1
|
Quach TK, Taylor MF, Currie PD, Eynon N, Ruparelia AA. Skeletal Muscle Aging: Lessons From Teleosts. J Gerontol A Biol Sci Med Sci 2025; 80:glae052. [PMID: 38367020 PMCID: PMC12080710 DOI: 10.1093/gerona/glae052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Indexed: 02/19/2024] Open
Abstract
Aging is the greatest risk factor for a multitude of age-related diseases including sarcopenia-the loss of skeletal muscle mass and strength-which occurs at remarkable rates each year. There is an unmet need not only to understand the mechanisms that drive sarcopenia but also to identify novel therapeutic strategies. Given the ease and affordability of husbandry, along with advances in genomics, genome editing technologies, and imaging capabilities, teleost models are increasingly used for aging and sarcopenia research. Here, we explain how teleost species such as zebrafish, African turquoise killifish, and medaka recapitulate many of the classical hallmarks of sarcopenia, and discuss the various dietary, pharmacological, and genetic approaches that have been used in teleosts to understand the mechanistic basis of sarcopenia.
Collapse
Affiliation(s)
- Tuyen K Quach
- Centre for Muscle Research, Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry, and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Megan F Taylor
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
- EMBL Australia, Victorian Node, Monash University, Clayton, Victoria, Australia
| | - Nir Eynon
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Avnika A Ruparelia
- Centre for Muscle Research, Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry, and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia (Biological Sciences Section)
| |
Collapse
|
2
|
Trace A, Wankell M, McFarlane C, Hebbard L. The challenges of using fish cells for cultivated seafood production. Food Sci Biotechnol 2025; 34:1565-1579. [PMID: 40129714 PMCID: PMC11929651 DOI: 10.1007/s10068-024-01786-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 03/26/2025] Open
Abstract
Continuing population growth is increasing nutritional demand and applying pressure to the world's finite resources. The current food systems struggle with sustainability, especially regarding protein sources. To address this, organisations have invested in developing novel sources of protein, such as lab-grown cultivated foods. Most of these efforts have focussed on the cultivated meat industry but neglect the emerging cultivated seafood industry. Arguably, seafood has a greater impact on protein availability and sustainability and should be a priority. Nonetheless, several technical barriers exist to produce cultivated seafood, and include a lack of established cell lines and specialised cell growth medium, that is affordable and sustainable. In addition, the application of this technology is difficult, due to public perception, ethical considerations, taste and food safety hurdles. Herein, we review the barriers that must be overcome by research institutions, companies, and stakeholders so that products can be introduced to the mainstream consumer market.
Collapse
Affiliation(s)
- Angela Trace
- Department of Biomedical Science and Molecular Biology, Australian Institute of Tropical Medicine and Health, College of Public Health, Medical and Veterinary Sciences, James Cook University, 1 James Cook Drive, Townsville, QLD 4811 Australia
| | - Miriam Wankell
- Department of Biomedical Science and Molecular Biology, Australian Institute of Tropical Medicine and Health, College of Public Health, Medical and Veterinary Sciences, James Cook University, 1 James Cook Drive, Townsville, QLD 4811 Australia
| | - Craig McFarlane
- Department of Biomedical Science and Molecular Biology, Australian Institute of Tropical Medicine and Health, College of Public Health, Medical and Veterinary Sciences, James Cook University, 1 James Cook Drive, Townsville, QLD 4811 Australia
| | - Lionel Hebbard
- Department of Biomedical Science and Molecular Biology, Australian Institute of Tropical Medicine and Health, College of Public Health, Medical and Veterinary Sciences, James Cook University, 1 James Cook Drive, Townsville, QLD 4811 Australia
| |
Collapse
|
3
|
Menard R, Morin E, Morse D, Halluin C, Pende M, Baanannou A, Grendler J, Fuqua H, Li J, Lancelot L, Drent J, Bonnet F, Graber JH, Murawala P, Dray C, Pradère JP, Coffman JA, Madelaine R. Zebrafish genetic model of neuromuscular degeneration associated with Atrogin-1 expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.07.642048. [PMID: 40196478 PMCID: PMC11975044 DOI: 10.1101/2025.03.07.642048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The degenerative loss of muscle associated with aging leading to muscular atrophy is called sarcopenia. Currently, practicing regular physical exercise is the only efficient way to delay sarcopenia onset. Identification of therapeutic targets to alleviate the symptoms of aging requires in vivo model organisms of accelerated muscle degeneration and atrophy. The zebrafish undergoes aging, with hallmarks including mitochondrial dysfunction, telomere shortening, and accumulation of senescent cells. However, zebrafish age slowly, and no specific zebrafish models of accelerated muscle atrophy associated with molecular events of aging are currently available. We have developed a new genetic tool to efficiently accelerate muscle-fiber degeneration and muscle-tissue atrophy in zebrafish larvae and adults. We used a gain-of-function strategy with a molecule that has been shown to be necessary and sufficient to induce muscle atrophy and a sarcopenia phenotype in mammals: Atrogin-1 (also named Fbxo32). We report the generation, validation, and characterization of a zebrafish genetic model of accelerated neuromuscular atrophy, the atrofish. We demonstrated that Atrogin-1 expression specifically in skeletal muscle tissue induces a muscle atrophic phenotype associated with locomotion dysfunction in both larvae and adult fish. We identified degradation of the myosin light chain as an event occurring prior to muscle-fiber degeneration. Biological processes associated with muscle aging such as proteolysis, inflammation, stress response, extracellular matrix (ECM) remodeling, and apoptosis are upregulated in the atrofish. Surprisingly, we observed a strong correlation between muscle-fiber degeneration and reduced numbers of neuromuscular junctions in the peripheral nervous system, as well as neuronal cell bodies in the spinal cord, suggesting that muscle atrophy could underly a neurodegenerative phenotype in the central nervous system. Finally, while atrofish larvae can recover locomotive functions, adult atrofish have impaired regenerative capacities, as is observed in mammals during muscle aging. In the future, the atrofish could serve as a platform for testing molecules aimed at treating or alleviating the symptoms of muscle aging, thereby opening new therapeutic avenues in the fight against sarcopenia.
Collapse
Affiliation(s)
- Romain Menard
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Elena Morin
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
- RESTORE Research Center, INSERM 1301, CNRS 5070, EFS, ENVT, Université Paul Sabatier, Toulouse, France
- IHU HealthAge, Toulouse, France
| | - Dexter Morse
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Caroline Halluin
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Marko Pende
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Aissette Baanannou
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Janelle Grendler
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Heath Fuqua
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Jijia Li
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
- CARe Graduate School, Université Paul Sabatier, Toulouse, France
| | - Laetitia Lancelot
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
- CARe Graduate School, Université Paul Sabatier, Toulouse, France
| | - Jessica Drent
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Frédéric Bonnet
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Joel H. Graber
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Prayag Murawala
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
- Department of nephrology and hypertension, Hannover Medical School, Hannover, Germany
| | - Cédric Dray
- RESTORE Research Center, INSERM 1301, CNRS 5070, EFS, ENVT, Université Paul Sabatier, Toulouse, France
- IHU HealthAge, Toulouse, France
| | - Jean-Philippe Pradère
- RESTORE Research Center, INSERM 1301, CNRS 5070, EFS, ENVT, Université Paul Sabatier, Toulouse, France
- IHU HealthAge, Toulouse, France
| | - James A. Coffman
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Romain Madelaine
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| |
Collapse
|
4
|
Zuo H, Jiang W, Gao J, Ma Z, Li C, Peng Y, Jin J, Zhan X, Lv W, Liu X, Hu J, Zhang M, Jia Y, Xu Z, Tang J, Zheng R, Zuo B. SYISL Knockout Promotes Embryonic Muscle Development of Offspring by Modulating Maternal Gut Microbiota and Fetal Myogenic Cell Dynamics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410953. [PMID: 39680624 PMCID: PMC11809340 DOI: 10.1002/advs.202410953] [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: 09/09/2024] [Revised: 12/02/2024] [Indexed: 12/18/2024]
Abstract
Embryonic muscle fiber formation determines post-birth muscle fiber totals. The previous research shows SYISL knockout significantly increases muscle fiber numbers and mass in mice, but the mechanism remains unclear. This study confirms that the SYISL gene, maternal gut microbiota, and their interaction significantly affect the number of muscle fibers in mouse embryos through distinct mechanisms, as SYISL knockout alters maternal gut microbiota composition and boosts butyrate levels in embryonic serum. Both fecal microbiota transplantation and butyrate feeding significantly increase muscle fiber numbers in offspring, with butyrate inhibiting histone deacetylases and increasing histone acetylation in embryonic muscle. Combined analysis of RNA-seq between wild-type and SYISL knockout mice with ChIP-seq for H3K9ac and H3K27ac reveals that SYISL and maternal microbiota interaction regulates myogenesis via the butyrate-HDAC-H3K9ac/H3K27ac pathway. Furthermore, scRNA-seq analysis shows that SYISL knockout alone significantly increases the number and proportion of myogenic cells and their dynamics, independently of regulating histone acetylation levels. Cell communication analysis suggests that this may be due to the downregulation of signaling pathways such as MSTN and TGFβ. Overall, multiple pathways are highlighted through which SYISL influences embryonic muscle development, offering valuable insights for treating muscle diseases and improving livestock production.
Collapse
Affiliation(s)
- Hao Zuo
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
- Hubei Hongshan LaboratoryWuhan430070China
| | - Wei Jiang
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Jianwei Gao
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Zhibo Ma
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Chen Li
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Yaxin Peng
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Jianjun Jin
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Xizhen Zhan
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Wei Lv
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Xiao Liu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Jingjing Hu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Mengdi Zhang
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Yiming Jia
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Zaiyan Xu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Department of Basic Veterinary MedicineCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
| | - Junming Tang
- Hubei Key Laboratory of Embryonic Stem Cell ResearchSchool of Basic Medicine ScienceHubei University of MedicineShiyan442000China
| | - Rong Zheng
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Bo Zuo
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
- Hubei Hongshan LaboratoryWuhan430070China
| |
Collapse
|
5
|
Coren L, Zaffryar-Eilot S, Odeh A, Kaganovsky A, Hasson P. Fibroblast diversification is an embryonic process dependent on muscle contraction. Cell Rep 2024; 43:115034. [PMID: 39636726 DOI: 10.1016/j.celrep.2024.115034] [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: 04/09/2024] [Revised: 09/18/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
Fibroblasts, the most common cell type found in connective tissues, play major roles in development, homeostasis, regeneration, and disease. Although specific fibroblast subpopulations have been associated with different biological processes, the mechanisms and unique activities underlying their diversity have not been thoroughly examined. Here, we set out to dissect the variation in skeletal-muscle-resident fibroblasts (mrFibroblasts) during development. Our results demonstrate that mrFibroblasts diversify following the transition from embryonic to fetal myogenesis prior to birth. We find that mrFibroblasts segregate into two major subpopulations occupying distinct niches, with interstitial fibroblasts residing between the muscle fibers and delineating fibroblasts sheathing the muscle. We further show that these subpopulations entail distinct cellular dynamics and transcriptomes. Notably, we find that mrFibroblast subpopulations exert distinct regulatory roles on myoblast proliferation and differentiation. Finally, we demonstrate that this diversification depends on muscle contraction. Altogether, these findings establish that mrFibroblasts diversify in a spatiotemporal embryonic process into distinct cell types, entailing different characteristics and roles.
