1
|
Roy BC, Coleman P, Markowsky M, Wang K, She Y, Richard C, Proctor SD, Bruce HL. Muscle Fiber, Connective Tissue and Meat Quality Characteristics of Pork from Low Birth Weight Pigs as Affected by Diet-Induced Increased Fat Absorption and Preferential Muscle Marbling. Food Sci Anim Resour 2024; 44:51-73. [PMID: 38229859 PMCID: PMC10789550 DOI: 10.5851/kosfa.2023.e56] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/07/2023] [Accepted: 09/15/2023] [Indexed: 01/18/2024] Open
Abstract
This study investigated how birth weight differences in piglets affected carcass and muscle fiber properties as well as meat quality at slaughter. Within litters, piglets were grouped according to their birth weight as either normal (NBW; 1.62-1.73 kg) or low (LBW; 1.18-1.29 kg). At 5 weeks of age, NBW piglets were randomly transitioned to control (C) or isocaloric high fat diets derived from non-dairy (HF), while LBW piglets were randomly transitioned to high fat diets derived from non-dairy (HF) or dairy sources (HFHD). Piglets were reared in individual pens under standardized housing and feeding conditions. Live weight was recorded weekly, and pigs were slaughtered at 12 weeks of age. Hot carcass weights, dressing percentages, lean meat yield, and primal cut proportions were determined. The m. longissimus thoracis was collected from the right side of the carcass for measurement of physical and chemical properties of meat and muscle fiber characteristics. Results indicated that LBW pigs compensated for their live weight compared to NBW pigs at 6 weeks of age. The mean muscle fiber diameter of LBW-HFHD group is significantly higher than NBW-C and NBW-HF group, and the type I muscle fiber diameter is significantly higher than NBW-C group. Dairy fat inclusion in LBW pig diet reduced carcass back fat thickness. This increased the calculated lean meat yield to be comparable to that of NBW pigs fed a commercial diet. Incorporating dairy-sourced high-fat into LBW pigs' diets appears to be an effective strategy for producing carcasses equivalent to NBW pigs.
Collapse
Affiliation(s)
- Bimol C. Roy
- Division of Animal Science, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
| | - Patience Coleman
- Division of Animal Science, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
| | - Meghan Markowsky
- Division of Animal Science, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
| | - Kun Wang
- Division of Animal Science, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
- Division of Human Nutrition, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
| | - Yongbo She
- Division of Animal Science, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
- Division of Human Nutrition, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
| | - Caroline Richard
- Division of Animal Science, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
- Division of Human Nutrition, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
| | - Spencer D. Proctor
- Division of Animal Science, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
- Division of Human Nutrition, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
| | - Heather L. Bruce
- Division of Animal Science, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
| |
Collapse
|
2
|
Ren P, Chen M, Li J, Lin Z, Yang C, Yu C, Zhang D, Liu Y. MYH1F promotes the proliferation and differentiation of chicken skeletal muscle satellite cells into myotubes. Anim Biotechnol 2023; 34:3074-3084. [PMID: 36244007 DOI: 10.1080/10495398.2022.2132953] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
In diploid organisms, interactions between alleles determine phenotypic variation. In previous experiments, only MYH1F was found to show both ASE (spatiotemporal allele-specific expression) and TRD (allelic transmission ratio distortion) characteristics in the pectoral muscle by comparing the genome-wide allele lists of hybrid populations (F1) of meat- and egg- type chickens. In addition, MYH1F is a member of the MYH gene family, which plays an important role in skeletal muscle and non-muscle cells of animals, but the specific expression and function of this gene in chickens are still unknown. Therefore, qRT-PCR was used to detect the expression of MYH1F in different tissues of chicken. Proliferation and differentiation of chicken skeletal muscle satellite cells (SMSCs) have been detected by transfection of MYH1F-specific small interfering RNA (siRNA). The results showed that the expression of MYH1F in chicken skeletal muscle was higher than that in other tissues. Combined with CCK-8 assay, EdU assay, immunofluorescence, and Western blot Assay, it was found that MYH1F knockdown could significantly suppress the proliferation of chicken SMSCs and depress the differentiation and fusion of the cells. These results suggest that MYH1F plays a critical role in myogenesis in poultry, which is of great significance for exploring the regulatory mechanisms of muscle development and improving animal productivity.
Collapse
Affiliation(s)
- Peng Ren
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, China
| | - Meiying Chen
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, China
| | - Jingjing Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhongzhen Lin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Chaowu Yang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Chunlin Yu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Donghao Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yiping Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Fujita R, Mizuno S, Sadahiro T, Hayashi T, Sugasawa T, Sugiyama F, Ono Y, Takahashi S, Ieda M. Generation of a MyoD knock-in reporter mouse line to study muscle stem cell dynamics and heterogeneity. iScience 2023; 26:106592. [PMID: 37250337 PMCID: PMC10214404 DOI: 10.1016/j.isci.2023.106592] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 02/19/2023] [Accepted: 03/31/2023] [Indexed: 05/31/2023] Open
Abstract
Myoblast determination protein 1 (MyoD) dynamics define the activation status of muscle stem cells (MuSCs), aiding in muscle tissue regeneration after injury. However, the lack of experimental platforms to monitor MyoD dynamics in vitro and in vivo has hampered the investigation of fate determination and heterogeneity of MuSCs. Herein, we report a MyoD knock-in (MyoD-KI) reporter mouse expressing tdTomato at the endogenous MyoD locus. Expression of tdTomato in MyoD-KI mice recapitulated the endogenous MyoD expression dynamics in vitro and during the early phase of regeneration in vivo. Additionally, we showed that tdTomato fluorescence intensity defines MuSC activation status without immunostaining. Based on these features, we developed a high-throughput screening system to assess the effects of drugs on the behavior of MuSCs in vitro. Thus, MyoD-KI mice are an invaluable resource for studying the dynamics of MuSCs, including their fate decisions and heterogeneity, and for drug screening in stem cell therapy.
Collapse
Affiliation(s)
- Ryo Fujita
- Division of Regenerative Medicine, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
- Department of Cardiology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Taketaro Sadahiro
- Department of Cardiology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Takuto Hayashi
- Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Takehito Sugasawa
- Laboratory of Clinical Examination and Sports Medicine, Department of Clinical Medicine, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Fumihiro Sugiyama
- Laboratory Animal Resource Center, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
- Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Masaki Ieda
- Department of Cardiology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
4
|
Bersin TV, Cordova KL, Saenger EK, Journey ML, Beckman BR, Lema SC. Nutritional status affects Igf1 regulation of skeletal muscle myogenesis, myostatin, and myofibrillar protein degradation pathways in gopher rockfish (Sebastes carnatus). Mol Cell Endocrinol 2023; 573:111951. [PMID: 37169322 DOI: 10.1016/j.mce.2023.111951] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/13/2023]
Abstract
Insulin-like growth factor-1 (Igf1) regulates skeletal muscle growth in fishes by increasing protein synthesis and promoting muscle hypertrophy. When fish experience periods of insufficient food intake, they undergo slower muscle growth or even muscle wasting, and those changes emerge in part from nutritional modulation of Igf1 signaling. Here, we examined how food deprivation (fasting) modulates Igf1 regulation of liver and skeletal muscle gene expression in gopher rockfish (Sebastes carnatus), a nearshore rockfish of importance for commercial and recreational fisheries in the northeastern Pacific Ocean, to understand how food limitation impacts Igf regulation of muscle growth pathways. Rockfish were either fed or fasted for 14 d, after which a subset of fish from each group was treated with recombinant Igf1 from sea bream (Sparus aurata). Fish that were fasted lost body mass and had lower body condition, reduced hepatosomatic index, and lower plasma Igf1 concentrations, as well as a decreased abundance of igf1 gene transcripts in the liver, increased hepatic mRNAs for Igf binding proteins igfbp1a, igfbp1b, and igfbp3a, and decreased mRNA abundances for igfbp2b and a putative Igf acid labile subunit (igfals) gene. In skeletal muscle, fasted fish showed a reduced abundance of intramuscular igf1 mRNAs but elevated gene transcripts encoding Igf1 receptors A (igf1ra) and B (igf1rb), which also showed downregulation by Igf1. Fasting increased skeletal muscle mRNAs for myogenin and myostatin1, as well as ubiquitin ligase F-box only protein 32 (fbxo32) and muscle RING-finger protein-1 (murf1) genes involved in muscle atrophy, while concurrently downregulating mRNAs for myoblast determination protein 2 (myod2), myostatin2, and myogenic factors 5 (myf5) and 6 (myf6 encoding Mrf4). Treatment with Igf1 downregulated muscle myostatin1 and fbxo32 under both feeding conditions, but showed feeding-dependent effects on murf1, myf5, and myf6/Mrf4 gene expression indicating that Igf1 effects on muscle growth and atrophy pathways is contingent on recent food consumption experience.
Collapse
Affiliation(s)
- Theresa V Bersin
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA, 93407, USA
| | - Kasey L Cordova
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA, 93407, USA
| | - E Kate Saenger
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA, 93407, USA
| | - Meredith L Journey
- Lynker Technology, 202 Church St SE #536, Leesburg, VA, 20175, USA; Under Contract to Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle, WA, 98112, USA
| | - Brian R Beckman
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle, WA, 98112, USA
| | - Sean C Lema
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA, 93407, USA.
| |
Collapse
|
5
|
Uchiyama H, Muramatsu D, Higashi H, Kida H, Iwai A. Effects of chondroitin sulfate oligosaccharides on osteoclast differentiation of RAW264 cells, and myotube differentiation of C2C12 cells. PLoS One 2023; 18:e0284343. [PMID: 37053208 PMCID: PMC10101473 DOI: 10.1371/journal.pone.0284343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
Chondroitin sulfate (CS) is a glycosaminoglycan, and CS derived from various animal species is used in drugs and food supplements to alleviate arthralgia. The CS is a high molecular weight compound, and hydrolysis of CS by intestinal microbiota is thought to be required for absorption in mammalians. Chondroitin sulfate oligosaccharides (Oligo-CS) are produced by hydrolysis with subcritical water from CS isolated from a species of skate, Raja pulchra for the improvement of bioavailability. The present study conducted in vitro experiments using murine cell lines, to compare the biological activities of Oligo-CS and high molecular weight CS composed with the similar disaccharide isomer units of D-glucuronic acid and N-acetyl-D-glucosamine (CS-C). The results show that Oligo-CS inhibits osteoclast differentiation of RAW264 cells significantly at lower concentrations than in CS. The cell viability of a myoblast cell line, C2C12 cells, was increased when the cells were grown in a differentiated medium for myotubes with Oligo-CS, where there were no effects on the cell viability in CS. These results suggest that in vitro Oligo-CS exhibits stronger bioactivity than high-molecular weight CS.
Collapse
Affiliation(s)
- Hirofumi Uchiyama
- Aureo Science Co., Ltd., Kita-ku, Sapporo, Hokkaido, Japan
- Division of Bioscience in Sapporo, Aureo Co., Ltd., Kita-ku, Sapporo, Hokkaido, Japan
| | - Daisuke Muramatsu
- Aureo Science Co., Ltd., Kita-ku, Sapporo, Hokkaido, Japan
- Division of Bioscience in Sapporo, Aureo Co., Ltd., Kita-ku, Sapporo, Hokkaido, Japan
| | - Hideaki Higashi
- Division of Infection and Immunity, International Institute for Zoonosis Control, Hokkaido University, Kita-ku, Sapporo, Hokkaido, Japan
| | - Hiroshi Kida
- International Institute for Zoonosis Control, Hokkaido University, Kita-ku, Sapporo, Hokkaido, Japan
| | - Atsushi Iwai
- Aureo Science Co., Ltd., Kita-ku, Sapporo, Hokkaido, Japan
- Division of Bioscience in Sapporo, Aureo Co., Ltd., Kita-ku, Sapporo, Hokkaido, Japan
| |
Collapse
|
6
|
Colasuonno F, Price R, Moreno S. Upper and Lower Motor Neurons and the Skeletal Muscle: Implication for Amyotrophic Lateral Sclerosis (ALS). ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2023; 236:111-129. [PMID: 37955773 DOI: 10.1007/978-3-031-38215-4_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
The relationships between motor neurons and the skeletal muscle during development and in pathologic contexts are addressed in this Chapter.We discuss the developmental interplay of muscle and nervous tissue, through neurotrophins and the activation of differentiation and survival pathways. After a brief overview on muscular regulatory factors, we focus on the contribution of muscle to early and late neurodevelopment. Such a role seems especially intriguing in relation to the epigenetic shaping of developing motor neuron fate choices. In this context, emphasis is attributed to factors regulating energy metabolism, which may concomitantly act in muscle and neural cells, being involved in common pathways.We then review the main features of motor neuron diseases, addressing the cellular processes underlying clinical symptoms. The involvement of different muscle-associated neurotrophic factors for survival of lateral motor column neurons, innervating MyoD-dependent limb muscles, and of medial motor column neurons, innervating Myf5-dependent back musculature is discussed. Among the pathogenic mechanisms, we focus on oxidative stress, that represents a common and early trait in several neurodegenerative disorders. The role of organelles primarily involved in reactive oxygen species scavenging and, more generally, in energy metabolism-namely mitochondria and peroxisomes-is discussed in the frame of motor neuron degeneration.We finally address muscular involvement in amyotrophic lateral sclerosis (ALS), a multifactorial degenerative disorder, hallmarked by severe weight loss, caused by imbalanced lipid metabolism. Even though multiple mechanisms have been recognized to play a role in the disease, current literature generally assumes that the primum movens is neuronal degeneration and that muscle atrophy is only a consequence of such pathogenic event. However, several lines of evidence point to the muscle as primarily involved in the disease, mainly through its role in energy homeostasis. Data from different ALS mouse models strongly argue for an early mitochondrial dysfunction in muscle tissue, possibly leading to motor neuron disturbances. Detailed understanding of skeletal muscle contribution to ALS pathogenesis will likely lead to the identification of novel therapeutic strategies.
