1
|
Tourel A, Reynaud-Dulaurier R, Brocard J, Fauré J, Marty I, Petiot A. RyR1 Is Involved in the Control of Myogenesis. Cells 2025; 14:158. [PMID: 39936950 PMCID: PMC11817019 DOI: 10.3390/cells14030158] [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: 12/14/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
The RyR1 calcium release channel is a key player in skeletal muscle excitation-contraction coupling. Mutations in the RYR1 gene are associated with congenital myopathies. Recently, a role of RyR1 in myotubes differentiation has been proposed and attributed to its calcium channel function, which nonetheless remains to be clearly demonstrated. In order to clarify RyR1 role in myogenesis, we have developed an in vitro model, the so-called RyR1-Rec myotubes, which are mouse primary myotubes with an inducible decrease in RyR1 protein amount and in RyR1-mediated calcium release. Using this model, we showed that the RyR1 protein decrease was responsible for an increase in both differentiation and fusion, from the RNA level to the morphological level, without affecting the myogenic factors MyoD and MyoG. Although an increase in mTOR pathway was observed in RyR1-Rec myotubes, it did not seem to be responsible for the role of RyR1 in myogenesis. Additionally, even if modulation of intracellular calcium level affected RyR1-Rec myotubes differentiation, we have shown that the role of RyR1 in myogenesis was independent of its calcium channel function. Therefore, our findings indicate that, besides its pivotal role as a calcium channel responsible for muscle contraction, RyR1 fulfills a calcium-independent inhibitor function of myogenesis.
Collapse
Affiliation(s)
| | | | | | | | - Isabelle Marty
- University Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institute Neurosciences, 38000 Grenoble, France (R.R.-D.); (J.B.); (J.F.)
| | - Anne Petiot
- University Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institute Neurosciences, 38000 Grenoble, France (R.R.-D.); (J.B.); (J.F.)
| |
Collapse
|
2
|
Jin H, Wang H, Wu J, Hu M, Zhou X, Yang S, Zhao A, He K. Asparagine synthetase regulates the proliferation and differentiation of chicken skeletal muscle satellite cells. Anim Biosci 2024; 37:1848-1862. [PMID: 39210809 PMCID: PMC11541025 DOI: 10.5713/ab.24.0271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/16/2024] [Accepted: 07/10/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE Asparagine synthetase (ASNS) is an aminotransferase responsible for the biosynthesis of aspartate by using aspartic acid and glutamine. ASNS is highly expressed in fast-growing broilers, but few studies have reported the regulatory role of ASNS in muscle development. METHODS To explore the function of ASNS in chicken muscle development, the expression of ASNS in different chicken breeds and tissues were first performed by real-time quantitative reverse transcription polymerase chain reaction (RT-PCR). Then, using real-time quantitative RT-PCR, western blot, EdU assay, cell cycle assay and immunofluorescence, the effects of ASNS on the proliferation and differentiation of chicken skeletal muscle satellite cell (SMSC) were investigated. Finally, potential mechanisms by which ASNS influences chicken muscle fiber differentiation were identified through RNA-Seq. RESULTS The mRNA expression pattern of ASNS in muscles mirrors trends in muscle fiber cross-sectional area, average daily weight gain, and muscle weight across different breeds. ASNS knockdown inhibited SMSC proliferation, while overexpression showed the opposite. Moreover, ASNS attenuated SMSC differentiation by activating the adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) pathway. Additionally, 5-aminoimidazole4-carboxamide1-β-D-ribofuranoside (AICAR) treatment suppressed the cell differentiation induced by siRNA-ASNS. RNA-Seq identified 1,968 differentially expressed genes (DEGs) during chicken SMSC differentiation when overexpression ASNS. Gene ontology (GO) enrichment analysis revealed that these DEGs primarily participated in 8 biological processes, 8 cellular components, and 4 molecular functions. Kyoto encyclopedia of genes and genomes (KEGG) pathway analysis identified several significantly enriched signaling pathways, such as the JAK-STAT signaling pathway, tumor necrosis factor signaling pathway, toll-like receptor signaling pathway, and PI3K-Akt signaling pathway. CONCLUSION ASNS promotes proliferation while inhibits the differentiation of chicken SMSCs. This study provides a theoretical basis for studying the role of ASNS in muscle development.
Collapse
Affiliation(s)
- Hangfeng Jin
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, 311300,
China
| | - Han Wang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, 311300,
China
| | - Jianqing Wu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, 311300,
China
| | - Moran Hu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, 311300,
China
| | - Xiaolong Zhou
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, 311300,
China
| | - Songbai Yang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, 311300,
China
| | - Ayong Zhao
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, 311300,
China
| | - Ke He
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, 311300,
China
| |
Collapse
|
3
|
Gu S, Huang Q, Jie Y, Sun C, Wen C, Yang N. Transcriptomic and epigenomic landscapes of muscle growth during the postnatal period of broilers. J Anim Sci Biotechnol 2024; 15:91. [PMID: 38961455 PMCID: PMC11223452 DOI: 10.1186/s40104-024-01049-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 05/12/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Broilers stand out as one of the fastest-growing livestock globally, making a substantial contribution to animal meat production. However, the molecular and epigenetic mechanisms underlying the rapid growth and development of broiler chickens are still unclear. This study aims to explore muscle development patterns and regulatory networks during the postnatal rapid growth phase of fast-growing broilers. We measured the growth performance of Cornish (CC) and White Plymouth Rock (RR) over a 42-d period. Pectoral muscle samples from both CC and RR were randomly collected at day 21 after hatching (D21) and D42 for RNA-seq and ATAC-seq library construction. RESULTS The consistent increase in body weight and pectoral muscle weight across both breeds was observed as they matured, with CC outpacing RR in terms of weight at each stage of development. Differential expression analysis identified 398 and 1,129 genes in the two dimensions of breeds and ages, respectively. A total of 75,149 ATAC-seq peaks were annotated in promoter, exon, intron and intergenic regions, with a higher number of peaks in the promoter and intronic regions. The age-biased genes and breed-biased genes of RNA-seq were combined with the ATAC-seq data for subsequent analysis. The results spotlighted the upregulation of ACTC1 and FDPS at D21, which were primarily associated with muscle structure development by gene cluster enrichment. Additionally, a noteworthy upregulation of MUSTN1, FOS and TGFB3 was spotted in broiler chickens at D42, which were involved in cell differentiation and muscle regeneration after injury, suggesting a regulatory role of muscle growth and repair. CONCLUSIONS This work provided a regulatory network of postnatal broiler chickens and revealed ACTC1 and MUSTN1 as the key responsible for muscle development and regeneration. Our findings highlight that rapid growth in broiler chickens triggers ongoing muscle damage and subsequent regeneration. These findings provide a foundation for future research to investigate the functional aspects of muscle development.