Collapse
Affiliation(s)
- Lavi Coren
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Shelly Zaffryar-Eilot
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Anas Odeh
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Anna Kaganovsky
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Peleg Hasson
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
6
|
Ranasinghe T, Seo Y, Park HC, Choe SK, Cha SH. Rotenone exposure causes features of Parkinson`s disease pathology linked with muscle atrophy in developing zebrafish embryo. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136215. [PMID: 39461288 DOI: 10.1016/j.jhazmat.2024.136215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024]
Abstract
Parkinson's disease (PD) is associated with both genetic and environmental factors; however, sporadic forms of PD account for > 90 % of cases, and PD prevalence has doubled in the past 25 years. Depending on the importance of the environmental factors, various neurotoxins are used to induce PD both in vivo and in vitro. Unlike other neurodegenerative diseases, PD can be induced in vivo using specific neurotoxic chemicals. However, no chemically induced PD model is available because of the sporadic nature of PD. Rotenone is a pesticide that accelerates the induction of PD and exhibits the highest toxicity in fish, unlike other pesticides. Therefore, in this study, we aimed to establish a model exhibiting PD pathologies such as dysfunction of DArgic neuron, aggregation of ɑ-synuclein, and behavioral abnormalities, which are known features of PD pathology, by rotenone exposure at an environmentally relevant concentration (30 nM) in developing zebrafish embryos. Our results provide direct evidence for the association between PD and muscle degeneration by confirming rotenone-induced muscle atrophy. Therefore, we conclude that the rotenone-induced model presents non-motor and motor defects with extensive studies related to muscle atrophy.
Collapse
Affiliation(s)
- Thilini Ranasinghe
- Department of Marine Bio and Medical Sciences, Hanseo University, Seosan-si 31962, Republic of Korea
| | - Yongbo Seo
- Department of Biomedical Sciences, Korea University, Ansan 15328, Republic of Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, Korea University, Ansan 15328, Republic of Korea
| | - Seong-Kyu Choe
- Department of Microbiology, Wonkwang University School of Medicine, Iksan 54538, Republic of Korea; Sacopenia Total Solution Center, Wonkwang University School of Medicine, Iksan 54538, Republic of Korea
| | - Seon-Heui Cha
- Department of Marine Bio and Medical Sciences, Hanseo University, Seosan-si 31962, Republic of Korea; Department of Aquatic Life Medicine, Hanseo University, Seosan-si 31962, Republic of Korea; Institute for International Fisheries Science, Hanseo University, Seosan-si 31962, Republic of Korea.
| |
Collapse
|
7
|
Diao Z, Jia S, Itoyama E, Yoshioka H, Murakami M, Funaba M. A possibility of uncoupling protein 1 induction with the enhancement of myogenesis related to ruminal fermentation. Sci Rep 2024; 14:29857. [PMID: 39622913 PMCID: PMC11612152 DOI: 10.1038/s41598-024-81272-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024] Open
Abstract
The expression of uncoupling protein 1 (UCP1), which regulates energy expenditure, is limited to brown/beige adipocytes in most mammals; however, it is also detected in the skeletal muscles of cattle. We previously observed a positive relationship between Ucp1 and fast-twitch myosin heavy chain (Myh) expression in bovine skeletal muscles. In the present study, we explored the regulatory expression of Ucp1 in bovine myogenic cells using cell culture. Vitamin C and high-dose capsaicin, which induce the formation of fast-twitch myotubes in murine myogenic cells, did not stimulate myogenesis in bovine myosatellite cells. Treatment with 4-phenylbutyric acid (PBA), a histone deacetylase inhibitor that enhances histone acetylation, upregulates the expression of all myogenic regulatory factors (MRFs), except Myog, in bovine myogenic cells. Consistent with this, PBA increased the expression levels of acetylated lysine 27 of histone 3 (H3K27), the fast-twitch component MYH1/2, and Ucp1 in bovine myogenic cells. SB203580, an inhibitor of p38 MAP kinase, blocked PBA-induced myogenesis and Ucp1 upregulation. PBA is a butyric acid-related molecule, and cattle produce large amounts of volatile fatty acids (VFAs), including acetic acid, propionic acid, and butyric acid, through ruminal fermentation. Propionic acid treatment stimulated H3K27 acetylation, myogenesis, and Ucp1 induction. Thus, the upregulation of muscular Ucp1 may be related to myogenic stimulation through the modulation of histone acetylation status in cattle; we propose that the cattle-specific expression of muscular UCP1 results from VFA production through ruminal fermentation.
Collapse
Affiliation(s)
- Zhicheng Diao
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kitashirakawa Oiwakecho, Kyoto, 606-8502, Japan
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Shunhua Jia
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kitashirakawa Oiwakecho, Kyoto, 606-8502, Japan
| | - Erina Itoyama
- Kyoto University Livestock Farm, Kyotanba, 622-0203, Japan
| | | | - Masaru Murakami
- Laboratory of Molecular Biology, Azabu University School of Veterinary Medicine, Sagamihara, 252-5201, Japan
| | - Masayuki Funaba
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kitashirakawa Oiwakecho, Kyoto, 606-8502, Japan.
- Kyoto University Livestock Farm, Kyotanba, 622-0203, Japan.
| |
Collapse
|
8
|
Masiero C, Aresi C, Forlino A, Tonelli F. Zebrafish Models for Skeletal and Extraskeletal Osteogenesis Imperfecta Features: Unveiling Pathophysiology and Paving the Way for Drug Discovery. Calcif Tissue Int 2024; 115:931-959. [PMID: 39320469 PMCID: PMC11607041 DOI: 10.1007/s00223-024-01282-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 08/27/2024] [Indexed: 09/26/2024]
Abstract
In the last decades, the easy genetic manipulation, the external fertilization, the high percentage of homology with human genes and the reduced husbandry costs compared to rodents, made zebrafish a valid model for studying human diseases and for developing new therapeutical strategies. Since zebrafish shares with mammals the same bone cells and ossification types, it became widely used to dissect mechanisms and possible new therapeutic approaches in the field of common and rare bone diseases, such as osteoporosis and osteogenesis imperfecta (OI), respectively. OI is a heritable skeletal disorder caused by defects in gene encoding collagen I or proteins/enzymes necessary for collagen I synthesis and secretion. Nevertheless, OI patients can be also characterized by extraskeletal manifestations such as dentinogenesis imperfecta, muscle weakness, cardiac valve and pulmonary abnormalities and skin laxity. In this review, we provide an overview of the available zebrafish models for both dominant and recessive forms of OI. An updated description of all the main similarities and differences between zebrafish and mammal skeleton, muscle, heart and skin, will be also discussed. Finally, a list of high- and low-throughput techniques available to exploit both larvae and adult OI zebrafish models as unique tools for the discovery of new therapeutic approaches will be presented.
Collapse
Affiliation(s)
- Cecilia Masiero
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy
| | - Carla Aresi
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy
| | - Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy.
| | - Francesca Tonelli
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy
| |
Collapse
|
9
|
Zeng W, Meng Y, Nie L, Cheng C, Gao Z, Liu L, Zhu X, Chu W. The Regulatory Role of Myomaker in the Muscle Growth of the Chinese Perch ( Siniperca chuatsi). Animals (Basel) 2024; 14:2448. [PMID: 39272233 PMCID: PMC11394465 DOI: 10.3390/ani14172448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
The fusion of myoblasts is a crucial stage in the growth and development of skeletal muscle. Myomaker is an important myoblast fusion factor that plays a crucial role in regulating myoblast fusion. However, the function of Myomaker in economic fish during posthatching has been poorly studied. In this study, we found that the expression of Myomaker in the fast muscle of Chinese perch (Siniperca chuatsi) was higher than that in other tissues. To determine the function of Myomaker in fast muscle, Myomaker-siRNA was used to knockdown Myomaker in Chinese perch and the effect on muscle growth was determined. The results showed that the growth of Chinese perch was significantly decreased in the Myomaker-siRNA group. Furthermore, both the diameter of muscle fibers and the number of nuclei in single muscle fibers were significantly reduced in the Myomaker-siRNA group, whereas there was no significant difference in the number of BrdU-positive cells (proliferating cells) between the control and the Myomaker-siRNA groups. Together, these findings indicate that Myomaker may regulate growth of fast muscle in Chinese perch juveniles by promoting myoblast fusion rather than proliferation.
Collapse
Affiliation(s)
- Wei Zeng
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China
| | - Yangyang Meng
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China
| | - Lingtao Nie
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China
| | - Congyi Cheng
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China
| | - Zexia Gao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Wuhan 430070, China
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Lusha Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Wuhan 430070, China
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Xin Zhu
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China
| | - Wuying Chu
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China
| |
Collapse
|
10
|
Tönißen K, Franz GP, Albrecht E, Lutze P, Bochert R, Grunow B. Pikeperch muscle tissues: a comparative study of structure, enzymes, genes, and proteins in wild and farmed fish. FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:1527-1544. [PMID: 38733450 PMCID: PMC11286731 DOI: 10.1007/s10695-024-01354-1] [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: 07/07/2023] [Accepted: 05/02/2024] [Indexed: 05/13/2024]
Abstract
Pikeperch (Sander lucioperca) is a freshwater species and an internationally highly demanded fish in aquaculture. Despite intensive research efforts on this species, fundamental knowledge of skeletal muscle biology and structural characteristics is missing. Therefore, we conducted a comprehensive analysis of skeletal muscle parameters in adult pikeperch from two different origins, wild-caught specimens from a lake and those reared in a recirculating aquaculture system. The analyses comprised the biochemical characteristics (nucleic acid, protein content), enzyme activities (creatine kinase, lactate dehydrogenase, NADP-dependent isocitrate dehydrogenase), muscle-specific gene and protein expression (related to myofibre formation, regeneration and permanent growth, muscle structure), and muscle fibre structure. The findings reveal distinct differences between the skeletal muscle of wild and farmed pikeperch. Specifically, nucleic acid content, enzyme activity, and protein expression varied significantly. The higher enzyme activity observed in wild pikeperch suggests greater metabolically activity in their muscles. Conversely, farmed pikeperch indicated a potential for pronounced muscle growth. As the data on pikeperch skeletal muscle characteristics is sparse, the purpose of our study is to gain fundamental insights into the characteristics of adult pikeperch muscle. The presented data serve as a foundation for further research on percids' muscle biology and have the potential to contribute to advancements and adaptations in aquaculture practices.
Collapse
Affiliation(s)
- Katrin Tönißen
- Fish Growth Physiology Workgroup, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| | - George P Franz
- Fish Growth Physiology Workgroup, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Elke Albrecht
- Working Group Muscle-Fat Crosstalk, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Philipp Lutze
- Fish Growth Physiology Workgroup, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Ralf Bochert
- Mecklenburg-Vorpommern Research Centre for Agriculture and Fisheries (LFA MV), Institute of Fisheries, Research Station Aquaculture, Born, Germany
| | - Bianka Grunow
- Fish Growth Physiology Workgroup, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| |
Collapse
|
11
|
Perez ÉS, Duran BOS, Zanella BTT, Dal-Pai-Silva M. Review: Understanding fish muscle biology in the indeterminate growth species pacu (Piaractus mesopotamicus). Comp Biochem Physiol A Mol Integr Physiol 2023; 285:111502. [PMID: 37572733 DOI: 10.1016/j.cbpa.2023.111502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023]
Abstract
The muscle phenotype of fish is regulated by numerous factors that, although widely explored, still need to be fully understood. In this context, several studies aimed to unravel how internal and external stimuli affect the muscle growth of these vertebrates. The pacu (Piaractus mesopotamicus) is a species of indeterminate muscular growth that quickly reaches high body weight. For this reason, it adds great importance to the productive sector, along with other round fish. In this context, we aimed to compile studies on fish biology and skeletal muscle growth, focusing on studies by our research group that used pacu as an experimental model along with other species. Based on these studies, new muscle phenotype regulators were identified and explored in vivo, in vitro, and in silico studies, which strongly contribute to advances in understanding muscle growth mechanisms with future applications in the productive sector.
