1
|
Fatemi SA, Levy AW, Peebles ED. The Expressions of the Immunity- and Muscle Development-Related Genes of 40-Day-Old Broilers Are Promoted in Response to the In Ovo and Dietary Supplemental Administration of Calcidiol in Conjunction with the In Ovo Administration of Marek's Disease Vaccine. Animals (Basel) 2024; 15:10. [PMID: 39794953 PMCID: PMC11718904 DOI: 10.3390/ani15010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/12/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
Effects of in ovo and dietary sources of calcidiol (25(OH)D3), combined with Marek's disease vaccine (MDV), on the expression of genes involved with the antioxidant activity, muscle deposition, and immunity in the pectoralis major (P. major) muscle and spleen of 40 d of age (doa) broilers were investigated. The in ovo treatments were as follows: (1) non-injected; (2) the injection of 50 μL of commercial MDV, (3) MDV + 1.2, or (4) 2.4 μg of 25(OH)D3. All birds received either a commercial diet containing no supplemental 25(OH)D3 (control) or the same diet supplemented with an additional 69 µg of 25(OH)D3 per kg of feed (Hy-D diet). At 40 doa, the pectoralis major (P. major) muscle and spleen of 48 birds (six replicates per diet x in ovo treatment combination) were collected. When compared to un-supplemented commercial diet-fed birds, in birds that were fed the Hy-D diet, the expression of the TGF-β4 gene in the spleen and P. major muscle, and the GSH-P1, GSH-P7, SOD2, MyoG, MyoD1, and Pax3 genes in the P. major muscle were up-regulated, whereas the expression of the IL-1β, IL-8, and CYP24A1 genes in the spleen and P. major muscle were down-regulated. Nevertheless, birds that received any of the in ovo injection doses of 25(OH)D3 exhibited a higher expression of the IL-10, TGF-β4, and CYP27B1 genes in the spleen and P. major muscle. Furthermore, in comparison to the MDV-injected control group, the CAT, MyoD1, and Pax3 genes in the P. major muscle were up-regulated, and the expression of the INF-γ, IL-1β and CYP24A1 genes in the spleen and the IL-8, and IL-1β genes in the P. major muscle were down-regulated. In conclusion, a significant improvement in the expression of genes responsible for enzymatic antioxidant activity, protein synthesis, and inflammatory reactions in 40-day-old broilers occurred in response to in ovo and dietary supplemental 25(OH)D3, and supplemental 25OHD3 provided via either route was used to enhance the expression of genes linked to vitamin D activity (CYP27B1, CYP24A1).
Collapse
|
2
|
Fatemi SA, Levy AW, Peebles ED. Enhancements in the expressions of genes associated with the immunity, muscle growth, and antioxidant activity of 14 d broilers in response to the in ovo injection of the Marek's disease vaccine alone or in conjunction with the in ovo and dietary supplemental administration of 25-hydroxycholecalciferol. Poult Sci 2024; 103:104372. [PMID: 39413703 PMCID: PMC11530893 DOI: 10.1016/j.psj.2024.104372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/13/2024] [Accepted: 09/25/2024] [Indexed: 10/18/2024] Open
Abstract
Influences the Marek's disease vaccine (MDV) alone or combined with the in ovo and dietary administration of 25-hydroxycholecalciferol (25OHD3) on the expression of genes associated with the breast muscle deposition, adaptive and innate immunity, and antioxidant and vitamin D activities of 14 d-old broilers were investigated. Four in ovo treatments were: noninjected; commercial MDV-alone-injected (50 μl); or 50 μl of MDV containing 1.2 (MDV+25OHD3-1.2) or 2.4 (MDV+25OHD3-2.4) μg of 25OHD3. Two dietary treatments were a commercial diet containing 25OHD3 (250 IU)/kg of feed (control) or the same diet supplemented with additional 25OHD3 (2,760 IU)/kg of feed (Hy-D diet). One bird per pen (48 total) was sampled at 14 d for determination of the expression of genes involved with the muscle deposition (MyoD1, MyoG, Pax3, and Mrf4), immunity (INF-γ, IL-10, IL-8, IL-1β, and TGF-β4), antioxidant capacity (SOD1, SOD2, GSH-P1, GSH-P7, and CAT), and vitamin D activity (VDR, 1α-hydroxylase, and 24-hydroxylase) in the spleen and pectoralis major (P.major) muscle. The treatment differences were considered significant at P ≤ 0.05. In the P. major, Mrf4 and MyoG were up-regulated in Hy-D-fed birds. Also, the in ovo and dietary 25OHD3 sources individually increased SOD2 gene expression in the P. major. In the spleen, the expressions of IL-1β and IL-8 were down-regulated and IL-10 and TGF-β4 gene expressions were up-regulated in Hy-D-fed birds than those commercial-fed broiler. In ovo and dietary 25OHD3 sources enhanced vitamin D gene (1α-hydroxylase and 24-hydroxylase) activities in the breast and spleen. In ovo x dietary treatment interactions were significant for the MyoD1, IL-8, Pax3, TGF-β4 genes of the P. major, in which the combined MDV with 1.2 μg of 25OHD3 enhanced their expressions in birds fed the Hy-D diet. In conclusion, both 25OHD3 sources promoted the expression of genes associated with immunity and P. major growth. It is recommended that both 25OHD3 sources can be used to promote the gene expression of 14-day-old broilers in the spleen and breast muscle when MDV administered in ovo.
Collapse
Affiliation(s)
- S A Fatemi
- Department of Poultry Science, Mississippi State University, MS 39762, USA.
| | - A W Levy
- DSM Nutritional Products, Parsippany, NJ, 07054, USA
| | - E D Peebles
- Department of Poultry Science, Mississippi State University, MS 39762, USA
| |
Collapse
|
3
|
Salman MI, Khalil EG, Almzaien AK, Hadi AA, Ahmed AA, Shaker HK, Al-Shammari AM. Promoting and accelerating muscle regeneration through cell therapy in a mouse model. J Taibah Univ Med Sci 2024; 19:1011-1023. [PMID: 39484055 PMCID: PMC11526084 DOI: 10.1016/j.jtumed.2024.09.004] [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: 05/06/2023] [Revised: 07/27/2024] [Accepted: 09/13/2024] [Indexed: 11/03/2024] Open
Abstract
Objectives Skeletal muscle injuries and disorders are universal clinical challenges with direct and indirect mechanisms and notable residual effects, such as prolonged, intense pain and physical disability. Stem cells, an innovative tool for cell therapy for musculoskeletal disorders, specifically promote skeletal muscle regeneration. This study was aimed at investigating the use of mesenchymal stem cells (MSCs) and their differentiated myocytes as a cell-based therapy to promote regeneration in damaged or diseased skeletal muscle. Methods Bone marrow mesenchymal stem cells (BM-MSCs) were isolated from the bone marrow of adult mice and grown in tissue culture flasks. The BM-MSCs were positive for CD90 and CD105, and negative for CD45 and CD34. These cells were induced with specific differentiation medium in vitro to differentiate into a skeletal muscle cell lineage over 7 days. Skeletal muscle differentiation was characterized according to morphology through hematoxylin and eosin staining, and scanning electron microscopy. Immunostaining for Myf-6, myosin heavy chain (MHC), and desmin-specific factors for skeletal muscle development-was performed to confirm skeletal muscle differentiation. An in vivo study in a muscle injury model was used to evaluate cell therapy based on naïve stem cells and differentiated myocytes. Results Cultured mouse BM-MSCS were positive for CD90 and CD105, and negative for CD45 and CD34. These cells developed into skeletal muscle with strong skeletal muscle differentiation potential, as confirmed by immunohistochemistry for the markers Myf6, MHC, and desmin. The differentiated myocytes showed better repair enhancement than undifferentiated stem cells after transplantations into a mouse model of skeletal muscle atrophy. Conclusions Myocytes derived from BM-MSCs may be incorporated into muscular atrophy treatment as a biological strategy for managing skeletal muscle diseases and injuries, thus advancing cell-based clinical treatments.
Collapse
Affiliation(s)
- Marwa I. Salman
- Biotechnology Department, College of Science, Baghdad University, Baghdad, Iraq
| | - Eman G. Khalil
- Biomedical Engineering Department, Engineering College, Al-Nahrain University, Baghdad, Iraq
| | - Aous K. Almzaien
- Experimental Therapy Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriyah University, Baghdad, Iraq
| | - Ali A. Hadi
- Department of Physical Education and Sports Sciences, Dijlah University College, Baghdad, Iraq
- Individual Sports Department, College of Physical Education and Sports Sciences, University of Baghdad, Iraq
| | - Aysar A. Ahmed
- Experimental Therapy Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriyah University, Baghdad, Iraq
| | - Hiba K. Shaker
- Experimental Therapy Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriyah University, Baghdad, Iraq
| | - Ahmed M. Al-Shammari
- Experimental Therapy Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriyah University, Baghdad, Iraq
| |
Collapse
|
4
|
Chand S, Tripathi AS, Dewani AP, Sheikh NWA. Molecular targets for management of diabetes: Remodelling of white adipose to brown adipose tissue. Life Sci 2024; 345:122607. [PMID: 38583857 DOI: 10.1016/j.lfs.2024.122607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/28/2024] [Accepted: 04/01/2024] [Indexed: 04/09/2024]
Abstract
Diabetes mellitus is a disorder characterised metabolic dysfunction that results in elevated glucose level in the bloodstream. Diabetes is of two types, type1 and type 2 diabetes. Obesity is considered as one of the major reasons intended for incidence of diabetes hence it turns out to be essential to study about the adipose tissue which is responsible for fat storage in body. Adipose tissues play significant role in maintaining the balance between energy stabilization and homeostasis. The three forms of adipose tissue are - White adipose tissue (WAT), Brown adipose tissue (BAT) and Beige adipose tissue (intermediate form). The amount of BAT gets reduced, and WAT starts to increase with the age. WAT when exposed to certain stimuli gets converted to BAT by the help of certain transcriptional regulators. The browning of WAT has been a matter of study to treat the metabolic disorders and to initiate the expenditure of energy. The three main regulators responsible for the browning of WAT are PRDM16, PPARγ and PGC-1α via various cellular and molecular mechanism. Presented review article includes the detailed elaborative aspect of genes and proteins involved in conversion of WAT to BAT.
Collapse
Affiliation(s)
- Shushmita Chand
- Amity Institute of Pharmacy, Amity University, Sector 125, Noida, Uttar Pradesh, India
| | - Alok Shiomurti Tripathi
- Department of Pharmacology, ERA College of Pharmacy, ERA University, Lucknow, Uttar Pradesh, India.
| | - Anil P Dewani
- Department of Pharmacology, P. Wadhwani College of Pharmacy, Yavatmal, Maharashtra, India
| | | |
Collapse
|
5
|
Fatemi SA, Mousstaaid A, Williams CJ, Deines J, Poudel S, Poudel I, Elliott KEC, Walters ER, Forcier N, Peebles ED. In ovo administration of the Marek's disease vaccine in conjunction with 25-hydroxyvitamin D 3 and its subsequent effects on the performance and immunity-related characteristics of Ross 708 broiler hatchlings 1,2,3. Poult Sci 2024; 103:103199. [PMID: 37939590 PMCID: PMC10665917 DOI: 10.1016/j.psj.2023.103199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 11/10/2023] Open
Abstract
The combined effects of the in ovo injection of commercial Marek's disease vaccine (MDV) and various levels of 25-hydroxyvitamin D3 (25OHD3) on the hatch variables, immunological measurements, and gene expression of Ross 708 hatchling broilers were investigated. A total of 5 in ovo injection treatments that were applied at 18 d of incubation (doi) included: 1) noninjected (control); or a 50 μL solution volume of 2) MDV alone; or MDV combined with 3) 0.6 μg of 25OHD3; 4) 1.2 μg of 25OHD3; or 5) 2.4 μg of 25OHD3. At hatch, hatchability of set and live embryonated eggs, hatchling body weight, hatch residue analysis, serum IgY and alpha-1 acid glycoprotein (AGP) concentrations, and the expression of genes related to immunity (INFα, INFβ, INFγ, TLR-3, and TLR-21) and vitamin D3 activity (1 α-hydroxylase, 24 hydroxylase, and vitamin D receptor) were determined. No significant treatment differences were observed for hatchability of set and live embryonated eggs, or for serum IgY and AGP concentrations. However, hatchling body weight was higher when MDV was combined with either 1.2 or 2.4 μg of 25OHD3 than when MDV was provided alone or in combination with 0.6 μg of 25OHD3. Also, in comparison to the noninjected treatment group, the expression of the genes for 1 α-hydroxylase and 24 hydroxylase was improved when MDV was combined with either 1.2 or 2.4 μg of 25OHD3. Lastly, expression of the genes linked to viral detection (TLR-3) and antibody production (INF-β) was increased in those treatments that contained any level of 25OHD3. These results indicate that in comparison to controls, the effects of MDV were observed to be greater on hatchling BW and splenic gene expression when it was administered in combination with the 1.2 or 2.4 μg doses of 25OHD3. Further research is needed to determine the posthatch effects of the administration of various levels of 25OHD3 in combination with MDV.