Collapse
Affiliation(s)
- Fiorella Colasuonno
- Department of Experimental Medicine , University of Rome "Tor Vergata", Rome, Italy
- Department of Science, LIME, University Roma Tre, Rome, Italy
| | - Rachel Price
- Department of Science, LIME, University Roma Tre, Rome, Italy
- Laboratory of Neurodevelopmental Biology, Neurogenetics and Molecular Neurobiology, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Sandra Moreno
- Department of Science, LIME, University Roma Tre, Rome, Italy.
- Laboratory of Neurodevelopmental Biology, Neurogenetics and Molecular Neurobiology, IRCCS Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
7
|
Murphy P, Rolfe RA. Building a Co-ordinated Musculoskeletal System: The Plasticity of the Developing Skeleton in Response to Muscle Contractions. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2023; 236:81-110. [PMID: 37955772 DOI: 10.1007/978-3-031-38215-4_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
The skeletal musculature and the cartilage, bone and other connective tissues of the skeleton are intimately co-ordinated. The shape, size and structure of each bone in the body is sculpted through dynamic physical stimuli generated by muscle contraction, from early development, with onset of the first embryo movements, and through repair and remodelling in later life. The importance of muscle movement during development is shown by congenital abnormalities where infants that experience reduced movement in the uterus present a sequence of skeletal issues including temporary brittle bones and joint dysplasia. A variety of animal models, utilising different immobilisation scenarios, have demonstrated the precise timing and events that are dependent on mechanical stimulation from movement. This chapter lays out the evidence for skeletal system dependence on muscle movement, gleaned largely from mouse and chick immobilised embryos, showing the many aspects of skeletal development affected. Effects are seen in joint development, ossification, the size and shape of skeletal rudiments and tendons, including compromised mechanical function. The enormous plasticity of the skeletal system in response to muscle contraction is a key factor in building a responsive, functional system. Insights from this work have implications for our understanding of morphological evolution, particularly the challenging concept of emergence of new structures. It is also providing insight for the potential of physical therapy for infants suffering the effects of reduced uterine movement and is enhancing our understanding of the cellular and molecular mechanisms involved in skeletal tissue differentiation, with potential for informing regenerative therapies.
Collapse
Affiliation(s)
- Paula Murphy
- School of Natural Sciences, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland.
| | - Rebecca A Rolfe
- School of Natural Sciences, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
| |
Collapse
|
8
|
Fung CW, Zhou S, Zhu H, Wei X, Wu Z, Wu AR. Cell fate determining molecular switches and signaling pathways in Pax7-expressing somitic mesoderm. Cell Discov 2022; 8:61. [PMID: 35764624 PMCID: PMC9240041 DOI: 10.1038/s41421-022-00407-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/28/2022] [Indexed: 11/09/2022] Open
Abstract
During development, different cell types originate from a common progenitor at well-defined time points. Previous lineage-tracing of Pax7+ progenitors from the somitic mesoderm has established its developmental trajectory towards the dermis, brown adipocytes, and skeletal muscle in the dorsal trunk; yet the molecular switches and mechanisms guiding the differentiation into different lineages remain unknown. We performed lineage-tracing of Pax7-expressing cells in mouse embryos at E9.5 and profiled the transcriptomes of Pax7-progenies on E12.5, E14.5, and E16.5 at single-cell level. Analysis of single-cell transcriptomic data at multiple time points showed temporal-specific differentiation events toward muscle, dermis, and brown adipocyte, identified marker genes for putative progenitors and revealed transcription factors that could drive lineage-specific differentiation. We then utilized a combination of surface markers identified in the single-cell data, Pdgfra, Thy1, and Cd36, to enrich brown adipocytes, dermal fibroblasts, and progenitors specific for these two cell types at E14.5 and E16.5. These enriched cell populations were then used for further culture and functional assays in vitro, in which Wnt5a and Rgcc are shown to be important factors that could alter lineage decisions during embryogenesis. Notably, we found a bipotent progenitor population at E14.5, having lineage potentials towards both dermal fibroblasts and brown adipocytes. They were termed eFAPs (embryonic fibro/adipogenic progenitors) as they functionally resemble adult fibro/adipogenic progenitors. Overall, this study provides further understanding of the Pax7 lineage during embryonic development using a combination of lineage tracing with temporally sampled single-cell transcriptomics.
Collapse
|
9
|
Kowalczyk K, Smyk M, Bartnik-Głaska M, Plaskota I, Wiśniowiecka-Kowalnik B, Bernaciak J, Chojnacka M, Paczkowska M, Niemiec M, Dutkiewicz D, Kozar A, Magdziak R, Krawczyk W, Pietras G, Michalak E, Klepacka T, Obersztyn E, Bal J, Nowakowska BA. Application of array comparative genomic hybridization (aCGH) for identification of chromosomal aberrations in the recurrent pregnancy loss. J Assist Reprod Genet 2022; 39:357-367. [PMID: 35079943 PMCID: PMC8956756 DOI: 10.1007/s10815-022-02400-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/13/2022] [Indexed: 12/03/2022] Open
Abstract
Spontaneous abortion occurs in 8–20% of recognized pregnancies and usually takes place in the first trimester (7–11 weeks). There are many causes of pregnancy loss, but the most important (about 75%) is the presence of chromosomal aberrations. We present the results of oligonucleotide array application in a cohort of 62 miscarriage cases. The inclusion criteria for the study were the loss after 8th week of pregnancy and the appearance of recurrent miscarriages. DNA was extracted from trophoblast or fetal skin fibroblasts. In the 62 tested materials from recurrent miscarriages, the detection rate was 56.5% (35/62). The most commonly found were aneuploidies (65%) (chromosomal trisomy 14, 16, 18, 21, and 22), Turner syndrome, and triploidy (17.1%). Other chromosomal abnormalities included pathogenic and likely pathogenic structural aberrations: 1) pathogenic: deletion 7p22.3p12.3 and duplication 9p24.3p13.2 inherited from the normal father, deletion 3q13.31q22.2 and deletion 3q22.3q23 of unknown inheritance and duplication of 17p12 inherited from father with foot malformation; 2) likely pathogenic variants: deletion 17p13.1 inherited from normal mother, deletion 5q14.3 of unknown inheritance and de novo deletion 1q21.1q21.2. Among these aberrations, six CNVs (copy number variants) were responsible for the miscarriage: deletion 7p22.3p12.3 and duplication 9p24.3p13.2, deletion 3q13.31q22.2 and deletion 3q22.3q23, and deletion 17p13.1 and deletion 1q21.1q21.2. Other two findings were classified as incidental findings (deletion 5q14.3 and 17p12 duplication). Our research shows that 17% of the aberrations (6/35 abnormal results) that cannot be identified by the routine kariotype analysis are structural aberrations containing genes important for fetal development, the mutations of which may cause spontaneous abortion.
Collapse
Affiliation(s)
- Katarzyna Kowalczyk
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland.
| | - Marta Smyk
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | | | - Izabela Plaskota
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | | | - Joanna Bernaciak
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | - Marta Chojnacka
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | | | - Magdalena Niemiec
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | - Daria Dutkiewicz
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | - Agata Kozar
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | - Róża Magdziak
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | - Wojciech Krawczyk
- Department of Obstetrics and Perinatology, Medical University of Lublin, Lublin, Poland
| | - Grzegorz Pietras
- Department of Obstetrics and Perinatology, Medical University of Lublin, Lublin, Poland
| | - Elżbieta Michalak
- Deparment of Pathomorphology, Institute of Mother and Child, Warsaw, Poland
| | - Teresa Klepacka
- Deparment of Pathomorphology, Institute of Mother and Child, Warsaw, Poland
| | - Ewa Obersztyn
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | - Jerzy Bal
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | | |
Collapse
|
10
|
Rodriguez-Outeiriño L, Hernandez-Torres F, Ramírez-de Acuña F, Matías-Valiente L, Sanchez-Fernandez C, Franco D, Aranega AE. Muscle Satellite Cell Heterogeneity: Does Embryonic Origin Matter? Front Cell Dev Biol 2021; 9:750534. [PMID: 34722534 PMCID: PMC8554119 DOI: 10.3389/fcell.2021.750534] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/27/2021] [Indexed: 12/25/2022] Open
Abstract
Muscle regeneration is an important homeostatic process of adult skeletal muscle that recapitulates many aspects of embryonic myogenesis. Satellite cells (SCs) are the main muscle stem cells responsible for skeletal muscle regeneration. SCs reside between the myofiber basal lamina and the sarcolemma of the muscle fiber in a quiescent state. However, in response to physiological stimuli or muscle trauma, activated SCs transiently re-enter the cell cycle to proliferate and subsequently exit the cell cycle to differentiate or self-renew. Recent evidence has stated that SCs display functional heterogeneity linked to regenerative capability with an undifferentiated subgroup that is more prone to self-renewal, as well as committed progenitor cells ready for myogenic differentiation. Several lineage tracing studies suggest that such SC heterogeneity could be associated with different embryonic origins. Although it has been established that SCs are derived from the central dermomyotome, how a small subpopulation of the SCs progeny maintain their stem cell identity while most progress through the myogenic program to construct myofibers is not well understood. In this review, we synthesize the works supporting the different developmental origins of SCs as the genesis of their functional heterogeneity.
Collapse
Affiliation(s)
- Lara Rodriguez-Outeiriño
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
| | - Francisco Hernandez-Torres
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, Granada, Spain
| | - F. Ramírez-de Acuña
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
| | - Lidia Matías-Valiente
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
| | - Cristina Sanchez-Fernandez
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
| | - Diego Franco
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
| | - Amelia Eva Aranega
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
| |
Collapse
|
11
|
Wang Z, Zhao Q, Li X, Yin Z, Chen S, Wu S, Yang N, Hou Z. MYOD1 inhibits avian adipocyte differentiation via miRNA-206/KLF4 axis. J Anim Sci Biotechnol 2021; 12:55. [PMID: 33952351 PMCID: PMC8101123 DOI: 10.1186/s40104-021-00579-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/01/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND A considerable number of muscle development-related genes were differentially expressed in the early stage of avian adipocyte differentiation. However, the functions of them in adipocyte differentiation remain largely known. In this study, the myoblast determination protein 1 (MYOD1) was selected as a representative of muscle development. We investigated its expression, function, and regulation in avian adipocyte differentiation. RESULTS The expression of MYOD1 decreased significantly in the early stage of avian adipocyte differentiation. CRISPR/Cas9-mediated deletion of MYOD1 induced adipocyte differentiation, whereas over-expression of MYOD1 inhibited adipogenesis. The mRNA-seq data showed that MYOD1 could perturb the lipid biosynthetic process during differentiation. Our results showed that MYOD1 directly up-regulates the miR-206 expression by binding the upstream 1200 bp region of miR-206. Then, over-expression of miR-206 can inhibit the adipogenesis. Furthermore, MYOD1 affected the expression of endogenous miR-206 and its target gene Kruppel-like factor 4 (KLF4), which is an important activator of adipogenesis. Accordingly, the inhibition of miR-206 or over-expression of KLF4 could counteract the inhibitory effect of MYOD1 on adipocyte differentiation. CONCLUSIONS Our results establish that MYOD1 inhibits adipocyte differentiation by up-regulating miR-206 to suppress the KLF4 expression. These findings identify a novel function of MYOD1 in adipocyte differentiation, suggesting a potential role in body-fat distribution regulation.
Collapse
Affiliation(s)
- Zheng Wang
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, MARA, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road No. 2, Beijing, 100193 China
| | - Qiangsen Zhao
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, MARA, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road No. 2, Beijing, 100193 China
| | - Xiaoqin Li
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, MARA, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road No. 2, Beijing, 100193 China
| | - Zhongtao Yin
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, MARA, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road No. 2, Beijing, 100193 China
| | - Sirui Chen
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, MARA, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road No. 2, Beijing, 100193 China
| | - Sen Wu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Beijing, 100193 China
| | - Ning Yang
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, MARA, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road No. 2, Beijing, 100193 China
| | - Zhuocheng Hou
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, MARA, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road No. 2, Beijing, 100193 China
| |
Collapse
|
12
|
Holzman MA, Ryckman A, Finkelstein TM, Landry-Truchon K, Schindler KA, Bergmann JM, Jeannotte L, Mansfield JH. HOXA5 Participates in Brown Adipose Tissue and Epaxial Skeletal Muscle Patterning and in Brown Adipocyte Differentiation. Front Cell Dev Biol 2021; 9:632303. [PMID: 33732701 PMCID: PMC7959767 DOI: 10.3389/fcell.2021.632303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/02/2021] [Indexed: 11/13/2022] Open
Abstract
Brown adipose tissue (BAT) plays critical thermogenic, metabolic and endocrine roles in mammals, and aberrant BAT function is associated with metabolic disorders including obesity and diabetes. The major BAT depots are clustered at the neck and forelimb levels, and arise largely within the dermomyotome of somites, from a common progenitor with skeletal muscle. However, many aspects of BAT embryonic development are not well understood. Hoxa5 patterns other tissues at the cervical and brachial levels, including skeletal, neural and respiratory structures. Here, we show that Hoxa5 also positively regulates BAT development, while negatively regulating formation of epaxial skeletal muscle. HOXA5 protein is expressed in embryonic preadipocytes and adipocytes as early as embryonic day 12.5. Hoxa5 null mutant embryos and rare, surviving adults show subtly reduced iBAT and sBAT formation, as well as aberrant marker expression, lower adipocyte density and altered lipid droplet morphology. Conversely, the epaxial muscles that arise from a common dermomyotome progenitor are expanded in Hoxa5 mutants. Conditional deletion of Hoxa5 with Myf5/Cre can reproduce both BAT and epaxial muscle phenotypes, indicating that HOXA5 is necessary within Myf5-positive cells for proper BAT and epaxial muscle development. However, recombinase-based lineage tracing shows that Hoxa5 does not act cell-autonomously to repress skeletal muscle fate. Interestingly, Hoxa5-dependent regulation of adipose-associated transcripts is conserved in lung and diaphragm, suggesting a shared molecular role for Hoxa5 in multiple tissues. Together, these findings establish a role for Hoxa5 in embryonic BAT development.
