1
|
Fraczek PM, Duran P, Yang BA, Ferre V, Alawieh L, Castor-Macias JA, Wong VT, Guzman SD, Piotto C, Itsani K, Larouche JA, Aguilar CA. Vitamin A retinoic acid contributes to muscle stem cell and mitochondrial function loss in old age. JCI Insight 2025; 10:e183706. [PMID: 40131371 DOI: 10.1172/jci.insight.183706] [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] [Indexed: 03/27/2025] Open
Abstract
Adult stem cells decline in number and function in old age, and identifying factors that can delay or revert age-associated adult stem cell dysfunction are vital for maintaining a healthy lifespan. Here we show that vitamin A, a micronutrient that is derived from diet and metabolized into retinoic acid, acts as an antioxidant and transcriptional regulator in muscle stem cells. We first show that obstruction of dietary vitamin A in young animals drives mitochondrial and cell cycle dysfunction in muscle stem cells that mimics old age. Next, we pharmacologically targeted retinoic acid signaling in myoblasts and aged muscle stem cells ex vivo and in vivo and observed reductions in oxidative damage, enhanced mitochondrial function, and improved maintenance of quiescence through fatty acid oxidation. We next detected that the receptor for vitamin A-derived retinol, stimulated by retinoic acid 6 or Stra6, was diminished with muscle stem cell activation and in old age. To understand the relevance of Stra6 loss, we knocked down Stra6 and observed an accumulation of mitochondrial reactive oxygen species, as well as changes in mitochondrial morphology and respiration. These results demonstrate that vitamin A regulates mitochondria and metabolism in muscle stem cells and highlight a unique mechanism connecting stem cell function with vitamin intake.
Collapse
Affiliation(s)
- Paula M Fraczek
- Department of Biomedical Engineering
- Biointerfaces Institute, and
| | - Pamela Duran
- Department of Biomedical Engineering
- Biointerfaces Institute, and
| | - Benjamin A Yang
- Department of Biomedical Engineering
- Biointerfaces Institute, and
| | - Valeria Ferre
- Department of Biomedical Engineering
- Biointerfaces Institute, and
| | - Leanne Alawieh
- Department of Biomedical Engineering
- Biointerfaces Institute, and
| | | | - Vivian T Wong
- Department of Biomedical Engineering
- Biointerfaces Institute, and
| | - Steve D Guzman
- Department of Biomedical Engineering
- Biointerfaces Institute, and
| | - Celeste Piotto
- Department of Biomedical Engineering
- Biointerfaces Institute, and
| | | | | | - Carlos A Aguilar
- Department of Biomedical Engineering
- Biointerfaces Institute, and
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
2
|
Lin WH, Tzeng CY, Kao FC, Tsao CW, Li N, Wu CC, Lee SH, Huang KF, Hu WW, Chen SL. The proliferation and differentiation of skeletal muscle stem cells are enhanced in a bioreactor. Biotechnol Bioeng 2025; 122:95-109. [PMID: 39369338 DOI: 10.1002/bit.28857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/07/2024]
Abstract
Skeletal muscle (SKM) is the largest organ in mammalian body and it can repair damages by using the residential myogenic stem cells (MuSC), but this repairing capacity reduces with age and in some genetic muscular dystrophy. Under these circumstances, artificial amplification of autologous MuSC in vitro might be necessary to repair the damaged SKM. The amplification of MuSC is highly dependent on myogenic signals, such as sonic hedgehog (Shh), Wnt3a, and fibroblast growth factors, so formulating an optimum myogenic kit composed of specific myogenic signals might increase the proliferation and differentiation of MuSC efficiently. In this study, various myogenic signals have been tested on C2C12 myoblasts and primary MuSC, and a myogenic kit consists of insulin, lithium chloride, T3, and retinoic acid has been formulated, and we found it significantly increased the fusion index and MHC expression level of both C2C12 and MuSC myotubes. A novel bioreactor providing cyclic stretching (CS) and electrical stimulation (ES) has been fabricated to enhance the myogenic differentiation of both C2C12 and MuSC. We further found that coating the bioreactor substratum with collagen gave the best effect on proliferation and differentiation of MuSC. Furthermore, combining the collagen coating and physical stimuli (CS + ES) in the bioreactor can generate more proliferative primary MuSC cells. Our results have demonstrated that the combination of myogenic kit and bioreactor can provide environment for efficient MuSC proliferation and differentiation. These MuSC and mature myotubes amplified in the bioreactor might be useful for clinical grafting into damaged SKM in the future.
Collapse
Affiliation(s)
- Wei-Hsuan Lin
- Department of Life Sciences, National Central University, Taoyuan, Taiwan
| | - Chung-Yuh Tzeng
- Department of Orthopedics, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Rehabilitation, Jen-Teh Junior College of Medicine, Nursing, and Management, Miaoli, Taiwan
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Fan-Che Kao
- Department of Life Sciences, National Central University, Taoyuan, Taiwan
| | - Chia-Wen Tsao
- Department of Mechanical Engineering, National Central University, Taoyuan, Taiwan
| | - Ning Li
- Department of Life Sciences, National Central University, Taoyuan, Taiwan
| | - Chuan-Che Wu
- Department of Life Sciences, National Central University, Taoyuan, Taiwan
| | - Sheng-Huei Lee
- Department of Electric Engineering, Chien Hsin University of Science and Technology, Taoyuan, Taiwan
| | - Kai-Fan Huang
- Department of Life Sciences, National Central University, Taoyuan, Taiwan
| | - Wei-Wen Hu
- Department of Chemical and Materials Engineering, National Central University, Taoyuan, Taiwan
| | - Shen-Liang Chen
- Department of Life Sciences, National Central University, Taoyuan, Taiwan
| |
Collapse
|
3
|
Dumas CE, Rousset C, De Bono C, Cortés C, Jullian E, Lescroart F, Zaffran S, Adachi N, Kelly RG. Retinoic acid signalling regulates branchiomeric neck muscle development at the head/trunk interface. Development 2024; 151:dev202905. [PMID: 39082789 DOI: 10.1242/dev.202905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/18/2024] [Indexed: 08/30/2024]
Abstract
Skeletal muscles of the head and trunk originate in distinct lineages with divergent regulatory programmes converging on activation of myogenic determination factors. Branchiomeric head and neck muscles share a common origin with cardiac progenitor cells in cardiopharyngeal mesoderm (CPM). The retinoic acid (RA) signalling pathway is required during a defined early time window for normal deployment of cells from posterior CPM to the heart. Here, we show that blocking RA signalling in the early mouse embryo also results in selective loss of the trapezius neck muscle, without affecting other skeletal muscles. RA signalling is required for robust expression of myogenic determination factors in posterior CPM and subsequent expansion of the trapezius primordium. Lineage-specific activation of a dominant-negative RA receptor reveals that trapezius development is not regulated by direct RA signalling to myogenic progenitor cells in CPM, or through neural crest cells, but indirectly through the somitic lineage, closely apposed with posterior CPM in the early embryo. These findings suggest that trapezius development is dependent on precise spatiotemporal interactions between cranial and somitic mesoderm at the head/trunk interface.
Collapse
Affiliation(s)
- Camille E Dumas
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | - Célia Rousset
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | | | - Claudio Cortés
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | - Estelle Jullian
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | | | - Stéphane Zaffran
- Aix-Marseille Université, INSERM, MMG U1251, 13005 Marseille, France
| | - Noritaka Adachi
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| |
Collapse
|
4
|
Fan SH, Li N, Huang KF, Chang YT, Wu CC, Chen SL. MyoD Over-Expression Rescues GST-bFGF Repressed Myogenesis. Int J Mol Sci 2024; 25:4308. [PMID: 38673893 PMCID: PMC11050597 DOI: 10.3390/ijms25084308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
During embryogenesis, basic fibroblast growth factor (bFGF) is released from neural tube and myotome to promote myogenic fate in the somite, and is routinely used for the culture of adult skeletal muscle (SKM) stem cells (MuSC, called satellite cells). However, the mechanism employed by bFGF to promote SKM lineage and MuSC proliferation has not been analyzed in detail. Furthermore, the question of if the post-translational modification (PTM) of bFGF is important to its stemness-promoting effect has not been answered. In this study, GST-bFGF was expressed and purified from E.coli, which lacks the PTM system in eukaryotes. We found that both GST-bFGF and commercially available bFGF activated the Akt-Erk pathway and had strong cell proliferation effect on C2C12 myoblasts and MuSC. GST-bFGF reversibly compromised the myogenesis of C2C12 myoblasts and MuSC, and it increased the expression of Myf5, Pax3/7, and Cyclin D1 but strongly repressed that of MyoD, suggesting the maintenance of myogenic stemness amid repressed MyoD expression. The proliferation effect of GST-bFGF was conserved in C2C12 over-expressed with MyoD (C2C12-tTA-MyoD), implying its independence of the down-regulation of MyoD. In addition, the repressive effect of GST-bFGF on myogenic differentiation was almost totally rescued by the over-expression of MyoD. Together, these evidences suggest that (1) GST-bFGF and bFGF have similar effects on myogenic cell proliferation and differentiation, and (2) GST-bFGF can promote MuSC stemness and proliferation by differentially regulating MRFs and Pax3/7, (3) MyoD repression by GST-bFGF is reversible and independent of the proliferation effect, and (4) GST-bFGF can be a good substitute for bFGF in sustaining MuSC stemness and proliferation.
