151
|
Petrany MJ, Millay DP. Cell Fusion: Merging Membranes and Making Muscle. Trends Cell Biol 2019; 29:964-973. [PMID: 31648852 PMCID: PMC7849503 DOI: 10.1016/j.tcb.2019.09.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 12/12/2022]
Abstract
Cell fusion is essential for the development of multicellular organisms, and plays a key role in the formation of various cell types and tissues. Recent findings have highlighted the varied protein machinery that drives plasma-membrane merger in different systems, which is characterized by diverse structural and functional elements. We highlight the discovery and activities of several key sets of fusion proteins that together offer an evolving perspective on cell membrane fusion. We also emphasize recent discoveries in vertebrate myoblast fusion in skeletal muscle, which is composed of numerous multinucleated myofibers formed by the fusion of progenitor cells during development.
Collapse
Affiliation(s)
- Michael J Petrany
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Douglas P Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA.
| |
Collapse
|
152
|
Dhanani ZN, Mann G, Adegoke OAJ. Depletion of branched-chain aminotransferase 2 (BCAT2) enzyme impairs myoblast survival and myotube formation. Physiol Rep 2019; 7:e14299. [PMID: 31833233 PMCID: PMC6908738 DOI: 10.14814/phy2.14299] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 12/15/2022] Open
Abstract
Much is known about the positive effects of branched-chain amino acids (BCAA) in regulating muscle protein metabolism. Comparatively much less is known about the effects of these amino acids and their metabolites in regulating myotube formation. Using cultured myoblasts, we showed that although leucine is required for myotube formation, this requirement is easily met by α-ketoisocaproic acid, the ketoacid of leucine. We then demonstrated increases in the expression of the first two enzymes in the catabolism of the three BCAA, branched-chain amino transferase (BCAT2) and branched-chain α-ketoacid dehydrogenase (BCKD), with ~3× increase in BCKD protein expression (p < .05) during differentiation. Furthermore, depletion of BCAT2 abolished myoblast differentiation, as indicated by reduction in the levels of myosin heavy chain-1, troponin and myogenin. Supplementation of incubation medium with branched-chain α-ketoacids or related metabolites derivable from BCAT2 functions did not rescue the defects. However, co-depletion of BCKD kinase partially rescued the defects. Collectively, our data indicate a requirement for BCAA catabolism during myotube formation and that this requirement for BCAT2 likely goes beyond the need for this enzyme to generate the α-ketoacids of the BCAA.
Collapse
Affiliation(s)
- Zameer N. Dhanani
- School of Kinesiology and Health ScienceMuscle Health Research CentreYork UniversityTorontoONCanada
| | - Gagandeep Mann
- School of Kinesiology and Health ScienceMuscle Health Research CentreYork UniversityTorontoONCanada
| | | |
Collapse
|
153
|
Geisbrecht ER, Baylies MK. In memoriam: Susan Abmayr (1956–2019) – “What do we do? Whatever it takes!”. Skelet Muscle 2019. [PMCID: PMC6882191 DOI: 10.1186/s13395-019-0215-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
154
|
NADPH Oxidase 4 Contributes to Myoblast Fusion and Skeletal Muscle Regeneration. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3585390. [PMID: 31827673 PMCID: PMC6885834 DOI: 10.1155/2019/3585390] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/24/2019] [Accepted: 10/08/2019] [Indexed: 01/28/2023]
Abstract
Myoblast fusion is an essential step in skeletal muscle development and regeneration. NADPH oxidase 4 (Nox4) regulates cellular processes such as proliferation, differentiation, and survival by producing reactive oxygen species (ROS). Insulin-like growth factor 1 induces muscle hypertrophy via Nox4, but its function in myoblast fusion remains elusive. Here, we report a ROS-dependent role of Nox4 in myoblast differentiation. Regenerating muscle fibers after injury by cardiotoxin had a lower cross-sectional area in Nox4-knockout (KO) mice than myofibers in wild-type (WT) mice. Diameters and fusion index values of myotubes differentiated from Nox4-KO primary myoblasts were significantly lower than those of myotubes derived from WT myoblasts. However, no difference was observed in the differentiation index and expression of MyoD, myogenin, and myosin heavy chain 3 (MHC) between KO and WT myotubes. The decreased fusion index was also observed during differentiation of primary myoblasts and C2C12 cells with suppressed Nox4 expression. In contrast, in C2C12 cells overexpressing Nox4, the fusion index was increased, whereas the differentiation index and MHC and myogenin protein expression were not affected compared to control. Interestingly, the expression of myomaker (Tmem8c), a fusogenic protein that controls myoblast fusion, was reduced in Nox4-knockdown C2C12 cells. The myomaker expression level was proportional to the cellular ROS level, which was regulated by of Nox4 expression level. These results suggests that Nox4 contributes to myoblast fusion, possibly through the regulation of myomaker expression via ROS production, and that Nox4-dependent ROS may promote skeletal muscle regeneration and growth.
Collapse
|
155
|
Recovery in the Myogenic Program of Congenital Myotonic Dystrophy Myoblasts after Excision of the Expanded (CTG) n Repeat. Int J Mol Sci 2019; 20:ijms20225685. [PMID: 31766224 PMCID: PMC6888582 DOI: 10.3390/ijms20225685] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 11/11/2019] [Indexed: 12/12/2022] Open
Abstract
The congenital form of myotonic dystrophy type 1 (cDM) is caused by the large-scale expansion of a (CTG•CAG)n repeat in DMPK and DM1-AS. The production of toxic transcripts with long trinucleotide tracts from these genes results in impairment of the myogenic differentiation capacity as cDM’s most prominent morpho-phenotypic hallmark. In the current in vitro study, we compared the early differentiation programs of isogenic cDM myoblasts with and without a (CTG)2600 repeat obtained by gene editing. We found that excision of the repeat restored the ability of cDM myoblasts to engage in myogenic fusion, preventing the ensuing myotubes from remaining immature. Although the cDM-typical epigenetic status of the DM1 locus and the expression of genes therein were not altered upon removal of the repeat, analyses at the transcriptome and proteome level revealed that early abnormalities in the temporal expression of differentiation regulators, myogenic progression markers, and alternative splicing patterns before and immediately after the onset of differentiation became normalized. Our observation that molecular and cellular features of cDM are reversible in vitro and can be corrected by repeat-directed genome editing in muscle progenitors, when already committed and poised for myogenic differentiation, is important information for the future development of gene therapy for different forms of myotonic dystrophy type 1 (DM1).
Collapse
|
156
|
Chen B, You W, Shan T. Myomaker, and Myomixer-Myomerger-Minion modulate the efficiency of skeletal muscle development with melatonin supplementation through Wnt/β-catenin pathway. Exp Cell Res 2019; 385:111705. [PMID: 31682812 DOI: 10.1016/j.yexcr.2019.111705] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/30/2019] [Accepted: 10/29/2019] [Indexed: 02/03/2023]
Abstract
Melatonin, a pleiotropic hormone secreted from the pineal gland, has been shown to exert beneficial effects in muscle regeneration and repair due to its functional diversity, including anti-inflammation, anti-apoptosis, and anti-oxidative activity. However, little is known about the negative role of melatonin in myogenesis. Here, using skeletal muscle cells, we found that melatonin promoted C2C12 cells proliferation and inhibits differentiation both in C2C12 cells and primary myoblasts in mice. Melatonin administration significantly down-regulated differentiation and fusion related genes and inhibited myotube formation both in C2C12 cells and primary myoblasts in mice. RNA-seq showed that melatonin down-regulated essential fusion pore components Myomaker and Myomixer-Myomerger-Minion. Moreover, melatonin suppressed Wnt/β-catenin signaling. Inhibition of GSK3β by LiCl rescued the influence of melatonin on differentiation efficiency, Myomaker, but not Myomxier in C2C12 cells. In conclusion, melatonin inhibits myogenic differentiation, Myomaker, and Myomixer through reducing Wnt/β-catenin signaling. These data establish a link between melatonin and fusogenic membrane proteins Myomaker and Myomixer, and suggest the new perspective of melatonin in treatment or preventment of muscular diseases.
Collapse
Affiliation(s)
- Bide Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, China; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Wenjing You
- College of Animal Sciences, Zhejiang University, Hangzhou, China; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, China; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China.
| |
Collapse
|
157
|
Tran MP, Tsutsumi R, Erberich JM, Chen KD, Flores MD, Cooper KL. Evolutionary loss of foot muscle during development with characteristics of atrophy and no evidence of cell death. eLife 2019; 8:50645. [PMID: 31612857 PMCID: PMC6855805 DOI: 10.7554/elife.50645] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 10/01/2019] [Indexed: 12/19/2022] Open
Abstract
Many species that run or leap across sparsely vegetated habitats, including horses and deer, evolved the severe reduction or complete loss of foot muscles as skeletal elements elongated and digits were lost, and yet the developmental mechanisms remain unknown. Here, we report the natural loss of foot muscles in the bipedal jerboa, Jaculus jaculus. Although adults have no muscles in their feet, newborn animals have muscles that rapidly disappear soon after birth. We were surprised to find no evidence of apoptotic or necrotic cell death during stages of peak myofiber loss, countering well-supported assumptions of developmental tissue remodeling. We instead see hallmarks of muscle atrophy, including an ordered disassembly of the sarcomere associated with upregulation of the E3 ubiquitin ligases, MuRF1 and Atrogin-1. We propose that the natural loss of muscle, which remodeled foot anatomy during evolution and development, involves cellular mechanisms that are typically associated with disease or injury. Intrinsic muscles are a group of muscles deep inside the hands and feet. They help to control the precise movements required, for example, for a pianist to play their instrument or for certain animals to climb with remarkable agility. Some animals, such as horses and deer, have evolved in such a way that they no longer grasp objects with hands and feet. Where intrinsic muscles were once present in the hands and feet of their ancestors, these animals now have strong ligaments that prevent over-extension of the wrist and ankle joints during hard landings. Given their size, it is difficult to study horses and deer in the laboratory and understand how they lost their intrinsic muscles during evolution. Tran et al. therefore focused on a small rodent called the lesser Egyptian jerboa, which also displays long legs with strong ligaments and no intrinsic muscles. Newborn jerboas have foot muscles that look very much like the intrinsic muscles found in mice, but these muscles disappear within 4 days of birth. A mechanism called programmed cell death is often responsible for specific tissues disappearing during development, but the experiments of Tran et al. revealed that this was not the case in jerboas. Instead, their intrinsic muscles were degraded by processes triggered by genes that disassemble underused muscles. In mice and humans, fasting, nerve injuries, or immobility trigger this type of muscle degradation, but in jerboas these processes appear to be a normal part of development. This unexpected discovery shows that development and disease-like processes are linked, and that more studies of nontraditional research animals may help scientists better understand these connections.
Collapse
Affiliation(s)
- Mai P Tran
- Division of Biological Sciences, Section of Cellular and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Rio Tsutsumi
- Division of Biological Sciences, Section of Cellular and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Joel M Erberich
- Division of Biological Sciences, Section of Cellular and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Kevin D Chen
- Division of Biological Sciences, Section of Cellular and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Michelle D Flores
- Division of Biological Sciences, Section of Cellular and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Kimberly L Cooper
- Division of Biological Sciences, Section of Cellular and Developmental Biology, University of California, San Diego, La Jolla, United States
| |
Collapse
|
158
|
Abstract
Cell-cell fusion is a fundamental process underlying fertilization, development, regeneration and physiology of metazoans. It is a multi-step process involving cell recognition and adhesion, actin cytoskeletal rearrangements, fusogen engagement, lipid mixing and fusion pore formation, ultimately resulting in the integration of two fusion partners. Here, we focus on the asymmetric actin cytoskeletal rearrangements at the site of fusion, known as the fusogenic synapse, which was first discovered during myoblast fusion in Drosophila embryos and later also found in mammalian muscle and non-muscle cells. At the asymmetric fusogenic synapse, actin-propelled invasive membrane protrusions from an attacking fusion partner trigger actomyosin-based mechanosensory responses in the receiving cell. The interplay between the invasive and resisting forces generated by the two fusion partners puts the fusogenic synapse under high mechanical tension and brings the two cell membranes into close proximity, promoting the engagement of fusogens to initiate fusion pore formation. In this Cell Science at a Glance article and the accompanying poster, we highlight the molecular, cellular and biophysical events at the asymmetric fusogenic synapse using Drosophila myoblast fusion as a model.