Collapse
Affiliation(s)
- S A Fatemi
- Department of Poultry Science, Mississippi State University, Mississippi State, MS 39762, USA.
| | - A Mousstaaid
- Department of Poultry Science, Mississippi State University, Mississippi State, MS 39762, USA
| | - C J Williams
- Zoetis Animal Health, Research Triangle Park, NC 27703, USA
| | - J Deines
- Zoetis Animal Health, Research Triangle Park, NC 27703, USA
| | - S Poudel
- Department of Poultry Science, Mississippi State University, Mississippi State, MS 39762, USA
| | - I Poudel
- Department of Poultry Science, Mississippi State University, Mississippi State, MS 39762, USA
| | - K E C Elliott
- Poultry Research Unit, USDA-ARS, Starkville, MS 39762, USA
| | - E R Walters
- Department of Poultry Science, Mississippi State University, Mississippi State, MS 39762, USA
| | - N Forcier
- Department of Poultry Science, Mississippi State University, Mississippi State, MS 39762, USA
| | - E D Peebles
- Department of Poultry Science, Mississippi State University, Mississippi State, MS 39762, USA
| |
Collapse
|
6
|
Papanikolaou NA, Nikolaidis M, Amoutzias GD, Fouza A, Papaioannou M, Pandey A, Papavassiliou AG. The Dynamic and Crucial Role of the Arginine Methylproteome in Myoblast Cell Differentiation. Int J Mol Sci 2023; 24:2124. [PMID: 36768448 PMCID: PMC9916730 DOI: 10.3390/ijms24032124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/14/2023] [Accepted: 01/17/2023] [Indexed: 01/25/2023] Open
Abstract
Protein arginine methylation is an extensive and functionally significant post-translational modification. However, little is known about its role in differentiation at the systems level. Using stable isotope labeling by amino acids in cell culture (SILAC) proteomics of whole proteome analysis in proliferating or five-day differentiated mouse C2C12 myoblasts, followed by high-resolution mass spectrometry, biochemical assays, and specific immunoprecipitation of mono- or dimethylated arginine peptides, we identified several protein families that were differentially methylated on arginine. Our study is the first to reveal global changes in the arginine mono- or dimethylation of proteins in proliferating myoblasts and differentiated myocytes and to identify enriched protein domains and novel short linear motifs (SLiMs). Our data may be crucial for dissecting the links between differentiation and cancer growth.
Collapse
Affiliation(s)
- Nikolaos A. Papanikolaou
- Laboratory of Biological Chemistry, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece
| | - Marios Nikolaidis
- Bioinformatics Laboratory, Department of Biochemistry and Biotechnology, University of Thessaly, 41500 Larisa, Greece
| | - Grigorios D. Amoutzias
- Bioinformatics Laboratory, Department of Biochemistry and Biotechnology, University of Thessaly, 41500 Larisa, Greece
| | - Ariadni Fouza
- Fifth Surgical Department, Ippokrateio General Hospital, School of Medicine, Aristotle University of Thessaloniki, 54643 Thessaloniki, Macedonia, Greece
| | - Maria Papaioannou
- Laboratory of Biological Chemistry, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
7
|
Hughes SM, Escaleira RC, Wanders K, Koth J, Wilkinson DG, Xu Q. Clonal behaviour of myogenic precursor cells throughout the vertebrate lifespan. Biol Open 2022; 11:276275. [PMID: 35972050 PMCID: PMC9399818 DOI: 10.1242/bio.059476] [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: 06/16/2022] [Accepted: 07/11/2022] [Indexed: 11/20/2022] Open
Abstract
To address questions of stem cell diversity during skeletal myogenesis, a Brainbow-like genetic cell lineage tracing method, dubbed Musclebow2, was derived by enhancer trapping in zebrafish. It is shown that, after initial formation of the primary myotome, at least 15 muscle precursor cells (mpcs) seed each somite, where they proliferate but contribute little to muscle growth prior to hatching. Thereafter, dermomyotome-derived mpc clones rapidly expand while some progeny undergo terminal differentiation, leading to stochastic clonal drift within the mpc pool. No evidence of cell-lineage-based clonal fate diversity was obtained. Neither fibre nor mpc death was observed in uninjured animals. Individual marked muscle fibres persist across much of the lifespan indicating low rates of nuclear turnover. In adulthood, early-marked mpc clones label stable blocks of tissue comprising a significant fraction of either epaxial or hypaxial somite. Fusion of cells from separate early-marked clones occurs in regions of clone overlap. Wounds are regenerated from several local mpcs; no evidence for specialised stem mpcs was obtained. In conclusion, our data indicate that most mpcs in muscle tissue contribute to local growth and repair and suggest that cellular turnover is low in the absence of trauma.
Collapse
Affiliation(s)
- Simon M Hughes
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, UK
| | - Roberta C Escaleira
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, UK
| | - Kees Wanders
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, UK
| | - Jana Koth
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, UK
| | | | - Qiling Xu
- Francis Crick Institute, London NW1 1AT, UK
| |
Collapse
|
8
|
Guo R, You X, Meng K, Sha R, Wang Z, Yuan N, Peng Q, Li Z, Xie Z, Chen R, Feng Y. Single-Cell RNA Sequencing Reveals Heterogeneity of Myf5-Derived Cells and Altered Myogenic Fate in the Absence of SRSF2. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105775. [PMID: 35460187 PMCID: PMC9218650 DOI: 10.1002/advs.202105775] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/05/2022] [Indexed: 06/14/2023]
Abstract
Splicing factor SRSF2 acts as a critical regulator for cell survival, however, it remains unknown whether SRSF2 is involved in myoblast proliferation and myogenesis. Here, knockdown of SRSF2 in myoblasts causes high rates of apoptosis and defective differentiation. Combined conditional knockout and lineage tracing approaches show that Myf5-cre mice lacking SRSF2 die immediately at birth and exhibit a complete absence of mature myofibers. Mutant Myf5-derived cells (tdtomato-positive cells) are randomly scattered in the myogenic and non-myogenic regions, indicating loss of the community effect required for skeletal muscle differentiation. Single-cell RNA-sequencing reveals high heterogeneity of myf5-derived cells and non-myogenic cells are significantly increased at the expense of skeletal muscle cells in the absence of SRSF2, reflecting altered cell fate. SRSF2 is demonstrated to regulate the entry of Myf5 cells into the myogenic program and ensures their survival by preventing precocious differentiation and apoptosis. In summary, SRSF2 functions as an essential regulator for Myf5-derived cells to respond correctly to positional cues and to adopt their myogenic fate.
Collapse
Affiliation(s)
- Ruochen Guo
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong ProvinceJining Medical UniversityJining272067P. R. China
| | - Xue You
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong ProvinceJining Medical UniversityJining272067P. R. China
| | - Kai Meng
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong ProvinceJining Medical UniversityJining272067P. R. China
| | - Rula Sha
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Zhenzhen Wang
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Ningyang Yuan
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Qian Peng
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Zhigang Li
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Zhiqin Xie
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Ruijiao Chen
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong ProvinceJining Medical UniversityJining272067P. R. China
| | - Ying Feng
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong ProvinceJining Medical UniversityJining272067P. R. China
| |
Collapse
|
9
|
Grimaldi A, Comai G, Mella S, Tajbakhsh S. Identification of bipotent progenitors that give rise to myogenic and connective tissues in mouse. eLife 2022; 11:70235. [PMID: 35225230 PMCID: PMC9020825 DOI: 10.7554/elife.70235] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 02/25/2022] [Indexed: 11/19/2022] Open
Abstract
How distinct cell fates are manifested by direct lineage ancestry from bipotent progenitors, or by specification of individual cell types is a key question for understanding the emergence of tissues. The interplay between skeletal muscle progenitors and associated connective tissue cells provides a model for examining how muscle functional units are established. Most craniofacial structures originate from the vertebrate-specific neural crest cells except in the dorsal portion of the head, where they arise from cranial mesoderm. Here, using multiple lineage-tracing strategies combined with single cell RNAseq and in situ analyses, we identify bipotent progenitors expressing Myf5 (an upstream regulator of myogenic fate) that give rise to both muscle and juxtaposed connective tissue. Following this bifurcation, muscle and connective tissue cells retain complementary signalling features and maintain spatial proximity. Disrupting myogenic identity shifts muscle progenitors to a connective tissue fate. The emergence of Myf5-derived connective tissue is associated with the activity of several transcription factors, including Foxp2. Interestingly, this unexpected bifurcation in cell fate was not observed in craniofacial regions that are colonised by neural crest cells. Therefore, we propose that an ancestral bi-fated program gives rise to muscle and connective tissue cells in skeletal muscles that are deprived of neural crest cells.
Collapse
Affiliation(s)
| | - Glenda Comai
- UMR 3738, Department of Developmental and Stem Cell Biology, CNRS, Paris, France
| | - Sebastien Mella
- Cytometry and Biomarkers UTechS, Institut Pasteur, Paris, France
| | | |
Collapse
|
10
|
Della Gaspera B, Weill L, Chanoine C. Evolution of Somite Compartmentalization: A View From Xenopus. Front Cell Dev Biol 2022; 9:790847. [PMID: 35111756 PMCID: PMC8802780 DOI: 10.3389/fcell.2021.790847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/26/2021] [Indexed: 11/13/2022] Open
Abstract
Somites are transitory metameric structures at the basis of the axial organization of vertebrate musculoskeletal system. During evolution, somites appear in the chordate phylum and compartmentalize mainly into the dermomyotome, the myotome, and the sclerotome in vertebrates. In this review, we summarized the existing literature about somite compartmentalization in Xenopus and compared it with other anamniote and amniote vertebrates. We also present and discuss a model that describes the evolutionary history of somite compartmentalization from ancestral chordates to amniote vertebrates. We propose that the ancestral organization of chordate somite, subdivided into a lateral compartment of multipotent somitic cells (MSCs) and a medial primitive myotome, evolves through two major transitions. From ancestral chordates to vertebrates, the cell potency of MSCs may have evolved and gave rise to all new vertebrate compartments, i.e., the dermomyome, its hypaxial region, and the sclerotome. From anamniote to amniote vertebrates, the lateral MSC territory may expand to the whole somite at the expense of primitive myotome and may probably facilitate sclerotome formation. We propose that successive modifications of the cell potency of some type of embryonic progenitors could be one of major processes of the vertebrate evolution.