Collapse
Affiliation(s)
| | | | | | | | | | - Shen-Liang Chen
- Department of Life Sciences, National Central University, Jhongli 32001, Taiwan; (S.-H.F.); (N.L.); (K.-F.H.); (Y.-T.C.); (C.-C.W.)
| |
Collapse
|
5
|
Chen SL, Wu CC, Li N, Weng TH. Post-transcriptional regulation of myogenic transcription factors during muscle development and pathogenesis. J Muscle Res Cell Motil 2024; 45:21-39. [PMID: 38206489 DOI: 10.1007/s10974-023-09663-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/29/2023] [Indexed: 01/12/2024]
Abstract
The transcriptional regulation of skeletal muscle (SKM) development (myogenesis) has been documented for over 3 decades and served as a paradigm for tissue-specific cell type determination and differentiation. Myogenic stem cells (MuSC) in embryos and adult SKM are regulated by the transcription factors Pax3 and Pax7 for their stem cell characteristics, while their lineage determination and terminal differentiation are both dictated by the myogenic regulatory factors (MRF) that comprise Mrf4, Myf5, Myogenin, and MyoD. The myocyte enhancer factor Mef2c is activated by MRF during terminal differentiation and collaborates with them to promote myoblast fusion and differentiation. Recent studies have found critical regulation of these myogenic transcription factors at mRNA level, including subcellular localization, stability, and translational regulation. Therefore, the regulation of Pax3/7, MRFs and Mef2c mRNAs by RNA-binding factors and non-coding RNAs (ncRNA), including microRNAs and long non-coding RNAs (lncRNA), will be the focus of this review and the impact of this regulation on myogenesis will be further addressed. Interestingly, the stem cell characteristics of MuSC has been found to be critically regulated by ncRNAs, implying the involvement of ncRNAs in SKM homeostasis and regeneration. Current studies have further identified that some ncRNAs are implicated in the etiology of some SKM diseases and can serve as valuable tools/indicators for prediction of prognosis. The roles of ncRNAs in the MuSC biology and SKM disease etiology will also be discussed in this review.
Collapse
Affiliation(s)
- Shen-Liang Chen
- Department of Life Sciences, National Central University, 300 Jhongda Rd, Jhongli, 32001, Taiwan.
| | - Chuan-Che Wu
- Department of Life Sciences, National Central University, 300 Jhongda Rd, Jhongli, 32001, Taiwan
| | - Ning Li
- Department of Life Sciences, National Central University, 300 Jhongda Rd, Jhongli, 32001, Taiwan
| | - Tzu-Han Weng
- Department of Life Sciences, National Central University, 300 Jhongda Rd, Jhongli, 32001, Taiwan
| |
Collapse
|
6
|
Zhang W, Peng Q, Zhang X, Guo J, Tong H, Li S. Vitamin A Promotes the Repair of Mice Skeletal Muscle Injury through RARα. Nutrients 2023; 15:3674. [PMID: 37686706 PMCID: PMC10490340 DOI: 10.3390/nu15173674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Vitamin A (VitA) is an important fat-soluble vitamin which plays an important role in cell growth and individual development. However, the effect of VitA on the repair process of muscle injury and its molecular mechanism are still unclear. In this study, VitA and RA were first added to the culture medium of differentiated cells. We then detected cell differentiation marker proteins and myotube fusion. Moreover, the effects of VitA on RARα expression and nuclear translocation were further examined. The results showed that VitA significantly promoted the differentiation of C2C12, and the expression of RARα was significantly increased. Furthermore, VitA was injected into skeletal muscle injury in mice. HE staining and Western Blot results showed that VitA could significantly accelerate the repair of skeletal muscle injury and VitA increase the expression of RARα in mice. This study provides a theoretical basis for elucidating the regulation mechanism of VitA-mediated muscle development and the development of therapeutic drugs for muscle diseases in animals.
Collapse
Affiliation(s)
- Wenjia Zhang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (W.Z.); (Q.P.); (X.Z.); (J.G.); (H.T.)
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin 150030, China
| | - Qingyun Peng
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (W.Z.); (Q.P.); (X.Z.); (J.G.); (H.T.)
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin 150030, China
| | - Xiaoyu Zhang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (W.Z.); (Q.P.); (X.Z.); (J.G.); (H.T.)
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin 150030, China
| | - Jiaxu Guo
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (W.Z.); (Q.P.); (X.Z.); (J.G.); (H.T.)
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin 150030, China
| | - Huili Tong
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (W.Z.); (Q.P.); (X.Z.); (J.G.); (H.T.)
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin 150030, China
| | - Shuang Li
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (W.Z.); (Q.P.); (X.Z.); (J.G.); (H.T.)
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
7
|
Eigler T, Zarfati G, Amzallag E, Sinha S, Segev N, Zabary Y, Zaritsky A, Shakked A, Umansky KB, Schejter ED, Millay DP, Tzahor E, Avinoam O. ERK1/2 inhibition promotes robust myotube growth via CaMKII activation resulting in myoblast-to-myotube fusion. Dev Cell 2021; 56:3349-3363.e6. [PMID: 34932950 PMCID: PMC8693863 DOI: 10.1016/j.devcel.2021.11.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 07/28/2021] [Accepted: 11/21/2021] [Indexed: 11/19/2022]
Abstract
Myoblast fusion is essential for muscle development and regeneration. Yet, it remains poorly understood how mononucleated myoblasts fuse with preexisting fibers. We demonstrate that ERK1/2 inhibition (ERKi) induces robust differentiation and fusion of primary mouse myoblasts through a linear pathway involving RXR, ryanodine receptors, and calcium-dependent activation of CaMKII in nascent myotubes. CaMKII activation results in myotube growth via fusion with mononucleated myoblasts at a fusogenic synapse. Mechanistically, CaMKII interacts with and regulates MYMK and Rac1, and CaMKIIδ/γ knockout mice exhibit smaller regenerated myofibers following injury. In addition, the expression of a dominant negative CaMKII inhibits the formation of large multinucleated myotubes. Finally, we demonstrate the evolutionary conservation of the pathway in chicken myoblasts. We conclude that ERK1/2 represses a signaling cascade leading to CaMKII-mediated fusion of myoblasts to myotubes, providing an attractive target for the cultivated meat industry and regenerative medicine.
Collapse
Affiliation(s)
- Tamar Eigler
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Giulia Zarfati
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Emmanuel Amzallag
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Sansrity Sinha
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Nadav Segev
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Yishaia Zabary
- Department of Software & Information Systems Engineering, Ben Gurion University, Be'er Sheva, Israel
| | - Assaf Zaritsky
- Department of Software & Information Systems Engineering, Ben Gurion University, Be'er Sheva, Israel
| | - Avraham Shakked
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Kfir-Baruch Umansky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal D Schejter
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Douglas P Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Ori Avinoam
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
8
|
Retinoic acid exerts sexually dimorphic effects on muscle energy metabolism and function. J Biol Chem 2021; 297:101101. [PMID: 34419449 PMCID: PMC8441203 DOI: 10.1016/j.jbc.2021.101101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/06/2021] [Accepted: 08/18/2021] [Indexed: 01/24/2023] Open
Abstract
The retinol dehydrogenase Rdh10 catalyzes the rate-limiting reaction that converts retinol into retinoic acid (RA), an autacoid that regulates energy balance and reduces adiposity. Skeletal muscle contributes to preventing adiposity, by consuming nearly half the energy of a typical human. We report sexually dimorphic differences in energy metabolism and muscle function in Rdh10+/- mice. Relative to wild-type (WT) controls, Rdh10+/- males fed a high-fat diet decrease reliance on fatty-acid oxidation and experience glucose intolerance and insulin resistance. Running endurance decreases 40%. Rdh10+/- females fed this diet increase fatty acid oxidation and experience neither glucose intolerance nor insulin resistance. Running endurance increases 220%. We therefore assessed RA function in the mixed-fiber type gastrocnemius muscles (GM), which contribute to running, rather than standing, and are similar to human GM. RA levels in Rdh10+/- male GM decrease 38% relative to WT. Rdh10+/- male GM increase expression of Myog and reduce Eif6 mRNAs, which reduce and enhance running endurance, respectively. Cox5A, complex IV activity, and ATP decrease. Increased centralized nuclei reveal existence of muscle malady and/or repair in GM fibers. Comparatively, RA in Rdh10+/- female GM decreases by less than half the male decrease, from a more modest decrease in Rdh10 and an increase in the estrogen-induced retinol dehydrogenase Dhrs9. Myog mRNA decreases. Cox5A, complex IV activity, and ATP increase. Centralized GM nuclei do not increase. We conclude that Rdh10/RA affects whole body energy use and insulin resistance partially through sexual dimorphic effects on skeletal muscle gene expression, structure, and mitochondria activity.
