1
|
Roberts BM, Geddis AV, Matheny RW. The dose-response effects of flurbiprofen, indomethacin, ibuprofen, and naproxen on primary skeletal muscle cells. J Int Soc Sports Nutr 2024; 21:2302046. [PMID: 38198469 PMCID: PMC10783825 DOI: 10.1080/15502783.2024.2302046] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Non-steroidal anti-inflammatory drugs (NSAIDs) like ibuprofen, flurbiprofen, naproxen sodium, and indomethacin are commonly employed for their pain-relieving and inflammation-reducing qualities. NSAIDs work by blocking COX-1 and/or COX-2, enzymes which play roles in inflammation, fever, and pain. The main difference among NSAIDs lies in their affinity to these enzymes, which in turn, influences prostaglandin secretion, and skeletal muscle growth and regeneration. The current study investigated the effects of NSAIDs on human skeletal muscle cells, focusing on myoblast proliferation, differentiation, and muscle protein synthesis signaling. METHODS Using human primary muscle cells, we examined the dose-response impact of flurbiprofen (25-200 µM), indomethacin (25-200 µM), ibuprofen (25-200 µM), and naproxen sodium (25-200 µM), on myoblast viability, myotube area, fusion, and prostaglandin production. RESULTS We found that supraphysiological concentrations of indomethacin inhibited myoblast proliferation (-74 ± 2% with 200 µM; -53 ± 3% with 100 µM; both p < 0.05) compared to control cells and impaired protein synthesis signaling pathways in myotubes, but only attenuated myotube fusion at the highest concentrations (-18 ± 2% with 200 µM, p < 0.05) compared to control myotubes. On the other hand, ibuprofen had no such effects. Naproxen sodium only increased cell proliferation at low concentrations (+36 ± 2% with 25 µM, p < 0.05), and flurbiprofen exhibited divergent impacts depending on the concentration whereby low concentrations improved cell proliferation (+17 ± 1% with 25 µM, p < 0.05) but high concentrations inhibited cell proliferation (-32 ± 1% with 200 µM, p < 0.05). CONCLUSION Our findings suggest that indomethacin, at high concentrations, may detrimentally affect myoblast proliferation and differentiation via an AKT-dependent mechanism, and thus provide new understanding of NSAIDs' effects on skeletal muscle cell development.
Collapse
Affiliation(s)
- Brandon M. Roberts
- Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
| | - Alyssa V. Geddis
- Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
| | - Ronald W. Matheny
- Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
- Military Operational Medicine Research Program, Detrick, MD, USA
| |
Collapse
|
2
|
Belal SA, Lee J, Park J, Kang D, Shim K. The Effects of Oleic Acid and Palmitic Acid on Porcine Muscle Satellite Cells. Foods 2024; 13:2200. [PMID: 39063284 PMCID: PMC11276066 DOI: 10.3390/foods13142200] [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: 05/26/2024] [Revised: 06/12/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
We aimed to determine the effects of oleic acid (OA) and palmitic acid (PA), alone or in combination, on proliferation, differentiation, triacylglycerol (TAG) content, and gene expression in porcine muscle satellite cells (PMSCs). Results revealed that OA-alone- and PA + OA-treated PMSCs showed significantly increased viability than those in the control or PA-alone-treated groups. No significant effects on apoptosis were observed in all three treatments, whereas necrosis was significantly lower in OA-alone- and PA + OA-treated groups than in the control and PA-alone-treated groups. Myotube formation significantly increased in OA-alone and PA + OA-treated PMSCs than in the control and PA-alone-treated PMSCs. mRNA expression of the myogenesis-related genes MyoD1 and MyoG and of the adipogenesis-related genes PPARα, C/EBPα, PLIN1, FABP4, and FAS was significantly upregulated in OA-alone- and PA + OA-treated cells compared to control and PA-alone-treated cells, consistent with immunoblotting results for MyoD1 and MyoG. Supplementation of unsaturated fatty acid (OA) with/without saturated fatty acid (PA) significantly stimulated TAG accumulation in treated cells compared to the control and PA-alone-treated PMSCs. These results indicate that OA (alone and with PA) promotes proliferation by inhibiting necrosis and promoting myotube formation and TAG accumulation, likely upregulating myogenesis- and adipogenesis-related gene expression by modulating the effects of PA in PMSCs.
Collapse
Affiliation(s)
- Shah Ahmed Belal
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju 54896, Republic of Korea; (S.A.B.); (D.K.)
- Department of Poultry Science, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | - Jeongeun Lee
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju 54896, Republic of Korea;
| | - Jinryong Park
- Food Processing Research Group, Korea Food Research Institute, Wanju 55365, Republic of Korea;
| | - Darae Kang
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju 54896, Republic of Korea; (S.A.B.); (D.K.)
| | - Kwanseob Shim
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju 54896, Republic of Korea; (S.A.B.); (D.K.)
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju 54896, Republic of Korea;
| |
Collapse
|
3
|
Moreno N, Sabater-Arcis M, Sevilla T, Alonso MP, Ohana J, Bargiela A, Artero R. Therapeutic potential of oleic acid supplementation in myotonic dystrophy muscle cell models. Biol Res 2024; 57:29. [PMID: 38760841 PMCID: PMC11100173 DOI: 10.1186/s40659-024-00496-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 04/05/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND We recently reported that upregulation of Musashi 2 (MSI2) protein in the rare neuromuscular disease myotonic dystrophy type 1 contributes to the hyperactivation of the muscle catabolic processes autophagy and UPS through a reduction in miR-7 levels. Because oleic acid (OA) is a known allosteric regulator of MSI2 activity in the biogenesis of miR-7, here we sought to evaluate endogenous levels of this fatty acid and its therapeutic potential in rescuing cell differentiation phenotypes in vitro. In this work, four muscle cell lines derived from DM1 patients were treated with OA for 24 h, and autophagy and muscle differentiation parameters were analyzed. RESULTS We demonstrate a reduction of OA levels in different cell models of the disease. OA supplementation rescued disease-related phenotypes such as fusion index, myotube diameter, and repressed autophagy. This involved inhibiting MSI2 regulation of direct molecular target miR-7 since OA isoschizomer, elaidic acid (EA) could not cause the same rescues. Reduction of OA levels seems to stem from impaired biogenesis since levels of the enzyme stearoyl-CoA desaturase 1 (SCD1), responsible for converting stearic acid to oleic acid, are decreased in DM1 and correlate with OA amounts. CONCLUSIONS For the first time in DM1, we describe a fatty acid metabolism impairment that originated, at least in part, from a decrease in SCD1. Because OA allosterically inhibits MSI2 binding to molecular targets, reduced OA levels synergize with the overexpression of MSI2 and contribute to the MSI2 > miR-7 > autophagy axis that we proposed to explain the muscle atrophy phenotype.
Collapse
Affiliation(s)
- Nerea Moreno
- Human Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
- CIBERER, IISCIII, Madrid, Spain
| | - Maria Sabater-Arcis
- Human Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
- CIBERER, IISCIII, Madrid, Spain
| | - Teresa Sevilla
- CIBERER, IISCIII, Madrid, Spain
- Neuromuscular and Ataxias Research Group, Health Research Institute Hospital, La Fe (IIS La Fe), Valencia, Spain
- Department of Medicine, University of Valencia, Valencia, Spain
| | - Manuel Perez Alonso
- Human Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
- CIBERER, IISCIII, Madrid, Spain
| | - Jessica Ohana
- Centre de Recherche en Myologie, Sorbonne Université, Inserm, Institut de Myologie, Paris, F-75013, France
| | - Ariadna Bargiela
- CIBERER, IISCIII, Madrid, Spain.
- Neuromuscular and Ataxias Research Group, Health Research Institute Hospital, La Fe (IIS La Fe), Valencia, Spain.
| | - Ruben Artero
- Human Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
- CIBERER, IISCIII, Madrid, Spain
| |
Collapse
|
4
|
Zeitler C, Braune S, Waldeck P, Jung CG, Küpper JH, Petrick I, Jung F. Response of Arthrospira platensis to nitrogen depletion and the effect of aqueous extracts on tumor and non-tumor cells. Clin Hemorheol Microcirc 2024; 88:S95-S111. [PMID: 39331099 PMCID: PMC11613083 DOI: 10.3233/ch-248108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
The microalgae Arthrospira platensis (AP), commonly known as Spirulina, has gained widespread popularity as a food supplement in recent years. AP is particularly abundant in protein, B vitamins, iron, magnesium, potassium, and various antioxidants. In this study we aimed to evaluate the effect of nitrate limitation in the AP culture medium on AP growth and composition. In addition, the cytotoxicity of the respective aqueous AP extracts on three different mammalian cell-lines (HepG2, Caco2, L929) was tested. AP was cultivated over a 10-day period under nitrogen-rich (Nrich: 1.8 g/L) and nitrogen-deficient (Nlimited: 0.2-0.4 g/L) conditions in two separate experiments, each with three biological replicates (three bioreactors). Throughout the cultivation, the kinetic progress of dry biomass, pH, pigment content, the levels of essential elements (sulphur, phosphate, and nitrate) and the composition of elements in the harvested biomass was determined. While the biomass slightly but significantly differed, the phycocyanin concentration differed considerably (around 10-fold higher in the Nrich medium, p < 0.05). Aqueous extracts of the Nrich medium had significantly stronger effects on the cell membrane integrity and the metabolic activity of the cells than extracts of the Nlimited medium. Particularly was the finding that AP had a significantly stronger toxic effect on the two tumour cell types (HepG2, Caco2) than on the non-tumour cells (L929). This study underscores the significance of nitrate content in the cultivation media of AP.
Collapse
Affiliation(s)
- Christina Zeitler
- Institute of Biotechnology, Phototrophic Biotechnology, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology, Cottbus-Senftenberg, Senftenberg, Germany
| | - Steffen Braune
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology, Cottbus-Senftenberg, Senftenberg, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Peter Waldeck
- Institute of Biotechnology, Phototrophic Biotechnology, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Conrad G.H. Jung
- Institute of Biotechnology, Phototrophic Biotechnology, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
- Carbon Biotech, Social Enterprise Stiftungs AG, Senftenberg, Germany
| | - Jan-Heiner Küpper
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology, Cottbus-Senftenberg, Senftenberg, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
- Carbon Biotech, Social Enterprise Stiftungs AG, Senftenberg, Germany
| | - Ingolf Petrick
- Institute of Biotechnology, Phototrophic Biotechnology, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Friedrich Jung
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology, Cottbus-Senftenberg, Senftenberg, Germany
| |
Collapse
|
5
|
Yan E, Guo J, Yin J. Nutritional regulation of skeletal muscle energy metabolism, lipid accumulation and meat quality in pigs. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 14:185-192. [PMID: 37808951 PMCID: PMC10556049 DOI: 10.1016/j.aninu.2023.04.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/12/2023] [Accepted: 04/05/2023] [Indexed: 10/10/2023]
Abstract
The quality of pork determines consumers' purchase intention, which directly affects the economic value of pork. Minimizing the proportion of inferior pork and producing high quality pork are the ultimate goals of the pig industry. Muscle energy metabolism, serving as a regulative hub in organism energy expenditure and storage as a fat deposit, is compatible with myofiber type composition, affecting meat color, intramuscular fat content, tenderness, pH values and drip loss. Increasing data illustrate that dietary nutrients and bioactive ingredients affect muscle energy metabolism, white adipose browning and fat distribution, and myofiber type composition in humans, and rodents. Recently, some studies have shown that modulating muscle energy metabolism and lipid accumulation through nutritional approaches could effectively improve meat quality. This article reviews the progress and development in this field, and specifically discusses the impacts of dietary supply of amino acids, lipids, and gut microbiota as well as maternal nutrition on skeletal muscle energy metabolism, lipid accumulation and meat quality of pigs, so as to provide comprehensive overview with respect to effective avenues for improving meat quality.
Collapse
Affiliation(s)
- Enfa Yan
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jianxin Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jingdong Yin
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
6
|
Son Y, Lorenz WW, Paton CM. Linoleic acid-induced ANGPTL4 inhibits C2C12 skeletal muscle differentiation by suppressing Wnt/β-catenin. J Nutr Biochem 2023; 116:109324. [PMID: 36963729 DOI: 10.1016/j.jnutbio.2023.109324] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 02/15/2023] [Accepted: 03/16/2023] [Indexed: 03/26/2023]
Abstract
Skeletal muscle differentiation is an essential process in embryonic development as well as regeneration and repair throughout the lifespan. It is well-known that dietary fat intake impacts biological and physiological function in skeletal muscle, however, understanding of the contribution of nutritional factors in skeletal muscle differentiation is limited. Therefore, the objective of the current study was to evaluate the effects of free fatty acids (FFAs) on skeletal muscle differentiation in vitro. We used C2C12 murine myoblasts and treated them with various FFAs, which revealed a unique response of angiopoietin-like protein-4 (ANGPTL4) with linoleic acid (LA) treatment that was associated with reduced differentiation. LA significantly inhibited myotube formation and lowered the protein expression of myogenic regulatory factors, including MyoD and MyoG and increased Pax7 during cell differentiation. Next, recombinant ANGPTL4 protein or siRNA knockdown of ANGPTL4 was employed to examine its role in skeletal muscle differentiation, and we confirmed that ANGPTL4 knockdown at day 2 and -6 of differentiation restored myotube formation in the presence of LA. RNA-sequencing analysis revealed that ANGPTL4-mediated inhibition of skeletal muscle differentiation at day 2 as well as LA at day 2 or -6 led to a reduction in Wnt/β-catenin signaling pathways. We confirmed that LA reduced Wnt11 and Axin2 while increasing expression of the Wnt inhibitor, Dkk2. ANGPTL4 knockdown increased β-catenin protein in the nucleus in response to LA and increased Axin2 and Wnt11 expression. Taken together, these results demonstrate that LA induced ANGPTL4 inhibits C2C12 differentiation by suppressing Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Yura Son
- Department of Nutritional Sciences
| | - W Walter Lorenz
- Georgia Genomics and Bioinformatics Core and Institute of Bioinformatics
| | - Chad M Paton
- Department of Nutritional Sciences; Department of Food Science & Technology, University of Georgia, Athens, GA, USA.
| |
Collapse
|
7
|
Gong S, Yin Y, Han M, Guo L, Duan Y, Guo Q, Yin J, Li F. Dietary leucine and fish oil cooperatively regulate skeletal myofiber type transformation via the CaMKII signaling pathway of pigs. Food Funct 2023; 14:133-147. [PMID: 36524418 DOI: 10.1039/d2fo03338k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The study investigated the effects of dietary leucine (Leu) and fish oil (FO) on skeletal myofiber type transformations in pigs and their potential interactions. The results showed that Leu increased the content of Leu, upregulated myocyte enhancer factor-2C (MEF2C) and activated the CaMKII-AMPK/SIRT1-PGC-1α pathway in the longissimus dorsi (LD) muscle. FO increased adiponectin and fatty acid beta-oxidation of LD muscle. Activation of the adiponectin signaling pathway by FO further enhanced the CaMKII pathway and upregulated the expression of MEF2C. Moreover, we found that Leu increased cyclic AMP and caffeine, and FO increased linoleic acid and glutamine in muscle metabolites, which may be the cause of myofiber conversion. In conclusion, this study demonstrated that dietary Leu and FO co-regulated the transformation from glycolytic to oxidative skeletal myofiber type. It is hypothesized that there is an interaction between amino acids and polyunsaturated fatty acids, possibly via the CaMKII signaling pathway to upregulate MEF2 and mitochondrial biogenesis.