Collapse
Affiliation(s)
- Érika Stefani Perez
- Department of Structural and Functional Biology, Institute of Bioscience of Botucatu, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil.
| | - Bruno Oliveira Silva Duran
- Department of Histology, Embryology and Cell Biology, Institute of Biological Sciences, Federal University of Goiás (UFG), Goiânia, Goiás, Brazil.
| | - Bruna Tereza Thomazini Zanella
- Department of Structural and Functional Biology, Institute of Bioscience of Botucatu, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil.
| | - Maeli Dal-Pai-Silva
- Department of Structural and Functional Biology, Institute of Bioscience of Botucatu, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil.
| |
Collapse
|
12
|
Wang P, Liu X, Yao Z, Chen Y, Luo L, Liang K, Tan JHE, Chua MWJ, Chua YJB, Ma S, Zhang L, Ma W, Liu S, Cao W, Guo L, Guang L, Wang Y, Zhao H, Ai N, Li Y, Li C, Wang RR, Teh BT, Jiang L, Yu K, Shyh-Chang N. Lin28a maintains a subset of adult muscle stem cells in an embryonic-like state. Cell Res 2023; 33:712-726. [PMID: 37188880 PMCID: PMC10474071 DOI: 10.1038/s41422-023-00818-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: 09/21/2022] [Accepted: 04/23/2023] [Indexed: 05/17/2023] Open
Abstract
During homeostasis and after injury, adult muscle stem cells (MuSCs) activate to mediate muscle regeneration. However, much remains unclear regarding the heterogeneous capacity of MuSCs for self-renewal and regeneration. Here, we show that Lin28a is expressed in embryonic limb bud muscle progenitors, and that a rare reserve subset of Lin28a+Pax7- skeletal MuSCs can respond to injury at adult stage by replenishing the Pax7+ MuSC pool to drive muscle regeneration. Compared with adult Pax7+ MuSCs, Lin28a+ MuSCs displayed enhanced myogenic potency in vitro and in vivo upon transplantation. The epigenome of adult Lin28a+ MuSCs showed resemblance to embryonic muscle progenitors. In addition, RNA-sequencing revealed that Lin28a+ MuSCs co-expressed higher levels of certain embryonic limb bud transcription factors, telomerase components and the p53 inhibitor Mdm4, and lower levels of myogenic differentiation markers compared to adult Pax7+ MuSCs, resulting in enhanced self-renewal and stress-response signatures. Functionally, conditional ablation and induction of Lin28a+ MuSCs in adult mice revealed that these cells are necessary and sufficient for efficient muscle regeneration. Together, our findings connect the embryonic factor Lin28a to adult stem cell self-renewal and juvenile regeneration.
Collapse
Affiliation(s)
- Peng Wang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xupeng Liu
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ziyue Yao
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yu Chen
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lanfang Luo
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kun Liang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jun-Hao Elwin Tan
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
- Laboratory of Cancer Therapeutics, Program in Cancer and Stem Cell Biology, Duke-National University of Singapore Medical School, Singapore, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Science, National Cancer Centre Singapore, Singapore, Singapore
| | - Min-Wen Jason Chua
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
- Laboratory of Cancer Therapeutics, Program in Cancer and Stem Cell Biology, Duke-National University of Singapore Medical School, Singapore, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Science, National Cancer Centre Singapore, Singapore, Singapore
| | - Yan-Jiang Benjamin Chua
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
- Laboratory of Cancer Therapeutics, Program in Cancer and Stem Cell Biology, Duke-National University of Singapore Medical School, Singapore, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Science, National Cancer Centre Singapore, Singapore, Singapore
| | - Shilin Ma
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Liping Zhang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wenwu Ma
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuqing Liu
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wenhua Cao
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Luyao Guo
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lu Guang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuefan Wang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - He Zhao
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Na Ai
- University of Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Yun Li
- University of Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Chunwei Li
- Department of Clinical Nutrition, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ruiqi Rachel Wang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Bin Tean Teh
- Laboratory of Cancer Therapeutics, Program in Cancer and Stem Cell Biology, Duke-National University of Singapore Medical School, Singapore, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Science, National Cancer Centre Singapore, Singapore, Singapore
| | - Lan Jiang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Kang Yu
- Department of Clinical Nutrition, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ng Shyh-Chang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
13
|
Zhao L, Liu X, Gomez NA, Gao Y, Son JS, Chae SA, Zhu MJ, Du M. Stage-specific nutritional management and developmental programming to optimize meat production. J Anim Sci Biotechnol 2023; 14:2. [PMID: 36597116 PMCID: PMC9809060 DOI: 10.1186/s40104-022-00805-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 11/23/2022] [Indexed: 01/04/2023] Open
Abstract
Over the past few decades, genetic selection and refined nutritional management have extensively been used to increase the growth rate and lean meat production of livestock. However, the rapid growth rates of modern breeds are often accompanied by a reduction in intramuscular fat deposition and increased occurrences of muscle abnormalities, impairing meat quality and processing functionality. Early stages of animal development set the long-term growth trajectory of offspring. However, due to the seasonal reproductive cycles of ruminant livestock, gestational nutrient deficiencies caused by seasonal variations, frequent droughts, and unfavorable geological locations negatively affect fetal development and their subsequent production efficiency and meat quality. Therefore, enrolling livestock in nutritional intervention strategies during gestation is effective for improving the body composition and meat quality of the offspring at harvest. These crucial early developmental stages include embryonic, fetal, and postnatal stages, which have stage-specific effects on subsequent offspring development, body composition, and meat quality. This review summarizes contemporary research in the embryonic, fetal, and neonatal development, and the impacts of maternal nutrition on the early development and programming effects on the long-term growth performance of livestock. Understanding the developmental and metabolic characteristics of skeletal muscle, adipose, and fibrotic tissues will facilitate the development of stage-specific nutritional management strategies to optimize production efficiency and meat quality.
Collapse
Affiliation(s)
- Liang Zhao
- grid.27871.3b0000 0000 9750 7019College of Animal Science and Technology, Nanjing Agricultural University, 210095 Nanjing, PR China ,grid.30064.310000 0001 2157 6568Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, WA 99164 Pullman, USA
| | - Xiangdong Liu
- grid.30064.310000 0001 2157 6568Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, WA 99164 Pullman, USA
| | - Noe A Gomez
- grid.30064.310000 0001 2157 6568Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, WA 99164 Pullman, USA
| | - Yao Gao
- grid.30064.310000 0001 2157 6568Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, WA 99164 Pullman, USA
| | - Jun Seok Son
- grid.30064.310000 0001 2157 6568Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, WA 99164 Pullman, USA ,grid.411024.20000 0001 2175 4264Laboratory of Perinatal Kinesioepigenetics, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, MD 21201 Baltimore, USA
| | - Song Ah Chae
- grid.30064.310000 0001 2157 6568Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, WA 99164 Pullman, USA
| | - Mei-Jun Zhu
- grid.30064.310000 0001 2157 6568School of Food Science, Washington State University, WA Pullman, USA
| | - Min Du
- grid.30064.310000 0001 2157 6568Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, WA 99164 Pullman, USA
| |
Collapse
|
14
|
Perelló-Amorós M, Otero-Tarrazón A, Jorge-Pedraza V, García-Pérez I, Sánchez-Moya A, Gabillard JC, Moshayedi F, Navarro I, Capilla E, Fernández-Borràs J, Blasco J, Chillarón J, García de la serrana D, Gutiérrez J. Myomaker and Myomixer Characterization in Gilthead Sea Bream under Different Myogenesis Conditions. Int J Mol Sci 2022; 23:ijms232314639. [PMID: 36498967 PMCID: PMC9737248 DOI: 10.3390/ijms232314639] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022] Open
Abstract
Skeletal muscle is formed by multinucleated myofibers originated by waves of hyperplasia and hypertrophy during myogenesis. Tissue damage triggers a regeneration process including new myogenesis and muscular remodeling. During myogenesis, the fusion of myoblasts is a key step that requires different genes' expression, including the fusogens myomaker and myomixer. The present work aimed to characterize these proteins in gilthead sea bream and their possible role in in vitro myogenesis, at different fish ages and during muscle regeneration after induced tissue injury. Myomaker is a transmembrane protein highly conserved among vertebrates, whereas Myomixer is a micropeptide that is moderately conserved. myomaker expression is restricted to skeletal muscle, while the expression of myomixer is more ubiquitous. In primary myocytes culture, myomaker and myomixer expression peaked at day 6 and day 8, respectively. During regeneration, the expression of both fusogens and all the myogenic regulatory factors showed a peak after 16 days post-injury. Moreover, myomaker and myomixer were present at different ages, but in fingerlings there were significantly higher transcript levels than in juveniles or adult fish. Overall, Myomaker and Myomixer are valuable markers of muscle growth that together with other regulatory molecules can provide a deeper understanding of myogenesis regulation in fish.
Collapse
Affiliation(s)
- Miquel Perelló-Amorós
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Aitor Otero-Tarrazón
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Violeta Jorge-Pedraza
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Isabel García-Pérez
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Albert Sánchez-Moya
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | | | - Fatemeh Moshayedi
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Isabel Navarro
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Encarnación Capilla
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Jaume Fernández-Borràs
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Josefina Blasco
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Josep Chillarón
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Daniel García de la serrana
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Joaquim Gutiérrez
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
- Correspondence: ; Tel.: +34-934-021-532
| |
Collapse
|
15
|
Tonelotto V, Consorti C, Facchinello N, Trapani V, Sabatelli P, Giraudo C, Spizzotin M, Cescon M, Bertolucci C, Bonaldo P. Collagen VI ablation in zebrafish causes neuromuscular defects during developmental and adult stages. Matrix Biol 2022; 112:39-61. [PMID: 35961424 DOI: 10.1016/j.matbio.2022.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/21/2022] [Accepted: 08/08/2022] [Indexed: 10/15/2022]
Abstract
Collagen VI (COL6) is an extracellular matrix protein exerting multiple functions in different tissues. In humans, mutations of COL6 genes cause rare inherited congenital disorders, primarily affecting skeletal muscles and collectively known as COL6-related myopathies, for which no cure is available yet. In order to get insights into the pathogenic mechanisms underlying COL6-related diseases, diverse animal models were produced. However, the roles exerted by COL6 during embryogenesis remain largely unknown. Here, we generated the first zebrafish COL6 knockout line through CRISPR/Cas9 site-specific mutagenesis of the col6a1 gene. Phenotypic characterization during embryonic and larval development revealed that lack of COL6 leads to neuromuscular defects and motor dysfunctions, together with distinctive alterations in the three-dimensional architecture of craniofacial cartilages. These phenotypic features were maintained in adult col6a1 null fish, which displayed defective muscle organization and impaired swimming capabilities. Moreover, col6a1 null fish showed autophagy defects and organelle abnormalities at both embryonic and adult stages, thus recapitulating the main features of patients affected by COL6-related myopathies. Mechanistically, lack of COL6 led to increased BMP signaling, and direct inhibition of BMP activity ameliorated the locomotor col6a1 null embryos. Finally performance of, treatment with salbutamol, a β2-adrenergic receptor agonist, elicited a significant amelioration of the neuromuscular and motility defects of col6a1 null fish embryos. Altogether, these findings indicate that this newly generated zebrafish col6a1 null line is a valuable in vivo tool to model COL6-related myopathies and suitable for drug screenings aimed at addressing the quest for effective therapeutic strategies for these disorders.