Collapse
|
11
|
Bjorke B, Weller KG, Jones LE, Robinson GE, Vesser M, Chen L, Gage PJ, Gould TW, Mastick GS. Oculomotor nerve guidance and terminal branching requires interactions with differentiating extraocular muscles. Dev Biol 2021; 476:272-281. [PMID: 33905720 PMCID: PMC8284410 DOI: 10.1016/j.ydbio.2021.04.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/20/2021] [Accepted: 04/20/2021] [Indexed: 11/25/2022]
Abstract
Muscle function is dependent on innervation by the correct motor nerves. Motor nerves are composed of motor axons which extend through peripheral tissues as a compact bundle, then diverge to create terminal nerve branches to specific muscle targets. As motor nerves approach their targets, they undergo a transition where the fasciculated nerve halts further growth then after a pause, the nerve later initiates branching to muscles. This transition point is potentially an intermediate target or guidepost to present specific cellular and molecular signals for navigation. Here we describe the navigation of the oculomotor nerve and its association with developing muscles in mouse embryos. We found that the oculomotor nerve initially grew to the eye three days prior to the appearance of any extraocular muscles. The oculomotor axons spread to form a plexus within a mass of cells, which included precursors of extraocular muscles and other orbital tissues and expressed the transcription factor Pitx2. The nerve growth paused in the plexus for more than two days, persisting during primary extraocular myogenesis, with a subsequent phase in which the nerve branched out to specific muscles. To test the functional significance of the nerve contact with Pitx2+ cells in the plexus, we used two strategies to genetically ablate Pitx2+ cells or muscle precursors early in nerve development. The first strategy used Myf5-Cre-mediated expression of diphtheria toxin A to ablate muscle precursors, leading to loss of extraocular muscles. The oculomotor axons navigated to the eye to form the main nerve, but subsequently largely failed to initiate terminal branches. The second strategy studied Pitx2 homozygous mutants, which have early apoptosis of Pitx2-expressing precursor cells, including precursors for extraocular muscles and other orbital tissues. Oculomotor nerve fibers also grew to the eye, but failed to stop to form the plexus, instead grew long ectopic projections. These results show that neither Pitx2 function nor Myf5-expressing cells are required for oculomotor nerve navigation to the eye. However, Pitx2 function is required for oculomotor axons to pause growth in the plexus, while Myf5-expressing cells are required for terminal branch initiation.
Collapse
Affiliation(s)
- Brielle Bjorke
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | | | - Lauren E Jones
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | - G Eric Robinson
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | - Michelle Vesser
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | - Lisheng Chen
- Department of Ophthalmology & Visual Science, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Philip J Gage
- Department of Ophthalmology & Visual Science, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Thomas W Gould
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, United States
| | - Grant S Mastick
- Department of Biology, University of Nevada, Reno, NV, 89557, USA.
| |
Collapse
|
12
|
Rauch A, Mandrup S. Transcriptional networks controlling stromal cell differentiation. Nat Rev Mol Cell Biol 2021; 22:465-482. [PMID: 33837369 DOI: 10.1038/s41580-021-00357-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2021] [Indexed: 02/02/2023]
Abstract
Stromal progenitors are found in many different tissues, where they play an important role in the maintenance of tissue homeostasis owing to their ability to differentiate into parenchymal cells. These progenitor cells are differentially pre-programmed by their tissue microenvironment but, when cultured and stimulated in vitro, these cells - commonly referred to as mesenchymal stromal cells (MSCs) - exhibit a marked plasticity to differentiate into many different cell lineages. Loss-of-function studies in vitro and in vivo have uncovered the involvement of specific signalling pathways and key transcriptional regulators that work in a sequential and coordinated fashion to activate lineage-selective gene programmes. Recent advances in omics and single-cell technologies have made it possible to obtain system-wide insights into the gene regulatory networks that drive lineage determination and cell differentiation. These insights have important implications for the understanding of cell differentiation, the contribution of stromal cells to human disease and for the development of cell-based therapeutic applications.
Collapse
Affiliation(s)
- Alexander Rauch
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark. .,Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark.
| | - Susanne Mandrup
- Center for Functional Genomics and Tissue Plasticity, Functional Genomics & Metabolism Research Unit, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
13
|
Hara A, Kato K, Ishihara T, Kobayashi H, Asai N, Mii S, Shiraki Y, Miyai Y, Ando R, Mizutani Y, Iida T, Takefuji M, Murohara T, Takahashi M, Enomoto A. Meflin defines mesenchymal stem cells and/or their early progenitors with multilineage differentiation capacity. Genes Cells 2021; 26:495-512. [PMID: 33960573 PMCID: PMC8360184 DOI: 10.1111/gtc.12855] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are the likely precursors of multiple lines of mesenchymal cells. The existence of bona fide MSCs with self‐renewal capacity and differentiation potential into all mesenchymal lineages, however, has been unclear because of the lack of MSC‐specific marker(s) that are not expressed by the terminally differentiated progeny. Meflin, a glycosylphosphatidylinositol‐anchored protein, is an MSC marker candidate that is specifically expressed in rare stromal cells in all tissues. Our previous report showed that Meflin expression becomes down‐regulated in bone marrow‐derived MSCs cultured on plastic, making it difficult to examine the self‐renewal and differentiation of Meflin‐positive cells at the single‐cell level. Here, we traced the lineage of Meflin‐positive cells in postnatal and adult mice, showing that those cells differentiated into white and brown adipocytes, osteocytes, chondrocytes and skeletal myocytes. Interestingly, cells derived from Meflin‐positive cells formed clusters of differentiated cells, implying the in situ proliferation of Meflin‐positive cells or their lineage‐committed progenitors. These results, taken together with previous findings that Meflin expression in cultured MSCs was lost upon their multilineage differentiation, suggest that Meflin is a useful potential marker to localize MSCs and/or their immature progenitors in multiple tissues.
Collapse
Affiliation(s)
- Akitoshi Hara
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Katsuhiro Kato
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toshikazu Ishihara
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Kobayashi
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoya Asai
- Department of Pathology, Fujita Health University, Toyoake, Japan
| | - Shinji Mii
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukihiro Shiraki
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuki Miyai
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryota Ando
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuyuki Mizutani
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tadashi Iida
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mikito Takefuji
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahide Takahashi
- International Center for Cell and Gene Therapy, Fujita Health University, Toyoake, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
14
|
Kannan S, Lock I, Ozenberger BB, Jones KB. Genetic drivers and cells of origin in sarcomagenesis. J Pathol 2021; 254:474-493. [DOI: 10.1002/path.5617] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/01/2020] [Accepted: 01/06/2021] [Indexed: 02/06/2023]
Affiliation(s)
- Sarmishta Kannan
- Departments of Orthopaedics and Oncological Sciences Huntsman Cancer Institute, University of Utah School of Medicine Salt Lake City UT USA
| | - Ian Lock
- Departments of Orthopaedics and Oncological Sciences Huntsman Cancer Institute, University of Utah School of Medicine Salt Lake City UT USA
| | - Benjamin B Ozenberger
- Departments of Orthopaedics and Oncological Sciences Huntsman Cancer Institute, University of Utah School of Medicine Salt Lake City UT USA
| | - Kevin B Jones
- Departments of Orthopaedics and Oncological Sciences Huntsman Cancer Institute, University of Utah School of Medicine Salt Lake City UT USA
| |
Collapse
|
15
|
Wei X, Franke J, Ost M, Wardelmann K, Börno S, Timmermann B, Meierhofer D, Kleinridders A, Klaus S, Stricker S. Cell autonomous requirement of neurofibromin (Nf1) for postnatal muscle hypertrophic growth and metabolic homeostasis. J Cachexia Sarcopenia Muscle 2020; 11:1758-1778. [PMID: 33078583 PMCID: PMC7749575 DOI: 10.1002/jcsm.12632] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/09/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Neurofibromatosis type 1 (NF1) is a multi-organ disease caused by mutations in neurofibromin 1 (NF1). Amongst other features, NF1 patients frequently show reduced muscle mass and strength, impairing patients' mobility and increasing the risk of fall. The role of Nf1 in muscle and the cause for the NF1-associated myopathy are mostly unknown. METHODS To dissect the function of Nf1 in muscle, we created muscle-specific knockout mouse models for NF1, inactivating Nf1 in the prenatal myogenic lineage either under the Lbx1 promoter or under the Myf5 promoter. Mice were analysed during prenatal and postnatal myogenesis and muscle growth. RESULTS Nf1Lbx1 and Nf1Myf5 animals showed only mild defects in prenatal myogenesis. Nf1Lbx1 animals were perinatally lethal, while Nf1Myf5 animals survived only up to approximately 25 weeks. A comprehensive phenotypic characterization of Nf1Myf5 animals showed decreased postnatal growth, reduced muscle size, and fast fibre atrophy. Proteome and transcriptome analyses of muscle tissue indicated decreased protein synthesis and increased proteasomal degradation, and decreased glycolytic and increased oxidative activity in muscle tissue. High-resolution respirometry confirmed enhanced oxidative metabolism in Nf1Myf5 muscles, which was concomitant to a fibre type shift from type 2B to type 2A and type 1. Moreover, Nf1Myf5 muscles showed hallmarks of decreased activation of mTORC1 and increased expression of atrogenes. Remarkably, loss of Nf1 promoted a robust activation of AMPK with a gene expression profile indicative of increased fatty acid catabolism. Additionally, we observed a strong induction of genes encoding catabolic cytokines in muscle Nf1Myf5 animals, in line with a drastic reduction of white, but not brown adipose tissue. CONCLUSIONS Our results demonstrate a cell autonomous role for Nf1 in myogenic cells during postnatal muscle growth required for metabolic and proteostatic homeostasis. Furthermore, Nf1 deficiency in muscle drives cross-tissue communication and mobilization of lipid reserves.
Collapse
Affiliation(s)
- Xiaoyan Wei
- Musculoskeletal Development and Regeneration Group, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Development and Disease Group, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Julia Franke
- Musculoskeletal Development and Regeneration Group, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Development and Disease Group, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Mario Ost
- Department of Physiology of Energy Metabolism, German Institute for Human Nutrition, Nuthetal, Germany.,Department of Neuropathology, University Hospital Leipzig, Leipzig, Germany
| | - Kristina Wardelmann
- Junior Research Group Central Regulation of Metabolism, German Institute for Human Nutrition, Nuthetal, Germany.,Institute of Nutritional Science, Department of Molecular and Experimental Nutritional Medicine, University of Potsdam, Potsdam, Germany
| | - Stefan Börno
- Sequencing Core Unit, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Bernd Timmermann
- Sequencing Core Unit, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - David Meierhofer
- Mass Spectrometry Core Unit, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Andre Kleinridders
- Junior Research Group Central Regulation of Metabolism, German Institute for Human Nutrition, Nuthetal, Germany.,Institute of Nutritional Science, Department of Molecular and Experimental Nutritional Medicine, University of Potsdam, Potsdam, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Susanne Klaus
- Department of Physiology of Energy Metabolism, German Institute for Human Nutrition, Nuthetal, Germany.,Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Sigmar Stricker
- Musculoskeletal Development and Regeneration Group, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Development and Disease Group, Max Planck Institute for Molecular Genetics, Berlin, Germany
| |
Collapse
|
16
|
Wood WM, Otis C, Etemad S, Goldhamer DJ. Development and patterning of rib primordia are dependent on associated musculature. Dev Biol 2020; 468:133-145. [PMID: 32768399 PMCID: PMC7669625 DOI: 10.1016/j.ydbio.2020.07.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/20/2020] [Accepted: 07/29/2020] [Indexed: 01/29/2023]
Abstract
The importance of skeletal muscle for rib development and patterning in the mouse embryo has not been resolved, largely because different experimental approaches have yielded disparate results. In this study, we utilize both gene knockouts and muscle cell ablation approaches to re-visit the extent to which rib growth and patterning are dependent on developing musculature. Consistent with previous studies, we show that rib formation is highly dependent on the MYOD family of myogenic regulatory factors (MRFs), and demonstrate that the extent of rib formation is gene-, allele-, and dosage-dependent. In the absence of Myf5 and MyoD, one allele of Mrf4 is sufficient for extensive rib growth, although patterning is abnormal. Under conditions of limiting MRF dosage, MyoD is identified as a positive regulator of rib patterning, presumably due to improved intercostal muscle development. In contrast to previous muscle ablation studies, we show that diphtheria toxin subunit A (DTA)-mediated ablation of muscle progenitors or differentiated muscle, using MyoDiCre or HSA-Cre drivers, respectively, profoundly disrupts rib development. Further, a comparison of three independently derived Rosa26-based DTA knockin alleles demonstrates that the degree of rib perturbations in MyoDiCre/+/DTA embryos is markedly dependent on the DTA allele used, and may in part explain discrepancies with previous findings. The results support the conclusion that the extent and quality of rib formation is largely dependent on the dosage of Myf5 and Mrf4, and that both early myotome-sclerotome interactions, as well as later muscle-rib interactions, are important for proper rib growth and patterning.