Collapse
Affiliation(s)
- Ji Hoon Kim
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elizabeth H Chen
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA .,Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
159
|
Der Vartanian A, Chabanais J, Carrion C, Maftah A, Germot A. Downregulation of POFUT1 Impairs Secondary Myogenic Fusion Through a Reduced NFATc2/IL-4 Signaling Pathway. Int J Mol Sci 2019; 20:ijms20184396. [PMID: 31500188 PMCID: PMC6770550 DOI: 10.3390/ijms20184396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 08/26/2019] [Accepted: 09/05/2019] [Indexed: 12/25/2022] Open
Abstract
Past work has shown that the protein O-fucosyltransferase 1 (POFUT1) is involved in mammal myogenic differentiation program. Pofut1 knockdown (Po –) in murine C2C12 cells leads to numerous elongated and thin myotubes, suggesting significant defects in secondary fusion. Among the few pathways involved in this process, NFATc2/IL-4 is described as the major one. To unravel the impact of POFUT1 on secondary fusion, we used wild-type (WT) C2C12 and Po – cell lines to follow Myf6, Nfatc2, Il-4 and Il-4rα expressions during a 120 h myogenic differentiation time course. Secreted IL-4 was quantified by ELISA. IL-4Rα expression and its labeling on myogenic cell types were investigated by Western blot and immunofluorescence, respectively. Phenotypic observations of cells treated with IL-4Rα blocking antibody were performed. In Po –, we found a decrease in nuclei number per myotube and a downexpression of Myf6. The observed downregulation of Nfatc2 is correlated to a diminution of secreted IL-4 and to the low level of IL-4Rα for reserve cells. Neutralization of IL-4Rα on WT C2C12 promotes myonuclear accretion defects, similarly to those identified in Po –. Thus, POFUT1 could be a new controller of myotube growth during myogenesis, especially through NFATc2/IL-4 signaling pathway.
Collapse
Affiliation(s)
- Audrey Der Vartanian
- PEIRENE, EA 7500, Glycosylation et différenciation cellulaire, Université de Limoges, F-87000 Limoges, France
- present address: INSERM, IMRB U955-E10, Faculté de Médecine, Université Paris Est Créteil, F-94000 Créteil, France
| | - Julien Chabanais
- PEIRENE, EA 7500, Glycosylation et différenciation cellulaire, Université de Limoges, F-87000 Limoges, France
| | - Claire Carrion
- UMR CNRS 7276, Contrôle de la Réponse Immune et des Lymphoproliférations, Université de Limoges, F-87000 Limoges, France
| | - Abderrahman Maftah
- PEIRENE, EA 7500, Glycosylation et différenciation cellulaire, Université de Limoges, F-87000 Limoges, France
| | - Agnès Germot
- PEIRENE, EA 7500, Glycosylation et différenciation cellulaire, Université de Limoges, F-87000 Limoges, France
- Correspondence: ; Tel.: +33-(0)5-55-45-76-57
| |
Collapse
|
160
|
Huang Y, Wu S, Zhang J, Wen H, Zhang M, He F. Methylation status and expression patterns of myomaker gene play important roles in postnatal development in the Japanese flounder (Paralichthys olivaceus). Gen Comp Endocrinol 2019; 280:104-114. [PMID: 31002826 DOI: 10.1016/j.ygcen.2019.04.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 02/28/2019] [Accepted: 04/16/2019] [Indexed: 01/01/2023]
Abstract
Myomaker is a membrane protein that plays a crucial role in the fusion of myoblasts during muscle growth. DNA methylation, a significant factor, regulates gene expression. The aim of this study was to examine the methylation and mRNA expression patterns of the myomaker gene during 8 different postnatal developmental stages in the Japanese flounder (L: 7 days post hatch (dph); M1: 21 dph; M2: 28 dph; M3: 35 dph; J1: 90 dph; J2: 180 dph; A1: 24 months; A2: 36 months). Muscle tissue samples were taken from Japanese flounder at different postnatal development stages to measure the extent of DNA methylation and gene expression. Methylation level in the promoter and exon 1 of myomaker was measured using bisulfite sequencing, and the relative expression of myomaker during each developmental stage was measured by quantitative PCR. The relative expression levels of myomaker were up-regulated from stages L to M2, M3 to J2, and methylation of myomaker was negatively correlated with mRNA expression. Furthermore, the CpG site located at -26 bp in the promoter was the lowest methylated region in all developmental stages. These results offer a basis for understanding the mechanism by which myomaker regulates muscle formation during postnatal development.
Collapse
Affiliation(s)
- Yajuan Huang
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, China
| | - Shuxian Wu
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, China
| | - Jingru Zhang
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, China
| | - Haishen Wen
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, China
| | - Meizhao Zhang
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, China
| | - Feng He
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, China.
| |
Collapse
|
161
|
Faust JJ, Balabiyev A, Heddleston JM, Podolnikova NP, Baluch DP, Chew TL, Ugarova TP. An actin-based protrusion originating from a podosome-enriched region initiates macrophage fusion. Mol Biol Cell 2019; 30:2254-2267. [PMID: 31242090 PMCID: PMC6743464 DOI: 10.1091/mbc.e19-01-0009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 05/15/2019] [Accepted: 06/17/2019] [Indexed: 01/24/2023] Open
Abstract
Macrophage fusion resulting in the formation of multinucleated giant cells occurs in a variety of chronic inflammatory diseases, yet the mechanism responsible for initiating this process is unknown. Here, we used live cell imaging to show that actin-based protrusions at the leading edge initiate macrophage fusion. Phase-contrast video microscopy demonstrated that in the majority of events, short protrusions (∼3 µm) between two closely apposed cells initiated fusion, but occasionally we observed long protrusions (∼12 µm). Using macrophages isolated from LifeAct mice and imaging with lattice light sheet microscopy, we further found that fusion-competent protrusions formed at sites enriched in podosomes. Inducing fusion in mixed populations of GFP- and mRFP-LifeAct macrophages showed rapid spatial overlap between GFP and RFP signal at the site of fusion. Cytochalasin B strongly reduced fusion and when rare fusion events occurred, protrusions were not observed. Fusion of macrophages deficient in Wiskott-Aldrich syndrome protein and Cdc42, key molecules involved in the formation of actin-based protrusions and podosomes, was also impaired both in vitro and in vivo. Finally, inhibiting the activity of the Arp2/3 complex decreased fusion and podosome formation. Together these data suggest that an actin-based protrusion formed at the leading edge initiates macrophage fusion.
Collapse
Affiliation(s)
- James J. Faust
- School of Life Sciences, Arizona State University, Tempe, AZ 85287
| | - Arnat Balabiyev
- School of Life Sciences, Arizona State University, Tempe, AZ 85287
| | - John M. Heddleston
- Advanced Imaging Center, HHMI Janelia Research Campus, Ashburn, VA 20147
| | | | - D. Page Baluch
- School of Life Sciences, Arizona State University, Tempe, AZ 85287
| | - Teng-Leong Chew
- Advanced Imaging Center, HHMI Janelia Research Campus, Ashburn, VA 20147
| | | |
Collapse
|
162
|
Ikeda K, Horie-Inoue K, Inoue S. Functions of estrogen and estrogen receptor signaling on skeletal muscle. J Steroid Biochem Mol Biol 2019; 191:105375. [PMID: 31067490 DOI: 10.1016/j.jsbmb.2019.105375] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/03/2019] [Indexed: 12/23/2022]
Abstract
Activity of estrogen, a sex steroid hormone, is not only limited to the reproductive organs but also involves other organs and tissues, including skeletal muscle. In postmenopausal women, estrogen decline causes endocrine and metabolic dysfunction, leading to a predisposition to osteoporosis, metabolic syndrome, and decreased muscle mass and strength. The decline in skeletal muscle mass often associates with sarcopenia, a popular condition observed in fragile elder people. In addition, varying estrogen levels associated with the menstrual phases may modulate exercise performance in women. Estrogen is thus considered to play a crucial role in skeletal muscle homeostasis and exercise capacity, although its precise mechanisms remain to be elucidated. In this article, we review the role of estrogen in the skeletal muscle, outlining the proposed molecular mechanisms. We especially focus on the current understanding of estrogen actions on mitochondria metabolism in skeletal muscle.
Collapse
Affiliation(s)
- Kazuhiro Ikeda
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Kuniko Horie-Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Satoshi Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan; Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.
| |
Collapse
|
163
|
Vajanthri KY, Sidu RK, Poddar S, Singh AK, Mahto SK. Combined substrate micropatterning and FFT analysis reveals myotube size control and alignment by contact guidance. Cytoskeleton (Hoboken) 2019; 76:269-285. [DOI: 10.1002/cm.21527] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/23/2019] [Accepted: 05/02/2019] [Indexed: 12/30/2022]
Affiliation(s)
- Kiran Yellappa Vajanthri
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical EngineeringIndian Institute of Technology (Banaras Hindu University) Varanasi Uttar Pradesh India
| | - Rakesh Kumar Sidu
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical EngineeringIndian Institute of Technology (Banaras Hindu University) Varanasi Uttar Pradesh India
| | - Suruchi Poddar
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical EngineeringIndian Institute of Technology (Banaras Hindu University) Varanasi Uttar Pradesh India
| | - Ashish Kumar Singh
- School of Biochemical EngineeringIndian Institute of Technology (Banaras Hindu University) Varanasi Uttar Pradesh India
| | - Sanjeev Kumar Mahto
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical EngineeringIndian Institute of Technology (Banaras Hindu University) Varanasi Uttar Pradesh India
- Center for Advanced Biomaterials and Tissue EngineeringIndian Institute of Technology (Banaras Hindu University) Varanasi Uttar Pradesh India
| |
Collapse
|
164
|
Vorontsova YE, Zavoloka EL, Cherezov RO, Simonova OB. Drosophila as a Model System Used for Searching the Genes, Signaling Pathways, and Mechanisms Controlling Cytoskeleton Formation. Russ J Dev Biol 2019. [DOI: 10.1134/s1062360419010065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
165
|
Shi J, Cai M, Si Y, Zhang J, Du S. Knockout of myomaker results in defective myoblast fusion, reduced muscle growth and increased adipocyte infiltration in zebrafish skeletal muscle. Hum Mol Genet 2019; 27:3542-3554. [PMID: 30016436 DOI: 10.1093/hmg/ddy268] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 07/13/2018] [Indexed: 01/08/2023] Open
Abstract
The fusion of myoblasts into multinucleated muscle fibers is vital to skeletal muscle development, maintenance and regeneration. Genetic mutations in the Myomaker (mymk) gene cause Carey-Fineman-Ziter syndrome (CFZS) in human populations. To study the regulation of mymk gene expression and function, we generated three mymk mutant alleles in zebrafish using Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) technology and analyzed the effects of mymk knockout on muscle development and growth. Our studies demonstrated that knockout of mymk resulted in defective myoblast fusion in zebrafish embryos and increased mortality at larval stage around 35-45 days post-fertilization. The viable homozygous mutants were smaller in size and weighed approximately one-third the weight of the wild type (WT) sibling at 3 months old. The homozygous mutants showed craniofacial deformities, resembling the facial defect observed in human populations with CFZS. Histological analysis revealed that skeletal muscles of mymk mutants contained mainly small-size fibers and substantial intramuscular adipocyte infiltration. Single fiber analysis revealed that myofibers in mymk mutant were predominantly single-nucleated fibers. However, myofibers with multiple myonuclei were observed, although the number of nuclei per fiber was much less compared with that in WT fibers. Overexpression of sonic Hedgehog inhibited mymk expression in zebrafish embryos and blocked myoblast fusion. Collectively, these studies demonstrated that mymk is essential for myoblast fusion during muscle development and growth.
