Next Article in Journal
Circadian Governance of Cardiac Growth
Next Article in Special Issue
Efficient Isolation of Lymphocytes and Myogenic Cells from the Tissue of Muscle Regeneration
Previous Article in Journal
The Lysosome in Malignant Melanoma: Biology, Function and Therapeutic Applications
Previous Article in Special Issue
Inflammatory Caspase Activity Mediates HMGB1 Release and Differentiation in Myoblasts Affected by Peripheral Arterial Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Factors Regulating or Regulated by Myogenic Regulatory Factors in Skeletal Muscle Stem Cells

1
Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu 803-8580, Japan
2
Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu 803-8580, Japan
3
Division of Anatomy, Department of Health Promotion, Kyushu Dental University, Kitakyushu 803-8580, Japan
*
Author to whom correspondence should be addressed.
Cells 2022, 11(9), 1493; https://doi.org/10.3390/cells11091493
Submission received: 14 February 2022 / Revised: 25 April 2022 / Accepted: 26 April 2022 / Published: 29 April 2022

Abstract

:
MyoD, Myf5, myogenin, and MRF4 (also known as Myf6 or herculin) are myogenic regulatory factors (MRFs). MRFs are regarded as master transcription factors that are upregulated during myogenesis and influence stem cells to differentiate into myogenic lineage cells. In this review, we summarize MRFs, their regulatory factors, such as TLE3, NF-κB, and MRF target genes, including non-myogenic genes such as taste receptors. Understanding the function of MRFs and the physiology or pathology of satellite cells will contribute to the development of cell therapy and drug discovery for muscle-related diseases.

1. Introduction

The skeletal muscle accounts for 40% of the total body weight, and the metabolism and motor functions of skeletal muscles are essential for human life. Loss of skeletal muscle mass leads to various negative effects, including obesity, insulin resistance, diabetes, decreased quality of life that may require dependence on others and hospital stays, and mortality [1]. As the number of elderly people in society is increasing, it is important to establish strategies for improving skeletal muscle mass and function in aged individuals. Therefore, understanding the physiology and pathology of satellite cells, such as skeletal muscle stem cells, is essential.
During skeletal muscle generation, known as myogenesis, myoblasts fuse with myofibers. Myogenesis is divided into two distinct phases [2]. In the embryonic stage, the structures derived from the mesoderm generate body muscle fibers; these fibers act as a template for the generation of additional fibers [3,4]. In the perinatal stage, myogenic progenitors residing in muscles initially proliferate extensively. Subsequently, the number of myonuclei reaches a steady state, and the protein synthesis in myofibers reaches a peak [5,6]. Once the muscle matures, these progenitors enter a period of quiescence and exist between the muscle fibers and basal lamina as satellite cells. In the adult stage, myogenesis depends on satellite cells to maintain tissue homeostasis, similar to in other organs that undergo renewal [7,8,9].
MyoD, Myf5, myogenin, and MRF4 (also known as Myf6 or herculin) are myogenic regulatory factors (MRFs), which act as master transcription factors that are upregulated during myogenesis and influence stem cells to differentiate into myogenic lineage cells [10,11,12,13]. MRFs regulate myogenesis by modulating the expression of both myogenic and non-myogenic genes. However, the mechanisms by which MRFs regulate non-myogenic genes remain unknown. Although signaling molecules mediate myogenesis and MRFs, these signaling mechanisms are not completely understood. In this review, we summarize MRFs and focus on their regulatory factors, such as transducin-like enhancer of Split3 (TLE3), nuclear factor-kappa B (NF-κB), and MRF target genes, including non-myogenic genes such as taste receptors.

2. Muscle Stem Cells and MyoD

Satellite cells are typically quiescent during the cell cycle; however, when these are activated by stimulation, such as by muscle injury, the expression of MRFs is upregulated and satellite cell progenitors commit to the myoblast lineage and proliferate rapidly [14,15]. Satellite cells are abundant during muscle regeneration and postnatal muscle growth [16]. In addition to satellite cells, mesangioblasts [17], Pw1+/Pax7+ interstitial cells [18], and TWIST2+ progenitors [19] are present in the population of muscle stem cells and can be directly converted to myogenic lineage cells. Although these cell classifications do not conform to the current characterization and many of these cells have overlapping markers and functions, the expression of MyoD is upregulated in all of these cells during myogenesis [20,21,22]. MyoD was first identified by the Harold M. Weintraub group. MyoD was cloned during a functional analysis of muscle fiber formation, and the expression of cDNA for the mouse MyoD protein converted murine embryonic fibroblast C3H10T1/2 into myogenic cells [22]. Ectopic expression of MyoD can also induce human induced pluripotent stem (iPS) cells or murine embryonic stem cells to differentiate into myogenic cells [23,24,25,26,27]. Therefore, introducing MyoD into iPS cells shows potential as a safe and effective approach for regenerative medicine applications in the muscle.

3. Structure and Function of MRFs

MyoD is an MRF transcription factor, along with Myf5 [10], myogenin [11], and MRF4 [12,13]. Despite variations in their lengths and amino acid sequences, each MRF is composed of three domain structures, including a highly conserved basic helix-loop-helix (bHLH) domain with myogenic potential [28]. This bHLH domain is flanked by an N-terminal region with a C/H domain and C-terminal region with a helix 3 domain that mediates the transcriptional region [29]. Phylogenetic analyses of the amino acid sequences of murine MRFs showed that the similarity between MyoD and Myf5 is 53%. The similarities between myogenin and MyoD and Myf5 are 38% and 40%, respectively. The similarities of MRF4 with MyoD and Myf5 are 43% and 40%, respectively (Figure 1) [30]. MRFs exhibit functional redundancy. Each MRF can induce myogenesis and is anthropogenically expressed in various non-myogenic cells such as fibroblasts, adipose, nerve, and liver cells, indicating a functional overlap in establishing the muscle cell lineage [31]. Experiments using MRF-null mice revealed hierarchical relations and overlapping functions as features of the regulatory network of MRFs [32]. Moreover, newborn mice null for both MyoD and Myf5 were completely deficient in myoblasts and myofibers. MyoD and Myf5 are essential for myogenic lineage cell commitment and act upstream of MRF4 and myogenin [12,33,34]. Further experiments focusing on these differences are required to understand the detailed functions of each MRF.

4. Role of MRFs during Myogenesis after Birth

Paired box gene 3 (Pax3) and paired box gene 7 (Pax7) are transcription factors. In postnatal muscle cells, Pax3 and Pax7 are markers of muscle stem cells that reside beneath the basal lamina of mature myofibers [35]. Pax7 is expressed in all satellite cells of myofibers after birth, whereas Pax3 is not. Chromatin immunoprecipitation (ChIP) sequencing of primary myoblasts revealed that although Pax3 and Pax7 recognize the same DNA motifs, Pax3 binds with a lower affinity compared to that of Pax7. Pax3 binds to a subset of Pax7 target genes that mainly regulate embryonic function and maintain undifferentiated satellite cells. In contrast, Pax7 activates the expression of genes involved in maintaining the postnatal satellite cell phenotype by regulating proliferation and inhibiting differentiation [36]. Satellite cells are typically quiescent, meaning that they are normally arrested in the G0 phase of the cell cycle [37]. During this phase, the cells can be divided into two groups, myogenic stem cells (~10%, Pax7+/Myf5) [38] and myogenic precursor cells (~90%, Pax7+/Myf5+) [39], based on their self-renewal ability (Figure 2) [40]. When satellite cells are activated by stimuli such as muscle injury, the expression of Pax7 [21] and Pax3 [41] gradually decreases in most satellite cells. In contrast, muscle satellite cells begin to increase MRF expression to ensure a myogenic program [42]. Myogenic stem cells (Pax7+/Myf5) exhibit asymmetric division, during which some daughter cells return to myogenic stem cells (Pax7+/Myf5) to serve as a stem cell reserve. Myogenic precursor cells (Pax7+/Myf5+) exhibit symmetrical division, in which every daughter cell is committed to the myoblast fate and begins to differentiate [39]. In addition to the expression of Myf5 and Pax7, satellite cells begin to express MyoD protein during the early stages of activation [43]. Pax7 is necessary to activate satellite cells by directly binding to the promoter and enhancer regions of MyoD and Myf5, respectively [36]. The basal lamina remnants guide activated satellite cell progeny that is committed to myogenesis to migrate and proliferate. Increased MyoD expression with MRF4 and myogenin expression stimulates the differentiation of these cells. Terminally differentiated satellite cells may fuse into either newly formed myofibers or pre-existing myofibers (Figure 2) [40]. The main function of MyoD is to withdraw cells from the cell cycle by enhancing the expression of myogenin and p21. p21 inhibits cyclin-dependent kinases, which inhibit MyoD. Therefore, MyoD expression enhances MyoD activity in satellite cells in a feedback and/or feedforward manner. Continuous expression of MyoD is required to sustain muscle-related gene expression [44] and is a critical effector of the fast-twitch muscle fiber phenotype (type IIA, IIB, and IIX) [45,46].