Collapse
|
9
|
Etienne J, Joanne P, Catelain C, Riveron S, Bayer AC, Lafable J, Punzon I, Blot S, Agbulut O, Vilquin JT. Aldehyde dehydrogenases contribute to skeletal muscle homeostasis in healthy, aging, and Duchenne muscular dystrophy patients. J Cachexia Sarcopenia Muscle 2020; 11:1047-1069. [PMID: 32157826 PMCID: PMC7432589 DOI: 10.1002/jcsm.12557] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 12/12/2019] [Accepted: 01/30/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Aldehyde dehydrogenases (ALDHs) are key players in cell survival, protection, and differentiation via the metabolism and detoxification of aldehydes. ALDH activity is also a marker of stem cells. The skeletal muscle contains populations of ALDH-positive cells amenable to use in cell therapy, whose distribution, persistence in aging, and modifications in myopathic context have not been investigated yet. METHODS The Aldefluor® (ALDEF) reagent was used to assess the ALDH activity of muscle cell populations, whose phenotypic characterizations were deepened by flow cytometry. The nature of ALDH isoenzymes expressed by the muscle cell populations was identified in complementary ways by flow cytometry, immunohistology, and real-time PCR ex vivo and in vitro. These populations were compared in healthy, aging, or Duchenne muscular dystrophy (DMD) patients, healthy non-human primates, and Golden Retriever dogs (healthy vs. muscular dystrophic model, Golden retriever muscular dystrophy [GRMD]). RESULTS ALDEF+ cells persisted through muscle aging in humans and were equally represented in several anatomical localizations in healthy non-human primates. ALDEF+ cells were increased in dystrophic individuals in humans (nine patients with DMD vs. five controls: 14.9 ± 1.63% vs. 3.6 ± 0.39%, P = 0.0002) and dogs (three GRMD dogs vs. three controls: 10.9 ± 2.54% vs. 3.7 ± 0.45%, P = 0.049). In DMD patients, such increase was due to the adipogenic ALDEF+ /CD34+ populations (11.74 ± 1.5 vs. 2.8 ± 0.4, P = 0.0003), while in GRMD dogs, it was due to the myogenic ALDEF+ /CD34- cells (3.6 ± 0.6% vs. 1.03 ± 0.23%, P = 0.0165). Phenotypic characterization associated the ALDEF+ /CD34- cells with CD9, CD36, CD49a, CD49c, CD49f, CD106, CD146, and CD184, some being associated with myogenic capacities. Cytological and histological analyses distinguished several ALDH isoenzymes (ALDH1A1, 1A2, 1A3, 1B1, 1L1, 2, 3A1, 3A2, 3B1, 3B2, 4A1, 7A1, 8A1, and 9A1) expressed by different cell populations in the skeletal muscle tissue belonging to multinucleated fibres, or myogenic, endothelial, interstitial, and neural lineages, designing them as potential new markers of cell type or of metabolic activity. Important modifications were noted in isoenzyme expression between healthy and DMD muscle tissues. The level of gene expression of some isoenzymes (ALDH1A1, 1A3, 1B1, 2, 3A2, 7A1, 8A1, and 9A1) suggested their specific involvement in muscle stability or regeneration in situ or in vitro. CONCLUSIONS This study unveils the importance of the ALDH family of isoenzymes in the skeletal muscle physiology and homeostasis, suggesting their roles in tissue remodelling in the context of muscular dystrophies.
Collapse
Affiliation(s)
- Jessy Etienne
- Sorbonne Université, INSERM, AIM, Centre de Recherche en Myologie, UMRS 974, AP-HP, Hôpital Pitié Salpêtrière, Paris, France.,Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, USA
| | - Pierre Joanne
- Sorbonne Université, CNRS, INSERM, Institut de Biologie Paris-Seine, IBPS, UMR 8256 Biological Adaptation and Ageing, Paris, France
| | - Cyril Catelain
- Sorbonne Université, INSERM, AIM, Centre de Recherche en Myologie, UMRS 974, AP-HP, Hôpital Pitié Salpêtrière, Paris, France
| | - Stéphanie Riveron
- Sorbonne Université, INSERM, AIM, Centre de Recherche en Myologie, UMRS 974, AP-HP, Hôpital Pitié Salpêtrière, Paris, France
| | - Alexandra Clarissa Bayer
- Sorbonne Université, INSERM, AIM, Centre de Recherche en Myologie, UMRS 974, AP-HP, Hôpital Pitié Salpêtrière, Paris, France
| | - Jérémy Lafable
- Sorbonne Université, INSERM, AIM, Centre de Recherche en Myologie, UMRS 974, AP-HP, Hôpital Pitié Salpêtrière, Paris, France
| | - Isabel Punzon
- Université Paris-Est Créteil, INSERM, Institut Mondor de Recherche Biomédicale, IMRB, École Nationale Vétérinaire d'Alfort, ENVA, U955-E10, Maisons-Alfort, France
| | - Stéphane Blot
- Université Paris-Est Créteil, INSERM, Institut Mondor de Recherche Biomédicale, IMRB, École Nationale Vétérinaire d'Alfort, ENVA, U955-E10, Maisons-Alfort, France
| | - Onnik Agbulut
- Sorbonne Université, CNRS, INSERM, Institut de Biologie Paris-Seine, IBPS, UMR 8256 Biological Adaptation and Ageing, Paris, France
| | - Jean-Thomas Vilquin
- Sorbonne Université, INSERM, AIM, Centre de Recherche en Myologie, UMRS 974, AP-HP, Hôpital Pitié Salpêtrière, Paris, France
| |
Collapse
|
10
|
Lee MP, Ratner N, Yutzey KE. Genome-wide Twist1 occupancy in endocardial cushion cells, embryonic limb buds, and peripheral nerve sheath tumor cells. BMC Genomics 2014; 15:821. [PMID: 25262113 PMCID: PMC4190347 DOI: 10.1186/1471-2164-15-821] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 09/22/2014] [Indexed: 11/10/2022] Open
Abstract
Background The basic helix-loop-helix transcription factor Twist1 has well-documented roles in progenitor populations of the developing embryo, including endocardial cushions (ECC) and limb buds, and also in cancer. Whether Twist1 regulates the same transcriptional targets in different tissue types is largely unknown. Results The tissue-specificity of Twist1 genomic occupancy was examined in mouse ECCs, limb buds, and peripheral nerve sheath tumor (PNST) cells using chromatin immunoprecipitation followed by sequencing (Chip-seq) analysis. Consistent with known Twist1 functions during development and in cancer cells, Twist1-DNA binding regions associated with genes related to cell migration and adhesion were detected in all three tissues. However, the vast majority of Twist1 binding regions were specific to individual tissue types. Thus, while Twist1 has similar functions in ECCs, limb buds, and PNST cells, the specific genomic sequences occupied by Twist1 were different depending on cellular context. Subgroups of shared genes, also predominantly related to cell adhesion and migration, were identified in pairwise comparisons of ECC, limb buds and PNST cells. Twist1 genomic occupancy was detected for six binding regions in all tissue types, and Twist1-binding sequences associated with Chst11, Litaf, Ror2, and Spata5 also bound the potential Twist1 cofactor RREB1. Pathway analysis of the genes associated with Twist1 binding suggests that Twist1 may regulate genes associated with the Wnt signaling pathway in ECCs and limb buds. Conclusions Together, these data indicate that Twist1 interacts with genes that regulate adhesion and migration in different tissues, potentially through distinct sets of target genes. In addition, there is a small subset of genes occupied by Twist1 in all three tissues that may represent a core group of Twist1 target genes in multiple cell types. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-821) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | - Katherine E Yutzey
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA.
| |
Collapse
|
11
|
Voronova A, Coyne E, Al Madhoun A, Fair JV, Bosiljcic N, St-Louis C, Li G, Thurig S, Wallace VA, Wiper-Bergeron N, Skerjanc IS. Hedgehog signaling regulates MyoD expression and activity. J Biol Chem 2013; 288:4389-4404. [PMID: 23266826 PMCID: PMC3567689 DOI: 10.1074/jbc.m112.400184] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 12/12/2012] [Indexed: 11/06/2022] Open
Abstract
The inhibition of MyoD expression is important for obtaining muscle progenitors that can replenish the satellite cell niche during muscle repair. Progenitors could be derived from either embryonic stem cells or satellite cells. Hedgehog (Hh) signaling is important for MyoD expression during embryogenesis and adult muscle regeneration. To date, the mechanistic understanding of MyoD regulation by Hh signaling is unclear. Here, we demonstrate that the Hh effector, Gli2, regulates MyoD expression and associates with MyoD gene elements. Gain- and loss-of-function experiments in pluripotent P19 cells show that Gli2 activity is sufficient and required for efficient MyoD expression during skeletal myogenesis. Inhibition of Hh signaling reduces MyoD expression during satellite cell activation in vitro. In addition to regulating MyoD expression, Hh signaling regulates MyoD transcriptional activity, and MyoD activates Hh signaling in myogenic conversion assays. Finally, Gli2, MyoD, and MEF2C form a protein complex, which enhances MyoD activity on skeletal muscle-related promoters. We therefore link Hh signaling to the function and expression of MyoD protein during myogenesis in stem cells.
