Fact-checked by Grok 2 weeks ago

MyoD

MyoD, also known as myoblast determination protein 1, is a basic helix-loop-helix (bHLH) that serves as a master regulator of development by initiating myogenic lineage commitment and activating muscle-specific . Discovered in 1987 through a pioneering screen in 10T1/2 fibroblasts treated with 5-azacytidine, MyoD was identified for its unique ability to convert non-muscle cells into stable myoblasts, marking the first demonstration of a single factor reprogramming cell fate toward . As a member of the myogenic regulatory factor (MRF) family—which includes Myf5, myogenin, and MRF4—MyoD shares structural homology in its bHLH domain, enabling dimerization with E proteins to bind DNA motifs (CANNTG) and orchestrate for . MyoD plays a pivotal role in embryonic muscle formation, where it is expressed starting around embryonic day 10.5 in the hypaxial and limb buds, regulated by signaling pathways including Wnt (promoting ) and BMP4 (inhibiting via antagonists like Noggin), driving the specification of myogenic progenitors from somites. In contrast to Myf5, which primarily handles early , MyoD excels at promoting both and progression to by recruiting complexes via interactions with BAF60c and Pbx proteins, thereby activating downstream targets like myogenin. Genetic studies reveal functional redundancy with Myf5 in myoblast commitment—double knockouts abolish all —but MyoD is essential for maintaining satellite cell numbers and function for postnatal muscle regeneration and repair. Beyond development, MyoD's regulatory functions extend to adult muscle , where it responds to signals from /7 and to reactivate quiescent satellite cells, ensuring tissue maintenance and adaptation to exercise or stress. Its activity is finely tuned by post-translational modifications and interactions with non-coding RNAs, preventing that could disrupt other lineages. Dysregulation of MyoD has been implicated in muscle disorders, including —where it promotes tumor survival via epigenetic mechanisms—and dystrophies. Recent research (as of 2025) has revealed MyoD's repressive transcriptional role, offering new avenues for therapies in .

Discovery and History

Initial Identification

MyoD was first identified in 1987 by researchers in Weintraub's laboratory at the Center through a functional approach aimed at isolating genes responsible for myoblast . Using subtractive hybridization, they enriched for cDNAs from myoblasts induced by 5-azacytidine treatment of C3H/10T1/2 mouse embryonic fibroblasts, specifically expressed in these myoblasts but absent in non-myogenic 10T1/2 fibroblasts, generating a library of candidate transcripts. These cDNAs were then transfected into 10T1/2 fibroblasts, and clones were screened for their ability to induce myogenic conversion, leading to the isolation of the MyoD gene as a single cDNA capable of activating the muscle program. Subsequent characterization revealed MyoD as a with a molecular weight of approximately 45 kDa, localized to the nuclei of proliferating myoblasts and differentiated myotubes but undetectable in fibroblasts or other non-muscle cells. Overexpression of MyoD in various non-myogenic cell types, including fibroblasts, resulted in their phenotypic conversion to myoblasts, marked by the formation of multinucleated myotubes and the expression of muscle-specific markers. This demonstrated MyoD's potent regulatory role in initiating from non-committed precursors. Early biochemical studies identified MyoD as a member of the basic helix-loop-helix (bHLH) family of transcription factors, featuring a conserved helix-loop-helix motif that facilitates dimerization and a basic region for DNA binding. MyoD binds specifically to the consensus sequence (CANNTG) in the regulatory regions of muscle genes, enabling sequence-specific transcriptional activation. Initial experiments confirmed that MyoD overexpression activates endogenous muscle-specific genes, such as the myosin heavy chain, in transfected fibroblasts, underscoring its function as a master regulator of myogenic differentiation.