5. Transcriptional Mechanism of MRFs

MRFs dimerize with other HLH-containing proteins through HLH–HLH interactions [47]. MRFs also form heterodimers with E proteins, including E47 and E12, through the bHLH domain and can bind to the E-box sequence motif containing the nucleotide CANNTG. Therefore, heterodimer formation activates transcription at the promoters of muscle-specific genes [48]. MRFs are thought to bind to hundreds of muscle-related gene promoters to result in stem cell myogenesis. Differences in the functions of MRF are related to their ability to induce different programs of gene expression. Studies are needed to understand how MRFs bind to the genome during skeletal muscle regeneration and development. The binding dynamics of MyoD have been extensively studied using cultured cells with high-throughput technologies, including DNA ChIP sequencing [49,50,51,52]. In vitro experiments using C2C12 cells revealed the MyoD-binding site during myoblast differentiation [53].
In addition to the regulatory elements of genes that are upregulated or downregulated during myoblast differentiation, MyoD recognizes and binds to a large number of E-box sites that are not related to myogenesis. Cao et al. compared the number of binding sites in myoblasts and myotubes and identified 23,000 and 26,000 MyoD binding sites in myoblasts and myotubes, respectively; they also showed that MyoD binding is more stable after differentiation. MyoD binding is correlated with regional histone acetylation rather than with local histone acetylation [53]. The biological importance and functions of these sites remain unclear because most of these sites are inactive when functional enhancer tests are performed in vitro. Mousavi et al. confirmed that the amount of bound MyoD in myotubes is higher than that in myoblasts (39,700 and 18,142, respectively) [54]. Both analyses revealed a discrepancy in the number of binding sites [53,54]. These differences are likely related to differences in the methods used to identify the peaks and boundaries of compartmentalization reading in the experiments. Using RNA-sequencing analysis, Mousavi et al. revealed that both MyoD and myogenin share most of the 35,000 binding sites [54]. In myotubes, these sites are also bound by polymerase II and are marked by H3K27Ac and H3K4me1 [54]. A total of 1053 Myf5 sites were identified in myoblasts, and 9300 and 1428 MyoD binding sites were identified in myotubes and myoblasts, respectively. The overlap between the binding sites in myoblasts is 30% [52].
To evaluate the dramatic differences in the number of MyoD-binding sites between myotubes and myoblasts, we focused on transcriptional repressors targeting the same DNA-binding motif as HLH proteins, including MRFs. Snail1 and Snail2 are expressed in myoblast lineage cells and repress transcription by recruiting histone deacetylases 1 and 2 [55,56,57]. Snail1 binds to E-boxes containing a G/C-rich nucleotide associated with genes expressed almost exclusively in myotubes. In contrast, Snail2 does not bind to E-boxes with A/T-rich nucleotides associated with gene expression in myoblasts, suggesting that Snail must be removed for MyoD to access the genes expressed in the myotube. The expression of Snail is also regulated by MRFs. MRFs induce the expression of miRNA-30 and miRNA-206, which suppress Snail1/2 expression in myotubes. Taken together, downregulation of Snail1/2 allows MyoD to access its binding sites [52].

6. Target Genes of MRFs

Taste is a guide and guardian of food intake and is necessary for maintaining human health [58]. Taste can be divided into five categories: salty, sweet, umami, sour, and bitter [59]. The salty, sweet, and umami tastes encourage the consumption of foods containing minerals, carbohydrates, and amino or nucleic acids, respectively, whereas sour and bitter tastes prevent ingestion of strong acids and potentially toxic substances, respectively [59]. The taste 1 receptor (T1R) family belongs to the G-protein-coupled receptor family and is composed of three members: T1R1, T1R2, and T1R3. These receptors form heterodimers with each other and recognize different ligands depending on the combination of receptors. T1R3 forms a dimer with T1R2 to act as the sweet taste receptor that responds to molecules, such as sugars, whereas T1R3 complexes with T1R1 form the umami taste receptor that responds to nuclear acids and amino acids [60,61,62,63]. Although these functions were originally thought to occur in the gustatory tissue inside the mouth, the first study showing that T1Rs and molecules involved in taste signal transduction are present in the stomach and intestine was reported in the late 1990s [64]. Subsequently, many researchers reported the expression and functional roles of T1R family members in various tissues and cell types, including in the adipose tissue [65], pancreatic β-cells [66], central nervous system [67], and heart [68]. Therefore, despite the name “taste receptor,” these G-protein-coupled receptors are now recognized as “nutrient sensors” with critical roles in chemoreception for detecting amino acids and glucose status [69]. We also reported the function of both T1R3 and T1R1, which are components of the umami receptor in the skeletal muscle [70,71,72]. The expression of T1R1 and T1R3 is regulated by MRFs [70,71,72]. ChIP analysis revealed that both MyoD and myogenin bind to the promoter region of T1R3 (Figure 3A). We also demonstrated that MyoD induces the promoter activity of T1R3, as assessed in a luciferase reporter assay. Comparative genomic analysis revealed that the gathering regions of MyoD-and myogenin-binding sites are highly conserved across species [70]. These observations are consistent with the fact that T1R3 expression increases with myogenic differentiation in vitro [70].
MyoD regulates the expression of T1R1 in both myoblasts and myotubes during myogenesis [50]. We determined the mechanism of the regulation of T1R1 expression through T1R1. Krüppel-like factor 5 (Klf5), a zinc finger protein, acts as a transcription factor [73] that regulates the transactivation of T1R1 during myoblast differentiation in C2C12 cells as Klf5 binds to the GT box (CCACCC) in the promoter region of T1R1 [71]. Klf5 also directly binds to MyoD and regulates muscle-specific genes associated with myoblast differentiation [74]. Based on these findings, we assessed the role of Klf5 in the transactivation of T1R1 in combination with MyoD. We examined the relationship between Klf5 and MyoD in regulating the gene expression of T1R1 during myoblast differentiation of C2C12 cells. Our results showed that MyoD binds to the T1R1 promoter region, which has three E-boxes (E-box 1–3). The heterodimer of MyoD and Tcf12 interacts with Klf5 and binds to E-box 1, activates transcription, and increases the expression levels of T1R1 during myoblast differentiation [72] (Figure 3B).
These findings agree with those of studies showing that T1R3 and T1R1 are highly expressed in the skeletal muscles. Skeletal muscle cells from T1R3-null mice exhibit impaired mammalian targets of rapamycin complex 1 signaling and an increase in autophagy [69,75,76]. This suggests that T1R3 forms a heterodimer with T1R1 as an umami receptor and plays a critical role in sensing the nutrient status. The skeletal muscle stores many amino acids and is an important source of amino acids under conditions of amino acid deprivation [77]. Signaling starting from the umami receptor (T1R1/T1R3) may be involved in the physiology of skeletal muscle metabolism and pathology of skeletal muscle diseases through the regulation of autophagy. Further studies are required to confirm this relationship.

7. Factors Regulating Myogenesis and Transcription of MRFs

Several signaling pathways play essential roles in the behaviors of skeletal muscle stem cells. Notch signaling is a major regulatory pathway in muscle progenitor pools. Notch signaling has been shown to inhibit myoblast differentiation in several models [78,79,80,81,82,83,84,85,86,87,88]. Bone morphogenetic protein (BMP) signaling is also involved in maintaining the proliferative status of muscle progenitors [89]. Loss- and gain-of-BMP function experiments in the limbs of fetal chicks showed that activation of BMP signaling leads to an increased number of Pax7-positive cells and increased muscle mass. Inhibition of BMP signaling impairs the growth of skeletal muscle, suggesting that BMP promotes the proliferation of muscle progenitor cells [90]. In mice, BMP regulates postnatal growth and adult muscle homeostasis by regulating the proliferation of satellite cells, whereas BMP inhibition impairs muscle growth [91,92]. Moreover, an inhibitor of DNA-binding protein 1, a BMP target gene, is necessary to maintain muscle satellite cells in an undifferentiated or proliferative status [93,94]. In an experiment using C2C12 cells, BMP induced osteoblast differentiation while suppressing myogenic differentiation [95]. Smad4, a downstream effector of BMP-Smad (canonical BMP) signaling, is a critical suppressor of BMP in myogenic differentiation [96].
The demand for Wnt signaling during myogenesis differs between the fetal and embryonic phases [97]. Embryonic myoblasts in the developing limbs of mice normally form progenitors and skeletal muscle fibers. In the fetal stages, progenitors that express a constitutively active form of β-catenin, an effector molecule of canonical Wnt signaling, increase the number of Pax7-positive cells [97]. Wnt7a enhances the symmetrical division of satellite cells during skeletal muscle regeneration by maintaining the stem cell pool [98]. Further studies reinforced the importance of BMP and Wnt signaling in myogenesis. Recapitulated myogenesis of pluripotent stem cells was achieved through the action of specific molecules at precise time points. When epiblast-like cells were treated with a BMP inhibitor (LDN193189) and Wnt activator (CHIR99021), commitment of the presomitic mesoderm was initiated [99]. Following the addition of a growth factor cocktail (fibroblast growth factor 2, hepatocyte growth factor, and insulin-like growth factor 1), the cell population displays a mixture of differentiated myocytes (myogenin-positive) and progenitor cells (Pax7-positive) [100,101]. Taken together, many growth factors and signaling molecules are involved in regulating the myogenesis and transcription of MRFs. Moreover, these factors and signals may be involved in cross-talk.