Collapse
Affiliation(s)
- William M Wood
- Department of Molecular and Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT, USA
| | - Chelsea Otis
- Department of Molecular and Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT, USA
| | - Shervin Etemad
- Department of Molecular and Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT, USA
| | - David J Goldhamer
- Department of Molecular and Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
17
|
Su S, Wang Y, Chen C, Suh M, Azain M, Kim WK. Fatty Acid Composition and Regulatory Gene Expression in Late-Term Embryos of ACRB and COBB Broilers. Front Vet Sci 2020; 7:317. [PMID: 32671107 PMCID: PMC7330006 DOI: 10.3389/fvets.2020.00317] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/07/2020] [Indexed: 12/20/2022] Open
Abstract
Cobb broilers (COBB) have been heavily selected for their production performance in the past several decades, while the Athens Canadian Random Bred (ACRB) chickens, a meat-type breed, have been kept as a non-selected control strain. The purpose of this study was to compare these two lines of chickens at late embryonic development and identify the molecular markers and fatty acid profiles underlining their differences in growth performance due to selection. Fertilized eggs of the ACRB (n = 6) and COBB (n = 6) were used at 14 and 18 embryonic days. Genes involved in lipogenesis and myogenesis were measured using quantitative real-time reverse transcroption-polymerase chain reaction (RT-PCR), and fatty acid (FA) compositions of egg yolk, muscle, and liver were measured using gas chromatography. COBB had higher egg weight, embryo weight, and breast and fat ratio. The gene expression in the liver showed an interaction between age and breed on FASN expression, with the highest level in COBB at E18. ACRB had higher ApoB and MTTP expression, but lower SREBP-1 expression compared to COBB. No difference was found in myogenesis gene expression in the muscle between two breeds. For the FA composition, muscle was largely affected by both breed and age. Yolk and liver were affected mainly by breed and age, respectively. Constant interaction effects in docosahexaenoic acid (DHA), indicating the highest level in all the tested tissues of ACRB at E14 and the constant main effects with higher myristic, palmitic, and gondoic, but lower linolenic acid in the liver and yolk of COBB compared to the levels in those of ACRB. Finally, fat accumulation in the liver had no obvious difference between the breeds but was higher when embryo was older. In conclusion, broiler breed affects egg, embryo, and tissue weight, as well as FA composition in initial egg yolk and throughout the embryonic development. The highest docosahexaenoic percentage was observed in ACRB, indicating that genetic selection may result in fatty acid profile changes such as lower DHA content in chicken tissues and eggs.
Collapse
Affiliation(s)
- Shengchen Su
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| | - Yidi Wang
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada.,Division of Neurodegenerative Disorders & Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - Chongxiao Chen
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| | - Miyoung Suh
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada.,Division of Neurodegenerative Disorders & Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - Michael Azain
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, United States
| | - Woo Kyun Kim
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| |
Collapse
|
18
|
Jung SM, Sanchez-Gurmaches J, Guertin DA. Brown Adipose Tissue Development and Metabolism. Handb Exp Pharmacol 2019; 251:3-36. [PMID: 30203328 DOI: 10.1007/164_2018_168] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Brown adipose tissue is well known to be a thermoregulatory organ particularly important in small rodents and human infants, but it was only recently that its existence and significance to metabolic fitness in adult humans have been widely realized. The ability of active brown fat to expend high amounts of energy has raised interest in stimulating thermogenesis therapeutically to treat metabolic diseases related to obesity and type 2 diabetes. In parallel, there has been a surge of research aimed at understanding the biology of rodent and human brown fat development, its remarkable metabolic properties, and the phenomenon of white fat browning, in which white adipocytes can be converted into brown like adipocytes with similar thermogenic properties. Here, we review the current understanding of the developmental and metabolic pathways involved in forming thermogenic adipocytes, and highlight some of the many unknown functions of brown fat that make its study a rich and exciting area for future research.
Collapse
Affiliation(s)
- Su Myung Jung
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Joan Sanchez-Gurmaches
- Division of Endocrinology, Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, USA. .,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - David A Guertin
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA. .,Molecular, Cell and Cancer Biology Program, University of Massachusetts Medical School, Worcester, MA, USA. .,Lei Weibo Institute for Rare Diseases, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
19
|
Chen JN, Chen Y, Wei YY, Raza MA, Zou Q, Xi XY, Zhu L, Tang GQ, Jiang YZ, Li XW. Regulation of m6A RNA Methylation and Its Effect on Myogenic Differentiation in Murine Myoblasts. Mol Biol 2019. [DOI: 10.1134/s002689331903004x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
20
|
Rion N, Castets P, Lin S, Enderle L, Reinhard JR, Eickhorst C, Rüegg MA. mTOR controls embryonic and adult myogenesis via mTORC1. Development 2019; 146:dev.172460. [PMID: 30872276 DOI: 10.1242/dev.172460] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 03/04/2019] [Indexed: 12/11/2022]
Abstract
The formation of multi-nucleated muscle fibers from progenitors requires the fine-tuned and coordinated regulation of proliferation, differentiation and fusion, both during development and after injury in the adult. Although some of the key factors that are involved in the different steps are well known, how intracellular signals are coordinated and integrated is largely unknown. Here, we investigated the role of the cell-growth regulator mTOR by eliminating essential components of the mTOR complexes 1 (mTORC1) and 2 (mTORC2) in mouse muscle progenitors. We show that inactivation of mTORC1, but not mTORC2, in developing muscle causes perinatal death. In the adult, mTORC1 deficiency in muscle stem cells greatly impinges on injury-induced muscle regeneration. These phenotypes are because of defects in the proliferation and fusion capacity of the targeted muscle progenitors. However, mTORC1-deficient muscle progenitors partially retain their myogenic function. Hence, our results show that mTORC1 and not mTORC2 is an important regulator of embryonic and adult myogenesis, and they point to alternative pathways that partially compensate for the loss of mTORC1.This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Nathalie Rion
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | | | - Shuo Lin
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | - Leonie Enderle
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | | | | | - Markus A Rüegg
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| |
Collapse
|
21
|
Baghdadi MB, Firmino J, Soni K, Evano B, Di Girolamo D, Mourikis P, Castel D, Tajbakhsh S. Notch-Induced miR-708 Antagonizes Satellite Cell Migration and Maintains Quiescence. Cell Stem Cell 2018; 23:859-868.e5. [PMID: 30416072 DOI: 10.1016/j.stem.2018.09.017] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 07/18/2018] [Accepted: 09/27/2018] [Indexed: 12/31/2022]
Abstract
Critical features of stem cells include anchoring within a niche and activation upon injury. Notch signaling maintains skeletal muscle satellite (stem) cell quiescence by inhibiting differentiation and inducing expression of extracellular components of the niche. However, the complete spectrum of how Notch safeguards quiescence is not well understood. Here, we perform Notch ChIP-sequencing and small RNA sequencing in satellite cells and identify the Notch-induced microRNA-708, which is a mirtron that is highly expressed in quiescent cells and sharply downregulated in activated cells. We employ in vivo and ex vivo functional studies, in addition to live imaging, to show that miR-708 regulates quiescence and self-renewal by antagonizing cell migration through targeting the transcripts of the focal-adhesion-associated protein Tensin3. Therefore, this study identifies a Notch-miR708-Tensin3 axis and suggests that Notch signaling can regulate satellite cell quiescence and transition to the activation state through dynamic regulation of the migratory machinery.
Collapse
Affiliation(s)
- Meryem B Baghdadi
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris 75015, France; CNRS UMR 3738, Institut Pasteur, Paris 75015, France; Sorbonne Universités, UPMC, University of Paris 06, IFD-ED 515, Paris 75252, France
| | - Joao Firmino
- Bioimaging and Optics platform (BIOP), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Kartik Soni
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris 75015, France; CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| | - Brendan Evano
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris 75015, France; CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| | - Daniela Di Girolamo
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris 75015, France; Dipartimento di Medicina Clinica e Chirurgica, Università degli Studi di Napoli Frederico II, 80131 Naples, Italy
| | | | - David Castel
- UMR8203 "Vectorologie et Thérapeutiques Anticancéreuses," CNRS, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France; Département de Cancérologie de l'Enfant et de l'Adolescent, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris 75015, France; CNRS UMR 3738, Institut Pasteur, Paris 75015, France.
| |
Collapse
|
22
|
Zecchini S, Giovarelli M, Perrotta C, Morisi F, Touvier T, Di Renzo I, Moscheni C, Bassi MT, Cervia D, Sandri M, Clementi E, De Palma C. Autophagy controls neonatal myogenesis by regulating the GH-IGF1 system through a NFE2L2- and DDIT3-mediated mechanism. Autophagy 2018; 15:58-77. [PMID: 30081710 DOI: 10.1080/15548627.2018.1507439] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Macroautophagy/autophagy is emerging as an important process in adult muscle stem cells functions: it regulates metabolic reprogramming during activation from a quiescent state, maintains stemness and prevents senescence. We now show that autophagy is specifically required for neonatal myogenesis and muscle development. Specific deletion of Atg7 in PAX7+ (paired box 7) precursors led in mice to a dwarf phenotype, with an effect restricted to the neonatal phase of muscle development. Atg7 knockdown suppressed neonatal satellite cell (nSC) proliferation and differentiation, downregulating the GH-IGF1 functions. When we disrupted autophagy, NFE2L2/NRF2 (nuclear factor, erythroid 2 like 2) accumulated in muscle and nSCs and negatively modulated DDIT3/CHOP (DNA-damage inducible transcript 3) expression. Lower levels of DDIT3 were responsible for reduced GHR expression leading to impaired local production of IGF1. Our results conclusively identify a novel autophagy-dependent pathway that regulates nSC behavior and indicate that autophagy is required for skeletal muscle development in the neonatal phase. Abbreviations: AKT/protein kinase B: Thymoma viral proto-oncogene; ASCs: adult stem cells; ATF4: activating transcription factor 4; ATG7: autophagy related 7; BAT: brown adipose tissue; BMP: bone morphogenetic protein; CEBPB: CCAAT/enhancer binding protein (C/EBP), beta; CSA: cross sectional area; CTNNB1: catenin (cadherin associated protein), beta 1; DDIT3: DNA-damage inducible transcript 3; DM: differentiation medium; E: embryonic stage; EIF2AK3/PERK; EIF4EBP1: eukaryotic translation initiation factor 2 alpha kinase 3; eukaryotic translation initiation factor 4E binding protein 1; ER: endoplasmic reticulum; FGF21: fibroblast growth factor 21; GH: growth hormone; GHR: growth hormone receptor; HSCs: hematopoietic stem cells; IGF1: insulin-like growth factor 1; ITGAM: integrin alpha M; KEAP1: kelch-like ECH-associated protein 1; LY6A/Sca-1; MAP1LC3: lymphocyte antigen 6 complex, locus A; microtubule-associated protein 1 light chain 3; MAPK1/ERK2: mitogen-activated protein kinase 1; MAPK3/ERK1: mitogen-activated protein kinase 3; miRNAs: microRNAs; MSCs: mesenchymal stem cells; MTOR: mechanistic target of rapamycin kinase; mtUPR: mitochondrial unfolded protein response; MYF5: myogenic factor 5; MYH: myosin, heavy polypeptide; MYOD1: myogenic differentiation 1; MYOG: myogenin; NFE2L2: nuclear factor, erythroid derived 2, like 2; nSC: neonatal satellite cells; NSCs: neuronal stem cells; P: postnatal day; PAX7: paired box 7; PECAM1: platelet/endothelial cell adhesion molecule 1; PPARG: peroxisome proliferator activated receptor gamma; PTPRC: protein tyrosine phosphatase, receptor type, C; ROS: reactive oxygen species; RPS6: ribosomal protein S6; SCs: adult satellite cells; SQSTM1: sequestosome 1; STAT5: signal transducer and activator of transcription 5; TGFB1: transforming growth factor beta 1; WAT: white adipose tissue; WT: wild type.
