.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 07/12/2015] [Indexed: 02/06/2023]
Abstract
The transcription factor nuclear factor-κB (NF-κB) plays an important role in regulating physiological processes such as immunity and inflammation. In addition to this primary role, NF-κB interacts physically with peroxisome proliferator-activated receptors regulating lipid metabolism-related gene expression and inhibits their transcriptional activity. Therefore, inhibition of NF-κB may promote fatty acid utilization, which could ameliorate obesity and improve endurance capacity. To test this hypothesis, we attempted to elucidate the energy metabolic status of mice lacking the p50 subunit of NF-κB (p50 KO mice) from the tissue to whole body level. p50 KO mice showed a significantly lower respiratory quotient throughout the day than did wild-type (WT) mice; this decrease was associated with increased fatty acid oxidation activity in liver and gastrocnemius muscle of p50 KO mice. p50 KO mice that were fed a high-fat diet were also resistant to fat accumulation and adipose tissue inflammation. Furthermore, p50 KO mice showed a significantly longer maximum running time compared with WT mice, with a lower respiratory exchange ratio during exercise as well as higher residual muscle glycogen content and lower blood lactate levels after exercise. These results suggest that p50 deletion facilitates fatty acid catabolism, leading to an anti-obesity and high-endurance phenotype of mice and supporting the idea that NF-κB is an important regulator of energy metabolism.
Collapse
Affiliation(s)
| | - Satoshi Haramizu
- Biological Science Laboratories, Kao Corporation, Tochigi, Japan
| | - Koichi Misawa
- Biological Science Laboratories, Kao Corporation, Tochigi, Japan
| | | | - Tadashi Hase
- Biological Science Laboratories, Kao Corporation, Tochigi, Japan
| | - Takatoshi Murase
- Biological Science Laboratories, Kao Corporation, Tochigi, Japan
| |
Collapse
|