Key Research Milestones

Following the foundational discovery of MyoD in 1987 as a protein capable of converting non-muscle cells into myoblasts, subsequent rapidly expanded the understanding of its regulatory network. In 1989, the identification of additional family members—Myf5, myogenin, and MRF4—established MyoD as part of a core set of myogenic regulatory factors (MRFs) that orchestrate determination and differentiation. Myf5 was isolated as a distinct bHLH inducing in non-muscle cells, while myogenin was characterized for its role in promoting terminal muscle differentiation, and MRF4 was recognized as another MRF with overlapping myogenic potential. These discoveries highlighted the MRF family's coordinated action as master regulators of . In the 1990s, genetic studies using knockout mice revealed functional redundancy among MRFs, particularly between MyoD and Myf5 in embryonic myogenesis. Single MyoD-null mice exhibited normal skeletal muscle development due to compensatory upregulation of Myf5, indicating MyoD's dispensability in embryogenesis but suggesting overlapping roles in myoblast specification. However, double MyoD/Myf5 knockouts resulted in a complete absence of skeletal myoblasts and body wall musculature at birth, confirming that at least one of these factors is essential for myogenic determination and progenitor cell viability during embryogenesis. These findings underscored the interchangeable yet critical functions of MyoD and Myf5 in initiating the myogenic lineage. During the , investigations elucidated MyoD's mechanisms in gene , focusing on its in and sequential activation of muscle genes during . MyoD was shown to recruit complexes to promoters like myogenin, facilitating eviction and transcriptional activation at silent loci. Further studies demonstrated that MyoD initiates early modifications at shared promoters, enabling subsequent MyoG-dependent activation of late genes, thus establishing a temporal hierarchy in myogenic progression. Additionally, homeodomain proteins like Pbx were found to mark MyoD target loci in inactive , enhancing MyoD's access and myogenic potential. These advancements revealed MyoD as a pioneer factor that reprograms landscapes for muscle-specific transcription. Post-2020 research has highlighted MyoD's evolving roles in adult muscle maintenance and , with 2025 studies emphasizing its interactions in the cell niche and metabolic regulation. In adult cells, MyoD-null conditions suppress expression of (ECM) components such as and collagen IV, impairing cell adherence to substrates like and delaying myoblast fusion during regeneration. Concurrently, MyoD has been implicated in regulating oxidative by cooperating with RelB to activate genes like PGC-1β, promoting in slow-twitch fibers and supporting metabolic adaptations in mature muscle. These findings illustrate MyoD's persistent influence on ECM dynamics and in adult .

Gene and Protein Structure

Genomic Organization

The human MYOD1 gene is located on the short arm of at position 11p15.1 and spans approximately 2.6 kb (from 17,719,571 to 17,722,136 bp on the GRCh38 assembly), comprising three exons separated by two introns. The promoter region of MYOD1 features two proximal motifs (CANNTG sequences) located near the transcription start site, which serve as binding sites for MyoD-E protein heterodimers to enable positive autoregulation of the gene. Upstream of the promoter, a core enhancer contains binding sites for paired-box transcription factors and PAX7, which activate MYOD1 expression in somitic and limb bud progenitors during early . The genomic organization of MYOD1 is highly conserved among vertebrates, with the mouse ortholog Myod1 similarly mapping to and exhibiting three exons within a compact ~2.6 kb span. This conservation extends to the promoter and enhancer regions, reflecting shared regulatory logic for myogenic commitment. MyoD orthologs first emerged in the lineage, as demonstrated by a single ancestral MyoD family gene in the Ciona intestinalis, which encodes two differentially expressed protein isoforms through alternative rather than splicing. In humans, MYOD1 primarily generates a single canonical mRNA transcript (NM_002478.5), with limited evidence of alternative splicing; however, minor isoforms involving intron retention have been reported in myogenic contexts, potentially influencing expression levels in differentiating muscle cells. In contrast, orthologs in other vertebrates, such as trout TmyoD1, produce multiple splice variants with tissue-specific patterns, including a non-coding γ-isoform enriched in fast-twitch muscle that regulates transcript stability via nonsense-mediated decay.

Protein Domains

The MyoD protein has a molecular weight of approximately 45 and consists of 320 residues in humans, exhibiting a modular architecture typical of basic helix-loop-helix (bHLH) transcription factors. This structure includes distinct domains that contribute to its biochemical properties, such as DNA recognition, protein-protein interactions, and subcellular localization, all encoded by the three-exon MYOD1 gene on chromosome 11. The core bHLH domain, spanning residues 109–160, is the defining feature of MyoD and comprises two subregions: a basic region (residues 109–125) and a helix-loop-helix motif (residues 126–160). The basic region forms an alpha-helix that directly contacts the major groove of DNA, binding specifically to E-box consensus sequences (CANNTG) found in the promoters of muscle-specific genes; structural studies reveal that key residues like Arg112 and Glu127 make hydrogen bonds with the DNA backbone and bases, ensuring sequence-specific recognition. The adjacent helix-loop-helix motif features two amphipathic alpha-helices separated by a short loop, which positions the helices for coiled-coil interactions that stabilize dimer formation, although detailed partner specificity is determined by flanking sequences. At the , residues 1–53 encompass the (TAD), an acidic region rich in glutamate and aspartate residues that serves as a platform for recruiting coactivators such as acetyltransferases and components of complex. This domain exhibits potent transcriptional activation potential when unmasked, with alanine-scanning identifying critical residues like Glu13 and Asp19 that interact with coactivator surfaces to facilitate and recruitment. MyoD also harbors nuclear localization signals (NLS) embedded within the basic region of the bHLH domain, specifically two bipartite motifs in the basic-helix 1 area (around residues 122–132 and 142–152) that mediate active import through the nuclear pore complex via importin-mediated transport; these signals operate independently and are essential for the protein's nuclear accumulation, as demonstrated by mutagenesis studies showing cytoplasmic retention upon their disruption. Phosphorylation sites within the bHLH domain, notably Ser200 and nearby residues like Ser199, represent key regulatory points influenced by MAPK signaling pathways. For instance, p38γ MAPK phosphorylates Ser199 and Ser200, enhancing MyoD's stability and transcriptional potency during muscle differentiation by altering its conformation and interactions. Similarly, Ser204 can be targeted by other kinases in the MAPK cascade, contributing to fine-tuned modulation of the protein's activity through charge alterations that affect DNA binding affinity and dimer stability.