Collapse
Affiliation(s)
- Jun Shi
- Institute of Marine and Environmental Technology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21202, USA
- Department of Bioengineering and Environmental Science, Changsha University, Hunan 410003, China
| | - Mengxin Cai
- Institute of Marine and Environmental Technology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21202, USA
- Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi' an 710062, China
| | - Yufeng Si
- Institute of Marine and Environmental Technology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21202, USA
| | - Jianshe Zhang
- Department of Bioengineering and Environmental Science, Changsha University, Hunan 410003, China
| | - Shaojun Du
- Institute of Marine and Environmental Technology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21202, USA
- Department of Bioengineering and Environmental Science, Changsha University, Hunan 410003, China
| |
Collapse
|
166
|
Chuang MC, Lin SS, Ohniwa RL, Lee GH, Su YA, Chang YC, Tang MJ, Liu YW. Tks5 and Dynamin-2 enhance actin bundle rigidity in invadosomes to promote myoblast fusion. J Cell Biol 2019; 218:1670-1685. [PMID: 30894403 PMCID: PMC6504888 DOI: 10.1083/jcb.201809161] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/22/2019] [Accepted: 03/04/2019] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle development requires the cell-cell fusion of differentiated myoblasts to form muscle fibers. The actin cytoskeleton is known to be the main driving force for myoblast fusion; however, how actin is organized to direct intercellular fusion remains unclear. Here we show that an actin- and dynamin-2-enriched protrusive structure, the invadosome, is required for the fusion process of myogenesis. Upon differentiation, myoblasts acquire the ability to form invadosomes through isoform switching of a critical invadosome scaffold protein, Tks5. Tks5 directly interacts with and recruits dynamin-2 to the invadosome and regulates its assembly around actin filaments to strengthen the stiffness of dynamin-actin bundles and invadosomes. These findings provide a mechanistic framework for the acquisition of myogenic fusion machinery during myogenesis and reveal a novel structural function for Tks5 and dynamin-2 in organizing actin filaments in the invadosome to drive membrane fusion.
Collapse
Affiliation(s)
- Mei-Chun Chuang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shan-Shan Lin
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ryosuke L Ohniwa
- Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Center for Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Gang-Hui Lee
- International Center of Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - You-An Su
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Chen Chang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Jer Tang
- International Center of Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan.,Department of Physiology, Medical College, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Wen Liu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan .,Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
167
|
McClure MJ, Ramey AN, Rashid M, Boyan BD, Schwartz Z. Integrin-α7 signaling regulates connexin 43, M-cadherin, and myoblast fusion. Am J Physiol Cell Physiol 2019; 316:C876-C887. [PMID: 30892939 DOI: 10.1152/ajpcell.00282.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Regenerative medicine treatments for severe skeletal muscle injuries are limited, resulting in persistent functional deficits. Clinical options include neglecting the wound with the expectation that fibrosis will develop or using an autologous muscle graft with minimal functional improvement. A regenerative matrix can be used, but muscle fiber development on these matrices remains a challenge in vivo. Here, we explored the fundamental mechanisms that mediate cell-substrate signaling and its effect on cell-cell communication during myoblast fusion and tube formation to improve outcomes following implantation of matrices used to stimulate muscle regeneration. We previously reported that integrin-α7 was increased on anisotropic biomaterials, suggesting a role for α7β1 signaling in myoblast communication via connexin 43 and M-cadherin. Our results demonstrated that α7 silencing blocked expression of myogenic differentiation factor 1 (Myod), myogenin (Myog), myogenic factor 6 (Myf6), myosin heavy chain type 1 (Myh1), and transmembrane protein 8c (Tmem8c), indicating that myoblast fusion was inhibited. Expression of α5 and M-cadherin decreased but β1 and connexin 43 increased. We examined protein production and observed reduced extracellular-signal regulated kinase 1/2 (ERK) in α7-silenced cells that correlated with upregulation of connexin 43 and M-cadherin, suggesting a compensatory pathway. These results indicate that α7 signaling plays a critical role in ex vivo fusion and implicates a relationship with connexin 43 and M-cadherin.
Collapse
Affiliation(s)
- Michael J McClure
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University , Richmond, Virginia
| | - Allison N Ramey
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University , Richmond, Virginia
| | - Mashaba Rashid
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University , Richmond, Virginia
| | - Barbara D Boyan
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University , Richmond, Virginia.,Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology , Atlanta, Georgia
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University , Richmond, Virginia.,Department of Periodontics, University of Texas Health Sciences Center at San Antonio , San Antonio, Texas
| |
Collapse
|
168
|
Takahashi K, Itakura E, Takano K, Endo T. DA-Raf, a dominant-negative regulator of the Ras–ERK pathway, is essential for skeletal myocyte differentiation including myoblast fusion and apoptosis. Exp Cell Res 2019; 376:168-180. [DOI: 10.1016/j.yexcr.2019.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/19/2019] [Accepted: 02/07/2019] [Indexed: 12/21/2022]
|
169
|
Khitun A, Ness TJ, Slavoff SA. Small open reading frames and cellular stress responses. Mol Omics 2019; 15:108-116. [PMID: 30810554 DOI: 10.1039/c8mo00283e] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Small open reading frames (smORFs) encoding polypeptides of less than 100 amino acids in eukaryotes (50 amino acids in prokaryotes) were historically excluded from genome annotation. However, recent advances in genomics, ribosome footprinting, and proteomics have revealed thousands of translated smORFs in genomes spanning evolutionary space. These smORFs can encode functional polypeptides, or act as cis-translational regulators. Herein we review evidence that some smORF-encoded polypeptides (SEPs) participate in stress responses in both prokaryotes and eukaryotes, and that some upstream ORFs (uORFs) regulate stress-responsive translation of downstream cistrons in eukaryotic cells. These studies provide insight into a regulated subclass of smORFs and suggest that at least some SEPs may participate in maintenance of cellular homeostasis under stress.
Collapse
Affiliation(s)
- Alexandra Khitun
- Chemical Biology Institute, Yale University, West Haven, CT 06516, USA. and Department of Chemistry, Yale University, New Haven, CT 06520, USA
| | - Travis J Ness
- Chemical Biology Institute, Yale University, West Haven, CT 06516, USA. and Department of Chemistry, Yale University, New Haven, CT 06520, USA
| | - Sarah A Slavoff
- Chemical Biology Institute, Yale University, West Haven, CT 06516, USA. and Department of Chemistry, Yale University, New Haven, CT 06520, USA and Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
170
|
Si Y, Wen H, Du S. Genetic Mutations in jamb, jamc, and myomaker Revealed Different Roles on Myoblast Fusion and Muscle Growth. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2019; 21:111-123. [PMID: 30467785 PMCID: PMC6467518 DOI: 10.1007/s10126-018-9865-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 11/15/2018] [Indexed: 05/08/2023]
Abstract
Myoblast fusion is a vital step for skeletal muscle development, growth, and regeneration. Loss of Jamb, Jamc, or Myomaker (Mymk) function impaired myoblast fusion in zebrafish embryos. In addition, mymk mutation hampered fish muscle growth. However, the effect of Jamb and Jamc deficiency on fish muscle growth is not clear. Moreover, whether jamb;jamc and jamb;mymk double mutations have stronger effects on myoblast fusion and muscle growth remains to be investigated. Here, we characterized the muscle development and growth in jamb, jamc, and mymk single and double mutants in zebrafish. We found that although myoblast fusion was compromised in jamb and jamc single or jamb;jamc double mutants, these mutant fish showed no defect in muscle cell fusion during muscle growth. The mutant fish were able to grow into adults that were indistinguishable from the wild-type sibling. In contrast, the jamb;mymk double mutants exhibited a stronger muscle phenotype compared to the jamb and jamc single and double mutants. The jamb;mymk double mutant showed reduced growth and partial lethality, similar to a mymk single mutant. Single fiber analysis of adult skeletal myofibers revealed that jamb, jamc, or jamb;jamc mutants contained mainly multinucleated myofibers, whereas jamb;mymk double mutants contained mostly mononucleated fibers. Significant intramuscular adipocyte infiltration was found in skeletal muscles of the jamb;mymk mutant. Collectively, these studies demonstrate that although Jamb, Jamc, and Mymk are all involved in myoblast fusion during early myogenesis, they have distinct roles in myoblast fusion during muscle growth. While Mymk is essential for myoblast fusion during both muscle development and growth, Jamb and Jamc are dispensable for myoblast fusion during muscle growth.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Cell Communication
- Cell Differentiation
- Cell Fusion
- Embryo, Nonmammalian
- Gene Expression Regulation, Developmental
- Junctional Adhesion Molecule B/deficiency
- Junctional Adhesion Molecule B/genetics
- Membrane Proteins/deficiency
- Membrane Proteins/genetics
- Muscle Development/genetics
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/metabolism
- Muscle Proteins/deficiency
- Muscle Proteins/genetics
- Muscle, Skeletal/cytology
- Muscle, Skeletal/growth & development
- Muscle, Skeletal/metabolism
- Mutation
- Myoblasts/cytology
- Myoblasts/metabolism
- Receptors, Cell Surface/deficiency
- Receptors, Cell Surface/genetics
- Zebrafish/genetics
- Zebrafish/growth & development
- Zebrafish/metabolism
- Zebrafish Proteins/deficiency
- Zebrafish Proteins/genetics
Collapse
Affiliation(s)
- Yufeng Si
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, 701 East Pratt Street, Baltimore, MD, 21202, USA
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Haishen Wen
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Shaojun Du
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, 701 East Pratt Street, Baltimore, MD, 21202, USA.
| |
Collapse
|
171
|
Sefton EM, Kardon G. Connecting muscle development, birth defects, and evolution: An essential role for muscle connective tissue. Curr Top Dev Biol 2019; 132:137-176. [PMID: 30797508 DOI: 10.1016/bs.ctdb.2018.12.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Skeletal muscle powers all movement of the vertebrate body and is distributed in multiple regions that have evolved distinct functions. Axial muscles are ancestral muscles essential for support and locomotion of the whole body. The evolution of the head was accompanied by development of cranial muscles essential for eye movement, feeding, vocalization, and facial expression. With the evolution of paired fins and limbs and their associated muscles, vertebrates gained increased locomotor agility, populated the land, and acquired fine motor skills. Finally, unique muscles with specialized functions have evolved in some groups, and the diaphragm which solely evolved in mammals to increase respiratory capacity is one such example. The function of all these muscles requires their integration with the other components of the musculoskeletal system: muscle connective tissue (MCT), tendons, bones as well as nerves and vasculature. MCT is muscle's closest anatomical and functional partner. Not only is MCT critical in the adult for muscle structure and function, but recently MCT in the embryo has been found to be crucial for muscle development. In this review, we examine the important role of the MCT in axial, head, limb, and diaphragm muscles for regulating normal muscle development, discuss how defects in MCT-muscle interactions during development underlie the etiology of a range of birth defects, and explore how changes in MCT development or communication with muscle may have led to the modification and acquisition of new muscles during vertebrate evolution.