Collapse
Affiliation(s)
- Saiming Gong
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China. .,Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
| | - Yunju Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China. .,Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
| | - Mengmeng Han
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China. .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liu Guo
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China. .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yehui Duan
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
| | - Qiuping Guo
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China.
| | - Fengna Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China. .,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
8
|
Coapplication of Magnesium Supplementation and Vibration Modulate Macrophage Polarization to Attenuate Sarcopenic Muscle Atrophy through PI3K/Akt/mTOR Signaling Pathway. Int J Mol Sci 2022; 23:ijms232112944. [PMID: 36361730 PMCID: PMC9654727 DOI: 10.3390/ijms232112944] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 11/23/2022] Open
Abstract
Sarcopenia is an age-related geriatric syndrome characterized by the gradual loss of muscle mass and function. Low-magnitude high-frequency vibration (LMHFV) was shown to be beneficial to structural and functional outcomes of skeletal muscles, while magnesium (Mg) is a cofactor associated with better indices of skeletal muscle mass and strength. We hypothesized that LMHFV, Mg and their combinations could suppress inflammation and sarcopenic atrophy, promote myogenesis via PI3k/Akt/mTOR pathway in senescence-accelerated mouse P8 (SAMP8) mice and C2C12 myoblasts. Results showed that Mg treatment and LMHFV could significantly decrease inflammatory expression (C/EBPα and LYVE1) and modulate a CD206-positive M2 macrophage population at month four. Mg treatment also showed significant inhibitory effects on FOXO3, MuRF1 and MAFbx mRNA expression. Coapplication showed a synergistic effect on suppression of type I fiber atrophy, with significantly higher IGF-1, MyoD, MyoG mRNA (p < 0.05) and pAkt protein expression (p < 0.0001) during sarcopenia. In vitro inhibition of PI3K/Akt and mTOR abolished the enhancement effects on myotube formation and inhibited MRF mRNA and p85, Akt, pAkt and mTOR protein expressions. The present study demonstrated that the PI3K/Akt/mTOR pathway is the predominant regulatory mechanism through which LMHFV and Mg enhanced muscle regeneration and suppressed atrogene upregulation.
Collapse
|
9
|
Liu XQ, Feng L, Wu P, Liu Y, Ren HM, Jin XW, Kuang SY, Li SW, Tang L, Zhang L, Mi HF, Zhou XQ, Jiang WD. Physicochemical property optimization and nutrient redistribution in the muscle of sub-adult grass carp (Ctenopharyngodon idella) by conjugated linoleic acid. Food Chem X 2022; 15:100412. [PMID: 36211744 PMCID: PMC9532757 DOI: 10.1016/j.fochx.2022.100412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 07/21/2022] [Accepted: 08/04/2022] [Indexed: 11/29/2022] Open
Abstract
The demand for high-quality fish products increasing from consumers. Conjugated linoleic acid (CLA) caused nutrient redistribution in fish muscle. CLA improved muscle beneficial fatty acids composition of fish. CLA increased shear force involved collagen synthesis and myofiber growth.
We studied the effects of conjugated linoleic acid (CLA) on the amount of nutrients, flavour substances, and healthcare fatty acids, the physicochemical properties, and the potential molecular mechanisms in the muscles of sub-adult grass carp (Ctenopharyngodon idella). Fish were fed graded levels of CLA (0.0, 3.1, 6.4, 9.6, 12.7, and 15.9 g/kg diets) for 60 days. Protein, glutamic acid, alanine, inosine monophosphate (IMP), eicosapentaenoic acid (EPA; 20:5n-3), docosahexaenoic acid (DHA; 22:6n-3), and total CLA contents (p < 0.05) increased in CLA 3.1 ∼ 12.7, 6.4 ∼ 9.6, 6.4 ∼ 9.6, 6.4 ∼ 15.9, 3.1 ∼ 9.6, 3.1 ∼ 9.6, and 3.1 ∼ 15.9 g/kg diet, respectively (p < 0.05). In addition, optimal CLA significantly increased pH24, shear force, collagen content, and myofibre density in the muscle (P < 0.05); however, it decreased myofibre diameter (p < 0.05). We concluded that 6–9 g/kg CLA in the diet could improve the flesh quality of sub-adult grass carp.
Collapse
Affiliation(s)
- Xiao-Qing Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production, University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Province, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production, University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production, University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, China
| | - Hong-Mei Ren
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiao-Wan Jin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Sheng-Yao Kuang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Shu-Wei Li
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Lu Zhang
- Tongwei Co., Ltd., Healthy Aquaculture Key Laboratory of Sichuan Province, Chengdu, Sichuan 610041, China
| | - Hai-Feng Mi
- Tongwei Co., Ltd., Healthy Aquaculture Key Laboratory of Sichuan Province, Chengdu, Sichuan 610041, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production, University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Province, China
- Corresponding authors at: Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production, University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, China
- Corresponding authors at: Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| |
Collapse
|
10
|
Sakai H, Matsumoto K, Urano T, Sakane F. Myristic acid selectively augments β-tubulin levels in C2C12 myotubes via diacylglycerol kinase δ. FEBS Open Bio 2022; 12:1788-1796. [PMID: 35856166 PMCID: PMC9527581 DOI: 10.1002/2211-5463.13466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/29/2022] [Accepted: 07/19/2022] [Indexed: 12/14/2022] Open
Abstract
Effective amelioration of type II diabetes requires therapies that increase both glucose uptake activity per cell and skeletal muscle mass. Myristic acid (14:0) increases diacylglycerol kinase (DGK) δ protein levels and enhances glucose uptake in myotubes in a DGKδ-dependent manner. However, it is still unclear whether myristic acid treatment affects skeletal muscle mass. In this study, we found that myristic acid treatment increased the protein level of β-tubulin, which constitutes microtubules and is closely related to muscle mass, in C2C12 myotubes but not in the proliferation stage in C2C12 myoblasts. However, lauric (12:0), palmitic (16:0) and oleic (18:1) acids failed to affect DGKδ and β-tubulin protein levels in C2C12 myotubes. Moreover, knockdown of DGKδ by siRNA significantly inhibited the increased protein level of β-tubulin in the presence of myristic acid, suggesting that the increase in β-tubulin protein by myristic acid depends on DGKδ. These results indicate that myristic acid selectively affects β-tubulin protein levels in C2C12 myotubes via DGKδ, suggesting that this fatty acid improves skeletal muscle mass in addition to increasing glucose uptake activity per cell.
Collapse
Affiliation(s)
- Hiromichi Sakai
- Department of Biosignaling and Radioisotope Experiment, Interdisciplinary Center for Science Research, Organization for Research and Academic InformationShimane UniversityIzumoJapan
| | - Ken‐ichi Matsumoto
- Department of Biosignaling and Radioisotope Experiment, Interdisciplinary Center for Science Research, Organization for Research and Academic InformationShimane UniversityIzumoJapan
| | - Takeshi Urano
- Department of BiochemistryShimane University School of MedicineIzumoJapan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of ScienceChiba UniversityJapan
| |
Collapse
|
11
|
Gope A, Mukhopadhyay A, Mukhopadhyay O, Chatterjee J. Regenerative repair of full thickness skin wound assisted by dual crosslinking percolative gel casting maneuvered alginate hydrogel embedded with honey ghee blend resembles standard cutaneous properties. J Tissue Viability 2022; 31:657-672. [PMID: 35870995 DOI: 10.1016/j.jtv.2022.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 04/12/2022] [Accepted: 07/15/2022] [Indexed: 10/17/2022]
Abstract
We report synergism in scarless cutaneous wound repair by alginate hydrogel (HGSAG) embedded with an optimized blend of characterized Jamun honey and characterized indigenously prepared ghee. Thorough screening and characterization of honey and ghee are carried out followed by obtaining a novel dual crosslinking percolative gel casting fabrication method to come up with HGSAG showing superior chemical stability, and mechanical strength (Nanoindentation study; lowest stiffness: 0.71 ± 0.19 μN/nm), and surface morphology (SEM; highest roughness: 0.13 ± 0.04 μm) to other variants. In vitro swelling study and degradation behavior study show intermediate swelling (swelling index: 0.59 ± 0.008 in 98 h) and required restricted degradation (PBS: 73.38 ± 0.55%, DMEM: 83.48 ± 0.69% in 10 days) for HGSAG which is necessary for providing nutrients to cells and in vivo therapeutic efficacy. We observe the remarkable antibacterial efficacy of HGSAG against Staphylococcus mutans and Escherichia coli. This particular substrate also shows decent 3T3 fibroblasts viability, cell-cell communication followed by cell-matrix interaction, and proliferation compared to other variants. Molecular gene expression studies by quantitative RT-PCR technique reveal strong upregulation of collagen I, CD26, and TGF-β3 while downregulation in the case of TGF-β1 which eventually substantiates scarless wound healing potential of HGSAG. Wound closure kinetics is most rapidly and successfully underpinned by HGSAG while compared to other alternatives including marketed healing patches. Regular close monitoring using histopathological studies and real-time imaging by Swept-Source Optical Coherence Tomography of in vivo wound model treated with HGSAG come up with the fascinating result of scarless healing (HGSAG treated epithelial thickness: 62.96 ± 0.67 μm, unwounded akin epithelial thickness: 62.56 ± 0.34 μm) within 12 days of wounding. Thus, the work highlights modified and stabilized alginate hydrogel embedded with honey and ghee blend as a potential scarless full-thickness cutaneous wound healing bio-scaffold.
Collapse
Affiliation(s)
- Ayan Gope
- Multimodal Imaging and Theranostics Laboratory, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, 721302, India.
| | - Anurup Mukhopadhyay
- Multimodal Imaging and Theranostics Laboratory, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, 721302, India.
| | - Oindrila Mukhopadhyay
- Multimodal Imaging and Theranostics Laboratory, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, 721302, India
| | - Jyotirmoy Chatterjee
- Multimodal Imaging and Theranostics Laboratory, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, 721302, India
| |
Collapse
|
12
|
Time-Series Clustering of lncRNA-mRNA Expression during the Adipogenic Transdifferentiation of Porcine Skeletal Muscle Satellite Cells. Curr Issues Mol Biol 2022; 44:2038-2053. [PMID: 35678667 PMCID: PMC9164044 DOI: 10.3390/cimb44050138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 11/16/2022] Open
Abstract
Skeletal muscle satellite cells (SMSCs), which are multifunctional muscle-derived stem cells, can differentiate into adipocytes. Long-chain non-coding RNA (lncRNA) has diverse biological functions, including the regulation of gene expression, chromosome silencing, and nuclear transport. However, the regulatory roles and mechanism of lncRNA during adipogenic transdifferentiation in muscle cells have not been thoroughly investigated. Here, porcine SMSCs were isolated, cultured, and induced for adipogenic differentiation. The expressions of lncRNA and mRNA at different time points during transdifferentiation were analysed using RNA-seq analysis. In total, 1005 lncRNAs and 7671 mRNAs showed significant changes in expression at differential differentiation stages. Time-series expression analysis showed that the differentially expressed (DE) lncRNAs and mRNAs were clustered into 5 and 11 different profiles with different changes, respectively. GO, KEGG, and REACTOME enrichment analyses revealed that DE mRNAs with increased expressions during the trans-differentiation were mainly enriched in the pathways for lipid metabolism and fat cell differentiation. The genes with decreased expressions were mainly enriched in the regulation of cell cycle and genetic information processing. In addition, 1883 DE mRNAs were regulated by 193 DE lncRNAs, and these genes were related to the controlling in cell cycle mainly. Notably, three genes in the fatty acid binding protein (FABP) family significantly and continuously increased during trans-differentiation, and 15, 13, and 11 lncRNAs may target FABP3, FABP4, and FABP5 genes by cis- or trans-regulation, respectively. In conclusion, these studies identify a set of new potential regulator for adipogenesis and cell fate and help us in better understanding the molecular mechanisms of trans-differentiation.
