101
|
Ling M, Lai X, Quan L, Li F, Lang L, Fu Y, Feng S, Yi X, Zhu C, Gao P, Zhu X, Wang L, Shu G, Jiang Q, Wang S. Knockdown of VEGFB/VEGFR1 Signaling Promotes White Adipose Tissue Browning and Skeletal Muscle Development. Int J Mol Sci 2022; 23:ijms23147524. [PMID: 35886871 PMCID: PMC9315609 DOI: 10.3390/ijms23147524] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 02/01/2023] Open
Abstract
It has been demonstrated that vascular endothelial growth factor B (VEGFB) and vascular endothelial growth factor receptor 1 (VEGFR1) play a vital role in regulating vascular biological function. However, the role of VEGFB and VEGFR1 in regulating fat deposition and skeletal muscle growth remains unclear. Therefore, this study was conducted to investigate the effects of VEGFB and VEGFR1 on fat deposition and skeletal muscle growth in mice. Our results showed that knockdown of VEGFB decreased body weight and iWAT index, stimulated the browning of mice iWAT with increased expression of UCP1, decreased the diameters of adipocytes, and elevated energy expenditure. In contrast, knockdown of VEGFB increased gastrocnemius (GAS) muscle index with increased proliferation of GAS muscle by expression of PCNA and Cyclin D1. Meanwhile, knockdown of endothelial VEGFR1 induced the browning of iWAT with increased expression of UCP1 and decreased diameters of adipocytes. By contrast, knockdown of endothelial VEGFR1 inhibited GAS muscle differentiation with decreased expression of MyoD. In conclusion, these results suggested that the loss of VEGFB/VEGFR1 signaling is associated with enhanced browning of inguinal white adipose tissue and skeletal muscle development. These results provided new insights into the regulation of skeletal muscle growth and regeneration, as well as fat deposition, suggesting the potential application of VEGFB/VEGFR1 as an intervention for the restriction of muscle diseases and obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Mingfa Ling
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (X.L.); (L.Q.); (F.L.); (L.L.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Xumin Lai
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (X.L.); (L.Q.); (F.L.); (L.L.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Lulu Quan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (X.L.); (L.Q.); (F.L.); (L.L.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Fan Li
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (X.L.); (L.Q.); (F.L.); (L.L.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Limin Lang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (X.L.); (L.Q.); (F.L.); (L.L.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Yiming Fu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (X.L.); (L.Q.); (F.L.); (L.L.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Shengchun Feng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (X.L.); (L.Q.); (F.L.); (L.L.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Xin Yi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (X.L.); (L.Q.); (F.L.); (L.L.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Canjun Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (X.L.); (L.Q.); (F.L.); (L.L.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Ping Gao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (X.L.); (L.Q.); (F.L.); (L.L.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Xiaotong Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (X.L.); (L.Q.); (F.L.); (L.L.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Lina Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (X.L.); (L.Q.); (F.L.); (L.L.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Gang Shu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (X.L.); (L.Q.); (F.L.); (L.L.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (X.L.); (L.Q.); (F.L.); (L.L.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Songbo Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.L.); (X.L.); (L.Q.); (F.L.); (L.L.); (Y.F.); (S.F.); (X.Y.); (C.Z.); (P.G.); (X.Z.); (L.W.); (G.S.); (Q.J.)
- National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Correspondence: ; Tel.: +86-135-7051-8681
| |
Collapse
|
102
|
CREB1 promotes proliferation and differentiation by mediating the transcription of CCNA2 and MYOG in bovine myoblasts. Int J Biol Macromol 2022; 216:32-41. [PMID: 35777504 DOI: 10.1016/j.ijbiomac.2022.06.177] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/17/2022] [Accepted: 06/26/2022] [Indexed: 11/23/2022]
Abstract
The cAMP response element binding protein 1 (CREB1) is an important nuclear transcription factor in eukaryotes. To explore the potential role of CREB1 on Qinchuan bovine skeletal myoblasts, we investigated the function of CREB1 on proliferation and differentiation. In this study, we found that CREB1 promoted cell proliferation by promoting DNA synthesis in S phase and cell division in G2 phase and promoted myogenic differentiation process in bovine myoblasts. Through dual luciferase experiments, we found that CREB1 can bind to the proximal promoter regions of CCNA2 and MyoG, indicating that CREB1 can play a positive regulatory role in the proliferation and differentiation of myoblasts by mediating the transcription of CCNA2 and MyoG. In addition, through downstream target gene analysis and transcriptome sequencing, we found that CREB1 plays a role in cell proliferation, myogenic differentiation, skeletal muscle repair and other related pathways.
Collapse
|
103
|
Kang K, Zhou N, Peng W, Peng F, Ma M, Li L, Fu F, Xiang S, Zhang H, He X, Song Z. Multi-Omics Analysis of the Microbiome and Metabolome Reveals the Relationship Between the Gut Microbiota and Wooden Breast Myopathy in Broilers. Front Vet Sci 2022; 9:922516. [PMID: 35812872 PMCID: PMC9260154 DOI: 10.3389/fvets.2022.922516] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/23/2022] [Indexed: 11/19/2022] Open
Abstract
Wooden breast (WB) is a widely prevalent myopathy in broiler chickens. However, the role of the gut microbiota in this myopathy remains largely unknown, in particular the regulatory effect of gut microbiota in the modulation of muscle metabolism. Totally, 300 1-day-old Arbor Acres broilers were raised until 49 days and euthanized, and the breast filets were classified as normal (NORM), mild (MILD), or severe wooden breast (SEV). Birds with WB comprised 27.02% of the individuals. Severe WB filets had a greater L* value, a* value, and dripping loss but a lower pH (P < 0.05). WB filets had abundant myofiber fragmentation, with a lower average myofiber caliber and more fibers with a diameter of <20 μm (P < 0.05). The diversity of the intestinal microflora was decreased in birds with severe WB, with decreases in Chao 1, and observed species indices. At the phylum level, birds with severe WB had a lower Firmicutes/Bacteroidetes ratio (P = 0.098) and a decreased abundance of Verrucomicrobia (P < 0.05). At the species level, gut microbiota were positively correlated with 131 digesta metabolites in pathways of glutamine and glutamate metabolism and arginine biosynthesis but were negatively correlated with 30 metabolites in the pathway of tyrosine metabolism. In plasma, WB induced five differentially expressed metabolites (DEMs), including anserine and choline, which were related to the severity of the WB lesion. The microbial-derived metabolites, including guanidoacetic acid, antiarol, and (2E)-decenoyl-ACP, which entered into plasma were related to meat quality traits and myofiber traits. In summary, WB filets differed in gut microbiota, digesta, and plasma metabolites. Gut microbiota respond to the wooden breast myopathy by driving dynamic changes in digesta metabolites that eventually enter the plasma.
Collapse
Affiliation(s)
- Kelang Kang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Ministry of Education Engineering Research Center of Feed Safety and Efficient Use, Changsha, China
| | - Nanxuan Zhou
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Ministry of Education Engineering Research Center of Feed Safety and Efficient Use, Changsha, China
| | - Weishi Peng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Ministry of Education Engineering Research Center of Feed Safety and Efficient Use, Changsha, China
| | - Fang Peng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Ministry of Education Engineering Research Center of Feed Safety and Efficient Use, Changsha, China
| | - Mengmeng Ma
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Ministry of Education Engineering Research Center of Feed Safety and Efficient Use, Changsha, China
| | - Liwei Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Ministry of Education Engineering Research Center of Feed Safety and Efficient Use, Changsha, China
| | - Fuyi Fu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Ministry of Education Engineering Research Center of Feed Safety and Efficient Use, Changsha, China
| | - Shuhan Xiang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Ministry of Education Engineering Research Center of Feed Safety and Efficient Use, Changsha, China
| | - Haihan Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Ministry of Education Engineering Research Center of Feed Safety and Efficient Use, Changsha, China
- Hunan Engineering Research Center of Poultry Production Safety, Changsha, China
| | - Xi He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Ministry of Education Engineering Research Center of Feed Safety and Efficient Use, Changsha, China
- Hunan Engineering Research Center of Poultry Production Safety, Changsha, China
- Hunan Co-Innovation Center of Animal Production Safety, Changsha, China
| | - Zehe Song
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Ministry of Education Engineering Research Center of Feed Safety and Efficient Use, Changsha, China
- Hunan Engineering Research Center of Poultry Production Safety, Changsha, China
- Hunan Co-Innovation Center of Animal Production Safety, Changsha, China
| |
Collapse
|
104
|
The mechanism of Megalobrama amblycephala muscle injury repair based on RNA-seq. Gene X 2022; 827:146455. [PMID: 35395368 DOI: 10.1016/j.gene.2022.146455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 01/19/2022] [Accepted: 03/25/2022] [Indexed: 11/22/2022] Open
Abstract
Skeletal muscle myogenesis and injury-induced muscle regeneration contribute to muscle formation. Skeletal muscle stem cells, termed satellite cells (SCs), proliferate to repair injured muscle. To identify the molecular mechanism of regeneration after muscle injury as well as the genes related to muscle development in fish, in this study, the immunohistochemistry and the high-throughput RNA sequencing (RNA-seq) analysis were performed after Megalobrama amblycephala muscle was injured by needle stab. The results showed that paired box7-positive (Pax7+) SCs increased, and peaked at 96 to 144 h-post injury (hpi). The 6729 differentially expressed genes (DEGs), including 2125 up-regulated and 4604 down-regulated genes were found. GO terms significantly enriched by DEGs contained intercellular connections, signaling transduction and enzyme activity. KEGG enrichment analysis showed that most of the pathways were related to immunity, metabolism and cells related molecules, including actin skeleton regulation, Epstein Barr virus infection and plaque adhesion. The WGCNA results revealed that actin cytoskeleton and lipid metabolism related genes probably played crucial roles during repair after muscle injury. Collectively, all these results will provide new insights into the molecular mechanisms underlying muscle injury repair of fish.
Collapse
|
105
|
Regulation of Non-Coding RNA in the Growth and Development of Skeletal Muscle in Domestic Chickens. Genes (Basel) 2022; 13:genes13061033. [PMID: 35741795 PMCID: PMC9222894 DOI: 10.3390/genes13061033] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/28/2022] [Accepted: 06/06/2022] [Indexed: 02/04/2023] Open
Abstract
Chicken is the most widely consumed meat product worldwide and is a high-quality source of protein for humans. The skeletal muscle, which accounts for the majority of chicken products and contains the most valuable components, is tightly correlated to meat product yield and quality. In domestic chickens, skeletal muscle growth is regulated by a complex network of molecules that includes some non-coding RNAs (ncRNAs). As a regulator of muscle growth and development, ncRNAs play a significant function in the development of skeletal muscle in domestic chickens. Recent advances in sequencing technology have contributed to the identification and characterization of more ncRNAs (mainly microRNAs (miRNAs), long non-coding RNAs (LncRNAs), and circular RNAs (CircRNAs)) involved in the development of domestic chicken skeletal muscle, where they are widely involved in proliferation, differentiation, fusion, and apoptosis of myoblasts and satellite cells, and the specification of muscle fiber type. In this review, we summarize the ncRNAs involved in the skeletal muscle growth and development of domestic chickens and discuss the potential limitations and challenges. It will provide a theoretical foundation for future comprehensive studies on ncRNA participation in the regulation of skeletal muscle growth and development in domestic chickens.
Collapse
|
106
|
Guo R, You X, Meng K, Sha R, Wang Z, Yuan N, Peng Q, Li Z, Xie Z, Chen R, Feng Y. Single-Cell RNA Sequencing Reveals Heterogeneity of Myf5-Derived Cells and Altered Myogenic Fate in the Absence of SRSF2. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105775. [PMID: 35460187 PMCID: PMC9218650 DOI: 10.1002/advs.202105775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/05/2022] [Indexed: 06/14/2023]
Abstract
Splicing factor SRSF2 acts as a critical regulator for cell survival, however, it remains unknown whether SRSF2 is involved in myoblast proliferation and myogenesis. Here, knockdown of SRSF2 in myoblasts causes high rates of apoptosis and defective differentiation. Combined conditional knockout and lineage tracing approaches show that Myf5-cre mice lacking SRSF2 die immediately at birth and exhibit a complete absence of mature myofibers. Mutant Myf5-derived cells (tdtomato-positive cells) are randomly scattered in the myogenic and non-myogenic regions, indicating loss of the community effect required for skeletal muscle differentiation. Single-cell RNA-sequencing reveals high heterogeneity of myf5-derived cells and non-myogenic cells are significantly increased at the expense of skeletal muscle cells in the absence of SRSF2, reflecting altered cell fate. SRSF2 is demonstrated to regulate the entry of Myf5 cells into the myogenic program and ensures their survival by preventing precocious differentiation and apoptosis. In summary, SRSF2 functions as an essential regulator for Myf5-derived cells to respond correctly to positional cues and to adopt their myogenic fate.