Collapse
Affiliation(s)
- Elizabeth M Sefton
- Department of Human Genetics, University of Utah, Salt Lake City, UT, United States
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT, United States.
| |
Collapse
|
172
|
Okamoto S, Asgar NF, Yokota S, Saito K, Minokoshi Y. Role of the α2 subunit of AMP-activated protein kinase and its nuclear localization in mitochondria and energy metabolism-related gene expressions in C2C12 cells. Metabolism 2019; 90:52-68. [PMID: 30359677 DOI: 10.1016/j.metabol.2018.10.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/07/2018] [Accepted: 10/16/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND AMP-activated protein kinase (AMPK), a heterotrimer with α1 or α2 catalytic subunits, acts as an energy sensor and regulates cellular homeostasis. Whereas AMPKα1 is necessary for myogenesis in skeletal muscle, the role of AMPKα2 in myogenic differentiation and energy metabolism-related gene expressions has remained unclear. We here examined the specific roles of AMPKα1 and AMPKα2 in the myogenic differentiation and mitochondria and energy metabolism-related gene expressions in C2C12 cells. MATERIALS AND METHODS Stable C2C12 cell lines expressing a scramble short hairpin RNA (shRNA) or shRNAs specific for AMPKα1 (shAMPKα1), AMPKα2 (shAMPKα2), or both AMPKα1 and AMPKα2 (shPanAMPK) were generated by lentivirus infection. Lentiviruses encoding wild-type AMPKα2 (WT-AMPKα2) or AMPKα2 with a mutated nuclear localization signal (ΔNLS-AMPKα2) were also constructed for introduction into myoblasts. Myogenesis was induced by culture of C2C12 myoblasts for 6 days in differentiation medium. RESULTS The amount of AMPKα2 increased progressively, whereas that of AMPKα1 remained constant, during the differentiation of myoblasts into myotubes. Expression of shPanAMPK or shAMPKα1, but not that of shAMPKα2, attenuated the proliferation of myoblasts as well as the phosphorylation of both acetyl-CoA carboxylase and the autophagy-initiating kinase ULK1 in myotubes. Up-regulation of myogenin mRNA, a marker for the middle stage of myogenesis, was attenuated in differentiating myotubes expressing shPanAMPK or shAMPKα1. In contrast, up-regulation of gene expression for muscle creatine kinase (MCK), a late-stage differentiation marker, as well as for genes related to mitochondrial biogenesis including the transcriptional coactivator peroxisome proliferator-activated receptor-γ coactivator-1α1 and α4 (PGC-1α1 and PGC-1α4) and mitochondria-specific genes such as cytochrome c were attenuated in myotubes expressing shAMPKα2 or shPanAMPK. The diameter of myotubes expressing shPanAMPK or shAMPKα2 was reduced, whereas that of those expressing shAMPKα1 was increased, compared with myotubes expressing scramble shRNA. A portion of AMPKα2 became localized to the nucleus during myogenic differentiation. The AMPK activator AICAR (5-aminoimidazole-4-carboxamide ribonucleotide) and 2-deoxyglucose (2DG) each induced the nuclear translocation of WT-AMPKα2, but not that of ΔNLS-AMPKα2. Finally, expression of WT-AMPKα2 increased the mRNA abundance of PGC-1α1 and MCK mRNAs as well as cell diameter and tended to increase that of PGC-1α4, whereas that of ΔNLS-AMPKα2 increased only the abundance of MCK mRNA, in myotubes depleted of endogenous AMPKα2. CONCLUSION TAMPKα1 and AMPKα2 have distinct roles in myogenic differentiation of C2C12 cells, with AMPKα1 contributing to the middle stage of myogenesis and AMPKα2 to the late stage. AMPKα2 regulates gene expressions including MCK, PGC-1α1 and PGC-1α4 and mitochondria-specific genes such as cytochrome c during the late stage of differentiation. Furthermore, the nuclear translocation of AMPKα2 is necessary for maintenance of PGC-1α1 mRNA during myogenesis.
Collapse
Affiliation(s)
- Shiki Okamoto
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan; Second Department of Internal Medicine (Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology), Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Nur Farehan Asgar
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan
| | - Shigefumi Yokota
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan
| | - Kumiko Saito
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan
| | - Yasuhiko Minokoshi
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan.
| |
Collapse
|
173
|
Coculture Method to Obtain Endothelial Networks Within Human Tissue-Engineered Skeletal Muscle. Methods Mol Biol 2019; 1889:169-183. [PMID: 30367414 DOI: 10.1007/978-1-4939-8897-6_10] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Skeletal muscle tissue engineering aims at creating functional skeletal muscle in vitro. Human muscle organoids can be used for potential applications in regenerative medicine, but also as an in vitro model for myogenesis or myopathology. However, the thickness of constructs is limited due to passive diffusion of nutrients and oxygen. Introduction of a vascular network in vitro may solve this limitation. Here, we describe tissue engineering of in vitro skeletal muscle consisting of human aligned myofibers with interspersed endothelial networks. To create bio-artificial muscle (BAM), human muscle progenitor cells are cocultured with human umbilical vein endothelial cells (HUVECs) in a fibrin hydrogel. The cell-gel mix is cast into silicone molds with end attachment sites and cultured in endothelial growth medium (EGM-2) for 1 week. The passive forces generated in the contracted hydrogel align the myogenic cells parallel to the long axis of the contracted gel such that they fuse into aligned multinucleated myofibers. This results in the formation of a 2 cm long and ~1.5 mm tick human BAM construct with endothelial networks.
Collapse
|
174
|
Ono S, Yoshida N, Maekawa D, Kitakaze T, Kobayashi Y, Kitano T, Fujita T, Okuwa‐Hayashi H, Harada N, Nakano Y, Yamaji R. 5-Hydroxy-7-methoxyflavone derivatives from Kaempferia parviflora induce skeletal muscle hypertrophy. Food Sci Nutr 2019; 7:312-321. [PMID: 30680186 PMCID: PMC6341173 DOI: 10.1002/fsn3.891] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/29/2018] [Accepted: 11/03/2018] [Indexed: 12/24/2022] Open
Abstract
Skeletal muscle plays a critical role in locomotion and energy metabolism. Maintenance or enhancement of skeletal muscle mass contributes to the improvement of mobility and prevents the development of metabolic diseases. The extracts from Kaempferia parviflora rhizomes contain at least ten methoxyflavone derivatives that exhibit enhancing effects on ATP production and glucose uptake in skeletal muscle cells. In the present study, we investigated the effects of ten K. parviflora-derived methoxyflavone derivatives (six 5,7-dimethoxyflavone (DMF) derivatives and four 5-hydroxy-7-methoxyflavone (HMF) derivatives) on skeletal muscle hypertrophy. Murine C2C12 myotubes and senescence-accelerated mouse-prone 1 (SAMP1) mice treated with methoxyflavones were used as experimental models to determine the effects of HMF derivatives on myotube diameter and size and muscle mass. The four HMF derivatives, but not the six DMF derivatives, increased myotube diameter. The 5-hydroxyflavone, 7-methoxyflavone, and 5,7-dihydroxyflavone had no influence on myotube size, a result that differed from HMF. Dietary administration of the mixture composed of the four HMF derivatives resulted in increase in the soleus muscle size and mass in SAMP1 mice. HMF derivatives also promoted protein synthesis in myotubes, and treatment with the intracellular Ca2+ chelator BAPTA-AM, which depletes intracellular Ca2+ levels, inhibited this promotion. Furthermore, BAPTA-AM inhibited HMF-promoted protein synthesis even when myotubes were incubated in Ca2+-free medium. These results indicate that HMF derivatives induce myotube hypertrophy and that both the 5-hydroxyl group and the 7-methoxy group in the flavones are necessary for myotube hypertrophy. Furthermore, these results suggest that HMF-induced protein synthesis requires intracellular Ca2+, but not extracellular Ca2+.
Collapse
Affiliation(s)
- Shintaro Ono
- Division of Applied Life SciencesGraduate School of Life and Environmental SciencesOsaka Prefecture UniversitySakaiOsakaJapan
| | - Naoki Yoshida
- Division of Applied Life SciencesGraduate School of Life and Environmental SciencesOsaka Prefecture UniversitySakaiOsakaJapan
| | - Daisuke Maekawa
- Division of Applied Life SciencesGraduate School of Life and Environmental SciencesOsaka Prefecture UniversitySakaiOsakaJapan
| | - Tomoya Kitakaze
- Division of Applied Life SciencesGraduate School of Life and Environmental SciencesOsaka Prefecture UniversitySakaiOsakaJapan
| | - Yasuyuki Kobayashi
- Division of Applied Life SciencesGraduate School of Life and Environmental SciencesOsaka Prefecture UniversitySakaiOsakaJapan
| | - Takehiro Kitano
- Division of Applied Life SciencesGraduate School of Life and Environmental SciencesOsaka Prefecture UniversitySakaiOsakaJapan
| | | | - Hirotaka Okuwa‐Hayashi
- Japan Tablet CorporationUjiKyotoJapan
- Center for Research and Development of BioresourcesOsaka Prefecture UniversitySakaiOsakaJapan
| | - Naoki Harada
- Division of Applied Life SciencesGraduate School of Life and Environmental SciencesOsaka Prefecture UniversitySakaiOsakaJapan
| | - Yoshihisa Nakano
- Center for Research and Development of BioresourcesOsaka Prefecture UniversitySakaiOsakaJapan
| | - Ryoichi Yamaji
- Division of Applied Life SciencesGraduate School of Life and Environmental SciencesOsaka Prefecture UniversitySakaiOsakaJapan
| |
Collapse
|
175
|
Zhang Z, Zhao LD, Johnson SE, Rhoads ML, Jiang H, Rhoads RP. Oxytocin is involved in steroid hormone-stimulated bovine satellite cell proliferation and differentiation in vitro. Domest Anim Endocrinol 2019; 66:1-13. [PMID: 30195176 DOI: 10.1016/j.domaniend.2018.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 07/12/2018] [Accepted: 07/28/2018] [Indexed: 11/17/2022]
Abstract
Sex steroid hormones are used in the meat industry due to their ability to regulate muscle hypertrophy. However, the mechanisms underlying their action are not fully elucidated. Recent reports demonstrate that steroid hormones increase oxytocin (OXT) expression in skeletal muscle, indicating that OXT may play a role in satellite cell activity. This hypothesis was tested using steroid hormones (17β-estradiol [E2]; trenbolone acetate [TBA]), tamoxifen (TAM), OXT, and atosiban (A: OXT receptor inhibitor) applied to bovine satellite cells (BSCs) to investigate BSC regulation by OXT. Oxytocin alone increased fusion index (P < 0.05) but not BSC proliferation. Oxytocin reduced (P < 0.05) apoptotic nuclei and stimulated migration rate (P < 0.05). Similarly, E2 and TBA increased (P < 0.05) BSC proliferation rate, fusion index, and migration and decreased (P < 0.05) apoptotic nuclei. 17β-Estradiol or TBA supplemented with A had lower (P < 0.05) BSC proliferation rate, fusion index, and migration and more (P < 0.05) apoptotic nuclei compared with E2 or TBA alone. Furthermore, OXT expression increased (P < 0.05) in E2 or TBA-treated proliferating BSC. Oxytocin, E2, and TBA increased (P < 0.05) MyoD and MyoG expression in proliferating BSC. During BSC differentiation, OXT expression increased (P < 0.05) with E2 or TBA treatments. MyoG expression increased (P < 0.05) in OXT, E2, and TBA compared with control. However, A, OXT + A, TAM, TAM + OXT, E2 + TAM, E2 + A, and TBA + A decreased (P < 0.05) MyoG expression during BSC differentiation. These results indicate that OXT is involved in steroid hormone-stimulated BSC activity.
