1
|
Wang X, Zhang J, Su J, Huang T, Lian L, Nie Q, Zhang X, Li J, Wang Y. Genome-wide mapping of the binding sites of myocyte enhancer factor 2A in chicken primary myoblasts. Poult Sci 2024; 103:104097. [PMID: 39094502 PMCID: PMC11345569 DOI: 10.1016/j.psj.2024.104097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 08/04/2024] Open
Abstract
Myocyte enhancer factor 2A (MEF2A) is a transcription factor that plays a critical role in cell proliferation, differentiation and apoptosis. In contrast to the wide characterization of its regulation mechanism in mammalian skeletal muscle, its role in chickens is limited. Especially, its wide target genes remain to be identified. Therefore, we utilized Cleavage Under Targets and Tagmentation (CUT&Tag) technology to reveal the genome-wide binding profile of MEF2A in chicken primary myoblasts thus gaining insights into its potential role in muscle development. Our results revealed that MEF2A binding sites were primarily distributed in intergenic and intronic regions. Within the promoter region, although only 8.87% of MEF2A binding sites were found, these binding sites were concentrated around the transcription start site (TSS). Following peak annotation, a total of 1903 genes were identified as potential targets of MEF2A. Gene Ontology (GO) enrichment analysis further revealed that MEF2A target genes may be involved in the regulation of embryonic development in multiple organ systems, including muscle development, gland development, and visual system development. Moreover, a comparison of the MEF2A target genes identified in chicken primary myoblasts with those in mouse C2C12 cells revealed 388 target genes are conserved across species, 1515 target genes are chicken specific. Among these conserved genes, ankyrin repeat and SOCS box containing 5 (ASB5), transmembrane protein 182 (TMEM182), myomesin 2 (MYOM2), leucyl and cystinyl aminopeptidase (LNPEP), actinin alpha 2 (ACTN2), sorbin and SH3 domain containing 1 (SORBS1), ankyrin 3 (ANK3), sarcoglycan delta (SGCD), and ORAI calcium release-activated calcium modulator 1 (ORAI1) exhibited consistent expression patterns with MEF2A during embryonic muscle development. Finally, TMEM182, as an important negative regulator of muscle development, has been validated to be regulated by MEF2A by dual-luciferase and quantitative real-time PCR (qPCR) assays. In summary, our study for the first time provides a wide landscape of MEF2A target genes in chicken primary myoblasts, which supports the active role of MEF2A in chicken muscle development.
Collapse
Affiliation(s)
- Xinglong Wang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, PR China
| | - Jiannan Zhang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, PR China
| | - Jiancheng Su
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, PR China
| | - Tianjiao Huang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, PR 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, PR China
| | - Qinghua Nie
- Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, PR China
| | - Xin Zhang
- Joint Nutrition Center for Animal Feeding of Sichuan University-Shengliyuan Group
| | - Juan Li
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, PR China; Joint Nutrition Center for Animal Feeding of Sichuan University-Shengliyuan Group
| | - Yajun Wang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, PR China; Joint Nutrition Center for Animal Feeding of Sichuan University-Shengliyuan Group.
| |
Collapse
|
2
|
Xiao W, Huang TE, Zhou J, Wang B, Wang X, Zeng W, Wang Q, Lan X, Xiang Y. Inhibition of MAT2A Impairs Skeletal Muscle Repair Function. Biomolecules 2024; 14:1098. [PMID: 39334864 PMCID: PMC11430595 DOI: 10.3390/biom14091098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/30/2024] Open
Abstract
The regenerative capacity of muscle, which primarily relies on anabolic processes, diminishes with age, thereby reducing the effectiveness of therapeutic interventions aimed at treating age-related muscle atrophy. In this study, we observed a decline in the expression of methionine adenosine transferase 2A (MAT2A), which synthesizes S-adenosylmethionine (SAM), in the muscle tissues of both aged humans and mice. Considering MAT2A's critical role in anabolism, we hypothesized that its reduced expression contributes to the impaired regenerative capacity of aging skeletal muscle. Mimicking this age-related reduction in the MAT2A level, either by reducing gene expression or inhibiting enzymatic activity, led to inhibiting their differentiation into myotubes. In vivo, inhibiting MAT2A activity aggravated BaCl2-induced skeletal muscle damage and decreased the number of satellite cells, whereas supplementation with SAM improved these effects. RNA-sequencing analysis further revealed that the Fas cell surface death receptor (Fas) gene was upregulated in Mat2a-knockdown C2C12 cells. Suppressing MAT2A expression or activity elevated Fas protein levels and increased the proportion of apoptotic cells. Additionally, inhibition of MAT2A expression or activity increased p53 expression. In conclusion, our findings demonstrated that impaired MAT2A expression or activity compromised the regeneration and repair capabilities of skeletal muscle, partially through p53-Fas-mediated apoptosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yang Xiang
- Metabolic Control and Aging—Jiangxi Key Laboratory of Aging and Diseases, Human Aging Research Institute (HARI), School of Life Science, Nanchang University, Nanchang 330031, China; (W.X.); (T.-E.H.); (J.Z.); (B.W.); (X.W.); (W.Z.); (Q.W.); (X.L.)
| |
Collapse
|
3
|
Li P, Feng X, Ma Z, Yuan Y, Jiang H, Xu G, Zhu Y, Yang X, Wang Y, Zhu C, Wang S, Gao P, Jiang Q, Shu G. Microbiota-derived 3-phenylpropionic acid promotes myotube hypertrophy by Foxo3/NAD + signaling pathway. Cell Biosci 2024; 14:62. [PMID: 38750565 PMCID: PMC11097579 DOI: 10.1186/s13578-024-01244-2] [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: 02/09/2024] [Accepted: 05/03/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Gut microbiota and their metabolites play a regulatory role in skeletal muscle growth and development, which be known as gut-muscle axis. 3-phenylpropionic acid (3-PPA), a metabolite produced by colonic microorganisms from phenylalanine in the gut, presents in large quantities in the blood circulation. But few study revealed its function in skeletal muscle development. RESULTS Here, we demonstrated the beneficial effects of 3-PPA on muscle mass increase and myotubes hypertrophy both in vivo and vitro. Further, we discovered the 3-PPA effectively inhibited protein degradation and promoted protein acetylation in C2C12 and chick embryo primary skeletal muscle myotubes. Mechanistically, we supported that 3-PPA reduced NAD+ synthesis and subsequently suppressed tricarboxylic acid cycle and the mRNA expression of SIRT1/3, thus promoting the acetylation of total protein and Foxo3. Moreover, 3-PPA may inhibit Foxo3 activity by directly binding. CONCLUSIONS This study firstly revealed the effect of 3-PPA on skeletal muscle growth and development, and newly discovered the interaction between 3-PPA and Foxo3/NAD+ which mechanically promote myotubes hypertrophy. These results expand new understanding for the regulation of gut microbiota metabolites on skeletal muscle growth and development.