Collapse
Affiliation(s)
- Miriam A. Holzman
- Department of Biology, Barnard College, Columbia University, New York, NY, United States
| | - Abigail Ryckman
- Department of Biology, Barnard College, Columbia University, New York, NY, United States
| | - Tova M. Finkelstein
- Department of Biology, Barnard College, Columbia University, New York, NY, United States
| | - Kim Landry-Truchon
- Centre de Recherche sur le Cancer de l’Université Laval, CRCHU de Québec-Université Laval (Oncology), Québec City, QC, Canada
| | - Kyra A. Schindler
- Department of Biology, Barnard College, Columbia University, New York, NY, United States
| | - Jenna M. Bergmann
- Department of Biology, Barnard College, Columbia University, New York, NY, United States
| | - Lucie Jeannotte
- Centre de Recherche sur le Cancer de l’Université Laval, CRCHU de Québec-Université Laval (Oncology), Québec City, QC, Canada
| | - Jennifer H. Mansfield
- Department of Biology, Barnard College, Columbia University, New York, NY, United States
| |
Collapse
|
13
|
Tompkins YH, Su S, Velleman SG, Kim WK. Effects of 20(S)-hydroxycholesterol on satellite cell proliferation and differentiation of broilers. Poult Sci 2021; 100:474-481. [PMID: 33518099 PMCID: PMC7858162 DOI: 10.1016/j.psj.2020.10.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/14/2020] [Accepted: 10/19/2020] [Indexed: 01/30/2023] Open
Abstract
In the modern poultry industry, with increasing product demand, muscle growth rate and meat yield in chickens have tremendously changed. Understanding the regulation of muscle development is important to maintain efficient growth and development in meat-type chickens. 20(S)-hydroxycholesterol (20S) is known as one of the naturally occurring osteogenic cholesterol derivatives due to its ability to induce osteogenic differentiation; however, no studies have evaluated myogenic response to 20S in chicken muscle cells. To determine the use of 20S in vitro for the proliferation and differentiation of chicken satellite cells, satellite cells were isolated from pectoralis major muscle of 4-week-old Ross 708 male chickens and subjected to 0.25, 0.5, and 1.0 μmol of 20S during their proliferation and differentiation stages. Cell proliferation and differentiation were measured every 24 h for 72 h by determining DNA concentration, the activity of creatine kinase, and the expressions of myogenic regulatory transcription factors. Together these results suggested that a lower concentration of 20S did not affect myogenesis but a high concentration of 1.0 μmol 20S can negatively affect proliferation and differentiation in chicken satellite cells.
Collapse
Affiliation(s)
- Yuguo H Tompkins
- Department of Poultry Science, University of Georgia, Athens, USA
| | - Shengchen Su
- Department of Poultry Science, University of Georgia, Athens, USA
| | - Sandra G Velleman
- The Ohio State University, Ohio Agricultural Research and Development Center, Wooster, USA
| | - Woo Kyun Kim
- Department of Poultry Science, University of Georgia, Athens, USA.
| |
Collapse
|
14
|
Riederer M, Wallner M, Schweighofer N, Fuchs-Neuhold B, Rath A, Berghold A, Eberhard K, Groselj-Strele A, Staubmann W, Peterseil M, Waldner I, Mayr JA, Rothe M, Holasek S, Maunz S, Pail E, van der Kleyn M. Distinct maternal amino acids and oxylipins predict infant fat mass and fat-free mass indices. Arch Physiol Biochem 2020; 129:563-574. [PMID: 33283558 DOI: 10.1080/13813455.2020.1846204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Interested in maternal determinants of infant fat mass index (FMI) and fat-free mass index (FFMI), considered as predictors for later development of obesity, we analysed amino acids (AA) and oxylipins in maternal serum and breast milk (BM). FMI and FFMI were calculated in 47 term infants aged 4 months (T4). Serum AA were analysed in pregnancy (T1, T2) and 6-8 weeks postpartum (T3). At T3, AA and oxylipins were analysed in BM. Biomarker-index-associations were identified by regression analysis. Infant FMI (4.1 ± 1.31 kg/m2; MW ± SD) was predicted by T2 proline (R2 adj.: 7.6%, p = .036) and T3 BM 11-hydroxy-eicosatetraenoic-acid (11-HETE) and 13-hydroxy-docosahexaenoic-acid (13-HDHA; together:35.5% R2 adj., p < .001). Maternal peripartum antibiotics (AB) emerged as confounders (+AB: 23.5% higher FMI; p = .025). Infant FFMI (12.1 ± 1.19 kg/m2; MW ± SD) was predicted by histidine (R2 adj.: 14.5%, p < .001) and 17-HDHA (BM, R2 adj.:19.3%, p < .001), determined at T3. Confirmed in a larger cohort, the parameters could elucidate connections between maternal metabolic status, nutrition, and infant body development.
Collapse
Affiliation(s)
- Monika Riederer
- Institute of Biomedical Science, University of Applied Sciences JOANNEUM, Graz, Austria
| | - Marlies Wallner
- Institute of Dietetics and Nutrition, Health Perception Lab, University of Applied Sciences JOANNEUM, Graz, Austria
| | | | - Bianca Fuchs-Neuhold
- Institute of Dietetics and Nutrition, Health Perception Lab, University of Applied Sciences JOANNEUM, Graz, Austria
| | - Anna Rath
- Institute of Midwifery, University of Applied Sciences JOANNEUM, Graz, Austria
| | - Andrea Berghold
- Institute for Medical Informatics, Statistics and Documentation, Medical University Graz, Graz, Austria
| | - Katharina Eberhard
- Core Facility Computational Bioanalytics, Center for Medical Research (ZMF), Medical University of Graz, Graz, Austria
| | - Andrea Groselj-Strele
- Core Facility Computational Bioanalytics, Center for Medical Research (ZMF), Medical University of Graz, Graz, Austria
| | - Wolfgang Staubmann
- Institute of Dietetics and Nutrition, Health Perception Lab, University of Applied Sciences JOANNEUM, Graz, Austria
| | - Marie Peterseil
- Institute of Dietetics and Nutrition, Health Perception Lab, University of Applied Sciences JOANNEUM, Graz, Austria
| | - Irmgard Waldner
- Institute of Midwifery, University of Applied Sciences JOANNEUM, Graz, Austria
| | - Johannes A Mayr
- University Clinic for Pediatrics and Adolescent Medicine Salzburg, Salzburg, Austria
| | | | - Sandra Holasek
- Department of Pathophysiology, Medical University Graz, Graz, Austria
| | - Susanne Maunz
- Institute of Dietetics and Nutrition, Health Perception Lab, University of Applied Sciences JOANNEUM, Graz, Austria
| | - Elisabeth Pail
- Institute of Dietetics and Nutrition, Health Perception Lab, University of Applied Sciences JOANNEUM, Graz, Austria
| | | |
Collapse
|
15
|
Peng Y, Yue F, Chen J, Xia W, Huang K, Yang G, Kuang S. Phosphatase orphan 1 inhibits myoblast proliferation and promotes myogenic differentiation. FASEB J 2020; 35:e21154. [PMID: 33140469 DOI: 10.1096/fj.202001672r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/29/2020] [Accepted: 10/16/2020] [Indexed: 01/08/2023]
Abstract
Myogenesis includes sequential stages of progenitor cell proliferation, myogenic commitment and differentiation, myocyte fusion, and myotube maturation. Different stages of myogenesis are orchestrated and regulated by myogenic regulatory factors and various downstream cellular signaling. Here we identify phosphatase orphan 1 (Phospho1) as a new player in myogenesis. During activation, proliferation, and differentiation of quiescent satellite cells, the expression of Phospho1 gradually increases. Overexpression of Phospho1 inhibits myoblast proliferation but promotes their differentiation and fusion. Conversely, knockdown of Phospho1 accelerates myoblast proliferation but impairs myotube formation. Moreover, knockdown of Phospho1 decreases the OXPHO protein levels and mitochondria density, whereas overexpression of Phospho1 upregulates OXPHO protein levels and promotes mitochondrial oxygen consumption. Finally, we show that Phospho1 expression is controlled by myogenin, which binds to the promoter of Phospho1 to regulate its transcription. These results indicate a key role of Phospho1 in regulating myogenic differentiation and mitochondrial function.
Collapse
Affiliation(s)
- Ying Peng
- College of Animal Science and Technology, Northwest A&F University, Yangling, China.,Department of Animal Sciences, Purdue University, West Lafayette, IN, USA
| | - Feng Yue
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA
| | - Jingjuan Chen
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA
| | - Wei Xia
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA.,College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| | - Kuilong Huang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China.,Department of Animal Sciences, Purdue University, West Lafayette, IN, USA
| | - Gongshe Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
16
|
Mani R, Balasubramanian S, Raghunath A, Perumal E. Chronic exposure to copper oxide nanoparticles causes muscle toxicity in adult zebrafish. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:27358-27369. [PMID: 31388954 DOI: 10.1007/s11356-019-06095-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 07/26/2019] [Indexed: 06/10/2023]
Abstract
Repeated deposition of copper oxide nanoparticles (CuO-NPs) into aquatic systems makes them a global threat since the NPs accumulate in various organs of the fish particularly skeletal muscle. In the present study, adult zebrafish were exposed to different concentrations of CuO-NPs (1 and 3 mg/L) for a period of 30 days. The status of functional markers (acetylcholinesterase, creatine kinase-MB, and lactate dehydrogenase) and oxidative stress markers (oxidants and antioxidants) were analyzed. The histological changes in muscle were studied followed by the immunohistochemistry expression for catalase. Further, the expression of myoD, myogenin, pax7, β-actin, and desmin was examined by semi-quantitative reverse transcriptase polymerase chain reaction. The results indicated that chronic exposure to CuO-NPs causes muscular damage as evidenced by elevated levels of functional markers. There was a significant increase in the oxidants with reduction in the antioxidant levels, implying that the antioxidant enzymes were unable to scavenge the free radicals induced by the CuO-NPs. The histopathological analysis showed degeneration and atrophy in the treated groups confirming muscle damage. The immunohistochemical catalase expression in the muscle was reduced in the treated groups further supporting the evidence that the antioxidant has suffered a decline. The altered gene expression indicates skeletal muscle damage due to the CuO-NPs exposure. Overall, the data suggest that chronic exposure to CuO-NPs caused muscular toxicity which may lead to muscle degeneration in adult zebrafish.
Collapse
Affiliation(s)
- Ramya Mani
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, 641 046, India
| | | | - Azhwar Raghunath
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, 641 046, India
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, 641 046, India.
| |
Collapse
|
17
|
Song J, Wang C, Long D, Li Z, You L, Brand-Saberi B, Wang G, Yang X. Dysbacteriosis-induced LPS elevation disturbs the development of muscle progenitor cells by interfering with retinoic acid signaling. FASEB J 2020; 34:6837-6853. [PMID: 32223025 DOI: 10.1096/fj.201902965r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/21/2020] [Accepted: 03/16/2020] [Indexed: 01/25/2023]
Abstract
Whether myogenesis is affected by the maternal gut dysbacteriosis still remains ambiguous. In this study, first we show the elevated level of lipopolysaccharides (LPS) in a gut microbiota dysbiosis mouse model. Second, we demonstrate that the diameter of muscle fibers, limb development, and somitogenesis were inhibited in both the gut microbiota dysbiosis and LPS exposed mice and chicken embryos. These might be due to LPS disturbed the cell survival and key genes which regulate the somitogenesis and myogenesis. RNA sequencing and subsequent validation experiments verified that retinoic acid (RA) signaling perturbation was mainly responsible for the aberrant somite formation and differentiation. Subsequently, we found that LPS-induced reactive oxygen species (ROS generation and antioxidant genes such as Nrf2, AKR1B10) contributed to the above -mentioned interference with RA signaling. These findings highlight that the gut microbiota homeostasis is also involved in regulating the development of muscle progenitor cells during pregnancy.