Collapse
|
13
|
Dalle S, Dupont J, Dedeyne L, Verschueren S, Tournoy J, Gielen E, Koppo K. Preliminary evidence of differential expression of myogenic and stress factors in skeletal muscle of older adults with low muscle strength. J Gerontol A Biol Sci Med Sci 2022; 77:1121-1129. [PMID: 34984449 DOI: 10.1093/gerona/glac002] [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: 08/30/2021] [Indexed: 11/12/2022] Open
Abstract
The age-related loss of muscle strength and mass, or sarcopenia, is a growing concern in the ageing population. Yet, it is not fully understood which molecular mechanisms underlie sarcopenia. Therefore, the present study compared the protein expression profile, such as catabolic, oxidative, stress-related and myogenic pathways, between older adults with preserved (8 ♀ and 5 ♂; 71.5 ±2.6 years) and low muscle strength (6 ♀ and 5 ♂; 78.0±5.0 years). Low muscle strength was defined as chair stand test time >15 seconds and/or handgrip strength <16kg (women) or <27kg (men) according the EWGSOP2 criteria. Catabolic signaling (i.e. FOXO1/3a, MuRF1, MAFbx, LC3b, Atg5, p62) was not differentially expressed between both groups, whereas the mitochondrial marker COX-IV, but not PGC1α and citrate synthase, was lower in the low muscle strength group. Stress factors CHOP and p-ERK1/2 were higher (~1.5-fold) in older adults with low muscle strength. Surprisingly, the inflammatory marker p-p65NF-κB was ~7-fold higher in older adults with preserved muscle strength. Finally, expression of myogenic factors (i.e. Pax7, MyoD, desmin; ~2-fold) was higher in adults with low muscle strength. To conclude, whereas the increased stress factors might reflect the age-related deterioration of tissue homeostasis, e.g. due to misfolded proteins (CHOP), upregulation of myogenic markers in the low strength group might be an attempt to compensate for the gradual loss in muscle quantity and quality. These data might provide valuable insights in the processes that underlie sarcopenia.
Collapse
Affiliation(s)
- Sebastiaan Dalle
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Belgium
| | - Jolan Dupont
- Geriatrics & Gerontology, Department of Public Health and Primary Care, KU Leuven, Belgium.,Department of Geriatric Medicine, UZ Leuven, Belgium
| | - Lenore Dedeyne
- Geriatrics & Gerontology, Department of Public Health and Primary Care, KU Leuven, Belgium
| | - Sabine Verschueren
- Research Group for Musculoskeletal Rehabilitation, Department of Movement Sciences, KU Leuven, Belgium
| | - Jos Tournoy
- Geriatrics & Gerontology, Department of Public Health and Primary Care, KU Leuven, Belgium.,Department of Geriatric Medicine, UZ Leuven, Belgium
| | - Evelien Gielen
- Geriatrics & Gerontology, Department of Public Health and Primary Care, KU Leuven, Belgium.,Department of Geriatric Medicine, UZ Leuven, Belgium
| | - Katrien Koppo
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Belgium
| |
Collapse
|
14
|
Rauen M, Hao D, Müller A, Mückter E, Bollheimer LC, Nourbakhsh M. Free Fatty Acid Species Differentially Modulate the Inflammatory Gene Response in Primary Human Skeletal Myoblasts. BIOLOGY 2021; 10:biology10121318. [PMID: 34943232 PMCID: PMC8698660 DOI: 10.3390/biology10121318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 12/21/2022]
Abstract
Simple Summary Epidemiological studies show that obesity increases the risk of muscle mass loss with age, a syndrome called sarcopenic obesity. Obesity leads to increased free fatty acids (FFAs) and excessive fat deposits, which impair the integrity of skeletal muscles by unknown mechanisms. This report indicates that FFAs directly affect human skeletal muscle cell replication and inflammatory gene expression. The structural characteristics of FFAs play a decisive role in triggering both processes. Thus, the characterization of abundant FFA species in the skeletal muscle of obese individuals may become a useful tool to predict the progression of sarcopenic obesity. Abstract Age-related loss of skeletal muscle is associated with obesity and inflammation. In animal models, intramuscular fat deposits compromise muscle integrity; however, the relevant fat components that mediate muscular inflammation are not known. Previously, we hypothesized that free fatty acids (FFAs) may directly induce inflammatory gene expression in skeletal muscle cells of obese rats. Here, we examined this hypothesis in primary human skeletal myoblasts (SkMs) using multiplex expression analysis of 39 inflammatory proteins in response to different FFA species. Multiplex mRNA quantification confirmed that the IL6, IL1RA, IL4, LIF, CXCL8, CXCL1, CXCL12 and CCL2 genes were differentially regulated by saturated and unsaturated C16 or C18 FFAs. Fluorescence staining revealed that only saturated C16 and C18 strongly interfere with myoblast replication independent of desmin expression, mitochondrial abundance and oxidative activity. Furthermore, we addressed the possible implications of 71 human receptor tyrosine kinases (RTKs) in FFA-mediated effects. Phosphorylated EphB6 and TNK2 were associated with impaired myoblast replication by saturated C16 and C18 FFAs. Our data suggest that abundant FFA species in human skeletal muscle tissue may play a decisive role in the progression of sarcopenic obesity by affecting inflammatory signals or myoblast replication.
Collapse
|
15
|
Ghzaiel I, Zarrouk A, Nury T, Libergoli M, Florio F, Hammouda S, Ménétrier F, Avoscan L, Yammine A, Samadi M, Latruffe N, Biressi S, Levy D, Bydlowski SP, Hammami S, Vejux A, Hammami M, Lizard G. Antioxidant Properties and Cytoprotective Effect of Pistacia lentiscus L. Seed Oil against 7β-Hydroxycholesterol-Induced Toxicity in C2C12 Myoblasts: Reduction in Oxidative Stress, Mitochondrial and Peroxisomal Dysfunctions and Attenuation of Cell Death. Antioxidants (Basel) 2021; 10:antiox10111772. [PMID: 34829643 PMCID: PMC8615043 DOI: 10.3390/antiox10111772] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/28/2021] [Accepted: 11/03/2021] [Indexed: 01/18/2023] Open
Abstract
Aging is characterized by a progressive increase in oxidative stress, which favors lipid peroxidation and the formation of cholesterol oxide derivatives, including 7β-hydroxycholesterol (7β-OHC). This oxysterol, which is known to trigger oxidative stress, inflammation, and cell death, could contribute to the aging process and age-related diseases, such as sarcopenia. Identifying molecules or mixtures of molecules preventing the toxicity of 7β-OHC is therefore an important issue. This study consists of determining the chemical composition of Tunisian Pistacia lentiscus L. seed oil (PLSO) used in the Tunisian diet and evaluating its ability to counteract the cytotoxic effects induced by 7β-OHC in murine C2C12 myoblasts. The effects of 7β-OHC (50 µM; 24 h), associated or not with PLSO, were studied on cell viability, oxidative stress, and on mitochondrial and peroxisomal damages induction. α-Tocopherol (400 µM) was used as the positive control for cytoprotection. Our data show that PLSO is rich in bioactive compounds; it contains polyunsaturated fatty acids, and several nutrients with antioxidant properties: phytosterols, α-tocopherol, carotenoids, flavonoids, and phenolic compounds. When associated with PLSO (100 µg/mL), the 7β-OHC-induced cytotoxic effects were strongly attenuated. The cytoprotection was in the range of those observed with α-tocopherol. This cytoprotective effect was characterized by prevention of cell death and organelle dysfunction (restoration of cell adhesion, cell viability, and plasma membrane integrity; prevention of mitochondrial and peroxisomal damage) and attenuation of oxidative stress (reduction in reactive oxygen species overproduction in whole cells and at the mitochondrial level; decrease in lipid and protein oxidation products formation; and normalization of antioxidant enzyme activities: glutathione peroxidase (GPx) and superoxide dismutase (SOD)). These results provide evidence that PLSO has similar antioxidant properties than α-tocopherol used at high concentration and contains a mixture of molecules capable to attenuate 7β-OHC-induced cytotoxic effects in C2C12 myoblasts. These data reinforce the interest in edible oils associated with the Mediterranean diet, such as PLSO, in the prevention of age-related diseases, such as sarcopenia.
Collapse
Affiliation(s)
- Imen Ghzaiel
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France; (I.G.); (T.N.); (A.Y.); (N.L.); (A.V.)
- Lab-NAFS ‘Nutrition—Functional Food & Vascular Health’, Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia; (S.H.); (S.H.); (M.H.)
- Faculty of Sciences of Tunis, University Tunis-El Manar, Tunis 2092, Tunisia
| | - Amira Zarrouk
- Lab-NAFS ‘Nutrition—Functional Food & Vascular Health’, Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia; (S.H.); (S.H.); (M.H.)
- Faculty of Medicine, University of Sousse, Sousse 4000, Tunisia
- Correspondence: (A.Z.); (G.L.); Tel.: +216-94-837-999 or +1-212-241 9304 (A.Z.); +33-380-396-256 (G.L.)
| | - Thomas Nury
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France; (I.G.); (T.N.); (A.Y.); (N.L.); (A.V.)
| | - Michela Libergoli
- Department of Cellular, Computational and Integrative Biology (CIBio) and Dulbecco Telethon Institute, University of Trento, 38123 Trento, Italy; (M.L.); (F.F.); (S.B.)
| | - Francesca Florio
- Department of Cellular, Computational and Integrative Biology (CIBio) and Dulbecco Telethon Institute, University of Trento, 38123 Trento, Italy; (M.L.); (F.F.); (S.B.)
| | - Souha Hammouda
- Lab-NAFS ‘Nutrition—Functional Food & Vascular Health’, Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia; (S.H.); (S.H.); (M.H.)
| | - Franck Ménétrier
- Centre des Sciences du Goût et de l’Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, 21065 Dijon, France;
| | - Laure Avoscan
- Agroécologie, AgroSup Dijon, CNRS, INRAE, University Bourgogne Franche-Comté, Plateforme DimaCell, 21000 Dijon, France;
| | - Aline Yammine
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France; (I.G.); (T.N.); (A.Y.); (N.L.); (A.V.)
| | - Mohammad Samadi
- LCPMC-A2, ICPM, Department of Chemistry, University Lorraine, Metz Technopôle, 57070 Metz, France;
| | - Norbert Latruffe
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France; (I.G.); (T.N.); (A.Y.); (N.L.); (A.V.)
| | - Stefano Biressi
- Department of Cellular, Computational and Integrative Biology (CIBio) and Dulbecco Telethon Institute, University of Trento, 38123 Trento, Italy; (M.L.); (F.F.); (S.B.)
| | - Débora Levy
- Lipids, Oxidation and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-900, Brazil; (D.L.); (S.P.B.)
| | - Sérgio Paulo Bydlowski
- Lipids, Oxidation and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-900, Brazil; (D.L.); (S.P.B.)
- National Institute of Science and Technology in Regenerative Medicine (INCT-Regenera), CNPq, Rio de Janeiro 21941-902, Brazil
| | - Sonia Hammami
- Lab-NAFS ‘Nutrition—Functional Food & Vascular Health’, Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia; (S.H.); (S.H.); (M.H.)
| | - Anne Vejux
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France; (I.G.); (T.N.); (A.Y.); (N.L.); (A.V.)
| | - Mohamed Hammami
- Lab-NAFS ‘Nutrition—Functional Food & Vascular Health’, Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia; (S.H.); (S.H.); (M.H.)
| | - Gérard Lizard
- Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France; (I.G.); (T.N.); (A.Y.); (N.L.); (A.V.)
- Correspondence: (A.Z.); (G.L.); Tel.: +216-94-837-999 or +1-212-241 9304 (A.Z.); +33-380-396-256 (G.L.)
| |
Collapse
|
16
|
Silva NC, Alvarez AM, DeOcesano-Pereira C, Fortes-Dias CL, Moreira V. Catalytically active phospholipase A 2 myotoxin from Crotalus durissus terrificus induces proliferation and differentiation of myoblasts dependent on prostaglandins produced by both COX-1 and COX-2 pathways. Int J Biol Macromol 2021; 187:603-613. [PMID: 34314795 DOI: 10.1016/j.ijbiomac.2021.07.121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 01/18/2023]
Abstract
Although crotoxin B (CB) is a well-established catalytically active secretory phospholipase A2 group IIA (sPLA2-IIA) myotoxin, we investigated its potential stimulatory effect on myogenesis with the involvement of prostaglandins (PGs) produced by cyclooxygenase (COX)-1 and -2 pathways. Myoblast C2C12 were cultured in proliferation or commitment protocols and incubated with CB followed by lumiracoxib (selective COX-2 inhibitor) or valeryl salicylate (selective COX-1 inhibitor) and subjected to analysis of PG release, cell proliferation and activation of myogenic regulatory factors (MRFs). Our data showed that CB in non-cytotoxic concentrations induces an increase of COX-2 protein expression and stimulates the activity of both COX isoforms to produce PGE2, PGD2 and 15d-PGJ2. CB induced an increase in the proliferation of C2C12 myoblast cells dependent on PGs from both COX-1 and COX-2 pathways. In addition, CB stimulated the activity of Pax7, MyoD, Myf5 and myogenin in proliferated cells. Otherwise, CB increased myogenin activity but not MyoD in committed cells. Our findings evidence the role of COX-1- and COX-2-derived PGs in modulating CB-induced activation of MRFs. This study contributes to the knowledge that CB promote early myogenic events via regulatory mechanisms on PG-dependent COX pathways, showing new concepts about the effect of sPLA2-IIA in skeletal muscle repair.