Collapse
Affiliation(s)
- Shuang Gu
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology China Agricultural University, Beijing, 100193, China
| | - Qiang Huang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology China Agricultural University, Beijing, 100193, China
| | - Yuchen Jie
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology China Agricultural University, Beijing, 100193, China
| | - Congjiao Sun
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology China Agricultural University, Beijing, 100193, China
- Sanya Institute of China Agricultural University, Hainan, 572025, China
| | - Chaoliang Wen
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology China Agricultural University, Beijing, 100193, China
- Sanya Institute of China Agricultural University, Hainan, 572025, China
| | - Ning Yang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China.
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology China Agricultural University, Beijing, 100193, China.
- Sanya Institute of China Agricultural University, Hainan, 572025, China.
| |
Collapse
|
4
|
Wang Y, Yang C, Wen J, Ju L, Ren Z, Zhang T, Liu Y. Whole-exome sequencing combined with postoperative data identify c.1614dup (CAMKK2) as a novel candidate monogenic obesity variant. Front Endocrinol (Lausanne) 2024; 15:1334342. [PMID: 38469147 PMCID: PMC10925648 DOI: 10.3389/fendo.2024.1334342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/09/2024] [Indexed: 03/13/2024] Open
Abstract
Early-onset obesity is a rising health concern influenced by heredity. However, many monogenic obesity variants (MOVs) remain to be discovered due to differences in ethnicity and culture. Additionally, patients with known MOVs have shown limited weight loss after bariatric surgery, suggesting it can be used as a screening tool for new candidates. In this study, we performed whole-exome sequencing (WES) combined with postoperative data to detect candidate MOVs in a cohort of 62 early-onset obesity and 9 late-onset obesity patients. Our findings demonstrated that patients with early-onset obesity preferred a higher BMI and waist circumference (WC). We confirmed the efficacy of the method by identifying a mutation in known monogenic obesity gene, PCSK1, which resulted in less weight loss after surgery. 5 genes were selected for further verification, and a frameshift variant in CAMKK2 gene: NM_001270486.1, c.1614dup, (p. Gly539Argfs*3) was identified as a novel candidate MOV. This mutation influenced the improvement of metabolism after bariatric surgery. In conclusion, our data confirm the efficacy of WES combined with postoperative data in detecting novel candidate MOVs and c.1614dup (CAMKK2) might be a promising MOV, which needs further confirmation. This study enriches the human monogenic obesity mutation database and provides a scientific basis for clinically accurate diagnosis and treatment.
Collapse
Affiliation(s)
- Yan Wang
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- Medical Research Center, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Chao Yang
- West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jun Wen
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Lingling Ju
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Zhengyun Ren
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Tongtong Zhang
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- Medical Research Center, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Yanjun Liu
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
5
|
Fan D, Yao Y, Liu Y, Yan C, Li F, Wang S, Yu M, Xie B, Tang Z. Regulation of myo-miR-24-3p on the Myogenesis and Fiber Type Transformation of Skeletal Muscle. Genes (Basel) 2024; 15:269. [PMID: 38540328 PMCID: PMC10970682 DOI: 10.3390/genes15030269] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 06/15/2024] Open
Abstract
Skeletal muscle plays critical roles in providing a protein source and contributing to meat production. It is well known that microRNAs (miRNAs) exert important effects on various biological processes in muscle, including cell fate determination, muscle fiber morphology, and structure development. However, the role of miRNA in skeletal muscle development remains incompletely understood. In this study, we observed a critical miRNA, miR-24-3p, which exhibited higher expression levels in Tongcheng (obese-type) pigs compared to Landrace (lean-type) pigs. Furthermore, we found that miR-24-3p was highly expressed in the dorsal muscle of pigs and the quadriceps muscle of mice. Functionally, miR-24-3p was found to inhibit proliferation and promote differentiation in muscle cells. Additionally, miR-24-3p was shown to facilitate the conversion of slow muscle fibers to fast muscle fibers and influence the expression of GLUT4, a glucose transporter. Moreover, in a mouse model of skeletal muscle injury, we demonstrated that overexpression of miR-24-3p promoted rapid myogenesis and contributed to skeletal muscle regeneration. Furthermore, miR-24-3p was found to regulate the expression of target genes, including Nek4, Pim1, Nlk, Pskh1, and Mapk14. Collectively, our findings provide evidence that miR-24-3p plays a regulatory role in myogenesis and fiber type conversion. These findings contribute to our understanding of human muscle health and have implications for improving meat production traits in livestock.
Collapse
Affiliation(s)
- Danyang Fan
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (D.F.); (Y.L.); (M.Y.)
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (C.Y.); (F.L.); (S.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China;
| | - Yilong Yao
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China;
| | - Yanwen Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (D.F.); (Y.L.); (M.Y.)
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (C.Y.); (F.L.); (S.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China;
| | - Chao Yan
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (C.Y.); (F.L.); (S.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China;
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Fanqinyu Li
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (C.Y.); (F.L.); (S.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China;
| | - Shilong Wang
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (C.Y.); (F.L.); (S.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China;
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Mei Yu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (D.F.); (Y.L.); (M.Y.)
| | - Bingkun Xie
- Guangxi Key Laboratory of Livestock Genetic Improvement, Guangxi Institute of Animal Sciences, Nanning 530001, China;
| | - Zhonglin Tang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (D.F.); (Y.L.); (M.Y.)