Collapse
Affiliation(s)
- Ruochen Guo
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong ProvinceJining Medical UniversityJining272067P. R. China
| | - Xue You
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong ProvinceJining Medical UniversityJining272067P. R. China
| | - Kai Meng
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong ProvinceJining Medical UniversityJining272067P. R. China
| | - Rula Sha
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Zhenzhen Wang
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Ningyang Yuan
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Qian Peng
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Zhigang Li
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Zhiqin Xie
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Ruijiao Chen
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong ProvinceJining Medical UniversityJining272067P. R. China
| | - Ying Feng
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong ProvinceJining Medical UniversityJining272067P. R. China
| |
Collapse
|
107
|
METTL3 promotes proliferation and myogenic differentiation through m6A RNA methylation/YTHDF1/2 signaling axis in myoblasts. Life Sci 2022; 298:120496. [DOI: 10.1016/j.lfs.2022.120496] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/17/2022] [Accepted: 03/19/2022] [Indexed: 01/11/2023]
|
108
|
Zhang Z, Deng K, Kang Z, Wang F, Fan Y. MicroRNA profiling reveals miR‐145‐5p inhibits goat myoblast differentiation by targeting the coding domain sequence of USP13. FASEB J 2022; 36:e22370. [DOI: 10.1096/fj.202200246r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/29/2022] [Accepted: 05/10/2022] [Indexed: 12/14/2022]
Affiliation(s)
- Zhen Zhang
- Institute of Sheep and Goat Science Nanjing Agricultural University Nanjing China
| | - Kaiping Deng
- Institute of Sheep and Goat Science Nanjing Agricultural University Nanjing China
| | - Ziqi Kang
- Institute of Sheep and Goat Science Nanjing Agricultural University Nanjing China
| | - Feng Wang
- Institute of Sheep and Goat Science Nanjing Agricultural University Nanjing China
| | - Yixuan Fan
- Institute of Sheep and Goat Science Nanjing Agricultural University Nanjing China
| |
Collapse
|
109
|
miR-377 Inhibits Proliferation and Differentiation of Bovine Skeletal Muscle Satellite Cells by Targeting FHL2. Genes (Basel) 2022; 13:genes13060947. [PMID: 35741709 PMCID: PMC9223022 DOI: 10.3390/genes13060947] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 02/01/2023] Open
Abstract
Non-coding RNAs, especially microRNAs (miRNAs), play an important role in skeletal muscle growth and development. miR-377 regulates many basic biological processes and plays a key role in tumor cell proliferation, migration and apoptosis. Nevertheless, the function of miR-377 during skeletal muscle development and how it regulates skeletal muscle satellite cells (SMSCs) remains unclear. In the present study, we proposed to elucidate the regulatory mechanism of miR-377 in the proliferation and differentiation of bovine primary SMSCs. Our results showed that miR-377 can significantly inhibit the proliferation of SMSCs. In addition, we found that miR-377 can reduce myotube formation and restrain skeletal myogenic differentiation. Moreover, the results obtained from the biosynthesis and dual luciferase experiments showed that FHL2 was the target gene of miR-377. We further probed the function of FHL2 in muscle development and found that FHL2 silencing significantly suppressed the proliferation and differentiation of SMSCS, which is contrary to the role of miR-377. Furthermore, FHL2 interacts with Dishevelled-2 (Dvl2) to enable Wnt/β-catenin signaling pathway, consequently regulating skeletal muscle development. miR-377 negatively regulates the Wnt/β-catenin signaling pathway by targeting FHL2-mediated Dvl2. Overall, these findings demonstrated that miR-377 regulates the bovine SMSCs proliferation and differentiation by targeting FHL2 and attenuating the Wnt/β-catenin signaling pathway.
Collapse
|
110
|
Yang X, Ning Y, Abbas Raza SH, Mei C, Zan L. MEF2C Expression Is Regulated by the Post-transcriptional Activation of the METTL3-m 6A-YTHDF1 Axis in Myoblast Differentiation. Front Vet Sci 2022; 9:900924. [PMID: 35573410 PMCID: PMC9096896 DOI: 10.3389/fvets.2022.900924] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/06/2022] [Indexed: 12/12/2022] Open
Abstract
N6-methyladenosine (m6A) plays an essential role in regulating gene expression. However, the effect of m6A on skeletal myoblast differentiation and the underlying mechanisms are still unclear. Here, we ascertained mRNA m6A methylation exhibited declined changes during bovine skeletal myoblast differentiation, and both MEF2C mRNA expression and m6A levels were significantly increased during myoblast differentiation. We found that MEF2C with mutated m6A sites significantly inhibited myoblast differentiation compared with wild-type MEF2C. METTL3 promoted MEF2C protein expression through posttranscriptional modification in an m6A-YTHDF1-dependent manner. Moreover, MEF2C promoted the expression of METTL3 by binding to its promoter. These results revealed that there is a positive feedback loop between these molecules in myoblast differentiation. Our study provided new insights into skeletal muscle differentiation and fusion, which may provide an RNA methylation-based approach for molecular genetics and breeding in livestock as well as for the treatment of muscle-related diseases.
Collapse
Affiliation(s)
- Xinran Yang
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | - Yue Ning
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China.,College of Chemistry and Chemical Engineering, Xianyang Normal University, Xianyang, China
| | | | - Chugang Mei
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China.,National Beef Cattle Improvement Center, Northwest A&F University, Xianyang, China
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China.,National Beef Cattle Improvement Center, Northwest A&F University, Xianyang, China
| |
Collapse
|
111
|
Identification of distinct non-myogenic skeletal-muscle-resident mesenchymal cell populations. Cell Rep 2022; 39:110785. [PMID: 35545045 PMCID: PMC9535675 DOI: 10.1016/j.celrep.2022.110785] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 02/23/2022] [Accepted: 04/13/2022] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal progenitors of the lateral plate mesoderm give rise to various cell fates within limbs, including a heterogeneous group of muscle-resident mesenchymal cells. Often described as fibro-adipogenic progenitors, these cells are key players in muscle development, disease, and regeneration. To further define this cell population(s), we perform lineage/reporter analysis, flow cytometry, single-cell RNA sequencing, immunofluorescent staining, and differentiation assays on normal and injured murine muscles. Here we identify six distinct Pdgfra+ non-myogenic muscle-resident mesenchymal cell populations that fit within a bipartite differentiation trajectory from a common progenitor. One branch of the trajectory gives rise to two populations of immune-responsive mesenchymal cells with strong adipogenic potential and the capability to respond to acute and chronic muscle injury, whereas the alternative branch contains two cell populations with limited adipogenic capacity and inherent mineralizing capabilities; one of the populations displays a unique neuromuscular junction association and an ability to respond to nerve injury. Leinroth et al. explore the heterogeneity of Pdgfra+ muscle-resident mesenchymal cells, demonstrating that Pdgfra+ subpopulations have unique gene expression profiles, exhibit two distinct cell trajectories from a common progenitor, differ in their abilities to respond to muscle injuries, and show variable adipogenic and mineralizing capacities.
Collapse
|
112
|
Zhou Z, Li K, Liu J, Zhang H, Fan Y, Chen Y, Han H, Yang J, Liu Y. Expression Profile Analysis to Identify Circular RNA Expression Signatures in Muscle Development of Wu'an Goat Longissimus Dorsi Tissues. Front Vet Sci 2022; 9:833946. [PMID: 35518637 PMCID: PMC9062782 DOI: 10.3389/fvets.2022.833946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
The growth and development of skeletal muscle is a physiological process regulated by a variety of genes and signaling pathways. As a posttranscriptional regulatory factor, circRNA plays a certain regulatory role in the development of animal skeletal muscle in the form of a miRNA sponge. However, the role of circRNAs in muscle development and growth in goats is still unclear. In our study, apparent differences in muscle fibers in Wu'an goats of different ages was firstly detected by hematoxylin-eosin (HE) staining, the circRNA expression profiles of longissimus dorsi muscles from 1-month-old (mon1) and 9-month-old (mon9) goats were screened by RNA-seq and verified by RT-qPCR. The host genes of differentially expressed (DE) circRNAs were predicted, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes analyses (KEGG) of host genes with DE circRNAs were performed to explore the functions of circRNAs. The circRNA-miRNA-mRNA networks were then constructed using Cytoscape software. Ten significantly differentially expressed circRNAs were also verified in the mon1 and mon9 groups by RT-qPCR. Luciferase Reporter Assay was used to verify the binding site between circRNA and its targeted miRNA. The results showed that a total of 686 DE circRNAs were identified between the mon9 and mon1 groups, of which 357 were upregulated and 329 were downregulated. Subsequently, the 467 host genes of DE circRNAs were predicted using Find_circ and CIRI software. The circRNA-miRNA-mRNA network contained 201 circRNAs, 85 miRNAs, and 581 mRNAs; the host mRNAs were associated with "muscle fiber development" and "AMPK signaling pathway" and were enriched in the FoxO signaling pathway. Competing endogenous RNA (ceRNA) network analysis showed that novel_circ_0005314, novel_circ_0005319, novel_circ_0009256, novel_circ_0009845, novel_circ_0005934 and novel_circ_0000134 may play important roles in skeletal muscle growth and development between the mon9 and mon1 groups. Luciferase Reporter Assay confirmed the combination between novel_circ_0005319 and chi-miR-199a-5p, novel_circ_0005934 and chi-miR-450-3p and novel_circ_0000134 and chi-miR-655. Our results provide specific information related to goat muscle development and a reference for the goat circRNA profile.
Collapse
Affiliation(s)
- Zuyang Zhou
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Kunyu Li
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Jiannan Liu
- School of Landscape and Ecological Engineering, Hebei University of Engineering, Handan, China
| | - Hui Zhang
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Yekai Fan
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Yulin Chen
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Haiyin Han
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Junqi Yang
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Yufang Liu
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| |
Collapse
|
113
|
Noda Y, Okada S, Suzuki T. Regulation of A-to-I RNA editing and stop codon recoding to control selenoprotein expression during skeletal myogenesis. Nat Commun 2022; 13:2503. [PMID: 35523818 PMCID: PMC9076623 DOI: 10.1038/s41467-022-30181-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 04/05/2022] [Indexed: 12/13/2022] Open
Abstract
Selenoprotein N (SELENON), a selenocysteine (Sec)-containing protein with high reductive activity, maintains redox homeostasis, thereby contributing to skeletal muscle differentiation and function. Loss-of-function mutations in SELENON cause severe neuromuscular disorders. In the early-to-middle stage of myoblast differentiation, SELENON maintains redox homeostasis and modulates endoplasmic reticulum (ER) Ca2+ concentration, resulting in a gradual reduction from the middle-to-late stages due to unknown mechanisms. The present study describes post-transcriptional mechanisms that regulate SELENON expression during myoblast differentiation. Part of an Alu element in the second intron of SELENON pre-mRNA is frequently exonized during splicing, resulting in an aberrant mRNA that is degraded by nonsense-mediated mRNA decay (NMD). In the middle stage of myoblast differentiation, ADAR1-mediated A-to-I RNA editing occurs in the U1 snRNA binding site at 5' splice site, preventing Alu exonization and producing mature mRNA. In the middle-to-late stage of myoblast differentiation, the level of Sec-charged tRNASec decreases due to downregulation of essential recoding factors for Sec insertion, thereby generating a premature termination codon in SELENON mRNA, which is targeted by NMD.
Collapse
Affiliation(s)
- Yuta Noda
- Department of Chemistry and Biotechnology, Graduate School of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Shunpei Okada
- Department of Chemistry and Biotechnology, Graduate School of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
- Department of Microbiology, Faculty of Medicine, Shimane University, 89-1 Enyacho, Izumo, Shimane, 693-8501, Japan
| | - Tsutomu Suzuki
- Department of Chemistry and Biotechnology, Graduate School of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan.
| |
Collapse
|
114
|
Lei Q, Hu X, Han H, Wang J, Liu W, Zhou Y, Cao D, Li F, Liu J. Integrative analysis of circRNA, miRNA, and mRNA profiles to reveal ceRNA regulation in chicken muscle development from the embryonic to post-hatching periods. BMC Genomics 2022; 23:342. [PMID: 35505302 PMCID: PMC9063329 DOI: 10.1186/s12864-022-08525-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 03/29/2022] [Indexed: 12/13/2022] Open
Abstract
Background The growth and development of skeletal muscle are regulated by protein-coding genes and non-coding RNA. Circular RNA (circRNA) is a type of non-coding RNA involved in a variety of biological processes, especially in post-transcriptional regulation. To better understand the regulatory mechanism of circRNAs during the development of muscle in chicken, we performed RNA-seq with linear RNA depletion for chicken breast muscle in 12 (E 12) and17 (E 17) day embryos, and 1 (D 1), 14 (D 14), 56 (D 56), and 98 (D 98) days post-hatch. Results We identified 5755 differentially expressed (DE)-circRNAs during muscle development. We profiled the expression of DE-circRNAs and mRNAs (identified in our previous study) at up to six time points during chicken muscle development and uncovered a significant profile (profile 16) for circRNA upregulation during aging in muscle tissues. To investigate competing endogenous RNA (ceRNA) regulation in muscle and identify muscle-related circRNAs, we constructed a circRNA-miRNA-mRNA regulatory network using the circRNAs and mRNAs from profile 16 and miRNAs identified in our previous study, which included 361 miRNAs, 68 circRNAs, 599 mRNAs, and 31,063 interacting pairs. Functional annotation showed that upregulated circRNAs might contribute to glycolysis/gluconeogenesis, biosynthesis of amino acids, pyruvate metabolism, carbon metabolism, glycogen and sucrose metabolism through the ceRNA network, and thus affected postnatal muscle development by regulating muscle protein deposition. Of them, circRNA225 and circRNA226 from the same host gene might be key circRNAs that could regulate muscle development by interacting with seven common miRNAs and 207 mRNAs. Our experiments also demonstrated that there were interactions among circRNA225, gga-miR-1306-5p, and heat shock protein alpha 8 (HSPA8). Conclusions Our results suggest that adequate supply of nutrients such as energy and protein after hatching may be a key factor in ensuring chicken yield, and provide several candidate circRNAs for future studies concerning ceRNA regulation during chicken muscle development. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08525-5.