Collapse
Affiliation(s)
- Nadine C Silva
- Pharmacology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP 04044-020, Brazil
| | - Angela M Alvarez
- Pharmacology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP 04044-020, Brazil; Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo, SP 05503-900, Brazil.
| | - Carlos DeOcesano-Pereira
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo, SP 05503-900, Brazil.
| | | | - Vanessa Moreira
- Pharmacology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP 04044-020, Brazil.
| |
Collapse
|
17
|
Monnard CR, Dulloo AG. Polyunsaturated fatty acids as modulators of fat mass and lean mass in human body composition regulation and cardiometabolic health. Obes Rev 2021; 22 Suppl 2:e13197. [PMID: 33471425 DOI: 10.1111/obr.13197] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/22/2022]
Abstract
It is now recognized that the amount and type of dietary fat consumed play an important role in metabolic health. In humans, high intake of polyunsaturated fatty acids (PUFAs) has been associated with reductions in cardiovascular disease risk, improvements in glucose homeostasis, and changes in body composition that involve reductions in central adiposity and, more recently, increases in lean body mass. There is also emerging evidence, which suggests that high intakes of the plant-based essential fatty acids (ePUFAs)-n-6 linoleic acid (LA) and n-3 α-linolenic acid (ALA)-have a greater impact on body composition (fat mass and lean mass) and on glucose homeostasis than the marine-derived long-chain n-3 PUFA-eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). In addition, high intake of both ePUFAs (LA and ALA) may also have anti-inflammatory effects in humans. The purpose of this review is to highlight the emerging evidence, from both epidemiological prospective studies and clinical intervention trials, of a role for PUFA, in particular ePUFA, in the long-term regulation of body weight and body composition, and their impact on cardiometabolic health. It also discusses current notions about the mechanisms by which PUFAs modulate fat mass and lean mass through altered control of energy intake, thermogenesis, or lean-fat partitioning.
Collapse
Affiliation(s)
- Cathriona R Monnard
- Faculty of Science and Medicine, Department of Endocrinology, Metabolism and Cardiovascular System, University of Fribourg, Fribourg, Switzerland
| | - Abdul G Dulloo
- Faculty of Science and Medicine, Department of Endocrinology, Metabolism and Cardiovascular System, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
18
|
PUFA Treatment Affects C2C12 Myocyte Differentiation, Myogenesis Related Genes and Energy Metabolism. Genes (Basel) 2021; 12:genes12020192. [PMID: 33525599 PMCID: PMC7910949 DOI: 10.3390/genes12020192] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/19/2021] [Accepted: 01/26/2021] [Indexed: 11/16/2022] Open
Abstract
Polyunsaturated fatty acids (PUFAs) are the main components of cell membrane affecting its fluidity, signaling processes and play a vital role in muscle cell development. The effects of docosahexaenoic acid (DHA) on myogenesis are well known, while the effects of arachidonic acid (AA) are largely unclear. The purpose of this study is to evaluate the effect of two PUFAs (DHA and AA) on cell fate during myogenic processes, Wnt signaling and energy metabolism by using the C2C12 cells. The cells were treated with different concentrations of AA or DHA for 48 h during the differentiation period. PUFA treatment increased mRNA level of myogenic factor 5 (Myf5), which is involved in early stage of myoblast proliferation. Additionally, PUFA treatment prevented myoblast differentiation, indicated by decreased myotube fusion index and differentiation index in parallel with reduced mRNA levels of myogenin (MyoG). After PUFA withdrawal, some changes in cell morphology and myosin heavy chain mRNA levels were still observed. Expression of genes associated with Wnt signaling pathway, and energy metabolism changed in PUFA treatment in a dose and time dependent manner. Our data suggests that PUFAs affect the transition of C2C12 cells from proliferation to differentiation phase by prolonging proliferation and preventing differentiation.
Collapse
|
19
|
Phycocyanin from Arthrospira platensis as Potential Anti-Cancer Drug: Review of In Vitro and In Vivo Studies. Life (Basel) 2021; 11:life11020091. [PMID: 33513794 PMCID: PMC7911896 DOI: 10.3390/life11020091] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 12/18/2022] Open
Abstract
The application of cytostatic drugs or natural substances to inhibit cancer growth and progression is an important and evolving subject of cancer research. There has been a surge of interest in marine bioresources, particularly algae, as well as cyanobacteria and their bioactive ingredients. Dried biomass products of Arthrospira and Chlorella have been categorized as “generally recognized as safe” (GRAS) by the US Food and Drug Administration (FDA). Of particular importance is an ingredient of Arthrospira: phycocyanin, a blue-red fluorescent, water-soluble and non-toxic biliprotein pigment. It is reported to be the main active ingredient of Arthrospira and was shown to have therapeutic properties, including anti-oxidant, anti-inflammatory, immune-modulatory and anti-cancer activities. In the present review, in vitro and in vivo data on the effects of phycocyanin on various tumor cells and on cells from healthy tissues are summarized. The existing knowledge of underlying molecular mechanisms, and strategies to improve the efficiency of potential phycocyanin-based anti-cancer therapies are discussed.
Collapse
|
20
|
Khazaeli P, Alaei M, Khaksarihadad M, Ranjbar M. Preparation of PLA/chitosan nanoscaffolds containing cod liver oil and experimental diabetic wound healing in male rats study. J Nanobiotechnology 2020; 18:176. [PMID: 33256764 PMCID: PMC7706058 DOI: 10.1186/s12951-020-00737-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/20/2020] [Indexed: 11/23/2022] Open
Abstract
Diabetes mellitus is one of the most common metabolic disorders. One of the important metabolic complications in diabetes is diabetic foot ulcer syndrome, which causes delayed and abnormal healing of the wound. The formulation of nanoscaffolds containing cod liver oil by altering the hemodynamic balance toward the vasodilators state, increasing wound blood supply, and altering plasma membrane properties, namely altering the membrane phospholipids composition, can be effective in wound healing. In this study, electrospinning method was used to produce poly lactic acid/chitosan nanoscaffolds as a suitable bio-substitute. After preparing the nanoscaffolds, the products were characterized with dynamic light scattering (DLS), transmission electron microscopy (TEM) and scanning electron microscopy (SEM). Also optical properties of polymer and comparison between adsorption between single polymer and polymer-drug calculated with UV−Vis spectra. The structure and functional groups of the final products were characterized by Fourier-transform infrared spectroscopy (FT-IR) and energy dispersive spectroscopy (EDAX) as elemental analysis. The results showed that the optimum formulation of cod liver oil was 30%, which formed a very thin fiber that rapidly absorbed to the wound and produced significant healing effects. According to the results, poly lactic acid/chitosan nanoscaffolds containing cod liver oil can be a suitable bio-product to be used in treating the diabetic foot ulcer syndrome.![]()
Collapse
Affiliation(s)
- Payam Khazaeli
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, P.O. Box: 76175-493, Kerman, 76169-11319, Iran.,Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Alaei
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksarihadad
- Neuroscience Research, and Physiology Research Centers, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehdi Ranjbar
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, P.O. Box: 76175-493, Kerman, 76169-11319, Iran.
| |
Collapse
|
21
|
Wang L, Shan T. Factors inducing transdifferentiation of myoblasts into adipocytes. J Cell Physiol 2020; 236:2276-2289. [PMID: 32989814 DOI: 10.1002/jcp.30074] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
Fat infiltration in skeletal muscle is observed in several myopathies, is associated with muscular dysfunction, and is strongly correlated with insulin resistance, diabetes, obesity, and aging. In animal production, skeletal muscle fat (also known as intermuscular and intramuscular fat) is positively related to meat quality including tenderness, flavor, and juiciness. Thus, understanding the cell origin and regulation mechanism of skeletal muscle fat infiltration is important for developing therapies against human myopathies as well as for improving meat quality. Notably, age, sarcopenia, oxidative stress, injury, and regeneration can activate adipogenic differentiation potential in myoblasts and affect fat accumulation in skeletal muscle. In addition, several transcriptional and nutritional factors can directly induce transdifferentiation of myoblasts into adipocytes. In this review, we focused on the recent progress in understanding the muscle-to-adipocyte differentiation and summarized and discussed the genetic, nutritional, and physiological factors that can induce transdifferentiation of myoblasts into adipocytes. Moreover, the regulatory roles and mechanisms of these factors during the transdifferentiation process were also discussed.
Collapse
Affiliation(s)
- Liyi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| |
Collapse
|
22
|
Mohammadi I, Mahdavi AH, Rabiee F, Nasr Esfahani MH, Ghaedi K. Positive effects of conjugated linoleic acid (CLA) on the PGC1-α expression under the inflammatory conditions induced by TNF-α in the C2C12 cell line. Gene 2020; 735:144394. [DOI: 10.1016/j.gene.2020.144394] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/17/2022]
|
23
|
Nguyen JH, Chung JD, Lynch GS, Ryall JG. The Microenvironment Is a Critical Regulator of Muscle Stem Cell Activation and Proliferation. Front Cell Dev Biol 2019; 7:254. [PMID: 31737625 PMCID: PMC6828616 DOI: 10.3389/fcell.2019.00254] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022] Open
Abstract
Skeletal muscle has a remarkable capacity to regenerate following injury, a property conferred by a resident population of muscle stem cells (MuSCs). In response to injury, MuSCs must double their cellular content to divide, a process requiring significant new biomass in the form of nucleotides, phospholipids, and amino acids. This new biomass is derived from a series of intracellular metabolic cycles and alternative routing of carbon. In this review, we examine the link between metabolism and skeletal muscle regeneration with particular emphasis on the role of the cellular microenvironment in supporting the production of new biomass and MuSC proliferation.
Collapse
Affiliation(s)
- John H Nguyen
- Department of Physiology, Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, Australia
| | - Jin D Chung
- Department of Physiology, Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, Australia
| | - Gordon S Lynch
- Department of Physiology, Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, Australia
| | - James G Ryall
- Department of Physiology, Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
24
|
Kuppusamy P, Soundharrajan I, Kim DH, Hwang I, Choi KC. 4-hydroxy-3-methoxy cinnamic acid accelerate myoblasts differentiation on C2C12 mouse skeletal muscle cells via AKT and ERK 1/2 activation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 60:152873. [PMID: 30879871 DOI: 10.1016/j.phymed.2019.152873] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/12/2019] [Accepted: 02/23/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND The dietary intake of plant-based supplements has a vital role in human health and development. However, the actions of secondary plant metabolites on cell growth, differentiation and their signaling mechanisms are still unclear. PURPOSE In this study, we aim to investigate the C2C12 myoblast cells proliferation and differentiation by 4-hydroxy-3-methoxy cinnamic acid (=HMCA, ferulic acid) in a dose-dependent manner and to reveal its underlying mechanism of action. METHODS The effect of HMCA on C2C12 cell proliferation and differentiation were evaluated by expression of BMP's marker genes (-2, -4, -6, -7) and related myogenic proteins were analyzed by quantitative PCR and western blot techniques, respectively. RESULTS The in vitro findings confirmed that the HMCA upregulates BMPs (including BMP-2, -4, -6, and-7), gene expression in C2C12 skeletal muscle cells. Exposure to the lower dose of HMCA caused a significantly greater induction of myogenic differentiation than the higher dose during three- and six-day treatments. Further, the C2C12 myogenic differentiation signaling proteins MyoD, myogenin, JAK-1, -2, -3, STAT -2, -3, AMPK-α, ERK(1/2), and AKT were more preferentially activated by HMCA exposure cells than by untreated models. Thus, the experiment with inhibitors revealed that the HMCA induced muscle cell proliferation and differentiation through AKT and ERK (1/2) signaling cascades. Also, HMCA enhanced the C2C12 muscle cell differentiation protein markers such as myogenin, AKT and ERK (1/2) significantly (p ≤ 0.05) at day three in chemical inhibitors of LY 294002 and PD98056 treated samples. CONCLUSION The HMCA has a significant effect on muscle cell differentiation through ERK(1/2) and AKT signaling activation. Also, the HMCA promotes C2C12 muscle cell proliferation and differentiation via activation of osteogenic genes and myogeneic protein markers. Therefore, this study suggests that the natural phenolic compound HMCA has a potent function in muscle cell proliferation, differentiation, and development.
Collapse
Affiliation(s)
- Palaniselvam Kuppusamy
- Grassland and Forage Division, National Institute of Animal Science, Rural Development Administration, Cheonan 330-801, Republic of Korea; Department of Animal Science, College of Agricultural and Life Science, Chonbuk National University, Jeonju 54896, Republic of Korea
| | - Ilavenil Soundharrajan
- Grassland and Forage Division, National Institute of Animal Science, Rural Development Administration, Cheonan 330-801, Republic of Korea
| | - Da Hye Kim
- Center for Research on Environmental Disease, Department of Animal Husbandry, University of Kentucky, Lexington, KY 40536, USA
| | - Inho Hwang
- Department of Animal Science, College of Agricultural and Life Science, Chonbuk National University, Jeonju 54896, Republic of Korea
| | - Ki Choon Choi
- Grassland and Forage Division, National Institute of Animal Science, Rural Development Administration, Cheonan 330-801, Republic of Korea.
| |
Collapse
|
25
|
Leng X, Jiang H. Effects of arachidonic acid and its major prostaglandin derivatives on bovine myoblast proliferation, differentiation, and fusion. Domest Anim Endocrinol 2019; 67:28-36. [PMID: 30677541 DOI: 10.1016/j.domaniend.2018.12.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 12/03/2018] [Accepted: 12/13/2018] [Indexed: 12/23/2022]
Abstract
Many studies have shown positive effects of prostaglandins (PGs) on various steps of skeletal muscle formation such as myoblast proliferation and myotube hypertrophy. In animals, PGs are synthesized through the action of the rate-limiting enzymes cyclooxygenase (COX) -1 and COX-2 from arachidonic acid (AA), a conditionally essential fatty acid. As a step toward exploring the possibility of using dietary supplementation of AA to improve skeletal muscle growth in cattle, which are major meat-producing animals, we determined the effects of AA and its major PG derivatives PGE2, PGF2α, and PGI2 on proliferation, differentiation, and fusion of primary bovine myoblasts in vitro. In the proliferation experiment, myoblasts were cultured in a growth medium to which was added 10 μM AA, 1 μM PGE2, 1 μM PGF2α, 1 μM PGI2, or vehicle control for 24 h, and the proliferating cells were identified by 5-ethynyl-2'-deoxyuridine (EdU) labeling. This experiment revealed that AA, PGE2, PGF2α, and PGI2 each increased the number of proliferating cells by 13%, 24%, 16%, and 16%, respectively, compared to the control (n = 7, P < 0.05). In the differentiation and fusion test, myoblasts were induced to differentiate and fuse into myotubes in the presence of the aforementioned treatments for 0, 24, 48, and 72 h. Based on quantitative reverse transcription PCR analyses of mRNAs of myoblast differentiation and fusion markers (myogenin; myosin heavy chain 3; creatine kinase, muscle; myomaker) at 0, 24, and 48 h of differentiation, AA, PGE2, and PGF2α promoted myoblast differentiation (n = 6, P < 0.05). Based on Giemsa staining and counting the number of myotubes at 72 h of differentiation, PGE2 enhanced the number of formed myotubes by 14% (P < 0.05) compared to the control. Treating the myoblasts with AA and either the COX-1 and COX-2 common inhibitor indomethacin or the COX-2-specific inhibitor NS-398 reversed the stimulatory effect of AA on myoblast proliferation (n = 4, P < 0.05). Overall, this study demonstrates that exogenous AA stimulates bovine myoblast proliferation and differentiation in culture. The results of this study suggest that AA stimulates myoblast proliferation through its metabolites PGE2, PGF2α, or PGI2, and that AA stimulates myoblast differentiation through PGE2.