Collapse
Affiliation(s)
- Jinhuan Song
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Jinan University, Guangzhou, China
| | - Chaojie Wang
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Jinan University, Guangzhou, China
| | - Denglu Long
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Jinan University, Guangzhou, China
| | - Ziguang Li
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Jinan University, Guangzhou, China
| | - Lingsen You
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Jinan University, Guangzhou, China
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Guang Wang
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Jinan University, Guangzhou, China
| | - Xuesong Yang
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Jinan University, Guangzhou, China.,Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| |
Collapse
|
18
|
Che D, Adams S, Zhao B, Qin G, Jiang H. Effects of Dietary L-arginine Supplementation from Conception to Post- Weaning in Piglets. Curr Protein Pept Sci 2019; 20:736-749. [PMID: 30678624 DOI: 10.2174/1389203720666190125104959] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 12/30/2018] [Accepted: 01/10/2019] [Indexed: 12/13/2022]
Abstract
Weaned piglets experience sudden changes in their dietary patterns such as withdrawal from the easily digestible watery milk to a coarse cereal diet with both systemic and intestinal disruptions coupling with the expression of pro-inflammatory proteins which affects the immune system and the concentrations of haptoglobin including both positive and negative acute-phase proteins in the plasma. L-arginine is an important protein amino acid for piglets, but its inadequate synthesis is a nutritional problem for both sows and piglets. Recent studies indicated that dietary supplementation of L-arginine increased feed intake, uterine growth, placental growth and nutrient transport, maternal growth and health, embryonic survival, piglets birth weight, piglet's growth, and productivity, and decreased stillbirths. L-arginine is essential in several important pathways involved in the growth and development of piglets such as nitric oxide synthesis, energy metabolism, polyamine synthesis, cellular protein production and muscle accretion, and the synthesis of other functional amino acids. However, the underlying molecular mechanism in these key pathways remains largely unresolved. This review was conducted on the general hypothesis that L-arginine increased the growth and survival of post-weaning piglets. We discussed the effects of dietary L-arginine supplementation during gestation, parturition, lactation, weaning, and post-weaning in pigs as each of these stages influences the health and survival of sows and their progenies. Therefore, the aim of this review was to discuss through a logical approach the effects of L-arginine supplementation on piglet's growth and survival from conception to postweaning.
Collapse
Affiliation(s)
- Dongsheng Che
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China.,Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun 130118, China.,Key Lab of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun, 130118 China
| | - Seidu Adams
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Bao Zhao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China.,Key Lab of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun, 130118 China
| | - Guixin Qin
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China.,Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun 130118, China.,Key Lab of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun, 130118 China
| | - Hailong Jiang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China.,Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun 130118, China.,Key Lab of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun, 130118 China
| |
Collapse
|
19
|
Rosero Salazar DH, Carvajal Monroy PL, Wagener FADTG, Von den Hoff JW. Orofacial Muscles: Embryonic Development and Regeneration after Injury. J Dent Res 2019; 99:125-132. [PMID: 31675262 PMCID: PMC6977159 DOI: 10.1177/0022034519883673] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Orofacial congenital defects such as cleft lip and/or palate are associated with impaired muscle regeneration and fibrosis after surgery. Also, other orofacial reconstructions or trauma may end up in defective muscle regeneration and fibrosis. The aim of this review is to discuss current knowledge on the development and regeneration of orofacial muscles in comparison to trunk and limb muscles. The orofacial muscles include the tongue muscles and the branchiomeric muscles in the lower face. Their main functions are chewing, swallowing, and speech. All orofacial muscles originate from the mesoderm of the pharyngeal arches under the control of cranial neural crest cells. Research in vertebrate models indicates that the molecular regulation of orofacial muscle development is different from that of trunk and limb muscles. In addition, the regenerative ability of orofacial muscles is lower, and they develop more fibrosis than other skeletal muscles. Therefore, specific approaches need to be developed to stimulate orofacial muscle regeneration. Regeneration may be stimulated by growth factors such fibroblast growth factors and hepatocyte growth factor, while fibrosis may be reduced by targeting the transforming growth factor β1 (TGFβ1)/myofibroblast axis. New approaches that combine these 2 aspects will improve the surgical treatment of orofacial muscle defects.
Collapse
Affiliation(s)
- D H Rosero Salazar
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - P L Carvajal Monroy
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Centre, Nijmegen, the Netherlands.,Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - F A D T G Wagener
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - J W Von den Hoff
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Centre, Nijmegen, the Netherlands
| |
Collapse
|
20
|
Baatar D, Hwang SG. Effect of testosterone on the differentiation control of stromal vascular cells isolated from longissimus muscle of Hanwoo beef cattle. Meat Sci 2019; 159:107916. [PMID: 31476680 DOI: 10.1016/j.meatsci.2019.107916] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 08/16/2019] [Accepted: 08/16/2019] [Indexed: 02/06/2023]
Abstract
Testosterone, as an influential factor in marbling score, requires strict management for uniform development of adipocytes in-between muscle bundles. Present study investigated effect of castration timing and testosterone levels on adipocyte development using SVCs. Isolated SVCs exhibited classical MSC markers, CD31-, CD34-, CD45-, CD90+, and CD105+. ELISA analysis indicated that serum testosterone concentration was highest in non-castrated calves while no significant difference was observed between female, early and late castrated calves. CCK-8 assay showed that concentration of testosterone had no effect on cell proliferation. However, the real-time PCR demonstrated that 20 ng/ml of testosterone suppressed expression of preadipocyte markers, pref-1 and zfp423, but encouraged expression of myoblast markers, myf5 and myoD, via the AR. Consequently, expression of adipogenic markers C/EBPα and PPARγ, as well as accumulation of triglyceride, were decreased in 20 ng/ml testosterone treatment under adipogenic conditions. These findings suggest that by castrating calves before level of testosterone increases, may improve marbling development in the Hanwoo beef industry.
Collapse
Affiliation(s)
- Delgerzul Baatar
- Department of Animal Life and Environmental Science, Hankyong National University, Anseong City 456-749, Republic of Korea
| | - Seong Gu Hwang
- Department of Animal Life and Environmental Science, Hankyong National University, Anseong City 456-749, Republic of Korea.
| |
Collapse
|
21
|
Han S, Cui C, Wang Y, He H, Shen X, Chen Y, Liu Z, Zhu Q, Li D, Yin H. FHL3 negatively regulates the differentiation of skeletal muscle satellite cells in chicken. 3 Biotech 2019; 9:206. [PMID: 31139537 DOI: 10.1007/s13205-019-1735-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/26/2019] [Indexed: 10/26/2022] Open
Abstract
As a member of four and a half LIM domain proteins, FHL3 gene-encoded protein (FHL3) plays an important role in skeletal muscle development and contraction. In this study, we determined the potential role of FHL3 in the proliferation and differentiation of primary satellite cells in chicken. RT-qPCR results showed that FHL3 mRNA was highly expressed in skeletal muscle in 12 chicken tissues. Four cell proliferation assays (CCK8 assay; EDU staining assay; flow cytometric detection of cell cycle assay; and detection of cell proliferation marker genes Ki67 and N-Ras assay) revealed that FHL3 knockdown had no effect on the proliferation rate of chicken satellite cells. FHL3 knockdown promoted the differentiation of satellite cells into myotubes, as evidenced by increased fusion index, number of nuclei per myotube, Myog, Myh7, Myf5, and Mrf4 mRNA expressions, and myog and myosin heavy chain protein expressions of myogenic markers (P < 0.05). These results showed that the FHL3 was a negative regulator of the differentiation and fusion of chicken satellite cells into myotubes. However, FHL3 expression was increased during the differentiation of chicken satellite cells into myotubes. The study suggested that FHL3 might have different functions in chicken myotubes compared with that in chicken satellite cells.
Collapse
|
22
|
Yoon N, Chu V, Gould M, Zhang M. Spatial and temporal changes in myogenic protein expression by the microenvironment after freeze injury. J Anat 2019; 234:359-367. [PMID: 30657171 DOI: 10.1111/joa.12925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2018] [Indexed: 11/26/2022] Open
Abstract
Skeletal muscle has the remarkable capability to regenerate itself following injury. Adult myogenic stem cells (MSCs) are responsible for the repair and regeneration, and their activity is controlled by intrinsic and extrinsic factors. The aim of this study was to examine and compare the expression levels of Pax3, Pax7, MRF and p38 proteins during the course of regeneration and in different areas of the focal freeze-lesion damaged adult rat TA muscle. Using the focal freeze injury model, immunohistochemistry, laser-capture micro-dissection and Western blot analysis were performed. The results show that (1) in the severely damaged area, the focal freeze-lesion injury significantly activated Pax7 and myogenin expression within 7 days and down-regulated Pax3, MyoD and Myf-5 within 1 or 3 days, and (2) the level of the p38 protein was strongly and transiently up-regulated in the whole muscle on day 7 following injury, whereas the level of the pp38 protein was down-regulated within 3 days in the severely damaged and non-damaged areas. These findings indicate that the temporal (e.g. the time course of regeneration) and spatial (e.g. three zones created by the focal freeze-lesion) cues in a regenerating muscle have a significant impact on the activity of the adult MSCs.
Collapse
Affiliation(s)
- Nara Yoon
- Anatomy Department, University of Otago, Dunedin, New Zealand
| | - Vivian Chu
- Anatomy Department, University of Otago, Dunedin, New Zealand
| | - Maree Gould
- Anatomy Department, University of Otago, Dunedin, New Zealand
| | - Ming Zhang
- Anatomy Department, University of Otago, Dunedin, New Zealand
| |
Collapse
|
23
|
Zhang S, Heng J, Song H, Zhang Y, Lin X, Tian M, Chen F, Guan W. Role of Maternal Dietary Protein and Amino Acids on Fetal Programming, Early Neonatal Development, and Lactation in Swine. Animals (Basel) 2019; 9:ani9010019. [PMID: 30642135 PMCID: PMC6356768 DOI: 10.3390/ani9010019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/08/2019] [Accepted: 01/09/2019] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Dietary protein is an important nutrient source for sows, necessary for not only growth and production, but also other physiological functions. Protein limitations in maternal diets have the potential to impair fetal myogenesis, while excess maternal dietary protein appears to only have minor effects on early fetal muscle formation. Effects of maternal protein deficiency on increased fat deposition in porcine neonates is inconsistent with gene expressions in the neonates. Sufficient maternal dietary protein can enhance porcine milk protein and fat concentration. Understanding the function of protein and amino acids in sows and the effects on their offspring can provide rational approaches for the regulation of piglet growth and further improvements in meat quality in the future. Abstract Maternal nutrition plays a vital role in fetal development, early development of neonates, and lactation and regulates the lifetime productivity of offspring. During pregnancy, maternal nutrition alters expression of the fetal genome and the development of tissues and organs via fetal programming. After parturition, maternal nutrition continues to regulate growth and development of piglets through maternal milk, which contains carbohydrates, lipids, proteins and oligosaccharides. Thus, deficiencies in maternal nutrition are detrimental to development of piglets, which can lead to inefficient growth and decreased carcass merit. Protein is an important nutritional component for sows, which not only functions in muscle development, but also plays a vital role in embryonic and neonatal development and lactation. Although effects of maternal undernutrition on neonatal development have been widely studied in sows, the function of different maternal dietary protein levels on fetal development, neonatal growth and lactation performance of sows is largely unknown. Determination of the effects and underlying mechanisms of maternal dietary protein levels on development of piglets is vital to the pork industry. Therefore, we summarized recent reports regarding mechanisms of effects of maternal protein levels on regulation of conceptus growth and early postnatal development though uterine fetal programming and lactation in swine.
Collapse
Affiliation(s)
- Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China.
| | - Jinghui Heng
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Hanqing Song
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Yufeng Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Xiaofeng Lin
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Min Tian
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Fang Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China.
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China.
- College of Animal Science, South China Agricultural University, Wushan Avenue, Tianhe District, Guangzhou 510642, China.
| |
Collapse
|
24
|
Roveimiab Z, Lin F, Anderson JE. Emerging Development of Microfluidics-Based Approaches to Improve Studies of Muscle Cell Migration. TISSUE ENGINEERING PART B-REVIEWS 2018; 25:30-45. [PMID: 30073911 DOI: 10.1089/ten.teb.2018.0181] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
IMPACT STATEMENT The essential interactions between and among cells in the three types of muscle tissue in development, wound healing, and regeneration of tissues, are underpinned by the ability of cardiac, smooth, and skeletal muscle cells to migrate in maintaining functional capacity after pathologies such as myocardial infarction, tissue grafting, and traumatic and postsurgical injury. Microfluidics-based devices now offer significant enhancement over conventional approaches to studying cell chemotaxis and haptotaxis that are inherent in migration. Advances in experimental approaches to muscle cell movement and tissue formation will contribute to innovations in tissue engineering for patching wound repair and muscle tissue replacement.
Collapse
Affiliation(s)
- Ziba Roveimiab
- 1 Department of Biological Sciences and University of Manitoba, Winnipeg, Canada.,2 Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
| | - Francis Lin
- 1 Department of Biological Sciences and University of Manitoba, Winnipeg, Canada.,2 Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
| | - Judy E Anderson
- 1 Department of Biological Sciences and University of Manitoba, Winnipeg, Canada
| |
Collapse
|
25
|
SNAIL is a key regulator of alveolar rhabdomyosarcoma tumor growth and differentiation through repression of MYF5 and MYOD function. Cell Death Dis 2018; 9:643. [PMID: 29844345 PMCID: PMC5974324 DOI: 10.1038/s41419-018-0693-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/30/2018] [Accepted: 05/10/2018] [Indexed: 12/13/2022]
Abstract
Rhabdomyosarcoma (RMS) is a mesenchymal tumor of soft tissue in children that originates from a myogenic differentiation defect. Expression of SNAIL transcription factor is elevated in the alveolar subtype of RMS (ARMS), characterized by a low myogenic differentiation status and high aggressiveness. In RMS patients SNAIL level increases with higher stage. Moreover, SNAIL level negatively correlates with MYF5 expression. The differentiation of human ARMS cells diminishes SNAIL level. SNAIL silencing in ARMS cells inhibits proliferation and induces differentiation in vitro, and thereby completely abolishes the growth of human ARMS xenotransplants in vivo. SNAIL silencing induces myogenic differentiation by upregulation of myogenic factors and muscle-specific microRNAs, such as miR-206. SNAIL binds to the MYF5 promoter suppressing its expression. SNAIL displaces MYOD from E-box sequences (CANNTG) that are associated with genes expressed during differentiation and G/C rich in their central dinucleotides. SNAIL silencing allows the re-expression of MYF5 and canonical MYOD binding, promoting ARMS cell myogenic differentiation. In differentiating ARMS cells SNAIL forms repressive complex with histone deacetylates 1 and 2 (HDAC1/2) and regulates their expression. Accordingly, in human myoblasts SNAIL silencing induces differentiation by upregulation of myogenic factors. Our data clearly point to SNAIL as a key regulator of myogenic differentiation and a new promising target for future ARMS therapies.