Collapse
Affiliation(s)
- Zhenhe Zhang
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Lidan D Zhao
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Sally E Johnson
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Michelle L Rhoads
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Honglin Jiang
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Robert P Rhoads
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
176
|
Vorontsova JE, Zavoloka EL, Cherezov RO, Simonova OB. Three Important Discoveries in the Field of the Cytoskeleton’s Proteins Functioning on the Drosophila melanogaster Model. Mol Biol 2019. [DOI: 10.1134/s0026893319010163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
177
|
Hall AE, Rose MD. Cell fusion in yeast is negatively regulated by components of the cell wall integrity pathway. Mol Biol Cell 2018; 30:441-452. [PMID: 30586320 PMCID: PMC6594448 DOI: 10.1091/mbc.e18-04-0236] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
During mating, Saccharomyces cerevisiae cells must degrade the intervening cell wall to allow fusion of the partners. Because improper timing or location of cell wall degradation would cause lysis, the initiation of cell fusion must be highly regulated. Here, we find that yeast cell fusion is negatively regulated by components of the cell wall integrity (CWI) pathway. Loss of the cell wall sensor, MID2, specifically causes “mating-induced death” after pheromone exposure. Mating-induced death is suppressed by mutations in cell fusion genes (FUS1, FUS2, RVS161, CDC42), implying that mid2Δ cells die from premature fusion without a partner. Consistent with premature fusion, mid2Δ shmoos had thinner cell walls and lysed at the shmoo tip. Normally, Cdc42p colocalizes with Fus2p to form a focus only when mating cells are in contact (prezygotes) and colocalization is required for cell fusion. However, Cdc42p was aberrantly colocalized with Fus2p to form a focus in mid2Δ shmoos. A hyperactive allele of the CWI kinase Pkc1p (PKC1*) caused decreased cell fusion and Cdc42p localization in prezygotes. In shmoos, PKC1* increased Cdc42p localization; however, it was not colocalized with Fus2p or associated with cell death. We conclude that Mid2p and Pkc1p negatively regulate cell fusion via Cdc42p and Fus2p.
Collapse
Affiliation(s)
- Allison E Hall
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Mark D Rose
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544.,Department of Biology, Georgetown University, Washington, DC 20057
| |
Collapse
|
178
|
Hsp70 Interacts with Mitogen-Activated Protein Kinase (MAPK)-Activated Protein Kinase 2 To Regulate p38MAPK Stability and Myoblast Differentiation during Skeletal Muscle Regeneration. Mol Cell Biol 2018; 38:MCB.00211-18. [PMID: 30275345 DOI: 10.1128/mcb.00211-18] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/26/2018] [Indexed: 12/24/2022] Open
Abstract
The regenerative process of injured muscle is dependent on the fusion and differentiation of myoblasts derived from muscle stem cells. Hsp70 is important for maintaining skeletal muscle homeostasis and regeneration, but the precise cellular mechanism remains elusive. In this study, we found that Hsp70 was upregulated during myoblast differentiation. Depletion or inhibition of Hsp70/Hsc70 impaired myoblast differentiation. Importantly, overexpression of p38 mitogen-activated protein kinase α (p38MAPKα) but not AKT1 rescued the impairment of myogenic differentiation in Hsp70- or Hsc70-depleted myoblasts. Moreover, Hsp70 interacted with MK2, a substrate of p38MAPK, to regulate the stability of p38MAPK. Knockdown of Hsp70 also led to downregulation of both MK2 and p38MAPK in intact muscles and during cardiotoxin-induced muscle regeneration. Hsp70 bound MK2 to regulate MK2-p38MAPK interaction in myoblasts. We subsequently identified the essential regions required for Hsp70-MK2 interaction. Functional analyses showed that MK2 is essential for both myoblast differentiation and skeletal muscle regeneration. Taken together, our findings reveal a novel role of Hsp70 in regulating myoblast differentiation by interacting with MK2 to stabilize p38MAPK.
Collapse
|
179
|
Castiglioni I, Caccia R, Garcia-Manteiga JM, Ferri G, Caretti G, Molineris I, Nishioka K, Gabellini D. The Trithorax protein Ash1L promotes myoblast fusion by activating Cdon expression. Nat Commun 2018; 9:5026. [PMID: 30487570 PMCID: PMC6262021 DOI: 10.1038/s41467-018-07313-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 10/24/2018] [Indexed: 12/17/2022] Open
Abstract
Myoblast fusion (MF) is required for muscle growth and repair, and its alteration contributes to muscle diseases. The mechanisms governing this process are incompletely understood, and no epigenetic regulator has been previously described. Ash1L is an epigenetic activator belonging to the Trithorax group of proteins and is involved in FSHD muscular dystrophy, autism and cancer. Its physiological role in skeletal muscle is unknown. Here we report that Ash1L expression is positively correlated with MF and reduced in Duchenne muscular dystrophy. In vivo, ex vivo and in vitro experiments support a selective and evolutionary conserved requirement for Ash1L in MF. RNA- and ChIP-sequencing indicate that Ash1L is required to counteract Polycomb repressive activity to allow activation of selected myogenesis genes, in particular the key MF gene Cdon. Our results promote Ash1L as an important epigenetic regulator of MF and suggest that its activity could be targeted to improve cell therapy for muscle diseases. Myoblast fusion in skeletal muscle is a complex process but how this is regulated is unclear. Here, the authors identify Ash1L, a histone methyltransferase, as modulating myoblast fusion via activation of the myogenesis gene Cdon, and observe decreased Ash1L expression in Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Ilaria Castiglioni
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, 20132, Italy
| | - Roberta Caccia
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, 20132, Italy
| | - Jose Manuel Garcia-Manteiga
- Center for Translational Genomics and BioInformatics, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, 20132, Italy
| | - Giulia Ferri
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, 20132, Italy
| | - Giuseppina Caretti
- Department of Biosciences, University of Milan, via Celoria 26, Milano, 20133, Italy
| | - Ivan Molineris
- Center for Translational Genomics and BioInformatics, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, 20132, Italy
| | - Kenichi Nishioka
- Department of Biomolecular Sciences, Division of Molecular Genetics and Epigenetics, Faculty of Medicine, Saga University, Saga, Japan.,Laboratory for Developmental Genetics, RIKEN IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa, 230-0045, Japan
| | - Davide Gabellini
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, 20132, Italy.
| |
Collapse
|
180
|
Isobe M, Lee S, Waguri S, Kametaka S. Clathrin adaptor GGA1 modulates myogenesis of C2C12 myoblasts. PLoS One 2018; 13:e0207533. [PMID: 30440034 PMCID: PMC6237421 DOI: 10.1371/journal.pone.0207533] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 11/01/2018] [Indexed: 12/23/2022] Open
Abstract
During myogenesis, myogenic stem cells undergo several sequential events, including cell division, migration, and cell-cell fusion, leading to the formation of multinuclear myotubes, which are the precursors of myofibers. To understand the molecular mechanisms underlying these complex processes, an RNA interference-based gene depletion approach was used. Golgi associated, gamma adaptin ear containing, ARF binding protein 1 (GGA1), a Golgi-resident monomeric clathrin adaptor, was found to be required for the process of myotube formation in C2C12 cells, a cultured murine myoblast cell line. Gga1 mRNA expression was upregulated during myogenesis, and Gga1 depletion prevented the formation of large multi-nucleated myotubes. Moreover, inhibition of lysosomal proteases in Gga1 knockdown myoblasts increased the amount of insulin receptor, suggesting that GGA1 is involved in the cell surface expression and sorting of the insulin receptor. These results suggested that GGA1 plays a significant role in the formation and maturation of myotubes by targeting the insulin receptor to the cell surface to establish functionally mature myofibers.
Collapse
Affiliation(s)
- Mari Isobe
- Department of Physical and Occupational Therapy, Graduate School of Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Sachiko Lee
- Department of Physical and Occupational Therapy, Graduate School of Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Satoshi Waguri
- Department of Anatomy and Histology, Fukushima Medical University, Fukushima, Fukushima, Japan
| | - Satoshi Kametaka
- Department of Physical and Occupational Therapy, Graduate School of Medicine, Nagoya University, Nagoya, Aichi, Japan
- * E-mail:
| |
Collapse
|
181
|
Chen X, Ouyang H, Wang Z, Chen B, Nie Q. A Novel Circular RNA Generated by FGFR2 Gene Promotes Myoblast Proliferation and Differentiation by Sponging miR-133a-5p and miR-29b-1-5p. Cells 2018; 7:cells7110199. [PMID: 30404220 PMCID: PMC6262629 DOI: 10.3390/cells7110199] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 10/26/2018] [Accepted: 11/02/2018] [Indexed: 12/19/2022] Open
Abstract
It is well known that fibroblast growth factor receptor 2 (FGFR2) interacts with its ligand of fibroblast growth factor (FGF) therefore exerting biological functions on cell proliferation and differentiation. In this study, we first reported that the FGFR2 gene could generate a circular RNA of circFGFR2, which regulates skeletal muscle development by sponging miRNA. In our previous study of circular RNA sequencing, we found that circFGFR2, generated by exon 3–6 of FGFR2 gene, differentially expressed during chicken embryo skeletal muscle development. The purpose of this study was to reveal the real mechanism of how circFGFR2 affects skeletal muscle development in chicken. In this study, cell proliferation was analyzed by both flow cytometry analysis of the cell cycle and 5-ethynyl-2′-deoxyuridine (EdU) assays. Cell differentiation was determined by analysis of the expression of the differentiation marker gene and Myosin heavy chain (MyHC) immunofluorescence. The results of flow cytometry analysis of the cell cycle and EdU assays showed that, overexpression of circFGFR2 accelerated the proliferation of myoblast and QM-7 cells, whereas knockdown of circFGFR2 with siRNA reduced the proliferation of both cells. Meanwhile, overexpression of circFGFR2 accelerated the expression of myogenic differentiation 1 (MYOD), myogenin (MYOG) and the formation of myotubes, and knockdown of circFGFR2 showed contrary effects in myoblasts. Results of luciferase reporter assay and biotin-coupled miRNA pull down assay further showed that circFGFR2 could directly target two binding sites of miR-133a-5p and one binding site of miR-29b-1-5p, and further inhibited the expression and activity of these two miRNAs. In addition, we demonstrated that both miR-133a-5p and miR-29b-1-5p inhibited myoblast proliferation and differentiation, while circFGFR2 could eliminate the inhibition effects of the two miRNAs as indicated by rescue experiments. Altogether, our data revealed that a novel circular RNA of circFGFR2 could promote skeletal muscle proliferation and differentiation by sponging miR-133a-5p and miR-29b-1-5p.