Collapse
Affiliation(s)
- Penglin Li
- State Key Laboratory of Swine and Poultry Breeding Industry, Tianhe District, 483 Wushan Road, Guangzhou, 510642, Guangdong, China
| | - Xiaohua Feng
- State Key Laboratory of Swine and Poultry Breeding Industry, Tianhe District, 483 Wushan Road, Guangzhou, 510642, Guangdong, China
| | - Zewei Ma
- State Key Laboratory of Swine and Poultry Breeding Industry, Tianhe District, 483 Wushan Road, Guangzhou, 510642, Guangdong, China
| | - Yexian Yuan
- State Key Laboratory of Swine and Poultry Breeding Industry, Tianhe District, 483 Wushan Road, Guangzhou, 510642, Guangdong, China
| | - Hongfeng Jiang
- State Key Laboratory of Swine and Poultry Breeding Industry, Tianhe District, 483 Wushan Road, Guangzhou, 510642, Guangdong, China
| | - Guli Xu
- State Key Laboratory of Swine and Poultry Breeding Industry, Tianhe District, 483 Wushan Road, Guangzhou, 510642, Guangdong, China
| | - Yunlong Zhu
- State Key Laboratory of Swine and Poultry Breeding Industry, Tianhe District, 483 Wushan Road, Guangzhou, 510642, Guangdong, China
| | - Xue Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, Tianhe District, 483 Wushan Road, Guangzhou, 510642, Guangdong, China
| | - Yujun Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, Tianhe District, 483 Wushan Road, Guangzhou, 510642, Guangdong, China
| | - Canjun Zhu
- State Key Laboratory of Swine and Poultry Breeding Industry, Tianhe District, 483 Wushan Road, Guangzhou, 510642, Guangdong, China
| | - Songbo Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, Tianhe District, 483 Wushan Road, Guangzhou, 510642, Guangdong, China
| | - Ping Gao
- State Key Laboratory of Swine and Poultry Breeding Industry, Tianhe District, 483 Wushan Road, Guangzhou, 510642, Guangdong, China
| | - Qingyan Jiang
- State Key Laboratory of Swine and Poultry Breeding Industry, Tianhe District, 483 Wushan Road, Guangzhou, 510642, Guangdong, China.
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Tianhe District, 483 Wushan Road, Guangzhou, 510642, Guangdong, China.
- Key Laboratory of Animal Nutritional Regulation, College of Animal Science, South China Agricultural University, Tianhe District, 483 Wushan Road, Guangzhou, 510642, Guangdong, China.
| | - Gang Shu
- State Key Laboratory of Swine and Poultry Breeding Industry, Tianhe District, 483 Wushan Road, Guangzhou, 510642, Guangdong, China.
- Guangdong Laboratory for Lingnan Modern Agricultural and Guangdong Province, Tianhe District, 483 Wushan Road, Guangzhou, 510642, Guangdong, China.
- Key Laboratory of Animal Nutritional Regulation, College of Animal Science, South China Agricultural University, Tianhe District, 483 Wushan Road, Guangzhou, 510642, Guangdong, China.
| |
Collapse
|
4
|
Jin C, Yan K, Wang M, Song W, Kong X, Zhang Z. Identification, Characterization and Functional Analysis of Fibroblast Growth Factors in Black Rockfish ( Sebastes schlegelii). Int J Mol Sci 2023; 24:ijms24043626. [PMID: 36835037 PMCID: PMC9958866 DOI: 10.3390/ijms24043626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/04/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
Fibroblast growth factors (FGFs) are short polypeptides that play essential roles in various cellular biological processes, including cell migration, proliferation, and differentiation, as well as tissue regeneration, immune response, and organogenesis. However, studies focusing on the characterization and function of FGF genes in teleost fishes are still limited. In this study, we identified and characterized expression patterns of 24 FGF genes in various tissues of embryonic and adult specimens of the black rockfish (Sebates schlegelii). Nine FGF genes were found to play essential roles in myoblast differentiation, as well as muscle development and recovery in juvelines of S. schlegelii. Moreover, sex-biased expression pattern of multiple FGF genes was recorded in the species' gonads during its development. Among them, expression of the FGF1 gene was recorded in interstitial and sertoli cells of testes, promoting germ-cell proliferation and differentiation. In sum, the obtained results enabled systematic and functional characterization of FGF genes in S. schlegelii, laying a foundation for further studies on FGF genes in other large teleost fishes.
Collapse
Affiliation(s)
- Chaofan Jin
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Kai Yan
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Mengya Wang
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
- Laboratory of Tropical Marine Germplasm Resources and Breeding Engineering, Sanya Oceanographic Institution, Ocean University of China, Sanya 572000, China
| | - Weihao Song
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Xiangfu Kong
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Zhengrui Zhang
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
- Correspondence:
| |
Collapse
|
5
|
Baldwin C, Kim J, Sivaraman S, Rao RR. Stem cell-based strategies for skeletal muscle tissue engineering. J Tissue Eng Regen Med 2022; 16:1061-1068. [PMID: 36223074 DOI: 10.1002/term.3355] [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: 01/04/2022] [Revised: 09/06/2022] [Accepted: 09/27/2022] [Indexed: 01/05/2023]
Abstract
Skeletal muscle tissue engineering has been a key area of focus over the years and has been of interest for developing regenerative strategies for injured or degenerative skeletal muscle tissue. Stem cells have gained increased attention as sources for developing skeletal muscle tissue for subsequent studies or potential treatments. Focus has been placed on understanding the molecular pathways that govern skeletal muscle formation in development to advance differentiation of stem cells towards skeletal muscle fates in vitro. Use of growth factors and transcription factors have long been the method for guiding skeletal muscle differentiation in vitro. However, further research in small molecule induced differentiation offers a xeno-free option that could result from use of animal derived factors.