Collapse
Affiliation(s)
| | - Erin Coyne
- From the Department of Biochemistry, Microbiology, and Immunology and
| | - Ashraf Al Madhoun
- From the Department of Biochemistry, Microbiology, and Immunology and
- Pancreatic Islet Biology and Transplantation Unit, Dasman Diabetes Institute, Dasman 15462, Kuwait and
| | - Joel V. Fair
- From the Department of Biochemistry, Microbiology, and Immunology and
| | - Neven Bosiljcic
- From the Department of Biochemistry, Microbiology, and Immunology and
| | - Catherine St-Louis
- the Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa and
| | - Grace Li
- the Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa and
| | - Sherry Thurig
- From the Department of Biochemistry, Microbiology, and Immunology and
- Ottawa Hospital Research Institute, Ottawa K1H 8M5, Canada
| | - Valerie A. Wallace
- From the Department of Biochemistry, Microbiology, and Immunology and
- Ottawa Hospital Research Institute, Ottawa K1H 8M5, Canada
| | - Nadine Wiper-Bergeron
- the Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa and
| | - Ilona S. Skerjanc
- From the Department of Biochemistry, Microbiology, and Immunology and
| |
Collapse
|
12
|
El Haddad M, Jean E, Turki A, Hugon G, Vernus B, Bonnieu A, Passerieux E, Hamade A, Mercier J, Laoudj-Chenivesse D, Carnac G. Glutathione peroxidase 3, a new retinoid target gene, is crucial for human skeletal muscle precursor cell survival. J Cell Sci 2012; 125:6147-56. [PMID: 23132926 DOI: 10.1242/jcs.115220] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Protection of satellite cells from cytotoxic damages is crucial to ensure efficient adult skeletal muscle regeneration and to improve therapeutic efficacy of cell transplantation in degenerative skeletal muscle diseases. It is therefore important to identify and characterize molecules and their target genes that control the viability of muscle stem cells. Recently, we demonstrated that high aldehyde dehydrogenase activity is associated with increased viability of human myoblasts. In addition to its detoxifying activity, aldehyde dehydrogenase can also catalyze the irreversible oxidation of vitamin A to retinoic acid; therefore, we examined whether retinoic acid is important for myoblast viability. We showed that when exposed to oxidative stress induced by hydrogen peroxide, adherent human myoblasts entered apoptosis and lost their capacity for adhesion. Pre-treatment with retinoic acid reduced the cytotoxic damage ex vivo and enhanced myoblast survival in transplantation assays. The effects of retinoic acid were maintained in dystrophic myoblasts derived from facioscapulohumeral patients. RT-qPCR analysis of antioxidant gene expression revealed glutathione peroxidase 3 (Gpx3), a gene encoding an antioxidant enzyme, as a potential retinoic acid target gene in human myoblasts. Knockdown of Gpx3 using short interfering RNA induced elevation in reactive oxygen species and cell death. The anti-cytotoxic effects of retinoic acid were impaired in GPx3-inactivated myoblasts, which indicates that GPx3 regulates the antioxidative effects of retinoic acid. Therefore, retinoid status and GPx3 levels may have important implications for the viability of human muscle stem cells.
Collapse
Affiliation(s)
- Marina El Haddad
- Inserm U1046, Université Montpellier 1, 34295 Montpellier, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Vella JB, Thompson SD, Bucsek MJ, Song M, Huard J. Murine and human myogenic cells identified by elevated aldehyde dehydrogenase activity: implications for muscle regeneration and repair. PLoS One 2011; 6:e29226. [PMID: 22195027 PMCID: PMC3240661 DOI: 10.1371/journal.pone.0029226] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 11/22/2011] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Despite the initial promise of myoblast transfer therapy to restore dystrophin in Duchenne muscular dystrophy patients, clinical efficacy has been limited, primarily by poor cell survival post-transplantation. Murine muscle derived stem cells (MDSCs) isolated from slowly adhering cells (SACs) via the preplate technique, induce greater muscle regeneration than murine myoblasts, primarily due to improved post-transplantation survival, which is conferred by their increased stress resistance capacity. Aldehyde dehydrogenase (ALDH) represents a family of enzymes with important morphogenic as well as oxidative damage mitigating roles and has been found to be a marker of stem cells in both normal and malignant tissue. In this study, we hypothesized that elevated ALDH levels could identify murine and human muscle derived cell (hMDC) progenitors, endowed with enhanced stress resistance and muscle regeneration capacity. METHODOLOGY/PRINCIPAL FINDINGS Skeletal muscle progenitors were isolated from murine and human skeletal muscle by a modified preplate technique and unfractionated enzymatic digestion, respectively. ALDH(hi) subpopulations isolated by fluorescence activate cell sorting demonstrated increased proliferation and myogenic differentiation capacities compared to their ALDH(lo) counterparts when cultivated in oxidative and inflammatory stress media conditions. This behavior correlated with increased intracellular levels of reduced glutathione and superoxide dismutase. ALDH(hi) murine myoblasts were observed to exhibit an increased muscle regenerative potential compared to ALDH(lo) myoblasts, undergo multipotent differentiation (osteogenic and chondrogenic), and were found predominately in the SAC fraction, characteristics that are also observed in murine MDSCs. Likewise, human ALDH(hi) hMDCs demonstrated superior muscle regenerative capacity compared to ALDH(lo) hMDCs. CONCLUSIONS The methodology of isolating myogenic cells on the basis of elevated ALDH activity yielded cells with increased stress resistance, a behavior that conferred increased regenerative capacity of dystrophic murine skeletal muscle. This result demonstrates the critical role of stress resistance in myogenic cell therapy as well as confirms the role of ALDH as a marker for rapid isolation of murine and human myogenic progenitors for cell therapy.
Collapse
Affiliation(s)
- Joseph B. Vella
- Department of Orthopedic Surgery, Stem Cell Research Center, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Seth D. Thompson
- Department of Orthopedic Surgery, Stem Cell Research Center, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mark J. Bucsek
- Department of Orthopedic Surgery, Stem Cell Research Center, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Minjung Song
- Department of Orthopedic Surgery, Stem Cell Research Center, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Johnny Huard
- Department of Orthopedic Surgery, Stem Cell Research Center, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- McGowen Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
14
|
Hayot M, Rodriguez J, Vernus B, Carnac G, Jean E, Allen D, Goret L, Obert P, Candau R, Bonnieu A. Myostatin up-regulation is associated with the skeletal muscle response to hypoxic stimuli. Mol Cell Endocrinol 2011; 332:38-47. [PMID: 20884321 DOI: 10.1016/j.mce.2010.09.008] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2010] [Revised: 09/16/2010] [Accepted: 09/16/2010] [Indexed: 10/19/2022]
Abstract
Myostatin and hypoxia signalling pathways are able to induce skeletal muscle atrophy, but whether a relationship between these two pathways exists is currently unknown. Here, we tested the hypothesis that a potential mechanism for hypoxia effect on skeletal muscle may be through regulation of myostatin. We reported an induction of myostatin expression in muscles of rats exposed to chronic hypoxia. Interestingly, we also demonstrated increased skeletal muscle myostatin protein expression in skeletal muscle of hypoxemic patients with severe chronic obstructive pulmonary disease (COPD). Parallel studies in human skeletal muscle cell cultures showed that induction of myostatin expression in myotubes treated with hypoxia-mimicking agent such as cobalt chloride (CoCl(2)) is associated with myotube atrophy. Furthermore, we demonstrated that inhibition of myostatin by means of genetic deletion of myostatin or treatment with blocking antimyostatin antibodies inhibits the CoCl(2)-induced atrophy in muscle cells. Finally, addition of recombinant myostatin restored the CoCl(2)-induced atrophy in myostatin deficient myotubes. These results strongly suggest that myostatin can play an essential role in the adaptation of skeletal muscle to hypoxic environment.