Expression Patterns

Embryonic Development

MyoD expression initiates in the embryo at embryonic day 10.5 (E10.5) in the ventrolateral (hypaxial) domain of newly formed , marking the early commitment of paraxial mesoderm cells to the myogenic lineage during primary formation. This onset follows the earlier activation of Myf5 at E8.0 in the same domain and is induced by somitogenesis-associated signals, particularly Wnt proteins secreted from the overlying surface , which preferentially activate MyoD in the hypaxial (ventrolateral) domain while Myf5 responds to dorsal signals. By E11.5, MyoD transcripts extend throughout the maturing , supporting the and of myogenic progenitors that contribute to both epaxial and hypaxial muscle masses. MyoD plays a critical role in the development of primary myotomes, which form the initial embryonic muscle layer, and later contributes to secondary myotome expansion around E12.5–E14.5, where additional myoblasts fuse to enlarge muscle fibers. Functional redundancy between MyoD and Myf5 ensures the robustness of this process; single mutants exhibit viable muscle formation, but double knockouts completely abolish all skeletal myoblasts, resulting in the absence of all skeletal muscles, highlighting their overlapping roles in myoblast specification and survival during embryonic myogenesis. This compensation is evident in Myf5-null embryos, where delayed primary myotome formation is rescued by ectopic MyoD activation starting at E10.5. Along the anterior-posterior axis, establish segment-specific patterning of MyoD expression by modulating myogenic regulatory enhancers in the somites, ensuring appropriate muscle identity in different vertebral levels. For instance, Hox group 6 proteins (e.g., Hoxb6) bind to the Myf5/MyoD locus at E9.5–E10.5 to upregulate expression in thoracic somites, promoting hypaxial development and rib-associated muscles, whereas Hox group 10 (e.g., Hoxa10) represses it in regions to prevent ectopic rib formation. In limb buds, MyoD expression emerges at E11.5 in the proximal of fore- and hindlimbs, driven by similar Hox-dependent cues that guide migrating somite-derived progenitors to form limb-specific muscles. Upon commitment, MyoD-expressing myoblasts in the embryonic undergo a transition from proliferative expansion to , as MyoD directly activates downstream targets like myogenin to enforce exit and initiate fusion into multinucleated myofibers. This switch is particularly pronounced in hypaxial progenitors around E10.5–E12.5, where MyoD overrides proliferative signals to coordinate the timely assembly of embryonic .