8. TLE3 Represses Transcription of MyoD and Suppresses Myogenesis

Groucho/TLE proteins are transcriptional cofactors that do not directly bind to DNA but play essential roles in cell differentiation [102]. Members of the Groucho/TLE family, including TLE3, suppress Wnt-β-catenin signaling as transcriptional co-repressors of Tcf/Lef, which are downstream effectors of Wnt-β-catenin signaling and subsequently repress transactivation of the Wnt target gene [102,103,104,105]. We previously identified Tcf/Lef-responsive elements in the 5′ untranslated regions of TLE3 through comparative genomic and functional analyses. We demonstrated that Wnt-β-catenin signaling directly increases the expression of TLE3 in W20-17 cells, a murine bone marrow mesenchymal stem cell line [106]. Groucho/TLE family proteins are composed of a five-domain structure [107]: the Q domain, which is glutamine-rich and highly conserved, forms two coiled-coil motifs that can oligomerize [108,109,110]; a glycine/proline-rich domain, which is critical for recruiting and interacting with histone deacetylases [109,111,112]; a CcN domain, which contains putative phosphorylation sites of cdc2 and casein kinase II and a nuclear localization sequence; a serine/proline-rich domain, which contains a region enriched by serine/proline [107,113,114,115]; and a WD40 domain, which is highly conserved and contains tandem repeats of tryptophan and aspartic acid. X-ray crystallography revealed that the WD40 domain forms a β-propeller and binds various transcription factors [102]. TLE3 is expressed in the myoblasts of murine embryos, and its expression overlaps with that of both MyoD and Myf5 [116]. TLE3 is also expressed in cells with the same mesenchymal lineage as myoblasts, including bone marrow mesenchymal stem cells [117,118,119], myoblasts, adipocytes, and osteoblasts. MyoD is a master transcription factor of myogenesis, and PPARγ and Runx2 are master transcription regulators of adipogenesis and osteoblastogenesis, respectively [120,121]. Villanueva et al. [122] identified TLE3 as a transcriptional coactivator of PPARγ in adipocytes, and it was later confirmed that TLE3 enhances the transcriptional activation of PPARγ and induces adipogenesis of bone marrow mesenchymal stem cells [118,119]. Interference with the interaction between PPARγ and PRDM16 causes TLE3 to shift the fate of adipose stem cells from brown fat cells to white fat cells, thus impairing fatty acid oxidation and thermogenesis [123]. Endogenous TLE3 interacts with MyoD in C2C12 cells. TLE suppresses myoblast differentiation in C2C12 cells and is induced by overexpression of MyoD in C3H10T1/2 cells [124]. To confirm whether the suppressive effect of TLE3 on myogenesis is due to repression of MyoD-dependent transactivation, we employed luciferase reporters driven by muscle creatine kinase (MCK0.8) and myogenin (MG185) promoters. TLE3 suppresses the transcriptional activation of MyoD and other MRFs including MRF4, Myf5, and myogenin. Moreover, ChIP assays showed that TLE3 disrupts the binding of MyoD to the promoter region of myogenin [124]. Typically, the WD domain of Groucho/TLE plays a critical role in transcriptional regulation [125]. We previously showed that the WD domain of TLE3 is necessary to repress the transcriptional activity of Runx2. However, the WD domain-deletion mutant still represses the transcriptional activity of MyoD, and both the Q and serine/proline-rich domains of TLE3 are essential for interacting with and repressing MyoD [124]. MRFs interact with E proteins, including E12, through their bHLH domains [126]. TLE3 also interacts with the bHLH domain of MyoD. Although overexpression of E12 dose-dependently counters the suppressive effect of TLE3 on transactivation of MyoD activity, TLE3 does not interact with E12 but rather disrupts the interaction between MyoD and E12. This result suggests that TLE3 masks the E protein-binding region on MyoD; thus, TLE3 competitively interferes with MRF and E protein interactions [124] (Figure 4). Together, TLE3 facilitates the differentiation of mesenchymal stem cells into the white adipocyte lineage by activating PPARγ, an adipogenic master regulator, and suppresses the transcription of Runx2 and MyoD, which are master regulators of osteoblastogenesis and myogenesis, respectively. The role of TLE3 in skeletal muscle in vivo remains unknown. Studies are needed to analyze muscle-specific TLE3 knockout mice because TLE3 null mice are embryonically lethal.

9. NF-κB Represses Transcription of MyoD and Suppresses Myogenesis

NF-κB is a transcription factor that is closely related to various physiological and pathological conditions, such as immune system, cell survival, and inflammatory processes. In response to stimuli such as tumor necrosis factor (TNF)-α, the inhibitor of κBα is degraded, and p65, the main subunit of NF-κB, forms a complex with p50. This heterodimer complex of p50/p65 is translocated from the cytoplasm to the nucleus. In the nucleus, the p50/p65 heterodimer binds to the responsive elements of NF-κB to regulate target gene expression [127]. In a mouse model, the regeneration of skeletal muscle following trauma was improved following systemic administration of the NF-κB inhibitor curcumin [128]. Several groups reported that activation of NF-κB suppresses the differentiation of skeletal muscle by reducing MyoD protein levels [129,130,131,132]. In contrast, NF-κB signaling is necessary for myoblast proliferation [129,130,131,132]. Because MyoD regulates the exit from the cell cycle, in part through transactivation of the cell cycle inhibitor p21, NF-κB suppresses myoblast differentiation through the sustained proliferation of myoblasts [129,133]. In addition, NF-κB mediates the suppression of myoblast differentiation by inducing cyclin D1, a member of the cyclin family of proteins, which increases cell cycle progression from the G1 to S phase [129]. We reported that treatment with a TNF-α ligand suppresses MG185-luciferase activity induced by overexpression of MyoD in a TNF-α concentration-dependent manner in vitro. To confirm this function, we generated C-terminal-truncated constructs of p65, the main subunit of NF-κB and downstream effector of TNF-α. p65 consists of transactivation domain 1, transactivation domain 2 (TA2), Rel homology domain, DNA-binding domain, and dimerization domain. Although the full-length and transactivation domain 1 domain-deleted p65 mutant suppressed the MG185-luciferase activity induced by MyoD induction, the TA2 domain-deleted mutant p65 did not. These results indicate that TNF-α suppresses transactivation of MyoD via the canonical NF-κB signaling effector p65-TA2 domain [132]. The DNA binding capacity and transcriptional activation of NF-κB decrease during myoblast differentiation [129,134]. Insulin-like growth factors stimulate the activity of NF-κB during myogenesis [135,136]. We previously determined the activity of NF-κB during skeletal muscle regeneration in a mouse model. NF-κB signaling was active from days 1 to 3 after injection of cardiotoxin, which corresponded to the proliferation stage of satellite cells. This behavior of NF-κB signaling was similar to the expression levels of cyclin A2 and was related to the progression of the S phase in the cell cycle [137]. NF-κB activation occurs prior to the increase in myogenin levels [132]. However, the role of NF-κB (p65) in the skeletal muscle in vivo remains unknown. Further studies of muscle-specific p65 knockout mice are needed because p65-null mice are embryonically lethal.

10. Conclusions

Maintaining skeletal muscle mass and function is important for a healthy lifestyle. Elderly populations in both developed and developing countries have increased, and strategies for regenerating and restoring skeletal muscle mass and function are urgently needed. Therefore, the research focus has rapidly shifted from the development of muscle to the regenerative medicine of skeletal muscle tissue, particularly using stem cells. Researchers have attempted to modify the expression of genes essential for generating muscles in vivo to recapitulate myogenesis ex vivo. In vitro myogenesis protocols are currently used to model muscle-associated diseases in vitro for drug discovery or cell therapy, in which functional muscles are produced in the laboratory for tissue replacement therapy. In this review, we highlight the function of MRFs and their modulators, such as TLE3 and NF-κB, and target genes, such as taste receptors. Further research is needed to understand the function of MRFs and the physiology or pathology of satellite cells, which will contribute to the development of cell therapies or drug discovery for muscle-related diseases.