Collapse
Affiliation(s)
| | - Chiara Consorti
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Nicola Facchinello
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Valeria Trapani
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Patrizia Sabatelli
- CNR - Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, 40136, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Chiara Giraudo
- Department of Medicine, Unit of Advanced Clinical and Translational Imaging, University of Padova, 35128 Padova, Italy
| | - Marianna Spizzotin
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Matilde Cescon
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Cristiano Bertolucci
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy; CRIBI Biotechnology Center, University of Padova, 35131 Padova, Italy.
| |
Collapse
|
16
|
Simó I, Faggiani M, Fernandez DA, Sciara AA, Arranz SE. The cellular basis of compensatory muscle growth in the teleost Odontesthes bonariensis. J Exp Biol 2021; 225:273693. [PMID: 34889453 DOI: 10.1242/jeb.242567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 12/06/2021] [Indexed: 11/20/2022]
Abstract
This study evaluates white muscle growth and in vivo cell proliferation during a fasting and refeeding trial, using pejerrey Odontesthes bonariensis as animal model, in order to better understand the cellular basis governing catch-up growth. Experiments consisted in two groups of fish, a control one continuously fed ad libitum, and a group fasted for 2 weeks and then fed for another 2 weeks. We examined how the formation of new muscle fibers and their increase in size were related to muscle precursor cell (MPC) proliferation under both experimental conditions. During fasting, the number of 5-ethynyl-2'-deoxyuridinepositive (EdU+) cells decreased along with myogenic regulatory factors (MRF) mRNA levels related to myoblast proliferation and differentiation, and the muscle stem cell-markerPax7 mRNA level increased. Analysis of myomere cross-sectional area, distribution of muscle fiber sizes and number of fibers per myomere showed that muscle hypertrophy but not hyperplasia was inhibited during fasting. Both higher igf2 mRNA level and the persistence of cell proliferation could be supporting new myofibre formation. On the other hand, an exacerbated MPC proliferation occurred during catch-up growth, and this increase in cell number could be contributing to the growth of both pre-existing and newly form small fibers. The finding that some MPCs proliferate during fasting and that muscle growth mechanisms, hyperplasia and hypertrophy, are differentially regulated could help to explain why re-fed fish could growth at higher rates, and why they return to the lost growth trajectory.
Collapse
Affiliation(s)
- Ignacio Simó
- Laboratorio Mixto de Biotecnología Acuática, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), Centro Científico, Tecnológico y Educativo Acuario del Río Paraná, Av. Eduardo Carrasco y Cordiviola s/n, Rosario, 2000, Argentina
| | - Mariano Faggiani
- Laboratorio Mixto de Biotecnología Acuática, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), Centro Científico, Tecnológico y Educativo Acuario del Río Paraná, Av. Eduardo Carrasco y Cordiviola s/n, Rosario, 2000, Argentina
| | - Daniel A Fernandez
- Instituto de Ciencias Polares, Ambiente y Recursos Naturales (ICPA), Universidad Nacional de Tierra del Fuego (UNTDF), Fuegiabasket 251, V9410BXE Ushuaia, Argentina.,Centro Austral de Investigaciones Científicas (CADIC-CONICET), Bernardo A. Houssay 200, V9410BXE Ushuaia, Argentina
| | - Andrés A Sciara
- Laboratorio Mixto de Biotecnología Acuática, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), Centro Científico, Tecnológico y Educativo Acuario del Río Paraná, Av. Eduardo Carrasco y Cordiviola s/n, Rosario, 2000, Argentina
| | - Silvia E Arranz
- Laboratorio Mixto de Biotecnología Acuática, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), Centro Científico, Tecnológico y Educativo Acuario del Río Paraná, Av. Eduardo Carrasco y Cordiviola s/n, Rosario, 2000, Argentina
| |
Collapse
|
17
|
Sun X, Li L, Wu B, Ge J, Zheng Y, Yu T, Zhou L, Zhang T, Yang A, Liu Z. Cell type diversity in scallop adductor muscles revealed by single-cell RNA-Seq. Genomics 2021; 113:3582-3598. [PMID: 34425225 DOI: 10.1016/j.ygeno.2021.08.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 07/26/2021] [Accepted: 08/18/2021] [Indexed: 11/28/2022]
Abstract
Studies on cell atlas in marine invertebrates provide a better understanding of cell types, stem cell maintenance, and lineages of cell differentiation. To investigate the molecular features of various cell types in molluscan muscles, we performed single-cell RNA sequencing (scRNA-seq) to map cell types in scallop adductor muscles. We uncovered the cell type-specific features of 20 cell clusters defined by the expression of multiple specific molecular markers. These cell clusters are mainly classified into four broad classes, including mesenchymal stem cells, muscle cells, neurons, and haemolymph cells. In particular, we identified a diverse repertoire of neurons in the striated adductor muscle, but not in the smooth muscle. We further reconstructed the cell differentiation events using all the cell clusters by single-cell pseudotemporal trajectories. By integrating dual BrdU-PCNA immunodetection, neuron-specific staining and electron microscopy observation, we showed the spatial distribution of mesenchymal stem cells and neurons in striated adductor muscle of scallops. The present findings will not only be useful to address the cell type-specific gene expression profiles in scallop muscles, but also provide valuable resources for cross-species comparison of marine organisms.
Collapse
Affiliation(s)
- Xiujun Sun
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Sustainable Development of Marine Fisheries, Ministry of Agriculture and Rural Affairs, Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, China
| | - Li Li
- Marine Biology Institute of Shandong Province, Qingdao 266104, China
| | - Biao Wu
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Sustainable Development of Marine Fisheries, Ministry of Agriculture and Rural Affairs, Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, China
| | - Jianlong Ge
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Sustainable Development of Marine Fisheries, Ministry of Agriculture and Rural Affairs, Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, China
| | - Yanxin Zheng
- Changdao Enhancement and Experiment Station, Chinese Academy of Fishery Sciences, Yantai 265800, China
| | - Tao Yu
- Changdao Enhancement and Experiment Station, Chinese Academy of Fishery Sciences, Yantai 265800, China
| | - Liqing Zhou
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Sustainable Development of Marine Fisheries, Ministry of Agriculture and Rural Affairs, Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, China
| | - Tianshi Zhang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Sustainable Development of Marine Fisheries, Ministry of Agriculture and Rural Affairs, Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, China
| | - Aiguo Yang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Sustainable Development of Marine Fisheries, Ministry of Agriculture and Rural Affairs, Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, China
| | - Zhihong Liu
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Sustainable Development of Marine Fisheries, Ministry of Agriculture and Rural Affairs, Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, China.
| |
Collapse
|
18
|
Histological and biochemical evaluation of skeletal muscle in the two salmonid species Coregonus maraena and Oncorhynchus mykiss. PLoS One 2021; 16:e0255062. [PMID: 34383783 PMCID: PMC8360549 DOI: 10.1371/journal.pone.0255062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/08/2021] [Indexed: 12/03/2022] Open
Abstract
The growth of fishes and their metabolism is highly variable in fish species and is an indicator for fish fitness. Therefore, somatic growth, as a main biological process, is ecologically and economically significant. The growth differences of two closely related salmonids, rainbow trout (Oncorhynchus mykiss) and maraena whitefsh (Coregonus maraena), have not been adequately studied as a comparative study and are therefore insufficiently understood. For this reason, our aim was to examine muscle growth in more detail and provide a first complex insight into the growth and muscle metabolism of these two fish species at slaughter size. In addition to skeletal muscle composition (including nuclear counting and staining of stem and progenitor cells), biochemical characteristics, and enzyme activity (creatine kinase, lactate dehydrogenase, isocitrate dehydrogenase) of rainbow trout and maraena whitefish were determined. Our results indicate that red muscle contains cells with a smaller diameter compared to white muscle and those fibres had more stem and progenitor cells as a proportion of total nuclei. Interestingly, numerous interspecies differences were identified; in rainbow trout muscle RNA content, intermediate fibres and fibre diameter and in whitefish red muscle cross-sectional area, creatine kinase activity were higher compared to the other species at slaughter weight. The proportional reduction in red muscle area, accompanied by an increase in DNA content and a lower activity of creatine kinase, exhibited a higher degree of hypertrophic growth in rainbow trout compared to maraena whitefish, which makes this species particularly successful as an aquaculture species.
Collapse
|
19
|
Function of Circular RNAs in Fish and Their Potential Application as Biomarkers. Int J Mol Sci 2021; 22:ijms22137119. [PMID: 34281172 PMCID: PMC8268770 DOI: 10.3390/ijms22137119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 11/16/2022] Open
Abstract
Circular RNAs (circRNAs) are an emerging class of regulatory RNAs with a covalently closed-loop structure formed during pre-mRNA splicing. Recent advances in high-throughput RNA sequencing and circRNA-specific computational tools have driven the development of novel approaches to their identification and functional characterization. CircRNAs are stable, developmentally regulated, and show tissue- and cell-type-specific expression across different taxonomic groups. They play a crucial role in regulating various biological processes at post-transcriptional and translational levels. However, the involvement of circRNAs in fish immunity has only recently been recognized. There is also broad evidence in mammals that the timely expression of circRNAs in muscle plays an essential role in growth regulation but our understanding of their expression and function in teleosts is still very limited. Here, we discuss the available knowledge about circRNAs and their role in growth and immunity in vertebrates from a comparative perspective, with emphasis on cultured teleost fish. We expect that the interest in teleost circRNAs will increase substantially soon, and we propose that they may be used as biomarkers for selective breeding of farmed fish, thus contributing to the sustainability of the aquaculture sector.
Collapse
|
20
|
Ghilardi A, Diana A, Bacchetta R, Santo N, Ascagni M, Prosperi L, Del Giacco L. Inner Ear and Muscle Developmental Defects in Smpx-Deficient Zebrafish Embryos. Int J Mol Sci 2021; 22:ijms22126497. [PMID: 34204426 PMCID: PMC8235540 DOI: 10.3390/ijms22126497] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/09/2021] [Accepted: 06/12/2021] [Indexed: 12/20/2022] Open
Abstract
The last decade has witnessed the identification of several families affected by hereditary non-syndromic hearing loss (NSHL) caused by mutations in the SMPX gene and the loss of function has been suggested as the underlying mechanism. In the attempt to confirm this hypothesis we generated an Smpx-deficient zebrafish model, pointing out its crucial role in proper inner ear development. Indeed, a marked decrease in the number of kinocilia together with structural alterations of the stereocilia and the kinocilium itself in the hair cells of the inner ear were observed. We also report the impairment of the mechanotransduction by the hair cells, making SMPX a potential key player in the construction of the machinery necessary for sound detection. This wealth of evidence provides the first possible explanation for hearing loss in SMPX-mutated patients. Additionally, we observed a clear muscular phenotype consisting of the defective organization and functioning of muscle fibers, strongly suggesting a potential role for the protein in the development of muscle fibers. This piece of evidence highlights the need for more in-depth analyses in search for possible correlations between SMPX mutations and muscular disorders in humans, thus potentially turning this non-syndromic hearing loss-associated gene into the genetic cause of dysfunctions characterized by more than one symptom, making SMPX a novel syndromic gene.
Collapse
Affiliation(s)
- Anna Ghilardi
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (A.G.); (A.D.); (L.P.)
| | - Alberto Diana
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (A.G.); (A.D.); (L.P.)
| | - Renato Bacchetta
- Department of Environmental Science and Policy, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Nadia Santo
- Unitech NOLIMITS, Università degli Studi di Milano, 20133 Milan, Italy; (N.S.); (M.A.)
| | - Miriam Ascagni
- Unitech NOLIMITS, Università degli Studi di Milano, 20133 Milan, Italy; (N.S.); (M.A.)
| | - Laura Prosperi
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (A.G.); (A.D.); (L.P.)
| | - Luca Del Giacco
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (A.G.); (A.D.); (L.P.)