Adult Tissues and Regeneration

In adult , MyoD exhibits low basal expression in quiescent satellite cells, which reside beneath the and maintain a dormant state characterized by the absence of detectable MyoD protein despite expressing Pax7 and Myf5 transcripts. Upon muscle injury or regenerative stimuli, satellite cells activate and rapidly upregulate MyoD, marking their commitment to the myogenic lineage and entry into the . This upregulation is modulated by signaling pathways such as , which sustains quiescence by inhibiting MyoD in resting cells but declines post-injury to permit MyoD induction and proliferation. Similarly, (IGF) signaling contributes to MyoD activation by enhancing Myf5 expression through PI3K/Akt and ERK pathways, thereby promoting satellite cell proliferation and myogenic progression during repair. MyoD plays a critical role in adult myonuclear accretion, the process by which fusing myoblasts add new nuclei to existing myofibers to support and repair, ensuring the myonuclear domain remains balanced for cytoplasmic demands. In this context, MyoD drives the and of activated cells, facilitating the of myonuclei into mature during regenerative responses. Additionally, MyoD contributes to type maintenance in muscle, with higher protein levels observed in fast-twitch fibers compared to slow-twitch ones, influencing isoform expression and contractile properties. This differential accumulation suggests MyoD helps sustain fiber-specific identities, potentially through regulation of gene programs that resist type shifts under stress. Satellite cell populations in adult muscle display heterogeneity with respect to MyoD expression, comprising distinct MyoD-negative (MyoD-) and MyoD-positive (MyoD+) progenitors that differ in self-renewal and differentiation potential. Quiescent satellite cells are predominantly MyoD-, serving as a reserve pool with high self-renewal capacity, while a subset of activated cells becomes MyoD+ and progresses toward myoblast differentiation and fusion. Approximately 10% of quiescent cells lack both MyoD and Myf5, representing true stem-like progenitors that asymmetrically divide to generate MyoD+ committed daughters, thereby preserving the stem cell niche during homeostasis. This MyoD-based dichotomy ensures a balanced output of progenitors for both renewal and myogenic expansion. With advancing age, MyoD expression in cells declines, particularly in their response, leading to impaired myogenic and reduced regenerative efficiency. Aged satellite cells exhibit delayed or diminished MyoD upregulation upon injury, correlating with a smaller pool of functional MyoD+ progenitors and increased propensity for . This age-related attenuation contributes to , the progressive loss of muscle mass and strength, by limiting myonuclear addition and fiber repair, ultimately exacerbating frailty in older adults. Recent studies as of 2025 indicate that exercise induces MyoD mRNA expression in human 3–12 hours post-exercise, with variations by age and sex influencing regenerative responses.

Function in Myogenesis

Myoblast Determination

MyoD plays a pivotal role in committing multipotent progenitors, particularly Pax3/7-positive cells, to the myogenic lineage by activating the skeletal muscle-specific transcriptional program. In these progenitors, Pax3 and Pax7 bind to regulatory elements of the MyoD promoter, such as the paired box site at -1502, and cooperate with FoxO3 to recruit RNA polymerase II, forming a pre-initiation complex that drives MyoD transcription. This activation ensures myoblast-specific expression of MyoD, suppressing alternative non-muscle fates through the inhibition of non-myogenic gene programs and promotion of muscle lineage specification. Experimental depletion of Pax3/7 reduces FoxO3 binding and MyoD levels, confirming their essential upstream regulatory function in myogenic commitment. MyoD cooperates with Myf5 during early myogenic , where the two basic helix-loop-helix factors redundantly specify myogenic progenitors, with MyoD compensating for Myf5 loss to maintain . In Myf5-null mutants, MyoD activates key growth-phase genes like Six1 and Runx1, as well as differentiation markers such as myosin heavy chain, albeit less efficiently than in wild-type contexts, enabling partial rescue of . Genome-wide studies reveal that Myf5 and MyoD occupy the same sites, but Myf5 primarily induces chromatin modifications like histone H4 acetylation without robust transcriptional activation, whereas MyoD recruits to drive and complete . This functional divergence underscores MyoD's role in advancing progenitors beyond initial specification toward stable myogenic identity. A critical aspect of MyoD-mediated commitment involves inhibition of the , achieved through upregulation of the cyclin-dependent kinase inhibitor p21, which promotes exit from and enforces irreversible myoblast arrest. MyoD directly activates p21 expression independently of , leading to sustained p21 levels that block s and prevent in differentiating myoblasts. This mechanism is evident in MyoD-transfected 10T1/2 fibroblasts, where p21 induction correlates with cell cycle withdrawal, and is preserved in myogenic cells from MyoD/myogenin-deficient mice, highlighting p21's broad role in terminal differentiation. Lineage tracing experiments further establish MyoD as a marker of stable commitment to the lineage, demonstrating that MyoD-expressing cells in the epiblast resist redirection to non-myogenic fates. In embryos, MyoD-positive epiblast cells microinjected into cardiac or neural environments continue to express MyoD mRNA and protein but fail to differentiate into cardiomyocytes or neurons, unlike MyoD-negative counterparts that adapt to the host tissue.