Author Contributions

T.S., T.T., A.I., T.M., T.K. and S.K., writing the manuscript; S.K., study design; S.K., literature review, article preparation, and document submission. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by JSPS KAKENHI Grant Numbers 22K17257, 21K21072.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Woo, J. Sarcopenia. Clin. Geriatr. Med. 2017, 33, 305–314. [Google Scholar] [CrossRef] [PubMed]
  2. Tajbakhsh, S. Skeletal muscle stem cells in developmental versus regenerative myogenesis. J. Intern. Med. 2009, 266, 372–389. [Google Scholar] [CrossRef] [PubMed]
  3. Parker, M.H.; Seale, P.; Rudnicki, M.A. Looking back to the embryo: Defining transcriptional networks in adult myogenesis. Nat. Rev. Genet. 2003, 4, 497–507. [Google Scholar] [CrossRef] [PubMed]
  4. Sambasivan, R.; Tajbakhsh, S. Skeletal muscle stem cell birth and properties. Semin. Cell. Dev. Biol. 2007, 18, 870–882. [Google Scholar] [CrossRef]
  5. Schultz, E. Satellite cell proliferative compartments in growing skeletal muscles. Dev. Biol. 1996, 175, 84–94. [Google Scholar] [CrossRef]
  6. Davis, T.A.; Fiorotto, M.L. Regulation of muscle growth in neonates. Curr. Opin. Clin. Nutr. Metab. Care 2009, 12, 78–85. [Google Scholar] [CrossRef] [Green Version]
  7. Schmalbruch, H.; Lewis, D.M. Dynamics of nuclei of muscle fibers and connective tissue cells in normal and denervated rat muscles. Muscle Nerve 2000, 23, 617–626. [Google Scholar] [CrossRef]
  8. Pellettieri, J.; Sanchez Alvarado, A. Cell turnover and adult tissue homeostasis: From humans to planarians. Annu. Rev. Genet. 2007, 41, 83–105. [Google Scholar] [CrossRef]
  9. Bentzinger, C.F.; Wang, Y.X.; Rudnicki, M.A. Building muscle: Molecular regulation of myogenesis. Cold Spring Harb. Perspect. Biol. 2012, 4, a008342. [Google Scholar] [CrossRef]
  10. Braun, T.; Buschhausen-Denker, G.; Bober, E.; Tannich, E.; Arnold, H.H. A novel human muscle factor related to but distinct from MyoD1 induces myogenic conversion in 10T1/2 fibroblasts. EMBO J. 1989, 8, 701–709. [Google Scholar] [CrossRef]
  11. Edmondson, D.G.; Olson, E.N. A gene with homology to the myc similarity region of MyoD1 is expressed during myogenesis and is sufficient to activate the muscle differentiation program. Genes Dev. 1989, 3, 628–640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Braun, T.; Rudnicki, M.A.; Arnold, H.H.; Jaenisch, R. Targeted inactivation of the muscle regulatory gene Myf-5 results in abnormal rib development and perinatal death. Cell 1992, 71, 369–382. [Google Scholar] [CrossRef]
  13. Rhodes, S.J.; Konieczny, S.F. Identification of MRF4: A new member of the muscle regulatory factor gene family. Genes Dev. 1989, 3, 2050–2061. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Ono, Y.; Urata, Y.; Goto, S.; Nakagawa, S.; Humbert, P.O.; Li, T.S.; Zammit, P.S. Muscle stem cell fate is controlled by the cell-polarity protein Scrib. Cell Rep. 2015, 10, 1135–1148. [Google Scholar] [CrossRef] [Green Version]
  15. Montarras, D.; L’Honore, A.; Buckingham, M. Lying low but ready for action: The quiescent muscle satellite cell. FEBS J. 2013, 280, 4036–4050. [Google Scholar] [CrossRef]
  16. Sacco, A.; Doyonnas, R.; Kraft, P.; Vitorovic, S.; Blau, H.M. Self-renewal and expansion of single transplanted muscle stem cells. Nature 2008, 456, 502–506. [Google Scholar] [CrossRef] [Green Version]
  17. Dellavalle, A.; Sampaolesi, M.; Tonlorenzi, R.; Tagliafico, E.; Sacchetti, B.; Perani, L.; Innocenzi, A.; Galvez, B.G.; Messina, G.; Morosetti, R.; et al. Pericytes of human skeletal muscle are myogenic precursors distinct from satellite cells. Nat. Cell Biol. 2007, 9, 255–267. [Google Scholar] [CrossRef] [Green Version]
  18. Mitchell, K.J.; Pannerec, A.; Cadot, B.; Parlakian, A.; Besson, V.; Gomes, E.R.; Marazzi, G.; Sassoon, D.A. Identification and characterization of a non-satellite cell muscle resident progenitor during postnatal development. Nat. Cell Biol. 2010, 12, 257–266. [Google Scholar] [CrossRef] [Green Version]
  19. Liu, N.; Garry, G.A.; Li, S.; Bezprozvannaya, S.; Sanchez-Ortiz, E.; Chen, B.; Shelton, J.M.; Jaichander, P.; Bassel-Duby, R.; Olson, E.N. A Twist2-dependent progenitor cell contributes to adult skeletal muscle. Nat. Cell Biol. 2017, 19, 202–213. [Google Scholar] [CrossRef] [Green Version]
  20. Harvey, N.; Dennison, E.; Cooper, C. Osteoporosis: Impact on health and economics. Nat. Rev. Rheumatol. 2010, 6, 99–105. [Google Scholar] [CrossRef]
  21. Zammit, P.S.; Golding, J.P.; Nagata, Y.; Hudon, V.; Partridge, T.A.; Beauchamp, J.R. Muscle satellite cells adopt divergent fates: A mechanism for self-renewal? J. Cell Biol. 2004, 166, 347–357. [Google Scholar] [CrossRef] [PubMed]
  22. Davis, R.L.; Weintraub, H.; Lassar, A.B. Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell 1987, 51, 987–1000. [Google Scholar] [CrossRef]
  23. Albini, S.; Coutinho, P.; Malecova, B.; Giordani, L.; Savchenko, A.; Forcales, S.V.; Puri, P.L. Epigenetic reprogramming of human embryonic stem cells into skeletal muscle cells and generation of contractile myospheres. Cell Rep. 2013, 3, 661–670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Dekel, I.; Magal, Y.; Pearson-White, S.; Emerson, C.P.; Shani, M. Conditional conversion of ES cells to skeletal muscle by an exogenous MyoD1 gene. New Biol. 1992, 4, 217–224. [Google Scholar] [PubMed]
  25. Goudenege, S.; Lebel, C.; Huot, N.B.; Dufour, C.; Fujii, I.; Gekas, J.; Rousseau, J.; Tremblay, J.P. Myoblasts derived from normal hESCs and dystrophic hiPSCs efficiently fuse with existing muscle fibers following transplantation. Mol. Ther. 2012, 20, 2153–2167. [Google Scholar] [CrossRef] [Green Version]
  26. Tanaka, A.; Woltjen, K.; Miyake, K.; Hotta, A.; Ikeya, M.; Yamamoto, T.; Nishino, T.; Shoji, E.; Sehara-Fujisawa, A.; Manabe, Y.; et al. Efficient and reproducible myogenic differentiation from human iPS cells: Prospects for modeling Miyoshi Myopathy in vitro. PLoS ONE 2013, 8, e61540. [Google Scholar] [CrossRef]
  27. Warren, L.; Manos, P.D.; Ahfeldt, T.; Loh, Y.H.; Li, H.; Lau, F.; Ebina, W.; Mandal, P.K.; Smith, Z.D.; Meissner, A.; et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell. 2010, 7, 618–630. [Google Scholar] [CrossRef] [Green Version]
  28. Davis, R.L.; Weintraub, H. Acquisition of myogenic specificity by replacement of three amino acid residues from MyoD into E12. Science 1992, 256, 1027–1030. [Google Scholar] [CrossRef]
  29. Hernandez-Hernandez, J.M.; Garcia-Gonzalez, E.G.; Brun, C.E.; Rudnicki, M.A. The myogenic regulatory factors, determinants of muscle development, cell identity and regeneration. Semin. Cell Dev. Biol. 2017, 72, 10–18. [Google Scholar] [CrossRef]
  30. Megeney, L.A.; Rudnicki, M.A. Determination versus differentiation and the MyoD family of transcription factors. Biochem. Cell Biol. 1995, 73, 723–732. [Google Scholar] [CrossRef]
  31. Rudnicki, M.A.; Jaenisch, R. The MyoD family of transcription factors and skeletal myogenesis. Bioessays 1995, 17, 203–209. [Google Scholar] [CrossRef] [PubMed]
  32. Charge, S.B.; Rudnicki, M.A. Cellular and molecular regulation of muscle regeneration. Physiol. Rev. 2004, 84, 209–238. [Google Scholar] [CrossRef] [PubMed]