- Correspondence:
| |
Collapse
|
21
|
Ferreira FJ, Carvalho L, Logarinho E, Bessa J. foxm1 Modulates Cell Non-Autonomous Response in Zebrafish Skeletal Muscle Homeostasis. Cells 2021; 10:cells10051241. [PMID: 34070077 PMCID: PMC8158134 DOI: 10.3390/cells10051241] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/01/2021] [Accepted: 05/11/2021] [Indexed: 12/23/2022] Open
Abstract
foxm1 is a master regulator of the cell cycle, contributing to cell proliferation. Recent data have shown that this transcription factor also modulates gene networks associated with other cellular mechanisms, suggesting non-proliferative functions that remain largely unexplored. In this study, we used CRISPR/Cas9 to disrupt foxm1 in the zebrafish terminally differentiated fast-twitching muscle cells. foxm1 genomic disruption increased myofiber death and clearance. Interestingly, this contributed to non-autonomous satellite cell activation and proliferation. Moreover, we observed that Cas9 expression alone was strongly deleterious to muscle cells. Our report shows that foxm1 modulates a muscle non-autonomous response to myofiber death and highlights underreported toxicity to high expression of Cas9 in vivo.
Collapse
Affiliation(s)
- Fábio J. Ferreira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (F.J.F.); (L.C.)
- Vertebrate Development and Regeneration Group, IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Aging and Aneuploidy Group, IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Graduate Program in Areas of Basic and Applied Biology (GABBA), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, 4050-313 Porto, Portugal
| | - Leonor Carvalho
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (F.J.F.); (L.C.)
- Vertebrate Development and Regeneration Group, IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Elsa Logarinho
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (F.J.F.); (L.C.)
- Aging and Aneuploidy Group, IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Correspondence: (E.L.); (J.B.)
| | - José Bessa
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (F.J.F.); (L.C.)
- Vertebrate Development and Regeneration Group, IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Correspondence: (E.L.); (J.B.)
| |
Collapse
|
22
|
Bhattacharya A, Champramary S, Tripathi T, Thakur D, Ioshikhes I, Singh SK, Nandi S. Identification of the conserved long non-coding RNAs in myogenesis. BMC Genomics 2021; 22:336. [PMID: 33971818 PMCID: PMC8112034 DOI: 10.1186/s12864-021-07615-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 04/14/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Our understanding of genome regulation is ever-evolving with the continuous discovery of new modes of gene regulation, and transcriptomic studies of mammalian genomes have revealed the presence of a considerable population of non-coding RNA molecules among the transcripts expressed. One such non-coding RNA molecule is long non-coding RNA (lncRNA). However, the function of lncRNAs in gene regulation is not well understood; moreover, finding conserved lncRNA across species is a challenging task. Therefore, we propose a novel approach to identify conserved lncRNAs and functionally annotate these molecules. RESULTS In this study, we exploited existing myogenic transcriptome data and identified conserved lncRNAs in mice and humans. We identified the lncRNAs expressing differentially between the early and later stages of muscle development. Differential expression of these lncRNAs was confirmed experimentally in cultured mouse muscle C2C12 cells. We utilized the three-dimensional architecture of the genome and identified topologically associated domains for these lncRNAs. Additionally, we correlated the expression of genes in domains for functional annotation of these trans-lncRNAs in myogenesis. Using this approach, we identified conserved lncRNAs in myogenesis and functionally annotated them. CONCLUSIONS With this novel approach, we identified the conserved lncRNAs in myogenesis in humans and mice and functionally annotated them. The method identified a large number of lncRNAs are involved in myogenesis. Further studies are required to investigate the reason for the conservation of the lncRNAs in human and mouse while their sequences are dissimilar. Our approach can be used to identify novel lncRNAs conserved in different species and functionally annotated them.
Collapse
Affiliation(s)
- Anupam Bhattacharya
- Division of Life Sciences, Institute of Advanced Study in Science and Technology, Vigyan Path, Paschim Boragaon, Garchuk, Guwahati, Assam, India
- Department of Molecular Biology and Biotechnology, Cotton University, Panbazar, Guwahati, Assam, India
| | - Simang Champramary
- University of Szeged Faculty of Science and Informatics, Szeged, 6720, Hungary
- Functional Genomics and Bionformatics, University of Sopron, Sopron, Hungary
| | - Tanya Tripathi
- Stem Cell & Cell Culture Lab, Centre For Advanced Research (CFAR), King George's Medical University, Lucknow, UP, India
| | - Debajit Thakur
- Division of Life Sciences, Institute of Advanced Study in Science and Technology, Vigyan Path, Paschim Boragaon, Garchuk, Guwahati, Assam, India
| | - Ilya Ioshikhes
- Ottawa Institute of Computational Biology and Bioinformatics (OICBB), Ottawa Institute of Systems Biology (OISB), Department of Biochemistry, Microbiology and Immunology (BMI),Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Satyendra Kumar Singh
- Stem Cell & Cell Culture Lab, Centre For Advanced Research (CFAR), King George's Medical University, Lucknow, UP, India
| | - Soumyadeep Nandi
- Data Sciences and Computational Biology Centre, Amity Institute of Integrative Sciences and Health, Amity University Haryana, Gurugram, Manesar, 122413, Haryana, India.
| |
Collapse
|
23
|
Zullo L, Bozzo M, Daya A, Di Clemente A, Mancini FP, Megighian A, Nesher N, Röttinger E, Shomrat T, Tiozzo S, Zullo A, Candiani S. The Diversity of Muscles and Their Regenerative Potential across Animals. Cells 2020; 9:cells9091925. [PMID: 32825163 PMCID: PMC7563492 DOI: 10.3390/cells9091925] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023] Open
Abstract
Cells with contractile functions are present in almost all metazoans, and so are the related processes of muscle homeostasis and regeneration. Regeneration itself is a complex process unevenly spread across metazoans that ranges from full-body regeneration to partial reconstruction of damaged organs or body tissues, including muscles. The cellular and molecular mechanisms involved in regenerative processes can be homologous, co-opted, and/or evolved independently. By comparing the mechanisms of muscle homeostasis and regeneration throughout the diversity of animal body-plans and life cycles, it is possible to identify conserved and divergent cellular and molecular mechanisms underlying muscle plasticity. In this review we aim at providing an overview of muscle regeneration studies in metazoans, highlighting the major regenerative strategies and molecular pathways involved. By gathering these findings, we wish to advocate a comparative and evolutionary approach to prompt a wider use of “non-canonical” animal models for molecular and even pharmacological studies in the field of muscle regeneration.
Collapse
Affiliation(s)
- Letizia Zullo
- Istituto Italiano di Tecnologia, Center for Micro-BioRobotics & Center for Synaptic Neuroscience and Technology (NSYN), 16132 Genova, Italy;
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- Correspondence: (L.Z.); (A.Z.)
| | - Matteo Bozzo
- Laboratory of Developmental Neurobiology, Department of Earth, Environment and Life Sciences, University of Genova, Viale Benedetto XV 5, 16132 Genova, Italy; (M.B.); (S.C.)
| | - Alon Daya
- Faculty of Marine Sciences, Ruppin Academic Center, Michmoret 40297, Israel; (A.D.); (N.N.); (T.S.)
| | - Alessio Di Clemente
- Istituto Italiano di Tecnologia, Center for Micro-BioRobotics & Center for Synaptic Neuroscience and Technology (NSYN), 16132 Genova, Italy;
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genova, Italy
| | | | - Aram Megighian
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy;
- Padova Neuroscience Center, University of Padova, 35131 Padova, Italy
| | - Nir Nesher
- Faculty of Marine Sciences, Ruppin Academic Center, Michmoret 40297, Israel; (A.D.); (N.N.); (T.S.)
| | - Eric Röttinger
- Institute for Research on Cancer and Aging (IRCAN), Université Côte d’Azur, CNRS, INSERM, 06107 Nice, France;
| | - Tal Shomrat
- Faculty of Marine Sciences, Ruppin Academic Center, Michmoret 40297, Israel; (A.D.); (N.N.); (T.S.)
| | - Stefano Tiozzo
- Laboratoire de Biologie du Développement de Villefranche-sur-Mer (LBDV), Sorbonne Université, CNRS, 06230 Paris, France;
| | - Alberto Zullo
- Department of Science and Technology, University of Sannio, 82100 Benevento, Italy;
- Correspondence: (L.Z.); (A.Z.)
| | - Simona Candiani
- Laboratory of Developmental Neurobiology, Department of Earth, Environment and Life Sciences, University of Genova, Viale Benedetto XV 5, 16132 Genova, Italy; (M.B.); (S.C.)
| |
Collapse
|
24
|
Carrera M, Piñeiro C, Martinez I. Proteomic Strategies to Evaluate the Impact of Farming Conditions on Food Quality and Safety in Aquaculture Products. Foods 2020; 9:E1050. [PMID: 32759674 PMCID: PMC7466198 DOI: 10.3390/foods9081050] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 02/08/2023] Open
Abstract
This review presents the primary applications of various proteomic strategies to evaluate the impact of farming conditions on food quality and safety in aquaculture products. Aquaculture is a quickly growing sector that represents 47% of total fish production. Food quality, dietary management, fish welfare, the stress response, food safety, and antibiotic resistance, which are covered by this review, are among the primary topics in which proteomic techniques and strategies are being successfully applied. The review concludes by outlining future directions and potential perspectives.
Collapse
Affiliation(s)
- Mónica Carrera
- Food Technology Department, Institute of Marine Research (IIM), Spanish National Research Council (CSIC), 36208 Vigo, Pontevedra, Spain
| | - Carmen Piñeiro
- Scientific Instrumentation and Quality Service (SICIM), Institute of Marine Research (IIM), Spanish National Research Council (CSIC), 36208 Vigo, Pontevedra, Spain;
| | - Iciar Martinez
- Research Centre for Experimental Marine Biology and Biotechnology—Plentzia Marine Station (PiE), University of the Basque Country UPV/EHU, 48620 Plentzia, Spain;
- IKERBASQUE Basque Foundation for Science, 48013 Bilbao, Spain
| |
Collapse
|
25
|
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
|
26
|
Ruparelia AA, Ratnayake D, Currie PD. Stem cells in skeletal muscle growth and regeneration in amniotes and teleosts: Emerging themes. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 9:e365. [PMID: 31743958 DOI: 10.1002/wdev.365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/22/2019] [Accepted: 10/03/2019] [Indexed: 12/19/2022]
Abstract
Skeletal muscle is a contractile, postmitotic tissue that retains the capacity to grow and regenerate throughout life in amniotes and teleost. Both muscle growth and regeneration are regulated by obligate tissue resident muscle stem cells. Given that considerable knowledge exists on the myogenic process, recent studies have focused on examining the molecular markers of muscle stem cells, and on the intrinsic and extrinsic signals regulating their function. From this, two themes emerge: firstly, muscle stem cells display remarkable heterogeneity not only with regards to their gene expression profile, but also with respect to their behavior and function; and secondly, the stem cell niche is a critical regulator of muscle stem cell function during growth and regeneration. Here, we will address the current understanding of these emerging themes with emphasis on the distinct processes used by amniotes and teleost, and discuss the challenges and opportunities in the muscle growth and regeneration fields. This article is characterized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Tissue Stem Cells and Niches Early Embryonic Development > Development to the Basic Body Plan Vertebrate Organogenesis > Musculoskeletal and Vascular.