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (C.Y.); (F.L.); (S.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China;
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| |
Collapse
|
6
|
Hong L, Xu D, Li W, Wang Y, Cao N, Fu X, Tian Y, Li Y, Li B. Non-coding RNA regulation of Magang geese skeletal muscle maturation via the MAPK signaling pathway. Front Physiol 2024; 14:1331974. [PMID: 38314139 PMCID: PMC10834734 DOI: 10.3389/fphys.2023.1331974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/30/2023] [Indexed: 02/06/2024] Open
Abstract
Skeletal muscle is a critical component of goose meat and a significant economic trait of geese. The regulatory roles of miRNAs and lncRNAs in the maturation stage of goose skeletal muscle are still unclear. Therefore, this study conducted experiments on the leg muscles of Magang geese at two stages: 3-day post-hatch (P3) and 3 months (M3). Morphological observations revealed that from P3 to M3, muscle fibers mainly underwent hypertrophy and maturation. The muscle fibers became thicker, nuclear density decreased, and nuclei moved towards the fiber edges. Additionally, this study analyzed the expression profiles of lncRNAs, miRNAs, and mRNAs during the skeletal muscle fiber maturation stage, identifying 1,949 differentially expressed mRNAs (DEMs), 21 differentially expressed miRNAs (DEMIs), and 172 differentially expressed lncRNAs (DELs). Furthermore, we performed enrichment analyses on DEMs, cis-regulatory genes of DELs, and target DEMs of DEMIs, revealing significant enrichment of signaling pathways including MAPK, PPAR, and mTOR signaling pathways. Among these, the MAPK signaling pathway was the only pathway enriched across all three types of differentially expressed RNAs, indicating its potentially more significant role in skeletal muscle maturation. Finally, this study integrated the targeting relationships between DELs, DEMs, and DEMIs from these two stages to construct a ceRNA regulatory network. These findings unveil the potential functions and mechanisms of lncRNAs and miRNAs in the growth and development of goose skeletal muscle and provide valuable references for further exploration of the mechanism underlying the maturation of Magang geese leg muscle.
Collapse
Affiliation(s)
- Longsheng Hong
- College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Danning Xu
- College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Wanyan Li
- College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Yifeng Wang
- College of Computer Science and Software Engineering, Shenzhen University, Shenzhen, China
| | - Nan Cao
- College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Xinliang Fu
- College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Yunbo Tian
- College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Yugu Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Bingxin Li
- College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| |
Collapse
|
7
|
Jacques M, Landen S, Romero JA, Hiam D, Schittenhelm RB, Hanchapola I, Shah AD, Voisin S, Eynon N. Methylome and proteome integration in human skeletal muscle uncover group and individual responses to high-intensity interval training. FASEB J 2023; 37:e23184. [PMID: 37698381 DOI: 10.1096/fj.202300840rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/17/2023] [Accepted: 08/24/2023] [Indexed: 09/13/2023]
Abstract
Exercise is a major beneficial contributor to muscle metabolism, and health benefits acquired by exercise are a result of molecular shifts occurring across multiple molecular layers (i.e., epigenome, transcriptome, and proteome). Identifying robust, across-molecular level targets associated with exercise response, at both group and individual levels, is paramount to develop health guidelines and targeted health interventions. Sixteen, apparently healthy, moderately trained (VO2 max = 51.0 ± 10.6 mL min-1 kg-1 ) males (age range = 18-45 years) from the Gene SMART (Skeletal Muscle Adaptive Responses to Training) study completed a longitudinal study composed of 12-week high-intensity interval training (HIIT) intervention. Vastus lateralis muscle biopsies were collected at baseline and after 4, 8, and 12 weeks of HIIT. DNA methylation (~850 CpG sites) and proteomic (~3000 proteins) analyses were conducted at all time points. Mixed models were applied to estimate group and individual changes, and methylome and proteome integration was conducted using a holistic multilevel approach with the mixOmics package. A total of 461 proteins significantly changed over time (at 4, 8, and 12 weeks), whilst methylome overall shifted with training only one differentially methylated position (DMP) was significant (adj.p-value < .05). K-means analysis revealed cumulative protein changes by clusters of proteins that presented similar changes over time. Individual responses to training were observed in 101 proteins. Seven proteins had large effect-sizes >0.5, among them are two novel exercise-related proteins, LYRM7 and EPN1. Integration analysis showed bidirectional relationships between the methylome and proteome. We showed a significant influence of HIIT on the epigenome and more so on the proteome in human muscle, and uncovered groups of proteins clustering according to similar patterns across the exercise intervention. Individual responses to exercise were observed in the proteome with novel mitochondrial and metabolic proteins consistently changed across individuals. Future work is required to elucidate the role of these proteins in response to exercise.
Collapse
Affiliation(s)
- Macsue Jacques
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Victoria, Australia
| | - Shanie Landen
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Victoria, Australia
| | - Javier Alvarez Romero
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Victoria, Australia
| | - Danielle Hiam
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Victoria, Australia
- Institute of Nutrition and Health Sciences, Deakin University, Melbourne, Victoria, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics & Metabolomics Facility, Monash University, Melbourne, Victoria, Australia
| | - Iresha Hanchapola
- Monash Proteomics & Metabolomics Facility, Monash University, Melbourne, Victoria, Australia
| | - Anup D Shah
- Monash Proteomics & Metabolomics Facility, Monash University, Melbourne, Victoria, Australia
| | - Sarah Voisin
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nir Eynon
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Victoria, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
8
|
Negoita F, Addinsall AB, Hellberg K, Bringas CF, Hafen PS, Sermersheim TJ, Agerholm M, Lewis CTA, Ahwazi D, Ling NXY, Larsen JK, Deshmukh AS, Hossain MA, Oakhill JS, Ochala J, Brault JJ, Sankar U, Drewry DH, Scott JW, Witczak CA, Sakamoto K. CaMKK2 is not involved in contraction-stimulated AMPK activation and glucose uptake in skeletal muscle. Mol Metab 2023; 75:101761. [PMID: 37380024 PMCID: PMC10362367 DOI: 10.1016/j.molmet.2023.101761] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 06/30/2023] Open
Abstract
OBJECTIVE The AMP-activated protein kinase (AMPK) gets activated in response to energetic stress such as contractions and plays a vital role in regulating various metabolic processes such as insulin-independent glucose uptake in skeletal muscle. The main upstream kinase that activates AMPK through phosphorylation of α-AMPK Thr172 in skeletal muscle is LKB1, however some studies have suggested that Ca2+/calmodulin-dependent protein kinase kinase 2 (CaMKK2) acts as an alternative kinase to activate AMPK. We aimed to establish whether CaMKK2 is involved in activation of AMPK and promotion of glucose uptake following contractions in skeletal muscle. METHODS A recently developed CaMKK2 inhibitor (SGC-CAMKK2-1) alongside a structurally related but inactive compound (SGC-CAMKK2-1N), as well as CaMKK2 knock-out (KO) mice were used. In vitro kinase inhibition selectivity and efficacy assays, as well as cellular inhibition efficacy analyses of CaMKK inhibitors (STO-609 and SGC-CAMKK2-1) were performed. Phosphorylation and activity of AMPK following contractions (ex vivo) in mouse skeletal muscles treated with/without CaMKK inhibitors or isolated from wild-type (WT)/CaMKK2 KO mice were assessed. Camkk2 mRNA in mouse tissues was measured by qPCR. CaMKK2 protein expression was assessed by immunoblotting with or without prior enrichment of calmodulin-binding proteins from skeletal muscle extracts, as well as by mass spectrometry-based proteomics of mouse skeletal muscle and C2C12 myotubes. RESULTS STO-609 and SGC-CAMKK2-1 were equally potent and effective in inhibiting CaMKK2 in cell-free and cell-based assays, but SGC-CAMKK2-1 was much more selective. Contraction-stimulated phosphorylation and activation of AMPK were not affected with CaMKK inhibitors or in CaMKK2 null muscles. Contraction-stimulated glucose uptake was comparable between WT and CaMKK2 KO muscle. Both CaMKK inhibitors (STO-609 and SGC-CAMKK2-1) and the inactive compound (SGC-CAMKK2-1N) significantly inhibited contraction-stimulated glucose uptake. SGC-CAMKK2-1 also inhibited glucose uptake induced by a pharmacological AMPK activator or insulin. Relatively low levels of Camkk2 mRNA were detected in mouse skeletal muscle, but neither CaMKK2 protein nor its derived peptides were detectable in mouse skeletal muscle tissue. CONCLUSIONS We demonstrate that pharmacological inhibition or genetic loss of CaMKK2 does not affect contraction-stimulated AMPK phosphorylation and activation, as well as glucose uptake in skeletal muscle. Previously observed inhibitory effect of STO-609 on AMPK activity and glucose uptake is likely due to off-target effects. CaMKK2 protein is either absent from adult murine skeletal muscle or below the detection limit of currently available methods.