  33. Rudnicki, M.A.; Braun, T.; Hinuma, S.; Jaenisch, R. Inactivation of MyoD in mice leads to up-regulation of the myogenic HLH gene Myf-5 and results in apparently normal muscle development. Cell 1992, 71, 383–390. [Google Scholar] [CrossRef]
  34. Rudnicki, M.A.; Schnegelsberg, P.N.; Stead, R.H.; Braun, T.; Arnold, H.H.; Jaenisch, R. MyoD or Myf-5 is required for the formation of skeletal muscle. Cell 1993, 75, 1351–1359. [Google Scholar] [CrossRef]
  35. Kassar-Duchossoy, L.; Giacone, E.; Gayraud-Morel, B.; Jory, A.; Gomes, D.; Tajbakhsh, S. Pax3/Pax7 mark a novel population of primitive myogenic cells during development. Genes Dev. 2005, 19, 1426–1431. [Google Scholar] [CrossRef] [Green Version]
  36. Soleimani, V.D.; Punch, V.G.; Kawabe, Y.; Jones, A.E.; Palidwor, G.A.; Porter, C.J.; Cross, J.W.; Carvajal, J.J.; Kockx, C.E.; van, I.W.F.; et al. Transcriptional dominance of Pax7 in adult myogenesis is due to high-affinity recognition of homeodomain motifs. Dev. Cell 2012, 22, 1208–1220. [Google Scholar] [CrossRef] [Green Version]
  37. Allouh, M.Z.; Yablonka-Reuveni, Z.; Rosser, B.W. Pax7 reveals a greater frequency and concentration of satellite cells at the ends of growing skeletal muscle fibers. J. Histochem. Cytochem. 2008, 56, 77–87. [Google Scholar] [CrossRef] [Green Version]
  38. Gayraud-Morel, B.; Chretien, F.; Jory, A.; Sambasivan, R.; Negroni, E.; Flamant, P.; Soubigou, G.; Coppee, J.Y.; Di Santo, J.; Cumano, A.; et al. Myf5 haploinsufficiency reveals distinct cell fate potentials for adult skeletal muscle stem cells. J. Cell Sci. 2012, 125, 1738–1749. [Google Scholar] [CrossRef] [Green Version]
  39. Kuang, S.; Kuroda, K.; Le Grand, F.; Rudnicki, M.A. Asymmetric self-renewal and commitment of satellite stem cells in muscle. Cell 2007, 129, 999–1010. [Google Scholar] [CrossRef] [Green Version]
  40. Asfour, H.A.; Allouh, M.Z.; Said, R.S. Myogenic regulatory factors: The orchestrators of myogenesis after 30 years of discovery. Exp. Biol. Med. 2018, 243, 118–128. [Google Scholar] [CrossRef]
  41. Relaix, F.; Montarras, D.; Zaffran, S.; Gayraud-Morel, B.; Rocancourt, D.; Tajbakhsh, S.; Mansouri, A.; Cumano, A.; Buckingham, M. Pax3 and Pax7 have distinct and overlapping functions in adult muscle progenitor cells. J. Cell Biol. 2006, 172, 91–102. [Google Scholar] [CrossRef] [PubMed]
  42. Olguin, H.C.; Olwin, B.B. Pax-7 up-regulation inhibits myogenesis and cell cycle progression in satellite cells: A potential mechanism for self-renewal. Dev. Biol. 2004, 275, 375–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Zammit, P.S.; Partridge, T.A.; Yablonka-Reuveni, Z. The skeletal muscle satellite cell: The stem cell that came in from the cold. J. Histochem. Cytochem. 2006, 54, 1177–1191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Fong, A.P.; Tapscott, S.J. Skeletal muscle programming and re-programming. Curr. Opin. Genet. Dev. 2013, 23, 568–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Hughes, S.M.; Koishi, K.; Rudnicki, M.; Maggs, A.M. MyoD protein is differentially accumulated in fast and slow skeletal muscle fibres and required for normal fibre type balance in rodents. Mech. Dev. 1997, 61, 151–163. [Google Scholar] [CrossRef]
  46. Ehlers, M.L.; Celona, B.; Black, B.L. NFATc1 controls skeletal muscle fiber type and is a negative regulator of MyoD activity. Cell Rep. 2014, 8, 1639–1648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Barndt, R.J.; Zhuang, Y. Controlling lymphopoiesis with a combinatorial E-protein code. Cold Spring. Harb. Symp. Quant. Biol. 1999, 64, 45–50. [Google Scholar] [CrossRef]
  48. Blackwell, T.K.; Weintraub, H. Differences and similarities in DNA-binding preferences of MyoD and E2A protein complexes revealed by binding site selection. Science 1990, 250, 1104–1110. [Google Scholar] [CrossRef] [Green Version]
  49. Ishibashi, J.; Perry, R.L.; Asakura, A.; Rudnicki, M.A. MyoD induces myogenic differentiation through cooperation of its NH2- and COOH-terminal regions. J. Cell Biol. 2005, 171, 471–482. [Google Scholar] [CrossRef] [Green Version]
  50. Blais, A.; Tsikitis, M.; Acosta-Alvear, D.; Sharan, R.; Kluger, Y.; Dynlacht, B.D. An initial blueprint for myogenic differentiation. Genes Dev. 2005, 19, 553–569. [Google Scholar] [CrossRef] [Green Version]
  51. Cao, Y.; Kumar, R.M.; Penn, B.H.; Berkes, C.A.; Kooperberg, C.; Boyer, L.A.; Young, R.A.; Tapscott, S.J. Global and gene-specific analyses show distinct roles for Myod and Myog at a common set of promoters. EMBO J. 2006, 25, 502–511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Soleimani, V.D.; Yin, H.; Jahani-Asl, A.; Ming, H.; Kockx, C.E.; van Ijcken, W.F.; Grosveld, F.; Rudnicki, M.A. Snail regulates MyoD binding-site occupancy to direct enhancer switching and differentiation-specific transcription in myogenesis. Mol. Cell 2012, 47, 457–468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Cao, Y.; Yao, Z.; Sarkar, D.; Lawrence, M.; Sanchez, G.J.; Parker, M.H.; MacQuarrie, K.L.; Davison, J.; Morgan, M.T.; Ruzzo, W.L.; et al. Genome-wide MyoD binding in skeletal muscle cells: A potential for broad cellular reprogramming. Dev. Cell 2010, 18, 662–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Mousavi, K.; Zare, H.; Dell’orso, S.; Grontved, L.; Gutierrez-Cruz, G.; Derfoul, A.; Hager, G.L.; Sartorelli, V. eRNAs promote transcription by establishing chromatin accessibility at defined genomic loci. Mol. Cell 2013, 51, 606–617. [Google Scholar] [CrossRef] [Green Version]
  55. Bolos, V.; Peinado, H.; Perez-Moreno, M.A.; Fraga, M.F.; Esteller, M.; Cano, A. The transcription factor Slug represses E-cadherin expression and induces epithelial to mesenchymal transitions: A comparison with Snail and E47 repressors. J. Cell Sci. 2003, 116, 499–511. [Google Scholar] [CrossRef] [Green Version]
  56. Hajra, K.M.; Chen, D.Y.; Fearon, E.R. The SLUG zinc-finger protein represses E-cadherin in breast cancer. Cancer Res. 2002, 62, 1613–1618. [Google Scholar]
  57. Peinado, H.; Olmeda, D.; Cano, A. Snail, Zeb and bHLH factors in tumour progression: An alliance against the epithelial phenotype? Nat. Rev. Cancer 2007, 7, 415–428. [Google Scholar] [CrossRef]
  58. Loper, H.B.; La Sala, M.; Dotson, C.; Steinle, N. Taste perception, associated hormonal modulation, and nutrient intake. Nutr. Rev. 2015, 73, 83–91. [Google Scholar] [CrossRef] [Green Version]
  59. Finger, T.E.; Kinnamon, S.C. Taste isn’t just for taste buds anymore. F1000 Biol. Rep. 2011, 3, 20. [Google Scholar] [CrossRef] [Green Version]
  60. Nelson, G.; Hoon, M.A.; Chandrashekar, J.; Zhang, Y.; Ryba, N.J.; Zuker, C.S. Mammalian sweet taste receptors. Cell 2001, 106, 381–390. [Google Scholar] [CrossRef] [Green Version]
  61. Nelson, G.; Chandrashekar, J.; Hoon, M.A.; Feng, L.; Zhao, G.; Ryba, N.J.; Zuker, C.S. An amino-acid taste receptor. Nature 2002, 416, 199–202. [Google Scholar] [CrossRef] [PubMed]
  62. Zhao, G.Q.; Zhang, Y.; Hoon, M.A.; Chandrashekar, J.; Erlenbach, I.; Ryba, N.J.; Zuker, C.S. The receptors for mammalian sweet and umami taste. Cell 2003, 115, 255–266. [Google Scholar] [CrossRef] [Green Version]
