1
|
Ruiz Daniels R, Salisbury SJ, Sveen L, Villamayor PR, Taylor RS, Vaadal M, Tengs T, Krasnov A, Monaghan SJ, Ballantyne M, Penaloza C, Fast MD, Bron JE, Houston R, Robinson N, Robledo D. Transcriptomic characterization of transitioning cell types in the skin of Atlantic salmon. BMC Biol 2025; 23:109. [PMID: 40289111 PMCID: PMC12036301 DOI: 10.1186/s12915-025-02196-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 03/21/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND The skin maintains the body's integrity and serves as the first line of defence against pathogens, stressors and mechanical injuries. Despite the global significance of salmon in aquaculture, how the transcriptomic profile of cells varies during wound healing remains unexplored. Teleost's skin contains adult pluripotent cells that differentiate into various tissues, including bone, cartilage, tendon, ligament, adipose, dermis, muscle and connective tissue within the skin. These cells are pivotal for preserving the integrity of skin tissue throughout an organism's lifespan and actively participate in the wound healing processes. In this study, we characterize the transcriptomic profiles of putative mesenchymal stromal cells (fibroblast-like adult stem cells) in healthy Atlantic salmon tissue and during the wound healing process. RESULTS Single-nucleus sequencing and spatial transcriptomics were used to detect transcriptomic changes occurring during wound healing that are commonly associated with mesenchymal stromal cells. We followed the transcriptomic activity of these cells during an in vivo wound healing time course study showing that these cells become more transcriptionally active during the remodelling stage of wound healing. The changes detected give insights into the potential differentiation pathways leading to osteogenic and fibroblast lineages in the skin of Atlantic salmon. CONCLUSIONS We chart the transcriptomic activity of subclusters of putative differentiating stromal cells during the process of wound healing for the first time, revealing different spatial niches of the various putative MSC subclusters, and setting the stage for further investigation of the manipulation of transitioning cell types to improve fish health.
Collapse
Affiliation(s)
- R Ruiz Daniels
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK.
- Institute of Aquaculture, University of Stirling, Stirling, UK.
| | - S J Salisbury
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | | | - P R Villamayor
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - R S Taylor
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | | | | | | | - S J Monaghan
- Institute of Aquaculture, University of Stirling, Stirling, UK
| | - M Ballantyne
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - C Penaloza
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
- Benchmark Genetics, Penicuik, UK
| | - M D Fast
- Hoplite Research Lab, Department of Pathology and Microbiology, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PEI, Canada
| | - J E Bron
- Institute of Aquaculture, University of Stirling, Stirling, UK
| | | | - N Robinson
- Nofima AS, Ås, Norway
- Sustainable Aquaculture Laboratory, Deakin University, Victoria, Australia
| | - D Robledo
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK.
- University of Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|
2
|
Du K, Wang DH, Hu SQ, Xia Y, Wu Q, Gu MQ, Chen XW. Genome-wide chromatin accessibility and selective signals of meat rabbits reveal key Cis-regulatory elements and variants during postnatal development of skeletal muscles in rabbits. BMC Genomics 2025; 26:296. [PMID: 40133827 PMCID: PMC11934498 DOI: 10.1186/s12864-025-11496-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND The development of skeletal muscles is intricately modulated by multiple genetic factors and significantly impacts the economic value of meat rabbits. However, our knowledge of epigenetics in rabbit skeletal muscles remains largely unknown. RESULTS In this study, we collected leg skeletal muscles of rabbits and performed assays for transposase-accessible chromatin with high throughput sequencing (ATAC-seq) to detect open chromatin across three developmental stages: birth (D1), weaning (D35), and adulthood (D75). A total of 126,959 accessible chromatin regions (ACRs) were identified across samples, and a broad increase and decrease in chromatin accessibility were found from D1 to D35 and D35 to D75, respectively. Integrative analysis of chromatin accessibility and transcriptome data revealed ACRs that were nearly closed at D1 but highly accessible at D35 and D75 were significantly enriched in skeletal muscle development. Cis-regulation analysis further revealed that genes dominated by enhancers mainly play roles in the neuron development of rabbit skeletal muscles. Moreover, the detection of selection signals of meat rabbits and the footprinting analysis of transcription factor at open chromatin revealed that both base transversion (Chr13:12144967 A-> G) and the dynamics of chromatin accessibility at the PRDM1 binding site might regulate ZSWIM5 during the development of skeletal muscles in rabbits. CONCLUSIONS Our study provided a category of potential cis-regulatory elements for understanding the development of skeletal muscles at the tissue level and might facilitate potential insights into growth regulation in rabbits.
Collapse
Affiliation(s)
- Kun Du
- Technology Research Center of Modern Breeding Development, Mianyang Normal University, Mianyang, 621000, China
| | - Dai-Hua Wang
- Technology Research Center of Modern Breeding Development, Mianyang Normal University, Mianyang, 621000, China
| | - Shen-Qiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Yu Xia
- Technology Research Center of Modern Breeding Development, Mianyang Normal University, Mianyang, 621000, China
| | - Qian Wu
- Technology Research Center of Modern Breeding Development, Mianyang Normal University, Mianyang, 621000, China
| | - Mao-Qing Gu
- Technology Research Center of Modern Breeding Development, Mianyang Normal University, Mianyang, 621000, China
| | - Xi-Wen Chen
- Technology Research Center of Modern Breeding Development, Mianyang Normal University, Mianyang, 621000, China.
| |
Collapse
|
3
|
Aranda-Martínez P, Sayed RKA, Fernández-Martínez J, Ramírez-Casas Y, Yang Y, Escames G, Acuña-Castroviejo D. Zebrafish as a Human Muscle Model for Studying Age-Dependent Sarcopenia and Frailty. Int J Mol Sci 2024; 25:6166. [PMID: 38892357 PMCID: PMC11172448 DOI: 10.3390/ijms25116166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Currently, there is an increase in the aging of the population, which represents a risk factor for many diseases, including sarcopenia. Sarcopenia involves progressive loss of mass, strength, and function of the skeletal muscle. Some mechanisms include alterations in muscle structure, reduced regenerative capacity, oxidative stress, mitochondrial dysfunction, and inflammation. The zebrafish has emerged as a new model for studying skeletal muscle aging because of its numerous advantages, including histological and molecular similarity to human skeletal muscle. In this study, we used fish of 2, 10, 30, and 60 months of age. The older fish showed a higher frailty index with a value of 0.250 ± 0.000 because of reduced locomotor activity and alterations in biometric measurements. We observed changes in muscle structure with a decreased number of myocytes (0.031 myocytes/μm2 ± 0.004 at 60 months) and an increase in collagen with aging up to 15% ± 1.639 in the 60-month group, corresponding to alterations in the synthesis, degradation, and differentiation pathways. These changes were accompanied by mitochondrial alterations, such as a nearly 50% reduction in the number of intermyofibrillar mitochondria, 100% mitochondrial damage, and reduced mitochondrial dynamics. Overall, we demonstrated a similarity in the aging processes of muscle aging between zebrafish and mammals.
Collapse
Affiliation(s)
- Paula Aranda-Martínez
- Centro de Investigación Biomédica, Facultad de Medicina, Departamento de Fisiología, Universidad de Granada, 18016 Granada, Spain; (P.A.-M.); (J.F.-M.); (Y.R.-C.); (G.E.)
- Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria (Ibs. Granada), Hospital Universitario San Cecilio, 18016 Granada, Spain
| | - Ramy K. A. Sayed
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt;
| | - José Fernández-Martínez
- Centro de Investigación Biomédica, Facultad de Medicina, Departamento de Fisiología, Universidad de Granada, 18016 Granada, Spain; (P.A.-M.); (J.F.-M.); (Y.R.-C.); (G.E.)
- Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria (Ibs. Granada), Hospital Universitario San Cecilio, 18016 Granada, Spain
| | - Yolanda Ramírez-Casas
- Centro de Investigación Biomédica, Facultad de Medicina, Departamento de Fisiología, Universidad de Granada, 18016 Granada, Spain; (P.A.-M.); (J.F.-M.); (Y.R.-C.); (G.E.)
- Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria (Ibs. Granada), Hospital Universitario San Cecilio, 18016 Granada, Spain
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi’an 710069, China;
| | - Germaine Escames
- Centro de Investigación Biomédica, Facultad de Medicina, Departamento de Fisiología, Universidad de Granada, 18016 Granada, Spain; (P.A.-M.); (J.F.-M.); (Y.R.-C.); (G.E.)
- Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria (Ibs. Granada), Hospital Universitario San Cecilio, 18016 Granada, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Darío Acuña-Castroviejo
- Centro de Investigación Biomédica, Facultad de Medicina, Departamento de Fisiología, Universidad de Granada, 18016 Granada, Spain; (P.A.-M.); (J.F.-M.); (Y.R.-C.); (G.E.)
- Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria (Ibs. Granada), Hospital Universitario San Cecilio, 18016 Granada, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- UGC de Laboratorios Clínicos, Hospital Universitario San Cecilio, 18016 Granada, Spain
| |
Collapse
|
4
|
Fan D, Yao Y, Liu Y, Yan C, Li F, Wang S, Yu M, Xie B, Tang Z. Regulation of myo-miR-24-3p on the Myogenesis and Fiber Type Transformation of Skeletal Muscle. Genes (Basel) 2024; 15:269. [PMID: 38540328 PMCID: PMC10970682 DOI: 10.3390/genes15030269] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 06/15/2024] Open
Abstract
Skeletal muscle plays critical roles in providing a protein source and contributing to meat production. It is well known that microRNAs (miRNAs) exert important effects on various biological processes in muscle, including cell fate determination, muscle fiber morphology, and structure development. However, the role of miRNA in skeletal muscle development remains incompletely understood. In this study, we observed a critical miRNA, miR-24-3p, which exhibited higher expression levels in Tongcheng (obese-type) pigs compared to Landrace (lean-type) pigs. Furthermore, we found that miR-24-3p was highly expressed in the dorsal muscle of pigs and the quadriceps muscle of mice. Functionally, miR-24-3p was found to inhibit proliferation and promote differentiation in muscle cells. Additionally, miR-24-3p was shown to facilitate the conversion of slow muscle fibers to fast muscle fibers and influence the expression of GLUT4, a glucose transporter. Moreover, in a mouse model of skeletal muscle injury, we demonstrated that overexpression of miR-24-3p promoted rapid myogenesis and contributed to skeletal muscle regeneration. Furthermore, miR-24-3p was found to regulate the expression of target genes, including Nek4, Pim1, Nlk, Pskh1, and Mapk14. Collectively, our findings provide evidence that miR-24-3p plays a regulatory role in myogenesis and fiber type conversion. These findings contribute to our understanding of human muscle health and have implications for improving meat production traits in livestock.
Collapse
Affiliation(s)
- Danyang Fan
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (D.F.); (Y.L.); (M.Y.)
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (C.Y.); (F.L.); (S.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China;
| | - Yilong Yao
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China;
| | - Yanwen Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (D.F.); (Y.L.); (M.Y.)
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (C.Y.); (F.L.); (S.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China;
| | - Chao Yan
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (C.Y.); (F.L.); (S.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China;
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Fanqinyu Li
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (C.Y.); (F.L.); (S.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China;
| | - Shilong Wang
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (C.Y.); (F.L.); (S.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China;
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Mei Yu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (D.F.); (Y.L.); (M.Y.)
| | - Bingkun Xie
- Guangxi Key Laboratory of Livestock Genetic Improvement, Guangxi Institute of Animal Sciences, Nanning 530001, China;
| | - Zhonglin Tang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (D.F.); (Y.L.); (M.Y.)
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China; (C.Y.); (F.L.); (S.W.)
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China;
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| |
Collapse
|
5
|
Oudhoff H, Hisler V, Baumgartner F, Rees L, Grepper D, Jaźwińska A. Skeletal muscle regeneration after extensive cryoinjury of caudal myomeres in adult zebrafish. NPJ Regen Med 2024; 9:8. [PMID: 38378693 PMCID: PMC10879182 DOI: 10.1038/s41536-024-00351-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 01/24/2024] [Indexed: 02/22/2024] Open
Abstract
Skeletal muscles can regenerate after minor injuries, but severe structural damage often leads to fibrosis in mammals. Whether adult zebrafish possess the capacity to reproduce profoundly destroyed musculature remains unknown. Here, a new cryoinjury model revealed that several myomeres efficiently regenerated within one month after wounding the zebrafish caudal peduncle. Wound clearance involved accumulation of the selective autophagy receptor p62, an immune response and Collagen XII deposition. New muscle formation was associated with proliferation of Pax7 expressing muscle stem cells, which gave rise to MyoD1 positive myogenic precursors, followed by myofiber differentiation. Monitoring of slow and fast muscles revealed their coordinated replacement in the superficial and profound compartments of the myomere. However, the final boundary between the muscular components was imperfectly recapitulated, allowing myofibers of different identities to intermingle. The replacement of connective with sarcomeric tissues required TOR signaling, as rapamycin treatment impaired new muscle formation, leading to persistent fibrosis. The model of zebrafish myomere restoration may provide new medical perspectives for treatment of traumatic injuries.