Collapse
Affiliation(s)
- Florentina Negoita
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Alex B Addinsall
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Kristina Hellberg
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Conchita Fraguas Bringas
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Paul S Hafen
- Department of Anatomy, Cell Biology & Physiology, and Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana Center for Diabetes & Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Division of Science, Indiana University Purdue University Columbus, Columbus, IN 47203, USA
| | - Tyler J Sermersheim
- Department of Anatomy, Cell Biology & Physiology, and Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana Center for Diabetes & Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Marianne Agerholm
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Christopher T A Lewis
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Danial Ahwazi
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Naomi X Y Ling
- Metabolic Signalling, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Jeppe K Larsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Atul S Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Mohammad A Hossain
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jonathan S Oakhill
- Metabolic Signalling, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia; Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
| | - Julien Ochala
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Jeffrey J Brault
- Department of Anatomy, Cell Biology & Physiology, and Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Uma Sankar
- Department of Anatomy, Cell Biology & Physiology, and Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - David H Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - John W Scott
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Melbourne, VIC 3052, Australia; The Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, VIC 3052, Australia; St Vincent's Institute of Medical Research, Fitzroy, Melbourne, VIC 3065, Australia
| | - Carol A Witczak
- Department of Anatomy, Cell Biology & Physiology, and Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana Center for Diabetes & Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark; The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
9
|
Abbadessa G, Maniscalco E, Grasso L, Popara J, Di Scipio F, Franco F, Mancardi D, Pigozzi F, Borrione P, Berta GN, Racca S. Metformin Protects Rat Skeletal Muscle from Physical Exercise-Induced Injury. Biomedicines 2023; 11:2334. [PMID: 37760776 PMCID: PMC10525561 DOI: 10.3390/biomedicines11092334] [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: 07/09/2023] [Revised: 07/26/2023] [Accepted: 08/16/2023] [Indexed: 09/29/2023] Open
Abstract
Metformin (Met) is a drug commonly prescribed in type 2 diabetes mellitus. Its efficacy is due to the suppression of hepatic gluconeogenesis, enhancement of peripheral glucose uptake and lower glucose absorption by the intestine. Recent studies have reported Met efficacy in other clinical applications, such as age-related diseases. Despite the wide clinical use of Met, its mechanism of action on muscle and its effect on muscle performance are unclear. We investigated the effects of Met combined with training on physical performance (PP) in healthy rats receiving Met for 8 weeks while undergoing daily moderate exercise. We evaluated the following: PP through graded endurance exercise test performed before the beginning of the training protocol and 48 h before the end of the training period; blood ALT, AST, LDH and CK-MB levels in order to address muscle damage; and several blood and muscle myokines and the expression of factors believed to be involved in muscle adaptation to exercise. Our data demonstrate that Met does not improve the positive effects of exercise on performance, although it protects myocytes from exercise-induced damage. Moreover, given that Met positively affects exercise-induced muscle adaptation, our data support the idea of the therapeutic application of Met when muscle function and structure are compromised.
Collapse
Affiliation(s)
- Giuliana Abbadessa
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.M.); (L.G.); (J.P.); (F.D.S.); (F.F.); (D.M.); (S.R.)
| | - Eleonora Maniscalco
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.M.); (L.G.); (J.P.); (F.D.S.); (F.F.); (D.M.); (S.R.)
| | - Loredana Grasso
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.M.); (L.G.); (J.P.); (F.D.S.); (F.F.); (D.M.); (S.R.)
| | - Jasmin Popara
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.M.); (L.G.); (J.P.); (F.D.S.); (F.F.); (D.M.); (S.R.)
| | - Federica Di Scipio
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.M.); (L.G.); (J.P.); (F.D.S.); (F.F.); (D.M.); (S.R.)
| | - Francesco Franco
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.M.); (L.G.); (J.P.); (F.D.S.); (F.F.); (D.M.); (S.R.)
| | - Daniele Mancardi
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.M.); (L.G.); (J.P.); (F.D.S.); (F.F.); (D.M.); (S.R.)
| | - Fabio Pigozzi
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy; (F.P.); (P.B.)
| | - Paolo Borrione
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy; (F.P.); (P.B.)
| | - Giovanni Nicolao Berta
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.M.); (L.G.); (J.P.); (F.D.S.); (F.F.); (D.M.); (S.R.)
| | - Silvia Racca
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.M.); (L.G.); (J.P.); (F.D.S.); (F.F.); (D.M.); (S.R.)