Collapse
Affiliation(s)
- Maurice Hayot
- INSERM, ERI 25-Muscle et Pathologies, Hôpital Arnaud de Villeneuve, Bât. A Craste de Paulet, F-34295 Montpellier, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Kennedy KAM, Porter T, Mehta V, Ryan SD, Price F, Peshdary V, Karamboulas C, Savage J, Drysdale TA, Li SC, Bennett SAL, Skerjanc IS. Retinoic acid enhances skeletal muscle progenitor formation and bypasses inhibition by bone morphogenetic protein 4 but not dominant negative beta-catenin. BMC Biol 2009; 7:67. [PMID: 19814781 PMCID: PMC2764571 DOI: 10.1186/1741-7007-7-67] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Accepted: 10/08/2009] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Understanding stem cell differentiation is essential for the future design of cell therapies. While retinoic acid (RA) is the most potent small molecule enhancer of skeletal myogenesis in stem cells, the stage and mechanism of its function has not yet been elucidated. Further, the intersection of RA with other signalling pathways that stimulate or inhibit myogenesis (such as Wnt and BMP4, respectively) is unknown. Thus, the purpose of this study is to examine the molecular mechanisms by which RA enhances skeletal myogenesis and interacts with Wnt and BMP4 signalling during P19 or mouse embryonic stem (ES) cell differentiation. RESULTS Treatment of P19 or mouse ES cells with low levels of RA led to an enhancement of skeletal myogenesis by upregulating the expression of the mesodermal marker, Wnt3a, the skeletal muscle progenitor factors Pax3 and Meox1, and the myogenic regulatory factors (MRFs) MyoD and myogenin. By chromatin immunoprecipitation, RA receptors (RARs) bound directly to regulatory regions in the Wnt3a, Pax3, and Meox1 genes and RA activated a beta-catenin-responsive promoter in aggregated P19 cells. In the presence of a dominant negative beta-catenin/engrailed repressor fusion protein, RA could not bypass the inhibition of skeletal myogenesis nor upregulate Meox1 or MyoD. Thus, RA functions both upstream and downstream of Wnt signalling. In contrast, it functions downstream of BMP4, as it abrogates BMP4 inhibition of myogenesis and Meox1, Pax3, and MyoD expression. Furthermore, RA downregulated BMP4 expression and upregulated the BMP4 inhibitor, Tob1. Finally, RA inhibited cardiomyogenesis but not in the presence of BMP4. CONCLUSION RA can enhance skeletal myogenesis in stem cells at the muscle specification/progenitor stage by activating RARs bound directly to mesoderm and skeletal muscle progenitor genes, activating beta-catenin function and inhibiting bone morphogenetic protein (BMP) signalling. Thus, a signalling pathway can function at multiple levels to positively regulate a developmental program and can function by abrogating inhibitory pathways. Finally, since RA enhances skeletal muscle progenitor formation, it will be a valuable tool for designing future stem cell therapies.
Collapse
Affiliation(s)
- Karen AM Kennedy
- Department of Biochemistry, Medical Sciences Building, The University of Western Ontario, London, Ontario, Canada
| | - Tammy Porter
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Virja Mehta
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Scott D Ryan
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada,Neural Regeneration Laboratory and Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Feodor Price
- Ottawa Health Research Institute, Molecular Medicine Program, Ottawa, Ontario, Canada
| | - Vian Peshdary
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada,Neural Regeneration Laboratory and Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Christina Karamboulas
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada,Department of Biochemistry, Medical Sciences Building, The University of Western Ontario, London, Ontario, Canada
| | - Josée Savage
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Thomas A Drysdale
- Department of Pediatrics and Physiology and Pharmacology, The University of Western Ontario, Children's Health Research Institute, London, Ontario, Canada
| | - Shun-Cheng Li
- Department of Biochemistry, Medical Sciences Building, The University of Western Ontario, London, Ontario, Canada
| | - Steffany AL Bennett
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada,Neural Regeneration Laboratory and Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Ilona S Skerjanc
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada,Department of Biochemistry, Medical Sciences Building, The University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
16
|
Lee S, Lee B, Lee JW, Lee SK. Retinoid signaling and neurogenin2 function are coupled for the specification of spinal motor neurons through a chromatin modifier CBP. Neuron 2009; 62:641-54. [PMID: 19524524 PMCID: PMC2705669 DOI: 10.1016/j.neuron.2009.04.025] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Revised: 03/08/2009] [Accepted: 04/30/2009] [Indexed: 11/15/2022]
Abstract
Extracellular signals and cell-intrinsic transcription factors cooperatively instruct generation of diverse neurons. However, little is known about how neural progenitors integrate both cues and orchestrate chromatin changes for neuronal specification. Here, we report that extrinsic signal retinoic acid (RA) and intrinsic transcription factor Neurogenin2 (Ngn2) collaboratively trigger transcriptionally active chromatin in spinal motor neuron genes during development. Retinoic acid receptor (RAR) binds Ngn2 and is thereby recruited to motor neuron genes targeted by Ngn2. RA then facilitates the recruitment of a histone acetyltransferase CBP to the Ngn2/RAR-complex, markedly inducing histone H3/H4-acetylation. Correspondingly, timely inactivation of CBP and its paralog p300 results in profound defects in motor neuron specification and motor axonal projection, accompanied by significantly reduced histone H3-acetylation of the motor neuron enhancer. Our study uncovers the mechanism by which extrinsic RA-signal and intrinsic transcription factor Ngn2 cooperate for cell fate specification through their synergistic activity to trigger transcriptionally active chromatin.
Collapse
Affiliation(s)
- Seunghee Lee
- Dept. Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Bora Lee
- Dept. Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Jae W. Lee
- Dept. Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Soo-Kyung Lee
- Dept. Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
17
|
Busson M, Daury L, Seyer P, Grandemange S, Pessemesse L, Casas F, Wrutniak-Cabello C, Cabello G. Avian MyoD and c-Jun coordinately induce transcriptional activity of the 3,5,3'-triiodothyronine nuclear receptor c-ErbAalpha1 in proliferating myoblasts. Endocrinology 2006; 147:3408-18. [PMID: 16556763 DOI: 10.1210/en.2006-0101] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although physical interactions with other receptors have been reported, heterodimeric complexes of T(3) nuclear receptors (TR) with retinoid X receptors (RXRs) are considered as major regulators of T(3) target gene expression. However, despite the potent T(3) influence in proliferating myoblasts, RXR isoforms are not expressed during proliferation, raising the question of the nature of the complex involved in TRalpha transcriptional activity. We have previously established that c-Jun induces TRalpha1 transcriptional activity in proliferating myoblasts not expressing RXR. This regulation is specific to the muscle lineage, suggesting the involvement of a muscle-specific factor. In this study, we found that MyoD expression in HeLa cells stimulates TRalpha1 activity, an influence potentiated by c-Jun coexpression. Similarly, in the absence of RXR, MyoD or c-Jun overexpression in myoblasts induces TRalpha1 transcriptional activity through a direct repeat 4 or an inverted palindrome 6 thyroid hormone response element. The highest rate of activity was recorded when c-Jun and MyoD were coexpressed. Using c-Jun-negative dominants, we established that MyoD influence on TRalpha1 activity needs c-Jun functionality. Furthermore, we demonstrated that TRalpha1 and MyoD physically interact in the hinge region of the receptor and the transactivation and basic helix loop helix domains of MyoD. RXR expression (spontaneously occurring at the onset of myoblast differentiation) in proliferating myoblasts abrogates these interactions. These data suggest that in the absence of RXR, TRalpha1 transcriptional activity in myoblasts is mediated through a complex including MyoD and c-Jun.
Collapse
Affiliation(s)
- Muriel Busson
- Unité d'Endocrinologie Cellulaire, Unité Mixte de Recherche Différenciation Cellulaire et Croissance, Institut National de la Recherche Agronomique, 2 place Viala, 34060 Montpellier Cedex 1, France
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Hamade A, Deries M, Begemann G, Bally-Cuif L, Genêt C, Sabatier F, Bonnieu A, Cousin X. Retinoic acid activates myogenesis in vivo through Fgf8 signalling. Dev Biol 2006; 289:127-40. [PMID: 16316642 DOI: 10.1016/j.ydbio.2005.10.019] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Revised: 09/13/2005] [Accepted: 10/21/2005] [Indexed: 11/21/2022]
Abstract
Retinoic acid (RA) has been shown to regulate muscle differentiation in vitro. Here, we have investigated the role of RA signalling during embryonic myogenesis in zebrafish. We have altered RA signalling from gastrulation stages onwards by either inhibiting endogenous RA synthesis using an inhibitor of retinaldehyde dehydrogenases (DEAB) or by addition of exogenous RA. DEAB reduces expression of the myogenic markers myoD and myogenin in somites, whereas RA induces increased expression of these genes and strongly induces premature myoD expression in the presomitic mesoderm (psm). The expression dynamics of myf5 in presomitic and somitic mesoderm suggest that RA promotes muscle differentiation, a role supported by the fact that RA activates expression of fast myosin, while DEAB represses it. We identify Fgf8 as a major relay factor in RA-mediated activation of myogenesis. We show that fgf8 expression in somites and anterior psm is regulated by RA, and find that in the absence of Fgf8 signalling in the acerebellar mutant RA fails to promote myoD expression. We propose that, in the developing embryo, localised synthesis of RA by Raldh2 in the anterior psm and in somites activates fgf8 expression which in turn induces the expression of myogenic genes and fast muscle differentiation.