Collapse
Affiliation(s)
- Silvia Zecchini
- a Unit of Clinical Pharmacology , University Hospital "Luigi Sacco"-ASST Fatebenefratelli Sacco , Milano , Italy
| | - Matteo Giovarelli
- b Department of Biomedical and Clinical Sciences "Luigi Sacco" , Università degli Studi di Milano , Milano , Italy
| | - Cristiana Perrotta
- b Department of Biomedical and Clinical Sciences "Luigi Sacco" , Università degli Studi di Milano , Milano , Italy
| | - Federica Morisi
- c Division of Genetics and Cell Biology , IRCCS Ospedale San Raffaele , Milano , Italy
| | - Thierry Touvier
- d Biology of Myelin Unit, Division of Genetics and Cell Biology , IRCCS Ospedale San Raffaele , Milano , Italy
| | - Ilaria Di Renzo
- b Department of Biomedical and Clinical Sciences "Luigi Sacco" , Università degli Studi di Milano , Milano , Italy
| | - Claudia Moscheni
- b Department of Biomedical and Clinical Sciences "Luigi Sacco" , Università degli Studi di Milano , Milano , Italy
| | - Maria Teresa Bassi
- e Laboratory of Molecular Biology , IRCCS Eugenio Medea , Bosisio Parini , Italy
| | - Davide Cervia
- f Department for Innovation in Biological, Agro-food and Forest systems , Università degli Studi della Tuscia , Viterbo , Italy
| | - Marco Sandri
- g Department of Biomedical Science , University of Padova , Padova , Italy.,h Laboratory of Molecular Biology , Venetian Institute of Molecular Medicine , Padova , Italy
| | - Emilio Clementi
- e Laboratory of Molecular Biology , IRCCS Eugenio Medea , Bosisio Parini , Italy.,i Department of Biomedical and Clinical Sciences "Luigi Sacco" , Università degli Studi di Milano , Milano , Italy
| | - Clara De Palma
- a Unit of Clinical Pharmacology , University Hospital "Luigi Sacco"-ASST Fatebenefratelli Sacco , Milano , Italy.,b Department of Biomedical and Clinical Sciences "Luigi Sacco" , Università degli Studi di Milano , Milano , Italy
| |
Collapse
|
23
|
Baghdadi MB, Castel D, Machado L, Fukada SI, Birk DE, Relaix F, Tajbakhsh S, Mourikis P. Reciprocal signalling by Notch-Collagen V-CALCR retains muscle stem cells in their niche. Nature 2018; 557:714-718. [PMID: 29795344 PMCID: PMC5985950 DOI: 10.1038/s41586-018-0144-9] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 04/06/2018] [Indexed: 12/31/2022]
Abstract
The microenvironment is critical for stem cell maintenance and can be of cellular and non-cellular composition, including secreted growth factors and extracellular matrix (ECM)1–3. Although Notch and other signalling pathways have been reported to regulate quiescence4–9, the composition and source of niche molecules remain largely unknown. Here, we show that adult muscle satellite (stem) cells produce ECM collagens to maintain quiescence cell-autonomously. By ChIP-sequencing we identified NOTCH/RBPJ-bound regulatory elements adjacent to specific collagen genes, whose expression is deregulated in Notch mutant mice. Moreover, we show that satellite cell produced collagen V (COLV) is a critical component of the quiescent niche, as conditional deletion of Col5a1 leads to anomalous cell cycle entry and gradual diminution of the stem cell pool. Notably, the interaction of COLV with satellite cells is mediated by CALCR, for which COLV acts as a surrogate local ligand. Strikingly, systemic administration of a calcitonin derivative is sufficient to rescue the quiescence and self-renewal defects scored in COLV null satellite cells. This study unveils a Notch/COLV/CALCR signalling cascade that cell-autonomously maintains the satellite cell quiescent state and raises the possibility of a similar reciprocal mechanism acting in diverse stem cell populations.
Collapse
Affiliation(s)
- Meryem B Baghdadi
- Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris, France.,CNRS UMR 3738, Institut Pasteur, Paris, France.,Sorbonne Universités, UPMC, University of Paris 06, Paris, France
| | - David Castel
- UMR8203, CNRS, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France.,Département de Cancérologie de l'Enfant et de l'Adolescent, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Léo Machado
- INSERM IMRB U955-E10, UPEC, ENVA, EFS, Créteil, France
| | - So-Ichiro Fukada
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - David E Birk
- Department of Molecular Pharmacology & Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | | | - Shahragim Tajbakhsh
- Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris, France. .,CNRS UMR 3738, Institut Pasteur, Paris, France.
| | | |
Collapse
|
24
|
Kheir E, Cusella G, Messina G, Cossu G, Biressi S. Reporter-Based Isolation of Developmental Myogenic Progenitors. Front Physiol 2018; 9:352. [PMID: 29674978 PMCID: PMC5895918 DOI: 10.3389/fphys.2018.00352] [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: 01/12/2018] [Accepted: 03/20/2018] [Indexed: 11/13/2022] Open
Abstract
The formation and activity of mammalian tissues entail finely regulated processes, involving the concerted organization and interaction of multiple cell types. In recent years the prospective isolation of distinct progenitor and stem cell populations has become a powerful tool in the hands of developmental biologists and has rendered the investigation of their intrinsic properties possible. In this protocol, we describe how to purify progenitors with different lineage history and degree of differentiation from embryonic and fetal skeletal muscle by fluorescence-activated cell sorting (FACS). The approach takes advantage of a panel of murine strains expressing fluorescent reporter genes specifically in the myogenic progenitors. We provide a detailed description of the dissection procedures and of the enzymatic dissociation required to maximize the yield of mononucleated cells for subsequent FACS-based purification. The procedure takes ~6–7 h to complete and allows for the isolation and the subsequent molecular and phenotypic characterization of developmental myogenic progenitors.
Collapse
Affiliation(s)
- Eyemen Kheir
- Centre for Integrative Biology (CIBIO), University of Trento, Trento, Italy.,Dulbecco Telethon Institute, University of Trento, Trento, Italy
| | - Gabriella Cusella
- Human Anatomy Unit, Department of Public Health, Experimental Medicine and Forensic, University of Pavia, Pavia, Italy.,Center for Health Technologies, University of Pavia, Pavia, Italy
| | | | - Giulio Cossu
- Division of Cell Matrix Biology and Regenerative Medicine, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Stefano Biressi
- Centre for Integrative Biology (CIBIO), University of Trento, Trento, Italy.,Dulbecco Telethon Institute, University of Trento, Trento, Italy
| |
Collapse
|
25
|
The Factor Inhibiting HIF Asparaginyl Hydroxylase Regulates Oxidative Metabolism and Accelerates Metabolic Adaptation to Hypoxia. Cell Metab 2018; 27:898-913.e7. [PMID: 29617647 PMCID: PMC5887987 DOI: 10.1016/j.cmet.2018.02.020] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 12/29/2017] [Accepted: 02/20/2018] [Indexed: 01/16/2023]
Abstract
Animals require an immediate response to oxygen availability to allow rapid shifts between oxidative and glycolytic metabolism. These metabolic shifts are highly regulated by the HIF transcription factor. The factor inhibiting HIF (FIH) is an asparaginyl hydroxylase that controls HIF transcriptional activity in an oxygen-dependent manner. We show here that FIH loss increases oxidative metabolism, while also increasing glycolytic capacity, and that this gives rise to an increase in oxygen consumption. We further show that the loss of FIH acts to accelerate the cellular metabolic response to hypoxia. Skeletal muscle expresses 50-fold higher levels of FIH than other tissues: we analyzed skeletal muscle FIH mutants and found a decreased metabolic efficiency, correlated with an increased oxidative rate and an increased rate of hypoxic response. We find that FIH, through its regulation of oxidation, acts in concert with the PHD/vHL pathway to accelerate HIF-mediated metabolic responses to hypoxia.
Collapse
|
26
|
Barrott JJ, Illum BE, Jin H, Hedberg ML, Wang Y, Grossmann A, Haldar M, Capecchi MR, Jones KB. Paracrine osteoprotegerin and β-catenin stabilization support synovial sarcomagenesis in periosteal cells. J Clin Invest 2017; 128:207-218. [PMID: 29202462 DOI: 10.1172/jci94955] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 10/10/2017] [Indexed: 01/30/2023] Open
Abstract
Synovial sarcoma (SS) is an aggressive soft-tissue sarcoma that is often discovered during adolescence and young adulthood. Despite the name, synovial sarcoma does not typically arise from a synoviocyte but instead arises in close proximity to bones. Previous work demonstrated that mice expressing the characteristic SS18-SSX fusion oncogene in myogenic factor 5-expressing (Myf5-expressing) cells develop fully penetrant sarcomagenesis, suggesting skeletal muscle progenitor cell origin. However, Myf5 is not restricted to committed myoblasts in embryos but is also expressed in multipotent mesenchymal progenitors. Here, we demonstrated that human SS and mouse tumors arising from SS18-SSX expression in the embryonic, but not postnatal, Myf5 lineage share an anatomic location that is frequently adjacent to bone. Additionally, we showed that SS can originate from periosteal cells expressing SS18-SSX alone and from preosteoblasts expressing the fusion oncogene accompanied by the added stabilization of β-catenin, which is a common secondary change in SS. Expression and secretion of the osteoclastogenesis inhibitory factor osteoprotegerin enabled early growth of SS18-SSX2-transformed cells, indicating a paracrine link between the bone and synovial sarcomagenesis. These findings explain the skeletal contact frequently observed in human SS and may provide alternate means of enabling SS18-SSX-driven oncogenesis in cells as differentiated as preosteoblasts.
Collapse
Affiliation(s)
| | - Benjamin E Illum
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA
| | - Huifeng Jin
- Departments of Orthopaedics and Oncological Sciences, and
| | - Matthew L Hedberg
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri, USA
| | - Yanliang Wang
- Departments of Orthopaedics and Oncological Sciences, and
| | - Allie Grossmann
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Malay Haldar
- Department of Pathology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mario R Capecchi
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA
| | - Kevin B Jones
- Departments of Orthopaedics and Oncological Sciences, and
| |
Collapse
|
27
|
Zammit PS. Function of the myogenic regulatory factors Myf5, MyoD, Myogenin and MRF4 in skeletal muscle, satellite cells and regenerative myogenesis. Semin Cell Dev Biol 2017; 72:19-32. [PMID: 29127046 DOI: 10.1016/j.semcdb.2017.11.011] [Citation(s) in RCA: 509] [Impact Index Per Article: 63.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 12/19/2022]
Abstract
Discovery of the myogenic regulatory factor family of transcription factors MYF5, MYOD, Myogenin and MRF4 was a seminal step in understanding specification of the skeletal muscle lineage and control of myogenic differentiation during development. These factors are also involved in specification of the muscle satellite cell lineage, which becomes the resident stem cell compartment inadult skeletal muscle. While MYF5, MYOD, Myogenin and MRF4 have subtle roles in mature muscle, they again play a crucial role in directing satellite cell function to regenerate skeletal muscle: linking the genetic control of developmental and regenerative myogenesis. Here, I review the role of the myogenic regulatory factors in developing and mature skeletal muscle, satellite cell specification and muscle regeneration.