Collapse
|
26
|
An Y, Wang G, Diao Y, Long Y, Fu X, Weng M, Zhou L, Sun K, Cheung TH, Ip NY, Sun H, Wang H, Wu Z. A Molecular Switch Regulating Cell Fate Choice between Muscle Progenitor Cells and Brown Adipocytes. Dev Cell 2017; 41:382-391.e5. [PMID: 28535373 DOI: 10.1016/j.devcel.2017.04.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 02/16/2017] [Accepted: 04/19/2017] [Indexed: 11/16/2022]
Abstract
During mouse embryo development, both muscle progenitor cells (MPCs) and brown adipocytes (BAs) are known to derive from the same Pax7+/Myf5+ progenitor cells. However, the underlying mechanisms for the cell fate control remain unclear. In Pax7-null MPCs from young mice, several BA-specific genes, including Prdm16 and Ucp1 and many other adipocyte-related genes, were upregulated with a concomitant reduction of Myod and Myf5, two muscle lineage-determining genes. This suggests a cell fate switch from MPC to BA. Consistently, freshly isolated Pax7-null but not wild-type MPCs formed lipid-droplet-containing UCP1+ BA in culture. Mechanistically, MyoD and Myf5, both known transcription targets of Pax7 in MPC, potently repress Prdm16, a BA-specific lineage-determining gene, via the E2F4/p107/p130 transcription repressor complex. Importantly, inducible Pax7 ablation in developing mouse embryos promoted brown fat development. Thus, the MyoD/Myf5-E2F4/p107/p130 axis functions in both the Pax7+/Myf5+ embryonic progenitor cells and postnatal myoblasts to repress the alternative BA fate.
Collapse
Affiliation(s)
- Yitai An
- Division of Life Science, Center for Stem Cell Research, Center of Systems Biology and Human Health, State Key Laboratory in Molecular Neuroscience, Hong Kong University of Science & Technology, Clearwater Bay, Kowloon, Hong Kong, China
| | - Gang Wang
- Division of Life Science, Center for Stem Cell Research, Center of Systems Biology and Human Health, State Key Laboratory in Molecular Neuroscience, Hong Kong University of Science & Technology, Clearwater Bay, Kowloon, Hong Kong, China
| | - Yarui Diao
- Division of Life Science, Center for Stem Cell Research, Center of Systems Biology and Human Health, State Key Laboratory in Molecular Neuroscience, Hong Kong University of Science & Technology, Clearwater Bay, Kowloon, Hong Kong, China
| | - Yanyang Long
- Division of Life Science, Center for Stem Cell Research, Center of Systems Biology and Human Health, State Key Laboratory in Molecular Neuroscience, Hong Kong University of Science & Technology, Clearwater Bay, Kowloon, Hong Kong, China
| | - Xinrong Fu
- Division of Life Science, Center for Stem Cell Research, Center of Systems Biology and Human Health, State Key Laboratory in Molecular Neuroscience, Hong Kong University of Science & Technology, Clearwater Bay, Kowloon, Hong Kong, China
| | - Mingxi Weng
- Division of Life Science, Center for Stem Cell Research, Center of Systems Biology and Human Health, State Key Laboratory in Molecular Neuroscience, Hong Kong University of Science & Technology, Clearwater Bay, Kowloon, Hong Kong, China
| | - Liang Zhou
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Kun Sun
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Tom H Cheung
- Division of Life Science, Center for Stem Cell Research, Center of Systems Biology and Human Health, State Key Laboratory in Molecular Neuroscience, Hong Kong University of Science & Technology, Clearwater Bay, Kowloon, Hong Kong, China
| | - Nancy Y Ip
- Division of Life Science, Center for Stem Cell Research, Center of Systems Biology and Human Health, State Key Laboratory in Molecular Neuroscience, Hong Kong University of Science & Technology, Clearwater Bay, Kowloon, Hong Kong, China
| | - Hao Sun
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Huating Wang
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhenguo Wu
- Division of Life Science, Center for Stem Cell Research, Center of Systems Biology and Human Health, State Key Laboratory in Molecular Neuroscience, Hong Kong University of Science & Technology, Clearwater Bay, Kowloon, Hong Kong, China.
| |
Collapse
|
27
|
Siqin Q, Nishiumi T, Yamada T, Wang S, Liu W, Wu R, Borjigin G. Relationships among muscle fiber type composition, fiber diameter and MRF gene expression in different skeletal muscles of naturally grazing Wuzhumuqin sheep during postnatal development. Anim Sci J 2017; 88:2033-2043. [PMID: 28730689 PMCID: PMC5763406 DOI: 10.1111/asj.12848] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 04/20/2017] [Indexed: 01/08/2023]
Abstract
The aim of this study was to determine the relationships among muscle fiber‐type composition, fiber diameter, and myogenic regulatory factor (MRF) gene expression in different skeletal muscles during development in naturally grazing Wuzhumuqin sheep. Three major muscles (i.e. the Longissimus dorsi (LD), Biceps femoris (BF) and Triceps brachii (TB)) were obtained from 20 Wuzhumuqin sheep and 20 castrated rams at each of the following ages: 1, 3, 6, 9, 12 and 18 months. Muscle fiber‐type composition and fiber diameter were measured using histochemistry and morphological analysis, and MRF gene expression levels were determined using real‐time PCR. In the LD muscle, changes in the proportion of each of different types of fiber (I, IIA and IIB) were relatively small. In the BF muscle, a higher proportion of type I and a 6.19‐fold lower proportion of type IIA fibers were observed (P < 0.05). In addition, the compositions of type I and IIA fibers continuously changed in the TB muscle (P < 0.05). Moreover, muscle diameter gradually increased throughout development (P < 0.05). Almost no significant difference was found in MRF gene expression patterns, which appeared to be relatively stable. These results suggest that changes in fiber‐type composition and increases in fiber size may be mutually interacting processes during muscle development.
Collapse
Affiliation(s)
- Qimuge Siqin
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Tadayuki Nishiumi
- Division of Life and Food Science, Graduate School of Science and Technology, Niigata University, Niigata, Japan
| | - Takahisa Yamada
- Division of Life and Food Science, Graduate School of Science and Technology, Niigata University, Niigata, Japan
| | - Shuiqing Wang
- Mongolian Sheep Animal Husbandry Co., Ltd, Hohhot, China
| | - Wenjun Liu
- Division of Life and Food Science, Graduate School of Science and Technology, Niigata University, Niigata, Japan
| | - Rihan Wu
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Gerelt Borjigin
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
28
|
Guo Y, Wang J, Zhu M, Zeng R, Xu Z, Li G, Zuo B. Identification of MyoD-Responsive Transcripts Reveals a Novel Long Non-coding RNA (lncRNA-AK143003) that Negatively Regulates Myoblast Differentiation. Sci Rep 2017; 7:2828. [PMID: 28588232 PMCID: PMC5460278 DOI: 10.1038/s41598-017-03071-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/21/2017] [Indexed: 02/04/2023] Open
Abstract
Myogenic differentiation factor (MyoD) is a master transcription factor in muscle development and differentiation. Although several long non-coding RNAs (lncRNAs) linked to MyoD have been found to influence muscle development, the functions of many lncRNAs have not been explored. Here we utilized lncRNA and mRNA microarray analysis to identify potential lncRNAs regulated by MyoD in muscle cells. A total of 997 differentially expressed lncRNAs (335 up-regulated and 662 down-regulated) and 1,817 differentially expressed mRNAs (148 up-regulated and 1,669 down-regulated) were identified after MyoD knockdown in C2C12 cells. Functional predictions suggested that most lncRNAs are involved in the biological pathways related to muscle differentiation and cell cycle with co-expressed genes. To gain further insight into the MyoD-mediated lncRNA expression in muscle differentiation, tissue expression profiles and MyoD overexpression were performed, and we found one of the candidate lncRNAs-AK143003 was significantly regulated by MyoD. Further analyses showed its noncoding ability and cytoplasmic localisation. Silencing of AK143003 stimulated the accumulation of myogenic marker genes, whereas AK143003 overexpression led to their decreased synthesis. This study identified a multitude of MyoD-mediated lncRNAs for further investigation and identified a novel lncRNA, lnc-AK143003, which plays a role in controlling muscle differentiation.
Collapse
Affiliation(s)
- Yiwen Guo
- 0000 0004 1790 4137grid.35155.37Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan, 430070 Hubei P.R. China
| | - Jingnan Wang
- 0000 0004 1790 4137grid.35155.37Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan, 430070 Hubei P.R. China
| | - Mingfei Zhu
- 0000 0004 1790 4137grid.35155.37Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan, 430070 Hubei P.R. China
| | - Rui Zeng
- 0000 0004 1790 4137grid.35155.37Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan, 430070 Hubei P.R. China
| | - Zaiyan Xu
- 0000 0004 1790 4137grid.35155.37Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan, 430070 Hubei P.R. China
| | - Guoliang Li
- 0000 0004 1790 4137grid.35155.37National Key Laboratory of Crop Genetic Improvement, Agricultural Bioinformatics Key Laboratory of Hubei Province, College of Informatics, Huazhong Agricultural University, Wuhan, 430070 Hubei P.R. China
| | - Bo Zuo
- 0000 0004 1790 4137grid.35155.37Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan, 430070 Hubei P.R. China ,grid.35155.370000 0004 1790 4137The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070 China
| |
Collapse
|
29
|
Ji Y, Wu Z, Dai Z, Wang X, Li J, Wang B, Wu G. Fetal and neonatal programming of postnatal growth and feed efficiency in swine. J Anim Sci Biotechnol 2017; 8:42. [PMID: 28484595 PMCID: PMC5420136 DOI: 10.1186/s40104-017-0173-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 04/18/2017] [Indexed: 01/10/2023] Open
Abstract
Maternal undernutrition or overnutrition during pregnancy alters organ structure, impairs prenatal and neonatal growth and development, and reduces feed efficiency for lean tissue gains in pigs. These adverse effects may be carried over to the next generation or beyond. This phenomenon of the transgenerational impacts is known as fetal programming, which is mediated by stable and heritable alterations of gene expression through covalent modifications of DNA and histones without changes in DNA sequences (namely, epigenetics). The mechanisms responsible for the epigenetic regulation of protein expression and functions include chromatin remodeling; DNA methylation (occurring at the 5´-position of cytosine residues within CpG dinucleotides); and histone modifications (acetylation, methylation, phosphorylation, and ubiquitination). Like maternal malnutrition, undernutrition during the neonatal period also reduces growth performance and feed efficiency (weight gain:feed intake; also known as weight-gain efficiency) in postweaning pigs by 5-10%, thereby increasing the days necessary to reach the market body-weight. Supplementing functional amino acids (e.g., arginine and glutamine) and vitamins (e.g., folate) play a key role in activating the mammalian target of rapamycin signaling and regulating the provision of methyl donors for DNA and protein methylation. Therefore, these nutrients are beneficial for the dietary treatment of metabolic disorders in offspring with intrauterine growth restriction or neonatal malnutrition. The mechanism-based strategies hold great promise for the improvement of the efficiency of pork production and the sustainability of the global swine industry.
Collapse
Affiliation(s)
- Yun Ji
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193 China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193 China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193 China
| | - Xiaolong Wang
- Henan Yinfa Animal Husbandry Co., Ltd., Xinzheng, Henan 451100 China
| | - Ju Li
- Henan Yinfa Animal Husbandry Co., Ltd., Xinzheng, Henan 451100 China
| | - Binggen Wang
- Henan Yinfa Animal Husbandry Co., Ltd., Xinzheng, Henan 451100 China
| | - Guoyao Wu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193 China.,Department of Animal Science and Center for Animal Genomics, Texas A&M University, Room 212, College Station, TX 77843 USA
| |
Collapse
|
30
|
Hernandez-Torres F, Rodríguez-Outeiriño L, Franco D, Aranega AE. Pitx2 in Embryonic and Adult Myogenesis. Front Cell Dev Biol 2017; 5:46. [PMID: 28507987 PMCID: PMC5410577 DOI: 10.3389/fcell.2017.00046] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/12/2017] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle is a heterogeneous tissue that represents between 30 and 38% of the human body mass and has important functions in the organism, such as maintaining posture, locomotor impulse, or pulmonary ventilation. The genesis of skeletal muscle during embryonic development is a process controlled by an elaborate regulatory network combining the interplay of extrinsic and intrinsic regulatory mechanisms that transform myogenic precursor cells into functional muscle fibers through a finely tuned differentiation program. However, the capacity of generating muscle still remains once these fibers have matured. Adult myogenesis resembles many of the embryonic morphogenetic episodes and depends on the activation of satellite cells that have the potential to differentiate into new muscle fibers. Pitx2 is a member of the bicoid family of homeodomain transcription factors that play an important role in morphogenesis. In the last decade, Pitx2 has emerged as a key element involved in the fine-tuning mechanism that regulates skeletal-muscle development as well as the differentiation and cell fate of satellite cells in adult muscle. Here we present an integrative view of all aspects of embryonic and adult myogenesis in which Pitx2 is involved, from embryonic development to satellite-cell proliferation, fate specification, and differentiation. Those new Pitx2 functions on satellite-cell biology might open new perspectives to develop therapeutic strategies for muscular disorders.