Collapse
Affiliation(s)
- Hendrik Oudhoff
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700, Fribourg, Switzerland
| | - Vincent Hisler
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700, Fribourg, Switzerland
| | - Florian Baumgartner
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700, Fribourg, Switzerland
| | - Lana Rees
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700, Fribourg, Switzerland
| | - Dogan Grepper
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700, Fribourg, Switzerland
| | - Anna Jaźwińska
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700, Fribourg, Switzerland.
| |
Collapse
|
6
|
Kahsay A, Dennhag N, Liu JX, Nord H, Rönnbäck H, Thorell AE, von Hofsten J, Pedrosa Domellöf F. Obscurin Maintains Myofiber Identity in Extraocular Muscles. Invest Ophthalmol Vis Sci 2024; 65:19. [PMID: 38334702 PMCID: PMC10860686 DOI: 10.1167/iovs.65.2.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/12/2024] [Indexed: 02/10/2024] Open
Abstract
Purpose The cytoskeleton of the extraocular muscles (EOMs) is significantly different from that of other muscles. We aimed to investigate the role of obscurin, a fundamental cytoskeletal protein, in the EOMs. Methods The distribution of obscurin in human and zebrafish EOMs was compared using immunohistochemistry. The two obscurin genes in zebrafish, obscna and obscnb, were knocked out using CRISPR/Cas9, and the EOMs were investigated using immunohistochemistry, qPCR, and in situ hybridization. The optokinetic reflex (OKR) in five-day-old larvae and adult obscna-/-;obscnb-/- and sibling control zebrafish was analyzed. Swimming distance was recorded at the same age. Results The obscurin distribution pattern was similar in human and zebrafish EOMs. The proportion of slow and fast myofibers was reduced in obscna-/-;obscnb-/- zebrafish EOMs but not in trunk muscle, whereas the number of myofibers containing cardiac myosin myh7 was significantly increased in EOMs of obscurin double mutants. Loss of obscurin resulted in less OKRs in zebrafish larvae but not in adult zebrafish. Conclusions Obscurin expression is conserved in normal human and zebrafish EOMs. Loss of obscurin induces a myofiber type shift in the EOMs, with upregulation of cardiac myosin heavy chain, myh7, showing an adaptation strategy in EOMs. Our model will facilitate further studies in conditions related to obscurin.
Collapse
Affiliation(s)
- Abraha Kahsay
- Department of Integrative Medical Biology (IMB), Umeå University, Umeå, Sweden
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| | - Nils Dennhag
- Department of Integrative Medical Biology (IMB), Umeå University, Umeå, Sweden
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| | - Jing-Xia Liu
- Department of Integrative Medical Biology (IMB), Umeå University, Umeå, Sweden
| | - Hanna Nord
- Department of Integrative Medical Biology (IMB), Umeå University, Umeå, Sweden
| | - Hugo Rönnbäck
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| | | | - Jonas von Hofsten
- Department of Integrative Medical Biology (IMB), Umeå University, Umeå, Sweden
| | - Fatima Pedrosa Domellöf
- Department of Integrative Medical Biology (IMB), Umeå University, Umeå, Sweden
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| |
Collapse
|
7
|
Ramírez-Vidal L, Molina-Villa T, Mendoza V, Peralta-Álvarez CA, Poot-Hernández AC, Dotov D, López-Casillas F. Betaglycan promoter activity is differentially regulated during myogenesis in zebrafish embryo somites. Dev Dyn 2023; 252:1162-1179. [PMID: 37222488 DOI: 10.1002/dvdy.602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 03/28/2023] [Accepted: 04/25/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Betaglycan, also known as the TGFβ type III receptor (Tgfbr3), is a co-receptor that modulates TGFβ family signaling. Tgfbr3 is upregulated during C2C12 myoblast differentiation and expressed in mouse embryos myocytes. RESULTS To investigate tgfbr3 transcriptional regulation during zebrafish embryonic myogenesis, we cloned a 3.2 kb promoter fragment that drives reporter transcription during C2C12 myoblasts differentiation and in the Tg(tgfbr3:mCherry) transgenic zebrafish. We detect tgfbr3 protein and mCherry expression in the adaxial cells concomitantly with the onset of their radial migration to become slow-twitch muscle fibers in the Tg(tgfbr3:mCherry). Remarkably, this expression displays a measurable antero-posterior somitic gradient expression. CONCLUSIONS tgfbr3 is transcriptionally regulated during somitic muscle development in zebrafish with an antero-posterior gradient expression that preferentially marks the adaxial cells and their descendants.
Collapse
Affiliation(s)
- Lizbeth Ramírez-Vidal
- Departmento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, UNAM, Mexico City, Mexico
| | - Tonatiuh Molina-Villa
- Departmento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, UNAM, Mexico City, Mexico
| | - Valentín Mendoza
- Departmento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, UNAM, Mexico City, Mexico
| | | | | | - Dobromir Dotov
- Psychology, Neuroscience & Behaviour, McMaster University, Hamilton, Canada
| | - Fernando López-Casillas
- Departmento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, UNAM, Mexico City, Mexico
| |
Collapse
|
8
|
Truong BT, Shull LC, Lencer E, Bend EG, Field M, Blue EE, Bamshad MJ, Skinner C, Everman D, Schwartz CE, Flanagan-Steet H, Artinger KB. PRDM1 DNA-binding zinc finger domain is required for normal limb development and is disrupted in split hand/foot malformation. Dis Model Mech 2023; 16:dmm049977. [PMID: 37083955 PMCID: PMC10151829 DOI: 10.1242/dmm.049977] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/09/2023] [Indexed: 04/22/2023] Open
Abstract
Split hand/foot malformation (SHFM) is a rare limb abnormality with clefting of the fingers and/or toes. For many individuals, the genetic etiology is unknown. Through whole-exome and targeted sequencing, we detected three novel variants in a gene encoding a transcription factor, PRDM1, that arose de novo in families with SHFM or segregated with the phenotype. PRDM1 is required for limb development; however, its role is not well understood and it is unclear how the PRDM1 variants affect protein function. Using transient and stable overexpression rescue experiments in zebrafish, we show that the variants disrupt the proline/serine-rich and DNA-binding zinc finger domains, resulting in a dominant-negative effect. Through gene expression assays, RNA sequencing, and CUT&RUN in isolated pectoral fin cells, we demonstrate that Prdm1a directly binds to and regulates genes required for fin induction, outgrowth and anterior/posterior patterning, such as fgfr1a, dlx5a, dlx6a and smo. Taken together, these results improve our understanding of the role of PRDM1 in the limb gene regulatory network and identified novel PRDM1 variants that link to SHFM in humans.
Collapse
Affiliation(s)
- Brittany T. Truong
- Human Medical Genetics & Genomics Graduate Program, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Lomeli C. Shull
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ezra Lencer
- Biology Department, Lafayette College, Easton, PA 18042, USA
| | - Eric G. Bend
- Greenwood Genetics Center, Greenwood, SC 29646, USA
| | - Michael Field
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, NSW 2298, AUS
| | - Elizabeth E. Blue
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA
| | - Michael J. Bamshad
- Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | - Kristin B. Artinger
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
9
|
Pipalia TG, Sultan SHA, Koth J, Knight RD, Hughes SM. Skeletal Muscle Regeneration in Zebrafish. Methods Mol Biol 2023; 2640:227-248. [PMID: 36995599 DOI: 10.1007/978-1-0716-3036-5_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Muscle regeneration models have revealed mechanisms of inflammation, wound clearance, and stem cell-directed repair of damage, thereby informing therapy. Whereas studies of muscle repair are most advanced in rodents, the zebrafish is emerging as an additional model organism with genetic and optical advantages. Various muscle wounding protocols (both chemical and physical) have been published. Here we describe simple, cheap, precise, adaptable, and effective wounding protocols and analysis methods for two stages of a larval zebrafish skeletal muscle regeneration model. We show examples of how muscle damage, ingression of muscle stem cells, immune cells, and regeneration of fibers can be monitored over an extended timecourse in individual larvae. Such analyses have the potential to greatly enhance understanding, by reducing the need to average regeneration responses across individuals subjected to an unavoidably variable wound stimulus.
Collapse
Affiliation(s)
- Tapan G Pipalia
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
| | - Sami H A Sultan
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Jana Koth
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
- Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | - Robert D Knight
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Simon M Hughes
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK.
| |
Collapse
|
10
|
Wu P, Yong P, Zhang Z, Xu R, Shang R, Shi J, Zhang J, Bi P, Chen E, Du S. Loss of Myomixer Results in Defective Myoblast Fusion, Impaired Muscle Growth, and Severe Myopathy in Zebrafish. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2022; 24:1023-1038. [PMID: 36083384 PMCID: PMC10112271 DOI: 10.1007/s10126-022-10159-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 08/25/2022] [Indexed: 06/15/2023]
Abstract
The development and growth of fish skeletal muscles require myoblast fusion to generate multinucleated myofibers. While zebrafish fast-twitch muscle can fuse to generate multinucleated fibers, the slow-twitch muscle fibers remain mononucleated in zebrafish embryos and larvae. The mechanism underlying the fiber-type-specific control of fusion remains elusive. Recent genetic studies using mice identified a long-sought fusion factor named Myomixer. To understand whether Myomixer is involved in the fiber-type specific fusion, we analyzed the transcriptional regulation of myomixer expression and characterized the muscle growth phenotype upon genetic deletion of myomixer in zebrafish. The data revealed that overexpression of Sonic Hedgehog (Shh) drastically inhibited myomixer expression and blocked myoblast fusion, recapitulating the phenotype upon direct genetic deletion of myomixer from zebrafish. The fusion defect in myomixer mutant embryos could be faithfully rescued upon re-expression of zebrafish myomixer gene or its orthologs from shark or human. Interestingly, myomixer mutant fish survived to adult stage though were notably smaller than wildtype siblings. Severe myopathy accompanied by the uncontrolled adipose infiltration was observed in both fast and slow muscle tissues of adult myomixer mutants. Collectively, our data highlight an indispensable role of myomixer gene for cell fusion during both embryonic muscle development and post-larval muscle growth.
Collapse
Affiliation(s)
- Ping Wu
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, USA
- Department of Bioengineering and Environmental Science, Changsha University, Changsha, China
| | - Pengzheng Yong
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, USA
| | - Zhanxiong Zhang
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, USA
| | - Rui Xu
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, USA
| | - Renjie Shang
- Center for Molecular Medicine & Department of Genetics, University of Georgia, Athens, USA
| | - Jun Shi
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, USA
- College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Jianshe Zhang
- Department of Bioengineering and Environmental Science, Changsha University, Changsha, China
| | - Pengpeng Bi
- Center for Molecular Medicine & Department of Genetics, University of Georgia, Athens, USA
| | - Elizabeth Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, USA
| | - Shaojun Du
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, USA.
| |
Collapse
|
11
|
Zeng C, Shi H, Kirkpatrick LT, Ricome A, Park S, Scheffler JM, Hannon KM, Grant AL, Gerrard DE. Driving an Oxidative Phenotype Protects Myh4 Null Mice From Myofiber Loss During Postnatal Growth. Front Physiol 2022; 12:785151. [PMID: 35283757 PMCID: PMC8908108 DOI: 10.3389/fphys.2021.785151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/28/2021] [Indexed: 12/17/2022] Open
Abstract
Postnatal muscle growth is accompanied by increases in fast fiber type compositions and hypertrophy, raising the possibility that a slow to fast transition may be partially requisite for increases in muscle mass. To test this hypothesis, we ablated the Myh4 gene, and thus myosin heavy chain IIB protein and corresponding fibers in mice, and examined its consequences on postnatal muscle growth. Wild-type and Myh4–/– mice had the same number of muscle fibers at 2 weeks postnatal. However, the gastrocnemius muscle lost up to 50% of its fibers between 2 and 4 weeks of age, though stabilizing thereafter. To compensate for the lack of functional IIB fibers, type I, IIA, and IIX(D) fibers increased in prevalence and size. To address whether slowing the slow-to-fast fiber transition process would rescue fiber loss in Myh4–/– mice, we stimulated the oxidative program in muscle of Myh4–/– mice either by overexpression of PGC-1α, a well-established model for fast-to-slow fiber transition, or by feeding mice AICAR, a potent AMP kinase agonist. Forcing an oxidative metabolism in muscle only partially protected the gastrocnemius muscle from loss of fibers in Myh4–/– mice. To explore whether traditional means of stimulating muscle hypertrophy could overcome the muscling deficits in postnatal Myh4–/– mice, myostatin null mice were bred with Myh4–/– mice, or Myh4–/– mice were fed the growth promotant clenbuterol. Interestingly, both genetic and pharmacological stimulations had little impact on mice lacking a functional Myh4 gene suggesting that the existing muscle fibers have maximized its capacity to enlarge to compensate for the lack of its neighboring IIB fibers. Curiously, however, cell signaling events responsible for IIB fiber formation remained intact in the tissue. These findings further show disrupting the slow-to-fast transition of muscle fibers compromises muscle growth postnatally and suggest that type IIB myosin heavy chain expression and its corresponding fiber type may be necessary for fiber maintenance, transition and hypertrophy in mice. The fact that forcing muscle metabolism toward a more oxidative phenotype can partially compensates for the lack of an intact Myh4 gene provides new avenues for attenuating the loss of fast-twitch fibers in aged or diseased muscles.