Collapse
Affiliation(s)
- Christofer Baldwin
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| | - Johntaehwan Kim
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Srikanth Sivaraman
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| | - Raj R Rao
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| |
Collapse
|
6
|
Cai MY, Xu YL, Rong H, Yang H. Low Level of PALMD Contributes to the Metastasis of Uveal Melanoma. Front Oncol 2022; 12:802941. [PMID: 35494064 PMCID: PMC9043551 DOI: 10.3389/fonc.2022.802941] [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: 10/28/2021] [Accepted: 03/16/2022] [Indexed: 11/13/2022] Open
Abstract
Uveal melanoma (UM) is a highly aggressive disease. There is an urgent need to develop the metastasis prediction markers of UM. This study aims to detect the key role of PALMD in UM metastasis. Transcriptome sequencing results of 2 sets of UM metastatic samples (GSE22138 and GSE156877) were downloaded from the Gene Expression Omnibus (GEO), and 18 overlapping differentially expressed genes were screened out, including PALMD. PALMD was significantly underexpressed in metastatic UM tissue. Low expression of PALMD was associated with poor prognosis in UM patients. The decreased expression of PALMD promoted the invasion and migration of 92-1 and Mel270 cells, while the high expression of PALMD inhibited the invasion and migration of UM cells. Furthermore, the levels of matrix metallopeptidase (MMP) 2 and MMP9 increased after transfection of siRNAs specifically targeting PALMD, whereas the levels of MMP2 and MMP9 were decreased after PALMD overexpression. However, PALMD did not affect the proliferation of UM cells. In addition, ZNF263 promoted the transcription of PALMD through the putative binding sequence using the JASPAR database, luciferase reporter gene analysis and chromatin immunoprecipitation assay. In summary, the expression of PALMD regulated by ZNF263 plays an important role in UM metastasis.
Collapse
Affiliation(s)
- Min-Yun Cai
- Department of Ophthalmology, Shanghai East Hospital, Shanghai, China
| | - Yue-Li Xu
- Department of Ophthalmology, Shanghai East Hospital, Shanghai, China
| | - Hua Rong
- Department of Ophthalmology, Shanghai Jiangong Hospital, Shanghai, China
| | - Hai Yang
- Department of Ophthalmology, Shanghai East Hospital, Shanghai, China
| |
Collapse
|
7
|
Wang S, Yu H, Gao J, Chen J, He P, Zhong H, Tan X, Staines KA, Macrae VE, Fu X, Jiang L, Zhu D. PALMD regulates aortic valve calcification via altered glycolysis and NF-κB-mediated inflammation. J Biol Chem 2022; 298:101887. [PMID: 35367413 PMCID: PMC9065630 DOI: 10.1016/j.jbc.2022.101887] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 11/25/2022] Open
Abstract
Recent genome-wide association and transcriptome-wide association studies have identified an association between the PALMD locus, encoding palmdelphin, a protein involved in myoblast differentiation, and calcific aortic valve disease (CAVD). Nevertheless, the function and underlying mechanisms of PALMD in CAVD remain unclear. We herein investigated whether and how PALMD affects the pathogenesis of CAVD using clinical samples from CAVD patients and a human valve interstitial cell (hVIC) in vitro calcification model. We showed that PALMD was upregulated in calcified regions of human aortic valves and calcified hVICs. Furthermore, silencing of PALMD reduced hVIC in vitro calcification, osteogenic differentiation, and apoptosis, whereas overexpression of PALMD had the opposite effect. RNA-Seq of PALMD-depleted hVICs revealed that silencing of PALMD reduced glycolysis and nuclear factor-κB (NF-κB)–mediated inflammation in hVICs and attenuated tumor necrosis factor α–induced monocyte adhesion to hVICs. Having established the role of PALMD in hVIC glycolysis, we examined whether glycolysis itself could regulate hVIC osteogenic differentiation and inflammation. Intriguingly, the inhibition of PFKFB3-mediated glycolysis significantly attenuated osteogenic differentiation and inflammation of hVICs. However, silencing of PFKFB3 inhibited PALMD-induced hVIC inflammation, but not osteogenic differentiation. Finally, we showed that the overexpression of PALMD enhanced hVIC osteogenic differentiation and inflammation, as opposed to glycolysis, through the activation of NF-κB. The present study demonstrates that the genome-wide association– and transcriptome-wide association–identified CAVD risk gene PALMD may promote CAVD development through regulation of glycolysis and NF-κB–mediated inflammation. We propose that targeting PALMD-mediated glycolysis may represent a novel therapeutic strategy for treating CAVD.
Collapse
Affiliation(s)
- Siying Wang
- Department of Basic Medical Research, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China; Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hongjiao Yu
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, China
| | - Jun Gao
- Department of Basic Medical Research, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Jiaxin Chen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Pengcheng He
- Guangdong Provincial Geriatrics Institute, and Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hui Zhong
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiao Tan
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Katherine A Staines
- Centre for Stress and Age-Related Disease, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK
| | - Vicky E Macrae
- Functional Genetics and Development, The Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Xiaodong Fu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Lei Jiang
- Guangdong Provincial Geriatrics Institute, and Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Dongxing Zhu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
8
|
Sáinz-Jaspeado M, Smith RO, Plunde O, Pawelzik SC, Jin Y, Nordling S, Ding Y, Aspenström P, Hedlund M, Bastianello G, Ascione F, Li Q, Demir CS, Fernando D, Daniel G, Franco-Cereceda A, Kroon J, Foiani M, Petrova TV, Kilimann MW, Bäck M, Claesson-Welsh L. Palmdelphin Regulates Nuclear Resilience to Mechanical Stress in the Endothelium. Circulation 2021; 144:1629-1645. [PMID: 34636652 PMCID: PMC8589083 DOI: 10.1161/circulationaha.121.054182] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Supplemental Digital Content is available in the text. PALMD (palmdelphin) belongs to the family of paralemmin proteins implicated in cytoskeletal regulation. Single nucleotide polymorphisms in the PALMD locus that result in reduced expression are strong risk factors for development of calcific aortic valve stenosis and predict severity of the disease.
Collapse
Affiliation(s)
- Miguel Sáinz-Jaspeado
- Rudbeck, Beijer and SciLifeLab Laboratories, Department of Immunology, Genetics and Pathology (M.S.-J., R.O.S., Y.J., S.N., Y.D., P.A., M.H., L.C.-W.), Uppsala University, Sweden
| | - Ross O Smith
- Rudbeck, Beijer and SciLifeLab Laboratories, Department of Immunology, Genetics and Pathology (M.S.-J., R.O.S., Y.J., S.N., Y.D., P.A., M.H., L.C.-W.), Uppsala University, Sweden
| | - Oscar Plunde
- Department of Medicine Solna, Karolinska Institutet and Department of Cardiology, Karolinska University Hospital Stockholm, Sweden (O.P., S.-C.P., M.B.)
| | - Sven-Christian Pawelzik
- Department of Medicine Solna, Karolinska Institutet and Department of Cardiology, Karolinska University Hospital Stockholm, Sweden (O.P., S.-C.P., M.B.)