Collapse
Affiliation(s)
- Peter S Zammit
- King's College London, Randall Centre for Cell and Molecular Biophysics, London, SE1 1UL, UK.
| |
Collapse
|
28
|
Shin JY, Méndez-López I, Hong M, Wang Y, Tanji K, Wu W, Shugol L, Krauss RS, Dauer WT, Worman HJ. Lamina-associated polypeptide 1 is dispensable for embryonic myogenesis but required for postnatal skeletal muscle growth. Hum Mol Genet 2017; 26:65-78. [PMID: 27798115 DOI: 10.1093/hmg/ddw368] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 10/21/2016] [Indexed: 12/17/2022] Open
Abstract
Lamina-associated polypeptide 1 (LAP1) is an integral protein of the inner nuclear membrane that has been implicated in striated muscle maintenance. Mutations in its gene have been linked to muscular dystrophy and cardiomyopathy. As germline deletion of the gene encoding LAP1 is perinatal lethal, we explored its potential role in myogenic differentiation and development by generating a conditional knockout mouse in which the protein is depleted from muscle progenitors at embryonic day 8.5 (Myf5-Lap1CKO mice). Although cultured myoblasts lacking LAP1 demonstrated defective terminal differentiation and altered expression of muscle regulatory factors, embryonic myogenesis and formation of skeletal muscle occurred in both mice with a Lap1 germline deletion and Myf5-Lap1CKO mice. However, skeletal muscle fibres were hypotrophic and their nuclei were morphologically abnormal with a wider perinuclear space than normal myonuclei. Myf5-Lap1CKO mouse skeletal muscle contained fewer satellite cells than normal and these cells had evidence of reduced myogenic potential. Abnormalities in signalling pathways required for postnatal hypertrophic growth were also observed in skeletal muscles of these mice. Our results demonstrate that early embryonic depletion of LAP1 does not impair myogenesis but that it is necessary for postnatal skeletal muscle growth.
Collapse
Affiliation(s)
- Ji-Yeon Shin
- Department of Medicine.,Department of Pathology and Cell Biology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Iván Méndez-López
- Department of Medicine.,Department of Pathology and Cell Biology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Mingi Hong
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yuexia Wang
- Department of Medicine.,Department of Pathology and Cell Biology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Kurenai Tanji
- Department of Pathology and Cell Biology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Wei Wu
- Department of Medicine.,Department of Pathology and Cell Biology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Leana Shugol
- Department of Medicine.,Department of Pathology and Cell Biology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Robert S Krauss
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William T Dauer
- Department of Neurology.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Howard J Worman
- Department of Medicine.,Department of Pathology and Cell Biology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| |
Collapse
|
29
|
Lee PL, Jung SM, Guertin DA. The Complex Roles of Mechanistic Target of Rapamycin in Adipocytes and Beyond. Trends Endocrinol Metab 2017; 28:319-339. [PMID: 28237819 PMCID: PMC5682923 DOI: 10.1016/j.tem.2017.01.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/20/2017] [Accepted: 01/23/2017] [Indexed: 01/01/2023]
Abstract
Having healthy adipose tissue is essential for metabolic fitness. This is clear from the obesity epidemic, which is unveiling a myriad of comorbidities associated with excess adipose tissue including type 2 diabetes, cardiovascular disease, and cancer. Lipodystrophy also causes insulin resistance, emphasizing the importance of having a balanced amount of fat. In cells, the mechanistic target of rapamycin (mTOR) complexes 1 and 2 (mTORC1 and mTORC2, respectively) link nutrient and hormonal signaling with metabolism, and recent studies are shedding new light on their in vivo roles in adipocytes. In this review, we discuss how recent advances in adipose tissue and mTOR biology are converging to reveal new mechanisms that maintain healthy adipose tissue, and discuss ongoing mysteries of mTOR signaling, particularly for the less understood complex mTORC2.
Collapse
Affiliation(s)
- Peter L Lee
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street, Worcester, MA 01605, USA
| | - Su Myung Jung
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street, Worcester, MA 01605, USA
| | - David A Guertin
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street, Worcester, MA 01605, USA.
| |
Collapse
|
30
|
Ignacio DL, Silvestre DHS, Anne-Palmer E, Bocco BMLC, Fonseca TL, Ribeiro MO, Gereben B, Bianco AC, Werneck-de-Castro JP. Early Developmental Disruption of Type 2 Deiodinase Pathway in Mouse Skeletal Muscle Does Not Impair Muscle Function. Thyroid 2017; 27:577-586. [PMID: 27967605 PMCID: PMC5385430 DOI: 10.1089/thy.2016.0392] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Myogenesis is positively regulated by thyroid hormone (triiodothyronine [T3]), which is amplified by the type 2 deiodinase (D2) activation of thyroxine to T3. Global inactivation of the Dio2 gene impairs skeletal muscle (SKM) differentiation and regeneration in response to muscle injury. Given that newborn and adult mice with late developmental SKM Dio2 disruption do not develop a significant phenotype, it was hypothesized that D2 plays an early role in this process. METHODS This was tested in mice with SKM disruption of Dio2 driven by two early developmental promoters: MYF5 and MYOD. RESULTS MYF5 myoblasts in culture differentiate normally into myotubes, despite loss of almost all D2 activity. Dio2 mRNA levels in developing SKM obtained from MYF5-D2KO embryos (E18.5) were about 54% of control littermates, but the expression of the T3-responsive genes Myh1 and 7 and Atp2a1 and 2 were not affected. In MYF5-D2KO and MYOD-D2KO neonatal hind-limb muscle, the expression of Myh1 and 7 and Atp2a2 remained unaffected, despite 60-70% loss in D2 activity and/or mRNA. Only in MYOD-D2KO neonatal muscle was there a 40% reduction in Atp2a1 mRNA. Postnatal growth of both mouse models and SKM function as assessed by exercise capacity and measurement of muscle strength were normal. Furthermore, an analysis of the adult soleus revealed no changes in the expression of T3-responsive genes, except for an about 18% increase in MYOD-D2KO SOL Myh7 mRNA. CONCLUSION Two mouse models of early developmental disruption of Dio2 in myocyte precursor exhibit no significant SKM phenotype.
Collapse
Affiliation(s)
- Daniele L Ignacio
- 1 Division of Endocrinology and Metabolism, Rush University Medical Center , Chicago, Illinois
- 2 Biophysics Institute and School of Physical Education and Sports, Federal University of Rio de Janeiro , Rio de Janeiro, Brazil
| | - Diego H S Silvestre
- 1 Division of Endocrinology and Metabolism, Rush University Medical Center , Chicago, Illinois
- 2 Biophysics Institute and School of Physical Education and Sports, Federal University of Rio de Janeiro , Rio de Janeiro, Brazil
- 3 Nutrition Institute Josué de Castro, Federal University of Rio de Janeiro , Rio de Janeiro, Brazil
| | - Elena Anne-Palmer
- 1 Division of Endocrinology and Metabolism, Rush University Medical Center , Chicago, Illinois
| | - Barbara M L C Bocco
- 1 Division of Endocrinology and Metabolism, Rush University Medical Center , Chicago, Illinois
- 4 Department of Translational Medicine, Federal University of São Paulo , São Paulo, Brazil
| | - Tatiana L Fonseca
- 1 Division of Endocrinology and Metabolism, Rush University Medical Center , Chicago, Illinois
| | - Miriam O Ribeiro
- 5 Developmental Disorders Program, Center for Biological and Health Sciences, Mackenzie Presbyterian University , São Paulo, Brazil
| | - Balázs Gereben
- 6 Department of Endocrine Neurobiology, Institute of Experimental Medicine , Hungarian Academy of Sciences, Budapest, Hungary
| | - Antonio C Bianco
- 1 Division of Endocrinology and Metabolism, Rush University Medical Center , Chicago, Illinois
| | - Joao P Werneck-de-Castro
- 1 Division of Endocrinology and Metabolism, Rush University Medical Center , Chicago, Illinois
- 2 Biophysics Institute and School of Physical Education and Sports, Federal University of Rio de Janeiro , Rio de Janeiro, Brazil
- 3 Nutrition Institute Josué de Castro, Federal University of Rio de Janeiro , Rio de Janeiro, Brazil
| |
Collapse
|
31
|
Tenente IM, Hayes MN, Ignatius MS, McCarthy K, Yohe M, Sindiri S, Gryder B, Oliveira ML, Ramakrishnan A, Tang Q, Chen EY, Petur Nielsen G, Khan J, Langenau DM. Myogenic regulatory transcription factors regulate growth in rhabdomyosarcoma. eLife 2017; 6. [PMID: 28080960 PMCID: PMC5231408 DOI: 10.7554/elife.19214] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 12/08/2016] [Indexed: 01/01/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is a pediatric malignacy of muscle with myogenic regulatory transcription factors MYOD and MYF5 being expressed in this disease. Consensus in the field has been that expression of these factors likely reflects the target cell of transformation rather than being required for continued tumor growth. Here, we used a transgenic zebrafish model to show that Myf5 is sufficient to confer tumor-propagating potential to RMS cells and caused tumors to initiate earlier and have higher penetrance. Analysis of human RMS revealed that MYF5 and MYOD are mutually-exclusively expressed and each is required for sustained tumor growth. ChIP-seq and mechanistic studies in human RMS uncovered that MYF5 and MYOD bind common DNA regulatory elements to alter transcription of genes that regulate muscle development and cell cycle progression. Our data support unappreciated and dominant oncogenic roles for MYF5 and MYOD convergence on common transcriptional targets to regulate human RMS growth. DOI:http://dx.doi.org/10.7554/eLife.19214.001
Collapse
Affiliation(s)
- Inês M Tenente
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital, Boston, United States.,Harvard Stem Cell Institute, Cambridge, United States.,GABBA Program, Abel Salazar Biomedical Sciences Institute, University of Porto, Porto, Portugal
| | - Madeline N Hayes
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital, Boston, United States.,Harvard Stem Cell Institute, Cambridge, United States
| | - Myron S Ignatius
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital, Boston, United States.,Harvard Stem Cell Institute, Cambridge, United States.,Molecular Medicine, Greehey Children's Cancer Research Institute, San Antonio, United States
| | - Karin McCarthy
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital, Boston, United States.,Harvard Stem Cell Institute, Cambridge, United States
| | - Marielle Yohe
- Oncogenomics Section, Pediatric Oncology Branch, Advanced Technology Center, National Cancer Institute, Gaithersburg, United States
| | - Sivasish Sindiri
- Oncogenomics Section, Pediatric Oncology Branch, Advanced Technology Center, National Cancer Institute, Gaithersburg, United States
| | - Berkley Gryder
- Oncogenomics Section, Pediatric Oncology Branch, Advanced Technology Center, National Cancer Institute, Gaithersburg, United States
| | - Mariana L Oliveira
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital, Boston, United States.,Harvard Stem Cell Institute, Cambridge, United States.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ashwin Ramakrishnan
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital, Boston, United States.,Harvard Stem Cell Institute, Cambridge, United States
| | - Qin Tang
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital, Boston, United States.,Harvard Stem Cell Institute, Cambridge, United States
| | - Eleanor Y Chen
- Department of Pathology, University of Washington, Seattle, United States
| | - G Petur Nielsen
- Department of Pathology, Massachusetts General Hospital, Boston, United States
| | - Javed Khan
- Oncogenomics Section, Pediatric Oncology Branch, Advanced Technology Center, National Cancer Institute, Gaithersburg, United States
| | - David M Langenau
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital, Boston, United States.,Harvard Stem Cell Institute, Cambridge, United States
| |
Collapse
|
32
|
Yin ZZ, Dong XY, Dong DJ, Ma YZ. Association of MYF5 and KLF15 gene polymorphisms with carcass traits in domestic pigeons (Columba livia). Br Poult Sci 2016; 57:612-618. [PMID: 27180898 DOI: 10.1080/00071668.2016.1190000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Single nucleotide polymorphisms (SNPs) in the exons of the myogenic factor 5 (MYF5) and Kruppel-like factor 15 (KLF15) genes were identified and analysed by using DNA sequencing methods in 60 female domestic pigeons (Columba livia). Five SNPs (T5067A, C5084T, C5101T, T5127A and C5154G) were detected in exon 3 of MYF5 and 6 SNPs (C1398T, C1464T, G1542A, C1929T, G1965A and A2355G) were found in exon 2 of KLF15, respectively. The analysis revealed three genotypes, in which the AA genotype was dominant and the A allele showed a dominant advantage. For the MYF5 gene, the C5084T and T5127A SNP genotypes were significantly associated with carcass traits of pigeons. Within those two SNPs, the BB genotype showed relatively higher trait association values than those of AA or AB genotypes. No significant association was observed between the KLF15 SNP genotypes and carcass traits. These results indicated that the MYF5 gene is a potential major gene affecting carcass traits in domestic pigeons. The BB genotype of the C5084T and T5127A SNPs could be a potential candidate genetic marker for marker-assisted selection in pigeon.