Differentiation and Fusion

Following myoblast determination, MyoD drives terminal maturation by orchestrating the expression of genes essential for cytoskeletal reorganization and contractile apparatus . During this phase, MyoD sequentially activates structural genes such as desmin, which supports formation in maturing myoblasts, and isoforms, which enable calcium-sensitive regulation of contraction. MyoD also induces contractile proteins like myosin heavy chain and skeletal α-actin, ensuring the progressive buildup of sarcomeric structures as myoblasts exit the proliferative state. This temporal progression in is governed by promoter-specific binding affinities of MyoD, which distinguish early genes (e.g., those for initial cytoskeletal components) from late genes (e.g., those for mature contractile elements). MyoD exhibits higher affinity for enhancers of early genes during initial commitment, facilitating chromatin opening, while late gene promoters require cooperative interactions with factors like myogenin for sustained activation, ensuring orderly maturation. MyoD further promotes myoblast fusion into multinucleated myotubes by binding to enhancers of genes that mediate and membrane remodeling, including M-cadherin (a key cadherin in myoblast recognition) and myomaker (a fusogenic protein essential for membrane fusion). Junctional adhesion molecules (JAMs), such as JAM-B and JAM-C, facilitate heterotypic interactions during fusion, complementing adhesion programs to align and merge myoblasts. studies demonstrate that MyoD overexpression in non-myogenic cells, such as fibroblasts, robustly induces and fusion, yielding elongated, multinucleated myotubes capable of contractile activity.

Molecular Mechanisms

Transcriptional Regulation

MyoD functions as a basic helix-loop-helix (bHLH) transcription factor that binds to canonical E-box sequences with the consensus motif CANNTG in the promoters and enhancers of muscle-specific genes, thereby initiating their transcriptional activation. This sequence-specific binding is mediated by the basic domain of MyoD, which recognizes the DNA motif, while the affinity for the E-box can be modulated by the flanking nucleotide sequences surrounding the core CANNTG. To achieve high-affinity DNA binding, MyoD heterodimerizes with E-proteins such as E47 or E12, forming active bHLH complexes that preferentially recognize and bind E-boxes in regulatory regions of target genes. These heterodimers are essential for MyoD's transcriptional activity, as MyoD homodimers exhibit low DNA-binding efficiency and transcriptional potency. The transactivation domain (TAD) of MyoD recruits RNA polymerase II (Pol II) and associated general transcription factors to the promoters of target genes, facilitating the assembly of the pre-initiation complex and promoting transcriptional initiation. This recruitment is a key step in MyoD-mediated gene activation, enabling the transition from poised to active transcription states at muscle-specific loci. In addition to activation, MyoD can function as a transcriptional by binding to non-E-box motifs, silencing non-myogenic genes to erase prior identities and facilitate toward the myogenic fate. This dual activator- role enhances MyoD's efficiency in muscle regeneration and . Representative examples of MyoD's transcriptional targets include MEF2C, which MyoD activates through direct binding to an essential in the skeletal muscle enhancer of the Mef2c gene, establishing synergistic of myogenic . Similarly, MyoD drives the expression of myogenin by targeting its promoter via elements and recruiting Pol II, thereby amplifying the myogenic regulatory network.

Chromatin Dynamics

MyoD functions as a transcription factor, enabling access to closed regions during the early stages of by binding to specific motifs within compacted nucleosomes. This capability allows MyoD to initiate chromatin reconfiguration prior to widespread transcriptional , distinguishing it from later-acting factors like myogenin that preferentially target more accessible sites. Studies have demonstrated that MyoD's basic helix-loop-helix domain facilitates this pioneer activity, promoting the opening of repressive chromatin landscapes in multipotent precursors to establish myogenic identity. Once bound, MyoD recruits such as p300 and CBP to target loci, catalyzing of tails to promote an open chromatin conformation conducive to myogenic . This recruitment enhances lysine 27 (H3K27ac) at enhancers and promoters, facilitating the assembly of active transcriptional complexes during . For instance, while p300/CBP-mediated is dispensable for early myogenic genes like myogenin, it is essential for late markers such as MHC and MCK, with differential roles observed where CBP and p300 regulate distinct gene sets. The activity of these enzymes is critical for terminal and myotube fusion, as mutants lacking this function impair myogenic progression despite intact protein recruitment. MyoD also cooperates with ATP-dependent SWI/SNF chromatin remodeling complexes to reposition nucleosomes and expose regulatory elements during muscle differentiation. Through interactions with subunits like BAF60c and Brg1, MyoD incorporates these complexes into promoter and enhancer regions, enabling nucleosome eviction and increased DNA accessibility at myogenic loci. This remodeling is temporally regulated, occurring post-MyoD binding to support sustained gene activation, and is vital for overcoming chromatin barriers in differentiating myoblasts. Dominant-negative SWI/SNF components block MyoD-induced differentiation, underscoring the complex's necessity in facilitating the transition from proliferation to fusion. Feedback mechanisms involving MyoD further reinforce dynamics through auto-regulatory loops mediated by enhancer-promoter interactions. MyoD binding at its own promoter and upstream enhancers promotes its expression, creating a circuit essential for maintaining myogenic commitment.