Collapse
Affiliation(s)
- Aline Hamade
- UMR866 Différenciation Cellulaire et Croissance, INRA, Montpellier, France
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Ricaud S, Vernus B, Bonnieu A. Response of human rhabdomyosarcoma cell lines to retinoic acid: relationship with induction of differentiation and retinoic acid sensitivity. Exp Cell Res 2005; 311:192-204. [PMID: 16236281 DOI: 10.1016/j.yexcr.2005.09.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2005] [Revised: 09/12/2005] [Accepted: 09/14/2005] [Indexed: 11/29/2022]
Abstract
The ability of retinoids to induce growth inhibition associated with differentiation of diverse cell types makes them potent anti-cancer agents. We examined the effect of retinoic acid (RA) in cell lines derived from rhabdomyosarcoma (RMS), a malignant soft-tissue tumor committed to the myogenic lineage, but arrested prior to terminal differentiation. We showed that several RMS derived cell lines, including RD human rhabdomyosarcoma cells, are resistant to the growth-inhibitory and differentiation effects of RA. We established that this RA-resistance correlates with reduced expression and activity of RA-receptors in RD cells. We stably expressed either RARalpha, RARbeta, RARgamma, or RXRalpha expression vector into RD cells and found that only RARbeta or RARgamma induced a significant RA growth arrest without promoting differentiation indicating that changes in the amounts of RARs and RXRs are not sufficient to determine the RA myogenic response of rhabdomyosarcoma cells. Activation of RD cell differentiation by ectopic MRF4 expression enhanced RA-receptor activity and led to RA induction of differentiation. These studies demonstrate that RA-resistance of RD cells is linked to their lack of differentiation and suggest that the differentiation-promoting activity of RA requires factors other than RAR-RXR heterodimers.
Collapse
Affiliation(s)
- Stéphanie Ricaud
- INRA, UMR 866-Différenciation Cellulaire et Croissance, 34060 Montpellier Cedex 1, France
| | | | | |
Collapse
|
20
|
Swales K, Kakizaki S, Yamamoto Y, Inoue K, Kobayashi K, Negishi M. Novel CAR-mediated mechanism for synergistic activation of two distinct elements within the human cytochrome P450 2B6 gene in HepG2 cells. J Biol Chem 2005; 280:3458-3466. [PMID: 15563456 DOI: 10.1074/jbc.m411318200] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The constitutive active receptor (CAR) regulates the induction of the cytochrome P450 2B6 (CYP2B6) gene by phenobarbital-type inducers, such as 1,4 bis[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP) via the distal phenobarbital-responsive enhancer module (PBREM, at -1732/-1685 bp). Activation of the PBREM by TCPOBOP generated a 10-fold induction of CYP2B6 mRNA in HepG2 cells stably expressing mouse CAR (Ym17). Co-treatment with the protein phosphatase inhibitor okadaic acid (OA) synergistically increased this induction over 100-fold without directly activating CAR or the PBREM. Although OA synergy required the presence of PBREM, deletion assays delineated the OA-responsive activity to a proximal 24-bp (-256/-233) sequence (OARE) in the CYP2B6 promoter. CAR did not directly bind to the OARE in electrophoretic mobility shift assays. However, both DNA affinity and chromatin immunoprecipitation assays showed a significant increase in CAR association with the OARE after co-treatment with TCPOBOP and OA, indicating the indirect binding of CAR to the OARE. The two cis-acting elements, the distal PBREM and the proximal OARE, within the chromatin structure are both regulated by CAR in response to TCPOBOP and OA, respectively, to maximally induce the CYP2B6 promoter. This functional interaction between the two sites expands the current understanding of the mechanism of CAR-mediated inducible transcription.
Collapse
Affiliation(s)
- Karen Swales
- Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | | | | | | | | | |
Collapse
|
21
|
Oh SY, Lee MY, Kim JM, Yoon S, Shin S, Park YN, Ahn YH, Kim KS. Alternative usages of multiple promoters of the acetyl-CoA carboxylase beta gene are related to differential transcriptional regulation in human and rodent tissues. J Biol Chem 2004; 280:5909-16. [PMID: 15590647 DOI: 10.1074/jbc.m409037200] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Acetyl-CoA carboxylase beta (ACCbeta) is a critical enzyme in the regulation of fatty acid oxidation and is dominantly expressed in the skeletal muscle, heart, and liver. It has been established that two promoters, P-I and P-II, control the transcription of the ACCbeta gene. However, the precise mechanism involved in controlling tissue-specific gene expression of ACCbeta is largely unknown yet. In this study we revealed that promoter P-I, active in the skeletal muscle and heart but not in the liver, could be activated by myogenic regulatory factors and retinoid X receptors in a synergistic manner. Moreover, P-I was also activated markedly by the cardiac-specific transcription factors, Csx/Nkx2.5 and GATA4. These results suggest that the proper stimulation of P-I by these tissue-specific transcription factors is important for the expression of ACCbeta according to the tissue types. In addition, CpG sites around human exon 1a transcribed by P-I are half-methylated in muscle but completely methylated in the liver, where P-I is absolutely inactive. In humans, the skeletal muscle uses P-II as well as P-I, whereas only P-I is active in rat skeletal muscle. The proximal myogenic regulatory factor-binding sites in human P-II, which are not conserved in rat P-II, might contribute to this difference in P-II usage between human and rat skeletal muscle. Hepatoma-derived cell lines primarily use another novel promoter located about 3 kilobases upstream of P-I, designated as P-O. This study is the first to explain the mechanisms underlying the differential regulation of ACCbeta gene expression between tissues in living organisms.
Collapse
Affiliation(s)
- So-Young Oh
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project for Medical Science, Institute of Genetic Science, Yonsei University College of Medicine, 134 Shinchondong Seodaemungu, Seoul 120-752, Korea
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Baldán A, Relat J, Marrero PF, Haro D. Functional interaction between peroxisome proliferator-activated receptors-alpha and Mef-2C on human carnitine palmitoyltransferase 1beta (CPT1beta) gene activation. Nucleic Acids Res 2004; 32:4742-9. [PMID: 15356291 PMCID: PMC519104 DOI: 10.1093/nar/gkh806] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Muscle-type carnitine palmitoyltransferase 1 (CPT1beta) is considered to be the gene that controls fatty acid mitochondrial beta-oxidation. A functional peroxisome proliferator-activated receptor (PPAR) responsive element (PPRE) and a myocite-specific (MEF2) site that binds MEF2A and MEF2C in the promoter of this gene had been previously identified. We investigated the roles of the PPRE and the MEF2 binding sites and the potential interaction between PPARalpha and MEF2C regulating the CPT1beta gene promoter. Mutation analysis indicated that the MEF2 site contributed to the activation of the CPT1beta promoter by PPAR in C2C12 cells. The reporter construct containing the PPRE and the MEF2C site was synergistically activated by co-expression of PPAR, retinoid X receptor (RXR) and MEF2C in non-muscle cells. Moreover, protein-binding assays demonstrated that MEF2C and PPAR specifically bound to one another in vitro. Also for the synergistic activation of the CPT1beta gene promoter by MEF2C and PPARalpha-RXRalpha, a precise arrangement of its binding sites was essential.
Collapse
Affiliation(s)
- Angel Baldán
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Barcelona, Avenue Diagonal 643, E-08028 Barcelona, Spain
| | | | | | | |
Collapse
|
23
|
Ricaud S, Vernus B, Duclos M, Bernardi H, Ritvos O, Carnac G, Bonnieu A. Inhibition of autocrine secretion of myostatin enhances terminal differentiation in human rhabdomyosarcoma cells. Oncogene 2003; 22:8221-32. [PMID: 14614446 DOI: 10.1038/sj.onc.1207177] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Rhabdomyosarcomas (RMSs) are one of the most common solid tumor of childhood. Rhabdomyosarcoma (RMS) cells fail to both complete the skeletal muscle differentiation program and irreversibly exit the cell cycle as a consequence of an active repression exerted on the muscle-promoting factor MyoD. Myostatin is a negative regulator of normal muscle growth, we have thus studied its possible role in RMS cells. Here, we present evidence that overexpression of myostatin is a common feature of RMS since both subtypes of RMS (embryonal RD and alveolar Rh30 cells) express high levels of myostatin when compared to nontumoral skeletal muscle cells. Interestingly, we found that inactivation of myostatin through overexpression of antisense myostatin or of follistatin (a myostatin antagonist) constructs enhanced differentiation of RD cells. In addition, RD and Rh30 cells treated with blocking antimyostatin antibodies progress into the myogenic terminal differentiation program. Finally, our results suggest that high levels of myostatin could impair MyoD function in RMS cells. These results show that an autocrine myostatin loop contributes to maintain RMS cells in an undifferentiating stage and suggest that new therapeutic approaches could be exploited for the treatment of RMS based on inactivation of myostatin protein.