| | - Yi Jin
- Rudbeck, Beijer and SciLifeLab Laboratories, Department of Immunology, Genetics and Pathology (M.S.-J., R.O.S., Y.J., S.N., Y.D., P.A., M.H., L.C.-W.), Uppsala University, Sweden
| | - Sofia Nordling
- Rudbeck, Beijer and SciLifeLab Laboratories, Department of Immunology, Genetics and Pathology (M.S.-J., R.O.S., Y.J., S.N., Y.D., P.A., M.H., L.C.-W.), Uppsala University, Sweden
| | - Yindi Ding
- Rudbeck, Beijer and SciLifeLab Laboratories, Department of Immunology, Genetics and Pathology (M.S.-J., R.O.S., Y.J., S.N., Y.D., P.A., M.H., L.C.-W.), Uppsala University, Sweden
| | - Pontus Aspenström
- Rudbeck, Beijer and SciLifeLab Laboratories, Department of Immunology, Genetics and Pathology (M.S.-J., R.O.S., Y.J., S.N., Y.D., P.A., M.H., L.C.-W.), Uppsala University, Sweden
| | - Marie Hedlund
- Rudbeck, Beijer and SciLifeLab Laboratories, Department of Immunology, Genetics and Pathology (M.S.-J., R.O.S., Y.J., S.N., Y.D., P.A., M.H., L.C.-W.), Uppsala University, Sweden
| | - Giulia Bastianello
- IFOM-FIRC (institute of molecular oncology - Fondazione italiana per la ricerca sul cancro), Milano, Italy (G.B., F.A., Q.L., M.F.).,University of Milan, Italy (G.B., M.F.)
| | - Flora Ascione
- IFOM-FIRC (institute of molecular oncology - Fondazione italiana per la ricerca sul cancro), Milano, Italy (G.B., F.A., Q.L., M.F.)
| | - Qingsen Li
- IFOM-FIRC (institute of molecular oncology - Fondazione italiana per la ricerca sul cancro), Milano, Italy (G.B., F.A., Q.L., M.F.)
| | - Cansaran Saygili Demir
- Department of Oncology, University of Lausanne, Switzerland (C.S.D., T.V.P.).,Ludwig Institute for Cancer Research Lausanne, Switzerland (C.S.D., T.V.P.)
| | - Dinesh Fernando
- Department of Forest Biomaterials and Technology, Swedish University of Agricultural Sciences, Uppsala (D.F., G.D.)
| | - Geoffrey Daniel
- Department of Forest Biomaterials and Technology, Swedish University of Agricultural Sciences, Uppsala (D.F., G.D.)
| | - Anders Franco-Cereceda
- Department of Molecular Medicine and Surgery, Karolinska Institutet, and Department of Cardiothoracic Surgery, Karolinska University Hospital, Stockholm, Sweden (A.F.-C.)
| | - Jeffrey Kroon
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam University Medical Center, The Netherlands (J.K.)
| | - Marco Foiani
- IFOM-FIRC (institute of molecular oncology - Fondazione italiana per la ricerca sul cancro), Milano, Italy (G.B., F.A., Q.L., M.F.).,University of Milan, Italy (G.B., M.F.)
| | - Tatiana V Petrova
- Department of Oncology, University of Lausanne, Switzerland (C.S.D., T.V.P.).,Ludwig Institute for Cancer Research Lausanne, Switzerland (C.S.D., T.V.P.)
| | - Manfred W Kilimann
- Department of Neuroscience (M.W.K.), Uppsala University, Sweden.,Department of Molecular Neurobiology, Max Planck Institute for Experimental Medicine, Göttingen, Germany (M.W.K.)
| | - Magnus Bäck
- Department of Medicine Solna, Karolinska Institutet and Department of Cardiology, Karolinska University Hospital Stockholm, Sweden (O.P., S.-C.P., M.B.)
| | - Lena Claesson-Welsh
- Rudbeck, Beijer and SciLifeLab Laboratories, Department of Immunology, Genetics and Pathology (M.S.-J., R.O.S., Y.J., S.N., Y.D., P.A., M.H., L.C.-W.), Uppsala University, Sweden
| |
Collapse
|
9
|
Major E, Keller I, Horváth D, Tamás I, Erdődi F, Lontay B. Smoothelin-Like Protein 1 Regulates the Thyroid Hormone-Induced Homeostasis and Remodeling of C2C12 Cells via the Modulation of Myosin Phosphatase. Int J Mol Sci 2021; 22:10293. [PMID: 34638630 PMCID: PMC8508602 DOI: 10.3390/ijms221910293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 11/28/2022] Open
Abstract
The pathological elevation of the active thyroid hormone (T3) level results in the manifestation of hyperthyroidism, which is associated with alterations in the differentiation and contractile function of skeletal muscle (SKM). Myosin phosphatase (MP) is a major cellular regulator that hydrolyzes the phosphoserine of phosphorylated myosin II light chain. MP consists of an MYPT1/2 regulatory and a protein phosphatase 1 catalytic subunit. Smoothelin-like protein 1 (SMTNL1) is known to inhibit MP by directly binding to MP as well as by suppressing the expression of MYPT1 at the transcriptional level. Supraphysiological vs. physiological concentration of T3 were applied on C2C12 myoblasts and differentiated myotubes in combination with the overexpression of SMTNL1 to assess the role and regulation of MP under these conditions. In non-differentiated myoblasts, MP included MYPT1 in the holoenzyme complex and its expression and activity was regulated by SMTNL1, affecting the phosphorylation level of MLC20 assessed using semi-quantitative Western blot analysis. SMTNL1 negatively influenced the migration and cytoskeletal remodeling of myoblasts measured by high content screening. In contrast, in myotubes, the expression of MYPT2 but not MYPT1 increased in a T3-dependent and SMTNL1-independent manner. T3 treatment combined with SMTNL1 overexpression impeded the activity of MP. In addition, MP interacted with Na+/K+-ATPase and dephosphorylated its inhibitory phosphorylation sites, identifying this protein as a novel MP substrate. These findings may help us gain a better understanding of myopathy, muscle weakness and the disorder of muscle regeneration in hyperthyroid patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Beáta Lontay
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (E.M.); (I.K.); (D.H.); (I.T.); (F.E.)
| |
Collapse
|
10
|
Chignon A, Rosa M, Boulanger MC, Argaud D, Devillers R, Bon-Baret V, Mkannez G, Li Z, Rufiange A, Gaudreault N, Gosselin D, Thériault S, Bossé Y, Mathieu P. Enhancer-associated aortic valve stenosis risk locus 1p21.2 alters NFATC2 binding site and promotes fibrogenesis. iScience 2021; 24:102241. [PMID: 33748722 PMCID: PMC7970363 DOI: 10.1016/j.isci.2021.102241] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/01/2021] [Accepted: 02/24/2021] [Indexed: 11/24/2022] Open
Abstract
Genome-wide association studies for calcific aortic valve stenosis (CAVS) previously reported strong signal for noncoding variants at 1p21.2. Previous study using Mendelian randomization suggested that the locus controls the expression of PALMD encoding Palmdelphin (PALMD). However, the molecular regulation at the locus and the impact of PALMD on the biology of the aortic valve is presently unknown. 3D genetic mapping and CRISPR activation identified rs6702619 as being located in a distant-acting enhancer, which controls the expression of PALMD. DNA-binding assay showed that the risk variant modified the DNA shape, which prevented the recruitment of NFATC2 and lowered the expression of PALMD. In co-expression network analysis, a module encompassing PALMD was enriched in actin-based process. Mass spectrometry and functional assessment showed that PALMD is a regulator of actin polymerization. In turn, lower level of PALMD promoted the activation of myocardin-related transcription factor and fibrosis, a key pathobiological process underpinning CAVS.