Collapse
Affiliation(s)
- Qiuxia Lei
- Poultry Institute, Shandong Academy of Agricultural Sciences, Ji'nan, 250023, China.,Poultry Breeding Engineering Technology Center of Shandong Province, Ji'nan, 250023, China
| | - Xin Hu
- Poultry Institute, Shandong Academy of Agricultural Sciences, Ji'nan, 250023, China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.,Molecular and Cellular Biology, Gembloux Agro-Bio Tech, University of Liège, 5030, Gembloux, Belgium
| | - Haixia Han
- Poultry Institute, Shandong Academy of Agricultural Sciences, Ji'nan, 250023, China
| | - Jie Wang
- Poultry Institute, Shandong Academy of Agricultural Sciences, Ji'nan, 250023, China
| | - Wei Liu
- Poultry Institute, Shandong Academy of Agricultural Sciences, Ji'nan, 250023, China
| | - Yan Zhou
- Poultry Institute, Shandong Academy of Agricultural Sciences, Ji'nan, 250023, China
| | - Dingguo Cao
- Poultry Institute, Shandong Academy of Agricultural Sciences, Ji'nan, 250023, China
| | - Fuwei Li
- Poultry Institute, Shandong Academy of Agricultural Sciences, Ji'nan, 250023, China
| | - Jie Liu
- Poultry Institute, Shandong Academy of Agricultural Sciences, Ji'nan, 250023, China. .,Poultry Breeding Engineering Technology Center of Shandong Province, Ji'nan, 250023, China.
| |
Collapse
|
115
|
LncR-133a Suppresses Myoblast Differentiation by Sponging miR-133a-3p to Activate the FGFR1/ERK1/2 Signaling Pathway in Goats. Genes (Basel) 2022; 13:genes13050818. [PMID: 35627202 PMCID: PMC9141198 DOI: 10.3390/genes13050818] [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: 04/16/2022] [Revised: 04/30/2022] [Accepted: 05/01/2022] [Indexed: 12/03/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are involved in a variety of biological processes and illnesses. While a considerable number of lncRNAs have been discovered in skeletal muscle to far, their role and underlying processes during myogenesis remain mostly unclear. In this study, we described a new functional lncRNA named lncR-133a. Gene overexpression and interference studies in goat skeletal muscle satellite cells (MuSCs) were used to establish its function. The molecular mechanism by which lncR-133a governs muscle differentiation was elucidated primarily using quantitative real-time PCR (qRT-PCR), Western blotting, dual-luciferase activity assays, RNA immunoprecipitation, biotin-labeled probe, and RNA fluorescence in situ hybridization analyses. LncR-133a was found to be substantially expressed in longissimus thoracis et lumborum muscle, and its expression levels changed during MuSC differentiation in goats. We validated that lncR-133a suppresses MuSC differentiation in vitro. Dual-luciferase reporter screening, Argonaute 2 (AGO2) RNA immunoprecipitation assays, biotin-labeled lncR-133a capture, and fluorescence in situ hybridization showed that lncR-133a interacted with miR-133a-3p. Additionally, miR-133a-3p facilitated MuSC differentiation, but lncR-133a reversed this effect. The luciferase reporter assay and functional analyses established that miR-133a-3p directly targets fibroblast growth factor receptor 1 (FGFR1). Moreover, lncR-133a directly reduced miR-133a-3p’s capacity to suppress FGFR1 expression, and positively regulated the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2). In summary, our results suggested that lncR-133a suppresses goat muscle differentiation by targeting miR-133a-3p and activating FGFR1/ERK1/2 signaling pathway.
Collapse
|
116
|
Iram T, Kern F, Kaur A, Myneni S, Morningstar AR, Shin H, Garcia MA, Yerra L, Palovics R, Yang AC, Hahn O, Lu N, Shuken SR, Haney MS, Lehallier B, Iyer M, Luo J, Zetterberg H, Keller A, Zuchero JB, Wyss-Coray T. Young CSF restores oligodendrogenesis and memory in aged mice via Fgf17. Nature 2022; 605:509-515. [PMID: 35545674 PMCID: PMC9377328 DOI: 10.1038/s41586-022-04722-0] [Citation(s) in RCA: 104] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 04/04/2022] [Indexed: 12/15/2022]
Abstract
Recent understanding of how the systemic environment shapes the brain throughout life has led to numerous intervention strategies to slow brain ageing1-3. Cerebrospinal fluid (CSF) makes up the immediate environment of brain cells, providing them with nourishing compounds4,5. We discovered that infusing young CSF directly into aged brains improves memory function. Unbiased transcriptome analysis of the hippocampus identified oligodendrocytes to be most responsive to this rejuvenated CSF environment. We further showed that young CSF boosts oligodendrocyte progenitor cell (OPC) proliferation and differentiation in the aged hippocampus and in primary OPC cultures. Using SLAMseq to metabolically label nascent mRNA, we identified serum response factor (SRF), a transcription factor that drives actin cytoskeleton rearrangement, as a mediator of OPC proliferation following exposure to young CSF. With age, SRF expression decreases in hippocampal OPCs, and the pathway is induced by acute injection with young CSF. We screened for potential SRF activators in CSF and found that fibroblast growth factor 17 (Fgf17) infusion is sufficient to induce OPC proliferation and long-term memory consolidation in aged mice while Fgf17 blockade impairs cognition in young mice. These findings demonstrate the rejuvenating power of young CSF and identify Fgf17 as a key target to restore oligodendrocyte function in the ageing brain.
Collapse
Affiliation(s)
- Tal Iram
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA,Correspondence to or
| | - Fabian Kern
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Chair for Clinical Bioinformatics, Saarland University, 66123 Saarbrücken, Germany.,Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Saarland University Campus E8.1, Saarbrücken, Germany
| | - Achint Kaur
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Saket Myneni
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Allison R. Morningstar
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Heather Shin
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Miguel A. Garcia
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Lakshmi Yerra
- Palo Alto Veterans Institute for Research, Palo Alto, CA 94304
| | - Robert Palovics
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Andrew C. Yang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Oliver Hahn
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Nannan Lu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Steven R. Shuken
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA,Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Michael s. Haney
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Benoit Lehallier
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Manasi Iyer
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Jian Luo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Palo Alto Veterans Institute for Research, Palo Alto, CA 94304
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Andreas Keller
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Chair for Clinical Bioinformatics, Saarland University, 66123 Saarbrücken, Germany.,Center for Bioinformatics, Saarland Informatics Campus, 66123 Saarbrücken, Germany
| | - J. Bradley Zuchero
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, California, USA.,Correspondence to or
| |
Collapse
|
117
|
Szilágyi A, Takács B, Szekeres R, Tarjányi V, Bombicz M, Priksz D, Kovács A, Juhász B, Frecska E, Szilvássy Z, Varga B. Therapeutic Properties of Ayahuasca Components in Ischemia/Reperfusion Injury of the Eye. Biomedicines 2022; 10:997. [PMID: 35625734 PMCID: PMC9138933 DOI: 10.3390/biomedicines10050997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/13/2022] [Accepted: 04/23/2022] [Indexed: 02/06/2023] Open
Abstract
Ischemic eye diseases are major causes of vision impairment. Thus, potential retinoprotective effects of N'N-dimethyltryptamine (DMT) were investigated. To inhibit its rapid breakdown by monoamine-oxidase A (MAO-A) enzyme, DMT was co-administered with harmaline, a β-carboline in the Amazonian Ayahuasca brew. Using ligation, 60 min of ischemia was provoked in eyes of rats, followed by 7 days of reperfusion whilst animals received harmaline alone, DMT + harmaline, or vehicle treatment. After 1 week of reperfusion, electroretinographical (ERG) measurements, histological analysis, and Western blot were performed. Harmaline alone exhibited retinoprotection in ischemia-reperfusion (I/R) which was, surprisingly, counterbalanced by DMT in case of co-administration. As both MAO-A inhibition and DMT increase serotoninergic tone synergistically, communicated to be anti-ischemic, thus, involvement of other pathways was investigated. Based on our experiments, DMT and harmaline exert opposite effects on important ocular proteins such as PARP1, NFκB, MMP9, or HSP70, each having a critical role in a different mechanism of eye-ischemia-related pathologies, e.g., cell death, inflammation, tissue destruction, and oxidative stress. Since DMT is proclaimed to be a promising drug candidate, its potentially undesirable effect on eye-ischemia should be further investigated. Meanwhile, this experiment revealed the potential therapeutic effect of MAO-A inhibitor harmaline in I/R-related eye diseases.
Collapse
Affiliation(s)
- Anna Szilágyi
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.S.); (B.T.); (R.S.); (V.T.); (M.B.); (D.P.); (B.J.); (Z.S.)
| | - Barbara Takács
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.S.); (B.T.); (R.S.); (V.T.); (M.B.); (D.P.); (B.J.); (Z.S.)
| | - Réka Szekeres
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.S.); (B.T.); (R.S.); (V.T.); (M.B.); (D.P.); (B.J.); (Z.S.)
| | - Vera Tarjányi
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.S.); (B.T.); (R.S.); (V.T.); (M.B.); (D.P.); (B.J.); (Z.S.)
| | - Mariann Bombicz
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.S.); (B.T.); (R.S.); (V.T.); (M.B.); (D.P.); (B.J.); (Z.S.)
| | - Dániel Priksz
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.S.); (B.T.); (R.S.); (V.T.); (M.B.); (D.P.); (B.J.); (Z.S.)
| | - Attila Kovács
- Department of Psychiatry, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.K.); (E.F.)
| | - Béla Juhász
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.S.); (B.T.); (R.S.); (V.T.); (M.B.); (D.P.); (B.J.); (Z.S.)
| | - Ede Frecska
- Department of Psychiatry, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.K.); (E.F.)
| | - Zoltán Szilvássy
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.S.); (B.T.); (R.S.); (V.T.); (M.B.); (D.P.); (B.J.); (Z.S.)
| | - Balázs Varga
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.S.); (B.T.); (R.S.); (V.T.); (M.B.); (D.P.); (B.J.); (Z.S.)
| |
Collapse
|
118
|
Kitahata K, Uchida T, Taniguchi R, Kato A, Sugiura K, Sakakibara I, Oarada M, Fukawa T, Junsoo P, Inho C, Nikawa T. Additional effects of simultaneous treatment with C14-Cblin and celastrol on the clinorotation-induced rat L6 myotube atrophy. THE JOURNAL OF MEDICAL INVESTIGATION 2022; 69:127-134. [PMID: 35466134 DOI: 10.2152/jmi.69.127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Two novel reagents, N-myristoylated Cbl-b inhibitory peptide (C14-Cblin) and celastrol, a quinone methide triterpene, are reported to be effective in preventing myotube atrophy. The combined effects of C14-Cblin and celastrol on rat L6 myotubes atrophy induced by 3D-clinorotation, a simulated microgravity model, was investigated in the present study. We first examined their effects on expression in atrogenes. Increase in MAFbx1/atrogin-1 and MuRF-1 by 3D-clinorotation was significantly suppressed by treatment with C14-Cblin or celastrol, but there was no additive effect of simultaneous treatment. However, celastrol significantly suppressed the upregulation of Cbl-b and HSP70 by 3D-clinorotation. Whereas 3D-clinorotation decreased the protein level of IRS-1 in L6 myotubes, C14-Cblin and celastrol inhibited the degradation of IRS-1. C14-Cblin and celastrol promoted the phosphorylation of FOXO3a even in microgravity condition. Simultaneous administration of C14-Cblin and celastrol had shown little additive effect in reversing the impairment of IGF-1 signaling by 3D-clinorotation. While 3D-clinorotation-induced marked oxidative stress in L6 myotubes, celastrol suppressed 3D-clinorotation-induced ROS production. Finally, the C14-Cblin and celastrol-treated groups were inhibited decrease in L6 myotube diameter and increased the protein content of slow-twitch MyHC cultured under 3D-clinorotation. The simultaneous treatment of C14-Cblin and celastrol additively prevented 3D-clinorotation-induced myotube atrophy than single treatment. J. Med. Invest. 69 : 127-134, February, 2022.