Collapse
Affiliation(s)
- Xiaolan Chen
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- National-Local Joint Engineering Research Center for Livestock Breeding, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and the Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China.
| | - Hongjia Ouyang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China.
| | - Zhijun Wang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- National-Local Joint Engineering Research Center for Livestock Breeding, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and the Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China.
| | - Biao Chen
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- National-Local Joint Engineering Research Center for Livestock Breeding, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and the Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China.
| | - Qinghua Nie
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- National-Local Joint Engineering Research Center for Livestock Breeding, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and the Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China.
| |
Collapse
|
182
|
Hamoud N, Tran V, Aimi T, Kakegawa W, Lahaie S, Thibault MP, Pelletier A, Wong GW, Kim IS, Kania A, Yuzaki M, Bouvier M, Côté JF. Spatiotemporal regulation of the GPCR activity of BAI3 by C1qL4 and Stabilin-2 controls myoblast fusion. Nat Commun 2018; 9:4470. [PMID: 30367035 PMCID: PMC6203814 DOI: 10.1038/s41467-018-06897-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 10/05/2018] [Indexed: 11/09/2022] Open
Abstract
Myoblast fusion is tightly regulated during development and regeneration of muscle fibers. BAI3 is a receptor that orchestrates myoblast fusion via Elmo/Dock1 signaling, but the mechanisms regulating its activity remain elusive. Here we report that mice lacking BAI3 display small muscle fibers and inefficient muscle regeneration after cardiotoxin-induced injury. We describe two proteins that repress or activate BAI3 in muscle progenitors. We find that the secreted C1q-like1-4 proteins repress fusion by specifically interacting with BAI3. Using a proteomic approach, we identify Stabilin-2 as a protein that interacts with BAI3 and stimulates its fusion promoting activity. We demonstrate that Stabilin-2 activates the GPCR activity of BAI3. The resulting activated heterotrimeric G-proteins contribute to the initial recruitment of Elmo proteins to the membrane, which are then stabilized on BAI3 through a direct interaction. Collectively, our results demonstrate that the activity of BAI3 is spatiotemporally regulated by C1qL4 and Stabilin-2 during myoblast fusion.
Collapse
Affiliation(s)
- Noumeira Hamoud
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, H2W 1R7, Canada.,Département de Médecine (Programmes de Biologie Moléculaire), Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Viviane Tran
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, H2W 1R7, Canada.,Département de Biochimie, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Takahiro Aimi
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JT), Tokyo, 102-0075, Japan
| | - Wataru Kakegawa
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JT), Tokyo, 102-0075, Japan
| | - Sylvie Lahaie
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, H2W 1R7, Canada.,Integrated Program in Neuroscience, McGill University, Montréal, QC, H3A 2B4, Canada
| | - Marie-Pier Thibault
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, H2W 1R7, Canada
| | - Ariane Pelletier
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, H2W 1R7, Canada
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.,Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - In-San Kim
- Biomedical Research Institute, Korea Institute Science and Technology, Seoul, 136-791, Republic of Korea.,KU-KIST school, Korea University, Seoul, 136-701, Republic of Korea
| | - Artur Kania
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, H2W 1R7, Canada.,Integrated Program in Neuroscience, McGill University, Montréal, QC, H3A 2B4, Canada.,Department of Anatomy and Cell Biology, McGill University, Montréal, QC, H3A 1A3, Canada
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JT), Tokyo, 102-0075, Japan
| | - Michel Bouvier
- Département de Biochimie, Université de Montréal, Montréal, QC, H3T 1J4, Canada.,Institut de Recherches en Immunologie et Cancérologie (IRIC), Université de Montréal, Montréal, QC, Canada, H3C 3J7
| | - Jean-François Côté
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, H2W 1R7, Canada. .,Département de Médecine (Programmes de Biologie Moléculaire), Université de Montréal, Montréal, QC, H3T 1J4, Canada. .,Département de Biochimie, Université de Montréal, Montréal, QC, H3T 1J4, Canada. .,Department of Anatomy and Cell Biology, McGill University, Montréal, QC, H3A 1A3, Canada.
| |
Collapse
|
183
|
Woo J, Kim HJ, Nam YR, Kim YK, Lee EJ, Choi I, Kim SJ, Lee W, Nam JH. Mitochondrial dysfunction reduces the activity of KIR2.1 K + channel in myoblasts via impaired oxidative phosphorylation. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2018; 22:697-703. [PMID: 30402030 PMCID: PMC6205933 DOI: 10.4196/kjpp.2018.22.6.697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 11/15/2022]
Abstract
Myoblast fusion depends on mitochondrial integrity and intracellular Ca2+ signaling regulated by various ion channels. In this study, we investigated the ionic currents associated with [Ca2+]i regulation in normal and mitochondrial DNA-depleted (ρ0) L6 myoblasts. The ρ0 myoblasts showed impaired myotube formation. The inwardly rectifying K+ current (IKir) was largely decreased with reduced expression of KIR2.1, whereas the voltage-operated Ca2+ channel and Ca2+-activated K+ channel currents were intact. Sustained inhibition of mitochondrial electron transport by antimycin A treatment (24 h) also decreased the IKir. The ρ0 myoblasts showed depolarized resting membrane potential and higher basal [Ca2+]i. Our results demonstrated the specific downregulation of IKir by dysfunctional mitochondria. The resultant depolarization and altered Ca2+ signaling might be associated with impaired myoblast fusion in ρ0 myoblasts.
Collapse
Affiliation(s)
- JooHan Woo
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hyun Jong Kim
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Korea.,Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Korea
| | - Yu Ran Nam
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Korea.,Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Korea
| | - Yung Kyu Kim
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea
| | - Sung Joon Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Wan Lee
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Korea.,Department of Biochemistry, Dongguk University College of Medicine, Gyeongju 38066, Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Korea.,Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Korea
| |
Collapse
|
184
|
Myogenin promotes myocyte fusion to balance fibre number and size. Nat Commun 2018; 9:4232. [PMID: 30315160 PMCID: PMC6185967 DOI: 10.1038/s41467-018-06583-6] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 08/31/2018] [Indexed: 01/01/2023] Open
Abstract
Each skeletal muscle acquires its unique size before birth, when terminally differentiating myocytes fuse to form a defined number of multinucleated myofibres. Although mice in which the transcription factor Myogenin is mutated lack most myogenesis and die perinatally, a specific cell biological role for Myogenin has remained elusive. Here we report that loss of function of zebrafish myog prevents formation of almost all multinucleated muscle fibres. A second, Myogenin-independent, fusion pathway in the deep myotome requires Hedgehog signalling. Lack of Myogenin does not prevent terminal differentiation; the smaller myotome has a normal number of myocytes forming more mononuclear, thin, albeit functional, fast muscle fibres. Mechanistically, Myogenin binds to the myomaker promoter and is required for expression of myomaker and other genes essential for myocyte fusion. Adult myog mutants display reduced muscle mass, decreased fibre size and nucleation. Adult-derived myog mutant myocytes show persistent defective fusion ex vivo. Myogenin is therefore essential for muscle homeostasis, regulating myocyte fusion to determine both muscle fibre number and size. Loss of the transcription factor Myogenin in mice reduces skeletal myogenesis and leads to perinatal death but how Myogenin regulates muscle formation is unclear. Here, the authors show that zebrafish Myogenin enhances Myomaker expression, muscle cell fusion and myotome size, yet decreases fast muscle fibre number.
Collapse
|
185
|
Chick embryonic cells as a source for generating in vitro model of muscle cell dystrophy. In Vitro Cell Dev Biol Anim 2018; 54:756-769. [DOI: 10.1007/s11626-018-0297-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 09/19/2018] [Indexed: 12/31/2022]
|
186
|
Whitlock JM, Yu K, Cui YY, Hartzell HC. Anoctamin 5/TMEM16E facilitates muscle precursor cell fusion. J Gen Physiol 2018; 150:1498-1509. [PMID: 30257928 PMCID: PMC6219693 DOI: 10.1085/jgp.201812097] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/12/2018] [Accepted: 09/10/2018] [Indexed: 12/19/2022] Open
Abstract
Limb-girdle muscular dystrophy type 2L arises from mutations in the anoctamin ANO5, whose role in muscle physiology is unknown. Whitlock et al. show that loss of ANO5 perturbs phosphatidylserine exposure and cell–cell fusion in muscle precursor cells, which is an essential step in muscle repair. Limb-girdle muscular dystrophy type 2L (LGMD2L) is a myopathy arising from mutations in ANO5; however, information about the contribution of ANO5 to muscle physiology is lacking. To explain the role of ANO5 in LGMD2L, we previously hypothesized that ANO5-mediated phospholipid scrambling facilitates cell–cell fusion of mononucleated muscle progenitor cells (MPCs), which is required for muscle repair. Here, we show that heterologous overexpression of ANO5 confers Ca2+-dependent phospholipid scrambling to HEK-293 cells and that scrambling is associated with the simultaneous development of a nonselective ionic current. MPCs isolated from adult Ano5−/− mice exhibit defective cell fusion in culture and produce muscle fibers with significantly fewer nuclei compared with controls. This defective fusion is associated with a decrease of Ca2+-dependent phosphatidylserine exposure on the surface of Ano5−/− MPCs and a decrease in the amplitude of Ca2+-dependent outwardly rectifying ionic currents. Viral introduction of ANO5 in Ano5−/− MPCs restores MPC fusion competence, ANO5-dependent phospholipid scrambling, and Ca2+-dependent outwardly rectifying ionic currents. ANO5-rescued MPCs produce myotubes having numbers of nuclei similar to wild-type controls. These data suggest that ANO5-mediated phospholipid scrambling or ionic currents play an important role in muscle repair.
Collapse
Affiliation(s)
- Jarred M Whitlock
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - Kuai Yu
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - Yuan Yuan Cui
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - H Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
187
|
Valer FB, Machado MCR, Silva-Junior RMP, Ramos RGP. Expression of Hbs, Kirre, and Rst during Drosophila ovarian development. Genesis 2018; 56:e23242. [PMID: 30114331 DOI: 10.1002/dvg.23242] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 07/22/2018] [Accepted: 07/23/2018] [Indexed: 12/16/2022]
Abstract
The Irre cell-recognition module (IRM) is a group of evolutionarily conserved and structurally related transmembrane glycoproteins of the immunoglobulin superfamily. In Drosophila melanogaster, it comprises the products of the genes roughest (rst; also known as irreC-rst), kin-of-irre (kirre; also known as duf), sticks-and-stones (sns), and hibris (hbs). In this model organism, the behavior of this group of proteins as a partly redundant functional unit mediating selective cell recognition was demonstrated in a variety of developmental contexts, but their possible involvement in ovarian development and oogenesis has not been investigated, notwithstanding the fact that some rst mutant alleles are also female sterile. Here, we show that IRM genes are dynamically and, to some extent, coordinately transcribed in both pupal and adult ovaries. Additionally, the spatial distribution of Hbs, Kirre, and Rst proteins indicates that they perform cooperative, although largely nonredundant, functions. Finally, phenotypical characterization of three different female sterile rst alleles uncovered two temporally separated and functionally distinct requirements for this locus in ovarian development: one in pupa, essential for the organization of peritoneal and epithelial sheaths that maintain the structural integrity of the adult organ and another, in mature ovarioles, needed for the progression of oogenesis beyond stage 10.
Collapse
Affiliation(s)
- Felipe Berti Valer
- Department of Cellular and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maiaro Cabral Rosa Machado
- Department of Cellular and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | | |
Collapse
|
188
|
Islam R, Yoon H, Shin HR, Bae HS, Kim BS, Yoon WJ, Woo KM, Baek JH, Lee YS, Ryoo HM. Peptidyl-prolyl cis-trans isomerase NIMA interacting 1 regulates skeletal muscle fusion through structural modification of Smad3 in the linker region. J Cell Physiol 2018; 233:9390-9403. [PMID: 30132832 PMCID: PMC6686165 DOI: 10.1002/jcp.26774] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 04/27/2018] [Indexed: 02/07/2023]
Abstract
Myoblast fusion is critical for muscle growth, regeneration, and repair. We previously reported that the enzyme peptidyl‐prolyl cis–trans isomerase NIMA interacting 1 (Pin1) is involved in osteoclast fusion. The objective of this study was to investigate the possibility that Pin1 also inhibits myoblast fusion. Here, we show the increased number of nuclei in the Pin1+/− mice muscle fiber compared to that in wild‐type mice. Moreover, we show that low dose of the Pin1 inhibitor dipentamethylene thiuram monosulfide treatment caused enhanced fusion in C2C12 cells. The R‐Smads are well‐known mediators of muscle hypertrophy and hyperplasia as well as being substrates of Pin1. We found that Pin1 is crucial for maintaining the stability of Smad3 (homologues of the Drosophila protein, mothers against decapentaplegic (Mad) and the Caenorhabditis elegans protein Sma). Our results show that serine 204 within Smad3 is the key Pin1‐binding site during inhibition of myoblast fusion and that both the transforming growth factor‐β receptor and extracellular signal‐regulated kinase (ERK)‐mediated phosphorylation are required for the interaction of Pin1 with Smad3. These findings suggest that a precise level of Pin1 activity is essential for regulating myoblast fusion during myogenesis and muscle regeneration.