Collapse
Affiliation(s)
- Arnaud Chignon
- Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, G1V-4G5, Québec City, QC, Canada
| | - Mickael Rosa
- Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, G1V-4G5, Québec City, QC, Canada
| | - Marie-Chloé Boulanger
- Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, G1V-4G5, Québec City, QC, Canada
| | - Déborah Argaud
- Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, G1V-4G5, Québec City, QC, Canada
| | - Romain Devillers
- Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, G1V-4G5, Québec City, QC, Canada
| | - Valentin Bon-Baret
- Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, G1V-4G5, Québec City, QC, Canada
| | - Ghada Mkannez
- Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, G1V-4G5, Québec City, QC, Canada
| | - Zhonglin Li
- Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, G1V-4G5, Québec City, QC, Canada
| | - Anne Rufiange
- Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, G1V-4G5, Québec City, QC, Canada
| | - Nathalie Gaudreault
- Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, G1V-4G5, Québec City, QC, Canada
| | - David Gosselin
- Department of Molecular Medicine, Laval University, Québec City, QC, Canada
| | - Sébastien Thériault
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Québec City, QC, Canada
| | - Yohan Bossé
- Department of Molecular Medicine, Laval University, Québec City, QC, Canada
| | - Patrick Mathieu
- Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, G1V-4G5, Québec City, QC, Canada
| |
Collapse
|
11
|
Xue M, Zhang F, Ji X, Yu H, Jiang X, Qiu Y, Yu J, Chen J, Yang F, Bao Z. Oleate Ameliorates Palmitate-Induced Impairment of Differentiative Capacity in C2C12 Myoblast Cells. Stem Cells Dev 2021; 30:289-300. [PMID: 33430700 DOI: 10.1089/scd.2020.0168] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
A common observation in metabolic disorders and aging is the elevation of free fatty acids (FFAs), which can form ectopic fat deposition and result in lipotoxicity. Ectopic fat deposition of skeletal muscle has been recognized as an important component of aging, frailty, and sarcopenia. Previous studies have suggested that lipotoxicity caused by FFAs mainly stemmed from saturated fatty acids and decreased unsaturated/saturated fatty acid ratio in serum are also observed among metabolic disorder patients. However, the different effects of saturated fatty acids and unsaturated fatty acids on skeletal muscle are not fully elucidated. In this study, we verified that palmitate (PA), a saturated fatty acid, could lead to impaired differentiative capacity of C2C12 myoblasts by affecting Pax7, MyoD, and myogenin (MyoG), which are master regulators of lineage specification and the myogenic program. Then, oleate (OA), a monounsaturated fatty acid, were added to culture medium together with PA. Results showed that OA could ameliorate the impairment of differentiative capacity in C2C12 myoblast cells. In addition, we found PI3K/Akt signaling pathway played an important role during the process by RNA sequencing and bioinformatics analysis. The positive effect of OA on myoblast differentiative capacity disappeared if PI3K inhibitor LY294002 was added. In conclusion, our study showed that PA could destroy differentiative capacity of C2C12 myoblasts by affecting the expression of Pax7, MyoD, and MyoG, and OA could improve this impairment through PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Mengjuan Xue
- Department of Geriatric Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, P.R. China
| | - Fan Zhang
- Department of Geriatric Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, P.R. China
| | - Xueying Ji
- Department of Geriatric Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, P.R. China
| | - Huiyuan Yu
- Department of Geriatric Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, P.R. China
| | - Xin Jiang
- Department of Geriatric Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, P.R. China
| | - Yixuan Qiu
- Department of Geriatric Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, P.R. China
| | - Jiaming Yu
- Department of Geriatric Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, P.R. China
| | - Jie Chen
- Department of Geriatric Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, P.R. China
| | - Fan Yang
- Department of Geriatric Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, P.R. China
| | - Zhijun Bao
- Department of Geriatric Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, P.R. China
| |
Collapse
|
12
|
Li Z, Gaudreault N, Arsenault BJ, Mathieu P, Bossé Y, Thériault S. Phenome-wide analyses establish a specific association between aortic valve PALMD expression and calcific aortic valve stenosis. Commun Biol 2020; 3:477. [PMID: 32859967 PMCID: PMC7455695 DOI: 10.1038/s42003-020-01210-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 08/04/2020] [Indexed: 12/27/2022] Open
Abstract
Calcific aortic valve stenosis (CAVS) is a frequent heart disease with significant morbidity and mortality. Recent genomic studies have identified a locus near the gene PALMD (palmdelphin) strongly associated with CAVS. Here, we show that genetically-determined expression of PALMD in the aortic valve is inversely associated with CAVS, with a stronger effect in women, in a meta-analysis of two large cohorts totaling 2359 cases and 350,060 controls. We further demonstrate the specificity of this relationship by showing the absence of other significant association between the genetically-determined expression of PALMD in 9 tissues and 852 phenotypes. Using genome-wide association studies meta-analyses of cardiovascular traits, we identify a significant colocalized positive association between genetically-determined expression of PALMD in four non-cardiac tissues (brain anterior cingulate cortex, esophagus muscularis, tibial nerve and subcutaneous adipose tissue) and atrial fibrillation. The present work further establishes PALMD as a promising molecular target for CAVS. Zhonglin Li et al. perform phenome-wide analyses to explore the genetic association between the locus near PALMD and calcific aortic valve stenosis (CAVS). Using previously reported aortic valve expression data and genotypes from large cohorts, they find a strong and specific association between genetically-determined PALMD expression in the aortic valve and CAVS as well as a novel association with atrial fibrillation in non-cardiac tissues.