Regulation of MyoD

Upstream Pathways

The upstream regulation of MyoD involves multiple signaling pathways that modulate its expression and activity during myogenic commitment and . The canonical Wnt/β-catenin pathway promotes MyoD expression in early myogenic progenitors within somites, driving lineage commitment and specification. However, in committed myoblasts such as cells, Wnt3a activation inhibits terminal by downregulating myogenin and other late myogenic targets, thereby favoring progression and over . This dual role helps balance progenitor expansion and progression during embryonic . Bone morphogenetic protein (BMP) and fibroblast growth factor (FGF) pathways similarly suppress MyoD in early myogenic stages to promote proliferation of undifferentiated cells. , primarily through and BMP-4, inhibits the transcriptional activity of MyoD without altering its nuclear localization or DNA-binding capability, leading to reduced expression of muscle-specific genes like muscle . In 10T1/2 fibroblasts stably expressing MyoD, treatment dose-dependently blocks myotube formation and heavy chain expression, highlighting its role in suppressing terminal during embryonic . Complementing this, delays MyoD expression in somitic progenitors and myoblasts by sustaining proliferative states; for example, in somites, reduced FGF8 activity elevates MyoD levels, accelerating myogenic commitment, while excess FGF maintains higher /7 expression and lower MyoD. These pathways thus coordinate to restrict MyoD induction until environmental cues, such as withdrawal, favor . In contrast, insulin-like growth factor-1 (IGF-1) and p38 mitogen-activated protein kinase (MAPK) provide activating inputs through post-translational modifications of MyoD. IGF-1 stimulates p38 MAPK phosphorylation in early , enhancing MyoD's ability to drive myoblast differentiation in normoxic conditions, as evidenced by p38 inhibition (e.g., via SB203580) blocking IGF-induced myotube formation in cells. Under , this pathway is dampened, shifting IGF-1 toward mitogenic effects, but normoxic activation of p38 by IGF-1 directly supports MyoD-dependent without relying on parallel PI3K/Akt signaling. p38 MAPK further potentiates MyoD activity indirectly by phosphorylating co-factors like MEF2, amplifying myogenic conversion in embryonic fibroblasts where p38α knockout reduces MyoD efficiency by approximately 45%. Notch signaling in adult satellite cells fine-tunes MyoD induction to balance self-renewal and . Active , driven by ligands like Delta-like 1 (DLL1), upregulates Pax7 while repressing MyoD expression through downstream effectors such as Hes/Hey repressors, which bind MyoD promoters to inhibit its transcription and maintain quiescence or . Upon muscle injury, declining Notch activity permits MyoD upregulation, enabling exit and myogenic progression; for example, constitutive Notch activation in cells elevates Pax7 and blocks MyoD, preserving the pool at the expense of . Oscillatory DLL1-Notch dynamics further ensure this equilibrium, preventing exhaustive during regeneration.

Coactivators and Repressors

MyoD's transcriptional activity is modulated by coactivators that enhance its ability to drive myogenic . The myocyte enhancer 2 (MEF2) members synergize with MyoD through direct physical interactions, enabling combinatorial control of muscle-specific promoters and amplifying in a cooperative manner. This synergy is particularly evident in the of genes required for , where MEF2 binding sites adjacent to MyoD-responsive E-boxes facilitate enhanced transactivation. Additionally, the p300 serves as a key coactivator by interacting with MyoD's amino-terminal domain, promoting acetylation of and MyoD itself to facilitate chromatin opening and transcriptional initiation. p300's role is critical for MyoD-dependent myoblast conversion, as its recruitment correlates with increased expression of muscle genes like myogenin. Recent single-cell studies have revealed heterogeneous MyoD expression patterns in adult muscle stem cells (MuSCs), highlighting additional regulatory layers such as copper-responsive factors like cysteine-rich intestinal protein 2 (CRIP2), which modulates MyoD in primary myoblasts under metal cues. Dual-specificity phosphatases 13 and 27 (DUSP13/27) also act as switches, dephosphorylating targets to transition MuSCs from to MyoD-driven . In contrast, repressors inhibit MyoD function by counteracting these activation mechanisms. Histone deacetylases (HDACs), particularly class II HDACs such as HDAC4 and HDAC5, associate with MyoD and MEF2 to deacetylate , thereby blocking access to myogenic promoters and preventing . This repression maintains myoblasts in a proliferative state until signals trigger HDAC dissociation. , a zinc-finger , represses MyoD during epithelial-mesenchymal transitions by competing for binding sites and recruiting corepressors, which suppresses myogenic conversion in contexts like tumor progression or development. SNAI2's interaction with MyoD enhances oncogenesis by overriding myogenic programs in cells. MyoD regulation exhibits context-dependent switching between activation and repression. For instance, the FOXP1-MTA1 complex represses in non-muscle cells by FOXP1 directly binding MyoD's bHLH domain to inhibit DNA binding, while MTA1 as part of the contributes to chromatin deacetylation for sustained repression during proliferative phases or in fibroblasts. This mechanism ensures MyoD activity is restricted outside committed myogenic lineages. MyoD levels are further controlled by ubiquitination-mediated , with the E3 TRIM32 playing a pivotal role in fine-tuning . TRIM32 targets inhibitors like c-Myc for ubiquitination and proteasomal , indirectly stabilizing MyoD function and promoting timely of muscle progenitors. Mutations in TRIM32 disrupt this balance, leading to impaired muscle regeneration as seen in limb-girdle type 2H.