Collapse
Affiliation(s)
- Z Z Yin
- a Animal Science College , Zhejiang University , Hangzhou , China
| | - X Y Dong
- a Animal Science College , Zhejiang University , Hangzhou , China
| | - D J Dong
- a Animal Science College , Zhejiang University , Hangzhou , China
| | - Y Z Ma
- a Animal Science College , Zhejiang University , Hangzhou , China
| |
Collapse
|
33
|
Wei Y, Tao X, Xu H, Chen Y, Zhu L, Tang G, Li M, Jiang A, Shuai S, Ma J, Jin L, Wen A, Wang Q, Zhu G, Xie M, Wu J, He T, Jiang Y, Li X. Role of miR-181a-5p and endoplasmic reticulum stress in the regulation of myogenic differentiation. Gene 2016; 592:60-70. [DOI: 10.1016/j.gene.2016.07.056] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 07/13/2016] [Accepted: 07/22/2016] [Indexed: 01/02/2023]
|
34
|
Conerly ML, Yao Z, Zhong JW, Groudine M, Tapscott SJ. Distinct Activities of Myf5 and MyoD Indicate Separate Roles in Skeletal Muscle Lineage Specification and Differentiation. Dev Cell 2016; 36:375-85. [PMID: 26906734 DOI: 10.1016/j.devcel.2016.01.021] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 12/22/2015] [Accepted: 01/26/2016] [Indexed: 01/03/2023]
Abstract
Most transcription factor families contain highly related paralogs generated by gene duplication, and functional divergence is generally accomplished by activation of distinct sets of genes by each member. Here we compare the molecular functions of Myf5 and MyoD, two highly related bHLH transcription factors that regulate skeletal muscle specification and differentiation. We find that MyoD and Myf5 bind the same sites genome-wide but have distinct functions: Myf5 induces histone acetylation without Pol II recruitment or robust gene activation, whereas MyoD induces histone acetylation, recruits Pol II, and robustly activates gene transcription. Therefore, the initial specification of the muscle lineage by Myf5 occurs without significant induction of gene transcription. Transcription of the skeletal muscle program is then achieved by the subsequent expression of MyoD, which binds to the same sites as Myf5, indicating that each factor regulates distinct steps in gene initiation and transcription at a shared set of binding sites.
Collapse
Affiliation(s)
- Melissa L Conerly
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Basic Science Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - Zizhen Yao
- Allen Brain Institute, Seattle, WA 98105, USA
| | - Jun Wen Zhong
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Mark Groudine
- Basic Science Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Stephen J Tapscott
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Neurology, University of Washington, School of Medicine, Seattle, WA 98105, USA.
| |
Collapse
|
35
|
Tierney MT, Sacco A. Satellite Cell Heterogeneity in Skeletal Muscle Homeostasis. Trends Cell Biol 2016; 26:434-444. [PMID: 26948993 DOI: 10.1016/j.tcb.2016.02.004] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/06/2016] [Accepted: 02/10/2016] [Indexed: 12/12/2022]
Abstract
The cellular turnover required for skeletal muscle maintenance and repair is mediated by resident stem cells, also termed satellite cells. Satellite cells normally reside in a quiescent state, intermittently entering the cell cycle to fuse with neighboring myofibers and replenish the stem cell pool. However, the mechanisms by which satellite cells maintain the precise balance between self-renewal and differentiation necessary for long-term homeostasis remain unclear. Recent work has supported a previously unappreciated heterogeneity in the satellite cell compartment that may underlie the observed variability in cell fate and function. In this review, we examine the work supporting this notion as well as the potential governing principles, developmental origins, and principal determinants of satellite cell heterogeneity.
Collapse
Affiliation(s)
- Matthew T Tierney
- Graduate School of Biomedical Sciences, Sanford Burnham Prebys Medical Discovery Institute, 10901N Torrey Pines Road, La Jolla, CA 92037, USA; Development, Aging, and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Alessandra Sacco
- Development, Aging, and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901N Torrey Pines Rd, La Jolla, CA 92037, USA.
| |
Collapse
|
36
|
Hu B, Simon-Keller K, Küffer S, Ströbel P, Braun T, Marx A, Porubsky S. Myf5 and Myogenin in the development of thymic myoid cells - Implications for a murine in vivo model of myasthenia gravis. Exp Neurol 2015; 277:76-85. [PMID: 26708556 DOI: 10.1016/j.expneurol.2015.12.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 11/24/2015] [Accepted: 12/15/2015] [Indexed: 01/24/2023]
Abstract
Myasthenia gravis (MG) is caused by autoantibodies against the neuromuscular junction of striated muscle. Most MG patients have autoreactive T- and B-cells directed to the acetylcholine receptor (AChR). To achieve immunologic tolerance, developing thymocytes are normally eliminated after recognition of self-antigen-derived peptides. Presentation of muscle-specific antigens is likely achieved through two pathways: on medullary thymic epithelial cells and on medullary dendritic cells cross-presenting peptides derived from a unique population of thymic myoid cells (TMC). Decades ago, it has been hypothesized that TMC play a key role in the induction of immunological tolerance towards skeletal muscle antigens. However, an experimental model to address this postulate has not been available. To generate such a model, we tested the hypothesis that the development of TMC depends on myogenic regulatory factors. To this end, we utilized Myf5-deficient mice, which lack the first wave of muscle cells but form normal skeletal muscles later during development, and Myogenin-deficient mice, which fail to form differentiated myofibers. We demonstrate for the first time that Myf5- and Myogenin-deficient mice showed a partial or complete, respectively, loss of TMC in an otherwise regularly structured thymus. To overcome early postnatal lethality of muscle-deficient, Myogenin-knockout mice we transplanted Myogenin-deficient fetal thymuses into Foxn1(nu/nu) mice that lack their own thymus anlage. We found that the transplants are functional but lack TMC. In combination with established immunization strategies (utilizing AChR or Titin), this model should enable us in the future testing the hypothesis that TMC play an indispensable role in the development of central tolerance towards striated muscle antigens.
Collapse
Affiliation(s)
- Bo Hu
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Katja Simon-Keller
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Stefan Küffer
- Institute of Pathology, University of Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Philipp Ströbel
- Institute of Pathology, University of Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Thomas Braun
- Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
| | - Alexander Marx
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Stefan Porubsky
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany.
| |
Collapse
|
37
|
Nagandla H, Lopez S, Yu W, Rasmussen TL, Tucker HO, Schwartz RJ, Stewart MD. Defective myogenesis in the absence of the muscle-specific lysine methyltransferase SMYD1. Dev Biol 2015; 410:86-97. [PMID: 26688546 DOI: 10.1016/j.ydbio.2015.12.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 12/07/2015] [Accepted: 12/07/2015] [Indexed: 11/19/2022]
Abstract
The SMYD (SET and MYND domain) family of lysine methyltransferases harbor a unique structure in which the methyltransferase (SET) domain is intervened by a zinc finger protein-protein interaction MYND domain. SMYD proteins methylate both histone and non-histone substrates and participate in diverse biological processes including transcriptional regulation, DNA repair, proliferation and apoptosis. Smyd1 is unique among the five family members in that it is specifically expressed in striated muscles. Smyd1 is critical for development of the right ventricle in mice. In zebrafish, Smyd1 is necessary for sarcomerogenesis in fast-twitch muscles. Smyd1 is expressed in the skeletal muscle lineage throughout myogenesis and in mature myofibers, shuttling from nucleus to cytosol during myoblast differentiation. Because of this expression pattern, we hypothesized that Smyd1 plays multiple roles at different stages of myogenesis. To determine the role of Smyd1 in mammalian myogenesis, we conditionally eliminated Smyd1 from the skeletal muscle lineage at the myoblast stage using Myf5(cre). Deletion of Smyd1 impaired myoblast differentiation, resulted in fewer myofibers and decreased expression of muscle-specific genes. Muscular defects were temporally restricted to the second wave of myogenesis. Thus, in addition to the previously described functions for Smyd1 in heart development and skeletal muscle sarcomerogenesis, these results point to a novel role for Smyd1 in myoblast differentiation.
Collapse
Affiliation(s)
- Harika Nagandla
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Suhujey Lopez
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Wei Yu
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Tara L Rasmussen
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA; Molecular Biosciences and Institute for Cellular and Molecular Biology, University of Texas, Austin, TX, USA
| | - Haley O Tucker
- Molecular Biosciences and Institute for Cellular and Molecular Biology, University of Texas, Austin, TX, USA
| | - Robert J Schwartz
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA; Stem Cell Engineering Department, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, TX, USA
| | - M David Stewart
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA.
| |
Collapse
|
38
|
Abstract
The tongue and mandible have common origins. They arise simultaneously from the mandibular arch and are coordinated in their development and growth, which is evident from several clinical conditions such as Pierre Robin sequence. Here, we review in detail the molecular networks controlling both mandible and tongue development. We also discuss their mechanical relationship and evolution as well as the potential for stem cell-based therapies for disorders affecting these organs.
Collapse
Affiliation(s)
- Carolina Parada
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, USA.
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
39
|
Mok GF, Mohammed RH, Sweetman D. Expression of myogenic regulatory factors in chicken embryos during somite and limb development. J Anat 2015; 227:352-60. [PMID: 26183709 PMCID: PMC4560569 DOI: 10.1111/joa.12340] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2015] [Indexed: 01/24/2023] Open
Abstract
The expression of the myogenic regulatory factors (MRFs), Myf5, MyoD, myogenin (Mgn) and MRF4 have been analysed during the development of chicken embryo somites and limbs. In somites, Myf5 is expressed first in somites and paraxial mesoderm at HH stage 9 followed by MyoD at HH stage 12, and Mgn and MRF4 at HH stage 14. In older somites, Myf5 and MyoD are also expressed in the ventrally extending myotome prior to Mgn and MRF4 expression. In limb muscles a similar temporal sequence is observed with Myf5 expression detected first in forelimbs at HH stage 22, MyoD at HH stage 23, Mgn at HH stage 24 and MRF4 at HH stage 30. This report describes the precise time of onset of expression of each MRF in somites and limbs during chicken embryo development, and provides a detailed comparative timeline of MRF expression in different embryonic muscle groups.
Collapse
Affiliation(s)
- Gi Fay Mok
- School of Biosciences, University of Nottingham, Sutton Bonington, UK
| | | | - Dylan Sweetman
- School of Biosciences, University of Nottingham, Sutton Bonington, UK
| |
Collapse
|
40
|
Yang X, Shao X, Gao L, Zhang S. Systematic DNA methylation analysis of multiple cell lines reveals common and specific patterns within and across tissues of origin. Hum Mol Genet 2015; 24:4374-84. [DOI: 10.1093/hmg/ddv172] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/05/2015] [Indexed: 12/23/2022] Open
|
41
|
|
42
|
Haldar M, Karan G, Watanabe S, Guenther S, Braun T, Capecchi MR. Response: Contributions of the Myf5-independent lineage to myogenesis. Dev Cell 2015; 31:539-41. [PMID: 25490265 DOI: 10.1016/j.devcel.2014.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
In this issue, Comai et al. present evidence that previous support for Myf5-independent myogenic cell lineages was confounded by inefficiencies in lineage marking and ablation. Here, Haldar et al. discuss other possible explanations for the inconsistencies between different data sets and reiterate their views on Myf5-independent myogenesis.