| |
Collapse
|
10
|
Lu T, Zhu Y, Guo J, Mo Z, Zhou Q, Hu CY, Wang C. MDFI regulates fast-to-slow muscle fiber type transformation via the calcium signaling pathway. Biochem Biophys Res Commun 2023; 671:215-224. [PMID: 37307704 DOI: 10.1016/j.bbrc.2023.05.053] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 06/14/2023]
Abstract
Muscle fiber is the basic unit of skeletal muscle with strong self-adaptability, and its type is closely related to meat quality. Myod family inhibitor (Mdfi) has the function of regulating myogenic regulatory factors during cell differentiation, but how Mdfi regulates muscle fiber type transformation in myoblasts is still unclear. In the present study, we constructed overexpressing and interfering with Mdfi C2C12 cell models by lipofection. The immunofluorescence, quantitative real-time PCR (qPCR), and western blot results show that the elevated MDFI promoted mitochondrial biogenesis, aerobic metabolism and the calcium level by activating CaMKK2 and AMPK phosphorylation and then stimulated the conversion of C2C12 cells from fast glycolytic to slow oxidative type. In addition, after inhibiting IP3R and RYR channels, the higher MDFI reversed the blockage of calcium release from the endoplasmic reticulum by calcium channel receptor inhibitors and increased intracellular calcium levels. Therefore, we propose that the higher MDFI promotes muscle fiber types conversion through the calcium signaling pathway. These findings further broaden our understanding of the regulatory mechanism of MDFI in muscle fiber type transformation. Furthermore, our results suggest potential therapeutic targets for skeletal muscle and metabolic-related diseases.
Collapse
Affiliation(s)
- Tingting Lu
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yifan Zhu
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiali Guo
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Ziyuan Mo
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Quan Zhou
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Ching Yuan Hu
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Chong Wang
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
11
|
Metformin regulates myoblast differentiation through an AMPK-dependent mechanism. PLoS One 2023; 18:e0281718. [PMID: 36763621 PMCID: PMC9916624 DOI: 10.1371/journal.pone.0281718] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
This study aims to investigate how metformin (Met) affects muscle tissue by evaluating the drug effects on proliferating, differentiating, and differentiated C2C12 cells. Moreover, we also investigated the role of 5'-adenosine monophosphate-activated protein kinase (AMPK) in the mechanism of action of Met. C2C12 myoblasts were cultured in growth medium with or without Met (250μM, 1mM and 10mM) for different times. Cell proliferation was evaluated by MTT assay, while cell toxicity was assessed by Trypan Blue exclusion test and Lactate Dehydrogenase release. Fluorescence Activated Cell Sorting analysis was performed to study cell cycle. Differentiating myoblasts were incubated in differentiation medium (DM) with or without 10mM Met. For experiments on myotubes, C2C12 were induced to differentiate in DM, and then treated with Met at scalar concentrations and for different times. Western blotting was performed to evaluate the expression of proteins involved in myoblast differentiation, muscle function and metabolism. In differentiating C2C12, Met inhibited cell differentiation, arrested cell cycle progression in G2/M phase and reduced the expression of cyclin-dependent kinase inhibitor 1. These effects were accompanied by activation of AMPK and modulation of the myogenic regulatory factors. Comparable results were obtained in myotubes. The use of Compound C, a specific inhibitor of AMPK, counteracted the above-mentioned Met effects. We reported that Met inhibits C2C12 differentiation probably by blocking cell-cycle progression and preventing cells permanent exit from cell-cycle. Moreover, our study provides solid evidence that most of the effects of Met on myoblasts and myotubes are mediated by AMPK.
Collapse
|
12
|
Wells C, Liang Y, Pulliam TL, Lin C, Awad D, Eduful B, O’Byrne S, Hossain MA, Catta-Preta CMC, Ramos PZ, Gileadi O, Gileadi C, Couñago RM, Stork B, Langendorf CG, Nay K, Oakhill JS, Mukherjee D, Racioppi L, Means AR, York B, McDonnell DP, Scott JW, Frigo DE, Drewry DH. SGC-CAMKK2-1: A Chemical Probe for CAMKK2. Cells 2023; 12:287. [PMID: 36672221 PMCID: PMC9856672 DOI: 10.3390/cells12020287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/31/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
The serine/threonine protein kinase calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) plays critical roles in a range of biological processes. Despite its importance, only a handful of inhibitors of CAMKK2 have been disclosed. Having a selective small molecule tool to interrogate this kinase will help demonstrate that CAMKK2 inhibition can be therapeutically beneficial. Herein, we disclose SGC-CAMKK2-1, a selective chemical probe that targets CAMKK2.
Collapse
Affiliation(s)
- Carrow Wells
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yi Liang
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Thomas L. Pulliam
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Chenchu Lin
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Dominik Awad
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Benjamin Eduful
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sean O’Byrne
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mohammad Anwar Hossain
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Carolina Moura Costa Catta-Preta
- Centro de Química Medicinal (CQMED), Centro de Biologia Molecular e Engenharia Genética (CBMEG), Universidade Estadual de Campinas (UNICAMP), Campinas 13083-886, Brazil
| | - Priscila Zonzini Ramos
- Centro de Química Medicinal (CQMED), Centro de Biologia Molecular e Engenharia Genética (CBMEG), Universidade Estadual de Campinas (UNICAMP), Campinas 13083-886, Brazil
| | - Opher Gileadi
- Centro de Química Medicinal (CQMED), Centro de Biologia Molecular e Engenharia Genética (CBMEG), Universidade Estadual de Campinas (UNICAMP), Campinas 13083-886, Brazil
| | - Carina Gileadi
- Centro de Química Medicinal (CQMED), Centro de Biologia Molecular e Engenharia Genética (CBMEG), Universidade Estadual de Campinas (UNICAMP), Campinas 13083-886, Brazil
| | - Rafael M. Couñago
- Centro de Química Medicinal (CQMED), Centro de Biologia Molecular e Engenharia Genética (CBMEG), Universidade Estadual de Campinas (UNICAMP), Campinas 13083-886, Brazil
| | - Brittany Stork
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Kevin Nay
- St Vincent’s Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC 3052, Australia
| | | | - Debarati Mukherjee
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27705, USA
| | - Luigi Racioppi
- Department of Medicine, Division of Hematological Malignancies and Cellular Therapy, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Anthony R. Means
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Brian York
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Donald P. McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27705, USA
| | - John W. Scott
- St Vincent’s Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC 3052, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia
| | - Daniel E. Frigo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77204, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - David H. Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
13
|
Wang Y, Lu J, Liu Y. Skeletal Muscle Regeneration in Cardiotoxin-Induced Muscle Injury Models. Int J Mol Sci 2022; 23:ijms232113380. [PMID: 36362166 PMCID: PMC9657523 DOI: 10.3390/ijms232113380] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Skeletal muscle injuries occur frequently in daily life and exercise. Understanding the mechanisms of regeneration is critical for accelerating the repair and regeneration of muscle. Therefore, this article reviews knowledge on the mechanisms of skeletal muscle regeneration after cardiotoxin-induced injury. The process of regeneration is similar in different mouse strains and is inhibited by aging, obesity, and diabetes. Exercise, microcurrent electrical neuromuscular stimulation, and mechanical loading improve regeneration. The mechanisms of regeneration are complex and strain-dependent, and changes in functional proteins involved in the processes of necrotic fiber debris clearance, M1 to M2 macrophage conversion, SC activation, myoblast proliferation, differentiation and fusion, and fibrosis and calcification influence the final outcome of the regenerative activity.