Collapse
Affiliation(s)
- Francisco Hernandez-Torres
- Cardiac and Skeletal Myogenesis Group, Departmento de Biología Experimental, Universidad de JaénJaén, Spain.,Cardiac and Skeletal Myogenesis Group, Fundación MEDINA Centro de Excelencia en Investigación de Medicamentos Innovadores en AndalucíaGranada, Spain
| | - Lara Rodríguez-Outeiriño
- Cardiac and Skeletal Myogenesis Group, Departmento de Biología Experimental, Universidad de JaénJaén, Spain.,Cardiac and Skeletal Myogenesis Group, Fundación MEDINA Centro de Excelencia en Investigación de Medicamentos Innovadores en AndalucíaGranada, Spain
| | - Diego Franco
- Cardiac and Skeletal Myogenesis Group, Departmento de Biología Experimental, Universidad de JaénJaén, Spain.,Cardiac and Skeletal Myogenesis Group, Fundación MEDINA Centro de Excelencia en Investigación de Medicamentos Innovadores en AndalucíaGranada, Spain
| | - Amelia E Aranega
- Cardiac and Skeletal Myogenesis Group, Departmento de Biología Experimental, Universidad de JaénJaén, Spain.,Cardiac and Skeletal Myogenesis Group, Fundación MEDINA Centro de Excelencia en Investigación de Medicamentos Innovadores en AndalucíaGranada, Spain
| |
Collapse
|
31
|
Myogenic Differentiation from MYOGENIN-Mutated Human iPS Cells by CRISPR/Cas9. Stem Cells Int 2017; 2017:9210494. [PMID: 28473859 PMCID: PMC5394914 DOI: 10.1155/2017/9210494] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 03/05/2017] [Indexed: 01/02/2023] Open
Abstract
It is well known that myogenic regulatory factors encoded by the Myod1 family of genes have pivotal roles in myogenesis, with partially overlapping functions, as demonstrated for the mouse embryo. Myogenin-mutant mice, however, exhibit severe myogenic defects without compensation by other myogenic factors. MYOGENIN might be expected to have an analogous function in human myogenic cells. To verify this hypothesis, we generated MYOGENIN-mutated human iPS cells by using CRISPR/Cas9 genome-editing technology. Our results suggest that MYOD1-independent or MYOD1-dependent mechanisms can compensate for the loss of MYOGENIN and that these mechanisms are likely to be crucial for regulating skeletal muscle differentiation and formation.
Collapse
|
32
|
Loss of MyoD Promotes Fate Transdifferentiation of Myoblasts Into Brown Adipocytes. EBioMedicine 2017; 16:212-223. [PMID: 28117277 PMCID: PMC5474440 DOI: 10.1016/j.ebiom.2017.01.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 01/10/2017] [Accepted: 01/10/2017] [Indexed: 02/08/2023] Open
Abstract
Brown adipose tissue (BAT) represents a promising agent to ameliorate obesity and other metabolic disorders. However, the abundance of BAT decreases with age and BAT paucity is a common feature of obese subjects. As brown adipocytes and myoblasts share a common Myf5 lineage origin, elucidating the molecular mechanisms underlying the fate choices of brown adipocytes versus myoblasts may lead to novel approaches to expand BAT mass. Here we identify MyoD as a key negative regulator of brown adipocyte development. CRISPR/CAS9-mediated deletion of MyoD in C2C12 myoblasts facilitates their adipogenic transdifferentiation. MyoD knockout downregulates miR-133 and upregulates the miR-133 target Igf1r, leading to amplification of PI3K–Akt signaling. Accordingly, inhibition of PI3K or Akt abolishes the adipogenic gene expression of MyoD null myoblasts. Strikingly, loss of MyoD converts satellite cell-derived primary myoblasts to brown adipocytes through upregulation of Prdm16, a target of miR-133 and key determinant of brown adipocyte fate. Conversely, forced expression of MyoD in brown preadipocytes blocks brown adipogenesis and upregulates the expression of myogenic genes. Importantly, miR-133a knockout significantly blunts the inhibitory effect of MyoD on brown adipogenesis. Our results establish MyoD as a negative regulator of brown adipocyte development by upregulating miR-133 to suppress Akt signaling and Prdm16. Loss of MyoD facilitates adipogenic transdifferentiation of myoblasts. Overexpression of MyoD transdifferentiate brown preadipocytes to myoblasts. MyoD acts partially through miR-133 to suppress brown adipocyte cell fate.
Brown fat burns fat to produce heat, and represents a promising agent to treat obesity and its related disorders. Brown fat cells and muscle cells share a common origin, but what controls the developmental separation of the two cell types is not well understood. This study reports that inhibition of “MyoD” gene in muscle progenitors promotes their differentiation into brown fat cells in mice. Conversely, forced expression of MyoD in brown fat progenitors converts them into muscle cells. This work suggests that inhibition of MyoD may represent a future direction to expand brown fat and alleviate obesity in humans.
Collapse
|
33
|
Deries M, Thorsteinsdóttir S. Axial and limb muscle development: dialogue with the neighbourhood. Cell Mol Life Sci 2016; 73:4415-4431. [PMID: 27344602 PMCID: PMC11108464 DOI: 10.1007/s00018-016-2298-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 06/03/2016] [Accepted: 06/21/2016] [Indexed: 11/29/2022]
Abstract
Skeletal muscles are part of the musculoskeletal system which also includes nerves, tendons, connective tissue, bones and blood vessels. Here we review the development of axial and limb muscles in amniotes within the context of their surrounding tissues in vivo. We highlight the reciprocal dialogue mediated by signalling factors between cells of these adjacent tissues and developing muscles and also demonstrate its importance from the onset of muscle cell differentiation well into foetal development. Early embryonic tissues secrete factors which are important regulators of myogenesis. However, later muscle development relies on other tissue collaborators, such as developing nerves and connective tissue, which are in turn influenced by the developing muscles themselves. We conclude that skeletal muscle development in vivo is a compelling example of the importance of reciprocal interactions between developing tissues for the complete and coordinated development of a functional system.
Collapse
Affiliation(s)
- Marianne Deries
- Centro de Ecologia, Evolução e Alterações Ambientais, Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal.
| | - Sólveig Thorsteinsdóttir
- Centro de Ecologia, Evolução e Alterações Ambientais, Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
34
|
Ilavenil S, Kim DH, Srigopalram S, Arasu MV, Lee KD, Lee JC, Lee JS, Renganathan S, Choi KC. Potential Application of p-Coumaric Acid on Differentiation of C2C12 Skeletal Muscle and 3T3-L1 Preadipocytes-An in Vitro and in Silico Approach. Molecules 2016; 21:molecules21080997. [PMID: 27490527 PMCID: PMC6274435 DOI: 10.3390/molecules21080997] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 07/26/2016] [Accepted: 07/27/2016] [Indexed: 01/10/2023] Open
Abstract
Coumaric acid (CA) is a phenolic acid of the hydroxycinnamic acid family, and it has many biological functions such as anti-oxidant, anti-inflammatory, antidiabetic, anti-ulcer, anti-platelet, anti-cancer activities, etc. In the present study, we planned to analyse the potential molecular function of CA on skeletal muscle and preadipocytes differentiation using PCR and Western blot techniques. First, we analysed the impact of CA on C2C12 skeletal muscle differentiation. It revealed that CA treatment inhibited horse serum-induced skeletal muscle differentiation as evidenced by the decreased expression of early myogenic differentiation markers such as Myogenin and myoD via the AMP activated protein kinase- alpha AMPK-α mediated pathway. Furthermore, the level of lipid accumulation and changes in genes and protein expressions that are associated with lipogenesis and lipolysis were analyzed in 3T3-L1 cells. The Oil Red O staining evidenced that CA treatment inhibited lipid accumulation at the concentration of 0.1 and 0.2 mM. Furthermore, coumaric acid treatment decreased the expression of main transcriptional factors such as CCAAT/enhancer binding protein-alpha (C/EBP-α) and peroxisome proliferator-activated receptor gamma-2 (PPAR-γ2). Subsequently, CA treatment decreased the expression of sterol regulatory element binding protein-1 (SREBP-1), fatty acid synthase (FAS), acetyl CoA carboxylase (ACC) and adiponectin. Finally, we identified conformational changes induced by CA in PPAR-γ2 using computational biology tools. It revealed that CA might downregulate the PPAR-γ2 expression by directly binding with amino acids of PPAR-γ2 by hydrogen at 3.26 distance and hydrophobic interactions at 3.90 contact distances. These data indicated that CA suppressed skeletal muscle and preadipocytes differentiation through downregulation of the main transcriptional factors and their downstream targets.
Collapse
Affiliation(s)
- Soundharrajan Ilavenil
- Grassland and Forage Division, National Institute of Animal Science, Rural Development Administration, Cheonan 330-801, Korea.
| | - Da Hye Kim
- Laboratory of Animal Physiology, Graduate School of Agricultural Science, Tohoku University, Aoba, Sendai 980-8577, Japan.
| | - Srisesharam Srigopalram
- Grassland and Forage Division, National Institute of Animal Science, Rural Development Administration, Cheonan 330-801, Korea.
| | - Mariadhas Valan Arasu
- Department of Botany and Microbiology, Addiriyah Chair for Environmental Studies, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Kyung Dong Lee
- Department of Oriental Medicine Materials, Dongsin University, Naju 520-714, Korea.
| | - Jeong Chae Lee
- Research Center of Bioactive Materials, Institute of Molecular Biology and Genetics, Chonbuk National University, Jeonju 561-756, Korea.
| | - Jong Suk Lee
- Biocenter, Gyeonggi Institute of Science and Technology, Suwon 443-270, Korea.
| | - Senthil Renganathan
- Department of Bioinformatics, Marudupandiyar College, Tamilnadu 613-403, India.
| | - Ki Choon Choi
- Grassland and Forage Division, National Institute of Animal Science, Rural Development Administration, Cheonan 330-801, Korea.
| |
Collapse
|
35
|
Sabillo A, Ramirez J, Domingo CR. Making muscle: Morphogenetic movements and molecular mechanisms of myogenesis in Xenopus laevis. Semin Cell Dev Biol 2016; 51:80-91. [PMID: 26853935 DOI: 10.1016/j.semcdb.2016.02.006] [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: 10/29/2015] [Accepted: 02/01/2016] [Indexed: 12/15/2022]
Abstract
Xenopus laevis offers unprecedented access to the intricacies of muscle development. The large, robust embryos make it ideal for manipulations at both the tissue and molecular level. In particular, this model system can be used to fate map early muscle progenitors, visualize cell behaviors associated with somitogenesis, and examine the role of signaling pathways that underlie induction, specification, and differentiation of muscle. Several characteristics that are unique to X. laevis include myogenic waves with distinct gene expression profiles and the late formation of dermomyotome and sclerotome. Furthermore, myogenesis in the metamorphosing frog is biphasic, facilitating regeneration studies. In this review, we describe the morphogenetic movements that shape the somites and discuss signaling and transcriptional regulation during muscle development and regeneration. With recent advances in gene editing tools, X. laevis remains a premier model organism for dissecting the complex mechanisms underlying the specification, cell behaviors, and formation of the musculature system.
Collapse
Affiliation(s)
- Armbien Sabillo
- Department of Molecular & Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Julio Ramirez
- Department of Biology, San Francisco State University, CA 94132, USA
| | - Carmen R Domingo
- Department of Biology, San Francisco State University, CA 94132, USA.
| |
Collapse
|
36
|
Panda AC, Abdelmohsen K, Martindale JL, Di Germanio C, Yang X, Grammatikakis I, Noh JH, Zhang Y, Lehrmann E, Dudekula DB, De S, Becker KG, White EJ, Wilson GM, de Cabo R, Gorospe M. Novel RNA-binding activity of MYF5 enhances Ccnd1/Cyclin D1 mRNA translation during myogenesis. Nucleic Acids Res 2016; 44:2393-408. [PMID: 26819411 PMCID: PMC4797292 DOI: 10.1093/nar/gkw023] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/08/2016] [Indexed: 01/07/2023] Open
Abstract
Skeletal muscle contains long multinucleated and contractile structures known as muscle fibers, which arise from the fusion of myoblasts into multinucleated myotubes during myogenesis. The myogenic regulatory factor (MRF) MYF5 is the earliest to be expressed during myogenesis and functions as a transcription factor in muscle progenitor cells (satellite cells) and myocytes. In mouse C2C12 myocytes, MYF5 is implicated in the initial steps of myoblast differentiation into myotubes. Here, using ribonucleoprotein immunoprecipitation (RIP) analysis, we discovered a novel function for MYF5 as an RNA-binding protein which associated with a subset of myoblast mRNAs. One prominent MYF5 target was Ccnd1 mRNA, which encodes the key cell cycle regulator CCND1 (Cyclin D1). Biotin-RNA pulldown, UV-crosslinking and gel shift experiments indicated that MYF5 was capable of binding the 3' untranslated region (UTR) and the coding region (CR) of Ccnd1 mRNA. Silencing MYF5 expression in proliferating myoblasts revealed that MYF5 promoted CCND1 translation and modestly increased transcription of Ccnd1 mRNA. Accordingly, overexpressing MYF5 in C2C12 cells upregulated CCND1 expression while silencing MYF5 reduced myoblast proliferation as well as differentiation of myoblasts into myotubes. Moreover, MYF5 silencing reduced myogenesis, while ectopically restoring CCND1 abundance partially rescued the decrease in myogenesis seen after MYF5 silencing. We propose that MYF5 enhances early myogenesis in part by coordinately elevating Ccnd1 transcription and Ccnd1 mRNA translation.