Collapse
Affiliation(s)
- X Leng
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - H Jiang
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
26
|
Zhang J, Xu X, Liu Y, Zhang L, Odle J, Lin X, Zhu H, Wang X, Liu Y. EPA and DHA Inhibit Myogenesis and Downregulate the Expression of Muscle-related Genes in C2C12 Myoblasts. Genes (Basel) 2019; 10:genes10010064. [PMID: 30669396 PMCID: PMC6356802 DOI: 10.3390/genes10010064] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 01/11/2019] [Indexed: 12/31/2022] Open
Abstract
This study was conducted to elucidate the biological effects of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) on cell proliferation, differentiation and gene expression in C2C12 myoblasts. C2C12 were treated with various concentrations of EPA or DHA under proliferation and differentiation conditions. Cell viability was analyzed using cell counting kit-8 assays (CCK-8). The Edu assays were performed to analyze cell proliferation. To analyze cell differentiation, the expressions of myogenic marker genes were determined at the transcriptional and translational levels by qRT-PCR, immunoblotting and immunofluorescence. Global gene expression patterns were characterized using RNA-sequencing. Phosphorylation levels of ERK and Akt were examined by immunoblotting. Cell viability and proliferation was significantly inhibited after incubation with EPA (50 and 100 μM) or DHA (100 μM). Both EPA and DHA suppressed C2C12 myoblasts differentiation. RNA-sequencing analysis revealed that some muscle-related genes were significantly downregulated following EPA or DHA (50 μM) treatment, including insulin-like growth factor 2 (IGF-2), troponin T3 (Tnnt3), myoglobin (Mb), myosin light chain phosphorylatable fast skeletal muscle (Mylpf) and myosin heavy polypeptide 3 (Myh3). IGF-2 was crucial for the growth and differentiation of skeletal muscle and could activate the PI3K/Akt and the MAPK/ERK cascade. We found that EPA and DHA (50 μM) decreased the phosphorylation levels of ERK1/2 and Akt in C2C12 myoblasts. Thus, this study suggested that EPA and DHA exerted an inhibitory effect on myoblast proliferation and differentiation and downregulated muscle-related genes expression.
Collapse
Affiliation(s)
- Jing Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Xin Xu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Yan Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Lin Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Jack Odle
- Laboratory of Development Nutrition, Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA.
| | - Xi Lin
- Laboratory of Development Nutrition, Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA.
| | - Huiling Zhu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Xiuying Wang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Yulan Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China.
| |
Collapse
|
27
|
Muthuramalingam K, Kim SY, Kim Y, Kim HS, Jeon YJ, Cho M. Bigbelly seahorse (Hippocampus abdominalis)-derived peptides enhance skeletal muscle differentiation and endurance performance via activated P38MAPK/AKT signalling pathway: An in vitro and in vivo analysis. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.10.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
28
|
Martacic J, Filipovic MK, Borozan S, Cvetkovic Z, Popovic T, Arsic A, Takic M, Vucic V, Glibetic M. N-acetyl-L-cysteine protects dental tissue stem cells against oxidative stress in vitro. Clin Oral Investig 2018; 22:2897-2903. [PMID: 29450735 DOI: 10.1007/s00784-018-2377-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 02/01/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVES The aim of our study was to investigate whether N-acetyl-L-cysteine (NAC) could protect stem cells from exfoliated deciduous teeth (SHED) against oxidative damage, during in vitro cultivation, to preserve regenerative potential of these cells. Accordingly, we examined the potential of cell culture supplementation with NAC in prevention of lipid peroxidation, unfavorable changes of total lipids fatty acid composition, and the effects on the activity of antioxidant enzymes. MATERIAL AND METHODS We analyzed the extent of oxidative damage in SHED after 48 h treatment with different NAC concentrations. Cellular lipid peroxidation was determined upon reaction with thiobarbituric acid. All enzyme activities were measured spectrophotometrically, based on published methods. Fatty acid methyl esters were analyzed by gas-liquid chromatography. RESULTS Concentration of 0.1 mM NAC showed the most profound effects on SHED, significantly decreasing levels of lipid peroxidation in comparison to control. This dose also diminished the activities of antioxidant enzymes. Furthermore, NAC treatment significantly changed fatty acid composition of cells, reducing levels of oleic acid and monounsaturated fatty acids and increasing linoleic acid, n-6, and total polyunsaturated fatty acid (PUFA) proportions. CONCLUSION Low dose of NAC significantly decreased lipid peroxidation and altered fatty acid composition towards increasing PUFA. The reduced oxidative damage of cellular lipids could be strongly related to improved SHED survival in vitro. CLINICAL RELEVANCE Low doses of antioxidants, applied during stem cells culturing and maintenance, could improve cellular characteristics in vitro. This is prerequisite for successful use of stem cells in various clinical applications.
Collapse
Affiliation(s)
- Jasmina Martacic
- Institute for Medical Research, University of Belgrade, Dr Subotica 4, Belgrade, 11000, Serbia
| | - Milica Kovacevic Filipovic
- Faculty of Veterinary Medicine, University of Belgrade, Bulevar oslobodjenja 18, Belgrade, 11000, Serbia
| | - Suncica Borozan
- Faculty of Veterinary Medicine, University of Belgrade, Bulevar oslobodjenja 18, Belgrade, 11000, Serbia
| | - Zorica Cvetkovic
- Department of Hematology, Clinical Hospital Center Zemun, Vukova 9, Belgrade, 11080, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, Belgrade, 11000, Serbia
| | - Tamara Popovic
- Institute for Medical Research, University of Belgrade, Dr Subotica 4, Belgrade, 11000, Serbia
| | - Aleksandra Arsic
- Institute for Medical Research, University of Belgrade, Dr Subotica 4, Belgrade, 11000, Serbia
| | - Marija Takic
- Institute for Medical Research, University of Belgrade, Dr Subotica 4, Belgrade, 11000, Serbia
| | - Vesna Vucic
- Institute for Medical Research, University of Belgrade, Dr Subotica 4, Belgrade, 11000, Serbia.
| | - Maria Glibetic
- Institute for Medical Research, University of Belgrade, Dr Subotica 4, Belgrade, 11000, Serbia
| |
Collapse
|
29
|
Belal SA, Sivakumar AS, Kang DR, Cho S, Choe HS, Shim KS. Modulatory effect of linoleic and oleic acid on cell proliferation and lipid metabolism gene expressions in primary bovine satellite cells. Anim Cells Syst (Seoul) 2018; 22:324-333. [PMID: 30460114 PMCID: PMC6171434 DOI: 10.1080/19768354.2018.1517824] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 08/16/2018] [Accepted: 08/19/2018] [Indexed: 12/16/2022] Open
Abstract
This study was performed to elucidate the effects of linoleic acid (LA), oleic acid (OA) and their combination (LA + OA) on cell proliferation, apoptosis, necrosis, and the lipid metabolism related gene expression in bovine satellite cells (BSCs), isolated from bovine muscles. Cell viability was significantly increased with the OA and LA treatment. Furthermore, LA + OA enhanced cell proliferation in a dose-dependent manner (10 to 100 µM), whereas it lowered at 250 µM. In addition, a cell-cycle analysis showed that 100 µM of LA and OA markedly decreased the G0/G1 phase proportion (62.58% and 61.33%, respectively), compared to controls (68.02%), whereas the S-phase cells' proportion was increased. The ratio of G2/M phase cells was not significantly different among the groups. Moreover, analyses with AO/EtBr staining showed that no apoptosis occurred. Necrosis were determined by flow cytometry using Annexin V-FITC/PI staining which revealed no early apoptosis in the cells pretreated with LA or OA, but occurred in the LA + OA group. We also analyzed the mRNA expression of lipid metabolizing genes such as peroxisome proliferator receptor alfa (PPARα), peroxisome proliferator receptor gamma (PPARγ), acyl-CoA oxidase (ACOX), lipoprotein lipase (LPL), carnitine palmitoyl transferase (CPT-1), and fatty-acid binding protein4 (FABP4), which were upregulated in LA or OA treated cells compared to the control group. In essence, LA and OA alone promote the cell proliferation without any apoptosis and necrosis, which might upregulate the lipid metabolism related gene expressions, and increase fatty-acid oxidation in the BSCs' lipid metabolism.
Collapse
Affiliation(s)
- Shah Ahmed Belal
- Department of Animal Biotechnology, Chonbuk National University, Jeonju-si, Republic of Korea.,Department of Poultry Science, Sylhet Agricultural University, Sylhet, Bangladesh
| | | | - Da Rae Kang
- Department of Animal Biotechnology, Chonbuk National University, Jeonju-si, Republic of Korea
| | - Sangbuem Cho
- Department of Animal Science, Chonbuk National University, Jeonju-si, Republic of Korea
| | - Ho Sung Choe
- Department of Animal Biotechnology, Chonbuk National University, Jeonju-si, Republic of Korea
| | - Kwan Seob Shim
- Department of Animal Biotechnology, Chonbuk National University, Jeonju-si, Republic of Korea
| |
Collapse
|
30
|
Lee H, Lim JY, Choi SJ. Role of l-carnitine and oleate in myogenic differentiation: implications for myofiber regeneration. J Exerc Nutrition Biochem 2018; 22:36-42. [PMID: 30149425 PMCID: PMC6058066 DOI: 10.20463/jenb.2018.0015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 06/18/2018] [Indexed: 01/07/2023] Open
Abstract
[Purpose] Myogenic progenitors play a critical role in injury-induced myofiber regeneration. The purpose of this study was to characterize the effects of oleate and L-carnitine on the overall behavior of proliferating myogenic progenitors (myoblasts) and its link to the mitochondrial biogenic process. [Methods] C2C12 myoblasts were cultured either with no treatment, oleate, L-carnitine, or their mixture. Proliferating myoblasts were investigated under a phase-contrast microscope. Myonuclei and myosin heavy chain were stained with DAPI and MF20 antibody, respectively, in differentiated myotubes and visualized under florescence microscopy. Mitochondrial biogenic markers and porin were assessed by qRT-PCR or immunoblotting. [Results] Increased proliferation rate was observed in myoblasts conditioned with oleate or a mixture of oleate and L-carnitine in contrast to that in non-treated (NT) and L-carnitine-treated myoblasts. Myoblast viability was not statistically different among all tested groups. Fusion index and myotube width were greater in oleate- or L-carnitine-conditioned myotubes than those in NT myotubes, with the greatest effect seen in myotubes conditioned with the mixture. The gene expressions of Pgc1-α, Nrf1, and Tfam were the greatest in myotubes conditioned with the mixture, whereas the level of Ncor1 expression was lower compared to those of the other groups. Protein level of porin was the greatest in myotubes conditioned with the mixture, followed by that of individually treated myotubes with oleate and L-carnitine. [Conclusion] These results provide a critical piece of cellular evidence that combined treatment of oleate and L-carnitine could serve as a potential therapeutic strategy to facilitate biological activation of myogenic progenitors.
Collapse
|
31
|
Wang L, Luo L, Zhao W, Yang K, Shu G, Wang S, Gao P, Zhu X, Xi Q, Zhang Y, Jiang Q, Wang L. Lauric Acid Accelerates Glycolytic Muscle Fiber Formation through TLR4 Signaling. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:6308-6316. [PMID: 29877088 DOI: 10.1021/acs.jafc.8b01753] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Lauric acid (LA), which is the primary fatty acid in coconut oil, was reported to have many metabolic benefits. TLR4 is a common receptor of lipopolysaccharides and involved mainly in inflammation responses. Here, we focused on the effects of LA on skeletal muscle fiber types and metabolism. We found that 200 μM LA treatment in C2C12 or dietary supplementation of 1% LA increased MHCIIb protein expression and the proportion of type IIb muscle fibers from 0.452 ± 0.0165 to 0.572 ± 0.0153, increasing the mRNA expression of genes involved in glycolysis, such as HK2 and LDH2 (from 1.00 ± 0.110 to 1.35 ± 0.0843 and from 1.00 ± 0.123 to 1.71 ± 0.302 in vivo, respectively), decreasing the catalytic activity of lactate dehydrogenase (LDH), and transforming lactic acid to pyruvic acid. Furthermore, LA activated TLR4 signaling, and TLR4 knockdown reversed the effect of LA on muscle fiber type and glycolysis. Thus, we inferred that LA promoted glycolytic fiber formation through TLR4 signaling.