Collapse
|
14
|
Yan Y, Li M, Lin J, Ji Y, Wang K, Yan D, Shen Y, Wang W, Huang Z, Jiang H, Sun H, Qi L. Adenosine monophosphate activated protein kinase contributes to skeletal muscle health through the control of mitochondrial function. Front Pharmacol 2022; 13:947387. [PMID: 36339617 PMCID: PMC9632297 DOI: 10.3389/fphar.2022.947387] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 10/06/2022] [Indexed: 11/26/2022] Open
Abstract
Skeletal muscle is one of the largest organs in the body and the largest protein repository. Mitochondria are the main energy-producing organelles in cells and play an important role in skeletal muscle health and function. They participate in several biological processes related to skeletal muscle metabolism, growth, and regeneration. Adenosine monophosphate-activated protein kinase (AMPK) is a metabolic sensor and regulator of systemic energy balance. AMPK is involved in the control of energy metabolism by regulating many downstream targets. In this review, we propose that AMPK directly controls several facets of mitochondrial function, which in turn controls skeletal muscle metabolism and health. This review is divided into four parts. First, we summarize the properties of AMPK signal transduction and its upstream activators. Second, we discuss the role of mitochondria in myogenesis, muscle atrophy, regeneration post-injury of skeletal muscle cells. Third, we elaborate the effects of AMPK on mitochondrial biogenesis, fusion, fission and mitochondrial autophagy, and discuss how AMPK regulates the metabolism of skeletal muscle by regulating mitochondrial function. Finally, we discuss the effects of AMPK activators on muscle disease status. This review thus represents a foundation for understanding this biological process of mitochondrial dynamics regulated by AMPK in the metabolism of skeletal muscle. A better understanding of the role of AMPK on mitochondrial dynamic is essential to improve mitochondrial function, and hence promote skeletal muscle health and function.
Collapse
Affiliation(s)
- Yan Yan
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Ming Li
- Department of Laboratory Medicine, Binhai County People’s Hospital Affiliated to Kangda College of Nanjing Medical University, Yancheng, China
| | - Jie Lin
- Department of Infectious Disease, Affiliated Hospital of Nantong University, Nantong, China
| | - Yanan Ji
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Kexin Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Dajun Yan
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Wei Wang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Department of Pathology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Zhongwei Huang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Haiyan Jiang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Haiyan Jiang, ; Hualin Sun, ; Lei Qi,
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
- *Correspondence: Haiyan Jiang, ; Hualin Sun, ; Lei Qi,
| | - Lei Qi
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Haiyan Jiang, ; Hualin Sun, ; Lei Qi,
| |
Collapse
|
15
|
Molecular Mechanisms Underlying Ca2+/Calmodulin-Dependent Protein Kinase Kinase Signal Transduction. Int J Mol Sci 2022; 23:ijms231911025. [PMID: 36232320 PMCID: PMC9570080 DOI: 10.3390/ijms231911025] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 12/03/2022] Open
Abstract
Ca2+/calmodulin-dependent protein kinase kinase (CaMKK) is the activating kinase for multiple downstream kinases, including CaM-kinase I (CaMKI), CaM-kinase IV (CaMKIV), protein kinase B (PKB/Akt), and 5′AMP-kinase (AMPK), through the phosphorylation of their activation-loop Thr residues in response to increasing the intracellular Ca2+ concentration, as CaMKK itself is a Ca2+/CaM-dependent enzyme. The CaMKK-mediated kinase cascade plays important roles in a number of Ca2+-dependent pathways, such as neuronal morphogenesis and plasticity, transcriptional activation, autophagy, and metabolic regulation, as well as in pathophysiological pathways, including cancer progression, metabolic syndrome, and mental disorders. This review focuses on the molecular mechanism underlying CaMKK-mediated signal transduction in normal and pathophysiological conditions. We summarize the current knowledge of the structural, functional, and physiological properties of the regulatory kinase, CaMKK, and the development and application of its pharmacological inhibitors.
Collapse
|
16
|
Targeting CAMKK2 and SOC Channels as a Novel Therapeutic Approach for Sensitizing Acute Promyelocytic Leukemia Cells to All-Trans Retinoic Acid. Cells 2021; 10:cells10123364. [PMID: 34943872 PMCID: PMC8699360 DOI: 10.3390/cells10123364] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 12/18/2022] Open
Abstract
Calcium ions (Ca2+) play important and diverse roles in the regulation of autophagy, cell death and differentiation. Here, we investigated the impact of Ca2+ in regulating acute promyelocytic leukemia (APL) cell fate in response to the anti-cancer agent all-trans retinoic acid (ATRA). We observed that ATRA promotes calcium entry through store-operated calcium (SOC) channels into acute promyelocytic leukemia (APL) cells. This response is associated with changes in the expression profiles of ORAI1 and STIM1, two proteins involved in SOC channels activation, as well as with a significant upregulation of several key proteins associated to calcium signaling. Moreover, ATRA treatment of APL cells led to a significant activation of calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) and its downstream effector AMP-activated protein kinase (AMPK), linking Ca2+ signaling to autophagy. Pharmacological inhibition of SOC channels and CAMKK2 enhanced ATRA-induced cell differentiation and death. Altogether, our results unravel an ATRA-elicited signaling pathway that involves SOC channels/CAMKK2 activation, induction of autophagy, inhibition of cellular differentiation and suppression of cell death. We suggest that SOC channels and CAMKK2 may constitute novel drug targets for potentiating the anti-cancer effect of ATRA in APL patients.