Protein Interactions

With Myogenic Regulatory Factors

MyoD coordinates through interactions with other myogenic regulatory factors (MRFs), including Myf5, myogenin, and MRF4, primarily via their shared basic helix-loop-helix (bHLH) domains. These domains enable the formation of higher-order protein complexes that enhance DNA to motifs (CANNTG) in target gene promoters, facilitating cooperative regulation of muscle-specific . Although MRFs predominantly dimerize with E proteins (such as E12 or E47) for high-affinity , they can also engage in homo- or heterodimerization among themselves, allowing MyoD to modulate the activity of family members during distinct phases of myoblast determination and differentiation. A key aspect of these interactions is the and compensatory mechanisms between MyoD and Myf5 in initiating . MyoD and Myf5 exhibit overlapping functions in committing multipotent progenitors to the myogenic , as evidenced by the viable and normal development in single MyoD-null or Myf5-null mice, where the remaining factor upregulates to compensate. In contrast, Myf5/MyoD double-null mice with intact MRF4 expression undergo delayed , forming small myotomes due to late activation of downstream targets like myogenin, demonstrating MRF4's partial compensatory role in myoblast specification. This ensures robust of the myogenic despite the loss of individual MRFs. MyoD further interacts with myogenin to promote and , acting sequentially by directly activating the myogenin promoter. Early expression of MyoD in proliferating myoblasts recruits the to the myogenin promoter, switching core promoter recognition and enabling robust transcription of myogenin, which then drives terminal . MyoD-myogenin enhance this process by binding shared enhancers, ensuring coordinated withdrawal from the and expression of contractile proteins like heavy chain. These interactions highlight MyoD's pivotal role in transitioning from (with Myf5) to (with myogenin) within the MRF network.

With Signaling Effectors

MyoD engages with signaling effectors from the p38 (MAPK) pathway to modulate its transcriptional activity during . Specifically, p38 MAPK phosphorylates the E-protein E47 at serine 140, which promotes the formation of MyoD/E47 heterodimers and enhances their binding to DNA sequences, thereby activating muscle-specific . Additionally, p38 activation stabilizes MyoD mRNA by phosphorylating the RNA-binding protein KSRP, inhibiting its decay-promoting function and increasing MyoD protein levels to support differentiation. In certain contexts, such as satellite cell activation, the p38γ isoform directly phosphorylates MyoD at serines 199 and 200, facilitating interactions that fine-tune myogenic progression. MyoD also interfaces with extracellular matrix (ECM) components through integrin-mediated adhesion in satellite cells, which is critical for muscle regeneration. During injury-induced repair, MyoD regulates the expression of basement membrane proteins and integrins, such as α7β1, enabling satellite cells to adhere to the ECM and migrate to sites of damage. Integrin signaling, in turn, activates focal adhesion kinase (FAK) and downstream pathways like p38 MAPK, which amplify MyoD activity to promote cell spreading and myoblast alignment necessary for efficient tissue rebuilding. This bidirectional interplay ensures proper niche interactions, preventing aberrant proliferation and supporting ordered regeneration without excessive fibrosis. Synergy between MyoD and the (pRb) drives exit in myoblasts, facilitating terminal differentiation. pRb binds to MyoD and cooperates to repress E2F-dependent proliferation genes while activating myogenic targets like p21, enforcing G1 arrest and commitment to the myotube fate. This interaction is essential in post-mitotic contexts, where pRb hypophosphorylation stabilizes MyoD function, preventing re-entry into the and ensuring synchronized muscle fiber formation. In hypertrophy scenarios, and HDAC5 interact with MEF2 to repress myogenic genes such as myogenin, prioritizing fiber growth over fusion in response to mechanical stress. This repressive mechanism, involving deacetylation, balances maintenance and adaptation in mature muscle by dampening aspects of the MyoD-driven program.