  63. Li, X.; Staszewski, L.; Xu, H.; Durick, K.; Zoller, M.; Adler, E. Human receptors for sweet and umami taste. Proc. Natl. Acad. Sci. USA 2002, 99, 4692–4696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Hofer, D.; Puschel, B.; Drenckhahn, D. Taste receptor-like cells in the rat gut identified by expression of α-gustducin. Proc. Natl. Acad. Sci. USA 1996, 93, 6631–6634. [Google Scholar] [CrossRef] [Green Version]
  65. Masubuchi, Y.; Nakagawa, Y.; Ma, J.; Sasaki, T.; Kitamura, T.; Yamamoto, Y.; Kurose, H.; Kojima, I.; Shibata, H. A novel regulatory function of sweet taste-sensing receptor in adipogenic differentiation of 3T3-L1 cells. PLoS ONE 2013, 8, e54500. [Google Scholar] [CrossRef] [Green Version]
  66. Oya, M.; Suzuki, H.; Watanabe, Y.; Sato, M.; Tsuboi, T. Amino acid taste receptor regulates insulin secretion in pancreatic β-cell line MIN6 cells. Genes Cells. 2011, 16, 608–616. [Google Scholar] [CrossRef]
  67. Ren, X.; Zhou, L.; Terwilliger, R.; Newton, S.S.; de Araujo, I.E. Sweet taste signaling functions as a hypothalamic glucose sensor. Front. Integr. Neurosci. 2009, 3, 12. [Google Scholar] [CrossRef] [Green Version]
  68. Foster, S.R.; Porrello, E.R.; Purdue, B.; Chan, H.W.; Voigt, A.; Frenzel, S.; Hannan, R.D.; Moritz, K.M.; Simmons, D.G.; Molenaar, P.; et al. Expression, regulation and putative nutrient-sensing function of taste GPCRs in the heart. PLoS ONE 2013, 8, e64579. [Google Scholar] [CrossRef] [Green Version]
  69. Miyamoto, T.; Wright, G.; Amrein, H. Nutrient sensors. Curr. Biol. 2013, 23, RP369-73. [Google Scholar] [CrossRef] [Green Version]
  70. Kokabu, S.; Lowery, J.W.; Toyono, T.; Seta, Y.; Hitomi, S.; Sato, T.; Enoki, Y.; Okubo, M.; Fukushima, Y.; Yoda, T. Muscle regulatory factors regulate T1R3 taste receptor expression. Biochem. Biophys. Res. Commun. 2015, 468, 568–573. [Google Scholar] [CrossRef]
  71. Hirata, Y.; Toyono, T.; Kokabu, S.; Obikane, Y.; Kataoka, S.; Nakatomi, M.; Masaki, C.; Hosokawa, R.; Seta, Y. Krüppel-like factor 5 (Klf5) regulates expression of mouse T1R1 amino acid receptor gene (Tas1r1) in C2C12 myoblast cells. Biomed. Res. 2019, 40, 67–78. [Google Scholar] [CrossRef] [Green Version]
  72. Obikane, Y.; Toyono, T.; Kokabu, S.; Matsuyama, K.; Kataoka, S.; Nakatomi, M.; Hosokawa, R.; Seta, Y. Myogenic differentiation 1 and transcription factor 12 activate the gene expression of mouse taste receptor type 1 member 1. J. Oral Biosci. 2021, 63, 420–428. [Google Scholar] [CrossRef] [PubMed]
  73. Sogawa, K.; Imataka, H.; Yamasaki, Y.; Kusume, H.; Abe, H.; Fujii-Kuriyama, Y. cDNA cloning and transcriptional properties of a novel GC box-binding protein, BTEB2. Nucleic Acids Res. 1993, 21, 1527–1532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Hayashi, S.; Manabe, I.; Suzuki, Y.; Relaix, F.; Oishi, Y. Klf5 regulates muscle differentiation by directly targeting muscle-specific genes in cooperation with MyoD in mice. Elife 2016, 5, e17462. [Google Scholar] [CrossRef] [PubMed]
  75. Wauson, E.M.; Zaganjor, E.; Lee, A.Y.; Guerra, M.L.; Ghosh, A.B.; Bookout, A.L.; Chambers, C.P.; Jivan, A.; McGlynn, K.; Hutchison, M.R.; et al. The G protein-coupled taste receptor T1R1/T1R3 regulates mTORC1 and autophagy. Mol. Cell 2012, 47, 851–862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Wauson, E.M.; Lorente-Rodriguez, A.; Cobb, M.H. Minireview: Nutrient sensing by G protein-coupled receptors. Mol. Endocrinol. 2013, 27, 1188–1197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Shimizu, N.; Yoshikawa, N.; Ito, N.; Maruyama, T.; Suzuki, Y.; Takeda, S.; Nakae, J.; Tagata, Y.; Nishitani, S.; Takehana, K.; et al. Crosstalk between glucocorticoid receptor and nutritional sensor mTOR in skeletal muscle. Cell Metab. 2011, 13, 170–182. [Google Scholar] [CrossRef] [Green Version]
  78. Bjornson, C.R.; Cheung, T.H.; Liu, L.; Tripathi, P.V.; Steeper, K.M.; Rando, T.A. Notch signaling is necessary to maintain quiescence in adult muscle stem cells. Stem Cells 2012, 30, 232–242. [Google Scholar] [CrossRef] [Green Version]
  79. Brohl, D.; Vasyutina, E.; Czajkowski, M.T.; Griger, J.; Rassek, C.; Rahn, H.P.; Purfurst, B.; Wende, H.; Birchmeier, C. Colonization of the satellite cell niche by skeletal muscle progenitor cells depends on Notch signals. Dev. Cell. 2012, 23, 469–481. [Google Scholar] [CrossRef] [Green Version]
  80. Delfini, M.C.; Hirsinger, E.; Pourquie, O.; Duprez, D. Delta 1-activated notch inhibits muscle differentiation without affecting Myf5 and Pax3 expression in chick limb myogenesis. Development 2000, 127, 5213–5224. [Google Scholar] [CrossRef]
  81. Esteves de Lima, J.; Bonnin, M.A.; Birchmeier, C.; Duprez, D. Muscle contraction is required to maintain the pool of muscle progenitors via YAP and NOTCH during fetal myogenesis. Elife 2016, 5, e15593. [Google Scholar] [CrossRef] [PubMed]
  82. Hirsinger, E.; Malapert, P.; Dubrulle, J.; Delfini, M.C.; Duprez, D.; Henrique, D.; Ish-Horowicz, D.; Pourquie, O. Notch signalling acts in postmitotic avian myogenic cells to control MyoD activation. Development 2001, 128, 107–116. [Google Scholar] [CrossRef] [PubMed]
  83. Lahmann, I.; Brohl, D.; Zyrianova, T.; Isomura, A.; Czajkowski, M.T.; Kapoor, V.; Griger, J.; Ruffault, P.L.; Mademtzoglou, D.; Zammit, P.S.; et al. Oscillations of MyoD and Hes1 proteins regulate the maintenance of activated muscle stem cells. Genes Dev. 2019, 33, 524–535. [Google Scholar] [CrossRef] [Green Version]
  84. Mourikis, P.; Sambasivan, R.; Castel, D.; Rocheteau, P.; Bizzarro, V.; Tajbakhsh, S. A critical requirement for notch signaling in maintenance of the quiescent skeletal muscle stem cell state. Stem Cells 2012, 30, 243–252. [Google Scholar] [CrossRef] [PubMed]
  85. Mourikis, P.; Gopalakrishnan, S.; Sambasivan, R.; Tajbakhsh, S. Cell-autonomous Notch activity maintains the temporal specification potential of skeletal muscle stem cells. Development 2012, 139, 4536–4548. [Google Scholar] [CrossRef] [Green Version]
  86. Pascoal, S.; Esteves de Lima, J.; Leslie, J.D.; Hughes, S.M.; Saude, L. Notch signalling is required for the formation of structurally stable muscle fibres in zebrafish. PLoS ONE 2013, 8, e68021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Schuster-Gossler, K.; Cordes, R.; Gossler, A. Premature myogenic differentiation and depletion of progenitor cells cause severe muscle hypotrophy in Delta1 mutants. Proc. Natl. Acad. Sci. USA 2007, 104, 537–542. [Google Scholar] [CrossRef] [Green Version]
  88. Vasyutina, E.; Lenhard, D.C.; Wende, H.; Erdmann, B.; Epstein, J.A.; Birchmeier, C. RBP-J (Rbpsuh) is essential to maintain muscle progenitor cells and to generate satellite cells. Proc. Natl. Acad. Sci. USA 2007, 104, 4443–4448. [Google Scholar] [CrossRef] [Green Version]
  89. Borok, M.J.; Mademtzoglou, D.; Relaix, F. Bu-M-P-ing Iron: How BMP Signaling Regulates Muscle Growth and Regeneration. J. Dev. Biol. 2020, 8, 4. [Google Scholar] [CrossRef] [Green Version]
  90. Wang, H.; Noulet, F.; Edom-Vovard, F.; Tozer, S.; Le Grand, F.; Duprez, D. Bmp signaling at the tips of skeletal muscles regulates the number of fetal muscle progenitors and satellite cells during development. Dev. Cell 2010, 18, 643–654. [Google Scholar] [CrossRef] [Green Version]
  91. Sartori, R.; Schirwis, E.; Blaauw, B.; Bortolanza, S.; Zhao, J.; Enzo, E.; Stantzou, A.; Mouisel, E.; Toniolo, L.; Ferry, A.; et al. BMP signaling controls muscle mass. Nat. Genet. 2013, 45, 1309–1318. [Google Scholar] [CrossRef] [PubMed]