Collapse
Affiliation(s)
- Stéphanie Ricaud
- INRA, UMR 866-Differenciation Cellulaire et Croissance, 34060 Montpellier Cedex 1, France
| | | | | | | | | | | | | |
Collapse
|
24
|
Lopez-Casillas F, Riquelme C, Perez-Kato Y, Ponce-Castaneda MV, Osses N, Esparza-Lopez J, Gonzalez-Nunez G, Cabello-Verrugio C, Mendoza V, Troncoso V, Brandan E. Betaglycan expression is transcriptionally up-regulated during skeletal muscle differentiation. Cloning of murine betaglycan gene promoter and its modulation by MyoD, retinoic acid, and transforming growth factor-beta. J Biol Chem 2003; 278:382-90. [PMID: 12399463 DOI: 10.1074/jbc.m208520200] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Betaglycan is a membrane-anchored proteoglycan co-receptor that binds transforming growth factor beta (TGF-beta) via its core protein and basic fibroblast growth factor through its glycosaminoglycan chains. In this study we evaluated the expression of betaglycan during the C(2)C(12) skeletal muscle differentiation. Betaglycan expression, as determined by Northern and Western blot, was up-regulated during the conversion of myoblasts to myotubes. The mouse betaglycan gene promoter was cloned, and its sequence showed putative binding sites for SP1, Smad3, Smad4, muscle regulatory factor elements such as MyoD and MEF2, and retinoic acid receptor. Transcriptional activity of the mouse betaglycan promoter reporter was also up-regulated in differentiating C(2)C(12) cells. We found that MyoD, but not myogenin, stimulated this transcriptional activity even in the presence of high serum. Betaglycan promoter activity was increased by RA and inhibited by the three isoforms of TGF-beta. On the other hand, basic fibroblast growth factor, BMP-2, and hepatocyte growth factor/scatter factor, which are inhibitors of myogenesis, had little effect. In myotubes, up-regulated betaglycan was also detectable by TGF-beta affinity labeling and immunofluorescence microscopy studies. The latter indicated that betaglycan was localized both on the cell surface and in the ECM. Forced expression of betaglycan in C(2)C(12) myoblasts increases their responsiveness to TGF-beta2, suggesting that it performs a TGF-beta presentation function in this cell lineage. These results indicate that betaglycan expression is up-regulated during myogenesis and that MyoD and RA modulate its expression by a mechanism that is independent of myogenin.
Collapse
Affiliation(s)
- Fernando Lopez-Casillas
- Instituto de Fisiologia Celular, Universidad Nacional Autónoma de México, Apartado Postal 70-246, México City, D.F., 04510, México
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Santalucía T, Moreno H, Palacín M, Yacoub MH, Brand NJ, Zorzano A. A novel functional co-operation between MyoD, MEF2 and TRalpha1 is sufficient for the induction of GLUT4 gene transcription. J Mol Biol 2001; 314:195-204. [PMID: 11718554 DOI: 10.1006/jmbi.2001.5091] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We report tripartite co-operation between MyoD, myocyte enhancer factor-2 (MEF2) and the thyroid hormone receptor (TRalpha1) that takes place in the context of an 82-bp muscle-specific enhancer in the rat insulin-responsive glucose transporter (GLUT4) gene that is active in both cardiac and skeletal muscle. In the L6E9 skeletal muscle cell line and in 10T1/2 fibroblasts, a powerful synergistic activation of the GLUT4 enhancer relied on the over-expression of MyoD, MEF2 and TRalpha1 and the integrity of their respective binding sites, and occurred when linked to either a heterologous promoter or in the context of the native GLUT4 promoter. In cardiac myocytes, enhancer activity was dependent on the binding sites for MEF2 and TRalpha1. Furthermore, we show that in 10T1/2 fibroblasts, the forced expression of MyoD, MEF2 and TRalpha1 induced the expression of the endogenous, otherwise silent, GLUT4 gene. In all, our results indicate a novel functional co-operation between these three factors which is required for full activation of GLUT4 transcription.
Collapse
MESH Headings
- Animals
- Base Sequence
- Binding Sites
- Cell Line
- Cells, Cultured
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Enhancer Elements, Genetic/genetics
- Fibroblasts/metabolism
- Genes, Reporter/genetics
- Glucose Transporter Type 4
- Humans
- MEF2 Transcription Factors
- Mice
- Monosaccharide Transport Proteins/genetics
- Monosaccharide Transport Proteins/metabolism
- Muscle Proteins
- Muscle, Skeletal/cytology
- Muscle, Skeletal/metabolism
- MyoD Protein/genetics
- MyoD Protein/metabolism
- Myocardium/cytology
- Myocardium/metabolism
- Myogenic Regulatory Factors
- Precipitin Tests
- Promoter Regions, Genetic/genetics
- Protein Binding
- Rats
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Thyroid Hormone
- Response Elements/genetics
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription, Genetic/genetics
- Transcriptional Activation
- Transfection
- Troponin I/genetics
Collapse
Affiliation(s)
- T Santalucía
- Department of Cardiothoracic Surgery, National Heart and Lung Institute, Faculty of Medicine, Imperial College of Science, Technology and Medicine, London, SW3 6LY, UK
| | | | | | | | | | | |
Collapse
|
26
|
Daury L, Busson M, Casas F, Cassar-Malek I, Wrutniak-Cabello C, Cabello G. The triiodothyronine nuclear receptor c-ErbAalpha1 inhibits avian MyoD transcriptional activity in myoblasts. FEBS Lett 2001; 508:236-40. [PMID: 11718722 DOI: 10.1016/s0014-5793(01)03063-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Thyroid hormone stimulates myoblast differentiation, through an inhibition of AP-1 activity occurring at the onset of differentiation. In this study we found that the T3 nuclear receptor c-ErbAalpha1 (T3Ralpha1) is involved in a mechanism preserving the duration of myoblast proliferation. Independently of the hormone presence, T3Ralpha1 represses avian MyoD transcriptional activity. Using several mutants of T3Ralpha1, we found that the hinge region plays a crucial role in the inhibition of MyoD activity. In particular, mutations of two small basic sequences included in alpha helices abrogate the T3Ralpha1/MyoD functional interaction. Similarly, the T3 receptor also represses myogenin transcriptional activity. Therefore, despite stimulating avian myoblast differentiation by a T3-dependent pathway not involving myogenic factors, T3Ralpha1 contributes to maintain an optimal myoblast proliferation period by inhibiting MyoD and myogenin activity.
Collapse
Affiliation(s)
- L Daury
- Unité d'Endocrinologie Cellulaire, UMR Différenciation Cellulaire et Croissance (INRA, Université Montpellier II, ENSAM), Institut National de la Recherche Agronomique (INRA), 2 place Viala, 34060 Montpellier Cedex 1, France
| | | | | | | | | | | |
Collapse
|
27
|
Hunter JG, van Delft MF, Rachubinski RA, Capone JP. Peroxisome proliferator-activated receptor gamma ligands differentially modulate muscle cell differentiation and MyoD gene expression via peroxisome proliferator-activated receptor gamma -dependent and -independent pathways. J Biol Chem 2001; 276:38297-306. [PMID: 11477074 DOI: 10.1074/jbc.m103594200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The effects of distinct classes of peroxisome proliferator-activated receptor gamma (PPARgamma) ligands on myogenesis and MyoD gene expression were examined in mouse skeletal muscle C2C12 myoblasts. Treatment of C2C12 cells with the PPARgamma ligand, 15-deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2), repressed morphologically defined myogenesis and reduced endogenous mRNA levels of the myogenic differentiation markers MyoD, myogenin, and alpha-actin. In contrast, two synthetic PPARgamma ligands, L-805645 and ciglitazone, exhibited no effects. In transient transfection assays, 15d-PGJ2 specifically inhibited the expression of a MyoD promoter-luciferase reporter gene (MyoDLuc) in a cell type- and promoter-specific manner, indicating that 15d-PGJ2 functions in part by repressing MyoD gene transcription. The inhibition of MyoD gene expression by 15d-PGJ2 is mediated by the distal region of the MyoD gene promoter. PPARgamma on its own also inhibited MyoDLuc expression and further augmented the 15d-PGJ2 response. In contrast, L-805645 and ciglitazone did not inhibit MyoDLuc expression on their own but did so in the presence of ectopically expressed PPARgamma. Interestingly, a transdominant inhibitor of PPARgamma (hPPARgamma2Delta500) had no effect on the 15d-PGJ2-dependent repression of MyoDLuc expression but overcame L-805645/PPARgamma-dependent repression. Finally, saturating concentrations of L-805645, which did not affect myogenesis, failed to ablate 15d-PGJ2-mediated repression of the myogenic program. Thus, distinct PPARgamma ligands may repress MyoD gene expression through PPARgamma-dependent and -independent pathways, and 15d-PGJ2 can inhibit the myogenic program independent of its cognate receptor, PPARgamma.
Collapse
Affiliation(s)
- J G Hunter
- Department of Biochemistry, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | | | | | | |
Collapse
|
28
|
Solanes G, Pedraza N, Iglesias R, Giralt M, Villarroya F. The human uncoupling protein-3 gene promoter requires MyoD and is induced by retinoic acid in muscle cells. FASEB J 2000; 14:2141-3. [PMID: 11024001 DOI: 10.1096/fj.00-0363fje] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The uncoupling protein-3 (UCP-3) gene encodes for a mitochondrial protein expressed preferentially in skeletal muscle. UCP-3 mRNA is expressed in cultured muscle cells (C2C12 or L6E9) only when differentiated, at which stage UCP-3 is highly induced by all-trans retinoic acid (RA). Here we report that human UCP-3 promoter activity is dependent on MyoD and inducible by all trans-RA. The action of all trans-RA is increased by co-transfection with RA receptor (RAR). We have characterized the RA response element that controls the induction by RA in the 5' noncoding region of the UCP-3 gene. Deletion and point-mutation analysis of the hUCP-3 promoter led us to identify a direct-repeat element with one base-pair spacing (DR1) at position -71/-59 responsible for the induction by RA of the activity of the promoter. This DR1 element bound a nuclear protein complex from muscle cells that contain RAR and retinoid X receptor (RXR). In the absence of this element, the promoter became unresponsive to RA, but it was still dependent on MyoD. In conclusion, it has been established that UCP-3 gene promoter activity is dependent on MyoD, and the first regulatory pathway for UCP-3 gene promoter regulation has been recognized by identifying RA as a transcriptional activator of the gene.