Collapse
Affiliation(s)
- Malay Haldar
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Goutam Karan
- MiRx Pharmaceuticals and Invenio Therapeutics, Lexington, KY 40502, USA
| | - Shuichi Watanabe
- Max-Planck-Institute for Heart and Lung Research, Department of Cardiac Development and Remodeling, Parkstrasse 1, D-61231 Bad Nauheim, Hessen, Germany
| | - Stefan Guenther
- Max-Planck-Institute for Heart and Lung Research, Department of Cardiac Development and Remodeling, Parkstrasse 1, D-61231 Bad Nauheim, Hessen, Germany
| | - Thomas Braun
- Max-Planck-Institute for Heart and Lung Research, Department of Cardiac Development and Remodeling, Parkstrasse 1, D-61231 Bad Nauheim, Hessen, Germany
| | - Mario R Capecchi
- Howard Hughes Medical Institute, University of Utah, Salt Lake City, UT 84112, USA; Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
43
|
Zhong Z, Zhao H, Mayo J, Chai Y. Different requirements for Wnt signaling in tongue myogenic subpopulations. J Dent Res 2015; 94:421-9. [PMID: 25576472 DOI: 10.1177/0022034514566030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The tongue is a muscular organ that is essential in vertebrates for important functions, such as food intake and communication. Little is known about regulation of myogenic progenitors during tongue development when compared with the limb or trunk region. In this study, we investigated the relationship between different myogenic subpopulations and the function of canonical Wnt signaling in regulating these subpopulations. We found that Myf5- and MyoD-expressing myogenic subpopulations exist during embryonic tongue myogenesis. In the Myf5-expressing myogenic progenitors, there is a cell-autonomous requirement for canonical Wnt signaling for cell migration and differentiation. In contrast, the MyoD-expressing subpopulation does not require canonical Wnt signaling during tongue myogenesis. Taken together, our results demonstrate that canonical Wnt signaling differentially regulates the Myf5- and MyoD-expressing subpopulations during tongue myogenesis.
Collapse
Affiliation(s)
- Z Zhong
- Department of Orthodontics, School of Stomatology, Peking University, Beijing, China Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - H Zhao
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - J Mayo
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Y Chai
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
44
|
Sasi SP, Rahimi L, Yan X, Silver M, Qin G, Losordo DW, Kishore R, Goukassian DA. Genetic deletion of TNFR2 augments inflammatory response and blunts satellite-cell-mediated recovery response in a hind limb ischemia model. FASEB J 2014; 29:1208-19. [PMID: 25466901 DOI: 10.1096/fj.14-249813] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 11/12/2014] [Indexed: 01/09/2023]
Abstract
We have previously shown that TNF-tumor necrosis factor receptor-2/p75 (TNFR2/p75) signaling plays a critical role in ischemia-induced neovascularization in skeletal muscle and heart tissues. To determine the role of TNF-TNFR2/p75 signaling in ischemia-induced inflammation and muscle regeneration, we subjected wild-type (WT) and TNFR2/p75 knockout (p75KO) mice to hind limb ischemia (HLI) surgery. Ischemia induced significant and long-lasting inflammation associated with considerable decrease in satellite-cell activation in p75KO muscle tissue up to 10 d after HLI surgery. To determine the possible additive negative roles of tissue aging and the absence of TNFR2/p75, either in the tissue or in the bone marrow (BM), we generated 2 chimeric BM transplantation (BMT) models where both young green fluorescent protein (GFP)-positive p75KO and WT BM-derived cells were transplanted into adult p75KO mice. HLI surgery was performed 1 mo after BMT, after confirming complete engraftment of the recipient BM with GFP donor cells. In adult p75KO with the WT-BMT, proliferative (Ki67(+)) cells were detected only by d 28 and were exclusively GFP(+), suggesting significantly delayed contribution of young WT-BM cell to adult p75KO ischemic tissue recovery. No GFP(+) young p75KO BM cells survived in adult p75KO tissue, signifying the additive negative roles of tissue aging combined with decreased/absent TNFR2/p75 signaling in postischemic recovery.
Collapse
Affiliation(s)
- Sharath P Sasi
- Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts, USA
| | - Layla Rahimi
- Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts, USA
| | - Xinhua Yan
- Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts, USA; Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Marcy Silver
- Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts, USA
| | - Gangjian Qin
- Feinberg Cardiovascular Institute, Feinberg School of Medicine Northwestern University, Chicago, Illinois, USA; and
| | - Douglas W Losordo
- Feinberg Cardiovascular Institute, Feinberg School of Medicine Northwestern University, Chicago, Illinois, USA; and
| | - Raj Kishore
- Center for Translational Medicine, Temple University School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - David A Goukassian
- Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts, USA; Tufts University School of Medicine, Boston, Massachusetts, USA;
| |
Collapse
|
45
|
Comai G, Sambasivan R, Gopalakrishnan S, Tajbakhsh S. Variations in the Efficiency of Lineage Marking and Ablation Confound Distinctions between Myogenic Cell Populations. Dev Cell 2014; 31:654-67. [DOI: 10.1016/j.devcel.2014.11.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 06/16/2014] [Accepted: 11/04/2014] [Indexed: 11/24/2022]
|
46
|
Francetic T, Li Q. Skeletal myogenesis and Myf5 activation. Transcription 2014; 2:109-114. [PMID: 21922054 DOI: 10.4161/trns.2.3.15829] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 04/12/2011] [Accepted: 04/13/2011] [Indexed: 11/19/2022] Open
Abstract
Myogenic regulatory factors (MRFs) are the master regulators of skeletal myogenesis. Among the MRFs, Myf5 is the earliest to be expressed and is regulated by a complex set of enhancers. The expression of Myf5 defines different muscle populations in the somite. Furthermore, Myf5 expression is also found in non-muscle tissues, such as preadipocytes and neurons. Here, we present a current view on the regulation of skeletal myogenesis by transcription factors and cellular signals, with an emphasis on the complexity of transcriptional activation of Myf5. We also discuss Myf5 expression in different populations of myoblasts, preadipocytes and neuronal tissue.
Collapse
Affiliation(s)
- Tanja Francetic
- Department of Cellular and Molecular Medicine; Faculty of Medicine; University of Ottawa; Ottawa, ON Canada
| | | |
Collapse
|
47
|
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: 18] [Impact Index Per Article: 1.6] [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
|
48
|
Stafford DA, Monica SD, Harland RM. Follistatin interacts with Noggin in the development of the axial skeleton. Mech Dev 2014; 131:78-85. [PMID: 24514266 DOI: 10.1016/j.mod.2013.10.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 09/25/2013] [Accepted: 10/07/2013] [Indexed: 10/26/2022]
Abstract
When compared to single mutants for Follistatin or Noggin, we find that double mutants display a dramatic further reduction in trunk cartilage formation, particularly in the vertebral bodies and proximal ribs. Consistent with these observations, expression of the early sclerotome markers Pax1 and Uncx is diminished in Noggin;Follistatin compound mutants. In contrast, Sim1 expression expands medially in double mutants. As the onset of Follistatin expression coincides with sclerotome specification, we argue that the effect of Follistatin occurs after sclerotome induction. We hypothesize that Follistatin aids in maintaining proper somite size, and consequently sclerotome progenitor numbers, by preventing paraxial mesoderm from adopting an intermediate/lateral plate mesodermal fate in the Noggin-deficient state.
Collapse
|
49
|
Genxi Z, Ying T, Tao Z, Jinyu W, Yongjuan W. Expression profiles and association analysis with growth traits of the MyoG and Myf5 genes in the Jinghai yellow chicken. Mol Biol Rep 2014; 41:7331-8. [PMID: 25098599 DOI: 10.1007/s11033-014-3619-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 07/16/2014] [Indexed: 11/26/2022]
Abstract
In order to analyze the association of single nucleotide polymorphisms (SNPs) in the MyoG and Myf5 genes with chicken growth traits, PCR-SSCP approach was used to detect the (SNPs). The general linear model was used to analyze gene interaction and genetic effects between different genotypes and growth traits of the Jinghai yellow chicken. For the MyoG gene, three genotypes (AA, AB and BB) were detected in the Jinghai yellow chicken population. Gene sequencing revealed one mutation (T36C) in the genotype BB in comparison to the genotype AA. For the Myf5 gene, three genotypes (CC, CD and DD) were detected in the Jinghai yellow chicken population. Gene sequencing revealed one mutation (A1313G) in the genotype DD in comparison to the genotype CC. Gene interaction effect has significant influence on 6, 8-week-weight and 300-day-weight. The least square analysis showed that individuals with BB genotype of the MyoG gene had higher bodyweight at 2, 4, 10, 12, 14 and 16 weeks compared to individuals with AA and AB genotypes. Individuals with CD genotype of the Myf5 gene had higher birth weight than individuals with CC genotype (P < 0.05). The interactive genotype AB*DD performs well at 6, 8 weeks and 300 days bodyweight. The results suggested that SNPs of the MyoG and Myf5 genes had certain effects on growth traits of the Jinghai yellow chicken.
Collapse
Affiliation(s)
- Zhang Genxi
- College of Animal Science and Technology, Yangzhou University, Jiangsu Yangzhou, 225009, China,
| | | | | | | | | |
Collapse
|
50
|
Grassot V, Da Silva A, Saliba J, Maftah A, Dupuy F, Petit JM. Highlights of glycosylation and adhesion related genes involved in myogenesis. BMC Genomics 2014; 15:621. [PMID: 25051993 PMCID: PMC4223822 DOI: 10.1186/1471-2164-15-621] [Citation(s) in RCA: 8] [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: 07/11/2014] [Accepted: 07/14/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Myogenesis is initiated by myoblast differentiation and fusion to form myotubes and muscle fibres. A population of myoblasts, known as satellite cells, is responsible for post-natal growth of muscle and for its regeneration. This differentiation requires many changes in cell behaviour and its surrounding environment. These modifications are tightly regulated over time and can be characterized through the study of changes in gene expression associated with this process. During the initial myogenesis steps, using the myoblast cell line C2C12 as a model, Janot et al. (2009) showed significant variations in expression for genes involved in pathways of glycolipid synthesis. In this study we used murine satellite cells (MSC) and their ability to differentiate into myotubes or early fat storage cells to select glycosylation related genes whose variation of expression is myogenesis specific. RESULTS The comparison of variant genes in both MSC differentiation pathways identified 67 genes associated with myogenesis. Comparison with data obtained for C2C12 revealed that only 14 genes had similar expression profiles in both cell types and that 17 genes were specifically regulated in MSC. Results were validated statistically by without a priori clustering. Classification according to protein function encoded by these 31 genes showed that the main regulated cellular processes during this differentiation were (i) remodeling of the extracellular matrix, particularly, sulfated structures, (ii) down-regulation of O-mannosyl glycan biosynthesis, and (iii) an increase in adhesion protein expression. A functional study was performed on Itga11 and Chst5 encoding two highly up-regulated proteins. The inactivation of Chst5 by specific shRNA delayed the fusion of MSC. By contrast, the inactivation of Itga11 by specific shRNA dramatically decreased the fusion ability of MSC. This result was confirmed by neutralization of Itga11 product by specific antibodies. CONCLUSIONS Our screening method detected 31 genes specific for myogenic differentiation out of the 383 genes studied. According to their function, interaction networks of the products of these selected genes converged to cell fusion. Functional studies on Itga11 and Chst5 demonstrated the robustness of this screening.
Collapse
Affiliation(s)
- Vincent Grassot
- INRA, UMR 1061 Unité de Génétique Moléculaire Animale, Université de Limoges, Faculté des Sciences et Techniques, 123 Avenue A. Thomas, Limoges 87060, France
| | - Anne Da Silva
- INRA, UMR 1061 Unité de Génétique Moléculaire Animale, Université de Limoges, Faculté des Sciences et Techniques, 123 Avenue A. Thomas, Limoges 87060, France
| | - James Saliba
- INRA, UMR 1061 Unité de Génétique Moléculaire Animale, Université de Limoges, Faculté des Sciences et Techniques, 123 Avenue A. Thomas, Limoges 87060, France
| | - Abderrahman Maftah
- INRA, UMR 1061 Unité de Génétique Moléculaire Animale, Université de Limoges, Faculté des Sciences et Techniques, 123 Avenue A. Thomas, Limoges 87060, France
| | - Fabrice Dupuy
- INRA, UMR 1061 Unité de Génétique Moléculaire Animale, Université de Limoges, Faculté des Sciences et Techniques, 123 Avenue A. Thomas, Limoges 87060, France
| | - Jean-Michel Petit
- INRA, UMR 1061 Unité de Génétique Moléculaire Animale, Université de Limoges, Faculté des Sciences et Techniques, 123 Avenue A. Thomas, Limoges 87060, France
| |
Collapse
|