Collapse
Affiliation(s)
- Rabia Islam
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Heein Yoon
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Hye-Rim Shin
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Han-Sol Bae
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Bong-Soo Kim
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Won-Joon Yoon
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Kyung-Mi Woo
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Jeong-Hwa Baek
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Yun-Sil Lee
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Hyun-Mo Ryoo
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
189
|
McClure MJ, Cohen DJ, Ramey AN, Bivens CB, Mallu S, Isaacs JE, Imming E, Huang YC, Sunwoo M, Schwartz Z, Boyan BD. Decellularized Muscle Supports New Muscle Fibers and Improves Function Following Volumetric Injury. Tissue Eng Part A 2018; 24:1228-1241. [DOI: 10.1089/ten.tea.2017.0386] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Michael J. McClure
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - David J. Cohen
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Allison N. Ramey
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Caroline B. Bivens
- Department of School of Art, Virginia Commonwealth University, Richmond, Virginia
| | - Satya Mallu
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Jonathan E. Isaacs
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Emily Imming
- MTF Biologics, Musculoskeletal Transplant Foundation, Edison, New Jersey
| | - Yen-Chen Huang
- MTF Biologics, Musculoskeletal Transplant Foundation, Edison, New Jersey
| | - MoonHae Sunwoo
- MTF Biologics, Musculoskeletal Transplant Foundation, Edison, New Jersey
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Barbara D. Boyan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
190
|
PPARγ Controls Ectopic Adipogenesis and Cross-Talks with Myogenesis During Skeletal Muscle Regeneration. Int J Mol Sci 2018; 19:ijms19072044. [PMID: 30011852 PMCID: PMC6073847 DOI: 10.3390/ijms19072044] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/06/2018] [Accepted: 07/09/2018] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscle is a regenerative tissue which can repair damaged myofibers through the activation of tissue-resident muscle stem cells (MuSCs). Many muscle diseases with impaired regeneration cause excessive adipose tissue accumulation in muscle, alter the myogenic fate of MuSCs, and deregulate the cross-talk between MuSCs and fibro/adipogenic progenitors (FAPs), a bi-potent cell population which supports myogenesis and controls intra-muscular fibrosis and adipocyte formation. In order to better characterize the interaction between adipogenesis and myogenesis, we studied muscle regeneration and MuSC function in whole body Pparg null mice generated by epiblast-specific Cre/lox deletion (PpargΔ/Δ). We demonstrate that deletion of PPARγ completely abolishes ectopic muscle adipogenesis during regeneration and impairs MuSC expansion and myogenesis after injury. Ex vivo assays revealed that perturbed myogenesis in PpargΔ/Δ mice does not primarily result from intrinsic defects of MuSCs or from perturbed myogenic support from FAPs. The immune transition from a pro- to anti-inflammatory MuSC niche during regeneration is perturbed in PpargΔ/Δ mice and suggests that PPARγ signaling in macrophages can interact with ectopic adipogenesis and influence muscle regeneration. Altogether, our study demonstrates that a PPARγ-dependent adipogenic response regulates muscle fat infiltration during regeneration and that PPARγ is required for MuSC function and efficient muscle repair.
Collapse
|
191
|
Drosophila pericardial nephrocyte ultrastructure changes during ageing. Mech Ageing Dev 2018; 173:9-20. [DOI: 10.1016/j.mad.2018.04.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 04/17/2018] [Accepted: 04/23/2018] [Indexed: 10/17/2022]
|
192
|
Tsutsumi R, Tran MP, Cooper KL. Changing While Staying the Same: Preservation of Structural Continuity During Limb Evolution by Developmental Integration. Integr Comp Biol 2018; 57:1269-1280. [PMID: 28992070 DOI: 10.1093/icb/icx092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
More than 150 years since Charles Darwin published "On the Origin of Species", gradual evolution by natural selection is still not fully reconciled with the apparent sudden appearance of complex structures, such as the bat wing, with highly derived functions. This is in part because developmental genetics has not yet identified the number and types of mutations that accumulated to drive complex morphological evolution. Here, we consider the experimental manipulations in laboratory model systems that suggest tissue interdependence and mechanical responsiveness during limb development conceptually reduce the genetic complexity required to reshape the structure as a whole. It is an exciting time in the field of evolutionary developmental biology as emerging technical approaches in a variety of non-traditional laboratory species are on the verge of filling the gaps between theory and evidence to resolve this sesquicentennial debate.
Collapse
Affiliation(s)
- Rio Tsutsumi
- Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093-0380, USA
| | - Mai P Tran
- Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093-0380, USA
| | - Kimberly L Cooper
- Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093-0380, USA
| |
Collapse
|
193
|
Duan R, Kim JH, Shilagardi K, Schiffhauer ES, Lee DM, Son S, Li S, Thomas C, Luo T, Fletcher DA, Robinson DN, Chen EH. Spectrin is a mechanoresponsive protein shaping fusogenic synapse architecture during myoblast fusion. Nat Cell Biol 2018; 20:688-698. [PMID: 29802406 PMCID: PMC6397639 DOI: 10.1038/s41556-018-0106-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 04/18/2018] [Indexed: 12/24/2022]
Abstract
Spectrin is a membrane skeletal protein best known for its structural role in maintaining cell shape and protecting cells from mechanical damage. Here, we report that α/βH-spectrin (βH is also called karst) dynamically accumulates and dissolves at the fusogenic synapse between fusing Drosophila muscle cells, where an attacking fusion partner invades its receiving partner with actin-propelled protrusions to promote cell fusion. Using genetics, cell biology, biophysics and mathematical modelling, we demonstrate that spectrin exhibits a mechanosensitive accumulation in response to shear deformation, which is highly elevated at the fusogenic synapse. The transiently accumulated spectrin network functions as a cellular fence to restrict the diffusion of cell-adhesion molecules and a cellular sieve to constrict the invasive protrusions, thereby increasing the mechanical tension of the fusogenic synapse to promote cell membrane fusion. Our study reveals a function of spectrin as a mechanoresponsive protein and has general implications for understanding spectrin function in dynamic cellular processes.
Collapse
Affiliation(s)
- Rui Duan
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Laboratory of Regenerative Medicine in Sports Science, School of Sports Science, South China Normal University, Guangzhou, China
| | - Ji Hoon Kim
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Khurts Shilagardi
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eric S Schiffhauer
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Donghoon M Lee
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sungmin Son
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | - Shuo Li
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Claire Thomas
- Departments of Biology and of Biochemistry and Molecular Biology, Penn State University, University Park, PA, USA
| | - Tianzhi Luo
- Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Daniel A Fletcher
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | - Douglas N Robinson
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth H Chen
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
194
|
Tsuchiya M, Hara Y, Okuda M, Itoh K, Nishioka R, Shiomi A, Nagao K, Mori M, Mori Y, Ikenouchi J, Suzuki R, Tanaka M, Ohwada T, Aoki J, Kanagawa M, Toda T, Nagata Y, Matsuda R, Takayama Y, Tominaga M, Umeda M. Cell surface flip-flop of phosphatidylserine is critical for PIEZO1-mediated myotube formation. Nat Commun 2018; 9:2049. [PMID: 29799007 PMCID: PMC5967302 DOI: 10.1038/s41467-018-04436-w] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/26/2018] [Indexed: 11/23/2022] Open
Abstract
Myotube formation by fusion of myoblasts and subsequent elongation of the syncytia is essential for skeletal muscle formation. However, molecules that regulate myotube formation remain elusive. Here we identify PIEZO1, a mechanosensitive Ca2+ channel, as a key regulator of myotube formation. During myotube formation, phosphatidylserine, a phospholipid that resides in the inner leaflet of the plasma membrane, is transiently exposed to cell surface and promotes myoblast fusion. We show that cell surface phosphatidylserine inhibits PIEZO1 and that the inward translocation of phosphatidylserine, which is driven by the phospholipid flippase complex of ATP11A and CDC50A, is required for PIEZO1 activation. PIEZO1-mediated Ca2+ influx promotes RhoA/ROCK-mediated actomyosin assemblies at the lateral cortex of myotubes, thus preventing uncontrolled fusion of myotubes and leading to polarized elongation during myotube formation. These results suggest that cell surface flip-flop of phosphatidylserine acts as a molecular switch for PIEZO1 activation that governs proper morphogenesis during myotube formation. Myotube formation by fusion of myoblasts is essential for skeletal muscle formation, but which molecules regulate this process remains elusive. Here authors identify the mechanosensitive PIEZO1 channel as a key element, whose activity is regulated by phosphatidylserine during myotube formation.
Collapse
Affiliation(s)
- Masaki Tsuchiya
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Yuji Hara
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan. .,AMED-PRIME, Japan Agency for Medical Research and Development, Tokyo, 100-0004, Japan.
| | - Masaki Okuda
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Karin Itoh
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Ryotaro Nishioka
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Akifumi Shiomi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Kohjiro Nagao
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Masayuki Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Junichi Ikenouchi
- Department of Biology, Faculty of Sciences, Kyushu University, Fukuoka, 819-0395, Japan
| | - Ryo Suzuki
- Institute for Integrated Cell-Material Sciences (WPI iCeMS), Kyoto University, Kyoto, 606-8501, Japan.,Center for Integrative Medicine and Physics, Institute for Advanced Study, Kyoto University, Kyoto, 606-8501, Japan
| | - Motomu Tanaka
- Institute for Integrated Cell-Material Sciences (WPI iCeMS), Kyoto University, Kyoto, 606-8501, Japan.,Physical Chemistry of Biosystems, Institute of Physical Chemistry, University of Heidelberg, Heidelberg, 69120, Germany.,Center for Integrative Medicine and Physics, Institute for Advanced Study, Kyoto University, Kyoto, 606-8501, Japan
| | - Tomohiko Ohwada
- Laboratory of Organic and Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 115-0033, Japan
| | - Junken Aoki
- Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Miyagi, 980-8578, Japan
| | - Motoi Kanagawa
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Tatsushi Toda
- Department of Neurology, Division of Neuroscience, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Yosuke Nagata
- Department of Life Science, Faculty of Science, Okayama University of Science, Okayama, 700-0005, Japan
| | - Ryoichi Matsuda
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, 153-8902, Japan
| | - Yasunori Takayama
- Division of Cell Signaling, Okazaki Institute for Integrative Bioscience, Aichi, 444-8787, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, Okazaki Institute for Integrative Bioscience, Aichi, 444-8787, Japan
| | - Masato Umeda
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan.
| |
Collapse
|
195
|
Domenighetti AA, Mathewson MA, Pichika R, Sibley LA, Zhao L, Chambers HG, Lieber RL. Loss of myogenic potential and fusion capacity of muscle stem cells isolated from contractured muscle in children with cerebral palsy. Am J Physiol Cell Physiol 2018; 315:C247-C257. [PMID: 29694232 DOI: 10.1152/ajpcell.00351.2017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cerebral palsy (CP) is the most common cause of pediatric neurodevelopmental and physical disability in the United States. It is defined as a group of motor disorders caused by a nonprogressive perinatal insult to the brain. Although the brain lesion is nonprogressive, there is a progressive, lifelong impact on skeletal muscles, which are shorter, spastic, and may develop debilitating contractures. Satellite cells are resident muscle stem cells that are indispensable for postnatal growth and regeneration of skeletal muscles. Here we measured the myogenic potential of satellite cells isolated from contractured muscles in children with CP. When compared with typically developing (TD) children, satellite cell-derived myoblasts from CP differentiated more slowly (slope: 0.013 (SD 0.013) CP vs. 0.091 (SD 0.024) TD over 24 h, P < 0.001) and fused less (fusion index: 21.3 (SD 8.6) CP vs. 81.3 (SD 7.7) TD after 48 h, P < 0.001) after exposure to low-serum conditions that stimulated myotube formation. This impairment was associated with downregulation of several markers important for myoblast fusion and myotube formation, including DNA methylation-dependent inhibition of promyogenic integrin-β 1D (ITGB1D) protein expression levels (-50% at 42 h), and ~25% loss of integrin-mediated focal adhesion kinase phosphorylation. The cytidine analog 5-Azacytidine (5-AZA), a demethylating agent, restored ITGB1D levels and promoted myogenesis in CP cultures. Our data demonstrate that muscle contractures in CP are associated with loss of satellite cell myogenic potential that is dependent on DNA methylation patterns affecting expression of genetic programs associated with muscle stem cell differentiation and muscle fiber formation.