Collapse
Affiliation(s)
- Caiyun Zeng
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Hao Shi
- Meat Science and Muscle Biology Research Group, Virginia Tech, Department of Animal and Poultry Sciences, Blacksburg, VA, United States
| | - Laila T. Kirkpatrick
- Meat Science and Muscle Biology Research Group, Virginia Tech, Department of Animal and Poultry Sciences, Blacksburg, VA, United States
| | - Aymeric Ricome
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Sungkwon Park
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Jason M. Scheffler
- Meat Science and Muscle Biology Research Group, Virginia Tech, Department of Animal and Poultry Sciences, Blacksburg, VA, United States
| | - Kevin M. Hannon
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States
| | - Alan L. Grant
- Meat Science and Muscle Biology Research Group, Virginia Tech, Department of Animal and Poultry Sciences, Blacksburg, VA, United States
| | - David E. Gerrard
- Meat Science and Muscle Biology Research Group, Virginia Tech, Department of Animal and Poultry Sciences, Blacksburg, VA, United States
- *Correspondence: David E. Gerrard,
| |
Collapse
|
12
|
Hasan S, Asakawa S, Watabe S, Kinoshita S. Regulation of the Expression of the Myosin Heavy Chain (MYH) Gene myh14 in Zebrafish Development. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2021; 23:821-835. [PMID: 34490548 DOI: 10.1007/s10126-021-10066-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 08/24/2021] [Indexed: 06/13/2023]
Abstract
The human sarcomeric myosin heavy chain gene MYH14 contains an intronic microRNA, miR-499. Our previous studies demonstrated divergent genomic organization and expression patterns of myh14/miR-499 among teleosts; however, the regulatory mechanism is partly known. In this study, we report the regulation of myh14 expression in zebrafish, Danio rerio. Zebrafish myh14 has three paralogs, myh14-1, myh14-2, and myh14-3. Detailed promoter analysis suggested that a 5710-bp 5'-flanking region of myh14-1 and a 5641-bp region of myh14-3 contain a necessary regulatory region to recapitulate specific expression during embryonic development. The 5'-flanking region of zebrafish myh14-1 and its torafugu ortholog shared two distal and a single proximal conserved region. The two distal conserved regions had no effect on zebrafish myh14-1 expression, in contrast to torafugu expression, suggesting an alternative regulatory mechanism among the myh14 orthologs. Comparison among the 5'-flanking regions of the myh14 paralogs revealed two conserved regions. Deletion of these conserved regions significantly reduced the promoter activity of myh14-3 but had no effect on myh14-1, indicating different cis-regulatory mechanisms of myh14 paralogs. Loss of function of miR-499 resulted in a marked reduction in slow muscle fibers in embryonic development. Our study identified different cis-regulatory mechanisms controlling the expression of myh14/miR-499 and an indispensable role of miR-499 in muscle fiber-type specification in zebrafish.
Collapse
Affiliation(s)
- Sharmin Hasan
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-8657, Japan.
- Department of Biology and Chemistry, Texas A&M International University, 5201 University Blvd., Laredo, TX, 78041, USA.
| | - Shuichi Asakawa
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-8657, Japan
| | - Shugo Watabe
- School of Marine Bioscience, Kitasato University, Minami-ku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Shigeharu Kinoshita
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-8657, Japan
| |
Collapse
|
13
|
Rasmussen M, Jin JP. Troponin Variants as Markers of Skeletal Muscle Health and Diseases. Front Physiol 2021; 12:747214. [PMID: 34733179 PMCID: PMC8559874 DOI: 10.3389/fphys.2021.747214] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/01/2021] [Indexed: 12/21/2022] Open
Abstract
Ca2 +-regulated contractility is a key determinant of the quality of muscles. The sarcomeric myofilament proteins are essential players in the contraction of striated muscles. The troponin complex in the actin thin filaments plays a central role in the Ca2+-regulation of muscle contraction and relaxation. Among the three subunits of troponin, the Ca2+-binding subunit troponin C (TnC) is a member of the calmodulin super family whereas troponin I (TnI, the inhibitory subunit) and troponin T (TnT, the tropomyosin-binding and thin filament anchoring subunit) are striated muscle-specific regulatory proteins. Muscle type-specific isoforms of troponin subunits are expressed in fast and slow twitch fibers and are regulated during development and aging, and in adaptation to exercise or disuse. TnT also evolved with various alternative splice forms as an added capacity of muscle functional diversity. Mutations of troponin subunits cause myopathies. Owing to their physiological and pathological importance, troponin variants can be used as specific markers to define muscle quality. In this focused review, we will explore the use of troponin variants as markers for the fiber contents, developmental and differentiation states, contractile functions, and physiological or pathophysiological adaptations of skeletal muscle. As protein structure defines function, profile of troponin variants illustrates how changes at the myofilament level confer functional qualities at the fiber level. Moreover, understanding of the role of troponin modifications and mutants in determining muscle contractility in age-related decline of muscle function and in myopathies informs an approach to improve human health.
Collapse
Affiliation(s)
- Monica Rasmussen
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Jian-Ping Jin
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
14
|
Nord H, Kahsay A, Dennhag N, Pedrosa Domellöf F, von Hofsten J. Genetic compensation between Pax3 and Pax7 in zebrafish appendicular muscle formation. Dev Dyn 2021; 251:1423-1438. [PMID: 34435397 DOI: 10.1002/dvdy.415] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 08/18/2021] [Accepted: 08/18/2021] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Migrating muscle progenitors delaminate from the somite and subsequently form muscle tissue in distant anatomical regions such as the paired appendages, or limbs. In amniotes, this process requires a signaling cascade including the transcription factor paired box 3 (Pax3). RESULTS In this study, we found that, unlike in mammals, pax3a/3b double mutant zebrafish develop near to normal appendicular muscle. By analyzing numerous mutant combinations of pax3a, pax3b and pax7a, and pax7b, we determined that there is a feedback system and a compensatory mechanism between Pax3 and Pax7 in this developmental process, even though Pax7 alone is not required for appendicular myogenesis. pax3a/3b/7a/7b quadruple mutant developed muscle-less pectoral fins. CONCLUSIONS We found that Pax3 and Pax7 are redundantly required during appendicular myogenesis in zebrafish, where Pax7 is able to activate the same developmental programs as Pax3 in the premigratory progenitor cells.
Collapse
Affiliation(s)
- Hanna Nord
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Abraha Kahsay
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Nils Dennhag
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Fatima Pedrosa Domellöf
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden.,Department of Clinical Science, Ophthalmology, Umeå University, Umeå, Sweden
| | - Jonas von Hofsten
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| |
Collapse
|
15
|
Dennhag N, Liu JX, Nord H, von Hofsten J, Pedrosa Domellöf F. Absence of Desmin in Myofibers of the Zebrafish Extraocular Muscles. Transl Vis Sci Technol 2020; 9:1. [PMID: 32953241 PMCID: PMC7476663 DOI: 10.1167/tvst.9.10.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/14/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose To study the medial rectus (MR) muscle of zebrafish (Daniorerio) with respect to the pattern of distribution of desmin and its correlation to distinct types of myofibers and motor endplates. Methods The MRs of zebrafish were examined using confocal microscopy in whole-mount longitudinal specimens and in cross sections processed for immunohistochemistry with antibodies against desmin, myosin heavy chain isoforms, and innervation markers. Desmin patterns were correlated to major myofiber type and type of innervation. A total of 1382 myofibers in nine MR muscles were analyzed. Results Four distinct desmin immunolabeling patterns were found in the zebrafish MRs. Approximately a third of all slow myofibers lacked desmin, representing 8.5% of the total myofiber population. The adult zebrafish MR muscle displayed en grappe, en plaque, and multiterminal en plaque neuromuscular junctions (NMJs) with intricate patterns of desmin immunolabeling. Conclusions The MRs of zebrafish showed important similarities with the human extraocular muscles with regard to the pattern of desmin distribution and presence of the major types of NMJs and can be regarded as an adequate model to further study the role of desmin and the implications of heterogeneity in cytoskeletal protein composition. Translational Relevance The establishment of a zebrafish model to study the cytoskeleton in muscles that are particularly resistant to muscle disease opens new avenues to understand human myopathies and muscle dystrophies and may provide clues to new therapies.
Collapse
Affiliation(s)
- Nils Dennhag
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Jing-Xia Liu
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Hanna Nord
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Jonas von Hofsten
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Fatima Pedrosa Domellöf
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden.,Department of Clinical Science, Ophthalmology, Umeå University, Umeå, Sweden
| |
Collapse
|
16
|
Iwanaga R, Truong BT, Hsu JY, Lambert KA, Vyas R, Orlicky D, Shellman YG, Tan AC, Ceol C, Artinger KB. Loss of prdm1a accelerates melanoma onset and progression. Mol Carcinog 2020; 59:1052-1063. [PMID: 32562448 PMCID: PMC7864383 DOI: 10.1002/mc.23236] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/04/2020] [Indexed: 12/19/2022]
Abstract
Melanoma is an aggressive, deadly skin cancer derived from melanocytes, a neural crest cell derivative. Melanoma cells mirror the developmental program of neural crest cells in that they exhibit the same gene expression patterns and utilize similar cellular mechanisms, including increased cell proliferation, epithelial-mesenchymal transition, and migration. Here we studied the role of neural crest regulator PRDM1 in melanoma onset and progression. In development, Prdm1a functions to promote neural crest progenitor fate, and in melanoma, we found that PRDM1 has reduced copy number and is recurrently deleted in both zebrafish and humans. When examining expression of neural crest and melanocyte development genes, we show that sox10 progenitor expression is high in prdm1a-/- mutants, while more differentiated melanocyte markers are reduced, suggesting that normally Prdm1a is required for differentiation. Data mining of human melanoma datasets indicates that high PRDM1 expression in human melanoma is correlated with better patient survival and decreased PRDM1 expression is common in metastatic tumors. When one copy of prdm1a is lost in the zebrafish melanoma model Tg(mitfa:BRAFV600E );p53-/- ;prdm1a+/- , melanoma onset occurs more quickly, and the tumors that form have a larger area with increased expression of sox10. These data demonstrate a novel role for PRDM1 as a tumor suppressor in melanoma.
Collapse
Affiliation(s)
- Ritsuko Iwanaga
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Brittany T. Truong
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
- Human Medical Genetics & Genomics Graduate Program, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Jessica Y. Hsu
- Pharmacology Graduate Program, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Karoline A. Lambert
- Department of Dermatology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Rajesh Vyas
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - David Orlicky
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Yiqun G. Shellman
- Department of Dermatology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Aik-Choon Tan
- Division of Medical Oncology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Craig Ceol
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Kristin Bruk Artinger
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
17
|
miRNA-mRNA network regulation in the skeletal muscle fiber phenotype of chickens revealed by integrated analysis of miRNAome and transcriptome. Sci Rep 2020; 10:10619. [PMID: 32606372 PMCID: PMC7326969 DOI: 10.1038/s41598-020-67482-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 06/08/2020] [Indexed: 12/23/2022] Open
Abstract
Skeletal muscle fibers are primarily categorized into oxidative and glycolytic fibers, and the ratios of different myofiber types are important factors in determining livestock meat quality. However, the molecular mechanism for determining muscle fiber types in chickens was hardly understood. In this study, we used RNA sequencing to systematically compare mRNA and microRNA transcriptomes of the oxidative muscle sartorius (SART) and glycolytic muscle pectoralis major (PMM) of Chinese Qingyuan partridge chickens. Among the 44,705 identified mRNAs in the two types of muscles, 3,457 exhibited significantly different expression patterns, including 2,364 up-regulated and 1,093 down-regulated mRNAs in the SART. A total of 698 chicken miRNAs were identified, including 189 novel miRNAs, among which 67 differentially expressed miRNAs containing 42 up-regulated and 25 down-regulated miRNAs in the SART were identified. Furthermore, function enrichment showed that the differentially expressed mRNAs and miRNAs were involved in energy metabolism, muscle contraction, and calcium, peroxisome proliferator-activated receptor (PPAR), insulin and adipocytokine signaling. Using miRNA-mRNA integrated analysis, we identified several candidate miRNA-gene pairs that might affect muscle fiber performance, viz, gga-miR-499-5p/SOX6 and gga-miR-196-5p/CALM1, which were supported by target validation using the dual-luciferase reporter system. This study revealed a mass of candidate genes and miRNAs involved in muscle fiber type determination, which might help understand the molecular mechanism underlying meat quality traits in chickens.
Collapse
|
18
|
Hromowyk KJ, Talbot JC, Martin BL, Janssen PML, Amacher SL. Cell fusion is differentially regulated in zebrafish post-embryonic slow and fast muscle. Dev Biol 2020; 462:85-100. [PMID: 32165147 PMCID: PMC7225055 DOI: 10.1016/j.ydbio.2020.03.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 02/08/2020] [Accepted: 03/03/2020] [Indexed: 12/13/2022]
Abstract
Skeletal muscle fusion occurs during development, growth, and regeneration. To investigate how muscle fusion compares among different muscle cell types and developmental stages, we studied muscle cell fusion over time in wild-type, myomaker (mymk), and jam2a mutant zebrafish. Using live imaging, we show that embryonic myoblast elongation and fusion correlate tightly with slow muscle cell migration. In wild-type embryos, only fast muscle fibers are multinucleate, consistent with previous work showing that the cell fusion regulator gene mymk is specifically expressed throughout the embryonic fast muscle domain. However, by 3 weeks post-fertilization, slow muscle fibers also become multinucleate. At this late-larval stage, mymk is not expressed in muscle fibers, but is expressed in small cells near muscle fibers. Although previous work showed that both mymk and jam2a are required for embryonic fast muscle cell fusion, we observe that muscle force and function is almost normal in mymk and jam2a mutant embryos, despite the lack of fast muscle multinucleation. We show that genetic requirements change post-embryonically, with jam2a becoming much less important by late-larval stages and mymk now required for muscle fusion and growth in both fast and slow muscle cell types. Correspondingly, adult mymk mutants perform poorly in sprint and endurance tests compared to wild-type and jam2a mutants. We show that adult mymk mutant muscle contains small mononucleate myofibers with average myonuclear domain size equivalent to that in wild type adults. The mymk mutant fibers have decreased Laminin expression and increased numbers of Pax7-positive cells, suggesting that impaired fiber growth and active regeneration contribute to the muscle phenotype. Our findings identify several aspects of muscle fusion that change with time in slow and fast fibers as zebrafish develop beyond embryonic stages.