Collapse
Affiliation(s)
- Avnika A Ruparelia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia.,EMBL Australia, Monash University, Melbourne, Victoria, Australia
| | - Dhanushika Ratnayake
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia.,EMBL Australia, Monash University, Melbourne, Victoria, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia.,EMBL Australia, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
27
|
Cassar S, Beekhuijzen M, Beyer B, Chapin R, Dorau M, Hoberman A, Krupp E, Leconte I, Stedman D, Stethem C, van den Oetelaar D, Tornesi B. A multi-institutional study benchmarking the zebrafish developmental assay for prediction of embryotoxic plasma concentrations from rat embryo-fetal development studies. Reprod Toxicol 2019; 86:33-44. [PMID: 30876927 DOI: 10.1016/j.reprotox.2019.02.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 12/24/2018] [Accepted: 02/08/2019] [Indexed: 01/11/2023]
Abstract
Predicting embryotoxicity of pharmaceutical compounds or industrial chemicals is crucial for public safety. Conventional studies which monitor embryo-fetal development in rats and rabbits are costly and time consuming. Alternative assays which are simpler and less costly are being pursued. The purpose of this research was to assess the capacity for the zebrafish development assay to predict mammalian plasma levels that are embryotoxic. Previously published data on rat plasma levels associated with embryotoxicity were used to guide concentration ranges for each of 25 chemicals dissolved in the media bathing developing zebrafish embryos. Embryotoxic media concentrations were compared to embryotoxic rat plasma concentrations. Assays were conducted in parallel at multiple sites as a consortium effort through the Health and Environmental Sciences Institute (HESI). Considering results from all sites, the zebrafish embryo development assay predicted (within 1-log) the rat maternal exposure levels associated with embryotoxicity 75% of the time.
Collapse
|
28
|
Pluripotent stem cell-derived myogenic progenitors remodel their molecular signature upon in vivo engraftment. Proc Natl Acad Sci U S A 2019; 116:4346-4351. [PMID: 30760602 DOI: 10.1073/pnas.1808303116] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Optimal cell-based therapies for the treatment of muscle degenerative disorders should not only regenerate fibers but provide a quiescent satellite cell pool ensuring long-term maintenance and regeneration. Conditional expression of Pax3/Pax7 in differentiating pluripotent stem cells (PSCs) allows the generation of myogenic progenitors endowed with enhanced regenerative capacity. To identify the molecular determinants underlying their regenerative potential, we performed transcriptome analyses of these cells along with primary myogenic cells from several developmental stages. Here we show that in vitro-generated PSC-derived myogenic progenitors possess a molecular signature similar to embryonic/fetal myoblasts. However, compared with fetal myoblasts, following transplantation they show superior myofiber engraftment and ability to seed the satellite cell niche, respond to multiple reinjuries, and contribute to long-term regeneration. Upon engraftment, the transcriptome of reisolated Pax3/Pax7-induced PSC-derived myogenic progenitors changes toward a postnatal molecular signature, particularly in genes involved in extracellular matrix remodeling. These findings demonstrate that Pax3/Pax7-induced myogenic progenitors remodel their molecular signature and functionally mature upon in vivo exposure to the adult muscle environment.
Collapse
|
29
|
Wang T, Wang X, Zhou H, Jiang H, Mai K, He G. The Mitotic and Metabolic Effects of Phosphatidic Acid in the Primary Muscle Cells of Turbot ( Scophthalmus maximus). Front Endocrinol (Lausanne) 2018; 9:221. [PMID: 29780359 PMCID: PMC5946094 DOI: 10.3389/fendo.2018.00221] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 04/19/2018] [Indexed: 12/11/2022] Open
Abstract
Searching for nutraceuticals and understanding the underlying mechanism that promote fish growth is at high demand for aquaculture industry. In this study, the modulatory effects of soy phosphatidic acids (PA) on cell proliferation, nutrient sensing, and metabolic pathways were systematically examined in primary muscle cells of turbot (Scophthalmus maximus). PA was found to stimulate cell proliferation and promote G1/S phase transition through activation of target of rapamycin signaling pathway. The expression of myogenic regulatory factors, including myoD and follistatin, was upregulated, while that of myogenin and myostatin was downregulated by PA. Furthermore, PA increased intracellular free amino acid levels and enhanced protein synthesis, lipogenesis, and glycolysis, while suppressed amino acid degradation and lipolysis. PA also was found to increased cellular energy production through stimulated tricarboxylic acid cycle and oxidative phosphorylation. Our results identified PA as a potential nutraceutical that stimulates muscle cell proliferation and anabolism in fish.
Collapse
Affiliation(s)
- Tingting Wang
- Key Laboratory of Aquaculture Nutrition, Ministry of Agriculture, Ocean University of China, Qingdao, China
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Xuan Wang
- Key Laboratory of Aquaculture Nutrition, Ministry of Agriculture, Ocean University of China, Qingdao, China
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Huihui Zhou
- Key Laboratory of Aquaculture Nutrition, Ministry of Agriculture, Ocean University of China, Qingdao, China
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Haowen Jiang
- Key Laboratory of Aquaculture Nutrition, Ministry of Agriculture, Ocean University of China, Qingdao, China
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition, Ministry of Agriculture, Ocean University of China, Qingdao, China
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Gen He
- Key Laboratory of Aquaculture Nutrition, Ministry of Agriculture, Ocean University of China, Qingdao, China
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
- *Correspondence: Gen He,
| |
Collapse
|
30
|
The histone code reader Spin1 controls skeletal muscle development. Cell Death Dis 2017; 8:e3173. [PMID: 29168801 PMCID: PMC5775400 DOI: 10.1038/cddis.2017.468] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/12/2017] [Accepted: 07/28/2017] [Indexed: 01/06/2023]
Abstract
While several studies correlated increased expression of the histone code reader Spin1 with tumor formation or growth, little is known about physiological functions of the protein. We generated Spin1M5 mice with ablation of Spin1 in myoblast precursors using the Myf5-Cre deleter strain. Most Spin1M5 mice die shortly after birth displaying severe sarcomere disorganization and necrosis. Surviving Spin1M5 mice are growth-retarded and exhibit the most prominent defects in soleus, tibialis anterior, and diaphragm muscle. Transcriptome analyses of limb muscle at embryonic day (E) 15.5, E16.5, and at three weeks of age provided evidence for aberrant fetal myogenesis and identified deregulated skeletal muscle (SkM) functional networks. Determination of genome-wide chromatin occupancy in primary myoblast revealed direct Spin1 target genes and suggested that deregulated basic helix-loop-helix transcription factor networks account for developmental defects in Spin1M5 fetuses. Furthermore, correlating histological and transcriptome analyses, we show that aberrant expression of titin-associated proteins, abnormal glycogen metabolism, and neuromuscular junction defects contribute to SkM pathology in Spin1M5 mice. Together, we describe the first example of a histone code reader controlling SkM development in mice, which hints at Spin1 as a potential player in human SkM disease.
Collapse
|
31
|
The circadian transcriptome of marine fish (Sparus aurata) larvae reveals highly synchronized biological processes at the whole organism level. Sci Rep 2017; 7:12943. [PMID: 29021622 PMCID: PMC5636797 DOI: 10.1038/s41598-017-13514-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 09/22/2017] [Indexed: 02/07/2023] Open
Abstract
The regulation of circadian gene expression remains largely unknown in farmed fish larvae. In this study, a high-density oligonucleotide microarray was used to examine the daily expression of 13,939 unique genes in whole gilthead sea bream (Sparus aurata) larvae with fast growth potentiality. Up to 2,229 genes were differentially expressed, and the first two components of Principal Component Analysis explained more than 81% of the total variance. Clustering analysis of differentially expressed genes identified 4 major clusters that were triggered sequentially, with a maximum expression at 0 h, 3 h, 9–15 h and 18-21 h zeitgeber time. Various core clock genes (per1, per2, per3, bmal1, cry1, cry2, clock) were identified in clusters 1–3, and their expression was significantly correlated with several genes in each cluster. Functional analysis revealed a daily consecutive activation of canonical pathways related to phototransduction, intermediary metabolism, development, chromatin remodeling, and cell cycle regulation. This daily transcriptome of whole larvae resembles a cell cycle (G1/S, G2/M, and M/G1 transitions) in synchronization with multicellular processes, such as neuromuscular development. This study supports that the actively feeding fish larval transcriptome is temporally organized in a 24-h cycle, likely for maximizing growth and development.
Collapse
|
32
|
Comprehensive analysis of lncRNAs and mRNAs in skeletal muscle of rainbow trout (Oncorhynchus mykiss) exposed to estradiol. Sci Rep 2017; 7:11780. [PMID: 28924252 PMCID: PMC5603547 DOI: 10.1038/s41598-017-12136-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 09/05/2017] [Indexed: 02/01/2023] Open
Abstract
Estradiol (E2) is a steroid hormone that negatively affects muscle growth in rainbow trout (Oncorhynchus mykiss), but the mechanisms directing with this response are not fully understood. To better characterize the effects of E2 in muscle, we identified differentially regulated mRNAs and lncRNAs in juvenile rainbow trout exposed to E2. Here, we performed next-generation RNA sequencing and comprehensive bioinformatics analyses to characterize the transcriptome profiles, including mRNAs and long noncoding RNAs (lncRNAs), in skeletal muscle of rainbow trout injected with E2. A total of 226 lncRNAs and 253 mRNAs were identified as differentially regulated. We identified crucial pathways, including several signal transduction pathways, hormone response, oxidative response and protein, carbon and fatty acid metabolism pathways. Subsequently, a functional lncRNA-mRNA co-expression network was constructed, which consisted of 681 co-expression relationships between 164 lncRNAs and 201 mRNAs. Moreover, a lncRNA-pathway network was constructed. A total of 65 key lncRNAs were identified that regulate 20 significantly enriched pathways. Overall, our analysis provides insights into mRNA and lncRNA networks in rainbow trout skeletal muscle and their regulation by E2 while understanding the molecular mechanism of lncRNAs.
Collapse
|
33
|
Fuentes EN, Zuloaga R, Almarza O, Mendez K, Valdés JA, Molina A, Pulgar J. Upwelling-derived oceanographic conditions impact growth performance and growth-related gene expression in intertidal fish. Comp Biochem Physiol B Biochem Mol Biol 2017; 214:12-18. [PMID: 28899845 DOI: 10.1016/j.cbpb.2017.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/29/2017] [Accepted: 09/05/2017] [Indexed: 12/30/2022]
Abstract
Growth is one of the main biological processes in aquatic organisms that is affected by environmental fluctuations such as upwelling (characterized by food-rich waters). In fish, growth is directly related with skeletal muscle increase; which represents the largest tissue of body mass. However, the effects of upwelling on growth, at the physiological and molecular level, are unknown. This study used Girella laevifrons (one of the most abundant intertidal fish in Eastern South Pacific) as a biological model, considering animals from upwelling (U) and non-upwelling (NU) areas. Here, we evaluated the effect of nutritional composition and food availability on growth performance and expression of key growth-related genes (insulin-kike growth factor 1 (igf1) and myosin heavy-chain (myhc)) and atrophy-related genes (muscle ring-finger 1 (murf1), F-box only protein 32 (atrogin-1) and BCL2/adenovirus E1B 19kDa-interacting protein 3 (bnip3)). We reported that, among zones, U fish displayed higher growth performance in response to nutritional composition, specifically between protein- and fiber-rich diets (~1g). We also found in NU fish that atrophy-related genes were upregulated with fiber-rich diet and during fasting (~2-fold at minimum respect U). In conclusion, our results suggest that the growth potential of upwelling fish may be a consequence of differential muscle gene expression. Our data provide a preliminary approach contributing on how upwelling influence fish growth at the physiological and molecular levels. Future studies are required to gain further knowledge about molecular differences between U and NU animals, as well as the possible applications of this knowledge in the aquaculture industry.