Collapse
Affiliation(s)
- Amaresh C Panda
- Laboratory of Genetics, National Institute on Aging, NIH, Baltimore, MD21224, USA
| | - Kotb Abdelmohsen
- Laboratory of Genetics, National Institute on Aging, NIH, Baltimore, MD21224, USA
| | | | - Clara Di Germanio
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Xiaoling Yang
- Laboratory of Genetics, National Institute on Aging, NIH, Baltimore, MD21224, USA
| | | | - Ji Heon Noh
- Laboratory of Genetics, National Institute on Aging, NIH, Baltimore, MD21224, USA
| | - Yongqing Zhang
- Laboratory of Genetics, National Institute on Aging, NIH, Baltimore, MD21224, USA
| | - Elin Lehrmann
- Laboratory of Genetics, National Institute on Aging, NIH, Baltimore, MD21224, USA
| | - Dawood B Dudekula
- Laboratory of Genetics, National Institute on Aging, NIH, Baltimore, MD21224, USA
| | - Supriyo De
- Laboratory of Genetics, National Institute on Aging, NIH, Baltimore, MD21224, USA
| | - Kevin G Becker
- Laboratory of Genetics, National Institute on Aging, NIH, Baltimore, MD21224, USA
| | - Elizabeth J White
- Department of Biochemistry and Molecular Biology and Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Gerald M Wilson
- Department of Biochemistry and Molecular Biology and Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Myriam Gorospe
- Laboratory of Genetics, National Institute on Aging, NIH, Baltimore, MD21224, USA
| |
Collapse
|
37
|
Endo T. Molecular mechanisms of skeletal muscle development, regeneration, and osteogenic conversion. Bone 2015; 80:2-13. [PMID: 26453493 DOI: 10.1016/j.bone.2015.02.028] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 02/18/2015] [Accepted: 02/28/2015] [Indexed: 12/21/2022]
Abstract
Both skeletal muscle and bone are of mesodermal origin and derived from somites during embryonic development. Somites differentiate into the dorsal dermomyotome and the ventral sclerotome, which give rise to skeletal muscle and bone, respectively. Extracellular signaling molecules, such as Wnt and Shh, secreted from the surrounding environment, determine the developmental fate of skeletal muscle. Dermomyotome cells are specified as trunk muscle progenitor cells by transcription factor networks involving Pax3. These progenitor cells delaminate and migrate to form the myotome, where they are determined as myoblasts that differentiate into myotubes or myofibers. The MyoD family of transcription factors plays pivotal roles in myogenic determination and differentiation. Adult skeletal muscle regenerates upon exercise, muscle injury, or degeneration. Satellite cells are muscle-resident stem cells and play essential roles in muscle growth and regeneration. Muscle regeneration recapitulates the process of muscle development in many aspects. In certain muscle diseases, ectopic calcification or heterotopic ossification, as well as fibrosis and adipogenesis, occurs in skeletal muscle. Muscle-resident mesenchymal progenitor cells, which may be derived from vascular endothelial cells, are responsible for the ectopic osteogenesis, fibrogenesis, and adipogenesis. The small GTPase M-Ras is likely to participate in the ectopic calcification and ossification, as well as in osteogenesis during development. This article is part of a Special Issue entitled "Muscle Bone Interactions".
Collapse
Affiliation(s)
- Takeshi Endo
- Department of Biology, Graduate School of Science, Chiba University, Yayoicho, Inageku, Chiba, Chiba 263-8522, Japan.
| |
Collapse
|
38
|
The influence of maternal energy status during mid-gestation on beef offspring tenderness, muscle characteristics, and gene expression. Meat Sci 2015; 110:201-11. [PMID: 26253836 DOI: 10.1016/j.meatsci.2015.07.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 07/06/2015] [Accepted: 07/23/2015] [Indexed: 01/24/2023]
Abstract
The objective of this study was to determine if maternal energy status during mid-gestation influences the expression of genes regulating muscle and fat development, and muscle characteristics that may impact meat tenderness. Cows grazed dormant, native range (Positive Energy Status [PES]) or were fed at 80% of maintenance energy requirements (Negative Energy Status [NES]) during mid-gestation. Steer offspring were harvested after 21 d in the feedlot (weaning subsample) or after 208 d in the feedlot (final subsample). Greater 21-d tenderness was observed in NES steers, resulting from reduced collagen content in longissimus lumborum steaks. In the semitendinosus, NES steers had greater soluble collagen, and down-regulated expression of MHC-IIA and TIMP-3 at weaning, while MHC-IIA expression was up-regulated in NES steers in the final harvest. Data show mid-gestational maternal energy status may impact offspring tenderness and collagen, but differences were not detected in expression of genes important in myogenesis and adipogenesis in muscle samples obtained from steers at weaning or slaughter.
Collapse
|
39
|
Abstract
There are three different types of adipose tissue (AT)-brown, white, and beige-that differ with stage of development, species, and anatomical location. Of these, brown AT (BAT) is the least abundant but has the greatest potential impact on energy balance. BAT is capable of rapidly producing large amounts of heat through activation of the unique uncoupling protein 1 (UCP1) located within the inner mitochondrial membrane. White AT is an endocrine organ and site of lipid storage, whereas beige AT is primarily white but contains some cells that possess UCP1. BAT first appears in the fetus around mid-gestation and is then gradually lost through childhood, adolescence, and adulthood. We focus on the interrelationships between adipocyte classification, anatomical location, and impact of diet in early life together with the extent to which fat development differs between the major species examined. Ultimately, novel dietary interventions designed to reactivate BAT could be possible.
Collapse
Affiliation(s)
- Michael E Symonds
- Division of Child Health, Obstetrics and Gynaecology, School of Medicine, University of Nottingham, Nottingham NG7 2UH, United Kingdom; , ,
| | | | | |
Collapse
|
40
|
Coles CA, Wadeson J, Leyton CP, Siddell JP, Greenwood PL, White JD, McDonagh MB. Proliferation rates of bovine primary muscle cells relate to liveweight and carcase weight in cattle. PLoS One 2015; 10:e0124468. [PMID: 25875203 PMCID: PMC4398453 DOI: 10.1371/journal.pone.0124468] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 03/11/2015] [Indexed: 12/03/2022] Open
Abstract
Muscling in cattle is largely influenced by genetic background, ultimately affecting beef yield and is of major interest to the beef industry. This investigation aimed to determine whether primary skeletal muscle cells isolated from different breeds of cattle with a varying genetic potential for muscling differ in their myogenic proliferative capacity. Primary skeletal muscle cells were isolated and cultured from the Longissimus muscle (LM) of 6 month old Angus, Hereford and Wagyu X Angus cattle. Cells were assessed for rate of proliferation and gene expression of PAX7, MYOD, MYF5, and MYOG. Proliferation rates were found to differ between breeds of cattle whereby myoblasts from Angus cattle were found to proliferate at a greater rate than those of Hereford and Wagyu X Angus during early stages of growth (5–20 hours in culture) in vitro (P < 0.05). The proliferation rates of myoblasts during early stages of culture in vitro were also found to be positively related to the liveweight and carcase weight of cattle (P < 0.05). Gene expression of MYF5 was also found to be significantly down-regulated in WagyuX compared with Angus cattle (P < 0.05). These findings suggest that early events during myogenesis are important for determining liveweight and caracase weights in cattle.
Collapse
Affiliation(s)
- Chantal A. Coles
- Department of Primary Industries Victoria, Discovery Technologies, Biosciences Research Division, Melbourne, Victoria 3083, Australia
- Department of Veterinary Science, University of Melbourne, Melbourne, Victoria 3010, Australia
- * E-mail:
| | - Jenny Wadeson
- Department of Primary Industries Victoria, Discovery Technologies, Biosciences Research Division, Melbourne, Victoria 3083, Australia
| | - Carolina P. Leyton
- Department of Primary Industries Victoria, Discovery Technologies, Biosciences Research Division, Melbourne, Victoria 3083, Australia
| | - Jason P. Siddell
- Cooperative Research Centre for Cattle and Beef Quality, University of New England, Armidale, NSW 2351, Australia
- New South Wales Department of Primary Industries, Beef Industry Centre of Excellence, Armidale, NSW 2351, Australia
| | - Paul L. Greenwood
- Cooperative Research Centre for Cattle and Beef Quality, University of New England, Armidale, NSW 2351, Australia
- New South Wales Department of Primary Industries, Beef Industry Centre of Excellence, Armidale, NSW 2351, Australia
| | - Jason D. White
- Department of Veterinary Science, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Matthew B. McDonagh
- Department of Primary Industries Victoria, Discovery Technologies, Biosciences Research Division, Melbourne, Victoria 3083, Australia
- Cooperative Research Centre for Cattle and Beef Quality, University of New England, Armidale, NSW 2351, Australia
- New South Wales Department of Primary Industries, Beef Industry Centre of Excellence, Armidale, NSW 2351, Australia
| |
Collapse
|
41
|
Kim M, Sung B, Kang YJ, Kim DH, Lee Y, Hwang SY, Yoon JH, Yoo MA, Kim CM, Chung HY, Kim ND. The combination of ursolic acid and leucine potentiates the differentiation of C2C12 murine myoblasts through the mTOR signaling pathway. Int J Mol Med 2015; 35:755-62. [PMID: 25529824 DOI: 10.3892/ijmm.2014.2046] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 12/08/2014] [Indexed: 11/06/2022] Open
Abstract
Aging causes phenotypic changes in skeletal muscle progenitor cells that lead to the progressive loss of myogenic differentiation and thus a decrease in muscle mass. The naturally occurring triterpene, ursolic acid, has been reported to be an effective agent for the prevention of muscle loss by suppressing degenerative muscular dystrophy. Leucine, a branched-chain amino acid, and its metabolite, β-hydroxy-β-methylbutyric acid, have been reported to enhance protein synthesis in skeletal muscle. Therefore, the aim of the present study was to investigate whether the combination of ursolic acid and leucine promotes greater myogenic differentiation compared to either agent alone in C2C12 murine myoblasts. Morphological changes were observed and creatine kinase (CK) activity analysis was performed to determine the conditions through which the combination of ursolic acid and leucine would exert the most prominent effects on muscle cell differentiation. The effect of the combination of ursolic acid and leucine on the expression of myogenic differentiation marker genes was examined by RT-PCR and western blot analysis. The combination of ursolic acid (0.5 µM) and leucine (10 µM) proved to be the most effective in promoting myogenic differentiation. The combination of ursolic acid and leucine significantly increased CK activity than treatment with either agent alone. The level of myosin heavy chain, a myogenic differentiation marker protein, was also enhanced by the combination of ursolic acid and leucine. The combination of ursolic acid and leucine significantly induced the expression of myogenic differentiation marker genes, such as myogenic differentiation 1 (MyoD) and myogenin, at both the mRNA and protein level. In addition, the number of myotubes and the fusion index were increased. These findings indicate that the combination of ursolic acid and leucine promotes muscle cell differentiation, thus suggesting that this combination of agents may prove to be beneficial in increasing muscle mass.
Collapse
Affiliation(s)
- Minjung Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609‑735, Republic of Korea
| | - Bokyung Sung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609‑735, Republic of Korea
| | - Yong Jung Kang
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609‑735, Republic of Korea
| | - Dong Hwan Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609‑735, Republic of Korea
| | - Yujin Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609‑735, Republic of Korea
| | - Seong Yeon Hwang
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609‑735, Republic of Korea
| | - Jeong-Hyun Yoon
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609‑735, Republic of Korea
| | - Mi-Ae Yoo
- Department of Molecular Biology, Pusan National University, Busan 609‑735, Republic of Korea
| | - Cheol Min Kim
- Research Center for Anti‑Aging Technology Development, Pusan National University, Busan 609‑735, Republic of Korea
| | - Hae Young Chung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609‑735, Republic of Korea
| | - Nam Deuk Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609‑735, Republic of Korea
| |
Collapse
|
42
|
Mok GF, Cardenas R, Anderton H, Campbell KHS, Sweetman D. Interactions between FGF18 and retinoic acid regulate differentiation of chick embryo limb myoblasts. Dev Biol 2014; 396:214-23. [PMID: 25446536 DOI: 10.1016/j.ydbio.2014.10.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 09/25/2014] [Accepted: 10/10/2014] [Indexed: 10/24/2022]
Abstract
During limb development Pax3 positive myoblasts delaminate from the hypaxial dermomyotome of limb level somites and migrate into the limb bud where they form the dorsal and ventral muscle masses. Only then do they begin to differentiate and express markers of myogenic commitment and determination such as Myf5 and MyoD. However the signals regulating this process remain poorly characterised. We show that FGF18, which is expressed in the distal mesenchyme of the limb bud, induces premature expression of both Myf5 and MyoD and that blocking FGF signalling also inhibits endogenous MyoD expression. This expression is mediated by ERK MAP kinase but not PI3K signalling. We also show that retinoic acid (RA) can inhibit the myogenic activity of FGF18 and that blocking RA signalling allows premature induction of MyoD by FGF18 at HH19. We propose a model where interactions between FGF18 in the distal limb and retinoic acid in the proximal limb regulate the timing of myogenic gene expression during limb bud development.