Collapse
Affiliation(s)
- Kimberly J Hromowyk
- Department of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA; Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH, 43210, USA; Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, 43210, USA
| | - Jared C Talbot
- Department of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA; Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH, 43210, USA.
| | - Brit L Martin
- Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH, 43210, USA; Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, 43210, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, 43210, USA
| | - Paul M L Janssen
- Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH, 43210, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, 43210, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Sharon L Amacher
- Department of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA; Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH, 43210, USA; Center for RNA Biology, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
19
|
Maire P, Dos Santos M, Madani R, Sakakibara I, Viaut C, Wurmser M. Myogenesis control by SIX transcriptional complexes. Semin Cell Dev Biol 2020; 104:51-64. [PMID: 32247726 DOI: 10.1016/j.semcdb.2020.03.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 02/07/2023]
Abstract
SIX homeoproteins were first described in Drosophila, where they participate in the Pax-Six-Eya-Dach (PSED) network with eyeless, eyes absent and dachsund to drive synergistically eye development through genetic and biochemical interactions. The role of the PSED network and SIX proteins in muscle formation in vertebrates was subsequently identified. Evolutionary conserved interactions with EYA and DACH proteins underlie the activity of SIX transcriptional complexes (STC) both during embryogenesis and in adult myofibers. Six genes are expressed throughout muscle development, in embryonic and adult proliferating myogenic stem cells and in fetal and adult post-mitotic myofibers, where SIX proteins regulate the expression of various categories of genes. In vivo, SIX proteins control many steps of muscle development, acting through feedforward mechanisms: in the embryo for myogenic fate acquisition through the direct control of Myogenic Regulatory Factors; in adult myofibers for their contraction/relaxation and fatigability properties through the control of genes involved in metabolism, sarcomeric organization and calcium homeostasis. Furthermore, during development and in the adult, SIX homeoproteins participate in the genesis and the maintenance of myofibers diversity.
Collapse
Affiliation(s)
- Pascal Maire
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014, Paris, France.
| | | | - Rouba Madani
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014, Paris, France
| | - Iori Sakakibara
- Research Center for Advanced Science and Technology, The University of Tokyo, Japan
| | - Camille Viaut
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014, Paris, France
| | - Maud Wurmser
- Department of Integrative Medical Biology (IMB), Umeå universitet, Sweden
| |
Collapse
|
20
|
Duran BODS, Dal-Pai-Silva M, Garcia de la Serrana D. Rainbow trout slow myoblast cell culture as a model to study slow skeletal muscle, and the characterization of mir-133 and mir-499 families as a case study. ACTA ACUST UNITED AC 2020; 223:jeb.216390. [PMID: 31871118 DOI: 10.1242/jeb.216390] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/17/2019] [Indexed: 12/14/2022]
Abstract
Muscle fibres are classified as fast, intermediate and slow. In vitro myoblast cell culture model from fast muscle is a very useful tool to study muscle growth and development; however, similar models for slow muscle do not exist. Owing to the compartmentalization of fish muscle fibres, we have developed a slow myoblast cell culture for rainbow trout (Oncorhynchus mykiss). Slow and fast muscle-derived myoblasts have similar morphology, but with differential expression of slow muscle markers such as slow myhc, sox6 and pgc-1α We also characterized the mir-133 and mir-499 microRNA families in trout slow and fast myoblasts as a case study during myogenesis and in response to electrostimulation. Three mir-133 (a-1a, a-1b and a-2) and four mir-499 (aa, ab, ba and bb) paralogues were identified for rainbow trout and named base on their phylogenetic relationship to zebrafish and Atlantic salmon orthologues. Omy-mir-499ab and omy-mir-499bb had 0.6 and 0.5-fold higher expression in slow myoblasts compared with fast myoblasts, whereas mir-133 duplicates had similar levels in both phenotypes and little variation during development. Slow myoblasts also showed increased expression for omy-mir-499b paralogues in response to chronic electrostimulation (7-fold increase for omy-mir-499ba and 2.5-fold increase for omy-mir-499bb). The higher expression of mir-499 paralogues in slow myoblasts suggests a role in phenotype determination, while the lack of significant differences of mir-133 copies during culture development might indicate a different role in fish compared with mammals. We have also found signs of sub-functionalization of mir-499 paralogues after electrostimulation, with omy-mir-499b copies more responsive to electrical signals.
Collapse
Affiliation(s)
- Bruno Oliveira da Silva Duran
- São Paulo State University (UNESP), Institute of Biosciences, Department of Morphology, Botucatu 18618-689, São Paulo, Brazil
| | - Maeli Dal-Pai-Silva
- São Paulo State University (UNESP), Institute of Biosciences, Department of Morphology, Botucatu 18618-689, São Paulo, Brazil
| | - Daniel Garcia de la Serrana
- University of St Andrews, Scottish Oceans Institute, School of Biology, St Andrews, Fife KY16 8LB, UK.,University of Barcelona, Faculty of Biology, Department of Cell Biology, Physiology and Immunology, 08028 Barcelona, Spain
| |
Collapse
|
21
|
Nord H, Dennhag N, Tydinger H, von Hofsten J. The zebrafish HGF receptor met controls migration of myogenic progenitor cells in appendicular development. PLoS One 2019; 14:e0219259. [PMID: 31287821 PMCID: PMC6615617 DOI: 10.1371/journal.pone.0219259] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 06/20/2019] [Indexed: 12/16/2022] Open
Abstract
The hepatocyte growth factor receptor C-met plays an important role in cellular migration, which is crucial for many developmental processes as well as for cancer cell metastasis. C-met has been linked to the development of mammalian appendicular muscle, which are derived from migrating muscle progenitor cells (MMPs) from within the somite. Mammalian limbs are homologous to the teleost pectoral and pelvic fins. In this study we used Crispr/Cas9 to mutate the zebrafish met gene and found that the MMP derived musculature of the paired appendages was severely affected. The mutation resulted in a reduced muscle fibre number, in particular in the pectoral abductor, and in a disturbed pectoral fin function. Other MMP derived muscles, such as the sternohyoid muscle and posterior hypaxial muscle were also affected in met mutants. This indicates that the role of met in MMP function and appendicular myogenesis is conserved within vertebrates.
Collapse
Affiliation(s)
- Hanna Nord
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Nils Dennhag
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Hanna Tydinger
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Jonas von Hofsten
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
- Umeå Centre for Molecular Medicine, Umeå University, Umeå, Sweden
- * E-mail:
| |
Collapse
|
22
|
Wang X, Chen X, Huang Z, Chen D, Yu B, Chen H, He J, Zheng P, Luo J, Yu J, Luo Y. Effects of active immunization against porcine Sox6 on meat quality and myosin heavy chain isoform expression in growing-finishing pigs. Anim Biotechnol 2018; 30:260-266. [PMID: 30264664 DOI: 10.1080/10495398.2018.1481416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
A feeding trial for 91 days was conducted to investigate effects of active immunization against porcine Sox6 (pSox6) on meat quality and myosin heavy chain (MyHC) isoform expression in growing-finishing pigs. Twenty-four castrated Duroc × Landrace × Yarkshire pigs were randomly divided into three groups: (1) Control group; (2) 1 mg/head pSox6 active immunity group; (3) 4 mg/head pSox6 active immunity group (4 mg/head group). The results showed that pigs in 4 mg/head group had a greater a* (Redness) and a higher marbling score, while no significant effect was observed in L* (Lightness), b* (Yellowness), intramuscular fat and cooking loss. Muscle succinic dehydrogenase activity in pSox6 active immunization groups was significantly increased, and muscle lactate dehydrogenase activity was significantly reduced. Meanwhile, active immunization against pSox6 upregulated the mRNA expression of MyHC I, while no effect was observed on the mRNA expressions of MyHC IIa, MyHC IIx, MyHC IIb. In addition, pigs in the 4 mg/head group exhibited lower Sox6 mRNA level and higher MyHC I protein level, while no significant influence was observed on MyHC IIb protein level. Together, our data imply that active immunization against pSox6 could improve the pork quality and promote the MyHC I expression in growing-finishing pigs.
Collapse
Affiliation(s)
- Xiaoqin Wang
- a Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education , Institute of Animal Nutrition, Sichuan Agricultural University , Chengdu , Sichuan , PR China
| | - Xiaoling Chen
- a Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education , Institute of Animal Nutrition, Sichuan Agricultural University , Chengdu , Sichuan , PR China
| | - Zhiqing Huang
- a Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education , Institute of Animal Nutrition, Sichuan Agricultural University , Chengdu , Sichuan , PR China
| | - Daiwen Chen
- a Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education , Institute of Animal Nutrition, Sichuan Agricultural University , Chengdu , Sichuan , PR China
| | - Bing Yu
- a Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education , Institute of Animal Nutrition, Sichuan Agricultural University , Chengdu , Sichuan , PR China
| | - Hong Chen
- b College of Food Science , Sichuan Agricultural University , Yaan , Sichuan , PR China
| | - Jun He
- a Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education , Institute of Animal Nutrition, Sichuan Agricultural University , Chengdu , Sichuan , PR China
| | - Ping Zheng
- a Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education , Institute of Animal Nutrition, Sichuan Agricultural University , Chengdu , Sichuan , PR China
| | - Junqiu Luo
- a Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education , Institute of Animal Nutrition, Sichuan Agricultural University , Chengdu , Sichuan , PR China
| | - Jie Yu
- a Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education , Institute of Animal Nutrition, Sichuan Agricultural University , Chengdu , Sichuan , PR China
| | - Yuheng Luo
- a Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education , Institute of Animal Nutrition, Sichuan Agricultural University , Chengdu , Sichuan , PR China
| |
Collapse
|
23
|
Xenopus SOX5 enhances myogenic transcription indirectly through transrepression. Dev Biol 2018; 442:262-275. [PMID: 30071218 DOI: 10.1016/j.ydbio.2018.07.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/16/2018] [Accepted: 07/28/2018] [Indexed: 02/06/2023]
Abstract
In anamniotes, somite compartimentalization in the lateral somitic domain leads simultaneously to myotome and dermomyotome formation. In the myotome, Xenopus Sox5 is co-expressed with Myod1 in the course of myogenic differentiation. Here, we studied the function of Sox5 using a Myod1-induced myogenic transcription assay in pluripotent cells of animal caps. We found that Sox5 enhances myogenic transcription of muscle markers Des, Actc1, Ckm and MyhE3. The use of chimeric transactivating or transrepressive Sox5 proteins indicates that Sox5 acts as a transrepressor and indirectly stimulates myogenic transcription except for the slow muscle-specific genes Myh7L, Myh7S, Myl2 and Tnnc1. We showed that this role is shared by Sox6, which is structurally similar to Sox5, both belonging to the SoxD subfamily of transcription factors. Moreover, Sox5 can antagonize the inhibitory function of Meox2 on myogenic differentiation. Meox2 which is a dermomyotome marker, represses myogenic transcription in Myod-induced myogenic transcription assay and in Nodal5-induced mesoderm from animal cap assay. The inhibitory function of Meox2 and the pro-myogenic function of Sox5 were confirmed during Xenopus normal development by the use of translation-blocking oligomorpholinos and dexamethasone inducible chimeric Sox5 and Meox2 proteins. We have therefore identified a new function for SoxD proteins in muscle cells, which can indirectly enhance myogenic transcription through transrepression, in addition to the previously identified function as a direct repressor of slow muscle-specific genes.
Collapse
|
24
|
Kinoshita S, Ceyhun SB, Md A, Siddique BS, Akolkar DB, Asakawa S, Watabe S. Promoter analysis of the fish gene of slow/cardiac-type myosin heavy chain implicated in specification of muscle fiber types. FISH PHYSIOLOGY AND BIOCHEMISTRY 2018; 44:679-691. [PMID: 29349631 DOI: 10.1007/s10695-018-0463-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 01/03/2018] [Indexed: 06/07/2023]
Abstract
Vertebrate skeletal muscles consist of heterogeneous tissues containing various types of muscle fibers, where specification of the fiber type is crucial for muscle development. Fish are an attractive experimental model to study the mechanisms of such fiber type specification because of the separated localization of slow and fast muscles in the trunk myotome. We examined regulation of expression of the torafugu gene of slow/cardiac-type myosin heavy chain, MYH M5 , and isolated an operational promoter in order to force its tissue-specific expression across different fish species via the transgenic approach in zebrafish and medaka. This promoter activity was observed in adaxial cell-derived superficial slow muscle fibers under the control of a hedgehog signal. We also uncovered coordinated expression of MYH M5 and Sox6b, which is an important transcriptional repressor for specification of muscle fiber types and participates in hedgehog signaling. Sequence comparison in the 5'-flanking region identified three conserved regions, CSR1-CSR3, between torafugu MYH M5 and its zebrafish ortholog. Analysis of deletion mutants showed that CSR1 significantly stimulates gene expression in slow muscle fibers. In contrast, deletion of CSR3 resulted in ectopic expression of a reporter gene in fast muscle fibers. CSR3 was found to contain a putative Sox family protein-binding site. These results indicate that the dual mechanism causing inhibition in fast muscle fibers and activation in slow muscle fibers is essential for slow muscle fiber-specific gene expression in fish.