Collapse
Affiliation(s)
- Zhonglin Li
- Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Nathalie Gaudreault
- Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Benoit J Arsenault
- Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, Quebec City, QC, G1V 0A6, Canada.,Department of Medicine, Laval University, Quebec City, QC, G1V 0A6, Canada
| | - Patrick Mathieu
- Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, Quebec City, QC, G1V 0A6, Canada.,Department of Surgery, Laval University, Quebec City, QC, G1V 0A6, Canada
| | - Yohan Bossé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, Quebec City, QC, G1V 0A6, Canada.,Department of Molecular Medicine, Laval University, Quebec City, QC, G1V 0A6, Canada
| | - Sébastien Thériault
- Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, Quebec City, QC, G1V 0A6, Canada. .,Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Quebec City, QC, G1V 0A6, Canada.
| |
Collapse
|
13
|
Long X, Gao Y, Liu W, Liu X, Hayashi T, Mizuno K, Hattori S, Ikejima T. Natural flavonoid silibinin promotes the migration and myogenic differentiation of murine C2C12 myoblasts via modulation of ROS generation and down-regulation of estrogen receptor α expression. Mol Cell Biochem 2020; 474:243-261. [PMID: 32789659 DOI: 10.1007/s11010-020-03849-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/20/2020] [Indexed: 01/11/2023]
Abstract
Skeletal muscle regeneration is a complex process, involving the proliferation, migration, and differentiation of myoblasts. Recent studies suggest that some natural flavanones stimulate myogenesis. However, the effect of plant estrogen, silibinin, on the regulation of myoblast behaviors is unclarified. In this study, we investigated the effects of silibinin on immortalized murine myoblast C2C12 in the aspects of proliferation, migration, differentiation along with underlying mechanisms. The results show that silibinin at concentrations below 50 μM enhanced the migration and differentiation of C2C12 cells, but had no effect on cell proliferation. Silibinin significantly promoted the production of ROS, which appeared to play important roles in the migration and differentiation of the myoblasts. Interestingly, among ROS, the superoxide anion and hydroxyl radical were associated with the migration, whereas hydrogen peroxide contributed to the myogenic differentiation. We used ER agonist and antagonist to explore whether estrogen receptors (ERs), which are affected by silibinin treatment in the silibinin-enhanced C2C12 migration and differentiation. Migration was independent of ERs, whereas the differentiation was associated with decreased ERα activity. In summary, silibinin treatment increases ROS levels, leading to the promotion of migration and myogenic differentiation. Negative regulation ERα of differentiation but not of migration may suggest that ERα represses hydrogen peroxide generation. The effect of silibinin on myoblast migration and differentiation suggests that silibinin may have therapeutic benefits for muscle regeneration.
Collapse
Affiliation(s)
- Xinyu Long
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, People's Republic of China
| | - Yanfang Gao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, People's Republic of China
| | - Weiwei Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, People's Republic of China
| | - Xiaoling Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, People's Republic of China
| | - Toshihiko Hayashi
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, People's Republic of China.,Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, 2665-1, Nakanomachi, Hachioji, Tokyo, 192-0015, Japan
| | - Kazunori Mizuno
- Nippi Research Institute of Biomatrix, Ibaraki, 649-1211, Japan
| | - Shunji Hattori
- Nippi Research Institute of Biomatrix, Ibaraki, 649-1211, Japan
| | - Takashi Ikejima
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, People's Republic of China. .,Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development Liaoning Province, 103 Wenhua Road, Shenyang, 110016, Liaoning, China. .,China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, China.
| |
Collapse
|
14
|
Active Compound of Pharbitis Semen ( Pharbitis nil Seeds) Suppressed KRAS-Driven Colorectal Cancer and Restored Muscle Cell Function during Cancer Progression. Molecules 2020; 25:molecules25122864. [PMID: 32580297 PMCID: PMC7356163 DOI: 10.3390/molecules25122864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/13/2020] [Accepted: 06/18/2020] [Indexed: 12/27/2022] Open
Abstract
Kirsten rat sarcoma viral oncogene homolog (KRAS)-driven colorectal cancer (CRC) is notorious to target with drugs and has shown ineffective treatment response. The seeds of Pharbitis nil, also known as morning glory, have been used as traditional medicine in East Asia. We focused on whether Pharbitis nil seeds have a suppressive effect on mutated KRAS-driven CRC as well as reserving muscle cell functions during CRC progression. Seeds of Pharbitis nil (Pharbitis semen) were separated by chromatography and the active compound of Pharbitis semen (PN) was purified by HPLC. The compound PN efficiently suppressed the proliferation of mutated KRAS-driven CRC cells and their clonogenic potentials in a concentration-dependent manner. It also induced apoptosis of SW480 human colon cancer cells and cell cycle arrest at the G2/M phase. The CRC related pathways, including RAS/ERK and AKT/mTOR, were assessed and PN reduced the phosphorylation of AKT and mTOR. Furthermore, PN preserved muscle cell proliferation and myotube formation in cancer conditioned media. In summary, PN significantly suppressed mutated KRAS-driven cell growth and reserved muscle cell function. Based on the current study, PN could be considered as a promising starting point for the development of a nature-derived drug against KRAS-mutated CRC progression.
Collapse
|
15
|
Alushi B, Curini L, Christopher MR, Grubitzch H, Landmesser U, Amedei A, Lauten A. Calcific Aortic Valve Disease-Natural History and Future Therapeutic Strategies. Front Pharmacol 2020; 11:685. [PMID: 32477143 PMCID: PMC7237871 DOI: 10.3389/fphar.2020.00685] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 04/27/2020] [Indexed: 12/20/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is the most frequent heart valve disorder. It is characterized by an active remodeling process accompanied with valve mineralization, that results in a progressive aortic valve narrowing, significant restriction of the valvular area, and impairment of blood flow.The pathophysiology of CAVD is a multifaceted process, involving genetic factors, chronic inflammation, lipid deposition, and valve mineralization. Mineralization is strictly related to the inflammatory process in which both, innate, and adaptive immunity are involved. The underlying pathophysiological pathways that go from inflammation to calcification and, finally lead to severe stenosis, remain, however, incompletely understood. Histopathological studies are limited to patients with severe CAVD and no samples are available for longitudinal studies of disease progression. Therefore, alternative routes should be explored to investigate the pathogenesis and progression of CAVD.Recently, increasing evidence suggests that epigenetic markers such as non-coding RNAs are implicated in the landscape of phenotypical changes occurring in CAVD. Furthermore, the microbiome, an essential player in several diseases, including the cardiovascular ones, has recently been linked to the inflammation process occurring in CAVD. In the present review, we analyze and discuss the CAVD pathophysiology and future therapeutic strategies, focusing on the real and putative role of inflammation, calcification, and microbiome.