  92. Stantzou, A.; Schirwis, E.; Swist, S.; Alonso-Martin, S.; Polydorou, I.; Zarrouki, F.; Mouisel, E.; Beley, C.; Julien, A.; Le Grand, F.; et al. BMP signaling regulates satellite cell-dependent postnatal muscle growth. Development 2017, 144, 2737–2747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Friedrichs, M.; Wirsdoerfer, F.; Flohe, S.B.; Schneider, S.; Wuelling, M.; Vortkamp, A. BMP signaling balances proliferation and differentiation of muscle satellite cell descendants. BMC Cell Biol. 2011, 12, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Ono, Y.; Calhabeu, F.; Morgan, J.E.; Katagiri, T.; Amthor, H.; Zammit, P.S. BMP signalling permits population expansion by preventing premature myogenic differentiation in muscle satellite cells. Cell Death Differ. 2011, 18, 222–234. [Google Scholar] [CrossRef] [Green Version]
  95. Katagiri, T.; Yamaguchi, A.; Komaki, M.; Abe, E.; Takahashi, N.; Ikeda, T.; Rosen, V.; Wozney, J.M.; Fujisawa-Sehara, A.; Suda, T. Bone morphogenetic protein-2 converts the differentiation pathway of C2C12 myoblasts into the osteoblast lineage. J. Cell Biol. 1994, 127, 1755–1766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Nojima, J.; Kanomata, K.; Takada, Y.; Fukuda, T.; Kokabu, S.; Ohte, S.; Takada, T.; Tsukui, T.; Yamamoto, T.S.; Sasanuma, H.; et al. Dual roles of smad proteins in the conversion from myoblasts to osteoblastic cells by bone morphogenetic proteins. J. Biol. Chem. 2010, 285, 15577–15586. [Google Scholar] [CrossRef] [Green Version]
  97. Hutcheson, D.A.; Zhao, J.; Merrell, A.; Haldar, M.; Kardon, G. Embryonic and fetal limb myogenic cells are derived from developmentally distinct progenitors and have different requirements for β-catenin. Genes Dev. 2009, 23, 997–1013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Le Grand, F.; Grifone, R.; Mourikis, P.; Houbron, C.; Gigaud, C.; Pujol, J.; Maillet, M.; Pages, G.; Rudnicki, M.; Tajbakhsh, S.; et al. Six1 regulates stem cell repair potential and self-renewal during skeletal muscle regeneration. J. Cell Biol. 2012, 198, 815–832. [Google Scholar] [CrossRef] [Green Version]
  99. Chal, J.; Al Tanoury, Z.; Hestin, M.; Gobert, B.; Aivio, S.; Hick, A.; Cherrier, T.; Nesmith, A.P.; Parker, K.K.; Pourquie, O. Generation of human muscle fibers and satellite-like cells from human pluripotent stem cells in vitro. Nat. Protoc. 2016, 11, 1833–1850. [Google Scholar] [CrossRef] [Green Version]
  100. Chal, J.; Oginuma, M.; Al Tanoury, Z.; Gobert, B.; Sumara, O.; Hick, A.; Bousson, F.; Zidouni, Y.; Mursch, C.; Moncuquet, P.; et al. Differentiation of pluripotent stem cells to muscle fiber to model Duchenne muscular dystrophy. Nat. Biotechnol. 2015, 33, 962–969. [Google Scholar] [CrossRef] [Green Version]
  101. Zhao, M.; Tazumi, A.; Takayama, S.; Takenaka-Ninagawa, N.; Nalbandian, M.; Nagai, M.; Nakamura, Y.; Nakasa, M.; Watanabe, A.; Ikeya, M.; et al. Induced Fetal Human Muscle Stem Cells with High Therapeutic Potential in a Mouse Muscular Dystrophy Model. Stem Cell Rep. 2020, 15, 80–94. [Google Scholar] [CrossRef]
  102. Jennings, B.H.; Ish-Horowicz, D. The Groucho/TLE/Grg family of transcriptional co-repressors. Genome Biol. 2008, 9, 205. [Google Scholar] [PubMed]
  103. Cavallo, R.A.; Cox, R.T.; Moline, M.M.; Roose, J.; Polevoy, G.A.; Clevers, H.; Peifer, M.; Bejsovec, A. Drosophila Tcf and Groucho interact to repress Wingless signalling activity. Nature 1998, 395, 604–608. [Google Scholar] [CrossRef] [PubMed]
  104. Roose, J.; Molenaar, M.; Peterson, J.; Hurenkamp, J.; Brantjes, H.; Moerer, P.; van de Wetering, M.; Destree, O.; Clevers, H. The Xenopus Wnt effector XTcf-3 interacts with Groucho-related transcriptional repressors. Nature 1998, 395, 608–612. [Google Scholar] [CrossRef] [PubMed]
  105. Daniels, D.L.; Weis, W.I. β-catenin directly displaces Groucho/TLE repressors from Tcf/Lef in Wnt-mediated transcription activation. Nat. Struct. Mol. Biol. 2005, 12, 364–371. [Google Scholar] [CrossRef]
  106. Kokabu, S.; Sato, T.; Ohte, S.; Enoki, Y.; Okubo, M.; Hayashi, N.; Nojima, J.; Tsukamoto, S.; Fukushima, Y.; Sakata, Y.; et al. Expression of TLE3 by bone marrow stromal cells is regulated by canonical Wnt signaling. FEBS Lett. 2014, 588, 614–619. [Google Scholar] [CrossRef] [Green Version]
  107. Stifani, S.; Blaumueller, C.M.; Redhead, N.J.; Hill, R.E.; Artavanis-Tsakonas, S. Human homologs of a Drosophila Enhancer of split gene product define a novel family of nuclear proteins. Nat. Genet. 1992, 2, 119–127. [Google Scholar] [CrossRef]
  108. Chen, G.; Nguyen, P.H.; Courey, A.J. A role for Groucho tetramerization in transcriptional repression. Mol. Cell Biol. 1998, 18, 7259–7268. [Google Scholar] [CrossRef] [Green Version]
  109. Pinto, M.; Lobe, C.G. Products of the grg (Groucho-related gene) family can dimerize through the amino-terminal Q domain. J. Biol. Chem. 1996, 271, 33026–33031. [Google Scholar] [CrossRef] [Green Version]
  110. Song, H.; Hasson, P.; Paroush, Z.; Courey, A.J. Groucho oligomerization is required for repression in vivo. Mol. Cell. Biol. 2004, 24, 4341–4350. [Google Scholar] [CrossRef] [Green Version]
  111. Grbavec, D.; Lo, R.; Liu, Y.; Stifani, S. Transducin-like Enhancer of split 2, a mammalian homologue of Drosophila Groucho, acts as a transcriptional repressor, interacts with Hairy/Enhancer of split proteins, and is expressed during neuronal development. Eur. J. Biochem. 1998, 258, 339–349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Brantjes, H.; Roose, J.; van De Wetering, M.; Clevers, H. All Tcf HMG box transcription factors interact with Groucho-related co-repressors. Nucleic Acids Res. 2001, 29, 1410–1419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Miyasaka, H.; Okabe, S.; Ishiguro, K.; Uchida, T.; Hirokawa, N. Interaction of the tail domain of high molecular weight subunits of neurofilaments with the COOH-terminal region of tubulin and its regulation by tau protein kinase II. J. Biol. Chem. 1993, 268, 22695–22702. [Google Scholar] [CrossRef]
  114. Fisher, A.L.; Caudy, M. Groucho proteins: Transcriptional corepressors for specific subsets of DNA-binding transcription factors in vertebrates and invertebrates. Genes Dev. 1998, 12, 1931–1940. [Google Scholar] [CrossRef] [Green Version]
  115. Parkhurst, S.M. Groucho: Making its Marx as a transcriptional co-repressor. Trends Genet. 1998, 14, 130–132. [Google Scholar] [CrossRef]
  116. Leon, C.; Lobe, C.G. Grg3, a murine Groucho-related gene, is expressed in the developing nervous system and in mesenchyme-induced epithelial structures. Dev. Dyn. 1997, 208, 11–24. [Google Scholar] [CrossRef]
  117. Chamberlain, G.; Fox, J.; Ashton, B.; Middleton, J. Concise review: Mesenchymal stem cells: Their phenotype, differentiation capacity, immunological features, and potential for homing. Stem. Cells 2007, 25, 2739–2749. [Google Scholar] [CrossRef] [Green Version]
  118. Kokabu, S.; Nguyen, T.; Ohte, S.; Sato, T.; Katagiri, T.; Yoda, T.; Rosen, V. TLE3, transducing-like enhancer of split 3, suppresses osteoblast differentiation of bone marrow stromal cells. Biochem. Biophys. Res. Commun. 2013, 438, 205–210. [Google Scholar] [CrossRef]