Collapse
Affiliation(s)
- Shigeharu Kinoshita
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-8657, Japan.
| | | | - Asaduzzamann Md
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-8657, Japan
| | - Bhuiyan Sharmin Siddique
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-8657, Japan
| | - Dadasaheb B Akolkar
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-8657, Japan
| | - Shuichi Asakawa
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-8657, Japan
| | - Shugo Watabe
- School of Marine Biosciences, Kitasato University, Minami, Sagamihara, Kanagawa, 252-0373, Japan
| |
Collapse
|
25
|
Wei W, Li B, Liu K, Jiang A, Dong C, Jia C, Chen J, Liu H, Wu W. Identification of key microRNAs affecting drip loss in porcine longissimus dorsi by RNA-Seq. Gene 2018; 647:276-282. [DOI: 10.1016/j.gene.2018.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/28/2017] [Accepted: 01/02/2018] [Indexed: 12/27/2022]
|
26
|
Kimelman D, Smith NL, Lai JKH, Stainier DYR. Regulation of posterior body and epidermal morphogenesis in zebrafish by localized Yap1 and Wwtr1. eLife 2017; 6:e31065. [PMID: 29283341 PMCID: PMC5773182 DOI: 10.7554/elife.31065] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 12/20/2017] [Indexed: 12/11/2022] Open
Abstract
The vertebrate embryo undergoes a series of dramatic morphological changes as the body extends to form the complete anterior-posterior axis during the somite-forming stages. The molecular mechanisms regulating these complex processes are still largely unknown. We show that the Hippo pathway transcriptional coactivators Yap1 and Wwtr1 are specifically localized to the presumptive epidermis and notochord, and play a critical and unexpected role in posterior body extension by regulating Fibronectin assembly underneath the presumptive epidermis and surrounding the notochord. We further find that Yap1 and Wwtr1, also via Fibronectin, have an essential role in the epidermal morphogenesis necessary to form the initial dorsal and ventral fins, a process previously thought to involve bending of an epithelial sheet, but which we now show involves concerted active cell movement. Our results reveal how the Hippo pathway transcriptional program, localized to two specific tissues, acts to control essential morphological events in the vertebrate embryo.
Collapse
Affiliation(s)
- David Kimelman
- Department of BiochemistryUniversity of WashingtonSeattleUnited States
| | - Natalie L Smith
- Department of BiochemistryUniversity of WashingtonSeattleUnited States
| | - Jason Kuan Han Lai
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Didier YR Stainier
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| |
Collapse
|
27
|
Taglietti V, Maroli G, Cermenati S, Monteverde S, Ferrante A, Rossi G, Cossu G, Beltrame M, Messina G. Nfix Induces a Switch in Sox6 Transcriptional Activity to Regulate MyHC-I Expression in Fetal Muscle. Cell Rep 2017; 17:2354-2366. [PMID: 27880909 PMCID: PMC5149531 DOI: 10.1016/j.celrep.2016.10.082] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/08/2016] [Accepted: 10/24/2016] [Indexed: 02/01/2023] Open
Abstract
Sox6 belongs to the Sox gene family and plays a pivotal role in fiber type differentiation, suppressing transcription of slow-fiber-specific genes during fetal development. Here, we show that Sox6 plays opposite roles in MyHC-I regulation, acting as a positive and negative regulator of MyHC-I expression during embryonic and fetal myogenesis, respectively. During embryonic myogenesis, Sox6 positively regulates MyHC-I via transcriptional activation of Mef2C, whereas during fetal myogenesis, Sox6 requires and cooperates with the transcription factor Nfix in repressing MyHC-I expression. Mechanistically, Nfix is necessary for Sox6 binding to the MyHC-I promoter and thus for Sox6 repressive function, revealing a key role for Nfix in driving Sox6 activity. This feature is evolutionarily conserved, since the orthologs Nfixa and Sox6 contribute to repression of the slow-twitch phenotype in zebrafish embryos. These data demonstrate functional cooperation between Sox6 and Nfix in regulating MyHC-I expression during prenatal muscle development. Sox6 has opposite roles in MyHC-I regulation during embryonic and fetal myogenesis In embryonic muscle, Sox6 enhances MyHC-I expression via regulation of Mef2C In fetal muscle, Nfix is required for Sox6-mediated repression of MyHC-I The Sox6 and Nfixa orthologs cooperate in repressing smyhc1 in zebrafish
Collapse
Affiliation(s)
| | - Giovanni Maroli
- Department of Biosciences, University of Milan, Milan 20133, Italy
| | - Solei Cermenati
- Department of Biosciences, University of Milan, Milan 20133, Italy
| | | | - Andrea Ferrante
- Department of Biosciences, University of Milan, Milan 20133, Italy
| | - Giuliana Rossi
- Department of Biosciences, University of Milan, Milan 20133, Italy
| | - Giulio Cossu
- Department of Biosciences, University of Milan, Milan 20133, Italy; Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Oxford Road, M13 9PL Manchester, UK
| | - Monica Beltrame
- Department of Biosciences, University of Milan, Milan 20133, Italy
| | | |
Collapse
|
28
|
Roy SD, Williams VC, Pipalia TG, Li K, Hammond CL, Knappe S, Knight RD, Hughes SM. Myotome adaptability confers developmental robustness to somitic myogenesis in response to fibre number alteration. Dev Biol 2017; 431:321-335. [PMID: 28887016 PMCID: PMC5667637 DOI: 10.1016/j.ydbio.2017.08.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 05/22/2017] [Accepted: 08/26/2017] [Indexed: 12/31/2022]
Abstract
Balancing the number of stem cells and their progeny is crucial for tissue development and repair. Here we examine how cell numbers and overall muscle size are tightly regulated during zebrafish somitic muscle development. Muscle stem/precursor cell (MPCs) expressing Pax7 are initially located in the dermomyotome (DM) external cell layer, adopt a highly stereotypical distribution and thereafter a proportion of MPCs migrate into the myotome. Regional variations in the proliferation and terminal differentiation of MPCs contribute to growth of the myotome. To probe the robustness of muscle size control and spatiotemporal regulation of MPCs, we compared the behaviour of wild type (wt) MPCs with those in mutant zebrafish that lack the muscle regulatory factor Myod. Myodfh261 mutants form one third fewer multinucleate fast muscle fibres than wt and show a significant expansion of the Pax7+ MPC population in the DM. Subsequently, myodfh261 mutant fibres generate more cytoplasm per nucleus, leading to recovery of muscle bulk. In addition, relative to wt siblings, there is an increased number of MPCs in myodfh261 mutants and these migrate prematurely into the myotome, differentiate and contribute to the hypertrophy of existing fibres. Thus, homeostatic reduction of the excess MPCs returns their number to normal levels, but fibre numbers remain low. The GSK3 antagonist BIO prevents MPC migration into the deep myotome, suggesting that canonical Wnt pathway activation maintains the DM in zebrafish, as in amniotes. BIO does not, however, block recovery of the myodfh261 mutant myotome, indicating that homeostasis acts on fibre intrinsic growth to maintain muscle bulk. The findings suggest the existence of a critical window for early fast fibre formation followed by a period in which homeostatic mechanisms regulate myotome growth by controlling fibre size. The feedback controls we reveal in muscle help explain the extremely precise grading of myotome size along the body axis irrespective of fish size, nutrition and genetic variation and may form a paradigm for wider matching of organ size. A critical window for early muscle fibre formation is proposed. Fish lacking MyoD1 form fewer muscle fibres, but have more myogenic stem cells. Stem cell numbers rapidly return to normal during subsequent development. GSK3 activity promotes and MyoD1 delays myoblast migration into the myotome. Compensatory fibre size increase ensures robustness of overall muscle size.
Collapse
Affiliation(s)
- Shukolpa D Roy
- Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Victoria C Williams
- Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Tapan G Pipalia
- Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Kuoyu Li
- Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Christina L Hammond
- Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Stefanie Knappe
- Division of Craniofacial Development and Stem Cell Biology, Guy's Hospital, King's College London, UK
| | - Robert D Knight
- Division of Craniofacial Development and Stem Cell Biology, Guy's Hospital, King's College London, UK
| | - Simon M Hughes
- Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK.
| |
Collapse
|
29
|
Duan Y, Li F, Tan B, Yao K, Yin Y. Metabolic control of myofibers: promising therapeutic target for obesity and type 2 diabetes. Obes Rev 2017; 18:647-659. [PMID: 28391659 DOI: 10.1111/obr.12530] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 01/13/2017] [Accepted: 01/26/2017] [Indexed: 02/02/2023]
Abstract
Mammalian skeletal muscles are composed of two major fibre types (I and II) that differ in terms of size, metabolism and contractile properties. In general, slow-twitch type I fibres are rich in mitochondria and have a greater insulin sensitivity than fast-twitch type II skeletal muscles. Although not widely appreciated, a forced induction of the slow skeletal muscle phenotype may inhibit the progress of obesity and diabetes. This potentially forms the basis for targeting slow/oxidative myofibers in the treatment of obesity. In this context, a better understanding of the molecular basis of fibre-type specification and plasticity may help to identify potential therapeutic targets for obesity and diabetes.
Collapse
Affiliation(s)
- Yehui Duan
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Fengna Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China.,Hunan Co-Innovation Center of Safety Animal Production, CICSAP, Changsha, China
| | - Bie Tan
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Kang Yao
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China.,Hunan Co-Innovation Center of Safety Animal Production, CICSAP, Changsha, China
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China.,Laboratory of Animal Nutrition and Human Health, School of Biology, Hunan Normal University, Changsha, China
| |
Collapse
|
30
|
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs that negatively regulate gene expression of target messenger RNAs (mRNAs) and miRNAs have been proven to play vital roles in skeletal muscle development. The miRNA-499-5p has been reported to be negatively related with the expression of Sox6, a critical transcription factor for the maintenance of fast-twitch skeletal muscle. In this study, we amplified a length of 2012-bp mRNA that contains a 1512-bp porcine Sox6 (pSox6) 3'UTR from skeletal muscle of a Duroc×Landrace×Yorkshire pig. By luciferase reporter assay we verified that pSox6 is a target of miR-499-5p. In extensor digitorum longus and Soleus muscles of pigs, the expression levels of miR-499-5p and pSox6 mRNA were also inversely correlated. Besides, overexpression of miR-499-5p in porcine satellite cells promoted the expression of MyHC I and MyHC IIa mRNA, along with a reduction of pSox6 mRNA. Taken together, these results indicate that miR-499-5p may facilitate the oxidative myofibers formation by downregulating pSox6 expression.
Collapse
|
31
|
Abstract
Skeletal muscle performs an essential function in human physiology with defects in genes encoding a variety of cellular components resulting in various types of inherited muscle disorders. Muscular dystrophies (MDs) are a severe and heterogeneous type of human muscle disease, manifested by progressive muscle wasting and degeneration. The disease pathogenesis and therapeutic options for MDs have been investigated for decades using rodent models, and considerable knowledge has been accumulated on the cause and pathogenetic mechanisms of this group of human disorders. However, due to some differences between disease severity and progression, what is learned in mammalian models does not always transfer to humans, prompting the desire for additional and alternative models. More recently, zebrafish have emerged as a novel and robust animal model for the study of human muscle disease. Zebrafish MD models possess a number of distinct advantages for modeling human muscle disorders, including the availability and ease of generating mutations in homologous disease-causing genes, the ability to image living muscle tissue in an intact animal, and the suitability of zebrafish larvae for large-scale chemical screens. In this chapter, we review the current understanding of molecular and cellular mechanisms involved in MDs, the process of myogenesis in zebrafish, and the structural and functional characteristics of zebrafish larval muscles. We further discuss the insights gained from the key zebrafish MD models that have been so far generated, and we summarize the attempts that have been made to screen for small molecules inhibitors of the dystrophic phenotypes using these models. Overall, these studies demonstrate that zebrafish is a useful in vivo system for modeling aspects of human skeletal muscle disorders. Studies using these models have contributed both to the understanding of the pathogenesis of muscle wasting disorders and demonstrated their utility as highly relevant models to implement therapeutic screening regimens.
Collapse
Affiliation(s)
- M Li
- Monash University, Clayton, VIC, Australia
| | - K J Hromowyk
- The Ohio State University, Columbus, OH, United States
| | - S L Amacher
- The Ohio State University, Columbus, OH, United States
| | - P D Currie
- Monash University, Clayton, VIC, Australia
| |
Collapse
|
32
|
An CI, Ichihashi Y, Peng J, Sinha NR, Hagiwara N. Transcriptome Dynamics and Potential Roles of Sox6 in the Postnatal Heart. PLoS One 2016; 11:e0166574. [PMID: 27832192 PMCID: PMC5104335 DOI: 10.1371/journal.pone.0166574] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 10/31/2016] [Indexed: 01/20/2023] Open
Abstract
The postnatal heart undergoes highly coordinated developmental processes culminating in the complex physiologic properties of the adult heart. The molecular mechanisms of postnatal heart development remain largely unexplored despite their important clinical implications. To gain an integrated view of the dynamic changes in gene expression during postnatal heart development at the organ level, time-series transcriptome analyses of the postnatal hearts of neonatal through adult mice (P1, P7, P14, P30, and P60) were performed using a newly developed bioinformatics pipeline. We identified functional gene clusters by principal component analysis with self-organizing map clustering which revealed organized, discrete gene expression patterns corresponding to biological functions associated with the neonatal, juvenile and adult stages of postnatal heart development. Using weighted gene co-expression network analysis with bootstrap inference for each of these functional gene clusters, highly robust hub genes were identified which likely play key roles in regulating expression of co-expressed, functionally linked genes. Additionally, motivated by the role of the transcription factor Sox6 in the functional maturation of skeletal muscle, the role of Sox6 in the postnatal maturation of cardiac muscle was investigated. Differentially expressed transcriptome analyses between Sox6 knockout (KO) and control hearts uncovered significant upregulation of genes involved in cell proliferation at postnatal day 7 (P7) in the Sox6 KO heart. This result was validated by detecting mitotically active cells in the P7 Sox6 KO heart. The current report provides a framework for the complex molecular processes of postnatal heart development, thus enabling systematic dissection of the developmental regression observed in the stressed and failing adult heart.
Collapse
Affiliation(s)
- Chung-Il An
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California Davis, Davis, California, United States of America
- * E-mail: (CA); (YI); (NH)
| | - Yasunori Ichihashi
- Department of Plant Biology, University of California Davis, Davis, California, United States of America
- * E-mail: (CA); (YI); (NH)
| | - Jie Peng
- Department of Statistics, University of California Davis, Davis, California, United States of America
| | - Neelima R. Sinha
- Department of Plant Biology, University of California Davis, Davis, California, United States of America
| | - Nobuko Hagiwara
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California Davis, Davis, California, United States of America
- * E-mail: (CA); (YI); (NH)
| |
Collapse
|
33
|
Omairi S, Matsakas A, Degens H, Kretz O, Hansson KA, Solbrå AV, Bruusgaard JC, Joch B, Sartori R, Giallourou N, Mitchell R, Collins-Hooper H, Foster K, Pasternack A, Ritvos O, Sandri M, Narkar V, Swann JR, Huber TB, Patel K. Enhanced exercise and regenerative capacity in a mouse model that violates size constraints of oxidative muscle fibres. eLife 2016; 5. [PMID: 27494364 PMCID: PMC4975572 DOI: 10.7554/elife.16940] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/19/2016] [Indexed: 12/28/2022] Open
Abstract
A central tenet of skeletal muscle biology is the existence of an inverse relationship between the oxidative fibre capacity and its size. However, robustness of this relationship is unknown. We show that superimposition of Estrogen-related receptor gamma (Errγ) on the myostatin (Mtn) mouse null background (Mtn-/-/ErrγTg/+) results in hypertrophic muscle with a high oxidative capacity thus violating the inverse relationship between fibre size and oxidative capacity. We also examined the canonical view that oxidative muscle phenotype positively correlate with Satellite cell number, the resident stem cells of skeletal muscle. Surprisingly, hypertrophic fibres from Mtn-/-/ErrγTg/+ mouse showed satellite cell deficit which unexpectedly did not affect muscle regeneration. These observations 1) challenge the concept of a constraint between fibre size and oxidative capacity and 2) indicate the important role of the microcirculation in the regenerative capacity of a muscle even when satellite cell numbers are reduced. DOI:http://dx.doi.org/10.7554/eLife.16940.001
Collapse
Affiliation(s)
- Saleh Omairi
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | | | - Hans Degens
- School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom.,Lithuanian Sports University, Kaunas, Lithuania
| | - Oliver Kretz
- Renal Division, University Medical Center Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kenth-Arne Hansson
- Centre for Integrative Neuroplasticity, Department of Biosciences, University of Oslo, Oslo, Norway
| | - Andreas Våvang Solbrå
- Centre for Integrative Neuroplasticity, Department of Biosciences, University of Oslo, Oslo, Norway.,Department of Physics, University of Oslo, Oslo, Norway
| | - Jo C Bruusgaard
- Centre for Integrative Neuroplasticity, Department of Biosciences, University of Oslo, Oslo, Norway.,Department of Health Sciences, Kristiania University College, Oslo, Norway
| | - Barbara Joch
- Department of Neuroanatomy, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Roberta Sartori
- Venetian Institute of Molecular Medicine, University of Padua, Padua, Italy
| | - Natasa Giallourou
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Robert Mitchell
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | | | - Keith Foster
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Arja Pasternack
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Olli Ritvos
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Marco Sandri
- Venetian Institute of Molecular Medicine, University of Padua, Padua, Italy
| | - Vihang Narkar
- Institute of Molecular Medicine, University of Health Science Center, Houston, Texas
| | - Jonathan R Swann
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Tobias B Huber
- Renal Division, University Medical Center Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,BIOSS Center for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Houston, Texas.,FRIAS, Freiburg Institute for Advanced Studies and Center for Biological System Analysis ZBSA, Freiburg, Germany
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, United Kingdom.,FRIAS, Freiburg Institute for Advanced Studies and Center for Biological System Analysis ZBSA, Freiburg, Germany
| |
Collapse
|
34
|
Liu X, Trakooljul N, Hadlich F, Muráni E, Wimmers K, Ponsuksili S. MicroRNA-mRNA regulatory networking fine-tunes the porcine muscle fiber type, muscular mitochondrial respiratory and metabolic enzyme activities. BMC Genomics 2016; 17:531. [PMID: 27485725 PMCID: PMC4970254 DOI: 10.1186/s12864-016-2850-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 06/20/2016] [Indexed: 02/02/2023] Open
Abstract
Background MicroRNAs (miRNAs) are small non-coding RNAs that play critical roles in diverse biological processes via regulation of gene expression including in skeletal muscles. In the current study, miRNA expression profile was investigated in longissimus muscle biopsies of malignant hyperthermia syndrome-negative Duroc and Pietrain pigs with distinct muscle metabolic properties in order to explore the regulatory role of miRNAs related to mitochondrial respiratory activity and metabolic enzyme activity in skeletal muscle. Results A comparative analysis of the miRNA expression profile between Duroc and Pietrain pigs was performed, followed by integration with mRNA profiles based on their pairwise correlation and computational target prediction. The identified target genes were enriched in protein ubiquitination pathway, stem cell pluripotency and geranylgeranyl diphosphate biosynthesis, as well as skeletal and muscular system development. Next, we analyzed the correlation between individual miRNAs and phenotypical traits including muscle fiber type, mitochondrial respiratory activity, metabolic enzyme activity and adenosine phosphate concentrations, and constructed the regulatory miRNA-mRNA networks associated with energy metabolism. It is noteworthy that miR-25 targeting BMPR2 and IRS1, miR-363 targeting USP24, miR-28 targeting HECW2 and miR-210 targeting ATP5I, ME3, MTCH1 and CPT2 were highly associated with slow-twitch oxidative fibers, fast-twitch oxidative fibers, ADP and ATP concentration suggesting an essential role of the miRNA-mRNA regulatory networking in modulating the mitochondrial energy expenditure in the porcine muscle. In the identified miRNA-mRNA network, a tight relationship between mitochondrial and ubiquitin proteasome system at the level of gene expression was observed. It revealed a link between these two systems contributing to energy metabolism of skeletal muscle under physiological conditions. Conclusions We assembled miRNA-mRNA regulatory networks based on divergent muscle properties between different pig breeds and further with the correlation analysis of expressed genes and phenotypic measurements. These complex networks relate to muscle fiber type, metabolic enzyme activity and ATP production and may contribute to divergent muscle phenotypes by fine-tuning the expression of genes. Altogether, the results provide an insight into a regulatory role of miRNAs in muscular energy metabolisms and may have an implication on meat quality and production. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2850-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xuan Liu
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Nares Trakooljul
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Frieder Hadlich
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Eduard Muráni
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Klaus Wimmers
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Siriluck Ponsuksili
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| |
Collapse
|
35
|
Güth R, Chaidez A, Samanta MP, Unguez GA. Properties of skeletal muscle in the teleost Sternopygus macrurus are unaffected by short-term electrical inactivity. Physiol Genomics 2016; 48:699-710. [PMID: 27449658 DOI: 10.1152/physiolgenomics.00068.2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 07/18/2016] [Indexed: 11/22/2022] Open
Abstract
Skeletal muscle is distinguished from other tissues on the basis of its shape, biochemistry, and physiological function. Based on mammalian studies, fiber size, fiber types, and gene expression profiles are regulated, in part, by the electrical activity exerted by the nervous system. To address whether similar adaptations to changes in electrical activity in skeletal muscle occur in teleosts, we studied these phenotypic properties of ventral muscle in the electric fish Sternopygus macrurus following 2 and 5 days of electrical inactivation by spinal transection. Our data show that morphological and biochemical properties of skeletal muscle remained largely unchanged after these treatments. Specifically, the distribution of type I and type II muscle fibers and the cross-sectional areas of these fiber types observed in control fish remained unaltered after each spinal transection survival period. This response to electrical inactivation was generally reflected at the transcript level in real-time PCR and RNA-seq data by showing little effect on the transcript levels of genes associated with muscle fiber type differentiation and plasticity, the sarcomere complex, and pathways implicated in the regulation of muscle fiber size. Data from this first study characterizing the acute influence of neural activity on muscle mass and sarcomere gene expression in a teleost are discussed in the context of comparative studies in mammalian model systems and vertebrate species from different lineages.
Collapse
Affiliation(s)
- Robert Güth
- Department of Biology, New Mexico State University, Las Cruces, New Mexico; and
| | - Alexander Chaidez
- Department of Biology, New Mexico State University, Las Cruces, New Mexico; and
| | | | - Graciela A Unguez
- Department of Biology, New Mexico State University, Las Cruces, New Mexico; and
| |
Collapse
|
36
|
Talbot J, Maves L. Skeletal muscle fiber type: using insights from muscle developmental biology to dissect targets for susceptibility and resistance to muscle disease. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2016; 5:518-34. [PMID: 27199166 DOI: 10.1002/wdev.230] [Citation(s) in RCA: 261] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 01/14/2016] [Accepted: 01/16/2016] [Indexed: 12/13/2022]
Abstract
Skeletal muscle fibers are classified into fiber types, in particular, slow twitch versus fast twitch. Muscle fiber types are generally defined by the particular myosin heavy chain isoforms that they express, but many other components contribute to a fiber's physiological characteristics. Skeletal muscle fiber type can have a profound impact on muscle diseases, including certain muscular dystrophies and sarcopenia, the aging-induced loss of muscle mass and strength. These findings suggest that some muscle diseases may be treated by shifting fiber type characteristics either from slow to fast, or fast to slow phenotypes, depending on the disease. Recent studies have begun to address which components of muscle fiber types mediate their susceptibility or resistance to muscle disease. However, for many diseases it remains largely unclear why certain fiber types are affected. A substantial body of work has revealed molecular pathways that regulate muscle fiber type plasticity and early developmental muscle fiber identity. For instance, recent studies have revealed many factors that regulate muscle fiber type through modulating the activity of the muscle regulatory transcription factor MYOD1. Future studies of muscle fiber type development in animal models will continue to enhance our understanding of factors and pathways that may provide therapeutic targets to treat muscle diseases. WIREs Dev Biol 2016, 5:518-534. doi: 10.1002/wdev.230 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Jared Talbot
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, USA
| | - Lisa Maves
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA.,Department of Pediatrics, University of Washington, Seattle, WA, USA
| |
Collapse
|
37
|
|
38
|
Pipalia TG, Koth J, Roy SD, Hammond CL, Kawakami K, Hughes SM. Cellular dynamics of regeneration reveals role of two distinct Pax7 stem cell populations in larval zebrafish muscle repair. Dis Model Mech 2016; 9:671-84. [PMID: 27149989 PMCID: PMC4920144 DOI: 10.1242/dmm.022251] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 04/27/2016] [Indexed: 12/25/2022] Open
Abstract
Heterogeneity of stem cells or their niches is likely to influence tissue regeneration. Here we reveal stem/precursor cell diversity during wound repair in larval zebrafish somitic body muscle using time-lapse 3D confocal microscopy on reporter lines. Skeletal muscle with incision wounds rapidly regenerates both slow and fast muscle fibre types. A swift immune response is followed by an increase in cells at the wound site, many of which express the muscle stem cell marker Pax7. Pax7(+) cells proliferate and then undergo terminal differentiation involving Myogenin accumulation and subsequent loss of Pax7 followed by elongation and fusion to repair fast muscle fibres. Analysis of pax7a and pax7b transgenic reporter fish reveals that cells expressing each of the duplicated pax7 genes are distinctly localised in uninjured larvae. Cells marked by pax7a only or by both pax7a and pax7b enter the wound rapidly and contribute to muscle wound repair, but each behaves differently. Low numbers of pax7a-only cells form nascent fibres. Time-lapse microscopy revealed that the more numerous pax7b-marked cells frequently fuse to pre-existing fibres, contributing more strongly than pax7a-only cells to repair of damaged fibres. pax7b-marked cells are more often present in rows of aligned cells that are observed to fuse into a single fibre, but more rarely contribute to nascent regenerated fibres. Ablation of a substantial portion of nitroreductase-expressing pax7b cells with metronidazole prior to wounding triggered rapid pax7a-only cell accumulation, but this neither inhibited nor augmented pax7a-only cell-derived myogenesis and thus altered the cellular repair dynamics during wound healing. Moreover, pax7a-only cells did not regenerate pax7b cells, suggesting a lineage distinction. We propose a modified founder cell and fusion-competent cell model in which pax7a-only cells initiate fibre formation and pax7b cells contribute to fibre growth. This newly discovered cellular complexity in muscle wound repair raises the possibility that distinct populations of myogenic cells contribute differentially to repair in other vertebrates.
Collapse
Affiliation(s)
- Tapan G Pipalia
- Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Jana Koth
- Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London SE1 1UL, UK Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford University, Oxford OX3 9DS, UK
| | - Shukolpa D Roy
- Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Christina L Hammond
- Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, and Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, Shizuoka 411-8540, Japan
| | - Simon M Hughes
- Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London SE1 1UL, UK
| |
Collapse
|
39
|
Naraballobh W, Trakooljul N, Muráni E, Brunner R, Krischek C, Janisch S, Wicke M, Ponsuksili S, Wimmers K. Immediate and long-term transcriptional response of hind muscle tissue to transient variation of incubation temperature in broilers. BMC Genomics 2016; 17:323. [PMID: 27142659 PMCID: PMC4855815 DOI: 10.1186/s12864-016-2671-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 04/25/2016] [Indexed: 12/20/2022] Open
Abstract
Background In oviparous species accidental variation of incubation temperatures may occur under natural conditions and mechanisms may have evolved by natural selection that facilitate coping with these stressors. However, under controlled artificial incubation modification of egg incubation temperature has been shown to have a wide-ranging impact on post-hatch development in several poultry species. Because developmental changes initiated in-ovo can affect poultry production, understanding the molecular routes and epigenetic alterations induced by incubation temperature differences may allow targeted modification of phenotypes. Results In order to identify molecular pathways responsive to variable incubation temperature, broiler eggs were incubated at a lower or higher temperature (36.8 °C, 38.8 °C) relative to control (37.8 °C) over two developmental intervals, embryonic days (E) 7–10 and 10–13. Global gene expression of M. gastrocnemius was assayed at E10, E13, and slaughter age [post-hatch day (D) 35] (6 groups; 3 time points; 8 animals each) by microarray analysis and treated samples were compared to controls within each time point. Transcript abundance differed for between 113 and 738 genes, depending on treatment group, compared to the respective control. In particular, higher incubation temperature during E7-10 immediately affected pathways involved in energy and lipid metabolism, cell signaling, and muscle development more so than did other conditions. But lower incubation temperature during E10-13 affected pathways related to cellular function and growth, and development of organ, tissue, and muscle as well as nutrient metabolism pathways at D35. Conclusion Shifts in incubation temperature provoke specific immediate and long-term transcriptional responses. Further, the transcriptional response to lower incubation temperature, which did not affect the phenotypes, mediates compensatory effects reflecting adaptability. In contrast, higher incubation temperature triggers gene expression and has long-term effects on the phenotype, reflecting considerable phenotypic plasticity. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2671-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Watcharapong Naraballobh
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, 18196, Dummerstorf, Germany
| | - Nares Trakooljul
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, 18196, Dummerstorf, Germany
| | - Eduard Muráni
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, 18196, Dummerstorf, Germany
| | - Ronald Brunner
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, 18196, Dummerstorf, Germany
| | - Carsten Krischek
- Institute of Food Quality and Food Safety, University of Veterinary Medicine Hannover, D-30173, Hannover, Germany
| | - Sabine Janisch
- Department of Animal Science, Quality of Food of Animal Origin, Georg-August-University Goettingen, D-37075, Goettingen, Germany
| | - Michael Wicke
- Department of Animal Science, Quality of Food of Animal Origin, Georg-August-University Goettingen, D-37075, Goettingen, Germany
| | - Siriluck Ponsuksili
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, 18196, Dummerstorf, Germany
| | - Klaus Wimmers
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, 18196, Dummerstorf, Germany.
| |
Collapse
|
40
|
Chechenova MB, Maes S, Cripps RM. Expression of the Troponin C at 41C Gene in Adult Drosophila Tubular Muscles Depends upon Both Positive and Negative Regulatory Inputs. PLoS One 2015; 10:e0144615. [PMID: 26641463 PMCID: PMC4671713 DOI: 10.1371/journal.pone.0144615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/20/2015] [Indexed: 12/05/2022] Open
Abstract
Most animals express multiple isoforms of structural muscle proteins to produce tissues with different physiological properties. In Drosophila, the adult muscles include tubular-type muscles and the fibrillar indirect flight muscles. Regulatory processes specifying tubular muscle fate remain incompletely understood, therefore we chose to analyze the transcriptional regulation of TpnC41C, a Troponin C gene expressed in the tubular jump muscles, but not in the fibrillar flight muscles. We identified a 300-bp promoter fragment of TpnC41C sufficient for the fiber-specific reporter expression. Through an analysis of this regulatory element, we identified two sites necessary for the activation of the enhancer. Mutations in each of these sites resulted in 70% reduction of enhancer activity. One site was characterized as a binding site for Myocyte Enhancer Factor-2. In addition, we identified a repressive element that prevents activation of the enhancer in other muscle fiber types. Mutation of this site increased jump muscle-specific expression of the reporter, but more importantly reporter expression expanded into the indirect flight muscles. Our findings demonstrate that expression of the TpnC41C gene in jump muscles requires integration of multiple positive and negative transcriptional inputs. Identification of the transcriptional regulators binding the cis-elements that we identified will reveal the regulatory pathways controlling muscle fiber differentiation.
Collapse
Affiliation(s)
- Maria B Chechenova
- Department of Biology, University of New Mexico, Albuquerque, NM, 87131, United States of America
| | - Sara Maes
- Department of Biology, University of New Mexico, Albuquerque, NM, 87131, United States of America
| | - Richard M Cripps
- Department of Biology, University of New Mexico, Albuquerque, NM, 87131, United States of America
| |
Collapse
|
41
|
Duran BODS, Fernandez GJ, Mareco EA, Moraes LN, Salomão RAS, Gutierrez de Paula T, Santos VB, Carvalho RF, Dal-Pai-Silvca M. Differential microRNA Expression in Fast- and Slow-Twitch Skeletal Muscle of Piaractus mesopotamicus during Growth. PLoS One 2015; 10:e0141967. [PMID: 26529415 PMCID: PMC4631509 DOI: 10.1371/journal.pone.0141967] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/15/2015] [Indexed: 11/26/2022] Open
Abstract
Pacu (Piaractus mesopotamicus) is a Brazilian fish with a high economic value in pisciculture due to its rusticity and fast growth. Postnatal growth of skeletal muscle in fish occurs by hyperplasia and/or hypertrophy, processes that are dependent on the proliferation and differentiation of myoblasts. A class of small noncoding RNAs, known as microRNAs (miRNAs), represses the expression of target mRNAs, and many studies have demonstrated that miR-1, miR-133, miR-206 and miR-499 regulate different processes in skeletal muscle through the mRNA silencing of hdac4 (histone deacetylase 4), srf (serum response factor), pax7 (paired box 7) and sox6 ((sex determining region Y)-box 6), respectively. The aim of our work was to evaluate the expression of these miRNAs and their putative target mRNAs in fast- and slow-twitch skeletal muscle of pacu during growth. We used pacus in three different development stages: larval (aged 30 days), juvenile (aged 90 days and 150 days) and adult (aged 2 years). To complement our study, we also performed a pacu myoblast cell culture, which allowed us to investigate miRNA expression in the progression from myoblast proliferation to differentiation. Our results revealed an inverse correlation between the expression of the miRNAs and their target mRNAs, and there was evidence that miR-1 and miR-206 may regulate the differentiation of myoblasts, whereas miR-133 may regulate the proliferation of these cells. miR-499 was highly expressed in slow-twitch muscle, which suggests its involvement in the specification of the slow phenotype in muscle fibers. The expression of these miRNAs exhibited variations between different development stages and between distinct muscle twitch phenotypes. This work provides the first identification of miRNA expression profiles in pacu skeletal muscle and suggests an important role of these molecules in muscle growth and in the maintenance of the muscle phenotype.
Collapse
Affiliation(s)
- Bruno Oliveira da Silva Duran
- Department of Morphology, Institute of Biosciences of Botucatu, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Geysson Javier Fernandez
- Department of Morphology, Institute of Biosciences of Botucatu, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Edson Assunção Mareco
- Department of Morphology, Institute of Biosciences of Botucatu, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Leonardo Nazario Moraes
- Department of Morphology, Institute of Biosciences of Botucatu, São Paulo State University, Botucatu, São Paulo, Brazil
| | | | - Tassiana Gutierrez de Paula
- Department of Morphology, Institute of Biosciences of Botucatu, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Vander Bruno Santos
- São Paulo Agency for Agribusiness Technology, Presidente Prudente, São Paulo, Brazil
| | - Robson Francisco Carvalho
- Department of Morphology, Institute of Biosciences of Botucatu, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Maeli Dal-Pai-Silvca
- Department of Morphology, Institute of Biosciences of Botucatu, São Paulo State University, Botucatu, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
42
|
Zannino DA, Sagerström CG. An emerging role for prdm family genes in dorsoventral patterning of the vertebrate nervous system. Neural Dev 2015; 10:24. [PMID: 26499851 PMCID: PMC4620005 DOI: 10.1186/s13064-015-0052-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/13/2015] [Indexed: 12/13/2022] Open
Abstract
The embryonic vertebrate neural tube is divided along its dorsoventral (DV) axis into eleven molecularly discrete progenitor domains. Each of these domains gives rise to distinct neuronal cell types; the ventral-most six domains contribute to motor circuits, while the five dorsal domains contribute to sensory circuits. Following the initial neurogenesis step, these domains also generate glial cell types—either astrocytes or oligodendrocytes. This DV pattern is initiated by two morphogens—Sonic Hedgehog released from notochord and floor plate and Bone Morphogenetic Protein produced in the roof plate—that act in concentration gradients to induce expression of genes along the DV axis. Subsequently, these DV-restricted genes cooperate to define progenitor domains and to control neuronal cell fate specification and differentiation in each domain. Many genes involved in this process have been identified, but significant gaps remain in our understanding of the underlying genetic program. Here we review recent work identifying members of the Prdm gene family as novel regulators of DV patterning in the neural tube. Many Prdm proteins regulate transcription by controlling histone modifications (either via intrinsic histone methyltransferase activity, or by recruiting histone modifying enzymes). Prdm genes are expressed in spatially restricted domains along the DV axis of the neural tube and play important roles in the specification of progenitor domains, as well as in the subsequent differentiation of motor neurons and various types of interneurons. Strikingly, Prdm proteins appear to function by binding to, and modulating the activity of, other transcription factors (particularly bHLH proteins). The identity of key transcription factors in DV patterning of the neural tube has been elucidated previously (e.g. the nkx, bHLH and pax families), but it now appears that an additional family is also required and that it acts in a potentially novel manner.
Collapse
Affiliation(s)
- Denise A Zannino
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 364 Plantation St./LRB815, Worcester, MA, 01605-2324, USA.
| | - Charles G Sagerström
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 364 Plantation St./LRB815, Worcester, MA, 01605-2324, USA.
| |
Collapse
|
43
|
Khodabukus A, Baar K. Contractile and metabolic properties of engineered skeletal muscle derived from slow and fast phenotype mouse muscle. J Cell Physiol 2015; 230:1750-7. [PMID: 25335966 DOI: 10.1002/jcp.24848] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 10/14/2014] [Indexed: 11/08/2022]
Abstract
Satellite cells derived from fast and slow muscles have been shown to adopt contractile and metabolic properties of their parent muscle. Mouse muscle shows less distinctive fiber-type profiles than rat or rabbit muscle. Therefore, in this study we sought to determine whether three-dimensional muscle constructs engineered from slow soleus (SOL) and fast tibialis anterior (TA) from mice would adopt the contractile and metabolic properties of their parent muscle. Time-to-peak tension (TPT) and half-relaxation time (1/2RT) was significantly slower in SOL constructs. In agreement with TPT, TA constructs contained significantly higher levels of fast myosin heavy chain (MHC) and fast troponin C, I, and T isoforms. Fast SERCA protein, both slow and fast calsequestrin isoforms and parvalbumin were found at higher levels in TA constructs. SOL constructs were more fatigue resistant and contained higher levels of the mitochondrial proteins SDH and ATP synthase and the fatty acid transporter CPT-1. SOL constructs contained lower levels of the glycolytic enzyme phosphofructokinase but higher levels of the β-oxidation enzymes LCAD and VLCAD suggesting greater fat oxidation. Despite no changes in PGC-1α protein, SOL constructs contained higher levels of SIRT1 and PRC. TA constructs contained higher levels of the slow-fiber program repressor SOX6 and the six transcriptional complex (STC) proteins Eya1 and Six4 which may underlie the higher in fast-fiber and lower slow-fiber program proteins. Overall, we have found that muscles engineered from predominantly slow and fast mouse muscle retain contractile and metabolic properties of their native muscle.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, California
| | | |
Collapse
|
44
|
Li Y, Li J, Zhang L, Yu C, Lin M, Gao F, Zhou G, Zhang Y, Fan Y, Nuldnali L. Effects of Dietary Energy Sources on Post Mortem Glycolysis, Meat Quality and Muscle Fibre Type Transformation of Finishing Pigs. PLoS One 2015; 10:e0131958. [PMID: 26125946 PMCID: PMC4488424 DOI: 10.1371/journal.pone.0131958] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 06/08/2015] [Indexed: 12/29/2022] Open
Abstract
Dietary energy source can influence muscle glycogen storage at slaughter. However, few studies have demonstrated whether the diet-induced change of muscle glycogen is achieved by the transformation of muscle fibre type. This study investigated the effects of dietary energy sources on meat quality, post mortem glycolysis and muscle fibre type transformation of finishing pigs. Seventy-two barrows with an average body weight of 65.0 ± 2.0 kg were selected and were allotted to three iso-energetic and iso-nitrogenous diets A, B or C, and each treatment consisted of three replicates (pens) of eight pigs each. Diet A contained 44.1% starch, 5.9% crude fat and 12.6% neutral detergent fiber (NDF); diet B contained 37.6% starch, 9.5% crude fat and 15.4% NDF; and diet C contained 30.9% starch, 14.3% crude fat and 17.8% NDF. The duration of the experiment was 28 days. After feed withdrawal 12 h, 24 pigs (eight per treatment) were slaughtered, samples from M. longissimus lumborum (LL) were collected for subsequent analysis. The results showed that pigs fed diet C had lesser average daily gain, average daily feed intake and back fat depth than those fed diet A (P<0.05). Diet C increased pH45min (P<0.05) and decreased drip loss (P<0.05) in LL muscles compared with diet A. Meat from pigs fed diet A showed increased contents of lactate and greater glycolytic potential (GP) compared with those fed diet C (P<0.05). Greater mRNA expression of myosin heavy-chain (MyHC)-I and IIa and lesser expression of MyHC-IIx and IIb (P<0.05) in LL muscles were found in pigs fed diet C, than in pigs fed diet A. In addition, pigs fed diet C resulted in downregulation of miR23a and upregulation of miR409 and miR208b (P<0.05), associated with conserved changes of their corresponding targets. These findings indicated that diets containing low starch and high fibre were beneficial in reducing muscle glycolysis, improving meat quality of finishing pigs. This reduction of GP may be partially associated with the improvement of oxidative fibre composition in LL muscle, and the change in myofibre type may be correlated with the change in the miRNA expression.