Collapse
Affiliation(s)
- Brunilda Alushi
- Department of Cardiology, Charite´ Universitätsmedizin Berlin and German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Department of General and Interventional Cardiology, Helios Klinikum Erfurt, Erfurt, Germany
| | - Lavinia Curini
- Department of Cardiology, Charite´ Universitätsmedizin Berlin and German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
| | - Mary Roxana Christopher
- Department of Cardiology, Charite´ Universitätsmedizin Berlin and German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Herko Grubitzch
- Berlin Institute of Health, Berlin, Germany
- Department of Cardiology, German Heart Centre Berlin (DHZB), Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Charite´ Universitätsmedizin Berlin and German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
- Sod of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi (AOUC), Florence, Italy
| | - Alexander Lauten
- Department of Cardiology, Charite´ Universitätsmedizin Berlin and German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Department of General and Interventional Cardiology, Helios Klinikum Erfurt, Erfurt, Germany
| |
Collapse
|
16
|
Sun YN, Huang JQ, Chen ZZ, Du M, Ren FZ, Luo J, Fang B. Amyotrophy Induced by a High-Fat Diet Is Closely Related to Inflammation and Protein Degradation Determined by Quantitative Phosphoproteomic Analysis in Skeletal Muscle of C57BL/6 J Mice. J Nutr 2020; 150:294-302. [PMID: 31618431 DOI: 10.1093/jn/nxz236] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 07/29/2019] [Accepted: 09/05/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Ectopic fat accumulation in skeletal muscle results in dysfunction and atrophy, but the underlying molecular mechanisms remain unclear. OBJECTIVE The aim of this study was to investigate the effects of a high-fat diet (HFD) in modulating the structure and energy metabolism of skeletal muscle and the underlying mechanisms in mice. METHODS Four-week-old male C57BL/6 J mice (n = 30) were allowed 1 wk for acclimatization. After 6 mice with low body weight were removed from the study, the remaining 24 mice were fed with a normal-fat diet (NFD; 10% energy from fat, n = 12) or an HFD (60% energy from fat, n = 12) for 24 wk. At the end of the experiment, serum glucose and lipid concentrations were measured, and skeletal muscle was collected for atrophy analysis, inflammation measurements, and phosphoproteomic analysis. RESULTS Compared with the NFD, the HFD increased (P < 0.05) body weight (35.8%), serum glucose (64.5%), and lipid (27.3%) concentrations, along with elevated (P < 0.05) expressions of the atrophy-related proteins muscle ring finger 1 (MURF1; 27.6%) and muscle atrophy F-box (MAFBX; 44.5%) in skeletal muscle. Phosphoproteomic analysis illustrated 64 proteins with differential degrees of phosphorylation between the HFD and NFD groups. These proteins were mainly involved in modulating cytoskeleton [adenylyl cyclase-associated protein 2 (CAP2) and actin-α skeletal muscle (ACTA1)], inflammation [NF-κB-activating protein (NKAP) and serine/threonine-protein kinase RIO3 (RIOK3)], glucose metabolism [Cdc42-interacting protein 4 (TRIP10); protein kinase C, and casein kinase II substrate protein 3 (PACSIN3)], and protein degradation [heat shock protein 90 kDa (HSP90AA1)]. The HFD-induced inhibitions of the insulin signaling pathway and activations of inflammation in skeletal muscle were verified by Western blot analysis. CONCLUSIONS Quantitative phosphoproteomic analysis in C57BL/6 J mice fed an NFD or HFD for 24 wk revealed that the phosphorylation of inflammatory proteins and proteins associated with glucose metabolism at specific serine residues may play critical roles in the regulation of skeletal muscle atrophy induced by an HFD. This work provides information regarding underlying molecular mechanisms for inflammation-induced dysfunction and atrophy in skeletal muscle.
Collapse
Affiliation(s)
- Ya-Nan Sun
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jia-Qiang Huang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zhong-Zhou Chen
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Min Du
- Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Fa-Zheng Ren
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing, China.,Beijing Laboratory of Food Quality and Safety, Beijing University of Agriculture, Beijing, China
| | - Jie Luo
- Beijing Laboratory of Food Quality and Safety, Beijing University of Agriculture, Beijing, China.,College of Food Science and Technology, Hunan Agricultural University, Changsha, China
| | - Bing Fang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
17
|
6-Bromoindirubin-3'-oxime intercepts GSK3 signaling to promote and enhance skeletal muscle differentiation affecting miR-206 expression in mice. Sci Rep 2019; 9:18091. [PMID: 31792344 PMCID: PMC6889408 DOI: 10.1038/s41598-019-54574-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 11/12/2019] [Indexed: 12/17/2022] Open
Abstract
Dystrophies are characterized by progressive skeletal muscle degeneration and weakness as consequence of their molecular abnormalities. Thus, new drugs for restoring skeletal muscle deterioration are critically needed. To identify new and alternative compounds with a functional role in skeletal muscle myogenesis, we screened a library of pharmacologically active compounds and selected the small molecule 6-bromoindirubin-3′-oxime (BIO) as an inhibitor of myoblast proliferation. Using C2C12 cells, we examined BIO’s effect during myoblast proliferation and differentiation showing that BIO treatment promotes transition from cell proliferation to myogenic differentiation through the arrest of cell cycle. Here, we show that BIO is able to promote myogenic differentiation in damaged myotubes in-vitro by enriching the population of newly formed skeletal muscle myotubes. Moreover, in-vivo experiments in CTX-damaged TA muscle confirmed the pro-differentiation capability of BIO as shown by the increasing of the percentage of myofibers with centralized nuclei as well as by the increasing of myofibers number. Additionally, we have identified a strong correlation of miR-206 with BIO treatment both in-vitro and in-vivo: the enhanced expression of miR-206 was observed in-vitro in BIO-treated proliferating myoblasts, miR-206 restored expression was observed in a forced miR-206 silencing conditions antagomiR-mediated upon BIO treatment, and in-vivo in CTX-injured muscles miR-206 enhanced expression was observed upon BIO treatment. Taken together, our results highlight the capacity of BIO to act as a positive modulator of skeletal muscle differentiation in-vitro and in-vivo opening up a new perspective for novel therapeutic targets to correct skeletal muscle defects.
Collapse
|
18
|
Andersson C, Lin H, Liu C, Levy D, Mitchell GF, Larson MG, Vasan RS. Integrated Multiomics Approach to Identify Genetic Underpinnings of Heart Failure and Its Echocardiographic Precursors. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 12:e002489. [DOI: 10.1161/circgen.118.002489] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background:
Heart failure (HF) may arise from alterations in metabolic, structural, and signaling pathways, but its genetic architecture is incompletely understood. To elucidate potential genetic contributors to cardiac remodeling and HF, we integrated genome-wide single-nucleotide polymorphisms, gene expression, and DNA methylation using a transomics analytical approach.
Methods:
We used robust rank aggregation (where the position of a certain gene in a rank order list [based on statistical significance level] is tested against a randomly shuffled rank order list) to derive an integrative transomic score for each annotated gene associated with a HF trait.