Collapse
Affiliation(s)
- Eduardo N Fuentes
- Interdisciplinary Center for Aquaculture Research (INCAR), Víctor Lamas 1290, PO Box 160-C, Concepción 4030000, Chile
| | - Rodrigo Zuloaga
- Interdisciplinary Center for Aquaculture Research (INCAR), Víctor Lamas 1290, PO Box 160-C, Concepción 4030000, Chile; Laboratorio de Biotecnología Molecular, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Av. Republica 217, Santiago 8370371, Chile
| | - Oscar Almarza
- Interdisciplinary Center for Aquaculture Research (INCAR), Víctor Lamas 1290, PO Box 160-C, Concepción 4030000, Chile; Laboratorio de Biotecnología Molecular, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Av. Republica 217, Santiago 8370371, Chile
| | - Katterinne Mendez
- Laboratorio de Biotecnología Molecular, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Av. Republica 217, Santiago 8370371, Chile
| | - Juan Antonio Valdés
- Interdisciplinary Center for Aquaculture Research (INCAR), Víctor Lamas 1290, PO Box 160-C, Concepción 4030000, Chile; Laboratorio de Biotecnología Molecular, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Av. Republica 217, Santiago 8370371, Chile
| | - Alfredo Molina
- Interdisciplinary Center for Aquaculture Research (INCAR), Víctor Lamas 1290, PO Box 160-C, Concepción 4030000, Chile; Laboratorio de Biotecnología Molecular, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Av. Republica 217, Santiago 8370371, Chile
| | - Jose Pulgar
- Departamento de Ecología & Biodiversidad, Universidad Andrés Bello, República 470, Santiago 8370371, Chile.
| |
Collapse
|
34
|
Quantification of Embryonic Myofiber Development by Immunofluorescence. Methods Mol Biol 2017. [PMID: 28842909 DOI: 10.1007/978-1-4939-7283-8_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Mammalian myogenesis occurs in two distinct phases, primary and secondary, which are temporally separated. The primary wave occurs shortly after somitogenesis producing embryonic myofibers. The secondary wave occurs after somitogenesis producing fetal myofibers that form adjacent to the embryonic myofibers. The myogenic cells that give rise to these two waves have distinct characteristics as do the myofibers they produce. The objective of this chapter is to describe our methods for quantifying embryonic and fetal myofiber development in mouse embryos using immunofluorescence.
Collapse
|
35
|
Lewandowski D, Dubińska-Magiera M, Posyniak E, Rupik W, Daczewska M. Does the grass snake (Natrix natrix) (Squamata: Serpentes: Natricinae) fit the amniotes-specific model of myogenesis? PROTOPLASMA 2017; 254:1507-1516. [PMID: 27834030 PMCID: PMC5487930 DOI: 10.1007/s00709-016-1040-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 10/27/2016] [Indexed: 06/06/2023]
Abstract
In the grass snake (Natrix natrix), the newly developed somites form vesicles that are located on both sides of the neural tube. The walls of the vesicles are composed of tightly connected epithelial cells surrounding the cavity (the somitocoel). Also, in the newly formed somites, the Pax3 protein can be observed in the somite wall cells. Subsequently, the somite splits into three compartments: the sclerotome, dermomyotome (with the dorsomedial [DM] and the ventrolateral [VL] lips) and the myotome. At this stage, the Pax3 protein is detected in both the DM and VL lips of the dermomyotome and in the mononucleated cells of the myotome, whereas the Pax7 protein is observed in the medial part of the dermomyotome and in some of the mononucleated cells of the myotome. The mononucleated cells then become elongated and form myotubes. As myogenesis proceeds, the myotome is filled with multinucleated myotubes accompanied by mononucleated, Pax7-positive cells (satellite cells) that are involved in muscle growth. The Pax3-positive progenitor muscle cells are no longer observed. Moreover, we have observed unique features in the differentiation of the muscles in these snakes. Specifically, our studies have revealed the presence of two classes of muscles in the myotomes. The first class is characterised by fast muscle fibres, with myofibrils equally distributed throughout the sarcoplasm. In the second class, composed of slow muscle fibres, the sarcoplasm is filled with lipid droplets. We assume that their storage could play a crucial role during hibernation in the adult snakes. We suggest that the model of myotomal myogenesis in reptiles, birds and mammals shows the same morphological and molecular character. We therefore believe that the grass snake, in spite of the unique features of its myogenesis, fits into the amniotes-specific model of trunk muscle development.
Collapse
Affiliation(s)
- Damian Lewandowski
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, Sienkiewicza 21, 50-335, Wroclaw, Poland
| | - Magda Dubińska-Magiera
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, Sienkiewicza 21, 50-335, Wroclaw, Poland
| | - Ewelina Posyniak
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, Sienkiewicza 21, 50-335, Wroclaw, Poland
| | - Weronika Rupik
- Department of Animal Histology and Embryology, University of Silesia, Bankowa 9, 40-007, Katowice, Poland
| | - Małgorzata Daczewska
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, Sienkiewicza 21, 50-335, Wroclaw, Poland.
| |
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
|
Kumar A, Singh N, Goswami M, Srivastava JK, Mishra AK, Lakra WS. Establishment and Characterization of a New Muscle Cell Line of Zebrafish (Danio rerio) as an In Vitro Model for Gene Expression Studies. Anim Biotechnol 2016; 27:166-73. [DOI: 10.1080/10495398.2016.1147455] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Amit Kumar
- Molecular Biology and Biotechnology Division, National Bureau of Fish Genetic Resources, Lucknow, India
| | - Neha Singh
- Amity University, Uttar Pradesh, Lucknow Campus, Lucknow, India
| | - Mukunda Goswami
- Central Institute of Fisheries Education, Versova, Mumbai, India
| | | | - Akhilesh K. Mishra
- Molecular Biology and Biotechnology Division, National Bureau of Fish Genetic Resources, Lucknow, India
| | - W. S. Lakra
- Central Institute of Fisheries Education, Versova, Mumbai, India
| |
Collapse
|
38
|
Khannoon ER, Rupik W, Lewandowski D, Dubińska-Magiera M, Swadźba E, Daczewska M. Unique features of myogenesis in Egyptian cobra (Naja haje) (Squamata: Serpentes: Elapidae). PROTOPLASMA 2016; 253:625-33. [PMID: 26025263 PMCID: PMC4783446 DOI: 10.1007/s00709-015-0840-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 05/21/2015] [Indexed: 06/04/2023]
Abstract
During early stages of myotomal myogenesis, the myotome of Egyptian cobra (Naja haje) is composed of homogenous populations of mononucleated primary myotubes. At later developmental phase, primary myotubes are accompanied by closely adhering mononucleated cells. Based on localization and morphology, we assume that mononucleated cells share features with satellite cells involved in muscle growth. An indirect morphological evidence of the fusion of mononucleated cells with myotubes is the presence of numerous vesicles in the subsarcolemmal region of myotubes adjacent to mononucleated cell. As differentiation proceeded, secondary muscle fibres appeared with considerably smaller diameter as compared to primary muscle fibre. Studies on N. haje myotomal myogenesis revealed some unique features of muscle differentiation. TEM analysis showed in the N. haje myotomes two classes of muscle fibres. The first class was characterized by typical for fast muscle fibres regular distribution of myofibrils which fill the whole volume of muscle fibre sarcoplasm. White muscle fibres in studied species were a prominent group of muscles in the myotome. The second class showed tightly paced myofibrils surrounding the centrally located nucleus accompanied by numerous vesicles of different diameter. The sarcoplasm of these cells was characterized by numerous lipid droplets. Based on morphological features, we believe that muscle capable of lipid storage belong to slow muscle fibres and the presence of lipid droplets in the sarcoplasm of these muscles during myogenesis might be a crucial adaptive mechanisms for subsequent hibernation in adults. This phenomenon was, for the first time, described in studies on N. haje myogenesis.
Collapse
Affiliation(s)
- Eraqi R Khannoon
- Zoology Department, Faculty of Science, Fayoum University, Fayoum, 63514, Egypt
| | - Weronika Rupik
- Department of Animal Histology and Embryology, University of Silesia, 9 Bankowa Str., 40-007, Katowice, Poland
| | - Damian Lewandowski
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wrocław, 21 Sienkiewicza Str., 53-335, Wrocław, Poland
| | - Magda Dubińska-Magiera
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wrocław, 21 Sienkiewicza Str., 53-335, Wrocław, Poland
| | - Elwira Swadźba
- Department of Animal Histology and Embryology, University of Silesia, 9 Bankowa Str., 40-007, Katowice, Poland
| | - Małgorzata Daczewska
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wrocław, 21 Sienkiewicza Str., 53-335, Wrocław, Poland.
| |
Collapse
|
39
|
Nagandla H, Lopez S, Yu W, Rasmussen TL, Tucker HO, Schwartz RJ, Stewart MD. Defective myogenesis in the absence of the muscle-specific lysine methyltransferase SMYD1. Dev Biol 2015; 410:86-97. [PMID: 26688546 DOI: 10.1016/j.ydbio.2015.12.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 12/07/2015] [Accepted: 12/07/2015] [Indexed: 11/19/2022]
Abstract
The SMYD (SET and MYND domain) family of lysine methyltransferases harbor a unique structure in which the methyltransferase (SET) domain is intervened by a zinc finger protein-protein interaction MYND domain. SMYD proteins methylate both histone and non-histone substrates and participate in diverse biological processes including transcriptional regulation, DNA repair, proliferation and apoptosis. Smyd1 is unique among the five family members in that it is specifically expressed in striated muscles. Smyd1 is critical for development of the right ventricle in mice. In zebrafish, Smyd1 is necessary for sarcomerogenesis in fast-twitch muscles. Smyd1 is expressed in the skeletal muscle lineage throughout myogenesis and in mature myofibers, shuttling from nucleus to cytosol during myoblast differentiation. Because of this expression pattern, we hypothesized that Smyd1 plays multiple roles at different stages of myogenesis. To determine the role of Smyd1 in mammalian myogenesis, we conditionally eliminated Smyd1 from the skeletal muscle lineage at the myoblast stage using Myf5(cre). Deletion of Smyd1 impaired myoblast differentiation, resulted in fewer myofibers and decreased expression of muscle-specific genes. Muscular defects were temporally restricted to the second wave of myogenesis. Thus, in addition to the previously described functions for Smyd1 in heart development and skeletal muscle sarcomerogenesis, these results point to a novel role for Smyd1 in myoblast differentiation.