Collapse
Affiliation(s)
- Yanjiao Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
- Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Jiaolong Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
- Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Lin Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
- Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Changning Yu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
- Synergetic Innovation Center of Food Safety and Nutrition, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Meng Lin
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
- Synergetic Innovation Center of Food Safety and Nutrition, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Feng Gao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
- Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
- * E-mail:
| | - Guanghong Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
- Synergetic Innovation Center of Food Safety and Nutrition, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Yu Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Yuanfang Fan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Lina Nuldnali
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| |
Collapse
|
45
|
Cheedipudi S, Puri D, Saleh A, Gala HP, Rumman M, Pillai MS, Sreenivas P, Arora R, Sellathurai J, Schrøder HD, Mishra RK, Dhawan J. A fine balance: epigenetic control of cellular quiescence by the tumor suppressor PRDM2/RIZ at a bivalent domain in the cyclin a gene. Nucleic Acids Res 2015; 43:6236-56. [PMID: 26040698 PMCID: PMC4513853 DOI: 10.1093/nar/gkv567] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 05/19/2015] [Indexed: 12/20/2022] Open
Abstract
Adult stem cell quiescence is critical to ensure regeneration while minimizing tumorigenesis. Epigenetic regulation contributes to cell cycle control and differentiation, but few regulators of the chromatin state in quiescent cells are known. Here we report that the tumor suppressor PRDM2/RIZ, an H3K9 methyltransferase, is enriched in quiescent muscle stem cells invivo and controls reversible quiescence in cultured myoblasts. We find that PRDM2 associates with >4400 promoters in G0 myoblasts, 55% of which are also marked with H3K9me2 and enriched for myogenic, cell cycle and developmental regulators. Knockdown of PRDM2 alters histone methylation at key promoters such as Myogenin and CyclinA2 (CCNA2), and subverts the quiescence program via global de-repression of myogenesis, and hyper-repression of the cell cycle. Further, PRDM2 acts upstream of the repressive PRC2 complex in G0. We identify a novel G0-specific bivalent chromatin domain in the CCNA2 locus. PRDM2 protein interacts with the PRC2 protein EZH2 and regulates its association with the bivalent domain in the CCNA2 gene. Our results suggest that induction of PRDM2 in G0 ensures that two antagonistic programs—myogenesis and the cell cycle—while stalled, are poised for reactivation. Together, these results indicate that epigenetic regulation by PRDM2 preserves key functions of the quiescent state, with implications for stem cell self-renewal.
Collapse
Affiliation(s)
- Sirisha Cheedipudi
- Institute for Stem Cell Biology and Regenerative Medicine, National Center for Biological Sciences, GKVK Post, Bellary Road, Bangalore 560065, India Council of Scientific and Industrial Research-Centre for Cellular and Molecular Biology, Hyderabad 500 007, India Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Deepika Puri
- Institute for Stem Cell Biology and Regenerative Medicine, National Center for Biological Sciences, GKVK Post, Bellary Road, Bangalore 560065, India Max Planck Institute of Immunobiology and Epigenetics, Freiburg D-79108, Germany
| | - Amena Saleh
- Institute for Stem Cell Biology and Regenerative Medicine, National Center for Biological Sciences, GKVK Post, Bellary Road, Bangalore 560065, India Manipal University, Manipal 576104 India
| | - Hardik P Gala
- Institute for Stem Cell Biology and Regenerative Medicine, National Center for Biological Sciences, GKVK Post, Bellary Road, Bangalore 560065, India Council of Scientific and Industrial Research-Centre for Cellular and Molecular Biology, Hyderabad 500 007, India
| | - Mohammed Rumman
- Institute for Stem Cell Biology and Regenerative Medicine, National Center for Biological Sciences, GKVK Post, Bellary Road, Bangalore 560065, India Manipal University, Manipal 576104 India
| | - Malini S Pillai
- Institute for Stem Cell Biology and Regenerative Medicine, National Center for Biological Sciences, GKVK Post, Bellary Road, Bangalore 560065, India
| | - Prethish Sreenivas
- Institute for Stem Cell Biology and Regenerative Medicine, National Center for Biological Sciences, GKVK Post, Bellary Road, Bangalore 560065, India Council of Scientific and Industrial Research-Centre for Cellular and Molecular Biology, Hyderabad 500 007, India
| | - Reety Arora
- Institute for Stem Cell Biology and Regenerative Medicine, National Center for Biological Sciences, GKVK Post, Bellary Road, Bangalore 560065, India
| | - Jeeva Sellathurai
- Institute of Clinical Research, SDU Muscle Research Cluster (SMRC), University of Southern Denmark, Odense 5000 C, Denmark
| | - Henrik Daa Schrøder
- Institute of Clinical Research, SDU Muscle Research Cluster (SMRC), University of Southern Denmark, Odense 5000 C, Denmark
| | - Rakesh K Mishra
- Council of Scientific and Industrial Research-Centre for Cellular and Molecular Biology, Hyderabad 500 007, India
| | - Jyotsna Dhawan
- Institute for Stem Cell Biology and Regenerative Medicine, National Center for Biological Sciences, GKVK Post, Bellary Road, Bangalore 560065, India Council of Scientific and Industrial Research-Centre for Cellular and Molecular Biology, Hyderabad 500 007, India
| |
Collapse
|
46
|
Nachtigall PG, Dias MC, Carvalho RF, Martins C, Pinhal D. MicroRNA-499 expression distinctively correlates to target genes sox6 and rod1 profiles to resolve the skeletal muscle phenotype in Nile tilapia. PLoS One 2015; 10:e0119804. [PMID: 25793727 PMCID: PMC4368118 DOI: 10.1371/journal.pone.0119804] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 02/02/2015] [Indexed: 11/22/2022] Open
Abstract
A class of small non-coding RNAs, the microRNAs (miRNAs), has been shown to be essential for the regulation of specific cell pathways, including skeletal muscle development, maintenance and homeostasis in vertebrates. However, the relative contribution of miRNAs for determining the red and white muscle cell phenotypes is far from being fully comprehended. To better characterize the role of miRNA in skeletal muscle cell biology, we investigated muscle-specific miRNA (myomiR) signatures in Nile tilapia fish. Quantitative (RT-qPCR) and spatial (FISH) expression analyses revealed a highly differential expression (forty-four-fold) of miR-499 in red skeletal muscle compared to white skeletal muscle, whereas the remaining known myomiRs were equally expressed in both muscle cell types. Detailed examination of the miR-499 targets through bioinformatics led us to the sox6 and rod1 genes, which had low expression in red muscle cells according to RT-qPCR, FISH, and protein immunofluorescence profiling experiments. Interestingly, we verified that the high expression of miR-499 perfectly correlates with a low expression of sox6 and rod1 target genes, as verified by a distinctive predominance of mRNA destabilization and protein translational decay to these genes, respectively. Through a genome-wide comparative analysis of SOX6 and ROD1 protein domains and through an in silico gene regulatory network, we also demonstrate that both proteins are essentially similar in vertebrate genomes, suggesting their gene regulatory network may also be widely conserved. Overall, our data shed light on the potential regulation of targets by miR-499 associated with the slow-twitch muscle fiber type phenotype. Additionally the results provide novel insights into the evolutionary dynamics of miRNA and target genes enrolled in a putative constrained molecular pathway in the skeletal muscle cells of vertebrates.
Collapse
Affiliation(s)
- Pedro G. Nachtigall
- Department of Genetics, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, Sao Paulo, 18618-970, Brazil
| | - Marcos C. Dias
- Department of Morphology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, Sao Paulo, 18618-970, Brazil
- Health Sciences Institute, Federal University of Mato Grosso (UFMT), Sinop, Mato Grosso, 78550-000, Brazil
| | - Robson F. Carvalho
- Department of Morphology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, Sao Paulo, 18618-970, Brazil
| | - Cesar Martins
- Department of Morphology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, Sao Paulo, 18618-970, Brazil
| | - Danillo Pinhal
- Department of Genetics, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, Sao Paulo, 18618-970, Brazil
| |
Collapse
|
47
|
Fong AP, Yao Z, Zhong JW, Johnson NM, Farr GH, Maves L, Tapscott SJ. Conversion of MyoD to a neurogenic factor: binding site specificity determines lineage. Cell Rep 2015; 10:1937-46. [PMID: 25801030 DOI: 10.1016/j.celrep.2015.02.055] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 01/16/2015] [Accepted: 02/23/2015] [Indexed: 10/23/2022] Open
Abstract
MyoD and NeuroD2, master regulators of myogenesis and neurogenesis, bind to a "shared" E-box sequence (CAGCTG) and a "private" sequence (CAGGTG or CAGATG, respectively). To determine whether private-site recognition is sufficient to confer lineage specification, we generated a MyoD mutant with the DNA-binding specificity of NeuroD2. This chimeric mutant gained binding to NeuroD2 private sites but maintained binding to a subset of MyoD-specific sites, activating part of both the muscle and neuronal programs. Sequence analysis revealed an enrichment for PBX/MEIS motifs at the subset of MyoD-specific sites bound by the chimera, and point mutations that prevent MyoD interaction with PBX/MEIS converted the chimera to a pure neurogenic factor. Therefore, redirecting MyoD binding from MyoD private sites to NeuroD2 private sites, despite preserved binding to the MyoD/NeuroD2 shared sites, is sufficient to change MyoD from a master regulator of myogenesis to a master regulator of neurogenesis.
Collapse
Affiliation(s)
- Abraham P Fong
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Pediatrics, Division of Hematology-Oncology, University of Washington School of Medicine, Seattle, WA 98105, USA.
| | - Zizhen Yao
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jun Wen Zhong
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Nathan M Johnson
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Gist H Farr
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Lisa Maves
- Department of Pediatrics, Division of Cardiology, University of Washington, Seattle, WA 98105, USA; Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Stephen J Tapscott
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Neurology, University of Washington School of Medicine, Seattle, WA 98105, USA.
| |
Collapse
|
48
|
Liu J, Liang X, Gan Z. Transcriptional regulatory circuits controlling muscle fiber type switching. SCIENCE CHINA-LIFE SCIENCES 2015; 58:321-7. [DOI: 10.1007/s11427-015-4833-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 12/17/2014] [Indexed: 12/16/2022]
|
49
|
Adaxial cell migration in the zebrafish embryo is an active cell autonomous property that requires the Prdm1a transcription factor. Differentiation 2015; 89:77-86. [DOI: 10.1016/j.diff.2015.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 02/17/2015] [Accepted: 03/17/2015] [Indexed: 02/06/2023]
|
50
|
Jackson HE, Ono Y, Wang X, Elworthy S, Cunliffe VT, Ingham PW. The role of Sox6 in zebrafish muscle fiber type specification. Skelet Muscle 2015; 5:2. [PMID: 25671076 PMCID: PMC4323260 DOI: 10.1186/s13395-014-0026-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 12/10/2014] [Indexed: 12/22/2022] Open
Abstract
Background The transcription factor Sox6 has been implicated in regulating muscle fiber type-specific gene expression in mammals. In zebrafish, loss of function of the transcription factor Prdm1a results in a slow to fast-twitch fiber type transformation presaged by ectopic expression of sox6 in slow-twitch progenitors. Morpholino-mediated Sox6 knockdown can suppress this transformation but causes ectopic expression of only one of three slow-twitch specific genes assayed. Here, we use gain and loss of function analysis to analyse further the role of Sox6 in zebrafish muscle fiber type specification. Methods The GAL4 binary misexpression system was used to express Sox6 ectopically in zebrafish embryos. Cis-regulatory elements were characterized using transgenic fish. Zinc finger nuclease mediated targeted mutagenesis was used to analyse the effects of loss of Sox6 function in embryonic, larval and adult zebrafish. Zebrafish transgenic for the GCaMP3 Calcium reporter were used to assay Ca2+ transients in wild-type and mutant muscle fibres. Results Ectopic Sox6 expression is sufficient to downregulate slow-twitch specific gene expression in zebrafish embryos. Cis-regulatory elements upstream of the slow myosin heavy chain 1 (smyhc1) and slow troponin c (tnnc1b) genes contain putative Sox6 binding sites required for repression of the former but not the latter. Embryos homozygous for sox6 null alleles expressed tnnc1b throughout the fast-twitch muscle whereas other slow-specific muscle genes, including smyhc1, were expressed ectopically in only a subset of fast-twitch fibers. Ca2+ transients in sox6 mutant fast-twitch fibers were intermediate in their speed and amplitude between those of wild-type slow- and fast-twitch fibers. sox6 homozygotes survived to adulthood and exhibited continued misexpression of tnnc1b as well as smaller slow-twitch fibers. They also exhibited a striking curvature of the spine. Conclusions The Sox6 transcription factor is a key regulator of fast-twitch muscle fiber differentiation in the zebrafish, a role similar to that ascribed to its murine ortholog. Electronic supplementary material The online version of this article (doi:10.1186/s13395-014-0026-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Harriet E Jackson
- ASTAR Institute of Molecular and Cell Biology, Proteos, 61 Biopolis Drive, Singapore, 138673 Republic of Singapore ; Bateson Centre, University of Sheffield, Western Bank, Sheffield, S10 2TN UK
| | - Yosuke Ono
- ASTAR Institute of Molecular and Cell Biology, Proteos, 61 Biopolis Drive, Singapore, 138673 Republic of Singapore
| | - Xingang Wang
- ASTAR Institute of Molecular and Cell Biology, Proteos, 61 Biopolis Drive, Singapore, 138673 Republic of Singapore
| | - Stone Elworthy
- Bateson Centre, University of Sheffield, Western Bank, Sheffield, S10 2TN UK
| | - Vincent T Cunliffe
- Bateson Centre, University of Sheffield, Western Bank, Sheffield, S10 2TN UK
| | - Philip W Ingham
- ASTAR Institute of Molecular and Cell Biology, Proteos, 61 Biopolis Drive, Singapore, 138673 Republic of Singapore ; Lee Kong Chian School of Medicine, Nanyang Technological University, Proteos, 61 Biopolis Drive, Singapore, 138673 Republic of Singapore ; Department of Medicine, Imperial College, South Kensington Campus, London, SW7 2AZ UK
| |
Collapse
|