Collapse
|
17
|
Jing J, Jiang X, Zhu C, Zheng Q, Ji Q, Yin H, Huang J, Zhu Y, Wang J, Qin S, Ling Y. Dynamic changes of miRNAs in skeletal muscle development at New Zealand rabbits. BMC Genomics 2021; 22:577. [PMID: 34315409 PMCID: PMC8314457 DOI: 10.1186/s12864-021-07896-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND miRNA is one of the crucial roles in the complex and dynamic network that regulates the development of skeletal muscle. The landscape of skeletal muscle miRNAs from fetus to adult in New Zealand rabbits has not been revealed yet. RESULTS In this study, nine RNA-seq libraries of fetus, child and adult rabbits' leg muscles were constructed. A total of 278 differentially expressed miRNAs (DEmiRNAs) were identified. In the fetus vs. child group, the main functional enrichments were involved in membrane and transport. Pathway enriched terms of up-regulated DEmiRNAs were connected with the differentiation and hypertrophy of skeletal muscle, and down-regulated ones were related to muscle structure and metabolic capacity. In the child vs. adult group, functions were associated to positioning and transportation, and pathways were relevant to ECM, muscle structure and hypertrophy. Finally, ocu-miR-185-3p and ocu-miR-370-3p, which had the most target genes, were identified as hub-miRNAs in these two groups. CONCLUSIONS In short, we summarized the highly expressed and uniquely expressed DEmiRNAs of fetus, child and adult rabbits' leg muscles. Besides, the potential functional changes of miRNAs in two consecutive stages have been explored. Among them, the ocu-miR-185-3p and ocu-miR-370-3p with the most target genes were selected as hub-miRNAs. These data improved the understanding of the regulatory molecules of meat rabbit development, and provided a novel perspective for molecular breeding of meat rabbits.
Collapse
Affiliation(s)
- Jing Jing
- College of Animal Science and Technology, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China.,Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio- Breeding, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China
| | - Xichun Jiang
- Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Anhui, 230031, Hefei, People's Republic of China
| | - Cuiyun Zhu
- College of Animal Science and Technology, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China.,Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio- Breeding, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China
| | - Qi Zheng
- College of Animal Science and Technology, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China.,Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio- Breeding, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China
| | - Qianyun Ji
- College of Animal Science and Technology, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China.,Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio- Breeding, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China
| | - Huiqun Yin
- Reproductive Medicine Center, The 901st Hospital, Anhui, 230031, Hefei, People's Republic of China
| | - Jingtong Huang
- College of Animal Science and Technology, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China.,Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio- Breeding, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China
| | - Yixiao Zhu
- College of Animal Science and Technology, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China.,Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio- Breeding, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China
| | - Jiao Wang
- College of Animal Science and Technology, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China.,Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio- Breeding, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China
| | - Shuaiqi Qin
- College of Animal Science and Technology, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China.,Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio- Breeding, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China
| | - Yinghui Ling
- College of Animal Science and Technology, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China. .,Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio- Breeding, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China.
| |
Collapse
|
18
|
A complete map of the Calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) signaling pathway. J Cell Commun Signal 2020; 15:283-290. [PMID: 33136287 DOI: 10.1007/s12079-020-00592-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 10/19/2020] [Indexed: 12/16/2022] Open
Abstract
Calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) is a serine/threonine-protein kinase belonging to the Ca2+/calmodulin-dependent protein kinase subfamily. CAMKK2 has an autocatalytic site, which gets exposed when Ca2+/calmodulin (CAM) binds to it. This results in autophosphorylation and complete activation of CAMKK2. The three major known downstream targets of CAMKK2 are 5'-adenosine monophosphate (AMP)-activated protein kinase (AMPKα), calcium/calmodulin-dependent protein kinase 1 (CAMK1) and calcium/calmodulin-dependent protein kinase 4 (CAMK4). Activation of these targets by CAMKK2 is important for the maintenance of different cellular and physiological processes within the cell. CAMKK2 is found to be important in neuronal development, bone remodeling, adipogenesis, and systemic glucose homeostasis, osteoclastgensis and postnatal myogensis. CAMKK2 is reported to be involved in pathologies like Duchenne muscular dystrophy, inflammation, osteoporosis and bone remodeling and is also reported to be overexpressed in prostate cancer, hepatic cancer, ovarian and gastric cancer. CAMKK2 is involved in increased cell proliferation and migration through CAMKK2/AMPK pathway in prostate cancer and activation of AKT in ovarian cancer. Although CAMKK2 is a molecule of great importance, a public resource of the CAMKK2 signaling pathway is currently lacking. Therefore, we carried out detailed data mining and documentation of the signaling events associated with CAMKK2 from published literature and developed an integrated reaction map of CAMKK2 signaling. This resulted in the cataloging of 285 reactions belonging to the CAMKK2 signaling pathway, which includes 33 protein-protein interactions, 74 post-translational modifications, 7 protein translocation events, and 22 activation/inhibition events. Besides, 124 gene regulation events and 25 activator/inhibitors involved in CAMKK2 activation were also cataloged. The CAMKK2 signaling pathway map data is made freely accessible through WikiPathway database ( https://www.wikipathways.org/index.php/Pathway:WP4874 ). We expect that data on a signaling map of CAMKK2 will provide the scientific community with an improved platform to facilitate further molecular as well as biomedical investigations on CAMKK2 and its utility in the development of biomarkers and therapeutic targets.
Collapse
|
19
|
Kun L, Lu L, Yongda L, Xingyue L, Guang H. Hyperbaric oxygen promotes mitophagy by activating CaMKK β/AMPK signal pathway in rats of neuropathic pain. Mol Pain 2020; 15:1744806919871381. [PMID: 31382832 PMCID: PMC6710678 DOI: 10.1177/1744806919871381] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Liu Kun
- 1 Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Li Lu
- 1 Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Liu Yongda
- 1 Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Li Xingyue
- 1 Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Han Guang
- 1 Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| |
Collapse
|
20
|
Torma F, Gombos Z, Jokai M, Takeda M, Mimura T, Radak Z. High intensity interval training and molecular adaptive response of skeletal muscle. SPORTS MEDICINE AND HEALTH SCIENCE 2019; 1:24-32. [PMID: 35782463 PMCID: PMC9219277 DOI: 10.1016/j.smhs.2019.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Increased cardiovascular fitness, V˙O2max, is associated with enhanced endurance capacity and a decreased rate of mortality. High intensity interval training (HIIT) is one of the best methods to increase V˙O2max and endurance capacity for top athletes and for the general public as well. Because of the high intensity of this type of training, the adaptive response is not restricted to Type I fibers, as found for moderate intensity exercise of long duration. Even with a short exercise duration, HIIT can induce activation of AMPK, PGC-1α, SIRT1 and ROS pathway as well as by the modulation of Ca2+ homeostasis, leading to enhanced mitochondrial biogenesis, and angiogenesis. The present review summarizes the current knowledge of the adaptive response of HIIT.