Collapse
Affiliation(s)
- Kanako Kitahata
- Departments of Nutritional Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Takayuki Uchida
- Departments of Nutritional Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Runa Taniguchi
- Departments of Nutritional Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Ayano Kato
- Departments of Nutritional Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Kosuke Sugiura
- Departments of Nutritional Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan.,Department of Orthopedics, Institute of Medical Biosciences, Tokushima University Graduate School, Tokushima, 770-8503, Japan
| | - Iori Sakakibara
- Departments of Nutritional Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Motoko Oarada
- Department of Nutrition Health, Faculty of Nutritional Science Sagami Women's University, Sagamihara, Kanagawa, 252-0383, Japan
| | - Tomoya Fukawa
- Department of Urology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Park Junsoo
- Division of Biological Science and Technology, College of Science and Technology, Yonsei University, Wonju, Gangwon-Do, Republic of Korea
| | - Choi Inho
- Division of Biological Science and Technology, College of Science and Technology, Yonsei University, Wonju, Gangwon-Do, Republic of Korea
| | - Takeshi Nikawa
- Departments of Nutritional Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| |
Collapse
|
119
|
Cai B, Ma M, Zhang J, Kong S, Zhou Z, Li Z, Abdalla BA, Xu H, Zhang X, Lawal RA, Nie Q. Long noncoding RNA ZFP36L2-AS functions as a metabolic modulator to regulate muscle development. Cell Death Dis 2022; 13:389. [PMID: 35449125 PMCID: PMC9023450 DOI: 10.1038/s41419-022-04772-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 03/17/2022] [Accepted: 03/25/2022] [Indexed: 01/17/2023]
Abstract
Skeletal muscle is the largest metabolic organ in the body, and its metabolic flexibility is essential for maintaining systemic energy homeostasis. Metabolic inflexibility in muscles is a dominant cause of various metabolic disorders, impeding muscle development. In our previous study, we found lncRNA ZFP36L2-AS (for “ZFP36L2-antisense transcript”) is specifically enriched in skeletal muscle. Here, we report that ZFP36L2-AS is upregulated during myogenic differentiation, and highly expressed in breast and leg muscle. In vitro, ZFP36L2-AS inhibits myoblast proliferation but promotes myoblast differentiation. In vivo, ZFP36L2-AS facilitates intramuscular fat deposition, as well as activates fast-twitch muscle phenotype and induces muscle atrophy. Mechanistically, ZFP36L2-AS interacts with acetyl-CoA carboxylase alpha (ACACA) and pyruvate carboxylase (PC) to induce ACACA dephosphorylation and damaged PC protein stability, thus modulating muscle metabolism. Meanwhile, ZFP36L2-AS can activate ACACA to reduce acetyl-CoA content, which enhances the inhibition of PC activity. Our findings present a novel model about the regulation of lncRNA on muscle metabolism.
Collapse
Affiliation(s)
- Bolin Cai
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, 510642, Guangdong, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, Guangdong, China
| | - Manting Ma
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, 510642, Guangdong, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, Guangdong, China
| | - Jing Zhang
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, 510642, Guangdong, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, Guangdong, China
| | - Shaofen Kong
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, 510642, Guangdong, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, Guangdong, China
| | - Zhen Zhou
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, 510642, Guangdong, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, Guangdong, China
| | - Zhenhui Li
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, 510642, Guangdong, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, Guangdong, China
| | - Bahareldin Ali Abdalla
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, 510642, Guangdong, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, Guangdong, China
| | - Haiping Xu
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, 510642, Guangdong, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, Guangdong, China
| | - Xiquan Zhang
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, 510642, Guangdong, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, Guangdong, China
| | | | - Qinghua Nie
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, 510642, Guangdong, China. .,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, Guangdong, China.
| |
Collapse
|
120
|
Siddiqui SH, Subramaniyan SA, Park J, Kang D, Khan M, Belal SA, Lee SC, Shim K. Modulatory effects of cell–cell interactions between porcine skeletal muscle satellite cells and fibroblasts on the expression of myogenesis-related genes. JOURNAL OF APPLIED ANIMAL RESEARCH 2022. [DOI: 10.1080/09712119.2022.2060986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Sharif Hasan Siddiqui
- Department of Animal Biotechnology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, Republic of Korea
| | - Sivakumar Allur Subramaniyan
- Department of Orthopaedic Surgery, Dongtan Sacred Heart Hospital, Hallym University, College of Medicine, Hwaseong, Republic of Korea
| | - Jinryong Park
- Department of Animal Biotechnology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, Republic of Korea
| | - Darae Kang
- Department of Animal Biotechnology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, Republic of Korea
| | - Mousumee Khan
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Shah Ahmed Belal
- Department of Poultry Science, Sylhet Agricultural University, Sylhet, Bangladesh
| | | | - Kwanseob Shim
- Department of Animal Biotechnology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
121
|
Jin Z, Da W, Zhao Y, Wang T, Xu H, Shu B, Gao X, Shi Q, Ma Y, Zhang Y, Wang Y, Tang D. Role of skeletal muscle satellite cells in the repair of osteoporotic fractures mediated by β-catenin. J Cachexia Sarcopenia Muscle 2022; 13:1403-1417. [PMID: 35178895 PMCID: PMC8977954 DOI: 10.1002/jcsm.12938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 01/08/2022] [Accepted: 01/17/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Osteoporosis is a metabolic disease, and osteoporotic fracture (OPF) is one of its most serious complications. It is often ignored that the influence of the muscles surrounding the fracture on the healing of OPF. We aimed to clarify the role of skeletal muscle satellite cells (SMSCs) in promoting OPF healing by β-catenin, to improve our understanding of SMSCs, and let us explore its potential as a therapeutic target. METHODS Skeletal muscles were obtained from control non-OPF or OPF patients for primary SMSCs culture (n = 3, 33% females, mean age 60 ± 15.52). Expression of SMSCs was measured. In vivo, 3-month-old female C57BL/6 mice underwent OVX surgery. Three months later, the left tibia fracture model was again performed. The control and the treatment group (n = 24, per group, female). The treatment group was treated with an agonist (osthole). Detection of SMSCs in muscles and fracture healing at 7, 14, and 28 three time points (n = 8, 8, 8, female). To further clarify the scientific hypothesis, we innovatively used Pax7-CreERT2/+ ;β-cateninfx/fx transgenic mice (n = 12, per group, male). Knock out β-catenin in SMSC to observe the proliferation and osteogenic differentiation of SMSCs, and OPF healing. In vitro primary cells of SMSCs from 3-month-old litter-negative β-cateninfx/fx transgenic mice. After adenovirus-CRE transfection, the myogenic and osteogenic differentiation of SMSC was observed. RESULTS We find that human SMSCs reduced proliferation and osteogenic differentiation in patients with OPF (-38.63%, P < 0.05). And through animal experiments, it was found that activation of β-catenin promoted the proliferation and osteogenic differentiation of SMSC at the fracture site, thereby accelerating the healing of the fracture site (189.47%, P < 0.05). To prove this point of view, in the in vivo Pax7-CreERT2/+ ;β-cateninfx/fx transgenic mouse experiment, we innovatively found that knocking out β-catenin in SMSC will cause a decrease in bone mass and bone microstructure, and accompanied by delayed fracture healing (-35.04%, P < 0.001). At the same time, through in vitro SMSC culture experiments, it was found that their myogenic (-66.89%, P < 0.01) and osteogenic differentiation (-16.5%, P < 0.05) ability decreased. CONCLUSIONS These results provide the first practical evidence for a direct contribution of SMSCs to promote the healing of OPF with important clinical implications as it may help in the treatment of delayed healing and non-union of OPFs, and mobilization of autologous stem cell therapy in orthopaedic applications.
Collapse
Affiliation(s)
- Zhenxiong Jin
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weiwei Da
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yongjian Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tengteng Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bing Shu
- Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiang Gao
- Department of Orthopedics, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Qi Shi
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong Ma
- Department of Orthopedics, Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu, China
| | - Yan Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yongjun Wang
- Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dezhi Tang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
122
|
Myokines and Resistance Training: A Narrative Review. Int J Mol Sci 2022; 23:ijms23073501. [PMID: 35408868 PMCID: PMC8998961 DOI: 10.3390/ijms23073501] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 01/27/2023] Open
Abstract
In the last few years, the muscular system has gained attention due to the discovery of the muscle-secretome and its high potency for retaining or regaining health. These cytokines, described as myokines, released by the working muscle, are involved in anti-inflammatory, metabolic and immunological processes. These are able to influence human health in a positive way and are a target of research in metabolic diseases, cancer, neurological diseases, and other non-communicable diseases. Therefore, different types of exercise training were investigated in the last few years to find associations between exercise, myokines and their effects on human health. Particularly, resistance training turned out to be a powerful stimulus to enhance myokine release. As there are different types of resistance training, different myokines are stimulated, depending on the mode of training. This narrative review gives an overview about resistance training and how it can be utilized to stimulate myokine production in order to gain a certain health effect. Finally, the question of why resistance training is an important key regulator in human health will be discussed.
Collapse
|
123
|
Buckley KH, Nestor-Kalinoski AL, Pizza FX. Positional Context of Myonuclear Transcription During Injury-Induced Muscle Regeneration. Front Physiol 2022; 13:845504. [PMID: 35492593 PMCID: PMC9040890 DOI: 10.3389/fphys.2022.845504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/02/2022] [Indexed: 01/31/2023] Open
Abstract
Fundamental aspects underlying downstream processes of skeletal muscle regeneration, such as myonuclear positioning and transcription are poorly understood. This investigation begins to address deficiencies in knowledge by examining the kinetics of myonuclear accretion, positioning, and global transcription during injury-induced muscle regeneration in mice. We demonstrate that myonuclear accretion plateaus within 7 days of an injury and that the majority (∼70%) of myonuclei are centrally aligned in linear arrays (nuclear chains) throughout the course of regeneration. Relatively few myonuclei were found in a peripheral position (∼20%) or clustered (∼10%) together during regeneration. Importantly, transcriptional activity of individual myonuclei in nuclear chains was high, and greater than that of peripheral or clustered myonuclei. Transcription occurring primarily in nuclear chains elevated the collective transcriptional activity of regenerating myofibers during the later stage of regeneration. Importantly, the number of myonuclei in chains and their transcriptional activity were statistically correlated with an increase in myofiber size during regeneration. Our findings demonstrate the positional context of transcription during regeneration and highlight the importance of centralized nuclear chains in facilitating hypertrophy of regenerating myofibers after injury.
Collapse
Affiliation(s)
- Kole H. Buckley
- School of Exercise and Rehabilitation Sciences, University of Toledo, Toledo, OH, United States
| | | | - Francis X. Pizza
- School of Exercise and Rehabilitation Sciences, University of Toledo, Toledo, OH, United States
| |
Collapse
|
124
|
m6A Methylases Regulate Myoblast Proliferation, Apoptosis and Differentiation. Animals (Basel) 2022; 12:ani12060773. [PMID: 35327170 PMCID: PMC8944832 DOI: 10.3390/ani12060773] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary N6-methyladenosine (m6A) is the most prevalent methylation modification in eukaryotic mRNA, and it plays an important role in regulating gene expression. Previous studies found that m6A methylation plays a role in mammalian skeletal muscle development. Skeletal muscle is an important factor that regulates livestock muscle quality and maintains metabolic homeostasis, and skeletal myogenesis is regulated by a series of transcription factors. However, the role of m6A in bovine skeletal myogenesis is unclear. In this study, we examined the expression patterns of the m6A methylase genes METTL3, METTL14, WTAP, FTO and ALKBH5 in bovine skeletal muscle tissue and during myogenesis in myoblasts. Furthermore, we used bovine skeletal muscle myoblasts as the object of study to discover the regulatory role of these genes in the process of skeletal myogenesis in vitro. Our findings indicate that these five m6A methylases have pronounced and diverse functions in regulating bovine skeletal myoblast proliferation, apoptosis and myogenic differentiation, which can contribute to further understanding the roles of m6A in skeletal muscle development. Abstract N6-methyladenosine (m6A) plays an important role in regulating gene expression. Previous studies found that m6A methylation affects skeletal muscle development. However, the effect of m6A methylases on bovine skeletal myogenesis is still unclear. Here, we found that the expression of m6A demethylases (FTO and ALKBH5) was significantly higher in the longissimus dorsi muscle of adult cattle than in newborn cattle. In contrast, the expression of m6A methyltransferases (METTL3, METTL14 and WTAP) was reduced. The mRNA expression of all five genes was found to be increased during the myogenesis of myoblasts in vitro. Knockdown of FTO or METTL3 promoted myoblast proliferation, inhibited myoblast apoptosis and suppressed myogenic differentiation, whereas ALKBH5 knockdown had the opposite effect. METTL14 knockdown enhanced myoblast proliferation and impaired myogenic differentiation. WTAP knockdown attenuated proliferation and contributed to apoptosis but did not affect differentiation. Furthermore, the functional domains of these five m6A methylases are conserved across species. Our results suggest that m6A methylases are involved in regulating skeletal muscle development and that there may be a complex network of m6A methylation regulating skeletal myogenesis.