Collapse
Affiliation(s)
- Harika Nagandla
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Suhujey Lopez
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Wei Yu
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Tara L Rasmussen
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA; Molecular Biosciences and Institute for Cellular and Molecular Biology, University of Texas, Austin, TX, USA
| | - Haley O Tucker
- Molecular Biosciences and Institute for Cellular and Molecular Biology, University of Texas, Austin, TX, USA
| | - Robert J Schwartz
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA; Stem Cell Engineering Department, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, TX, USA
| | - M David Stewart
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA.
| |
Collapse
|
40
|
Aedo JE, Maldonado J, Aballai V, Estrada JM, Bastias-Molina M, Meneses C, Gallardo-Escarate C, Silva H, Molina A, Valdés JA. mRNA-seq reveals skeletal muscle atrophy in response to handling stress in a marine teleost, the red cusk-eel (Genypterus chilensis). BMC Genomics 2015; 16:1024. [PMID: 26626593 PMCID: PMC4667402 DOI: 10.1186/s12864-015-2232-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 11/19/2015] [Indexed: 01/07/2023] Open
Abstract
Background Fish reared under intensive conditions are repeatedly exposed to stress, which negatively impacts growth. Although most fish follow a conserved pattern of stress response, with increased concentrations of cortisol, each species presents specificities in the cell response and stress tolerance. Therefore, culturing new species requires a detailed knowledge of these specific responses. The red cusk-eel (Genypterus chilensis) is a new economically important marine species for the Chilean aquaculture industry. However, there is no information on the stress- and cortisol-induced mechanisms that decrease skeletal muscle growth in this teleost. Results Using Illumina RNA-seq technology, skeletal muscle sequence reads for G. chilensis were generated under control and handling stress conditions. Reads were mapped onto a reference transcriptome, resulting in the in silico identification of 785 up-regulated and 167 down-regulated transcripts. Gene ontology enrichment analysis revealed a significant up-regulation of catabolic genes associated with skeletal muscle atrophy. These results were validated by RT-qPCR analysis for ten candidates genes involved in ubiquitin-mediated proteolysis, autophagy and skeletal muscle growth. Additionally, using a primary culture of fish skeletal muscle cells, the effect of cortisol was evaluated in relation to red cusk-eel skeletal muscle atrophy. Conclusions The present data demonstrated that handling stress promotes skeletal muscle atrophy in the marine teleost G. chilensis through the expression of components of the ubiquitin-proteasome and autophagy-lysosome systems. Furthermore, cortisol was a powerful inductor of skeletal muscle atrophy in fish myotubes. This study is an important step towards understanding the atrophy system in non-model teleost species and provides novel insights on the cellular and molecular mechanisms that control skeletal muscle growth in early vertebrates. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2232-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jorge E Aedo
- Laboratorio de Biotecnología Molecular, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago, Chile
| | - Jonathan Maldonado
- Departamento de Producción Agrícola, Laboratorio de Genómica Funcional & Bioinformática, Universidad de Chile, Facultad de Ciencias Agronómicas, Av. Santa Rosa 11315, La Pintana, 8820808, Santiago, Chile
| | - Víctor Aballai
- Laboratorio de Biotecnología Molecular, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago, Chile
| | - Juan M Estrada
- Centro de Investigación Marina Quintay (CIMARQ), Universidad Andrés Bello, Quintay, Chile
| | - Macarena Bastias-Molina
- Centro de Biotecnología Vegetal, Facultad Ciencias Biológicas, Universidad Andrés Bello, Santiago, Chile
| | - Claudio Meneses
- Centro de Biotecnología Vegetal, Facultad Ciencias Biológicas, Universidad Andrés Bello, Santiago, Chile
| | - Cristian Gallardo-Escarate
- Laboratory of Biotechnology and Aquatic Genomics, Universidad de Concepción, Concepción, Chile.,Interdisciplinary Center for Aquaculture Research (INCAR), P.O. Box 160-C, Concepción, Chile
| | - Herman Silva
- Departamento de Producción Agrícola, Laboratorio de Genómica Funcional & Bioinformática, Universidad de Chile, Facultad de Ciencias Agronómicas, Av. Santa Rosa 11315, La Pintana, 8820808, Santiago, Chile
| | - Alfredo Molina
- Laboratorio de Biotecnología Molecular, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago, Chile.,Interdisciplinary Center for Aquaculture Research (INCAR), P.O. Box 160-C, Concepción, Chile.,Centro de Investigación Marina Quintay (CIMARQ), Universidad Andrés Bello, Quintay, Chile
| | - Juan A Valdés
- Laboratorio de Biotecnología Molecular, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago, Chile. .,Interdisciplinary Center for Aquaculture Research (INCAR), P.O. Box 160-C, Concepción, Chile. .,Centro de Investigación Marina Quintay (CIMARQ), Universidad Andrés Bello, Quintay, Chile.
| |
Collapse
|
41
|
Role of PRDM16 and its PR domain in the epigenetic regulation of myogenic and adipogenic genes during transdifferentiation of C2C12 cells. Gene 2015; 570:191-8. [DOI: 10.1016/j.gene.2015.06.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/02/2015] [Accepted: 06/05/2015] [Indexed: 12/18/2022]
|
42
|
Dyment NA, Galloway JL. Regenerative biology of tendon: mechanisms for renewal and repair. ACTA ACUST UNITED AC 2015; 1:124-131. [PMID: 26389023 DOI: 10.1007/s40610-015-0021-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Understanding the molecular and cellular mechanisms underlying tissue turnover and repair are essential towards addressing pathologies in aging, injury and disease. Each tissue has distinct means of maintaining homeostasis and healing after injury. For some, resident stem cell populations mediate both of these processes. These stem cells, by definition, are self renewing and give rise to all the differentiated cells of that tissue. However, not all organs fit with this traditional stem cell model of regeneration, and some do not appear to harbor somatic stem or progenitor cells capable of multilineage in vivo reconstitution. Despite recent progress in tendon progenitor cell research, our current knowledge of the mechanisms regulating tendon cell homeostasis and injury response is limited. Understanding the role of resident tendon cell populations is of great importance for regenerative medicine based approaches to tendon injuries and disease. The goal of this review is to bring to light our current knowledge regarding tendon progenitor cells and their role in tissue maintenance and repair. We will focus on pressing questions in the field and the new tools, including model systems, available to address them.
Collapse
Affiliation(s)
- Nathaniel A Dyment
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center
| | - Jenna L Galloway
- Center for Regenerative Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Harvard Stem Cell Institute
| |
Collapse
|
43
|
Oncorhynchus mykiss pax7 sequence variations with comparative analyses against other teleost species. SPRINGERPLUS 2015; 4:263. [PMID: 26090310 PMCID: PMC4469688 DOI: 10.1186/s40064-015-1030-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 05/12/2015] [Indexed: 12/24/2022]
Abstract
The paired box-7 (pax7) transcription factor expressed in satellite cells (SCs) is an essential regulator of skeletal muscle growth and regeneration in vertebrates including fish. Characterization of rainbow trout (Oncorhynchus mykiss) pax7 gene/s may offer novel insights into skeletal myogenesis by SCs in this indeterminate growth species. Further, evaluation of promoters for cis-regulatory regions may shed light on the evolutionary fate of the duplicated genes. Employing standard PCR, cloning and computational approach, we identified and report complete coding sequences of two pax7 paralogs of rainbow trout (rt); rtpax7α and rtpax7β. Both genes show significant identity in the nucleotide (97%) and the predicted amino acid (98%) sequences, and bear the characteristic paired domain (PD), octapeptide (OP) and homeodomain (HD) motifs. We further report several splice variants of each gene and nucleotide differences in coding sequence that predicts six putative amino acid changes between the two genes. Additionally, we noted a trinucleotide deletion in rtpax7β that results in putative serine elimination at the N-terminus and a single nucleotide polymorphism (SNP) in majority of the rtpax7β variants (6/10) that predicts an arginine substitution for a lysine. We also deciphered the genomic organization up to the first three exons and the upstream putative promoter regions of both genes. Comparative in silico analysis of both the trout pax7 promoters with that of zebrafish pax7 duplicates; zfpax7a and zfpax7b; predicts several important cis-elements/transcription factor binding sites (TFBS) in these teleost pax7 promoter regions.
Collapse
|
44
|
Kim JL, Kong HJ, Kim HS, Kim WJ, Kim DG, Nam BH, Kim YO, An CM. Fish Myogenic Regulatory Protein LUC7L: Characterization and Expression Analysis in Korean Rose Bitterling (Rhodeus uyekii). Dev Reprod 2015; 18:251-8. [PMID: 25949195 DOI: 10.12717/devrep.2014.18.4.251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 11/20/2014] [Accepted: 11/24/2014] [Indexed: 11/17/2022]
Abstract
Serine-arginine-rich nuclear protein LUC7L plays an important role in the regulation of myogenesis in mice. In the present study, we isolated and characterized the Korean rose bitterling Rhodeus uyekii Luc7l cDNA, designated RuLuc7l. The RuLuc7l cDNA is 1,688 bp long and encodes a 364-amino-acid polypeptide containing serine/arginine-rich region at the C-terminus. The deduced RuLuc7l protein has high amino acid identity (71-97%) with those of other species including human. Phylogenetic analysis revealed that RuLUC7L clustered with fish LUC7L proteins. The expression of RuLuc7l mRNA was high in the brain, kidney, and stomach of Korean rose bitterling. Expression of the RuLuc7l mRNA was detected from 1 day post-fertilization (dpf) and moderately increased until 21 dpf during the early development. Further investigations are required to elucidate the functional role of RuLUC7L in myogenesis in R. uyekii.
Collapse
Affiliation(s)
- Ju Lan Kim
- Biotechnology Research Division, National Fisheries Research and Development Institute, Busan 619-705, Korea
| | - Hee Jeong Kong
- Biotechnology Research Division, National Fisheries Research and Development Institute, Busan 619-705, Korea
| | - Hyung Soo Kim
- Biotechnology Research Division, National Fisheries Research and Development Institute, Busan 619-705, Korea
| | - Woo-Jin Kim
- Biotechnology Research Division, National Fisheries Research and Development Institute, Busan 619-705, Korea
| | - Dong-Gyun Kim
- Biotechnology Research Division, National Fisheries Research and Development Institute, Busan 619-705, Korea
| | - Bo-Hye Nam
- Biotechnology Research Division, National Fisheries Research and Development Institute, Busan 619-705, Korea
| | - Young-Ok Kim
- Biotechnology Research Division, National Fisheries Research and Development Institute, Busan 619-705, Korea
| | - Cheul Min An
- Biotechnology Research Division, National Fisheries Research and Development Institute, Busan 619-705, Korea
| |
Collapse
|
45
|
Du M, Wang B, Fu X, Yang Q, Zhu MJ. Fetal programming in meat production. Meat Sci 2015; 109:40-7. [PMID: 25953215 DOI: 10.1016/j.meatsci.2015.04.010] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 04/14/2015] [Accepted: 04/16/2015] [Indexed: 12/23/2022]
Abstract
Nutrient fluctuations during the fetal stage affects fetal development, which has long-term impacts on the production efficiency and quality of meat. During the early development, a pool of mesenchymal progenitor cells proliferate and then diverge into either myogenic or adipogenic/fibrogenic lineages. Myogenic progenitor cells further develop into muscle fibers and satellite cells, while adipogenic/fibrogenic lineage cells develop into adipocytes, fibroblasts and resident fibro-adipogenic progenitor cells. Enhancing the proliferation and myogenic commitment of progenitor cells during fetal development enhances muscle growth and lean production in offspring. On the other hand, promoting the adipogenic differentiation of adipogenic/fibrogenic progenitor cells inside the muscle increases intramuscular adipocytes and reduces connective tissue, which improves meat marbling and tenderness. Available studies in mammalian livestock, including cattle, sheep and pigs, clearly show the link between maternal nutrition and the quantity and quality of meat production. Similarly, chicken muscle fibers develop before hatching and, thus, egg and yolk sizes and hatching temperature affect long-term growth performance and meat production of chicken. On the contrary, because fishes are able to generate new muscle fibers lifelong, the impact of early nutrition on fish growth performance is expected to be minor, which requires further studies.
Collapse
Affiliation(s)
- Min Du
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States.
| | - Bo Wang
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Xing Fu
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Qiyuan Yang
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA 99164, United States
| |
Collapse
|
46
|
Kim JL, Kong HJ, Kim HS, Kim WJ, Kim DG, Nam BH, Kim YO, An CM. Fish Myogenic Regulatory Protein LUC7L: Characterization and Expression Analysis in Korean Rose Bitterling (Rhodeus uyekii). DEVELOPMENT & REPRODUCITON 2014. [PMID: 25949195 PMCID: PMC4415636 DOI: 10.12717/dr.2014.18.4.251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
| | - Hee Jeong Kong
- Corresponding Author : Hee Jeong Kong,
Biotechnology Research Division, National Fisheries Research and Development
Institute (NFRDI), 216 Haean-ro, Gijang-up, Gijang-gun, Busan 619-705, Korea.
Tel. :+82-51-720-2453, Fax :+82-51-720-2456, E-mail :
| | | | | | | | | | | | | |
Collapse
|