Collapse
Affiliation(s)
- Ferenc Torma
- Research Center of Molecular Exercise Science, University of Physical Education, Budapest, Hungary
| | - Zoltan Gombos
- Research Center of Molecular Exercise Science, University of Physical Education, Budapest, Hungary
| | - Matyas Jokai
- Research Center of Molecular Exercise Science, University of Physical Education, Budapest, Hungary
| | - Masaki Takeda
- Faculty of Health and Sports Science, Doshisha University, Kyotanabe, Japan
| | - Tatsuya Mimura
- Faculty of Sport and Health Sciences, Osaka Sangyo University, Osaka, Japan
| | - Zsolt Radak
- Research Center of Molecular Exercise Science, University of Physical Education, Budapest, Hungary
- Corresponding author. Alkotas u. 44, Budapest, H-1123, Hungary.
| |
Collapse
|
21
|
The Role of AMPK in the Regulation of Skeletal Muscle Size, Hypertrophy, and Regeneration. Int J Mol Sci 2018; 19:ijms19103125. [PMID: 30314396 PMCID: PMC6212977 DOI: 10.3390/ijms19103125] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 12/22/2022] Open
Abstract
AMPK (5’-adenosine monophosphate-activated protein kinase) is heavily involved in skeletal muscle metabolic control through its regulation of many downstream targets. Because of their effects on anabolic and catabolic cellular processes, AMPK plays an important role in the control of skeletal muscle development and growth. In this review, the effects of AMPK signaling, and those of its upstream activator, liver kinase B1 (LKB1), on skeletal muscle growth and atrophy are reviewed. The effect of AMPK activity on satellite cell-mediated muscle growth and regeneration after injury is also reviewed. Together, the current data indicate that AMPK does play an important role in regulating muscle mass and regeneration, with AMPKα1 playing a prominent role in stimulating anabolism and in regulating satellite cell dynamics during regeneration, and AMPKα2 playing a potentially more important role in regulating muscle degradation during atrophy.
Collapse
|
22
|
Nanjappa V, Sathe GJ, Jain AP, Rajagopalan P, Raja R, Subbannayya T, Patil AH, Kumar P, Prasad TSK, Mathur PP, Sidransky D, Gowda H, Chatterjee A. Investigation of curcumin-mediated signalling pathways in head and neck squamous cell carcinoma. TRANSLATIONAL RESEARCH IN ORAL ONCOLOGY 2017. [DOI: 10.1177/2057178x17743142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
| | - Gajanan J. Sathe
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal University, Madhav Nagar, Manipal, India
| | - Ankit P. Jain
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| | - Pavithra Rajagopalan
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| | - Remya Raja
- Institute of Bioinformatics, International Technology Park, Bangalore, India
| | | | - Arun H. Patil
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| | - Prashant Kumar
- Institute of Bioinformatics, International Technology Park, Bangalore, India
| | - T. S. Keshava Prasad
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- YU-IOB Center for Systems Biology and Molecular Medicine, Yenepoya University, Mangalore, India
- NIMHANS-IOB Proteomics and Bioinformatics Laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | - Premendu Prakash Mathur
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
- Department of Biochemistry & Molecular Biology, Pondicherry University, Pondicherry, India
| | - David Sidransky
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Harsha Gowda
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- YU-IOB Center for Systems Biology and Molecular Medicine, Yenepoya University, Mangalore, India
| | - Aditi Chatterjee
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- YU-IOB Center for Systems Biology and Molecular Medicine, Yenepoya University, Mangalore, India
| |
Collapse
|
23
|
Jianfei L, Min W, Chunlai M, Bicui C, Jiming Z, Bin W. The Ca 2+/CaMKK2 axis mediates the telbivudine induced upregulation of creatine kinase: Implications for mechanism of antiviral nucleoside analogs' side effect. Biochem Pharmacol 2017; 146:224-232. [PMID: 29038020 DOI: 10.1016/j.bcp.2017.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 10/12/2017] [Indexed: 12/24/2022]
Abstract
Telbivudine (LdT), a widely prescribed anti-hepatitis B virus (HBV) drug for the treatment of chronic Hepatitis B (CHB), causes adverse reactions ranging from creatine kinase (CK) elevation to myopathy. The purpose of this study was to explore the mechanism(s) of LdT induced CK elevation. The effects of LdT on mitochondrial morphology and proteins (TK2 and β-actin), oxidative stress, intracellular Ca2+ levels, Ca2+-related signaling pathway (CaMKK2/AMPK), and Ca2+-related biomarkers such as superoxide dismutase (SOD) and malondialdehyde (MDA) were assessed in human skeletal muscle cells (HSKMCs). The results showed that LdT induced a dose-dependent increase in CK activity in HSKMCs, without affecting mitochondrial morphology, and TK2 and β-actin protein levels, following 72 h of treatment. In addition, LdT increased Ca2+ production, ROS generation, MDA and lipid peroxide (LPO) levels, and activated the CaMKK2/AMPK signaling pathway. Moreover, these effects were attenuated by the BAPIA-AM (the calcium chelator). We also confirmed the presence of relevant markers (MDA, LPO, and SOD) in serum from CHB patients after LdT treatment, and found that CK was positively correlated with MDA and LPO, and negatively associated with SOD. These findings indicate that LdT induces CK elevation and oxidative stress associated with imbalance of intracellular Ca2+ in HSKMCs, suggesting that Ca2+/CaMKK2 axis imbalance may underlie human LdT-induced CK elevation. The present findings provide a solid basis for assessing the mechanism of drug-induced CK elevation, which can help develop new tools for the prevention and treatment of diseases associated with drug-induced CK elevation.
Collapse
Affiliation(s)
- Long Jianfei
- Department of Pharmacy, HuaShan Hospital, Fudan University, Shanghai, China
| | - Wang Min
- College of Pharmacy, Fudan University, Shanghai, China
| | - Ma Chunlai
- Department of Pharmacy, HuaShan Hospital, Fudan University, Shanghai, China
| | - Chen Bicui
- Department of Pharmacy, HuaShan Hospital, Fudan University, Shanghai, China
| | - Zhang Jiming
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China.
| | - Wang Bin
- Department of Pharmacy, HuaShan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|