Collapse
|
32
|
Mitchell CJ, D'Souza RF, Figueiredo VC, Chan A, Aasen K, Durainayagam B, Mitchell S, Sinclair AJ, Egner IM, Raastad T, Cameron-Smith D, Markworth JF. Effect of dietary arachidonic acid supplementation on acute muscle adaptive responses to resistance exercise in trained men: a randomized controlled trial. J Appl Physiol (1985) 2018; 124:1080-1091. [DOI: 10.1152/japplphysiol.01100.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Arachidonic acid (ARA), a polyunsaturated ω-6 fatty acid, acts as precursor to a number of prostaglandins with potential roles in muscle anabolism. It was hypothesized that ARA supplementation might enhance the early anabolic response to resistance exercise (RE) by increasing muscle protein synthesis (MPS) via mammalian target of rapamycin (mTOR) pathway activation and/or the late anabolic response by modulating ribosome biogenesis and satellite cell expansion. Nineteen men with ≥1 yr of resistance-training experience were randomized to consume either 1.5 g daily ARA or a corn-soy-oil placebo in a double-blind manner for 4 wk. Participants then undertook fasted RE (8 sets each of leg press and extension at 80% 1-repetition maximum), with vastus lateralis biopsies obtained before exercise, immediately postexercise, and at 2, 4, and 48 h of recovery. MPS (measured via stable isotope infusion) was not different between groups ( P = 0.212) over the 4-h recovery period. mTOR pathway members p70 S6 kinase and S6 ribosomal protein were phosphorylated postexercise ( P < 0.05), with no difference between groups. 45S preribosomal RNA increased 48 h after exercise only in ARA ( P = 0.012). Neural cell adhesion molecule-positive satellite cells per fiber increased 48 h after exercise ( P = 0.013), with no difference between groups ( P = 0.331). Prior ARA supplementation did not alter the acute anabolic response to RE in previously resistance-trained men; however, at 48 h of recovery, ribosome biogenesis was stimulated only in the ARA group. The findings do not support a mechanistic link between ARA and short-term anabolism, but ARA supplementation in conjunction with resistance training may stimulate increases in translational capacity. NEW & NOTEWORTHY Four weeks of daily arachidonic acid supplementation in trained men did not alter their acute muscle protein synthetic or anabolic signaling response to resistance exercise. However, 48 h after exercise, men supplemented with arachidonic acid showed greater ribosome biogenesis and a trend toward greater change in satellite cell content. Chronic arachidonic acid supplementation does not appear to regulate the acute anabolic response to resistance exercise but may augment muscle adaptation in the following days of recovery.
Collapse
Affiliation(s)
| | | | - Vandre C. Figueiredo
- Liggins Institute, University of Auckland, Auckland, New Zealand
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | - Alex Chan
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Kirsten Aasen
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | - Sarah Mitchell
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | | | - Truls Raastad
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - David Cameron-Smith
- Liggins Institute, University of Auckland, Auckland, New Zealand
- Food & Bio-based Products Group, AgResearch, Palmerston North, New Zealand
- Riddet Institute, Palmerston North, New Zealand
| | - James F. Markworth
- Liggins Institute, University of Auckland, Auckland, New Zealand
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
33
|
Potential Roles of n-3 PUFAs during Skeletal Muscle Growth and Regeneration. Nutrients 2018; 10:nu10030309. [PMID: 29510597 PMCID: PMC5872727 DOI: 10.3390/nu10030309] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 03/01/2018] [Accepted: 03/02/2018] [Indexed: 01/06/2023] Open
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFAs), which are commonly found in fish oil supplements, are known to possess anti-inflammatory properties and more recently alter skeletal muscle function. In this review, we discuss novel findings related to how n-3 PUFAs modulate molecular signaling responsible for growth and hypertrophy as well as the activity of muscle stem cells. Muscle stem cells commonly known as satellite cells, are primarily responsible for driving the skeletal muscle repair process to potentially damaging stimuli, such as mechanical stress elicited by exercise contraction. To date, there is a paucity of human investigations related to the effects of n-3 PUFAs on satellite cell content and activity. Based on current in vitro investigations, this review focuses on novel mechanisms linking n-3 PUFA’s to satellite cell activity and how they may improve muscle repair. Understanding the role of n-3 PUFAs during muscle growth and regeneration in association with exercise could lead to the development of novel supplementation strategies that increase muscle mass and strength, therefore possibly reducing the burden of muscle wasting with age.
Collapse
|
34
|
Fatty acids promote bovine skeletal muscle satellite cell differentiation by regulating ELOVL3 expression. Cell Tissue Res 2018; 373:499-508. [DOI: 10.1007/s00441-018-2812-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 01/25/2018] [Indexed: 10/18/2022]
|
35
|
Markworth JF, Mitchell CJ, D'Souza RF, Aasen KMM, Durainayagam BR, Mitchell SM, Chan AHC, Sinclair AJ, Garg M, Cameron-Smith D. Arachidonic acid supplementation modulates blood and skeletal muscle lipid profile with no effect on basal inflammation in resistance exercise trained men. Prostaglandins Leukot Essent Fatty Acids 2018; 128:74-86. [PMID: 29413364 DOI: 10.1016/j.plefa.2017.12.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 11/02/2017] [Accepted: 12/07/2017] [Indexed: 12/12/2022]
Abstract
Arachidonic acid (ARA), an omega-6 polyunsaturated fatty acid (PUFA), is the metabolic precursor to the eicosanoid family of lipid mediators. Eicosanoids have potent pro-inflammatory actions, but also act as important autocrine/paracrine signaling molecules in skeletal muscle growth and development. Whether dietary ARA is incorporated into skeletal muscle phospholipids and the resulting impact on intramuscular inflammatory and adaptive processes in-vivo is not known. In the current study, resistance trained men (≥1 year) received dietary supplementation with 1.5g/day ARA (n=9, 24 ± 1.5 years) or placebo (n=10, 26 ± 1.3 years) for 4-weeks while continuing their normal training regimen. Plasma and vastus lateralis muscle biopsies were collected in an overnight fasted state at baseline and week 4. ARA supplementation increased plasma content of ARA and gamma-linolenic acid, while decreasing relative abundance of linoleic acid, eicosapentaenoic acid, and dihomo-gamma-linolenic acid. In skeletal muscle, ARA and dihomo-gamma-linolenic acid content increased, whereas alpha-linolenic-acid was reduced. Compared to placebo, ARA supplementation reduced circulating platelet and monocyte number, and decreased the mRNA expression of the immune cell surface markers; neutrophil elastase/CD66b and interleukin 1-beta, in peripheral blood mononuclear cells. In muscle, ARA supplementation increased mRNA expression of the myogenic regulatory factors; MyoD and myogenin, but had no effect on a range of immune cell markers or inflammatory cytokines. These data show that dietary ARA supplementation can rapidly and safely modulate plasma and muscle fatty acid profile and promote myogenic gene expression in resistance trained men, without a risk of increasing basal systemic or intramuscular inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Alex H C Chan
- Liggins Institute, University of Auckland, New Zealand
| | | | - Manohar Garg
- Nutraceuticals Research Program, School of Biomedical Sciences & Pharmacy, University of Newcastle, Australia
| | - David Cameron-Smith
- Liggins Institute, University of Auckland, New Zealand; Food & Bio-based Products Group, AgResearch, Palmerston North, New Zealand; Riddet Institute, Palmerston North, New Zealand.
| |
Collapse
|
36
|
Terruzzi I, Vacante F, Senesi P, Montesano A, Codella R, Luzi L. Effect of Hazelnut Oil on Muscle Cell Signalling and Differentiation. J Oleo Sci 2018; 67:1315-1326. [DOI: 10.5650/jos.ess18086] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Ileana Terruzzi
- Diabetes Research Institute, Metabolism, Nutrigenomics and Cellular Differentiation Unit, San Raffaele Scientific Institute
| | | | - Pamela Senesi
- Metabolism Research Center, IRCCS Policlinico San Donato
| | - Anna Montesano
- Department of Biomedical Sciences for Health, Università degli Studi di Milano
| | - Roberto Codella
- Metabolism Research Center, IRCCS Policlinico San Donato
- Department of Biomedical Sciences for Health, Università degli Studi di Milano
| | - Livio Luzi
- Metabolism Research Center, IRCCS Policlinico San Donato
- Department of Biomedical Sciences for Health, Università degli Studi di Milano
| |
Collapse
|
37
|
Abreu P, Leal-Cardoso JH, Ceccatto VM, Hirabara SM. Regulation of muscle plasticity and trophism by fatty acids: A short review. Rev Assoc Med Bras (1992) 2017; 63:148-155. [PMID: 28355376 DOI: 10.1590/1806-9282.63.02.148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 06/26/2016] [Indexed: 11/22/2022] Open
Abstract
The skeletal muscle tissue has a remarkable ability to alter its plastic structural and functional properties after a harmful stimulus, regulating the expression of proteins in complex events such as muscle regeneration. In this context, considering that potential therapeutic agents have been widely studied, nutritional strategies have been investigated in order to improve the regenerative capacity of skeletal muscle. There is evidence of the modulatory action of fatty acids, such that oleic and linoleic acids, that are abundant in Western diets, on muscle function and trophism. Thus, fatty acids appear to be potential candidates to promote or impair the recovery of muscle mass and function during regeneration, since they modulate intracellular pathways that regulate myogenesis. This study is the first to describe and discuss the effect of fatty acids on muscle plasticity and trophism, with emphasis on skeletal muscle regeneration and in vitro differentiation of muscle cells.
Collapse
Affiliation(s)
- Phablo Abreu
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, SP, Brazil
| | - José Henrique Leal-Cardoso
- Department of Physiology, Institute for Biomedical Sciences, Universidade Estadual do Ceará, Fortaleza, CE, Brazil
| | - Vânia Marilande Ceccatto
- Department of Physiology, Institute for Biomedical Sciences, Universidade Estadual do Ceará, Fortaleza, CE, Brazil
| | - Sandro Massao Hirabara
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, SP, Brazil.,Interdisciplinary Graduate Program in Health Sciences, Institute of Physical Activity and Sport Sciences, Universidade Cruzeiro do Sul, São Paulo, SP, Brazil
| |
Collapse
|
38
|
Alom N, Peto H, Kirkham GR, Shakesheff KM, White LJ. Bone extracellular matrix hydrogel enhances osteogenic differentiation of C2C12 myoblasts and mouse primary calvarial cells. J Biomed Mater Res B Appl Biomater 2017; 106:900-908. [PMID: 28429412 DOI: 10.1002/jbm.b.33894] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 02/01/2017] [Accepted: 03/25/2017] [Indexed: 12/20/2022]
Abstract
Hydrogel scaffolds derived from the extracellular matrix (ECM) of mammalian tissues have been successfully used to promote tissue repair in vitro and in vivo. The objective of this study was to evaluate the osteogenic potential of ECM hydrogels prepared from demineralized and decellularized bovine bone in the presence and absence of osteogenic medium. Culture of C2C12 and mouse primary calvarial cells (mPCs) on decellularized bone ECM (bECM) and demineralized bone matrix (DBM) gels resulted in increased expression of osteogenic gene markers, including a 3.6- and 13.4-fold increase in osteopontin and 15.7- and 27.1-fold increase in osteocalcin when mPCs were cultured upon bECM with basal and osteogenic media, respectively. bECM hydrogels stimulated the osteogenic differentiation of C2C12 and mPCs even in the absence of osteogenic medium. These results suggest that bECM hydrogel scaffolds may have great utility in future clinical applications for bone tissue engineering. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 900-908, 2018.
Collapse
Affiliation(s)
- Noura Alom
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham University Park, Nottingham, United Kingdom
| | - Heather Peto
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham University Park, Nottingham, United Kingdom
| | - Glen R Kirkham
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham University Park, Nottingham, United Kingdom
| | - Kevin M Shakesheff
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham University Park, Nottingham, United Kingdom
| | - Lisa J White
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham University Park, Nottingham, United Kingdom
| |
Collapse
|
39
|
Akirin2 regulates proliferation and differentiation of porcine skeletal muscle satellite cells via ERK1/2 and NFATc1 signaling pathways. Sci Rep 2017; 7:45156. [PMID: 28327665 PMCID: PMC5361102 DOI: 10.1038/srep45156] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 02/21/2017] [Indexed: 12/16/2022] Open
Abstract
Akirin2, a novel nuclear factor, plays an important role in myogenesis. To investigate the role of Akirin2 in proliferation and differentiation of porcine skeletal muscle satellite cells, Akirin2 overexpression and Akirin2 silence technologies were employed. Our results showed that overexpression of Akirin2 markedly enhanced the proliferation and differentiation of porcine skeletal muscle satellite cells, whereas silencing of Akirin2 got the opposite results. Furthermore, our results showed that Akirin2 affected proliferation and differentiation of porcine skeletal muscle satellite cells through extracellular-signal regulated kinase-1/2 (ERK1/2) and NFATc1 signaling pathways. These results indicate that Akirin2 can effectively promote skeletal muscle satellite cells proliferation and differentiation, acting through ERK1/2- and NFATc1-dependent mechanisms.
Collapse
|
40
|
Abreu P, Pinheiro CHJ, Vitzel KF, Vasconcelos DAA, Torres RP, Fortes MS, Marzuca-Nassr GN, Mancini-Filho J, Hirabara SM, Curi R. Contractile function recovery in severely injured gastrocnemius muscle of rats treated with either oleic or linoleic acid. Exp Physiol 2016; 101:1392-1405. [PMID: 27579497 DOI: 10.1113/ep085899] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/18/2016] [Indexed: 12/31/2022]
Abstract
NEW FINDINGS What is the central question of this study? Oleic and linoleic acids modulate fibroblast proliferation and myogenic differentiation in vitro. However, their in vivo effects on muscle regeneration have not yet been examined. We investigated the effects of either oleic or linoleic acid on a well-established model of muscle regeneration after severe laceration. What is the main finding and its importance? We found that linoleic acid increases fibrous tissue deposition and impairs muscle regeneration and recovery of contractile function, whereas oleic acid has the opposite effects in severely injured gastrocnemius muscle, suggesting that linoleic acid has a harmful effect and oleic acid a potential therapeutic effect on muscle regeneration. Oleic and linoleic acids control fibroblast proliferation and myogenic differentiation in vitro; however, there was no study in skeletal muscle in vivo. The aim of this study was to evaluate the effects of either oleic or linoleic acid on the fibrous tissue content (collagen deposition) of muscle and recovery of contractile function in rat gastrocnemius muscle after being severely injured by laceration. Rats were supplemented with either oleic or linoleic acid for 4 weeks after laceration [0.44 g (kg body weight)-1 day-1 ]. Muscle injury led to an increase in oleic-to-stearic acid and palmitoleic-to-palmitic acid ratios, suggesting an increase in Δ9 desaturase activity. Increased fibrous tissue deposition and reduced isotonic and tetanic specific forces and resistance to fatigue were observed in the injured muscle. Supplementation with linoleic acid increased the content of eicosadienoic (20:2, n-6) and arachidonic (20:4, n-6) acids, reduced muscle mass and fibre cross-sectional areas, increased fibrous tissue deposition and further reduced the isotonic and tetanic specific forces and resistance to fatigue induced by laceration. Supplementation with oleic acid increased the content of docosahexaenoic acid (22:6, n-3) and abolished the increase in fibrous tissue area and the decrease in isotonic and tetanic specific forces and resistance to fatigue induced by muscle injury. We concluded that supplementation with linoleic acid impairs muscle regeneration and increases fibrous tissue deposition, resulting in impaired recovery of contractile function. Oleic acid supplementation reduced fibrous tissue deposition and improved recovery of contractile function, attenuating the tissue damage caused by muscle injury.
Collapse
Affiliation(s)
- Phablo Abreu
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Carlos H J Pinheiro
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Kaio F Vitzel
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | | - Rosângela P Torres
- Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Marco S Fortes
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | | - Jorge Mancini-Filho
- Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Sandro M Hirabara
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.,Institute of Physical Activity Sciences and Sports, Cruzeiro do Sul University, São Paulo, SP, Brazil
| | - Rui Curi
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
41
|
Marzuca-Nassr GN, Vitzel KF, De Sousa LG, Murata GM, Crisma AR, Rodrigues Junior CF, Abreu P, Torres RP, Mancini-Filho J, Hirabara SM, Newsholme P, Curi R. Effects of high EPA and high DHA fish oils on changes in signaling associated with protein metabolism induced by hindlimb suspension in rats. Physiol Rep 2016; 4:e12958. [PMID: 27650250 PMCID: PMC5037913 DOI: 10.14814/phy2.12958] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 08/09/2016] [Accepted: 08/11/2016] [Indexed: 12/16/2022] Open
Abstract
The effects of either eicosapentaenoic (EPA)- or docosahexaenoic (DHA)-rich fish oils on hindlimb suspension (HS)-induced muscle disuse atrophy were compared. Daily oral supplementations (0.3 mL/100 g b.w.) with mineral oil (MO) or high EPA or high DHA fish oils were performed in adult rats. After 2 weeks, the animals were subjected to HS for further 2 weeks. The treatments were maintained alongside HS At the end of 4 weeks, we evaluated: body weight gain, muscle mass and fat depots, composition of fatty acids, cross-sectional areas (CSA) of the soleus muscle and soleus muscle fibers, activities of cathepsin L and 26S proteasome, and content of carbonylated proteins in the soleus muscle. Signaling pathway activities associated with protein synthesis (Akt, p70S6K, S6, 4EBP1, and GSK3-beta) and protein degradation (atrogin-1/MAFbx, and MuRF1) were evaluated. HS decreased muscle mass, CSA of soleus muscle and soleus muscle fibers, and altered signaling associated with protein synthesis (decreased) and protein degradation (increased). The treatment with either fish oil decreased the ratio of omega-6/omega-3 fatty acids and changed protein synthesis-associated signaling. EPA-rich fish oil attenuated the changes induced by HS on 26S proteasome activity, CSA of soleus muscle fibers, and levels of p-Akt, total p70S6K, p-p70S6K/total p70S6K, p-4EBP1, p-GSK3-beta, p-ERK2, and total ERK 1/2 proteins. DHA-rich fish oil attenuated the changes induced by HS on p-4EBP1 and total ERK1 levels. The effects of EPA-rich fish oil on protein synthesis signaling were more pronounced. Both EPA- and DHA-rich fish oils did not impact skeletal muscle mass loss induced by non-inflammatory HS.
Collapse
Affiliation(s)
- Gabriel Nasri Marzuca-Nassr
- Department of Physiology and Biophysics, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil
| | - Kaio Fernando Vitzel
- Department of Physiology and Biophysics, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil Massey Institute of Food Science and Technology, College of Health Massey University, Albany, New Zealand
| | - Luís Gustavo De Sousa
- Department of Physiology and Biophysics, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil
| | - Gilson M Murata
- Department of Physiology and Biophysics, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil
| | - Amanda Rabello Crisma
- Department of Physiology and Biophysics, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil
| | | | - Phablo Abreu
- Department of Physiology and Biophysics, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil
| | - Rosângela Pavan Torres
- Department of Lipids Laboratory, Food Science & Nutrition, Faculty of Pharmaceutical Science University of São Paulo, São Paulo, Brazil
| | - Jorge Mancini-Filho
- Department of Lipids Laboratory, Food Science & Nutrition, Faculty of Pharmaceutical Science University of São Paulo, São Paulo, Brazil
| | - Sandro M Hirabara
- Institute of Physical Activity Sciences and Sport, Cruzeiro do Sul University, São Paulo, Brazil
| | - Philip Newsholme
- School of Biomedical Sciences, CHIRI Biosciences Curtin University, Perth, Australia
| | - Rui Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil
| |
Collapse
|
42
|
Grabiec K, Majewska A, Wicik Z, Milewska M, Błaszczyk M, Grzelkowska-Kowalczyk K. The effect of palmitate supplementation on gene expression profile in proliferating myoblasts. Cell Biol Toxicol 2016; 32:185-98. [PMID: 27114085 PMCID: PMC4882353 DOI: 10.1007/s10565-016-9324-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 03/28/2016] [Indexed: 12/20/2022]
Abstract
High-fat diet, exposure to saturated fatty acids, or the presence of adipocytes in myoblast microenvironment affects skeletal muscle growth and function. The aim of the present study was to investigate the effect of palmitate supplementation on transcriptomic profile of mouse C2C12 myoblasts. Global gene expression was evaluated using whole mouse genome oligonucleotide microarrays, and the results were validated through qPCR. A total of 4047 genes were identified as differentially expressed, including 3492 downregulated and 555 upregulated genes, during a 48-h exposure to palmitate (0.1 mmol/l). Functional classification showed the involvement of these genes in several processes which regulate cell growth. In conclusion, the addition of palmitate modifies the expression of genes associated with (1) myoblast responsiveness to hormones and growth factors, (2) cytokine and growth factor expression, and (3) regulation of cell-cell and cell-matrix communication. Such alterations can affect myoblast growth and differentiation; however, further studies in this field are required.
Collapse
Affiliation(s)
- K Grabiec
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Nowoursynowska 159, 02-776, Warsaw, Poland
| | - A Majewska
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Nowoursynowska 159, 02-776, Warsaw, Poland
| | - Z Wicik
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Nowoursynowska 159, 02-776, Warsaw, Poland
| | - M Milewska
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Nowoursynowska 159, 02-776, Warsaw, Poland
| | - M Błaszczyk
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Nowoursynowska 159, 02-776, Warsaw, Poland
| | - K Grzelkowska-Kowalczyk
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Nowoursynowska 159, 02-776, Warsaw, Poland.
| |
Collapse
|
43
|
Lee YH, Kim WJ, Lee MH, Kim SY, Seo DH, Kim HS, Gelinsky M, Kim TJ. Anti-skeletal muscle atrophy effect of Oenothera odorata root extract via reactive oxygen species-dependent signaling pathways in cellular and mouse model. Biosci Biotechnol Biochem 2015; 80:80-8. [PMID: 26613402 DOI: 10.1080/09168451.2015.1075861] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Skeletal muscle atrophy can be defined as a decrease of muscle volume caused by injury or lack of use. This condition is associated with reactive oxygen species (ROS), resulting in various muscular disorders. We acquired 2D and 3D images using micro-computed tomography in gastrocnemius and soleus muscles of sciatic-denervated mice. We confirmed that sciatic denervation-small animal model reduced muscle volume. However, the intraperitoneal injection of Oenothera odorata root extract (EVP) delayed muscle atrophy compared to a control group. We also investigated the mechanism of muscle atrophy's relationship with ROS. EVP suppressed expression of SOD1, and increased expression of HSP70, in both H2O2-treated C2C12 myoblasts and sciatic-denervated mice. Moreover, EVP regulated apoptotic signals, including caspase-3, Bax, Bcl-2, and ceramide. These results indicate that EVP has a positive effect on reducing the effect of ROS on muscle atrophy.
Collapse
Affiliation(s)
- Yong-Hyeon Lee
- a Division of Biological Science and Technology, Yonsei-Fraunhofer Medical Device Lab , College of Science and Technology, Yonsei University , Wonju , Korea
| | - Wan-Joong Kim
- a Division of Biological Science and Technology, Yonsei-Fraunhofer Medical Device Lab , College of Science and Technology, Yonsei University , Wonju , Korea
| | - Myung-Hun Lee
- a Division of Biological Science and Technology, Yonsei-Fraunhofer Medical Device Lab , College of Science and Technology, Yonsei University , Wonju , Korea
| | - Sun-Young Kim
- a Division of Biological Science and Technology, Yonsei-Fraunhofer Medical Device Lab , College of Science and Technology, Yonsei University , Wonju , Korea
| | - Dong-Hyun Seo
- b Department of Biomedical Engineering, Yonsei-Fraunhofer Medical Device Lab , College of Health Science, Yonsei University , Wonju , Korea
| | - Han-Sung Kim
- b Department of Biomedical Engineering, Yonsei-Fraunhofer Medical Device Lab , College of Health Science, Yonsei University , Wonju , Korea
| | - Michael Gelinsky
- c Center for Translational Bone, Joint and Soft Tissue Research, Medical Faculty and University Hospital, Technische Universität Dresden , Dresden , Germany
| | - Tack-Joong Kim
- a Division of Biological Science and Technology, Yonsei-Fraunhofer Medical Device Lab , College of Science and Technology, Yonsei University , Wonju , Korea
| |
Collapse
|
44
|
Qi R, Chen Y, Peng H, Jiang S, Xiao R, Huang J. Conjugated linoleic acid supplementation during late gestation and lactation of sows affects myofiber type in their litters. Livest Sci 2015. [DOI: 10.1016/j.livsci.2015.04.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
45
|
Blondelle J, Ohno Y, Gache V, Guyot S, Storck S, Blanchard-Gutton N, Barthélémy I, Walmsley G, Rahier A, Gadin S, Maurer M, Guillaud L, Prola A, Ferry A, Aubin-Houzelstein G, Demarquoy J, Relaix F, Piercy RJ, Blot S, Kihara A, Tiret L, Pilot-Storck F. HACD1, a regulator of membrane composition and fluidity, promotes myoblast fusion and skeletal muscle growth. J Mol Cell Biol 2015; 7:429-40. [PMID: 26160855 PMCID: PMC4589950 DOI: 10.1093/jmcb/mjv049] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/21/2015] [Indexed: 01/04/2023] Open
Abstract
The reduced diameter of skeletal myofibres is a hallmark of several congenital myopathies, yet the underlying cellular and molecular mechanisms remain elusive. In this study, we investigate the role of HACD1/PTPLA, which is involved in the elongation of the very long chain fatty acids, in muscle fibre formation. In humans and dogs, HACD1 deficiency leads to a congenital myopathy with fibre size disproportion associated with a generalized muscle weakness. Through analysis of HACD1-deficient Labradors, Hacd1-knockout mice, and Hacd1-deficient myoblasts, we provide evidence that HACD1 promotes myoblast fusion during muscle development and regeneration. We further demonstrate that in normal differentiating myoblasts, expression of the catalytically active HACD1 isoform, which is encoded by a muscle-enriched splice variant, yields decreased lysophosphatidylcholine content, a potent inhibitor of myoblast fusion, and increased concentrations of ≥C18 and monounsaturated fatty acids of phospholipids. These lipid modifications correlate with a reduction in plasma membrane rigidity. In conclusion, we propose that fusion impairment constitutes a novel, non-exclusive pathological mechanism operating in congenital myopathies and reveal that HACD1 is a key regulator of a lipid-dependent muscle fibre growth mechanism.
Collapse
Affiliation(s)
- Jordan Blondelle
- Inserm, IMRB U955-E10, 94000 Créteil, France Université Paris-Est, Ecole nationale vétérinaire d'Alfort (EnvA), 94700 Maisons-Alfort, France Université Paris-Est Créteil, Faculté de médecine, 94000 Créteil, France
| | - Yusuke Ohno
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Vincent Gache
- Inserm, IMRB U955-E10, 94000 Créteil, France Université Paris-Est, Ecole nationale vétérinaire d'Alfort (EnvA), 94700 Maisons-Alfort, France Université Paris-Est Créteil, Faculté de médecine, 94000 Créteil, France
| | - Stéphane Guyot
- Université de Bourgogne, UMR A 02.102 PAM-EPMB, AgroSup Dijon, 21000 Dijon, France
| | - Sébastien Storck
- Institut Necker-Enfants Malades, INSERM U1151-CNRS UMR 8253, Sorbonne Paris Cité, Université Paris Descartes, Faculté de Médecine-Site Broussais, 75015 Paris, France
| | - Nicolas Blanchard-Gutton
- Inserm, IMRB U955-E10, 94000 Créteil, France Université Paris-Est, Ecole nationale vétérinaire d'Alfort (EnvA), 94700 Maisons-Alfort, France Université Paris-Est Créteil, Faculté de médecine, 94000 Créteil, France
| | - Inès Barthélémy
- Inserm, IMRB U955-E10, 94000 Créteil, France Université Paris-Est, Ecole nationale vétérinaire d'Alfort (EnvA), 94700 Maisons-Alfort, France Université Paris-Est Créteil, Faculté de médecine, 94000 Créteil, France
| | - Gemma Walmsley
- Comparative Neuromuscular Disease Laboratory, Department of Clinical Sciences and Services, Royal Veterinary College, London NW1 0TU, UK
| | - Anaëlle Rahier
- Inserm, IMRB U955-E10, 94000 Créteil, France Université Paris-Est, Ecole nationale vétérinaire d'Alfort (EnvA), 94700 Maisons-Alfort, France Université Paris-Est Créteil, Faculté de médecine, 94000 Créteil, France
| | - Stéphanie Gadin
- Inserm, IMRB U955-E10, 94000 Créteil, France Université Paris-Est, Ecole nationale vétérinaire d'Alfort (EnvA), 94700 Maisons-Alfort, France Université Paris-Est Créteil, Faculté de médecine, 94000 Créteil, France
| | - Marie Maurer
- Inserm, IMRB U955-E10, 94000 Créteil, France Université Paris-Est, Ecole nationale vétérinaire d'Alfort (EnvA), 94700 Maisons-Alfort, France Université Paris-Est Créteil, Faculté de médecine, 94000 Créteil, France
| | - Laurent Guillaud
- Inserm, IMRB U955-E10, 94000 Créteil, France Université Paris-Est, Ecole nationale vétérinaire d'Alfort (EnvA), 94700 Maisons-Alfort, France Université Paris-Est Créteil, Faculté de médecine, 94000 Créteil, France
| | - Alexandre Prola
- Inserm, IMRB U955-E10, 94000 Créteil, France Université Paris-Est, Ecole nationale vétérinaire d'Alfort (EnvA), 94700 Maisons-Alfort, France Université Paris-Est Créteil, Faculté de médecine, 94000 Créteil, France
| | - Arnaud Ferry
- Thérapie des maladies du muscle strié INSERM U974 - CNRS UMR7215 - UPMC UM76 - Institut de Myologie, Université Pierre et Marie Curie - Université Paris Descartes, 75000 Paris, France
| | - Geneviève Aubin-Houzelstein
- Inserm, IMRB U955-E10, 94000 Créteil, France Université Paris-Est, Ecole nationale vétérinaire d'Alfort (EnvA), 94700 Maisons-Alfort, France Université Paris-Est Créteil, Faculté de médecine, 94000 Créteil, France
| | - Jean Demarquoy
- Université de Bourgogne, Faculté des Sciences Gabriel, Bio-PeroxIL, 21000 Dijon, France
| | - Frédéric Relaix
- Inserm, IMRB U955-E10, 94000 Créteil, France Université Paris-Est, Ecole nationale vétérinaire d'Alfort (EnvA), 94700 Maisons-Alfort, France Université Paris-Est Créteil, Faculté de médecine, 94000 Créteil, France
| | - Richard J Piercy
- Comparative Neuromuscular Disease Laboratory, Department of Clinical Sciences and Services, Royal Veterinary College, London NW1 0TU, UK
| | - Stéphane Blot
- Inserm, IMRB U955-E10, 94000 Créteil, France Université Paris-Est, Ecole nationale vétérinaire d'Alfort (EnvA), 94700 Maisons-Alfort, France Université Paris-Est Créteil, Faculté de médecine, 94000 Créteil, France
| | - Akio Kihara
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Laurent Tiret
- Inserm, IMRB U955-E10, 94000 Créteil, France Université Paris-Est, Ecole nationale vétérinaire d'Alfort (EnvA), 94700 Maisons-Alfort, France Université Paris-Est Créteil, Faculté de médecine, 94000 Créteil, France
| | - Fanny Pilot-Storck
- Inserm, IMRB U955-E10, 94000 Créteil, France Université Paris-Est, Ecole nationale vétérinaire d'Alfort (EnvA), 94700 Maisons-Alfort, France Université Paris-Est Créteil, Faculté de médecine, 94000 Créteil, France
| |
Collapse
|
46
|
Abstract
DNA damage is induced in many types of cells by internal and external cell stress. When DNA is damaged, DNA Damage Response (DDR) programs are activated to repair the DNA lesions in order to preserve genomic integrity and suppress subsequent malignant transformation. Among these programs is cell cycle checkpoint that ensures cell cycle arrest and subsequent repair of the damaged DNA, apoptosis and senescence in various phases of the cell cycle. Moreover, recent studies have established the cell differentiation checkpoint, the other type of the checkpoint that is specifically activated in the course of differentiation. We will discuss the evidences that support the link between DNA damage proteins and C2C12 cell differentiation.
Collapse
Affiliation(s)
- Sara Cuesta Sancho
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY14263, USA
| | - Toru Ouchi
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY14263, USA
| |
Collapse
|
47
|
Grabiec K, Milewska M, Błaszczyk M, Gajewska M, Grzelkowska-Kowalczyk K. Palmitate exerts opposite effects on proliferation and differentiation of skeletal myoblasts. Cell Biol Int 2015; 39:1044-52. [PMID: 25857830 DOI: 10.1002/cbin.10477] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 03/31/2015] [Indexed: 12/27/2022]
Abstract
The purpose of the study was to examine mechanisms controlling cell cycle progression/arrest and differentiation of mouse C2C12 myoblasts exposed to long-chain saturated fatty acid salt, palmitate. Treatment of proliferating myoblasts with palmitate (0.1 mmol/l) markedly decreased myoblast number. Cyclin A and cyclin D1 levels decreased, whereas total p21 and p21 complexed with cyclin-dependent kinase-4 (cdk4) increased in myoblasts treated with palmitate. In cells induced to differentiation addition of palmitate augmented the level of cyclin D3, the early (myogenin) and late (α-actinin, myosin heavy chain) markers of myogenesis, and caused an increase of myotube diameter. In conclusion, exposure to palmitate inhibits proliferation of myoblasts through a decrease in cyclin A and cyclin D1 levels and an increase of p21-cdk4 complex formation; however, it promotes cell cycle exit, myogenic differentiation and myotube growth.
Collapse
Affiliation(s)
- Kamil Grabiec
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Nowoursynowska 159, 02-776, Warsaw, Poland
| | - Marta Milewska
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Nowoursynowska 159, 02-776, Warsaw, Poland
| | - Maciej Błaszczyk
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Nowoursynowska 159, 02-776, Warsaw, Poland
| | - Małgorzata Gajewska
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Nowoursynowska 159, 02-776, Warsaw, Poland
| | - Katarzyna Grzelkowska-Kowalczyk
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Nowoursynowska 159, 02-776, Warsaw, Poland
| |
Collapse
|
48
|
Effects of dietary n-6:n-3 PUFA ratio on fatty acid composition, free amino acid profile and gene expression of transporters in finishing pigs. Br J Nutr 2015; 113:739-48. [PMID: 25704496 DOI: 10.1017/s0007114514004346] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Revealing the expression patterns of fatty acid and amino acid transporters as affected by dietary n-6:n-3 PUFA ratio would be useful for further clarifying the importance of the balance between n-6 and n-3 PUFA. A total of ninety-six finishing pigs were fed one of four diets with the ratio of 1:1, 2·5:1, 5:1 and 10:1. Pigs fed the dietary n-6:n-3 PUFA ratio of 5:1 had the highest (P< 0·05) daily weight gain, and those fed the dietary n-6:n-3 PUFA ratio of 1:1 had the largest loin muscle area (P< 0·01). The concentration of n-3 PUFA was raised as the ratio declined (P< 0·05) in the longissimus dorsi and subcutaneous adipose tissue. The contents of tryptophan, tasty amino acids and branched-chain amino acids in the longissimus dorsi were enhanced in pigs fed the dietary n-6:n-3 PUFA ratios of 1:1-5:1. The mRNA expression level of the fatty acid transporter fatty acid transport protein-1 (FATP-1) was declined (P< 0·05) in the longissimus dorsi of pigs fed the dietary n-6:n-3 PUFA ratios of 1:1-5:1, and increased (P< 0·05) in the subcutaneous adipose tissue of pigs fed the dietary n-6:n-3 PUFA ratios of 5:1 and 10:1. The expression profile of FATP-4 was similar to those of FATP-1 in the adipose tissue. The mRNA expression level of the amino acid transceptors LAT1 and SNAT2 was up-regulated (P< 0·05) in the longissimus dorsi of pigs fed the dietary n-6:n-3 PUFA ratios of 1:1 and 2·5:1. In conclusion, maintaining the dietary n-6:n-3 PUFA ratios of 1:1-5:1 would facilitate the absorption and utilisation of fatty acids and free amino acids, and result in improved muscle and adipose composition.
Collapse
|
49
|
Kim J, Carlson ME, Watkins BA. Docosahexaenoyl ethanolamide improves glucose uptake and alters endocannabinoid system gene expression in proliferating and differentiating C2C12 myoblasts. Front Physiol 2014; 5:100. [PMID: 24711795 PMCID: PMC3968752 DOI: 10.3389/fphys.2014.00100] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 02/27/2014] [Indexed: 11/26/2022] Open
Abstract
Skeletal muscle is a major storage site for glycogen and a focus for understanding insulin resistance and type-2-diabetes. New evidence indicates that overactivation of the peripheral endocannabinoid system (ECS) in skeletal muscle diminishes insulin sensitivity. Specific n-6 and n-3 polyunsaturated fatty acids (PUFA) are precursors for the biosynthesis of ligands that bind to and activate the cannabinoid receptors. The function of the ECS and action of PUFA in skeletal muscle glucose uptake was investigated in proliferating and differentiated C2C12 myoblasts treated with either 25 μM of arachidonate (AA) or docosahexaenoate (DHA), 25 μM of EC [anandamide (AEA), 2-arachidonoylglycerol (2-AG), docosahexaenoylethanolamide (DHEA)], 1 μM of CB1 antagonist NESS0327, and CB2 inverse agonist AM630. Compared to the BSA vehicle control cell cultures in both proliferating and differentiated myoblasts those treated with DHEA, the EC derived from the n-3 PUFA DHA, had higher 24 h glucose uptake, while AEA and 2-AG, the EC derived from the n-6 PUFA AA, had lower basal glucose uptake. Adenylyl cyclase mRNA was higher in myoblasts treated with DHA in both proliferating and differentiated states while those treated with AEA or 2-AG were lower compared to the control cell cultures. Western blot and qPCR analysis showed higher expression of the cannabinoid receptors in differentiated myoblasts treated with DHA while the opposite was observed with AA. These findings indicate a compensatory effect of DHA and DHEA compared to AA-derived ligands on the ECS and associated ECS gene expression and higher glucose uptake in myoblasts.
Collapse
Affiliation(s)
- Jeffrey Kim
- Center on Aging, University of Connecticut Health Center Farmington, CT, USA
| | - Morgan E Carlson
- Center on Aging, University of Connecticut Health Center Farmington, CT, USA
| | - Bruce A Watkins
- Center on Aging, University of Connecticut Health Center Farmington, CT, USA
| |
Collapse
|
50
|
Gershon E, Vale WW. CRF type 2 receptors mediate the metabolic effects of ghrelin in C2C12 cells. Obesity (Silver Spring) 2014; 22:380-9. [PMID: 23804489 PMCID: PMC4170921 DOI: 10.1002/oby.20535] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 05/17/2013] [Indexed: 11/07/2022]
Abstract
OBJECTIVE Ghrelin is known to regulate appetite control and cellular metabolism. The corticotropin-releasing factor (CRF) family is also known to regulate energy balance. In this study, the links between ghrelin and the CRF family in C2C12 cells, a mouse myoblast cell line was investigated. DESIGN AND METHODS C2C12 cells were treated with ghrelin in the presence or absence of CRF receptor antagonists and then subjected to different metabolic analyses. RESULTS Ghrelin enhanced glucose uptake by C2C12 cells, induced GLUT4 translocation to the cell surface and decreased RBP4 expression. A CRF-R2 selective antagonist, anti-sauvagine-30, blocked ghrelin-induced glucose uptake, Ghrelin upregulated CRF-R2 but not CRF-R1 levels. Moreover, ghrelin-treated C2C12 cells displayed a cAMP and pERK activation in response to Ucn3, a CRF-R2 specific ligand, but not in response to CRF or stressin, CRF-R1 specific ligands. Ghrelin also induced UCP2 and UCP3 expression, which were blocked by anti- sauvagine-30. Ghrelin did not induce fatty acids uptake by C2C12 cells or ACC expression. Even though C2C12 cells clearly exhibited responses to ghrelin, the known ghrelin receptor, GHSR1a, was not detectable in C2C12 cells. CONCLUSION The results suggest that, ghrelin plays a role in regulating muscle glucose and, raise the possibility that suppression of the CRF-R2 pathway might provide benefits in high ghrelin states.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/pharmacology
- Biological Transport/drug effects
- Cell Line
- Cell Membrane/drug effects
- Cell Membrane/metabolism
- Cell Membrane/ultrastructure
- Gene Expression Regulation/drug effects
- Ghrelin/metabolism
- Glucose/metabolism
- Glucose Transporter Type 4/metabolism
- Ion Channels/agonists
- Ion Channels/genetics
- Ion Channels/metabolism
- Mice
- Mitochondrial Proteins/agonists
- Mitochondrial Proteins/genetics
- Mitochondrial Proteins/metabolism
- Myoblasts/drug effects
- Myoblasts/metabolism
- Myoblasts/ultrastructure
- Protein Transport/drug effects
- Pyrimidines/pharmacology
- Pyrroles/pharmacology
- Receptors, Corticotropin-Releasing Hormone/agonists
- Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors
- Receptors, Corticotropin-Releasing Hormone/genetics
- Receptors, Corticotropin-Releasing Hormone/metabolism
- Receptors, Ghrelin/genetics
- Receptors, Ghrelin/metabolism
- Retinol-Binding Proteins, Plasma/antagonists & inhibitors
- Retinol-Binding Proteins, Plasma/genetics
- Retinol-Binding Proteins, Plasma/metabolism
- Signal Transduction/drug effects
- Uncoupling Protein 2
- Uncoupling Protein 3
- Urocortins/metabolism
Collapse
Affiliation(s)
- Eran Gershon
- To whom correspondence should be addressed: Eran Gershon, Department of Ruminant Science, The Volcani Center, P.O.Box 6, Bet Dagan 50250, ISRAEL,
| | | |
Collapse
|