Collapse
Affiliation(s)
- Gi Fay Mok
- Division of Animal Sciences, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Sutton Bonington LE12 5RD, UK
| | - Ryan Cardenas
- Division of Animal Sciences, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Sutton Bonington LE12 5RD, UK
| | - Helen Anderton
- Division of Animal Sciences, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Sutton Bonington LE12 5RD, UK
| | - Keith H S Campbell
- Division of Animal Sciences, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Sutton Bonington LE12 5RD, UK
| | - Dylan Sweetman
- Division of Animal Sciences, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Sutton Bonington LE12 5RD, UK.
| |
Collapse
|
43
|
Ge X, Zhang Y, Park S, Cong X, Gerrard DE, Jiang H. Stac3 inhibits myoblast differentiation into myotubes. PLoS One 2014; 9:e95926. [PMID: 24788338 PMCID: PMC4005754 DOI: 10.1371/journal.pone.0095926] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 04/01/2014] [Indexed: 12/19/2022] Open
Abstract
The functionally undefined Stac3 gene, predicted to encode a SH3 domain- and C1 domain-containing protein, was recently found to be specifically expressed in skeletal muscle and essential to normal skeletal muscle development and contraction. In this study we determined the potential role of Stac3 in myoblast proliferation and differentiation, two important steps of muscle development. Neither siRNA-mediated Stac3 knockdown nor plasmid-mediated Stac3 overexpression affected the proliferation of C2C12 myoblasts. Stac3 knockdown promoted the differentiation of C2C12 myoblasts into myotubes as evidenced by increased fusion index, increased number of nuclei per myotube, and increased mRNA and protein expression of myogenic markers including myogenin and myosin heavy chain. In contrast, Stac3 overexpression inhibited the differentiation of C2C12 myoblasts into myotubes as evidenced by decreased fusion index, decreased number of nuclei per myotube, and decreased mRNA and protein expression of myogenic markers. Compared to wild-type myoblasts, myoblasts from Stac3 knockout mouse embryos showed accelerated differentiation into myotubes in culture as evidenced by increased fusion index, increased number of nuclei per myotube, and increased mRNA expression of myogenic markers. Collectively, these data suggest an inhibitory role of endogenous Stac3 in myoblast differentiation. Myogenesis is a tightly controlled program; myofibers formed from prematurely differentiated myoblasts are dysfunctional. Thus, Stac3 may play a role in preventing precocious myoblast differentiation during skeletal muscle development.
Collapse
Affiliation(s)
- Xiaomei Ge
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Yafei Zhang
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Sungwon Park
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Xiaofei Cong
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, Virginia, United States of America
| | - David E. Gerrard
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Honglin Jiang
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, Virginia, United States of America
- * E-mail:
| |
Collapse
|
44
|
Zhang H, Anderson JE. Satellite cell activation and populations on single muscle-fiber cultures from adult zebrafish (Danio rerio). ACTA ACUST UNITED AC 2014; 217:1910-7. [PMID: 24577448 DOI: 10.1242/jeb.102210] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Satellite cells (SCs), stem cells in skeletal muscle, are mitotically quiescent in adult mammals until activated for growth or regeneration. In mouse muscle, SCs are activated by nitric oxide (NO), hepatocyte growth factor (HGF) and the mechanically induced NO-HGF signaling cascade. Here, the SC population on fibers from the adult, ectothermic zebrafish and SC responsiveness to activating stimuli were assessed using the model system of isolated fibers cultured at 27 and 21°C. SCs were identified by immunostaining for the HGF receptor, c-met, and activation was determined using bromodeoxyuridine uptake in culture or in vivo. In dose-response studies, SC activation was increased by treatment with the NO-donor drug isosorbide dinitrate (1 mmol l(-1)) or HGF (10 ng ml(-1)) to maximum activation at lower concentrations of both than in previous studies of mouse fibers. HGF-induced activation was blocked by anti-c-met antibody, and reduced by culture at 21°C. The effect of cyclical stretch (3 h at 4 cycles per minute) increased activation and was blocked by nitric oxide synthase inhibition and reduced by culture at 21°C. The number of c-met+ SCs per fiber increased rapidly (by 3 h) after stretching. The character of signaling in SC activation on zebrafish fibers, in particular temperature-dependent responses to HGF and stretch, gives new insights into the influence of ectothermy on regulation of muscle growth in teleosts and suggests the use of the single-fiber model system to explore the basis of fiber hyperplasia and the conservation of regulatory pathways between species.
Collapse
Affiliation(s)
- Helia Zhang
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, Canada R3T 2N2
| | - Judy E Anderson
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, Canada R3T 2N2
| |
Collapse
|
45
|
Montesano A, Luzi L, Senesi P, Mazzocchi N, Terruzzi I. Resveratrol promotes myogenesis and hypertrophy in murine myoblasts. J Transl Med 2013; 11:310. [PMID: 24330398 PMCID: PMC3867424 DOI: 10.1186/1479-5876-11-310] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 12/05/2013] [Indexed: 12/30/2022] Open
Abstract
Background Nutrigenomics elucidate the ability of bioactive food components to influence gene expression, protein synthesis, degradation and post-translational modifications. Resveratrol (RSV), natural polyphenol found in grapes and in other fruits, has a plethora of health benefits in a variety of human diseases: cardio- and neuroprotection, immune regulation, cancer chemoprevention, DNA repair, prevention of mitochondrial disorder, avoidance of obesity-related diseases. In skeletal muscle, RSV acts on protein catabolism and muscle function, conferring resistance against oxidative stress, injury and cell death, but its action mechanisms and protein targets in myogenesis process are not completely known. Myogenesis is a dynamic multistep process regulated by Myogenic Regulator Factors (MRFs), responsible of the commitment of myogenic cell into skeletal muscle: mononucleated undifferentiated myoblasts break free from cell cycle, elongate and fuse to form multinucleated myotubes. Skeletal muscle hypertrophy can be defined as a result of an increase in the size of pre-existing skeletal muscle fibers accompanied by increased protein synthesis, mainly regulated by Insulin Like Growth Factor 1 (IGF-1), PI3-K/AKT signaling pathways. Aim of this work was the study of RSV effects on proliferation, differentiation process and hypertrophy in C2C12 murine cells. Methods To study proliferative phase, cells were incubated in growth medium with/without RSV (0.1 or 25 μM) until reaching sub confluence condition (24, 48, 72 h). To examine differentiation, at 70% confluence, cells were transferred in differentiation medium both with/without RSV (0.1 or 25 μM) for 24, 48, 72, 96 hours. After 72 hours of differentiation, the genesis of hypertrophy in neo-formed myotubes was analyzed. Results Data showed that RSV regulates cell cycle exit and induces C2C12 muscle differentiation. Furthermore, RSV might control MRFs and muscle-specific proteins synthesis. In late differentiation, RSV has positive effects on hypertrophy: RSV stimulates IGF-1 signaling pathway, in particular AKT and ERK 1/2 protein activation, AMPK protein level and induces hypertrophic morphological changes in neo-formed myotubes modulating cytoskeletal proteins expression. Conclusions RSV might control cell cycle promoting myogenesis and hypertrophy in vitro, opening a novel field of application of RSV in clinical conditions characterized by chronic functional and morphological muscle impairment.
Collapse
Affiliation(s)
| | | | | | | | - Ileana Terruzzi
- Division of Metabolic and Cardiovascular Sciences, Metabolism, Nutrigenomics and Cellular Differentiation Unit, DIBIT-San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
46
|
Venhoranta H, Bauersachs S, Taponen J, Lohi H, Taira T, Andersson M, Kind A, Schnieke A, Flisikowski K. Fetal growth restriction caused by MIMT1 deletion alters brain transcriptome in cattle. Int J Dev Neurosci 2013; 31:463-7. [PMID: 23726833 DOI: 10.1016/j.ijdevneu.2013.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 04/25/2013] [Accepted: 05/13/2013] [Indexed: 02/07/2023] Open
Abstract
We examined levels of gene expression in the brains of bovine fetuses carrying a truncated MIMT1 allele, MIMT1(Del), shown to cause late abortion and stillbirth as a result of fetal growth restriction. MIMT1 is a non-protein coding gene that forms part of the imprinted PEG3 (paternally expressed gene 3) domain. Microarray analysis of brain cortex samples from mid-gestation MIMT1(Del/WT) bovine fetuses and wild-type siblings was performed to study the effect of fetal growth restriction on brain gene expression. Statistical analysis revealed 134 genes with increased mRNA levels and 22 with reduced levels in MIMT1(Del/WT) fetuses. Gene set enrichment analysis identified a relatively small number of significant functional clusters representing three major biological processes: response to oxidative stress, angiogenesis, and epithelial cell proliferation. Gene expression microarray analyses identified increased expression of VIPR2, HTRA1, S100A4 and MYH8 in fetuses carrying the deletion and decreased expression of DRD2, ADAM18, miR345, ZNF585A. ADAM18, DRD2 and S100A4 are known to be involved in prenatal brain development. ZNF585A, miR-345, VIPR2, HTRA1, and MYH8 are known to be involved in cell growth and differentiation, but any role in neural developmental has yet to be elucidated.
Collapse
Affiliation(s)
- Heli Venhoranta
- Department of Production Animal Medicine, University of Helsinki, Saarentaus, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Obesity represents a major risk factor for the development of several of our most common medical conditions, including Type 2 diabetes, dyslipidaemia, non-alcoholic fatty liver, cardiovascular disease and even some cancers. Although increased fat mass is the main feature of obesity, not all fat depots are created equal. Adipocytes found in white adipose tissue contain a single large lipid droplet and play well-known roles in energy storage. By contrast, brown adipose tissue is specialized for thermogenic energy expenditure. Owing to its significant capacity to dissipate energy and regulate triacylglycerol (triglyceride) and glucose metabolism, and its demonstrated presence in adult humans, brown fat could be a potential target for the treatment of obesity and metabolic syndrome. Undoubtedly, fundamental knowledge about the formation of brown fat and regulation of its activity is imperatively needed to make such therapeutics possible. In the present review, we integrate the recent advancements on the regulation of brown fat formation and activity by developmental and hormonal signals in relation to its metabolic function.
Collapse
|
48
|
Abstract
Adult skeletal muscle in mammals is a stable tissue under normal circumstances but has remarkable ability to repair after injury. Skeletal muscle regeneration is a highly orchestrated process involving the activation of various cellular and molecular responses. As skeletal muscle stem cells, satellite cells play an indispensible role in this process. The self-renewing proliferation of satellite cells not only maintains the stem cell population but also provides numerous myogenic cells, which proliferate, differentiate, fuse, and lead to new myofiber formation and reconstitution of a functional contractile apparatus. The complex behavior of satellite cells during skeletal muscle regeneration is tightly regulated through the dynamic interplay between intrinsic factors within satellite cells and extrinsic factors constituting the muscle stem cell niche/microenvironment. For the last half century, the advance of molecular biology, cell biology, and genetics has greatly improved our understanding of skeletal muscle biology. Here, we review some recent advances, with focuses on functions of satellite cells and their niche during the process of skeletal muscle regeneration.
Collapse
Affiliation(s)
- Hang Yin
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | | | | |
Collapse
|
49
|
Borensztein M, Monnier P, Court F, Louault Y, Ripoche MA, Tiret L, Yao Z, Tapscott SJ, Forné T, Montarras D, Dandolo L. Myod and H19-Igf2 locus interactions are required for diaphragm formation in the mouse. Development 2013; 140:1231-9. [PMID: 23406902 DOI: 10.1242/dev.084665] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The myogenic regulatory factor Myod and insulin-like growth factor 2 (Igf2) have been shown to interact in vitro during myogenic differentiation. In order to understand how they interact in vivo, we produced double-mutant mice lacking both the Myod and Igf2 genes. Surprisingly, these mice display neonatal lethality due to severe diaphragm atrophy. Alteration of diaphragm muscle development occurs as early as 15.5 days post-coitum in the double-mutant embryos and leads to a defect in the terminal differentiation of muscle progenitor cells. A negative-feedback loop was detected between Myod and Igf2 in embryonic muscles. Igf2 belongs to the imprinted H19-Igf2 locus. Molecular analyses show binding of Myod on a mesodermal enhancer (CS9) of the H19 gene. Chromatin conformation capture experiments reveal direct interaction of CS9 with the H19 promoter, leading to increased H19 expression in the presence of Myod. In turn, the non-coding H19 RNA represses Igf2 expression in trans. In addition, Igf2 also negatively regulates Myod expression, possibly by reducing the expression of the Srf transcription factor, a known Myod activator. In conclusion, Igf2 and Myod are tightly co-regulated in skeletal muscles and act in parallel pathways in the diaphragm, where they affect the progression of myogenic differentiation. Igf2 is therefore an essential player in the formation of a functional diaphragm in the absence of Myod.
Collapse
Affiliation(s)
- Maud Borensztein
- Genetics and Development Department, Inserm U1016, CNRS UMR 8104, University of Paris Descartes, Institut Cochin, 75014 Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Anderson C, Williams VC, Moyon B, Daubas P, Tajbakhsh S, Buckingham ME, Shiroishi T, Hughes SM, Borycki AG. Sonic hedgehog acts cell-autonomously on muscle precursor cells to generate limb muscle diversity. Genes Dev 2012; 26:2103-17. [PMID: 22987640 DOI: 10.1101/gad.187807.112] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
How muscle diversity is generated in the vertebrate body is poorly understood. In the limb, dorsal and ventral muscle masses constitute the first myogenic diversification, as each gives rise to distinct muscles. Myogenesis initiates after muscle precursor cells (MPCs) have migrated from the somites to the limb bud and populated the prospective muscle masses. Here, we show that Sonic hedgehog (Shh) from the zone of polarizing activity (ZPA) drives myogenesis specifically within the ventral muscle mass. Shh directly induces ventral MPCs to initiate Myf5 transcription and myogenesis through essential Gli-binding sites located in the Myf5 limb enhancer. In the absence of Shh signaling, myogenesis is delayed, MPCs fail to migrate distally, and ventral paw muscles fail to form. Thus, Shh production in the limb ZPA is essential for the spatiotemporal control of myogenesis and coordinates muscle and skeletal development by acting directly to regulate the formation of specific ventral muscles.
Collapse
Affiliation(s)
- Claire Anderson
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|