Collapse
Affiliation(s)
- G Solanes
- Departament de Bioquimica i Biologia Molecular, Universitat de Barcelona, Avda Diagonal 645, 08028 Barcelona, Spain
| | | | | | | | | |
Collapse
|
29
|
Puri PL, Sartorelli V. Regulation of muscle regulatory factors by DNA-binding, interacting proteins, and post-transcriptional modifications. J Cell Physiol 2000; 185:155-73. [PMID: 11025438 DOI: 10.1002/1097-4652(200011)185:2<155::aid-jcp1>3.0.co;2-z] [Citation(s) in RCA: 232] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Skeletal muscle differentiation is influenced by multiple pathways, which regulate the activity of myogenic regulatory factors (MRFs)-the myogenic basic helix-loop-helix proteins and the MEF2-family members-in positive or negative ways. Here we will review and discuss the network of signals that regulate MRF function during myocyte proliferation, differentiation, and post-mitotic growth. Elucidating the mechanisms governing muscle-specific transcription will provide important insight in better understanding the embryonic development of muscle at the molecular level and will have important implications in setting out strategies aimed at muscle regeneration. Since the activity of MRFs are compromised in tumors of myogenic derivation-the rhabdomyosarcomas-the studies summarized in this review can provide a useful tool to uncover the molecular basis underlying the formation of these tumors.
Collapse
Affiliation(s)
- P L Puri
- Department of Biology, University of California San Diego, La Jolla, California, USA.
| | | |
Collapse
|
30
|
Schaefer L, Beermann ML, Miller JB. Coding sequence, genomic organization, chromosomal localization, and expression pattern of the signalosome component Cops2: the mouse homologue of Drosophila alien. Genomics 1999; 56:310-6. [PMID: 10087198 DOI: 10.1006/geno.1998.5728] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The Drosophila alien gene is highly homologous to the human thyroid receptor interacting protein, TRIP15/COPS2, which is a component of the recently identified signalosome protein complex. We identified the mouse homologue of Drosophila alien through homology searches of the EST database. We found that the mouse cDNA encodes a predicted 443-amino-acid protein, which migrates at approximately 50 kDa. The gene for the mouse alien homologue, named Cops2, includes 12 coding exons spanning approximately 30 kb of genomic DNA on the central portion of mouse chromosome 2. Mouse Cops2 is widely expressed in embryonic, fetal, and adult tissues beginning as early as E7.5. Mouse Cops2 cDNA hybridizes to two mRNA bands in all tissues at approximately 2.3 and approximately 4 kb, with an additional approximately 1.9-kb band in liver. Immunostaining of native and epitope tagged proteins localized the mouse Cops2 protein in both the cytoplasm and the nucleus, with larger amounts in the nucleus in some cells.
Collapse
Affiliation(s)
- L Schaefer
- Myogenesis Research Laboratory, Massachusetts General Hospital, 149 13th Street, Charlestown, Massachusetts 02129, USA
| | | | | |
Collapse
|
31
|
Lau P, Bailey P, Dowhan DH, Muscat GE. Exogenous expression of a dominant negative RORalpha1 vector in muscle cells impairs differentiation: RORalpha1 directly interacts with p300 and myoD. Nucleic Acids Res 1999; 27:411-20. [PMID: 9862959 PMCID: PMC148194 DOI: 10.1093/nar/27.2.411] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
ROR/RZR is an orphan nuclear receptor that has no known ligand in the 'classical sense'. In the present study we demonstrate that RORalpha is constitutively expressed during the differentiation of proliferating myoblasts to post-mitotic multinucleated myotubes, that have acquired a contractile phenotype. Exogenous expression of dominant negative RORalpha1DeltaE mRNA in myogenic cells significantly reduces the endogenous expression of RORalpha1 mRNA, represses the accumu-lation and delays the activation of mRNAs encoding MyoD and myogenin [the muscle-specific basic helix-loop-helix (bHLH) proteins] and p21(Waf-1/Cip-1) (a cdk inhibitor). Immunohistochemistry demonstrates that morpho-logical differentiation is delayed in cells expressing the RORDeltaE transcript. Furthermore, the size and development of mutlinucleated myotubes is impaired. The E region of RORalpha1 interacts with p300, a cofactor that functions as a coactivator in nuclear receptor and MyoD-mediated transactivation. Consistent with the functional role of RORalpha1 in myogenesis, we observed that RORalpha1 directly interacts with the bHLH protein MyoD. This interaction was mediated by the N-terminal activation domain of the bHLH protein, MyoD, and the RORalpha1 DNA binding domain/C region. Furthermore, we demonstrated that p300, RORalpha1 and MyoD interact in a non-competitive manner. In conclusion, this study provides evidence for a biological role and positive influence of RORalpha1 in the cascade of events involved in the activation of myogenic-specific markers and cell cycle regulators and suggests that crosstalk between theretinoid-relatedorphan (ROR) nuclear receptors and the myogenic bHLH proteins has functional consequences for differentiation.
Collapse
Affiliation(s)
- P Lau
- University of Queensland, Centre for Molecular and Cellular Biology, Ritchie Research Laboratories, B402A, St Lucia, 4072, Queensland, Australia
| | | | | | | |
Collapse
|
32
|
Bailey P, Sartorelli V, Hamamori Y, Muscat GE. The orphan nuclear receptor, COUP-TF II, inhibits myogenesis by post-transcriptional regulation of MyoD function: COUP-TF II directly interacts with p300 and myoD. Nucleic Acids Res 1998; 26:5501-10. [PMID: 9826778 PMCID: PMC147985 DOI: 10.1093/nar/26.23.5501] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
COUP-TF II is an orphan nuclear receptor that has no known ligand in the 'classical sense'. COUP-TF interacts with the corepressors N-CoR, SMRT and RIP13, and silences transcription by active repression and trans-repression. Forced expression of the orphan nuclear receptor COUP-TF II in mouse C2 myogenic cells has been demonstrated to inhibit morphological differentiation, and to repress the expression of: (i) the myoD gene family which encodes myogenic basic helix-loop-helix (bHLH) proteins; and (ii) the cell cycle regulator, p21(Waf-1/Cip-1). In the present study, we show that COUP-TF II efficiently inhibits the myoD -mediated myogenic conversion of pluripotential C3H10T1/2 cells by post-transcriptional mechanisms. Furthermore, repression of MyoD-dependent transcription by COUP-TF II occurs in the absence of the nuclear receptor cognate binding motif. The inhibition of MyoD-mediated trans-activation involves the direct binding of the DNA binding domain/C-region and hinge/D-regions [i.e. amino acid (aa) residues 78-213] of COUP-TF II to the N-terminal activation domain of MyoD. Over-expression of the cofactor p300, which functions as a coactivator of myoD-mediated transcription, alleviated repression by COUP-TF II. Further binding analysis demonstrated that COUP-TF II interacted with the N-terminal 149 aa residues of p300 which encoded the receptor interaction domain of the coactivator. Finally we observed that COUP-TF II, MyoD and p300 interact in a competitive manner, and that increasing amounts of COUP-TF II have the ability to reduce the interaction between myoD and p300 invitro. The experiments presented herein suggest thatCOUP-TF II post-transcriptionally regulates myoD activity/function, and that crosstalk between orphan nuclear receptors and the myogenic bHLH proteins has functional consequences for differentiation.
Collapse
MESH Headings
- 3T3 Cells
- Animals
- Binding, Competitive/genetics
- COUP Transcription Factors
- Cell Differentiation/genetics
- Cells, Cultured
- DNA-Binding Proteins/physiology
- E1A-Associated p300 Protein
- Gene Expression Regulation, Developmental
- Genes, Reporter
- Humans
- Male
- Mice
- Mice, Inbred C3H
- Muscle, Skeletal/cytology
- Muscle, Skeletal/metabolism
- MyoD Protein/metabolism
- MyoD Protein/physiology
- Nuclear Proteins/physiology
- Peptide Fragments/physiology
- RNA Processing, Post-Transcriptional
- Receptors, Steroid
- Repressor Proteins/physiology
- Trans-Activators/physiology
- Transcription Factors/physiology
- Transcriptional Activation
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- P Bailey
- University of Queensland, Centre for Molecular and Cellular Biology, Ritchie Research Laboratories, B402A, St Lucia, 4072 Queensland, Australia
| | | | | | | |
Collapse
|