Collapse
Affiliation(s)
- Andrea A Domenighetti
- The Shirley Ryan AbilityLab, Chicago, Illinois.,Department of Physical Medicine & Rehabilitation, Northwestern University , Chicago, Illinois.,Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, California
| | - Margie A Mathewson
- Bioengineering Department, University of California, San Diego, La Jolla, California
| | | | | | - Leyna Zhao
- ACEA Biosciences Incorporated, San Diego, California
| | | | - Richard L Lieber
- The Shirley Ryan AbilityLab, Chicago, Illinois.,Department of Physical Medicine & Rehabilitation, Northwestern University , Chicago, Illinois.,Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, California
| |
Collapse
|
196
|
Weiler J, Mohr M, Zänker KS, Dittmar T. Matrix metalloproteinase-9 (MMP9) is involved in the TNF-α-induced fusion of human M13SV1-Cre breast epithelial cells and human MDA-MB-435-pFDR1 cancer cells. Cell Commun Signal 2018; 16:14. [PMID: 29636110 PMCID: PMC5894245 DOI: 10.1186/s12964-018-0226-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/02/2018] [Indexed: 01/07/2023] Open
Abstract
Background In addition to physiological events such as fertilisation, placentation, osteoclastogenesis, or tissue regeneration/wound healing, cell fusion is involved in pathophysiological conditions such as cancer. Cell fusion, which applies to both the proteins and conditions that induce the merging of two or more cells, is not a fully understood process. Inflammation/pro-inflammatory cytokines might be a positive trigger for cell fusion. Using a Cre-LoxP-based cell fusion assay we demonstrated that the fusion between human M13SV1-Cre breast epithelial cells and human MDA-MB-435-pFDR1 cancer cells was induced by the pro-inflammatory cytokine tumour necrosis factor-α (TNF-α). Methods The gene expression profile of the cells in the presence of TNF-α and under normoxic and hypoxic conditions was analysed by cDNA microarray analysis. cDNA microarray data were verified by qPCR, PCR, Western blot and zymography. Quantification of cell fusion events was determined by flow cytometry. Proteins of interest were either blocked or knocked-down using a specific inhibitor, siRNA or a blocking antibody. Results The data showed an up-regulation of various genes, including claudin-1 (CLDN1), ICAM1, CCL2 and MMP9 in M13SV1-Cre and/or MDA-MB-435-pFDR1 cells. Inhibition of these proteins using a blocking ICAM1 antibody, CLDN1 siRNA or an MMP9 inhibitor showed that only the blockage of MMP9 was correlated with a decreased fusion rate of the cells. Likewise, the tetracycline-based antibiotic minocycline, which exhibits anti-inflammatory properties, was also effective in both inhibiting the TNF-α-induced MMP9 expression in M13SV1-Cre cells and blocking the TNF-α-induced fusion frequency of human M13SV1-Cre breast epithelial cells and human MDA-MB-435-pFDR1 cancer cells. Conclusions The matrix metalloproteinase-9 (MMP9) is most likely involved in the TNF-α-mediated fusion of human M13SV1-Cre breast epithelial cells and human MDA-MB-435-pFDR1 cancer cells. Likewise, our data indicate that the tetracycline-based antibiotic minocycline might exhibit anti-fusogenic properties because it inhibits a cell fusion-related mechanism. Electronic supplementary material The online version of this article (10.1186/s12964-018-0226-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julian Weiler
- Institute of Immunology, Centre of Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, 58448, Witten, Germany
| | - Marieke Mohr
- BioGenes GmbH, Köpenicker Str. 325, 12555, Berlin, Germany
| | - Kurt S Zänker
- Institute of Immunology, Centre of Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, 58448, Witten, Germany
| | - Thomas Dittmar
- Institute of Immunology, Centre of Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, 58448, Witten, Germany.
| |
Collapse
|
197
|
Bi P, McAnally JR, Shelton JM, Sánchez-Ortiz E, Bassel-Duby R, Olson EN. Fusogenic micropeptide Myomixer is essential for satellite cell fusion and muscle regeneration. Proc Natl Acad Sci U S A 2018; 115:3864-3869. [PMID: 29581287 PMCID: PMC5899482 DOI: 10.1073/pnas.1800052115] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Regeneration of skeletal muscle in response to injury occurs through fusion of a population of stem cells, known as satellite cells, with injured myofibers. Myomixer, a muscle-specific membrane micropeptide, cooperates with the transmembrane protein Myomaker to regulate embryonic myoblast fusion and muscle formation. To investigate the role of Myomixer in muscle regeneration, we used CRISPR/Cas9-mediated genome editing to generate conditional knockout Myomixer alleles in mice. We show that genetic deletion of Myomixer in satellite cells using a tamoxifen-regulated Cre recombinase transgene under control of the Pax7 promoter abolishes satellite cell fusion and prevents muscle regeneration, resulting in severe muscle degeneration after injury. Satellite cells devoid of Myomixer maintain expression of Myomaker, demonstrating that Myomaker alone is insufficient to drive myoblast fusion. These findings, together with prior studies demonstrating the essentiality of Myomaker for muscle regeneration, highlight the obligatory partnership of Myomixer and Myomaker for myofiber formation throughout embryogenesis and adulthood.
Collapse
Affiliation(s)
- Pengpeng Bi
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - John R McAnally
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - John M Shelton
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Efrain Sánchez-Ortiz
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Eric N Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390;
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
198
|
Camuglia JM, Mandigo TR, Moschella R, Mark J, Hudson CH, Sheen D, Folker ES. An RNAi based screen in Drosophila larvae identifies fascin as a regulator of myoblast fusion and myotendinous junction structure. Skelet Muscle 2018; 8:12. [PMID: 29625624 PMCID: PMC5889537 DOI: 10.1186/s13395-018-0159-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 03/22/2018] [Indexed: 02/08/2023] Open
Abstract
Background A strength of Drosophila as a model system is its utility as a tool to screen for novel regulators of various functional and developmental processes. However, the utility of Drosophila as a screening tool is dependent on the speed and simplicity of the assay used. Methods Here, we use larval locomotion as an assay to identify novel regulators of skeletal muscle function. We combined this assay with muscle-specific depletion of 82 genes to identify genes that impact muscle function by their expression in muscle cells. The data from the screen were supported with characterization of the muscle pattern in embryos and larvae that had disrupted expression of the strongest hit from the screen. Results With this assay, we showed that 12/82 tested genes regulate muscle function. Intriguingly, the disruption of five genes caused an increase in muscle function, illustrating that mechanisms that reduce muscle function exist and that the larval locomotion assay is sufficiently quantitative to identify conditions that both increase and decrease muscle function. We extended the data from this screen and tested the mechanism by which the strongest hit, fascin, impacted muscle function. Compared to controls, animals in which fascin expression was disrupted with either a mutant allele or muscle-specific expression of RNAi had fewer muscles, smaller muscles, muscles with fewer nuclei, and muscles with disrupted myotendinous junctions. However, expression of RNAi against fascin only after the muscle had finished embryonic development did not recapitulate any of these phenotypes. Conclusions These data suggest that muscle function is reduced due to impaired myoblast fusion, muscle growth, and muscle attachment. Together, these data demonstrate the utility of Drosophila larval locomotion as an assay for the identification of novel regulators of muscle development and implicate fascin as necessary for embryonic muscle development. Electronic supplementary material The online version of this article (10.1186/s13395-018-0159-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Torrey R Mandigo
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA
| | | | - Jenna Mark
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA
| | | | - Derek Sheen
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA
| | - Eric S Folker
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA.
| |
Collapse
|
199
|
Pélisse M, Der Vartanian A, Germot A, Maftah A. Protein O-Glucosyltransferase 1 Expression Influences Formation of Differentiated Myotubes in C2C12 Cell Line. DNA Cell Biol 2018; 37:359-372. [DOI: 10.1089/dna.2017.4052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Mélanie Pélisse
- Université de Limoges, INRA, UMR1061 Génétique Moléculaire Animale, FR 3503 GEIST, Limoges, France
| | - Audrey Der Vartanian
- Université de Limoges, INRA, UMR1061 Génétique Moléculaire Animale, FR 3503 GEIST, Limoges, France
- Faculté de Médecine, INSERM, IMRB U955-E10, Université Paris Est, Créteil, France
| | - Agnès Germot
- Université de Limoges, INRA, UMR1061 Génétique Moléculaire Animale, FR 3503 GEIST, Limoges, France
| | - Abderrahman Maftah
- Université de Limoges, INRA, UMR1061 Génétique Moléculaire Animale, FR 3503 GEIST, Limoges, France
| |
Collapse
|
200
|
Thomas JD, Oliveira R, Sznajder ŁJ, Swanson MS. Myotonic Dystrophy and Developmental Regulation of RNA Processing. Compr Physiol 2018; 8:509-553. [PMID: 29687899 PMCID: PMC11323716 DOI: 10.1002/cphy.c170002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Myotonic dystrophy (DM) is a multisystemic disorder caused by microsatellite expansion mutations in two unrelated genes leading to similar, yet distinct, diseases. DM disease presentation is highly variable and distinguished by differences in age-of-onset and symptom severity. In the most severe form, DM presents with congenital onset and profound developmental defects. At the molecular level, DM pathogenesis is characterized by a toxic RNA gain-of-function mechanism that involves the transcription of noncoding microsatellite expansions. These mutant RNAs disrupt key cellular pathways, including RNA processing, localization, and translation. In DM, these toxic RNA effects are predominantly mediated through the modulation of the muscleblind-like and CUGBP and ETR-3-like factor families of RNA binding proteins (RBPs). Dysfunction of these RBPs results in widespread RNA processing defects culminating in the expression of developmentally inappropriate protein isoforms in adult tissues. The tissue that is the focus of this review, skeletal muscle, is particularly sensitive to mutant RNA-responsive perturbations, as patients display a variety of developmental, structural, and functional defects in muscle. Here, we provide a comprehensive overview of DM1 and DM2 clinical presentation and pathology as well as the underlying cellular and molecular defects associated with DM disease onset and progression. Additionally, fundamental aspects of skeletal muscle development altered in DM are highlighted together with ongoing and potential therapeutic avenues to treat this muscular dystrophy. © 2018 American Physiological Society. Compr Physiol 8:509-553, 2018.
Collapse
Affiliation(s)
- James D. Thomas
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida, USA
| | - Ruan Oliveira
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida, USA
| | - Łukasz J. Sznajder
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida, USA
| | - Maurice S. Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida, USA
| |
Collapse
|