Collapse
|
125
|
Cai B, Ma M, Zhang J, Wang Z, Kong S, Zhou Z, Lian L, Zhang J, Li J, Wang Y, Li H, Zhang X, Nie Q. LncEDCH1 improves mitochondrial function to reduce muscle atrophy by interacting with SERCA2. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:319-334. [PMID: 35024244 PMCID: PMC8717430 DOI: 10.1016/j.omtn.2021.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 12/07/2021] [Indexed: 12/25/2022]
Abstract
Skeletal muscle is a regulator of the body's energy expenditure and metabolism. Abnormal regulation of skeletal muscle-specific genes leads to various muscle diseases. Long non-coding RNAs (lncRNAs) have been demonstrated to play important roles in muscle growth and muscle atrophy. To explore the potential function of muscle-associated lncRNA, we analyzed our previous RNA-sequencing data and selected the lncRNA (LncEDCH1) as the research object. In this study, we report that LncEDCH1 is specifically enriched in skeletal muscle, and its transcriptional activity is positively regulated by transcription factor SP1. LncEDCH1 regulates myoblast proliferation and differentiation in vitro. In vivo, LncEDCH1 reduces intramuscular fat deposition, activates slow-twitch muscle phenotype, and inhibits muscle atrophy. Mechanistically, LncEDCH1 binds to sarcoplasmic/ER calcium ATPase 2 (SERCA2) protein to enhance SERCA2 protein stability and increase SERCA2 activity. Meanwhile, LncEDCH1 improves mitochondrial efficiency possibly through a SERCA2-mediated activation of the AMPK pathway. Our findings provide a strategy for using LncEDCH1 as an effective regulator for the treatment of muscle atrophy and energy metabolism.
Collapse
Affiliation(s)
- Bolin Cai
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Manting Ma
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Jing Zhang
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Zhijun Wang
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Shaofen Kong
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Zhen Zhou
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Ling Lian
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jiannan Zhang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China
| | - Juan Li
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China
| | - Yajun Wang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China
| | - Hongmei Li
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Xiquan Zhang
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Qinghua Nie
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| |
Collapse
|
126
|
Howard JJ, Graham K, Shortland AP. Understanding skeletal muscle in cerebral palsy: a path to personalized medicine? Dev Med Child Neurol 2022; 64:289-295. [PMID: 34499350 DOI: 10.1111/dmcn.15018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/02/2021] [Accepted: 07/02/2021] [Indexed: 12/11/2022]
Abstract
Until recently, there has been little interest in understanding the intrinsic features associated with the pathomorphology of skeletal muscle in cerebral palsy (CP). Coupled with emerging evidence that challenges the role of spasticity as a determinant of gross motor function and in the development of fixed muscle contractures, it has become increasingly important to further elucidate the underlying mechanisms responsible for muscle alterations in CP. This knowledge can help clinicians to understand and apply treatment modalities that take these aspects into account. Thus, the inherent heterogeneity of the CP phenotype allows for the potential of personalized medicine through the understanding of muscle pathomorphology on an individual basis and tailoring treatment approaches accordingly. This review aims to summarize recent developments in the understanding of CP muscle and their relationship to musculoskeletal manifestations, in addition to proposing a treatment paradigm that incorporates this new knowledge.
Collapse
Affiliation(s)
- Jason J Howard
- Department of Orthopaedic Surgery, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Kerr Graham
- Department of Orthopaedic Surgery, University of Melbourne, Melbourne, Victoria, Australia.,Hugh Williamson Gait Laboratory, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Adam P Shortland
- Paediatric Neurosciences, Guy's and St Thomas' Foundation NHS Trust, Evelina Children's Hospital, London, UK
| |
Collapse
|
127
|
Chang YB, Ahn Y, Suh HJ, Jo K. Yeast hydrolysate ameliorates dexamethasone-induced muscle atrophy by suppressing MuRF-1 expression in C2C12 cells and C57BL/6 mice. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.104985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
128
|
Attwaters M, Hughes SM. Cellular and molecular pathways controlling muscle size in response to exercise. FEBS J 2022; 289:1428-1456. [PMID: 33755332 DOI: 10.1111/febs.15820] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/27/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022]
Abstract
From the discovery of ATP and motor proteins to synaptic neurotransmitters and growth factor control of cell differentiation, skeletal muscle has provided an extreme model system in which to understand aspects of tissue function. Muscle is one of the few tissues that can undergo both increase and decrease in size during everyday life. Muscle size depends on its contractile activity, but the precise cellular and molecular pathway(s) by which the activity stimulus influences muscle size and strength remain unclear. Four correlates of muscle contraction could, in theory, regulate muscle growth: nerve-derived signals, cytoplasmic calcium dynamics, the rate of ATP consumption and physical force. Here, we summarise the evidence for and against each stimulus and what is known or remains unclear concerning their molecular signal transduction pathways and cellular effects. Skeletal muscle can grow in three ways, by generation of new syncytial fibres, addition of nuclei from muscle stem cells to existing fibres or increase in cytoplasmic volume/nucleus. Evidence suggests the latter two processes contribute to exercise-induced growth. Fibre growth requires increase in sarcolemmal surface area and cytoplasmic volume at different rates. It has long been known that high-force exercise is a particularly effective growth stimulus, but how this stimulus is sensed and drives coordinated growth that is appropriately scaled across organelles remains a mystery.
Collapse
Affiliation(s)
- Michael Attwaters
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, UK
| | - Simon M Hughes
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, UK
| |
Collapse
|
129
|
Zeng C, Shi H, Kirkpatrick LT, Ricome A, Park S, Scheffler JM, Hannon KM, Grant AL, Gerrard DE. Driving an Oxidative Phenotype Protects Myh4 Null Mice From Myofiber Loss During Postnatal Growth. Front Physiol 2022; 12:785151. [PMID: 35283757 PMCID: PMC8908108 DOI: 10.3389/fphys.2021.785151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/28/2021] [Indexed: 12/17/2022] Open
Abstract
Postnatal muscle growth is accompanied by increases in fast fiber type compositions and hypertrophy, raising the possibility that a slow to fast transition may be partially requisite for increases in muscle mass. To test this hypothesis, we ablated the Myh4 gene, and thus myosin heavy chain IIB protein and corresponding fibers in mice, and examined its consequences on postnatal muscle growth. Wild-type and Myh4–/– mice had the same number of muscle fibers at 2 weeks postnatal. However, the gastrocnemius muscle lost up to 50% of its fibers between 2 and 4 weeks of age, though stabilizing thereafter. To compensate for the lack of functional IIB fibers, type I, IIA, and IIX(D) fibers increased in prevalence and size. To address whether slowing the slow-to-fast fiber transition process would rescue fiber loss in Myh4–/– mice, we stimulated the oxidative program in muscle of Myh4–/– mice either by overexpression of PGC-1α, a well-established model for fast-to-slow fiber transition, or by feeding mice AICAR, a potent AMP kinase agonist. Forcing an oxidative metabolism in muscle only partially protected the gastrocnemius muscle from loss of fibers in Myh4–/– mice. To explore whether traditional means of stimulating muscle hypertrophy could overcome the muscling deficits in postnatal Myh4–/– mice, myostatin null mice were bred with Myh4–/– mice, or Myh4–/– mice were fed the growth promotant clenbuterol. Interestingly, both genetic and pharmacological stimulations had little impact on mice lacking a functional Myh4 gene suggesting that the existing muscle fibers have maximized its capacity to enlarge to compensate for the lack of its neighboring IIB fibers. Curiously, however, cell signaling events responsible for IIB fiber formation remained intact in the tissue. These findings further show disrupting the slow-to-fast transition of muscle fibers compromises muscle growth postnatally and suggest that type IIB myosin heavy chain expression and its corresponding fiber type may be necessary for fiber maintenance, transition and hypertrophy in mice. The fact that forcing muscle metabolism toward a more oxidative phenotype can partially compensates for the lack of an intact Myh4 gene provides new avenues for attenuating the loss of fast-twitch fibers in aged or diseased muscles.
Collapse
Affiliation(s)
- Caiyun Zeng
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Hao Shi
- Meat Science and Muscle Biology Research Group, Virginia Tech, Department of Animal and Poultry Sciences, Blacksburg, VA, United States
| | - Laila T. Kirkpatrick
- Meat Science and Muscle Biology Research Group, Virginia Tech, Department of Animal and Poultry Sciences, Blacksburg, VA, United States
| | - Aymeric Ricome
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Sungkwon Park
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Jason M. Scheffler
- Meat Science and Muscle Biology Research Group, Virginia Tech, Department of Animal and Poultry Sciences, Blacksburg, VA, United States
| | - Kevin M. Hannon
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States
| | - Alan L. Grant
- Meat Science and Muscle Biology Research Group, Virginia Tech, Department of Animal and Poultry Sciences, Blacksburg, VA, United States
| | - David E. Gerrard
- Meat Science and Muscle Biology Research Group, Virginia Tech, Department of Animal and Poultry Sciences, Blacksburg, VA, United States
- *Correspondence: David E. Gerrard,
| |
Collapse
|
130
|
Luo M, Yang H, Wu D, You X, Huang S, Song Y. Tent5a modulates muscle fiber formation in adolescent idiopathic scoliosis via maintenance of myogenin expression. Cell Prolif 2022; 55:e13183. [PMID: 35137485 PMCID: PMC8891553 DOI: 10.1111/cpr.13183] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 01/04/2021] [Accepted: 01/02/2022] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Paravertebral muscle asymmetry may be involved in the pathogenesis of adolescent idiopathic scoliosis (AIS), and the Tent5a protein was recently identified as a novel active noncanonical poly(A) polymerase. We, therefore, explored the function of the AIS susceptibility gene Tent5a in myoblasts. MATERIALS AND METHODS RNA-seq of AIS paravertebral muscle was performed, and the molecular differences in paravertebral muscle were investigated. Twenty-four AIS susceptibility genes were screened, and differential expression of Tent5a in paravertebral muscles was confirmed with qPCR and Western blot. After the knockdown of Tent5a, the functional effects of Tent5a on C2C12 cell proliferation, migration, and apoptosis were detected by Cell Counting Kit-8 assay, wound-healing assay, and TUNEL assay, respectively. Myogenic differentiation markers were tested with immunofluorescence and qPCR in vitro, and muscle fiber formation was compared in vivo. RESULTS The AIS susceptibility gene Tent5a was differentially expressed in AIS paravertebral muscles. Tent5a knockdown inhibited the proliferation and migration of C2C12 cells and inhibited the maturation of type I muscle fibers in vitro and in vivo. Mechanistically, the expression of myogenin was decreased along with the suppression of Tent5a. CONCLUSIONS Tent5a plays an important role in the proliferation and migration of myoblasts, and it regulates muscle fiber maturation by maintaining the stability of myogenin. Tent5a may be involved in the pathogenesis of AIS by regulating the formation of muscle fiber type I.
Collapse
Affiliation(s)
- Ming Luo
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.,Department of Orthopedics, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Huiliang Yang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Diwei Wu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Xuanhe You
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Shishu Huang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Yueming Song
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
131
|
Noncoding RNAs-associated ceRNA networks involved in the amelioration of skeletal muscle aging after whey protein supplementation. J Nutr Biochem 2022; 104:108968. [DOI: 10.1016/j.jnutbio.2022.108968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 01/06/2022] [Accepted: 01/19/2022] [Indexed: 11/23/2022]
|
132
|
Biogenesis and Function of Extracellular Vesicles in Pathophysiological Processes Skeletal Muscle Atrophy. Biochem Pharmacol 2022; 198:114954. [DOI: 10.1016/j.bcp.2022.114954] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/08/2022] [Accepted: 02/08/2022] [Indexed: 12/13/2022]
|
133
|
Yoshida S, Fujimoto T, Takahashi T, Sugimoto K, Akasaka H, Tanaka M, Huang Y, Yasunobe Y, Xie K, Ohnishi Y, Minami T, Takami Y, Yamamoto K, Rakugi H. IL-15RA regulates IL-15 localization and protein expression in skeletal muscle cells. Exp Physiol 2022; 107:222-232. [PMID: 35100657 DOI: 10.1113/ep090205] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/24/2022] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? How are the dynamics of IL-15 and its receptors altered during the differentiation of myoblasts into myotubes, and how is IL-15 regulated? What is the main finding and its importance? ABSTRACT Interleukin-15 (IL-15) is a myokine in the Interleukin-2 (IL-2) family that is generated in the skeletal muscle during exercise. The functional effect of IL-15 involves muscle regeneration and metabolic regulation in skeletal muscle. Reports have indicated that the mechanism of Interleukin-15 receptor subunit alpha (IL-15RA) regulates IL-15 localization in immune cells. However, the dynamic of IL-15 and its receptors, which regulate the IL-15 pathway in skeletal muscle differentiation, have not yet been clarified. This study investigated the mechanism of IL-15 regulation using a mouse skeletal muscle cell line, C2C12 cells. We found that the mRNA expression of IL-15, Interleukin 2 Receptor Subunit Beta (IL-2RB) (CD122), and Interleukin 2 Receptor Subunit Gamma (IL-2RG) (CD132) increased, but that IL-15RA exhibits different kinetics as differentiation progresses. We also found that IL-15, mainly localized in the cytosol, preassembled with IL-15RA in the cytosol and fused to the plasma membrane. Moreover, IL-15RA increased IL-15 protein levels. Our findings suggest that genes comprising the IL-15 signaling complex are enhanced with the differentiation of myotubes and that IL-15RA regulates the protein kinetics of IL-15 signaling in skeletal muscle. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Shino Yoshida
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Taku Fujimoto
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.,Institute for Biogenesis Research, Department of Anatomy Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, 96813, USA
| | - Toshimasa Takahashi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Ken Sugimoto
- Department of General and Geriatric Medicine, Kawasaki Medical University, Okayama, 700-8505, Japan
| | - Hiroshi Akasaka
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Minoru Tanaka
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.,Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Kobe, Hyogo, 654-0142, Japan.,Department of Rehabilitation Science, Osaka Health Science University, Osaka, 530-0043, Japan
| | - Yibin Huang
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Yukiko Yasunobe
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Keyu Xie
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Yuri Ohnishi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Tomohiro Minami
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Yoichi Takami
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Koichi Yamamoto
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
134
|
Luo Q, Yu Y, Lan X. SIGNET: single-cell RNA-seq-based gene regulatory network prediction using multiple-layer perceptron bagging. Brief Bioinform 2022; 23:bbab547. [PMID: 34962260 PMCID: PMC8769917 DOI: 10.1093/bib/bbab547] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/13/2021] [Accepted: 11/25/2021] [Indexed: 11/17/2022] Open
Abstract
High-throughput single-cell RNA-seq data have provided unprecedented opportunities for deciphering the regulatory interactions among genes. However, such interactions are complex and often nonlinear or nonmonotonic, which makes their inference using linear models challenging. We present SIGNET, a deep learning-based framework for capturing complex regulatory relationships between genes under the assumption that the expression levels of transcription factors participating in gene regulation are strong predictors of the expression of their target genes. Evaluations based on a variety of real and simulated scRNA-seq datasets showed that SIGNET is more sensitive to ChIP-seq validated regulatory interactions in different types of cells, particularly rare cells. Therefore, this process is more effective for various downstream analyses, such as cell clustering and gene regulatory network inference. We demonstrated that SIGNET is a useful tool for identifying important regulatory modules driving various biological processes.
Collapse
Affiliation(s)
- Qinhuan Luo
- School of Medicine, Tsinghua University, Beijing, China
| | - Yongzhen Yu
- School of Medicine, Tsinghua University, Beijing, China
| | - Xun Lan
- School of Medicine,and the Tsinghua-Peking Center for Life science, MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing, China
| |
Collapse
|
135
|
Chen MM, Zhao YP, Zhao Y, Deng SL, Yu K. Regulation of Myostatin on the Growth and Development of Skeletal Muscle. Front Cell Dev Biol 2022; 9:785712. [PMID: 35004684 PMCID: PMC8740192 DOI: 10.3389/fcell.2021.785712] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/08/2021] [Indexed: 01/01/2023] Open
Abstract
Myostatin (MSTN), a member of the transforming growth factor-β superfamily, can negatively regulate the growth and development of skeletal muscle by autocrine or paracrine signaling. Mutation of the myostatin gene under artificial or natural conditions can lead to a significant increase in muscle quality and produce a double-muscle phenotype. Here, we review the similarities and differences between myostatin and other members of the transforming growth factor-β superfamily and the mechanisms of myostatin self-regulation. In addition, we focus extensively on the regulation of myostatin functions involved in myogenic differentiation, myofiber type conversion, and skeletal muscle protein synthesis and degradation. Also, we summarize the induction of reactive oxygen species generation and oxidative stress by myostatin in skeletal muscle. This review of recent insights into the function of myostatin will provide reference information for future studies of myostatin-regulated skeletal muscle formation and may have relevance to agricultural fields of study.
Collapse
Affiliation(s)
- Ming-Ming Chen
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yi-Ping Zhao
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Yue Zhao
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shou-Long Deng
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Kun Yu
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
136
|
Zhou X, Yan Q, Liu L, Chen G, Tang S, He Z, Tan Z. Maternal undernutrition alters the skeletal muscle development and methylation of myogenic factors in goat offspring. Anim Biosci 2022; 35:847-857. [PMID: 34991223 PMCID: PMC9066034 DOI: 10.5713/ab.21.0285] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/08/2021] [Indexed: 11/27/2022] Open
Abstract
Objective The effects of maternal undernutrition during midgestation on muscle fiber histology, myosin heavy chain (MyHC) expression, methylation modification of myogenic factors, and the mammalian target of rapamycin (mTOR) signaling pathway in the skeletal muscles of prenatal and postnatal goats were examined. Methods Twenty-four pregnant goats were assigned to a control (100% of the nutrients requirement, n = 12) or a restricted group (60% of the nutrients requirement, n = 12) between 45 and 100 days of gestation. Descendants were harvested at day 100 of gestation and at day 90 after birth to collect the femoris muscle tissue. Results Maternal undernutrition increased (p<0.05) the fiber area of the vastus muscle in the fetuses and enhanced (p<0.01) the proportions of MyHCI and MyHCIIA fibers in offspring, while the proportion of MyHCIIX fibers was decreased (p<0.01). DNA methylation at the +530 cytosine-guanine dinucleotide (CpG) site of the myogenic factor 5 (MYF5) promoter in restricted fetuses was increased (p<0.05), but the methylation of the MYF5 gene at the +274,280 CpG site and of the myogenic differentiation (MYOD) gene at the +252 CpG site in restricted kids was reduced (p<0.05). mTOR protein signals were downregulated (p<0.05) in the restricted offspring. Conclusion Maternal undernutrition altered the muscle fiber type in offspring, but its relationship with methylation in the promoter regions of myogenic genes needs to be elucidated.
Collapse
Affiliation(s)
- Xiaoling Zhou
- College of Animal Science, Tarim University, Alaer 843300, China.,Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha 410125, China
| | - Qiongxian Yan
- Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha 410125, China.,Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha 410128, China
| | - Liling Liu
- College of Animal Science, Tarim University, Alaer 843300, China
| | - Genyuan Chen
- College of Animal Science, Tarim University, Alaer 843300, China
| | - Shaoxun Tang
- Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha 410125, China.,University of Chinese Academy of Science, Beijing 100049, China
| | - Zhixiong He
- Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha 410125, China.,Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha 410128, China
| | - Zhiliang Tan
- Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha 410125, China.,Hunan Co-Innovation Center of Animal Production Safety, CICAPS, Changsha 410128, China
| |
Collapse
|
137
|
Ipulan-Colet LA. Sexual dimorphism through androgen signaling; from external genitalia to muscles. Front Endocrinol (Lausanne) 2022; 13:940229. [PMID: 35983512 PMCID: PMC9379613 DOI: 10.3389/fendo.2022.940229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Sexual dimorphisms can be seen in many organisms with some exhibiting subtle differences while some can be very evident. The difference between male and female can be seen on the morphological level such as discrepancies in body mass, presence of body hair in distinct places, or through the presence of specific reproductive structures. It is known that the development of the reproductive structures is governed by hormone signaling, most commonly explained through the actions of androgen signaling. The developmental program of the male and female external genitalia involves a common anlage, the genital tubercle or GT, that later on develop into a penis and clitoris, respectively. Androgen signaling involvement can be seen in the different tissues in the GT that express Androgen receptor and the different genes that are regulated by androgen in the mesenchyme and endoderm component of the GT. Muscles are also known to be responsive to androgen signaling with male and female muscles exhibiting different capabilities. However, the occurrence of sexual dimorphism in muscle development is unclear. In this minireview, a summary on the role of androgen in the sexually dimorphic development of the genital tubercle was provided. This was used as a framework on analyzing the different mechanism employed by androgen signaling to regulate the sexual dimorphism in muscle development.
Collapse
|
138
|
Cheng J, Wang L, Wang S, Chen R, Zhang T, Ma H, Lu H, Yuan G. Transcriptomic analysis of thigh muscle of Lueyang black-bone chicken in free-range and caged feeding. Anim Biotechnol 2021:1-11. [PMID: 34965837 DOI: 10.1080/10495398.2021.1993235] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Lueyang black-bone chicken is free-range in hilly areas and has unique genetic characteristics and excellent muscle quality. However, the molecular mechanisms of breeding mode influence growth and meat quality in Lueyang black-bone chicken are still unclear. Here we analyzed the meat quality and transcriptome data of thigh muscle by comparing free-range and caged modes at the age of 60 and 120 days in Lueyang black-boned chicken. The results demonstrated that the free-range mode could improve the pH value, tenderness, and reducing the hardness of the thigh muscle. Intramuscular fat (IMF) content of the thigh muscle was markedly higher in the caged chickens compared with free-range animals at the age of 60 days. Functional pathway analysis illustrated that tight junction signaling was associated with the formation of slow-twitch fibers in free-range chickens at age of 120 days. All research data proved that the free-range mode could improve muscle quality by promoting the formation of slow-twitch fibers and IMF in thigh muscle in Lueyang black-bone chicken. Based on the animal benefit and healthy, the free-range feeding should be considered during the breeding process of broiler chicken. The results provide good knowledge of the functional molecular mechanisms associated with muscle quality in Lueyang black-bone chicken.
Collapse
Affiliation(s)
- Jia Cheng
- School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, China
| | - Ling Wang
- School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, China
| | - Shanshan Wang
- School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, China
| | - Rui Chen
- School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, China
| | - Tao Zhang
- School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, China
| | - Haidong Ma
- School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, China
| | - Hongzhao Lu
- School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, China
| | - Guoqiang Yuan
- Shaanxi Baiweiyuan Network Technology Co., Ltd, Hanzhong, China
| |
Collapse
|
139
|
Yang X, Wang J, Ma X, Du J, Mei C, Zan L. Transcriptome-wide N 6-Methyladenosine Methylome Profiling Reveals m 6A Regulation of Skeletal Myoblast Differentiation in Cattle ( Bos taurus). Front Cell Dev Biol 2021; 9:785380. [PMID: 34938736 PMCID: PMC8685427 DOI: 10.3389/fcell.2021.785380] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/12/2021] [Indexed: 12/28/2022] Open
Abstract
N 6 -methyladenosine (m6A) is the most prevalent methylation modification of eukaryotic mRNA, and it plays an important role in regulating gene expression. Previous studies have found that m6A methylation plays a role in mammalian skeletal muscle development. However, the effect of m6A on bovine skeletal myogenesis are still unclear. Here, we selected proliferating myoblasts (GM) and differentiated myotubes (on the 4th day of differentiation, DM) for m6A-seq and RNA-seq to explore the m6A methylation modification pattern during bovine skeletal myogenesis. m6A-seq analysis revealed that m6A methylation was an abundant modification of the mRNA in bovine myoblasts and myotubes. We scanned 5,691-8,094 m6A-modified transcripts, including 1,437 differentially methylated genes (DMGs). GO and KEGG analyses revealed that DMGs were primarily involved in transcriptional regulation and RNA metabolism, as well as insulin resistance and metabolic pathways related to muscle development. The combined analysis further identified 268 genes that had significant changes at both m6A and mRNA levels, suggesting that m6A modification may regulate myoblast differentiation by mediating the expression of these genes. Furthermore, we experimentally confirmed four genes related to myogenesis, including MYOZ2, TWIST1, KLF5 and MYOD1, with differential changes in both m6A and mRNA levels during bovine myoblast differentiation, indicating that they can be potential candidate targets for m6A regulation of skeletal myogenesis. Our results may provide new insight into molecular genetics and breeding of beef cattle, and provide a reference for investigating the mechanism of m6A regulating skeletal muscle development.
Collapse
Affiliation(s)
- Xinran Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Jianfang Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xinhao Ma
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Jiawei Du
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chugang Mei
- College of Animal Science and Technology, Northwest A&F University, Yangling, China.,National Beef Cattle Improvement Center, Northwest A&F University, Yangling, China
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, China.,National Beef Cattle Improvement Center, Northwest A&F University, Yangling, China
| |
Collapse
|
140
|
Genome-wide identification of enhancers and transcription factors regulating the myogenic differentiation of bovine satellite cells. BMC Genomics 2021; 22:901. [PMID: 34915843 PMCID: PMC8675486 DOI: 10.1186/s12864-021-08224-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/29/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Satellite cells are the myogenic precursor cells in adult skeletal muscle. The objective of this study was to identify enhancers and transcription factors that regulate gene expression during the differentiation of bovine satellite cells into myotubes. RESULTS Chromatin immunoprecipitation followed by deep sequencing (ChIP-seq) was performed to identify genomic regions where lysine 27 of H3 histone is acetylated (H3K27ac), i.e., active enhancers, from bovine satellite cells before and during differentiation into myotubes. A total of 19,027 and 47,669 H3K27ac-marked enhancers were consistently identified from two biological replicates of before- and during-differentiation bovine satellite cells, respectively. Of these enhancers, 5882 were specific to before-differentiation, 35,723 to during-differentiation, and 13,199 common to before- and during-differentiation bovine satellite cells. Whereas most of the before- or during-differentiation-specific H3K27ac-marked enhancers were located distally to the transcription start site, the enhancers common to before- and during-differentiation were located both distally and proximally to the transcription start site. The three sets of H3K27ac-marked enhancers were associated with functionally different genes and enriched with different transcription factor binding sites. Specifically, many of the H3K27ac-marked enhancers specific to during-differentiation bovine satellite cells were associated with genes involved in muscle structure and development, and were enriched with binding sites for the MyoD, AP-1, KLF, TEAD, and MEF2 families of transcription factors. A positive role was validated for Fos and FosB, two AP-1 family transcription factors, in the differentiation of bovine satellite cells into myotubes by siRNA-mediated knockdown. CONCLUSIONS Tens of thousands of H3K27ac-marked active enhancers have been identified from bovine satellite cells before or during differentiation. These enhancers contain binding sites not only for transcription factors whose role in satellite cell differentiation is well known but also for transcription factors whose role in satellite cell differentiation is unknown. These enhancers and transcription factors are valuable resources for understanding the complex mechanism that mediates gene expression during satellite cell differentiation. Because satellite cell differentiation is a key step in skeletal muscle growth, the enhancers, the transcription factors, and their target genes identified in this study are also valuable resources for identifying and interpreting skeletal muscle trait-associated DNA variants in cattle.
Collapse
|
141
|
Liao Y, Li D, Zhou X, Peng Z, Meng Z, Liu R, Yang W. Pyruvate Might Bridge Gut Microbiota and Muscle Health in Aging Mice After Chronic High Dose of Leucine Supplementation. Front Med (Lausanne) 2021; 8:755803. [PMID: 34881260 PMCID: PMC8645596 DOI: 10.3389/fmed.2021.755803] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/25/2021] [Indexed: 12/23/2022] Open
Abstract
Background: The previous studies demonstrated that there might be complex and close relationships among leucine supplementation, gut microbiota, and muscle health, which still needs further investigation. Aims: This study aimed to explore the associations of gut microbiota with muscle health after leucine intake. Methods: In this study, 19-month-old male C57BL/6j mice (n = 12/group) were supplemented with ultrapure water, low dose of leucine (500 mg/kg·d), and high dose of leucine (1,250 mg/kg·d) for 12 weeks by oral gavage. The mice fecal samples in each group before and after supplementation were collected for baseline and endpoint gut microbiota analysis by using 16S rDNA amplicon sequencing. Meanwhile, ultrasound measurement, H&E staining, myofiber cross-sectional area (CSA) measurement, and western blotting were performed in the quadriceps subsequently. The pyruvate levels were detected in feces. Results: Improvement in muscle of histology and ultrasonography were observed after both low and high dose of leucine supplementation. High dose of leucine supplementation could promote skeletal muscle health in aging mice via regulating AMPKα/SIRT1/PGC-1α. The richness and diversities of microbiota as well as enriched metabolic pathways were altered after leucine supplementation. Firmicutes-Bacteroidetes ratio was significantly decreased in high-leucine group. Moreover, pyruvate fermentation to propanoate I were negatively associated with differential species and the pyruvate levels were significantly increased in feces after high dose of leucine supplementation. Conclusions: Chronic high dose of leucine supplementation changed gut microbiota composition and increased pyruvate levels in the feces, which possibly provides a novel direction for promoting muscle health in aging mice.
Collapse
Affiliation(s)
- Yuxiao Liao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaolei Zhou
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zitong Meng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Liu
- Department of Preventive Medicine, School of Medicine, Jianghan University, Wuhan, China
| | - Wei Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
142
|
Rodrigues GDA, Júnior DTV, Soares MH, da Silva CB, Fialho FA, Barbosa LMDR, Neves MM, Rocha GC, Duarte MDS, Saraiva A. L-Arginine Supplementation for Nulliparous Sows during the Last Third of Gestation. Animals (Basel) 2021; 11:ani11123476. [PMID: 34944251 PMCID: PMC8698042 DOI: 10.3390/ani11123476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/30/2021] [Accepted: 12/03/2021] [Indexed: 11/16/2022] Open
Abstract
We evaluated the effects of L-arginine supplementation during the last third of gestation on molecular mechanisms related to skeletal muscle development of piglets and litter traits at birth. Twenty-three nulliparous sows averaging 205.37 ± 11.50 kg of body weight were randomly assigned to the following experimental treatments: control (CON), where pregnant sows were fed diets to meet their nutritional requirements; arginine (ARG), where sows where fed CON + 1.0% L-arginine. Skeletal muscle from piglets born from sows from ARG group had greater mRNA expression of MYOD (p = 0.043) and MYOG (p ≤ 0.01), and tended to present greater mRNA expression (p = 0.06) of IGF-2 gene compared to those born from CON sows. However, there were no differences (p > 0.05) in the histomorphometric variables of fetuses' skeletal muscle. The total weight of born piglets, total weight of born alive piglets, piglet weight at birth, coefficient of variation of birth weight, and the incidence of intrauterine growth restriction (IUGR) piglets did not differ between groups. No stillborn piglets (p < 0.01) were verified in the ARG sows compared to CON group. The blood levels of estradiol (p = 0.035) and urea (p = 0.03) were higher in ARG sows compared to those from the CON group. In summary, our data show that arginine supplementation of nulliparous sows at late gestation enhance mRNA expression of key myogenic regulatory factors, which likely contribute to improve animal growth rates in later stages of development.
Collapse
Affiliation(s)
- Gustavo de Amorim Rodrigues
- Department of Animal Sciences, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (G.d.A.R.); (D.T.V.J.); (M.H.S.); (C.B.d.S.); (F.A.F.); (L.M.d.R.B.); (M.M.N.); (G.C.R.)
- Muscle Biology and Nutrigenomics Laboratory, Department of Animal Sciences, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil
| | - Dante Teixeira Valente Júnior
- Department of Animal Sciences, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (G.d.A.R.); (D.T.V.J.); (M.H.S.); (C.B.d.S.); (F.A.F.); (L.M.d.R.B.); (M.M.N.); (G.C.R.)
- Muscle Biology and Nutrigenomics Laboratory, Department of Animal Sciences, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil
| | - Marcos Henrique Soares
- Department of Animal Sciences, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (G.d.A.R.); (D.T.V.J.); (M.H.S.); (C.B.d.S.); (F.A.F.); (L.M.d.R.B.); (M.M.N.); (G.C.R.)
- Muscle Biology and Nutrigenomics Laboratory, Department of Animal Sciences, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil
| | - Caroline Brito da Silva
- Department of Animal Sciences, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (G.d.A.R.); (D.T.V.J.); (M.H.S.); (C.B.d.S.); (F.A.F.); (L.M.d.R.B.); (M.M.N.); (G.C.R.)
- Muscle Biology and Nutrigenomics Laboratory, Department of Animal Sciences, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil
| | - Fernanda Abranches Fialho
- Department of Animal Sciences, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (G.d.A.R.); (D.T.V.J.); (M.H.S.); (C.B.d.S.); (F.A.F.); (L.M.d.R.B.); (M.M.N.); (G.C.R.)
- Muscle Biology and Nutrigenomics Laboratory, Department of Animal Sciences, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil
| | - Lívia Maria dos Reis Barbosa
- Department of Animal Sciences, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (G.d.A.R.); (D.T.V.J.); (M.H.S.); (C.B.d.S.); (F.A.F.); (L.M.d.R.B.); (M.M.N.); (G.C.R.)
| | - Mariana Machado Neves
- Department of Animal Sciences, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (G.d.A.R.); (D.T.V.J.); (M.H.S.); (C.B.d.S.); (F.A.F.); (L.M.d.R.B.); (M.M.N.); (G.C.R.)
- Structural Biology Laboratory, Department of Animal Sciences, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil
| | - Gabriel Cipriano Rocha
- Department of Animal Sciences, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (G.d.A.R.); (D.T.V.J.); (M.H.S.); (C.B.d.S.); (F.A.F.); (L.M.d.R.B.); (M.M.N.); (G.C.R.)
- Muscle Biology and Nutrigenomics Laboratory, Department of Animal Sciences, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil
| | | | - Alysson Saraiva
- Department of Animal Sciences, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (G.d.A.R.); (D.T.V.J.); (M.H.S.); (C.B.d.S.); (F.A.F.); (L.M.d.R.B.); (M.M.N.); (G.C.R.)
- Muscle Biology and Nutrigenomics Laboratory, Department of Animal Sciences, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil
- Correspondence:
| |
Collapse
|
143
|
Choi HK, Kim CH, Lee SN, Kim TH, Oh BK. Nano-sized graphene oxide coated nanopillars on microgroove polymer arrays that enhance skeletal muscle cell differentiation. NANO CONVERGENCE 2021; 8:40. [PMID: 34862954 PMCID: PMC8643291 DOI: 10.1186/s40580-021-00291-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 11/22/2021] [Indexed: 05/04/2023]
Abstract
The degeneration or loss of skeletal muscles, which can be caused by traumatic injury or disease, impacts most aspects of human activity. Among various techniques reported to regenerate skeletal muscle tissue, controlling the external cellular environment has been proven effective in guiding muscle differentiation. In this study, we report a nano-sized graphene oxide (sGO)-modified nanopillars on microgroove hybrid polymer array (NMPA) that effectively controls skeletal muscle cell differentiation. sGO-coated NMPA (sG-NMPA) were first fabricated by sequential laser interference lithography and microcontact printing methods. To compensate for the low adhesion property of polydimethylsiloxane (PDMS) used in this study, graphene oxide (GO), a proven cytophilic nanomaterial, was further modified. Among various sizes of GO, sGO (< 10 nm) was found to be the most effective not only for coating the surface of the NM structure but also for enhancing the cell adhesion and spreading on the fabricated substrates. Remarkably, owing to the micro-sized line patterns that guide cellular morphology to an elongated shape and because of the presence of sGO-modified nanostructures, mouse myoblast cells (C2C12) were efficiently differentiated into skeletal muscle cells on the hybrid patterns, based on the myosin heavy chain expression levels. Therefore, the developed sGO coated polymeric hybrid pattern arrays can serve as a potential platform for rapid and highly efficient in vitro muscle cell generation.
Collapse
Affiliation(s)
- Hye Kyu Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04170, South Korea
| | - Cheol-Hwi Kim
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Korea
| | | | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Korea.
| | - Byung-Keun Oh
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04170, South Korea.
| |
Collapse
|
144
|
Liu D, Xia J, Yang Z, Zhao X, Li J, Hao W, Yang X. Identification of Chimeric RNAs in Pig Skeletal Muscle and Transcriptomic Analysis of Chimeric RNA TNNI2-ACTA1 V1. Front Vet Sci 2021; 8:742593. [PMID: 34778431 PMCID: PMC8578878 DOI: 10.3389/fvets.2021.742593] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/27/2021] [Indexed: 12/11/2022] Open
Abstract
Chimeric RNA was considered a special marker of cancer. However, recent studies have demonstrated that chimeric RNAs also exist in non-cancerous cells and tissues. Here, we analyzed and predicted jointly 49 chimeric RNAs by Star-Fusion and FusionMap. One chimeric RNA, we named TNNI2-ACTA1, and its eight transcript variants were identified by reverse transcriptase–polymerase chain reaction. The overexpression of TNNI2-ACTA1 V1 inhibited the proliferation of porcine skeletal muscle satellite cells through down-regulating the mRNA expression levels of cell cycle–related genes cyclinD1. However, as parental genes, there is no such effect in the TNNI2 and ACTA1. To explore the underlying mechanism for this phenomenon, we used RNA-seq to profile the transcriptomes of PSCs with overexpression. Compared with the negative control group, 1,592 differentially expressed genes (DEGs) were upregulated and 1,077 DEGs downregulated in TNNI2 group; 1,226 DEGs were upregulated and 902 DEGs downregulated in ACTA1 group; and 13 DEGs were upregulated and 16 DEGs downregulated in TNNI2-ACTA1 V1 group, respectively. Compared with the parental gene groups, three specific genes were enriched in the TNNI2-ACTA1 V1 group (NCOA3, Radixin, and DDR2). These three genes may be the key to TNNI2-ACTA1 V1 regulating cell proliferation. Taken together, our study explores the role of chimeric RNAs in normal tissues. In addition, our study as the first research provides the foundation for the mechanism of chimeric RNAs regulating porcine skeletal muscle growth.