Results:
We evaluated ≤8372 FHS (Framingham Heart Study) participants (54% women; mean age, 55±17 years). Of these, 62 (0.7%) and 35 (0.4%) had prevalent HF with reduced ejection fraction and HF with preserved left ventricular ejection fraction, respectively. During a mean follow-up of 8.5 years (minimum–maximum, 0.005–18.6 years), 223 (2.7%) and 234 (2.8%) individuals developed incident HF with reduced ejection fraction and HF with reduced ejection fraction, respectively. Top genes included
MMP20
and
MTSS1
(promotes actin assembly at intercellular junctions) for left ventricular systolic function;
ITGA9
(receptor for
VCAM1
[vascular cell protein 1]) and
C5
for left ventricular remodeling;
NUP210
(expressed during myogenic differentiation) and
ANK1
(cytoskeletal protein) for diastolic function;
TSPAN16
and
RAB11FIP3
(involved in regulation of actin cytoskeleton) for prevalent HF with reduced ejection fraction;
ANKRD13D
and
TRIM69
for incident HF with reduced ejection fraction;
HPCAL1
and
PTTG1IP
for prevalent HF with reduced ejection fraction; and
ZNF146
(close to the
COX7A1
enzyme) and
ZFP3
(close to
SLC52A1
—the riboflavin transporter) for incident HF with reduced ejection fraction. We tested the HF-related top single-nucleotide polymorphisms in the UK biobank, where
rs77059055
in
TPM1
(minor allele frequency, 0.023; odds ratio, 0.83;
P
=0.002) remained statistically significant upon Bonferroni correction.
Conclusions:
Our integrative transomics approach offers insights into potential molecular and genetic contributors to HF and its precursors. Although several of our candidate genes have been implicated in HF in animal models, independent replication is warranted.
Collapse
Affiliation(s)
- Charlotte Andersson
- Framingham Heart Study, MA (C.A., H.L., C.L., D.L., M.G.L., R.S.V.)
- Department of Cardiology, Herlev and Gentofte Hospital, Herlev, Denmark (C.A.)
| | - Honghuang Lin
- Framingham Heart Study, MA (C.A., H.L., C.L., D.L., M.G.L., R.S.V.)
- Section of Computational Biomedicine, Department of Medicine (H.L.), Boston University School of Medicine, MA
| | - Chunyu Liu
- Framingham Heart Study, MA (C.A., H.L., C.L., D.L., M.G.L., R.S.V.)
- Department of Biostatistics (C.L., M.G.L.), Boston University School of Public Health, MA
| | - Daniel Levy
- Framingham Heart Study, MA (C.A., H.L., C.L., D.L., M.G.L., R.S.V.)
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (D.L.)
| | | | - Martin G. Larson
- Framingham Heart Study, MA (C.A., H.L., C.L., D.L., M.G.L., R.S.V.)
- Department of Biostatistics (C.L., M.G.L.), Boston University School of Public Health, MA
| | - Ramachandran S. Vasan
- Framingham Heart Study, MA (C.A., H.L., C.L., D.L., M.G.L., R.S.V.)
- Sections of Preventive Medicine and Epidemiology and Cardiology, Department of Medicine (R.S.V.), Boston University School of Medicine, MA
- Department of Epidemiology (R.S.V.), Boston University School of Public Health, MA
| |
Collapse
|
19
|
Kim SH, Yi SJ, Lee H, Kim JH, Oh MJ, Song EJ, Kim K, Jhun BH. β 2-Adrenergic receptor (β 2-AR) agonist formoterol suppresses differentiation of L6 myogenic cells by blocking PI3K-AKT pathway. Anim Cells Syst (Seoul) 2019; 23:18-25. [PMID: 30834155 PMCID: PMC6394304 DOI: 10.1080/19768354.2018.1561516] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/20/2018] [Accepted: 12/05/2018] [Indexed: 01/01/2023] Open
Abstract
β2-Adrenergic receptor (β2-AR) is implicated in muscle metabolic activities such as glycogen metabolism, glucose uptake, lipolysis and muscle growth. However, the functional role of β2-AR in the differentiation of skeletal muscle is largely unknown. Here, we examined the functional role of β2-AR in L6 myoblast differentiation using the long-term-acting β2-AR-specific agonist formoterol. We observed that formoterol treatment strongly suppressed L6 myoblast differentiation and the expression of myosin heavy chain (MHC) in a dose- and time-dependent manner. Showing that both long-acting agonist (formoterol) and short-acting agonist (terbutaline) inhibited the induction of MHC protein, whereas β2-AR antagonist (ICI-118,551) upregulated MHC expression, we clearly demonstrated that β2-AR is involved in L6 myoblast differentiation. Furthermore, our pharmacological inhibition study revealed that the PI3K–AKT pathway is the main signaling pathway for myotube formation. Formoterol inhibited the activation of PI3K–AKT signaling, but not that of ERK signaling. Moreover, formoterol selectively inhibited AKT activation by IGF-I, but not by insulin. Collectively, our findings reveal a previously undocumented role of β2-AR activation in modulating the differentiation of L6 myoblasts.
Collapse
Affiliation(s)
- So-Hyeon Kim
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, Republic of Korea
| | - Sun-Ju Yi
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Hyerim Lee
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Ji-Hyun Kim
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, Republic of Korea
| | - Myung-Ju Oh
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, Republic of Korea
| | - Eun-Ju Song
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, Republic of Korea
| | - Kyunghwan Kim
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Byung H Jhun
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
20
|
A transcriptome-wide association study identifies PALMD as a susceptibility gene for calcific aortic valve stenosis. Nat Commun 2018; 9:988. [PMID: 29511167 PMCID: PMC5840407 DOI: 10.1038/s41467-018-03260-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 01/31/2018] [Indexed: 12/21/2022] Open
Abstract
Calcific aortic valve stenosis (CAVS) is a common and life-threatening heart disease and the current treatment options cannot stop or delay its progression. A GWAS on 1009 cases and 1017 ethnically matched controls was combined with a large-scale eQTL mapping study of human aortic valve tissues (n = 233) to identify susceptibility genes for CAVS. Replication was performed in the UK Biobank, including 1391 cases and 352,195 controls. A transcriptome-wide association study (TWAS) reveals PALMD (palmdelphin) as significantly associated with CAVS. The CAVS risk alleles and increasing disease severity are both associated with decreased mRNA expression levels of PALMD in valve tissues. The top variant identified shows a similar effect and strong association with CAVS (P = 1.53 × 10−10) in UK Biobank. The identification of PALMD as a susceptibility gene for CAVS provides insights into the genetic nature of this disease, opens avenues to investigate its etiology and to develop much-needed therapeutic options. Progressive remodeling and calcification of the aortic valve leads to calcific aortic valve stenosis (CAVS) and, ultimately, heart failure. In a combined GWAS and TWAS approach, Thériault et al. identify PALMD as a candidate causal gene for CAVS, which is further supported by Mendelian randomization.
Collapse
|