  119. Kokabu, S.; Lowery, J.W.; Jimi, E. Cell Fate and Differentiation of Bone Marrow Mesenchymal Stem Cells. Stem Cells Int. 2016, 2016, 3753581. [Google Scholar] [CrossRef] [Green Version]
  120. Tontonoz, P.; Hu, E.; Spiegelman, B.M. Stimulation of adipogenesis in fibroblasts by PPARγ2, a lipid-activated transcription factor. Cell 1994, 79, 1147–1156. [Google Scholar] [CrossRef]
  121. Komori, T.; Yagi, H.; Nomura, S.; Yamaguchi, A.; Sasaki, K.; Deguchi, K.; Shimizu, Y.; Bronson, R.T.; Gao, Y.H.; Inada, M.; et al. Targeted disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts. Cell 1997, 89, 755–764. [Google Scholar] [CrossRef] [Green Version]
  122. Villanueva, C.J.; Waki, H.; Godio, C.; Nielsen, R.; Chou, W.L.; Vargas, L.; Wroblewski, K.; Schmedt, C.; Chao, L.C.; Boyadjian, R.; et al. TLE3 is a dual-function transcriptional coregulator of adipogenesis. Cell Metab. 2011, 13, 413–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Villanueva, C.J.; Vergnes, L.; Wang, J.; Drew, B.G.; Hong, C.; Tu, Y.; Hu, Y.; Peng, X.; Xu, F.; Saez, E.; et al. Adipose subtype-selective recruitment of TLE3 or Prdm16 by PPARγ specifies lipid storage versus thermogenic gene programs. Cell Metab. 2013, 17, 423–435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Kokabu, S.; Nakatomi, C.; Matsubara, T.; Ono, Y.; Addison, W.N.; Lowery, J.W.; Urata, M.; Hudnall, A.M.; Hitomi, S.; Nakatomi, M.; et al. The transcriptional co-repressor TLE3 regulates myogenic differentiation by repressing the activity of the MyoD transcription factor. J. Biol. Chem. 2017, 292, 12885–12894. [Google Scholar] [CrossRef] [Green Version]
  125. Jennings, B.H.; Pickles, L.M.; Wainwright, S.M.; Roe, S.M.; Pearl, L.H.; Ish-Horowicz, D. Molecular recognition of transcriptional repressor motifs by the WD domain of the Groucho/TLE corepressor. Mol. Cell 2006, 22, 645–655. [Google Scholar] [CrossRef]
  126. Tapscott, S.J. The circuitry of a master switch: Myod and the regulation of skeletal muscle gene transcription. Development 2005, 132, 2685–2695. [Google Scholar] [CrossRef] [Green Version]
  127. Jimi, E.; Fei, H.; Nakatomi, C. NF-κB Signaling Regulates Physiological and Pathological Chondrogenesis. Int. J. Mol. Sci. 2019, 20, 6275. [Google Scholar] [CrossRef] [Green Version]
  128. Thaloor, D.; Miller, K.J.; Gephart, J.; Mitchell, P.O.; Pavlath, G.K. Systemic administration of the NF-κB inhibitor curcumin stimulates muscle regeneration after traumatic injury. Am. J. Physiol. 1999, 277, C320–C329. [Google Scholar] [CrossRef]
  129. Guttridge, D.C.; Albanese, C.; Reuther, J.Y.; Pestell, R.G.; Baldwin, A.S., Jr. NF-κB controls cell growth and differentiation through transcriptional regulation of cyclin D1. Mol. Cell Biol. 1999, 19, 5785–5799. [Google Scholar] [CrossRef] [Green Version]
  130. Guttridge, D.C.; Mayo, M.W.; Madrid, L.V.; Wang, C.Y.; Baldwin, A.S., Jr. NF-κB-induced loss of MyoD messenger RNA: Possible role in muscle decay and cachexia. Science 2000, 289, 2363–2366. [Google Scholar] [CrossRef] [Green Version]
  131. Langen, R.C.; Schols, A.M.; Kelders, M.C.; Wouters, E.F.; Janssen-Heininger, Y.M. Inflammatory cytokines inhibit myogenic differentiation through activation of nuclear factor-κB. FASEB J. 2001, 15, 1169–1180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Shirakawa, T.; Rojasawasthien, T.; Inoue, A.; Matsubara, T.; Kawamoto, T.; Kokabu, S. Tumor necrosis factor alpha regulates myogenesis to inhibit differentiation and promote proliferation in satellite cells. Biochem. Biophys. Res. Commun. 2021, 580, 35–40. [Google Scholar] [CrossRef] [PubMed]
  133. Guo, K.; Wang, J.; Andres, V.; Smith, R.C.; Walsh, K. MyoD-induced expression of p21 inhibits cyclin-dependent kinase activity upon myocyte terminal differentiation. Mol. Cell Biol. 1995, 15, 3823–3829. [Google Scholar] [PubMed] [Green Version]
  134. Lehtinen, S.K.; Rahkila, P.; Helenius, M.; Salminen, A. Down-regulation of nuclear binding activities of OXBOX-REBOX transcription factors during cellular senescence. Biochem. Biophys. Res. Commun. 1996, 226, 403–406. [Google Scholar] [CrossRef]
  135. Kaliman, P.; Canicio, J.; Testar, X.; Palacin, M.; Zorzano, A. Insulin-like growth factor-II, phosphatidylinositol 3-kinase, nuclear factor-κB and inducible nitric-oxide synthase define a common myogenic signaling pathway. J. Biol. Chem. 1999, 274, 17437–17444. [Google Scholar] [CrossRef] [Green Version]
  136. Conejo, R.; de Alvaro, C.; Benito, M.; Cuadrado, A.; Lorenzo, M. Insulin restores differentiation of Ras-transformed C2C12 myoblasts by inducing NF-κB through an AKT/P70S6K/p38-MAPK pathway. Oncogene 2002, 21, 3739–3753. [Google Scholar] [CrossRef]
  137. Yam, C.H.; Fung, T.K.; Poon, R.Y. Cyclin A in cell cycle control and cancer. Cell Mol. Life Sci. 2002, 59, 1317–1326. [Google Scholar] [CrossRef]
Figure 1. Protein structure of myogenic regulatory factors.
Figure 1. Protein structure of myogenic regulatory factors.
Cells 11 01493 g001
Figure 2. Expression of myogenic regulatory factors during different developmental stages of satellite cells. Cells can divide into two groups, myogenic stem cells (Pax7+/Myf5) (A) and myogenic precursor cells (Pax7+/Myf5+) (B). The figures are referred from the review paper by Asfour et al. [40].
Figure 2. Expression of myogenic regulatory factors during different developmental stages of satellite cells. Cells can divide into two groups, myogenic stem cells (Pax7+/Myf5) (A) and myogenic precursor cells (Pax7+/Myf5+) (B). The figures are referred from the review paper by Asfour et al. [40].
Cells 11 01493 g002
Figure 3. Myogenic regulatory factors regulate the expression levels of T1R1 and T1R3 during myogenesis. MyoD and myogenin binding the promoter region of T1R3 (A). The heterodimer of MyoD and Tcf12 interacts with Klf5 and binds to promoter region of T1R1 (B).
Figure 3. Myogenic regulatory factors regulate the expression levels of T1R1 and T1R3 during myogenesis. MyoD and myogenin binding the promoter region of T1R3 (A). The heterodimer of MyoD and Tcf12 interacts with Klf5 and binds to promoter region of T1R1 (B).
Cells 11 01493 g003
Figure 4. Model for transcriptional regulation of MyoD by the transcription cofactor, TLE3.
Figure 4. Model for transcriptional regulation of MyoD by the transcription cofactor, TLE3.
Cells 11 01493 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Shirakawa, T.; Toyono, T.; Inoue, A.; Matsubara, T.; Kawamoto, T.; Kokabu, S. Factors Regulating or Regulated by Myogenic Regulatory Factors in Skeletal Muscle Stem Cells. Cells 2022, 11, 1493. https://doi.org/10.3390/cells11091493

AMA Style

Shirakawa T, Toyono T, Inoue A, Matsubara T, Kawamoto T, Kokabu S. Factors Regulating or Regulated by Myogenic Regulatory Factors in Skeletal Muscle Stem Cells. Cells. 2022; 11(9):1493. https://doi.org/10.3390/cells11091493

Chicago/Turabian Style

Shirakawa, Tomohiko, Takashi Toyono, Asako Inoue, Takuma Matsubara, Tatsuo Kawamoto, and Shoichiro Kokabu. 2022. "Factors Regulating or Regulated by Myogenic Regulatory Factors in Skeletal Muscle Stem Cells" Cells 11, no. 9: 1493. https://doi.org/10.3390/cells11091493

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop