1
|
Cai S, Yin N. Single-cell transcriptome and chromatin accessibility mapping of upper lip and primary palate fusion. J Cell Mol Med 2024; 28:e70128. [PMID: 39392189 PMCID: PMC11467802 DOI: 10.1111/jcmm.70128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/17/2024] [Accepted: 09/20/2024] [Indexed: 10/12/2024] Open
Abstract
Cleft lip and/or primary palate (CL/P) represent a prevalent congenital malformation, the aetiology of which is highly intricate. Although it is generally accepted that the condition arises from failed fusion between the upper lip and primary palate, the precise mechanism underlying this fusion process remains enigmatic. In this study, we utilized transposase-accessible chromatin sequencing (scATAC-seq) and single-cell RNA sequencing (scRNA-seq) to interrogate lambdoidal junction tissue derived from C57BL/6J mouse embryos at critical stages of embryogenesis (10.5, 11.5 and 12.5 embryonic days). We successfully identified distinct subgroups of mesenchymal and ectodermal cells involved in the fusion process and characterized their unique transcriptional profiles. Furthermore, we conducted cell differentiation trajectory analysis, revealing a dynamic repertoire of genes that are sequentially activated or repressed during pseudotime, facilitating the transition of relevant cell types. Additionally, we employed scATAC data to identify key genes associated with the fusion process and demonstrated differential chromatin accessibility across major cell types. Finally, we constructed a dynamic intercellular communication network and predicted upstream transcriptional regulators of critical genes involved in important signalling pathways. Our findings provide a valuable resource for future studies on upper lip and primary palate development, as well as congenital defects.
Collapse
Affiliation(s)
- Sini Cai
- The Department of Cleft Lip and Palate of Plastic Surgery HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Medical Cosmetic Center of Dermatology Hospital of Southern Medical UniversityGuangdong Provincial Dermatology HospitalGuangzhouChina
| | - Ningbei Yin
- The Department of Cleft Lip and Palate of Plastic Surgery HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
2
|
Wang M, Yang J, Wu Y, Li H, Zhong Y, Luo Y, Xie R. Curcumin-activated Wnt5a pathway mediates Ca 2+ channel opening to affect myoblast differentiation and skeletal muscle regeneration. J Cachexia Sarcopenia Muscle 2024; 15:1834-1849. [PMID: 38982896 PMCID: PMC11446719 DOI: 10.1002/jcsm.13535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 05/17/2024] [Accepted: 06/03/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Skeletal muscle injury is one of the most common sports injuries; if not properly treated or not effective rehabilitation treatment after injury, it can be transformed into chronic cumulative injury. Curcumin, an herbal ingredient, has been found to promote skeletal muscle injury repair and regeneration. The Wnt5a pathway is related to the expression of myogenic regulatory factors, and Ca2+ promotes the differentiation and fusion process of myoblasts. This study explored the effect and mechanism of curcumin on myoblast differentiation during the repair and regeneration of injured skeletal muscle and its relationship with the Wnt5a pathway and Ca2+ channel. METHODS Myogenic differentiation of C2C12 cells was induced with 2% horse serum, and a mouse (male, 10 weeks old) model of acute skeletal muscle injury was established using cardiotoxin (20 μL). In addition, we constructed a Wnt5a knockdown C2C12 cell model and a Wnt5a knockout mouse model. Besides, curcumin was added to the cell culture solution (80 mg/L) and fed to the mice (50 mg/kg). Fluorescence microscopy was used to determine the concentration of Ca2+. Western blot and RT-qPCR were used to detect the protein and mRNA levels of Wnt5a, CaN, NFAT2, MyoD, Myf5, Pax7, and Myogenin. The expression levels of MyoD, Myf5, Myogenin, MHC, Desmin, and NFAT2 were detected using immunofluorescence techniques. In addition, MyoD expression was observed using immunohistochemistry, and morphological changes in mouse muscle tissue were observed using HE staining. RESULTS During myoblast differentiation and muscle regeneration, Wnt5a expression was upregulated (P < 0.001) and the Wnt5a signalling pathway was activated. Wnt5a overexpression promoted the expression of MyoD, Myf5, Myogenin, MHC, and Desmin (P < 0.05), and conversely, knockdown of Wnt5a inhibited their expression (P < 0.001). The Wnt5a pathway mediated the opening of Ca2+ channels, regulated the expression levels of CaN, NFAT2, MyoD, Myf5, Myogenin, MHC, and Desmin (P < 0.01) and promoted the differentiation of C2C12 myoblasts and the repair and regeneration of injured skeletal muscle. The expression of Wnt5a, CaN, NFAT2, MyoD, Myogenin, Myf5, and MHC in C2C12 myoblast was significantly increased after curcumin intervention (P < 0.05); however, their expression decreased significantly after knocking down Wnt5a on the basis of curcumin intervention (P < 0.05). Similarly, in Wnt5a knockout mice, the promotion of muscle regeneration by curcumin was significantly attenuated. CONCLUSIONS Curcumin can activate the Wnt5a signalling pathway and mediate the opening of Ca2+ channels to accelerate the myogenic differentiation of C2C12 cells and the repair and regeneration of injured skeletal muscle.
Collapse
Affiliation(s)
- Mao‐yuan Wang
- Department of Rehabilitation MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouChina
- Ganzhou Key Laboratory of Rehabilitation MedicineGanzhouChina
| | - Jia‐ming Yang
- Department of Rehabilitation MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouChina
| | - Yi Wu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of EducationGannan Medical UniversityGanzhouChina
| | - Hai Li
- Department of Rehabilitation MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouChina
| | - Yan‐biao Zhong
- Department of Rehabilitation MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouChina
- Ganzhou Intelligent Rehabilitation Technology Innovation CenterGanzhouChina
| | - Yun Luo
- Department of Rehabilitation MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouChina
| | - Rui‐lian Xie
- Department of OncologyFirst Affiliated Hospital of Gannan Medical UniversityGanzhouChina
| |
Collapse
|
3
|
Guo L, Huang W, Wen Q, Zhang S, Bordbar F, Xiao Z, Nie Q. The first embryonic landscape of G-quadruplexes related to myogenesis. BMC Biol 2024; 22:194. [PMID: 39256800 PMCID: PMC11389323 DOI: 10.1186/s12915-024-01993-z] [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: 03/06/2024] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND DNA G-quadruplexes (G4s) represent a distinctive class of non-canonical DNA secondary structures. Despite their recognition as potential therapeutic targets in some cancers, the developmental role of G4 structures remains enigmatic. Mammalian embryonic myogenesis studies are hindered by limitations, prompting the use of chicken embryo-derived myoblasts as a model to explore G4 dynamics. This study aims to reveal the embryonic G4s landscape and elucidate the underlying mechanisms for candidate G4s that influence embryonic myogenesis. RESULTS This investigation unveils a significant reduction in G4s abundance during myogenesis. G4s stabilizer pyridostatin impedes embryonic myogenesis, emphasizing the regulatory role of G4s in this process. G4 Cut&Tag sequencing and RNA-seq analyses identify potential G4s and DEGs influencing embryonic myogenesis. Integration of G4 and DEG candidates identifies 32 G4s located in promoter regions capable of modulating gene transcription. WGBS elucidates DNA methylation dynamics during embryonic myogenesis. Coordinating transcriptome data with DNA G4s and DNA methylation profiles constructs a G4-DMR-DEG network, revealing nine interaction pairs. Notably, the NFATC2 promoter region sequence is confirmed to form a G4 structure, reducing promoter mCpG content and upregulating NFATC2 transcriptional activity. CONCLUSIONS This comprehensive study unravels the first embryonic genomic G4s landscape, highlighting the regulatory role of NFATC2 G4 in orchestrating transcriptional activity through promoter DNA methylation during myogenesis.
Collapse
Affiliation(s)
- Lijin Guo
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding & Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, 510642, China
- Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan, 512005, China
| | - Weiling Huang
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding & Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, 510642, China
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
| | - Qi Wen
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding & Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, 510642, China
| | - Siyu Zhang
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding & Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, 510642, China
| | - Farhad Bordbar
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding & Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, 510642, China
| | - Zhengzhong Xiao
- Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan, 512005, China
| | - Qinghua Nie
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding & Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
4
|
Hao X, Fu Y, Li S, Nie J, Zhang B, Zhang H. Porcine transient receptor potential channel 1 (TRPC1) regulates muscle growth via the Wnt/β-catenin and Wnt/Ca 2+ pathways. Int J Biol Macromol 2024; 265:130855. [PMID: 38490377 DOI: 10.1016/j.ijbiomac.2024.130855] [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: 06/04/2023] [Revised: 03/09/2024] [Accepted: 03/11/2024] [Indexed: 03/17/2024]
Abstract
Transient receptor potential canonical (TRPC) channels allow the intracellular entry of Ca2+ and play important roles in several physio-pathological processes. In this study, we constructed transgenic mice expressing porcine TRPC1 (Tg-pTRPC1) to verify the effects of TRPC1 on skeletal muscle growth and elucidate the underlying mechanism. Porcine TRPC1 increased the muscle mass, fiber cross-sectional area, and exercise endurance of mice and accelerated muscle repair and regeneration. TRPC1 overexpression enhanced β-catenin expression and promoted myogenesis, which was partly reversed by inhibitors of β-catenin. TRPC1 facilitated the accumulation of intracellular Ca2+ and nuclear translocation of the NFATC2/NFATC2IP complex involved in the Wnt/Ca2+ pathway, promoting muscle growth. Paired related homeobox 1 (Prrx1) promoted the expression of TRPC1, NFATC2, and NFATC2IP that participate in the regulation of muscle growth. Taken together, our findings indicate that porcine TRPC1 promoted by Prrx1 could regulate muscle development through activating the canonical Wnt/β-catenin and non-canonical Wnt/Ca2+ pathways.
Collapse
Affiliation(s)
- Xin Hao
- State Key Laboratory of animal biotech breeding, Beijing Key Laboratory of animal genetic engineering, China Agricultural University, Beijing 100193, China
| | - Yu Fu
- State Key Laboratory of animal biotech breeding, Beijing Key Laboratory of animal genetic engineering, China Agricultural University, Beijing 100193, China
| | - Shixin Li
- State Key Laboratory of animal biotech breeding, Beijing Key Laboratory of animal genetic engineering, China Agricultural University, Beijing 100193, China
| | - Jingru Nie
- State Key Laboratory of animal biotech breeding, Beijing Key Laboratory of animal genetic engineering, China Agricultural University, Beijing 100193, China
| | - Bo Zhang
- State Key Laboratory of animal biotech breeding, Beijing Key Laboratory of animal genetic engineering, China Agricultural University, Beijing 100193, China
| | - Hao Zhang
- State Key Laboratory of animal biotech breeding, Beijing Key Laboratory of animal genetic engineering, China Agricultural University, Beijing 100193, China; Sanya Institute of China Agricultural University, Sanya, Hainan 572025, China.
| |
Collapse
|
5
|
Mao H, Wang M, Ke Z, Wang J, Raza SHA, Dong X, An J, Yin Z, Qi L. Association of variants and expression levels of MYOD1 gene with carcass and muscle characteristic traits in domestic pigeons. Anim Biotechnol 2023; 34:4927-4937. [PMID: 37199180 DOI: 10.1080/10495398.2023.2213263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
This study was to investigate the correlations of myogenic differentiation 1 (MYOD1) gene polymorphisms with carcass traits and its expression with breast muscle development in pigeons. Four SNPs were found in the pigeon MYOD1 gene. Correlation analysis showed that individuals with AA genotype at both SNPs g.2967A > G (p < .01) and g.3044G > A (p < .05) have significantly higher live weight (LW), carcass weight (CW), semi-eviscerated weight (SEW), eviscerated weight (EW) and breast muscle weight (BMW). Moreover, the two SNPs also had the same significant effects on MYOD1 mRNA expression levels in breast muscle of pigeons, ie, the AA genotype showed higher MYOD1 mRNA expression levels. The diameter and cross-section area of muscle fibers continuously increased from 0w to 4w (p < .05), accompanied with the increasing expression of MYOD1 gene, while the density decreased (p < .05) dramatically from 0w to 1w and continuously fell over in the next few weeks (p > .05). What's more, the expression level of MYOD1 gene was positively correlated with a diameter (r = 0.937, p < .05) and cross-sectional area (r = 0.956, p < .01) of myofiber, and negatively correlated with density (r = -0.769, p < .01). The results showed that individuals with AA genotype at both SNPs g.2967A > G and g.3044G > A have showed higher carcass traits (LW, CW, SEW, EW, and BMW) and higher MYOD1 mRNA expression level in breast muscle than AB and BB genotypes. Moreover, the expression level of MYOD1 gene was closely correlated with muscle characteristic traits, indicating variants of MYOD1 gene was closely related to muscle development and could be a potential candidate gene in marker-assisted selection of pigeons.
Collapse
Affiliation(s)
- Haiguang Mao
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, Zhejiang, China
- College of Animal Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Mengting Wang
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, Zhejiang, China
| | - Zhijian Ke
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, Zhejiang, China
| | - Jinbo Wang
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, Zhejiang, China
| | - Sayed Haidar Abbas Raza
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xianggui Dong
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Jing An
- Institute of Plant Sciences Paris-Saclay (IPS2), Université Paris-Saclay, CNRS, INRAE, Université Evry, Orsay, Paris, France
| | - Zhaozheng Yin
- College of Animal Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lili Qi
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, Zhejiang, China
| |
Collapse
|
6
|
Kumar P, Zehra A, Saini M, Mathew SJ. Zeb1 and Tle3 are trans-factors that differentially regulate the expression of myosin heavy chain-embryonic and skeletal muscle differentiation. FASEB J 2023; 37:e23074. [PMID: 37392376 PMCID: PMC7615532 DOI: 10.1096/fj.202201698rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/03/2023]
Abstract
Myosin heavy chain-embryonic encoded by the Myh3 gene is a skeletal muscle-specific contractile protein expressed during mammalian development and regeneration, essential for proper myogenic differentiation and function. It is likely that multiple trans-factors are involved in this precise temporal regulation of Myh3 expression. We identify a 4230 bp promoter-enhancer region that drives Myh3 transcription in vitro during C2C12 myogenic differentiation and in vivo during muscle regeneration, including sequences both upstream and downstream of the Myh3 TATA-box that are necessary for complete Myh3 promoter activity. Using C2C12 mouse myogenic cells, we find that Zinc-finger E-box binding homeobox 1 (Zeb1) and Transducin-like Enhancer of Split 3 (Tle3) proteins are crucial trans-factors that interact and differentially regulate Myh3 expression. Loss of Zeb1 function results in earlier expression of myogenic differentiation genes and accelerated differentiation, whereas Tle3 depletion leads to reduced expression of myogenic differentiation genes and impaired differentiation. Tle3 knockdown resulted in downregulation of Zeb1, which could be mediated by increased expression of miR-200c, a microRNA that binds to Zeb1 transcript and degrades it. Tle3 functions upstream of Zeb1 in regulating myogenic differentiation since double knockdown of Zeb1 and Tle3 resulted in effects seen upon Tle3 depletion. We identify a novel E-box in the Myh3 distal promoter-enhancer region, where Zeb1 binds to repress Myh3 expression. In addition to regulation of myogenic differentiation at the transcriptional level, we uncover post-transcriptional regulation by Tle3 to regulate MyoG expression, mediated by the mRNA stabilizing Human antigen R (HuR) protein. Thus, Tle3 and Zeb1 are essential trans-factors that differentially regulate Myh3 expression and C2C12 cell myogenic differentiation in vitro.
Collapse
Affiliation(s)
- Pankaj Kumar
- Developmental Genetics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad, India
- Affiliated to Manipal Academy of Higher Education (MAHE), Manipal University, Manipal, India
- Developmental Genetics—III, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Aatifa Zehra
- Developmental Genetics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad, India
| | - Masum Saini
- Developmental Genetics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad, India
| | - Sam J. Mathew
- Developmental Genetics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad, India
- Affiliated to Manipal Academy of Higher Education (MAHE), Manipal University, Manipal, India
| |
Collapse
|
7
|
Hoh JFY. Developmental, physiologic and phylogenetic perspectives on the expression and regulation of myosin heavy chains in mammalian skeletal muscles. J Comp Physiol B 2023:10.1007/s00360-023-01499-0. [PMID: 37277594 DOI: 10.1007/s00360-023-01499-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 06/07/2023]
Abstract
The kinetics of myosin controls the speed and power of muscle contraction. Mammalian skeletal muscles express twelve kinetically different myosin heavy chain (MyHC) genes which provides a wide range of muscle speeds to meet different functional demands. Myogenic progenitors from diverse craniofacial and somitic mesoderm specify muscle allotypes with different repertoires for MyHC expression. This review provides a brief synopsis on the historical and current views on how cell lineage, neural impulse patterns, and thyroid hormone influence MyHC gene expression in muscles of the limb allotype during development and in adult life and the molecular mechanisms thereof. During somitic myogenesis, embryonic and foetal myoblast lineages form slow and fast primary and secondary myotube ontotypes which respond differently to postnatal neural and thyroidal influences to generate fully differentiated fibre phenotypes. Fibres of a given phenotype may arise from myotubes of different ontotypes which retain their capacity to respond differently to neural and thyroidal influences during postnatal life. This gives muscles physiological plasticity to adapt to fluctuations in thyroid hormone levels and patterns of use. The kinetics of MyHC isoforms vary inversely with animal body mass. Fast 2b fibres are specifically absent in muscles involved in elastic energy saving in hopping marsupials and generally absent in large eutherian mammals. Changes in MyHC expression are viewed in the context of the physiology of the whole animal. The roles of myoblast lineage and thyroid hormone in regulating MyHC gene expression are phylogenetically the most ancient while that of neural impulse patterns the most recent.
Collapse
Affiliation(s)
- Joseph Foon Yoong Hoh
- Discipline of Physiology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia.
- , PO Box 152, Killara, NSW, 2071, Australia.
| |
Collapse
|
8
|
Zhang RN, Bao X, Liu Y, Wang Y, Li XY, Tan G, Mbadhi MN, Xu W, Yang Q, Yao LY, Chen L, Zhao XY, Hu CQ, Zhang JX, Zheng HT, Wu Y, Li S, Chen SJ, Chen SY, Lv J, Shi LL, Tang JM. The spatiotemporal matching pattern of Ezrin/Periaxin involved in myoblast differentiation and fusion and Charcot-Marie-Tooth disease-associated muscle atrophy. J Transl Med 2023; 21:173. [PMID: 36870952 PMCID: PMC9985213 DOI: 10.1186/s12967-023-04016-7] [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: 10/07/2022] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND Clinically, Charcot-Marie-Tooth disease (CMT)-associated muscle atrophy still lacks effective treatment. Deletion and mutation of L-periaxin can be involved in CMT type 4F (CMT4F) by destroying the myelin sheath form, which may be related to the inhibitory role of Ezrin in the self-association of L-periaxin. However, it is still unknown whether L-periaxin and Ezrin are independently or interactively involved in the process of muscle atrophy by affecting the function of muscle satellite cells. METHOD A gastrocnemius muscle atrophy model was prepared to mimic CMT4F and its associated muscle atrophy by mechanical clamping of the peroneal nerve. Differentiating C2C12 myoblast cells were treated with adenovirus-mediated overexpression or knockdown of Ezrin. Then, overexpression of L-periaxin and NFATc1/c2 or knockdown of L-periaxin and NFATc3/c4 mediated by adenovirus vectors were used to confirm their role in Ezrin-mediated myoblast differentiation, myotube formation and gastrocnemius muscle repair in a peroneal nerve injury model. RNA-seq, real-time PCR, immunofluorescence staining and Western blot were used in the above observation. RESULTS For the first time, instantaneous L-periaxin expression was highest on the 6th day, while Ezrin expression peaked on the 4th day during myoblast differentiation/fusion in vitro. In vivo transduction of adenovirus vectors carrying Ezrin, but not Periaxin, into the gastrocnemius muscle in a peroneal nerve injury model increased the numbers of muscle myosin heavy chain (MyHC) I and II type myofibers, reducing muscle atrophy and fibrosis. Local muscle injection of overexpressed Ezrin combined with incubation of knockdown L-periaxin within the injured peroneal nerve or injection of knockdown L-periaxin into peroneal nerve-injured gastrocnemius muscle not only increased the number of muscle fibers but also recovered their size to a relatively normal level in vivo. Overexpression of Ezrin promoted myoblast differentiation/fusion, inducing increased MyHC-I+ and MyHC-II + muscle fiber specialization, and the specific effects could be enhanced by the addition of adenovirus vectors for knockdown of L-periaxin by shRNA. Overexpression of L-periaxin did not alter the inhibitory effects on myoblast differentiation and fusion mediated by knockdown of Ezrin by shRNA in vitro but decreased myotube length and size. Mechanistically, overexpressing Ezrin did not alter protein kinase A gamma catalytic subunit (PKA-γ cat), protein kinase A I alpha regulatory subunit (PKA reg Iα) or PKA reg Iβ levels but increased PKA-α cat and PKA reg II α levels, leading to a decreased ratio of PKA reg I/II. The PKA inhibitor H-89 remarkably abolished the effects of overexpressing-Ezrin on increased myoblast differentiation/fusion. In contrast, knockdown of Ezrin by shRNA significantly delayed myoblast differentiation/fusion accompanied by an increased PKA reg I/II ratio, and the inhibitory effects could be eliminated by the PKA reg activator N6-Bz-cAMP. Meanwhile, overexpressing Ezrin enhanced type I muscle fiber specialization, accompanied by an increase in NFATc2/c3 levels and a decrease in NFATc1 levels. Furthermore, overexpressing NFATc2 or knocking down NFATc3 reversed the inhibitory effects of Ezrin knockdown on myoblast differentiation/fusion. CONCLUSIONS The spatiotemporal pattern of Ezrin/Periaxin expression was involved in the control of myoblast differentiation/fusion, myotube length and size, and myofiber specialization, which was related to the activated PKA-NFAT-MEF2C signaling pathway, providing a novel L-Periaxin/Ezrin joint strategy for the treatment of muscle atrophy induced by nerve injury, especially in CMT4F.
Collapse
Affiliation(s)
- Ruo-Nan Zhang
- Faculty of Basic Medical Sciences, Postgraduate Union Training Basement of Jin Zhou Medical University, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.,Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research,Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.,Emergency Comprehensive Department, Shiyan Maternal and Child Health Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Xin Bao
- Faculty of Basic Medical Sciences, Postgraduate Union Training Basement of Jin Zhou Medical University, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.,Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research,Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.,Experimental Medical Center, Dongfeng Hospital, Hubei University of Medicine, Shiyan, China
| | - Yun Liu
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research,Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Yan Wang
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research,Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Xing-Yuan Li
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research,Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.,Department of Physiology, Faculty of Basic Medical Sciences, Zunyi Medical University, Zunyi, 563006, Guizhou, People's Republic of China
| | - Ge Tan
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research,Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Magdaleena Naemi Mbadhi
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research,Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Wei Xu
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research,Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Qian Yang
- Institute of Anesthesiology, Department of Anesthesiology, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.,Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research,Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Lu-Yuan Yao
- Institute of Anesthesiology, Department of Anesthesiology, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.,Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research,Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Long Chen
- Experimental Medical Center, Dongfeng Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiao-Ying Zhao
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research,Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Chang-Qing Hu
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research,Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Jing-Xuan Zhang
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research,Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Hong-Tao Zheng
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research,Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Yan Wu
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research,Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Shan Li
- Department of Biochemistry, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Shao-Juan Chen
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Shi-You Chen
- Department of Surgery, University of Missouri, Columbia, USA
| | - Jing Lv
- Institute of Anesthesiology, Department of Anesthesiology, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.
| | - Liu-Liu Shi
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research,Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.
| | - Jun-Ming Tang
- Faculty of Basic Medical Sciences, Postgraduate Union Training Basement of Jin Zhou Medical University, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China. .,Institute of Anesthesiology, Department of Anesthesiology, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China. .,Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research,Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.
| |
Collapse
|
9
|
Notch1 Is Involved in Physiologic Cardiac Hypertrophy of Mice via the p38 Signaling Pathway after Voluntary Running. Int J Mol Sci 2023; 24:ijms24043212. [PMID: 36834623 PMCID: PMC9966550 DOI: 10.3390/ijms24043212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/26/2023] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
Appropriate exercise such as voluntary wheel-running can induce physiological cardiac hypertrophy. Notch1 plays an important role in cardiac hypertrophy; however, the experimental results are inconsistent. In this experiment, we aimed to explore the role of Notch1 in physiological cardiac hypertrophy. Twenty-nine adult male mice were randomly divided into a Notch1 heterozygous deficient control (Notch1+/- CON) group, a Notch1 heterozygous deficient running (Notch1+/- RUN) group, a wild type control (WT CON) group, and a wild type running (WT RUN) group. Mice in the Notch1+/- RUN and WT RUN groups had access to voluntary wheel-running for two weeks. Next, the cardiac function of all of the mice was examined by echocardiography. The H&E staining, Masson trichrome staining, and a Western blot assay were carried out to analyze cardiac hypertrophy, cardiac fibrosis, and the expression of proteins relating to cardiac hypertrophy. After two-weeks of running, the Notch1 receptor expression was decreased in the hearts of the WT RUN group. The degree of cardiac hypertrophy in the Notch1+/- RUN mice was lower than that of their littermate control. Compared to the Notch1+/- CON group, Notch1 heterozygous deficiency could lead to a decrease in Beclin-1 expression and the ratio of LC3II/LC3I in the Notch1+/- RUN group. The results suggest that Notch1 heterozygous deficiency could partly dampen the induction of autophagy. Moreover, Notch1 deficiency may lead to the inactivation of p38 and the reduction of β-catenin expression in the Notch1+/- RUN group. In conclusion, Notch1 plays a critical role in physiologic cardiac hypertrophy through the p38 signaling pathway. Our results will help to understand the underlying mechanism of Notch1 on physiological cardiac hypertrophy.
Collapse
|
10
|
Continuous exposure to isoprenaline reduced myotube size by delaying myoblast differentiation and fusion through the NFAT-MEF2C signaling pathway. Sci Rep 2023; 13:436. [PMID: 36624121 PMCID: PMC9829891 DOI: 10.1038/s41598-022-22330-w] [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: 01/23/2022] [Accepted: 10/13/2022] [Indexed: 01/11/2023] Open
Abstract
We aimed to explore whether superfluous sympathetic activity affects myoblast differentiation, fusion, and myofiber types using a continuous single-dose isoprenaline exposure model in vitro and to further confirm the role of distinct NFATs in ISO-mediated effects. Compared with delivery of single and interval single, continuous single-dose ISO most obviously diminished myotube size while postponing myoblast differentiation/fusion in a time- and dose-dependent pattern, accompanied by an apparent decrease in nuclear NFATc1/c2 levels and a slight increase in nuclear NFATc3/c4 levels. Overexpression of NFATc1 or NFATc2, particularly NFATc1, markedly abolished the inhibitory effects of ISO on myoblast differentiation/fusion, myotube size and Myh7 expression, which was attributed to a remarkable increase in the nuclear NFATc1/c2 levels and a reduction in the nuclear NFATc4 levels and the associated increase in the numbers of MyoG and MEF2C positive nuclei within more than 3 nuclei myotubes, especially in MEF2C. Moreover, knockdown of NFATc3 by shRNA did not alter the inhibitory effect of ISO on myoblast differentiation/fusion or myotube size but partially recovered the expression of Myh7, which was related to the slightly increased nuclear levels of NFATc1/c2, MyoG and MEF2C. Knockdown of NFATc4 by shRNA prominently increased the number of MyHC +, MyoG or MEF2C + myoblast cells with 1 ~ 2 nuclei, causing fewer numbers and smaller myotube sizes. However, NFATc4 knockdown further deteriorated the effects of ISO on myoblast fusion and myotube size, with more than 5 nuclei and Myh1/2/4 expression, which was associated with a decrease in nuclear NFATc2/c3 levels. Therefore, ISO inhibited myoblast differentiation/fusion and myotube size through the NFAT-MyoG-MEF2C signaling pathway.
Collapse
|
11
|
Lim JY, Kim E, Douglas CM, Wirianto M, Han C, Ono K, Kim SY, Ji JH, Tran CK, Chen Z, Esser KA, Yoo SH. The circadian E3 ligase FBXL21 regulates myoblast differentiation and sarcomere architecture via MYOZ1 ubiquitination and NFAT signaling. PLoS Genet 2022; 18:e1010574. [PMID: 36574402 PMCID: PMC9829178 DOI: 10.1371/journal.pgen.1010574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 01/09/2023] [Accepted: 12/14/2022] [Indexed: 12/28/2022] Open
Abstract
Numerous molecular and physiological processes in the skeletal muscle undergo circadian time-dependent oscillations in accordance with daily activity/rest cycles. The circadian regulatory mechanisms underlying these cyclic processes, especially at the post-transcriptional level, are not well defined. Previously, we reported that the circadian E3 ligase FBXL21 mediates rhythmic degradation of the sarcomere protein TCAP in conjunction with GSK-3β, and Psttm mice harboring an Fbxl21 hypomorph allele show reduced muscle fiber diameter and impaired muscle function. To further elucidate the regulatory function of FBXL21 in skeletal muscle, we investigated another sarcomere protein, Myozenin1 (MYOZ1), that we identified as an FBXL21-binding protein from yeast 2-hybrid screening. We show that FBXL21 binding to MYOZ1 led to ubiquitination-mediated proteasomal degradation. GSK-3β co-expression and inhibition were found to accelerate and decelerate FBXL21-mediated MYOZ1 degradation, respectively. Previously, MYOZ1 has been shown to inhibit calcineurin/NFAT signaling important for muscle differentiation. In accordance, Fbxl21 KO and MyoZ1 KO in C2C12 cells impaired and enhanced myogenic differentiation respectively compared with control C2C12 cells, concomitant with distinct effects on NFAT nuclear localization and NFAT target gene expression. Importantly, in Psttm mice, both the levels and diurnal rhythm of NFAT2 nuclear localization were significantly diminished relative to wild-type mice, and circadian expression of NFAT target genes associated with muscle differentiation was also markedly dampened. Furthermore, Psttm mice exhibited significant disruption of sarcomere structure with a considerable excess of MYOZ1 accumulation in the Z-line. Taken together, our study illustrates a pivotal role of FBXL21 in sarcomere structure and muscle differentiation by regulating MYOZ1 degradation and NFAT2 signaling.
Collapse
Affiliation(s)
- Ji Ye Lim
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Eunju Kim
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Collin M. Douglas
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Marvin Wirianto
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Chorong Han
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Kaori Ono
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Sun Young Kim
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Justin H. Ji
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Celia K. Tran
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Zheng Chen
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Karyn A. Esser
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| |
Collapse
|
12
|
Mao H, Yin Z, Wang M, Zhang W, Raza SHA, Althobaiti F, Qi L, Wang J. Expression of DGAT2 Gene and Its Associations With Intramuscular Fat Content and Breast Muscle Fiber Characteristics in Domestic Pigeons (Columba livia). Front Vet Sci 2022; 9:847363. [PMID: 35754541 PMCID: PMC9227834 DOI: 10.3389/fvets.2022.847363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 04/26/2022] [Indexed: 12/13/2022] Open
Abstract
Diacylglycerol acyltransferase 2 (DGAT2) catalyzes the final step in triglyceride synthesis and plays an important role in the synthesis of fat, but the effects of its expression on intramuscular fat (IMF) content and muscle development are still unknown. In this study, we investigated the expression of the DGAT2 gene and its associations with IMF content and breast muscle fiber characteristics in pigeons. The spatiotemporal expression profile of the pigeon DGAT2 gene in breast muscle showed that the mRNA expression level of DGAT2 gene in subcutaneous fat was the highest (p < 0.01) among eight tissues from 0 to 4 weeks of age, and showed an upward trend week by week, followed by liver (p < 0.05). Moreover, both mRNA and protein levels of the DGAT2 gene in breast muscle showed an upward trend from 0 to 4 weeks (p < 0.05), accompanied by the upregulation of MYOD1 and MSTN. In addition, the paraffin section analysis results revealed that the diameter and cross-sectional area of pectoralis muscle fiber significantly increased with age (p < 0.05), and a significant positive correlation was shown between the DGAT2 gene expression level and muscle fiber diameter (p < 0.05). Furthermore, correlation analysis suggested that the mRNA expression level of the pigeon DGAT2 gene was significantly (p < 0.01) correlated with IMF content in breast muscle. These results imply that the DGAT2 gene has a close relationship with IMF content and breast muscle fiber characteristics in pigeons, indicating that the DGAT2 gene might be used as a candidate gene marker-assisted breeding in pigeons.
Collapse
Affiliation(s)
- Haiguang Mao
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, China
| | - Zhaozheng Yin
- College of Animal Science, Zhejiang University, Hangzhou, China
| | - Mengting Wang
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, China
| | - Wenwen Zhang
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, China
| | | | - Fayez Althobaiti
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
| | - Lili Qi
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, China
- *Correspondence: Lili Qi
| | - Jinbo Wang
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, China
- Jinbo Wang
| |
Collapse
|
13
|
A fast Myosin super enhancer dictates muscle fiber phenotype through competitive interactions with Myosin genes. Nat Commun 2022; 13:1039. [PMID: 35210422 PMCID: PMC8873246 DOI: 10.1038/s41467-022-28666-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 02/04/2022] [Indexed: 12/15/2022] Open
Abstract
The contractile properties of adult myofibers are shaped by their Myosin heavy chain isoform content. Here, we identify by snATAC-seq a 42 kb super-enhancer at the locus regrouping the fast Myosin genes. By 4C-seq we show that active fast Myosin promoters interact with this super-enhancer by DNA looping, leading to the activation of a single promoter per nucleus. A rainbow mouse transgenic model of the locus including the super-enhancer recapitulates the endogenous spatio-temporal expression of adult fast Myosin genes. In situ deletion of the super-enhancer by CRISPR/Cas9 editing demonstrates its major role in the control of associated fast Myosin genes, and deletion of two fast Myosin genes at the locus reveals an active competition of the promoters for the shared super-enhancer. Last, by disrupting the organization of fast Myosin, we uncover positional heterogeneity within limb skeletal muscles that may underlie selective muscle susceptibility to damage in certain myopathies. The contractile properties of adult myofibers are shaped by their Myosin heavy chain isoform content. Here the authors show that a super enhancer controls the spatiotemporal expression of the genes at the fast myosin heavy chain locus by DNA looping and that this expression profile is recapitulated in a rainbow transgenic mouse model of the locus.
Collapse
|
14
|
Chapotte-Baldacci CA, Cognard C, Bois P, Chatelier A, Sebille S. Handling a mature calcium signature through optogenetics improves the differentiation of primary murine myotubes. Cell Calcium 2022; 103:102546. [DOI: 10.1016/j.ceca.2022.102546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 12/15/2022]
|
15
|
Jan MI, Ali T, Ishtiaq A, Mushtaq I, Murtaza I. Prospective Advances in Non-coding RNAs Investigation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:385-426. [PMID: 32285426 DOI: 10.1007/978-981-15-1671-9_24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Non-coding RNAs (ncRNAs) play significant roles in numerous physiological cellular processes and molecular alterations during pathological conditions including heart diseases, cancer, immunological disorders and neurological diseases. This chapter is focusing on the basis of ncRNA relation with their functions and prospective advances in non-coding RNAs particularly miRNAs investigation in the cardiovascular disease management.The field of ncRNAs therapeutics is a very fascinating and challenging too. Scientists have opportunity to develop more advanced therapeutics as well as diagnostic approaches for cardiovascular conditions. Advanced studies are critically needed to deepen the understanding of the molecular biology, mechanism and modulation of ncRNAs and chemical formulations for managing CVDs.
Collapse
Affiliation(s)
- Muhammad Ishtiaq Jan
- Department of Biochemistry, Signal Transduction Laboratory, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Tahir Ali
- Department of Biochemistry, Signal Transduction Laboratory, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Ayesha Ishtiaq
- Department of Biochemistry, Signal Transduction Laboratory, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Iram Mushtaq
- Department of Biochemistry, Signal Transduction Laboratory, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Iram Murtaza
- Department of Biochemistry, Signal Transduction Laboratory, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|
16
|
Cho IC, Park HB, Ahn JS, Han SH, Lee JB, Lim HT, Yoo CK, Jung EJ, Kim DH, Sun WS, Ramayo-Caldas Y, Kim SG, Kang YJ, Kim YK, Shin HS, Seong PN, Hwang IS, Park BY, Hwang S, Lee SS, Ryu YC, Lee JH, Ko MS, Lee K, Andersson G, Pérez-Enciso M, Lee JW. A functional regulatory variant of MYH3 influences muscle fiber-type composition and intramuscular fat content in pigs. PLoS Genet 2019; 15:e1008279. [PMID: 31603892 PMCID: PMC6788688 DOI: 10.1371/journal.pgen.1008279] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 07/01/2019] [Indexed: 11/18/2022] Open
Abstract
Muscle development and lipid accumulation in muscle critically affect meat quality of livestock. However, the genetic factors underlying myofiber-type specification and intramuscular fat (IMF) accumulation remain to be elucidated. Using two independent intercrosses between Western commercial breeds and Korean native pigs (KNPs) and a joint linkage-linkage disequilibrium analysis, we identified a 488.1-kb region on porcine chromosome 12 that affects both reddish meat color (a*) and IMF. In this critical region, only the MYH3 gene, encoding myosin heavy chain 3, was found to be preferentially overexpressed in the skeletal muscle of KNPs. Subsequently, MYH3-transgenic mice demonstrated that this gene controls both myofiber-type specification and adipogenesis in skeletal muscle. We discovered a structural variant in the promotor/regulatory region of MYH3 for which Q allele carriers exhibited significantly higher values of a* and IMF than q allele carriers. Furthermore, chromatin immunoprecipitation and cotransfection assays showed that the structural variant in the 5'-flanking region of MYH3 abrogated the binding of the myogenic regulatory factors (MYF5, MYOD, MYOG, and MRF4). The allele distribution of MYH3 among pig populations worldwide indicated that the MYH3 Q allele is of Asian origin and likely predates domestication. In conclusion, we identified a functional regulatory sequence variant in porcine MYH3 that provides novel insights into the genetic basis of the regulation of myofiber type ratios and associated changes in IMF in pigs. The MYH3 variant can play an important role in improving pork quality in current breeding programs.
Collapse
Affiliation(s)
- In-Cheol Cho
- National Institute of Animal Science, Rural Development Administration, Jeju, Republic of Korea
| | - Hee-Bok Park
- Department of Animal Resources Science, College of Industrial Sciences, Kongju National University, Yesan, Republic of Korea
| | - Jin Seop Ahn
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Sang-Hyun Han
- Educational Science Research Institute, Jeju National University, Jeju, Republic of Korea
| | - Jae-Bong Lee
- Korea Zoonosis Research Institute, Chonbuk National University, Iksan, Republic of Korea
| | - Hyun-Tae Lim
- Department of Animal Science, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea
- Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Chae-Kyoung Yoo
- Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Eun-Ji Jung
- Bio-Medical Science Co., Ltd., Gimpo, Republic of Korea
| | - Dong-Hwan Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
| | - Wu-Sheng Sun
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Yuliaxis Ramayo-Caldas
- Génétique Animale et Biologie Intégrative (GABI), INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
- Animal Breeding and Genetics Program, Institute for Research and Technology in Food and Agriculture (IRTA), Torre Marimon, Caldes de Montbui, Spain
| | - Sang-Geum Kim
- National Institute of Animal Science, Rural Development Administration, Jeju, Republic of Korea
| | - Yong-Jun Kang
- National Institute of Animal Science, Rural Development Administration, Jeju, Republic of Korea
| | - Yoo-Kyung Kim
- Educational Science Research Institute, Jeju National University, Jeju, Republic of Korea
| | - Hyun-Sook Shin
- National Institute of Animal Science, Rural Development Administration, Jeju, Republic of Korea
| | - Pil-Nam Seong
- National Institute of Animal Science, Rural Development Administration, Jeju, Republic of Korea
| | - In-Sul Hwang
- National Institute of Animal Science, Rural Development Administration, Wanju, Republic of Korea
| | - Beom-Young Park
- National Institute of Animal Science, Rural Development Administration, Wanju, Republic of Korea
| | - Seongsoo Hwang
- National Institute of Animal Science, Rural Development Administration, Wanju, Republic of Korea
| | - Sung-Soo Lee
- National Institute of Animal Science, Rural Development Administration, Namwon, Republic of Korea
| | - Youn-Chul Ryu
- Division of Biotechnology, SARI, Jeju National University, Jeju, Republic of Korea
| | - Jun-Heon Lee
- Division of Animal and Dairy Science, Chungnam National University, Deajeon, Republic of Korea
| | - Moon-Suck Ko
- National Institute of Animal Science, Rural Development Administration, Jeju, Republic of Korea
| | - Kichoon Lee
- Department of Animal Sciences, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Columbus, OH, United States of America
| | - Göran Andersson
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Miguel Pérez-Enciso
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB Consortium, Barcelona, Spain
- Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Barcelona, Spain
- ICREA, Carrer de Lluís Companys, Barcelona, Spain
| | - Jeong-Woong Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
17
|
Multiple transcription factors mediating the expressional regulation of myosin heavy chain gene involved in the indeterminate muscle growth of fish. Gene 2019; 687:308-318. [PMID: 30453072 DOI: 10.1016/j.gene.2018.11.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/05/2018] [Accepted: 11/15/2018] [Indexed: 11/22/2022]
Abstract
Torafugu myosin heavy chain gene, MYHM2528-1, is specifically expressed in neonatal slow and fast muscle fibers, suggesting its functional role in indeterminate muscle growth in fish. However, the transcriptional regulatory mechanisms of MYHM2528-1 involved in indeterminate muscle growth in fish remained unknown. We previously isolated a 2100 bp 5'- flanking sequence of torafugu MYHM2528-1 that showed sufficient promoter activity to allow specific gene expression in neonatal muscle fibers of zebrafish. Here, we examined the cis-regulatory mechanism of 2100 bp 5'-flanking region of torafugu MYHM2528-1 using deletion-mutation analysis in zebrafish embryo. We discovered that myoblast determining factor (MyoD) binding elements play a key role and participate in the transcriptional regulation of MYHM2528-1 expression in zebrafish embryos. We further discovered that paired box protein (Pax3) are required for promoting MYHM2528-1 expression and myocyte enhancer factor-2 (MEF2) binding sites participate in the transcriptional regulation of MYHM2528-1 expression in slow/fast skeletal muscles. Our study also confirmed that the nuclear factor of activated T-cell (NFAT) binding sites take part in the transcriptional regulation of MYHM2528-1 expression in slow and fast muscles fiber in relation to indeterminate muscle growth. These results obviously confirmed that multiple cis-elements in the 5'-flanking region of MYHM2528-1 function in the transcriptional regulation of its expression.
Collapse
|
18
|
Dong X, Cao H, Mao H, Hong Q, Yin Z. Association of MyoD1 Gene Polymorphisms with Meat Quality Traits in Domestic Pigeons ( Columba livia). J Poult Sci 2019; 56:20-26. [PMID: 32055192 PMCID: PMC6993886 DOI: 10.2141/jpsa.0170182] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 04/27/2018] [Indexed: 01/03/2023] Open
Abstract
Myogenic differentiation 1 (MyoD1) belongs to the MyoD family and plays a key role in myogenesis and consequently, in determining muscle fiber characteristics. In this study, single nucleotide polymorphisms (SNPs) in the exons of MyoD1 were identified in 200 domestic pigeons (Columba livia) by direct DNA sequencing, and the association between MyoD1 polymorphisms and meat quality traits was analyzed. We found four novel variations (A2967G, G3044A, A3164C, and C3311G) in exon 3. The SNP A2967G is a synonymous mutation, while the other 3 SNPs are located in the 3' untranslated region. The analysis revealed 3 genotypes, in which allele A was the predominant allele in the SNP A2967G, while allele B was the predominant allele in the SNPs G3044A and A3164C. The mutations A2967G and G3044A were significantly associated with meat quality traits in pigeon. Pigeons with AA or AB genotypes had higher breast muscle concentrations of inosinic acid and intramuscular fat than those of BB genotype. Moreover, these 2 SNPs had significant effects on MyoD1 mRNA expression. The SNPs A2967G and G3044A were organized into 4 haplotypes, which formed 7 diplotypes. Association analysis showed that the diplotypes were not significantly associated with meat quality traits. Our results implied that associations do exist between MyoD1 gene polymorphisms and meat quality traits in domestic pigeons, and the AA and AB genotypes could be applied as genetic markers in marker-aid pigeon breeding.
Collapse
Affiliation(s)
- Xinyang Dong
- Animal Science College, Zhejiang University, Zijingang Campus, Hangzhou, 310058, China
| | - Haiyue Cao
- Animal Science College, Zhejiang University, Zijingang Campus, Hangzhou, 310058, China
| | - Haiguang Mao
- Animal Science College, Zhejiang University, Zijingang Campus, Hangzhou, 310058, China
| | - Qihua Hong
- Animal Science College, Zhejiang University, Zijingang Campus, Hangzhou, 310058, China
| | - Zhaozheng Yin
- Animal Science College, Zhejiang University, Zijingang Campus, Hangzhou, 310058, China
| |
Collapse
|
19
|
|
20
|
Comprehensive analysis of lncRNAs and mRNAs with associated co-expression and ceRNA networks in C2C12 myoblasts and myotubes. Gene 2018; 647:164-173. [PMID: 29331478 DOI: 10.1016/j.gene.2018.01.039] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/29/2017] [Accepted: 01/09/2018] [Indexed: 12/30/2022]
Abstract
Long non-coding RNAs (lncRNAs) are emerging as important regulators in the modulation of muscle development and muscle-related diseases. To explore potential regulators of muscle differentiation, we determined the expression profiles of lncRNAs and mRNAs in C2C12 mouse myoblast cell line using microarray analysis. Gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were performed to explore their function. We also constructed co-expression, cis/trans-regulation, and competing endogenous RNA (ceRNA) networks with bioinformatics methods. We found that 3067 lncRNAs and 3235 mRNAs were differentially regulated (fold change ≥2.0). Bioinformatics analysis indicated that the principal functions of the transcripts were related to muscle structure development and morphogenesis. Co-expression analysis showed 261 co-expression relationships between 233 lncRNAs and 10 mRNAs, and nine lncRNAs interacted with myog and MEF2C collectively. Cis/trans-regulation prediction revealed that lncRNA Myh6 could be a valuable gene via cis-regulation, and lncRNAs such as 2310043L19Ris, V00821, and AK139352 may participate in particular pathways regulated by transcription factors, including myog, myod1, and foxo1. The myog-specific ceRNA network covered 10 lncRNAs, 378 miRNAs, and 1960 edges. The upregulated lncRNAs Filip1, Myl1, and 2310043L19Rik may promote myog expression by acting as ceRNAs. Our results offer a new perspective on the modulation of lncRNAs in muscle differentiation.
Collapse
|
21
|
Wang JS, Infante CR, Park S, Menke DB. PITX1 promotes chondrogenesis and myogenesis in mouse hindlimbs through conserved regulatory targets. Dev Biol 2017; 434:186-195. [PMID: 29273440 DOI: 10.1016/j.ydbio.2017.12.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 12/05/2017] [Accepted: 12/18/2017] [Indexed: 10/18/2022]
Abstract
The PITX1 transcription factor is expressed during hindlimb development, where it plays a critical role in directing hindlimb growth and the specification of hindlimb morphology. While it is known that PITX1 regulates hindlimb formation, in part, through activation of the Tbx4 gene, other transcriptional targets remain to be elucidated. We have used a combination of ChIP-seq and RNA-seq to investigate enhancer regions and target genes that are directly regulated by PITX1 in embryonic mouse hindlimbs. In addition, we have analyzed PITX1 binding sites in hindlimbs of Anolis lizards to identify ancient PITX1 regulatory targets. We find that PITX1-bound regions in both mouse and Anolis hindlimbs are strongly associated with genes implicated in limb and skeletal system development. Gene expression analyses reveal a large number of misexpressed genes in the hindlimbs of Pitx1-/- mouse embryos. By intersecting misexpressed genes with genes that have neighboring mouse PITX1 binding sites, we identified 440 candidate targets of PITX1. Of these candidates, 68 exhibit ultra-conserved PITX1 binding events that are shared between mouse and Anolis hindlimbs. Among the ancient targets of PITX1 are important regulators of cartilage and skeletal muscle development, including Sox9 and Six1. Our data suggest that PITX1 promotes chondrogenesis and myogenesis in the hindlimb by direct regulation of several key members of the cartilage and muscle transcriptional networks.
Collapse
Affiliation(s)
- Jialiang S Wang
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Carlos R Infante
- Department of Genetics, University of Georgia, Athens, GA 30602, USA; Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
| | - Sungdae Park
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Douglas B Menke
- Department of Genetics, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
22
|
Miao C, Lv Y, Zhang W, Chai X, Feng L, Fang Y, Liu X, Zhang X. Pyrrolidine Dithiocarbamate (PDTC) Attenuates Cancer Cachexia by Affecting Muscle Atrophy and Fat Lipolysis. Front Pharmacol 2017; 8:915. [PMID: 29311924 PMCID: PMC5733020 DOI: 10.3389/fphar.2017.00915] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 11/30/2017] [Indexed: 12/18/2022] Open
Abstract
Cancer cachexia is a kind of whole body metabolic disorder syndrome accompanied with severe wasting of muscle and adipose tissue. NF-κB signaling plays an important role during skeletal muscle atrophy and fat lipolysis. As an inhibitor of NF-κB signaling, Pyrrolidine dithiocarbamate (PDTC) was reported to relieve cancer cachexia; however, its mechanism remains largely unknown. In our study, we showed that PDTC attenuated cancer cachexia symptom in C26 tumor bearing mice models in vivo without influencing tumor volume. What’s more, PDTC inhibited muscle atrophy and lipolysis in cells models in vitro induced by TNFα and C26 tumor medium. PDTC suppressed atrophy of myotubes differentiated from C2C12 by reducing MyoD and upregulating MuRF1, and preserving the expression of perilipin as well as blocking the activation of HSL in 3T3-L1 mature adipocytes. Meaningfully, we observed that PDTC also inhibited p38 MAPK signaling besides the NF-κB signaling in cancer cachexia in vitro models. In addition, PDTC also influenced the protein synthesis of skeletal muscle by activating AKT signaling and regulated fat energy metabolism by inhibiting AMPK signaling. Therefore, PDTC primarily influenced different pathways in different tissues. The study not only established a simple and reliable screening drugs model of cancer cachexia in vitro but also provided new theoretical basis for future treatment of cancer cachexia.
Collapse
Affiliation(s)
- Chunxiao Miao
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, College of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Yuanyuan Lv
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, College of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Wanli Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, College of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Xiaoping Chai
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, College of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Lixing Feng
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, College of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China.,Institute of Interdisciplinary Integrative Biomedical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanfen Fang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, College of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China.,Division of Anti-tumor Pharmacology, Shanghai Institute of Materia Medica (CAS), Chinese Academy of Sciences, Shanghai, China
| | - Xuan Liu
- Institute of Interdisciplinary Integrative Biomedical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiongwen Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, College of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| |
Collapse
|
23
|
Taglietti V, Maroli G, Cermenati S, Monteverde S, Ferrante A, Rossi G, Cossu G, Beltrame M, Messina G. Nfix Induces a Switch in Sox6 Transcriptional Activity to Regulate MyHC-I Expression in Fetal Muscle. Cell Rep 2017; 17:2354-2366. [PMID: 27880909 PMCID: PMC5149531 DOI: 10.1016/j.celrep.2016.10.082] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/08/2016] [Accepted: 10/24/2016] [Indexed: 02/01/2023] Open
Abstract
Sox6 belongs to the Sox gene family and plays a pivotal role in fiber type differentiation, suppressing transcription of slow-fiber-specific genes during fetal development. Here, we show that Sox6 plays opposite roles in MyHC-I regulation, acting as a positive and negative regulator of MyHC-I expression during embryonic and fetal myogenesis, respectively. During embryonic myogenesis, Sox6 positively regulates MyHC-I via transcriptional activation of Mef2C, whereas during fetal myogenesis, Sox6 requires and cooperates with the transcription factor Nfix in repressing MyHC-I expression. Mechanistically, Nfix is necessary for Sox6 binding to the MyHC-I promoter and thus for Sox6 repressive function, revealing a key role for Nfix in driving Sox6 activity. This feature is evolutionarily conserved, since the orthologs Nfixa and Sox6 contribute to repression of the slow-twitch phenotype in zebrafish embryos. These data demonstrate functional cooperation between Sox6 and Nfix in regulating MyHC-I expression during prenatal muscle development. Sox6 has opposite roles in MyHC-I regulation during embryonic and fetal myogenesis In embryonic muscle, Sox6 enhances MyHC-I expression via regulation of Mef2C In fetal muscle, Nfix is required for Sox6-mediated repression of MyHC-I The Sox6 and Nfixa orthologs cooperate in repressing smyhc1 in zebrafish
Collapse
Affiliation(s)
| | - Giovanni Maroli
- Department of Biosciences, University of Milan, Milan 20133, Italy
| | - Solei Cermenati
- Department of Biosciences, University of Milan, Milan 20133, Italy
| | | | - Andrea Ferrante
- Department of Biosciences, University of Milan, Milan 20133, Italy
| | - Giuliana Rossi
- Department of Biosciences, University of Milan, Milan 20133, Italy
| | - Giulio Cossu
- Department of Biosciences, University of Milan, Milan 20133, Italy; Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Oxford Road, M13 9PL Manchester, UK
| | - Monica Beltrame
- Department of Biosciences, University of Milan, Milan 20133, Italy
| | | |
Collapse
|
24
|
Yokoyama S, Ohno Y, Egawa T, Yasuhara K, Nakai A, Sugiura T, Ohira Y, Yoshioka T, Okita M, Origuchi T, Goto K. Heat shock transcription factor 1-associated expression of slow myosin heavy chain in mouse soleus muscle in response to unloading with or without reloading. Acta Physiol (Oxf) 2016; 217:325-37. [PMID: 27084024 DOI: 10.1111/apha.12692] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 11/28/2015] [Accepted: 04/11/2016] [Indexed: 12/28/2022]
Abstract
AIM The effects of heat shock transcription factor 1 (HSF1) deficiency on the fibre type composition and the expression level of nuclear factor of activated T cells (NFAT) family members (NFATc1, NFATc2, NFATc3 and NFATc4), phosphorylated glycogen synthase kinase 3α (p-GSK3α) and p-GSK3β, microRNA-208b (miR-208b), miR-499 and slow myosin heavy chain (MyHC) mRNAs (Myh7 and Myh7b) of antigravitational soleus muscle in response to unloading with or without reloading were investigated. METHODS HSF1-null and wild-type mice were subjected to continuous 2-week hindlimb suspension followed by 2- or 4-week ambulation recovery. RESULTS In wild-type mice, the relative population of slow type I fibres, the expression level of NFATc2, p-GSK3 (α and β), miR-208b, miR-499 and slow MyHC mRNAs (Myh7 and Myh7b) were all decreased with hindlimb suspension, but recovered after it. Significant interactions between train and time (the relative population of slow type I fibres; P = 0.01, the expression level of NFATc2; P = 0.001, p-GSKβ; P = 0.009, miR-208b; P = 0.002, miR-499; P = 0.04) suggested that these responses were suppressed in HSF1-null mice. CONCLUSION HSF1 may be a molecule in the regulation of the expression of slow MyHC as well as miR-208b, miR-499, NFATc2 and p-GSK3 (α and β) in mouse soleus muscle.
Collapse
Affiliation(s)
- S. Yokoyama
- Department of Locomotive Rehabilitation Science; Unit of Rehabilitation Sciences; Nagasaki University Graduate School of Biomedical Sciences; Nagasaki Japan
- Laboratory of Physiology; School of Health Science; Toyohashi SOZO University; Toyohashi Japan
| | - Y. Ohno
- Laboratory of Physiology; School of Health Science; Toyohashi SOZO University; Toyohashi Japan
| | - T. Egawa
- Department of Physiology; Graduate School of Health Science; Toyohashi SOZO University; Toyohashi Japan
| | - K. Yasuhara
- Department of Orthopaedic Surgery; St. Marianna University School of Medicine; Kawasaki Japan
| | - A. Nakai
- Department of Molecular Biology; Graduate School of Medicine; Yamaguchi University; Ube Japan
| | - T. Sugiura
- Faculty of Education; Yamaguchi University; Yamaguchi Japan
| | - Y. Ohira
- Faculty and Graduate School of Health and Sports Sciences; Doshisha University; Kyotanabe Japan
| | | | - M. Okita
- Department of Locomotive Rehabilitation Science; Unit of Rehabilitation Sciences; Nagasaki University Graduate School of Biomedical Sciences; Nagasaki Japan
| | - T. Origuchi
- Department of Locomotive Rehabilitation Science; Unit of Rehabilitation Sciences; Nagasaki University Graduate School of Biomedical Sciences; Nagasaki Japan
| | - K. Goto
- Laboratory of Physiology; School of Health Science; Toyohashi SOZO University; Toyohashi Japan
- Department of Physiology; Graduate School of Health Science; Toyohashi SOZO University; Toyohashi Japan
| |
Collapse
|
25
|
Ahammad AS, Asaduzzaman M, Asakawa S, Watabe S, Kinoshita S. Regulation of gene expression mediating indeterminate muscle growth in teleosts. Mech Dev 2015; 137:53-65. [DOI: 10.1016/j.mod.2015.02.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 01/19/2015] [Accepted: 02/02/2015] [Indexed: 01/13/2023]
|
26
|
Blais A. Myogenesis in the Genomics Era. J Mol Biol 2015; 427:2023-38. [DOI: 10.1016/j.jmb.2015.02.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/04/2015] [Accepted: 02/05/2015] [Indexed: 01/06/2023]
|