Collapse
Affiliation(s)
- Dongyu Liu
- College of Animal Sciences and Technology, Northeast Agricultural University, Harbin, China
| | - Jiqiao Xia
- College of Animal Sciences and Technology, Northeast Agricultural University, Harbin, China
| | - Zewei Yang
- College of Animal Sciences and Technology, Northeast Agricultural University, Harbin, China
| | - Xuelian Zhao
- College of Animal Sciences and Technology, Northeast Agricultural University, Harbin, China
| | - Jiaxin Li
- College of Animal Sciences and Technology, Northeast Agricultural University, Harbin, China
| | - Wanjun Hao
- College of Animal Sciences and Technology, Northeast Agricultural University, Harbin, China
| | - Xiuqin Yang
- College of Animal Sciences and Technology, Northeast Agricultural University, Harbin, China
| |
Collapse
|
145
|
Yang Y, Yan J, Fan X, Chen J, Wang Z, Liu X, Yi G, Liu Y, Niu Y, Zhang L, Wang L, Li S, Li K, Tang Z. The genome variation and developmental transcriptome maps reveal genetic differentiation of skeletal muscle in pigs. PLoS Genet 2021; 17:e1009910. [PMID: 34780471 PMCID: PMC8629385 DOI: 10.1371/journal.pgen.1009910] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 11/29/2021] [Accepted: 10/25/2021] [Indexed: 12/13/2022] Open
Abstract
Natural and artificial directional selections have resulted in significantly genetic and phenotypic differences across breeds in domestic animals. However, the molecular regulation of skeletal muscle diversity remains largely unknown. Here, we conducted transcriptome profiling of skeletal muscle across 27 time points, and performed whole-genome re-sequencing in Landrace (lean-type) and Tongcheng (obese-type) pigs. The transcription activity decreased with development, and the high-resolution transcriptome precisely captured the characterizations of skeletal muscle with distinct biological events in four developmental phases: Embryonic, Fetal, Neonatal, and Adult. A divergence in the developmental timing and asynchronous development between the two breeds was observed; Landrace showed a developmental lag and stronger abilities of myoblast proliferation and cell migration, whereas Tongcheng had higher ATP synthase activity in postnatal periods. The miR-24-3p driven network targeting insulin signaling pathway regulated glucose metabolism. Notably, integrated analysis suggested SATB2 and XLOC_036765 contributed to skeletal muscle diversity via regulating the myoblast migration and proliferation, respectively. Overall, our results provide insights into the molecular regulation of skeletal muscle development and diversity in mammals.
Collapse
Affiliation(s)
- Yalan Yang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Kunpeng Institute of Modern Agriculture at Foshan, Foshan, China
| | - Junyu Yan
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Xinhao Fan
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Jiaxing Chen
- Department of Computer Science, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Zishuai Wang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Department of Computer Science, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Xiaoqin Liu
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Guoqiang Yi
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Kunpeng Institute of Modern Agriculture at Foshan, Foshan, China
- Guangxi Engineering Centre for Resource Development of Bama Xiang Pig, Bama, China
| | - Yuwen Liu
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Kunpeng Institute of Modern Agriculture at Foshan, Foshan, China
| | | | - Longchao Zhang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lixian Wang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - ShuaiCheng Li
- Department of Computer Science, City University of Hong Kong, Kowloon, Hong Kong, China
- * E-mail: (SCL); (KL); (ZLT)
| | - Kui Li
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Research Centre of Animal Nutritional Genomics, State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Shenzhen, China
- * E-mail: (SCL); (KL); (ZLT)
| | - Zhonglin Tang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Kunpeng Institute of Modern Agriculture at Foshan, Foshan, China
- Guangxi Engineering Centre for Resource Development of Bama Xiang Pig, Bama, China
- Research Centre of Animal Nutritional Genomics, State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Shenzhen, China
- * E-mail: (SCL); (KL); (ZLT)
| |
Collapse
|
146
|
Skeletal Muscle Regeneration by the Exosomes of Adipose Tissue-Derived Mesenchymal Stem Cells. Curr Issues Mol Biol 2021; 43:1473-1488. [PMID: 34698065 PMCID: PMC8929094 DOI: 10.3390/cimb43030104] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 09/28/2021] [Accepted: 10/01/2021] [Indexed: 12/17/2022] Open
Abstract
Profound skeletal muscle loss can lead to severe disability and cosmetic deformities. Mesenchymal stem cell (MSC)-derived exosomes have shown potential as an effective therapeutic tool for tissue regeneration. This study aimed to determine the regenerative capacity of MSC-derived exosomes for skeletal muscle regeneration. Exosomes were isolated from human adipose tissue-derived MSCs (AD-MSCs). The effects of MSC-derived exosomes on satellite cells were investigated using cell viability, relevant genes, and protein analyses. Moreover, NOD-SCID mice were used and randomly assigned to the healthy control (n = 4), muscle defect (n = 6), and muscle defect + exosome (n = 6) groups. Muscle defects were created using a biopsy punch on the quadriceps of the hind limb. Four weeks after the surgery, the quadriceps muscles were harvested, weighed, and histologically analyzed. MSC-derived exosome treatment increased the proliferation and expression of myocyte-related genes, and immunofluorescence analysis for myogenin revealed a similar trend. Histologically, MSC-derived exosome-treated mice showed relatively preserved shapes and sizes of the muscle bundles. Immunohistochemical staining revealed greater expression of myogenin and myoblast determination protein 1 in the MSC-derived exosome-treated group. These results indicate that exosomes extracted from AD-MSCs have the therapeutic potential for skeletal muscle regeneration.
Collapse
|
147
|
Becker R, Vergarajauregui S, Billing F, Sharkova M, Lippolis E, Mamchaoui K, Ferrazzi F, Engel FB. Myogenin controls via AKAP6 non-centrosomal microtubule-organizing center formation at the nuclear envelope. eLife 2021; 10:65672. [PMID: 34605406 PMCID: PMC8523159 DOI: 10.7554/elife.65672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 10/01/2021] [Indexed: 12/22/2022] Open
Abstract
Non-centrosomal microtubule-organizing centers (MTOCs) are pivotal for the function of multiple cell types, but the processes initiating their formation are unknown. Here, we find that the transcription factor myogenin is required in murine myoblasts for the localization of MTOC proteins to the nuclear envelope. Moreover, myogenin is sufficient in fibroblasts for nuclear envelope MTOC (NE-MTOC) formation and centrosome attenuation. Bioinformatics combined with loss- and gain-of-function experiments identified induction of AKAP6 expression as one central mechanism for myogenin-mediated NE-MTOC formation. Promoter studies indicate that myogenin preferentially induces the transcription of muscle- and NE-MTOC-specific isoforms of Akap6 and Syne1, which encodes nesprin-1α, the NE-MTOC anchor protein in muscle cells. Overexpression of AKAP6β and nesprin-1α was sufficient to recruit endogenous MTOC proteins to the nuclear envelope of myoblasts in the absence of myogenin. Taken together, our results illuminate how mammals transcriptionally control the switch from a centrosomal MTOC to an NE-MTOC and identify AKAP6 as a novel NE-MTOC component in muscle cells.
Collapse
Affiliation(s)
- Robert Becker
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Silvia Vergarajauregui
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Florian Billing
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Maria Sharkova
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Eleonora Lippolis
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Kamel Mamchaoui
- Sorbonne Universités UPMC Université Paris 06, INSERM U974, CNRS FRE3617, Center for Research in Myology, GH Pitié Salpêtrière, 47 Boulevard de l'Hôpital, Paris, France
| | - Fulvia Ferrazzi
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.,Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.,Muscle Research Center Erlangen (MURCE), Erlangen, Germany
| |
Collapse
|
148
|
Ong C, Lee JH, Leow MKS, Puthucheary ZA. A narrative review of skeletal muscle atrophy in critically ill children: pathogenesis and chronic sequelae. Transl Pediatr 2021; 10:2763-2777. [PMID: 34765499 PMCID: PMC8578782 DOI: 10.21037/tp-20-298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/18/2020] [Indexed: 11/10/2022] Open
Abstract
Muscle wasting is now recognized as a growing, debilitating problem in critically ill adults, resulting in long-term deficits in function and an impaired quality of life. Ultrasonography has demonstrated decreases in skeletal muscle size during pediatric critical illness, although variations exist. However, muscle protein turnover patterns during pediatric critical illness are unclear. Understanding muscle protein turnover during critical illness is important in guiding interventions to reduce muscle wasting. The aim of this review was to explore the possible protein synthesis and breakdown patterns in pediatric critical illness. Muscle protein turnover studies in critically ill children are lacking, with the exception of those with burn injuries. Children with burn injuries demonstrate an elevation in both muscle protein breakdown (MPB) and synthesis during critical illness. Extrapolations from animal models and whole-body protein turnover studies in children suggest that children may be more dependent on anabolic factors (e.g., nutrition and growth factors), and may experience greater muscle degradation in response to insults than adults. Yet, children, particularly the younger ones, are more responsive to anabolic agents, suggesting modifiable muscle wasting during critical illness. There is a lack of evidence for muscle wasting in critically ill children and its correlation with outcomes, possibly due to current available methods to study muscle protein turnover in children-most of which are invasive or tedious. In summary, children may experience muscle wasting during critical illness, which may be more reversible by the appropriate anabolic agents than adults. Age appears an important determinant of skeletal muscle turnover. Less invasive methods to study muscle protein turnover and associations with long-term outcome would strengthen the evidence for muscle wasting in critically ill children.
Collapse
Affiliation(s)
- Chengsi Ong
- Nutrition and Dietetics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Jan Hau Lee
- Children's Intensive Care Unit, KK Women's Children's Hospital, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Melvin K S Leow
- Duke-NUS Medical School, Singapore, Singapore.,Clinical Nutrition Research Center, Agency for Science, Technology and Research, Singapore, Singapore.,Department of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Zudin A Puthucheary
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Adult Critical Care Unit, Royal London Hospital, London, UK
| |
Collapse
|
149
|
Vasyukova OV, Kasyanova YV, Okorokov PL, Bezlepkina OB. [Myokines and adipomyokines: inflammatory mediators or unique molecules of targeted therapy for obesity?]. ACTA ACUST UNITED AC 2021; 67:36-45. [PMID: 34533012 DOI: 10.14341/probl12779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/02/2021] [Accepted: 08/10/2021] [Indexed: 11/06/2022]
Abstract
Skeletal muscles make up about 25% of the total mass in children and more than 40% in adults. Studies of the last twenty years have shown that along with the main functions, muscle tissue has hormonal activity. It was found that myocytes are able to release signaling molecules-myokines. They act auto-and paracrine within the muscle, and at a high level-through the systemic circulation, carrying out interactions between skeletal muscles and various organs and tissues, such as the liver, bone and adipose tissue, the brain. It is proved that the key factor in the expression of myokines is physical activity, and their level largely depends on physical fitness, the amount of skeletal muscle mass and its composition (the ratio of fast and slow fibers), on the intensity and duration of physical activity. Myokines have a wide range of physiological effects: myostatin suppresses the growth and differentiation of muscle tissue, and decorin, acting as its antagonist, promotes muscle hypertrophy. Interleukin 6 provides an energy substrate for contracting muscle fibers, fibroblast growth factor 21 activates the mechanisms of energy production during fasting and improves tissue sensitivity to insulin; irisin stimulates thermogenesis, glucose uptake by myocytes, and also contributes to an increase in bone mineral density. The study of myokines is one of the key links in understanding the mechanisms underlying obesity and metabolic complications, the consequences of a sedentary lifestyle, as well as the implementation of the action of physical activity. Taking into account the physiological effects of myokines in the body, in the future they can become therapeutic targets for the treatment of these conditions.
Collapse
|
150
|
Siddiqui SH, Park J, Kang D, Khan M, Shim K. Cortisol differentially affects the viability and myogenesis of mono- and co-cultured porcine gluteal muscles satellite cells and fibroblasts. Tissue Cell 2021; 73:101615. [PMID: 34419738 DOI: 10.1016/j.tice.2021.101615] [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: 04/22/2021] [Revised: 07/26/2021] [Accepted: 07/29/2021] [Indexed: 12/19/2022]
Abstract
Cortisol is a ubiquitously expressed stress hormone. In this study, we investigated the effects of exogenous cortisol on porcine gluteal muscles primary cultured satellite cells and fibroblasts. Satellite cells and fibroblasts were mono-or co-cultured, and cells in each type of culture were categorized into the control and cortisol-treated (treatment) groups. We selected 28 μmol mL-1 cortisol for treatment based on their efficacy. Cortisol treatment reduced viability of monocultured satellite cells and fibroblasts. In both monocultured and co-cultured cells, the nucleus in the treatment group was damaged than that control group. Moreover, the total cell cycle duration was shorter in the treatment group than the control group. PAX-7 expression was upregulated in the control group of co-cultured satellite cells and fibroblasts than those remaining groups. Moreover, MyoD expression was downregulated in the cortisol treated group of both mono-and co-cultured satellite cells compared with that in the control group. In co-cultured fibroblasts, MyoD and MyoG expression was upregulated than those remaining groups. The Cyto-C expression was upregulated in the treatment group compared to the control mono-and co-cultured both cells. These results suggest that the selected experimental dose of cortisol reduced cell viability and myogenesis-related gene expression in the monoculture compared to that in the co-culture of satellite cells and fibroblasts.
Collapse
Affiliation(s)
- Sharif Hasan Siddiqui
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Jinryong Park
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Darae Kang
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Mousumee Khan
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University, Jeonju, 54907, Republic of Korea
| | - Kwanseob Shim
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju, 54896, Republic of Korea; Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, 54896, Republic of Korea.
| |
Collapse
|