Broader Physiological Roles

Muscle Maintenance and Metabolism

In adult , MyoD maintains low-level expression primarily in fast-twitch fibers, where it accumulates in the nuclei of the fastest fiber classes (IIB/IIX), contributing to the preservation of their contractile properties. This bias toward fast-twitch maintenance is evident from studies showing that MyoD disruption in shifts fast muscles toward a slower while slow muscles adopt faster characteristics, underscoring its role in fiber type . MyoD further supports muscle maintenance by regulating oxidative metabolism through direct transcriptional activation of genes involved in , such as PGC-1β, a key coactivator that coordinates energy production pathways including oxidation and the . In mature myofibers, this MyoD-mediated control, often in cooperation with alternative signaling, ensures adequate ATP availability for sustained contraction and prevents metabolic imbalances during routine activity. Adaptation to exercise involves transient upregulation of MyoD in cells, which activates their and fusion to existing fibers without inducing overt damage. Following resistance training, MyoD mRNA levels rise significantly within 24 hours post-exercise, amplifying as marked by increased Pax7+/MyoD+ cells per fiber, thereby facilitating hypertrophic responses and long-term muscle plasticity. MyoD also influences whole-body metabolism by enhancing insulin sensitivity and glucose uptake in myofibers via its promotion of myogenic . Enhancement of MyoD expression, as seen with certain hypoglycemic agents, activates the PI3K/AKT pathway to improve insulin responsiveness and lower blood glucose in insulin-resistant models. Additionally, MyoD directly regulates transcription of the glucose transporter through muscle-specific enhancers, optimizing postprandial glucose handling in .

Response to Injury

Upon muscle injury, satellite cells, the primary s responsible for repair, rapidly activate and upregulate MyoD expression to initiate the regenerative process. This upregulation occurs in response to damage signals, such as those from cardiotoxin-induced injury models, where MyoD coordinates the of activated satellite cells and their subsequent into myoblasts; asymmetric division allows a subset to self-renew and return to quiescence to replenish the stem cell pool. MyoD's role in this coordination ensures balanced expansion of progenitors without depleting the quiescent reserve, as evidenced by impaired and delayed regeneration in MyoD-deficient models. MyoD works in coordination with Pax7, a key regulator of satellite cell identity, to drive asymmetric cell divisions that maintain stem cell heterogeneity during regeneration. In these divisions, one daughter cell retains high Pax7 and low MyoD expression to self-renew as a quiescent , while the other upregulates MyoD alongside Pax7 to commit to proliferation and myogenic differentiation. This process, observed in injury models, relies on polarity cues like the Par complex, which asymmetrically activates signaling pathways (e.g., p38 MAPK) to induce MyoD in the committed progeny, thereby sustaining long-term regenerative capacity. MyoD contributes to scarless muscle healing by promoting efficient myogenic repair that suppresses excessive fibrotic deposition through regulation of genes favoring myoblast fusion over stromal accumulation. In experimental cardiotoxin-induced damage, MyoD- and Myf5-null cells fail to mount effective regeneration, resulting in persistent and increased fibrotic tissue formation, underscoring its essential role in anti-fibrotic outcomes. This regenerative efficiency contrasts with the steady-state metabolic maintenance in uninjured muscle, where MyoD levels remain low.

Clinical and Research Implications

Role in Muscular Disorders

In (DMD), the absence of results in significantly reduced MyoD expression in proliferating myoblasts, leading to dysregulation of over 170 genes involved in muscle development and function. This downregulation, observed in both mouse Dmd<sup>mdx</sup> models and human DMD-derived myoblasts, impairs cell regeneration by altering , , and dynamics, thereby exacerbating muscle degeneration through cell-autonomous defects. Consequently, the compromised regenerative capacity of cells contributes to progressive muscle wasting and characteristic of DMD. Therapeutic approaches targeting MyoD enhancement in s show promise for restoring muscle repair in DMD. For instance, strategies utilizing lentiviral vectors to overexpress MyoD in fibroblasts from mdx mice reprogram these s into induced myogenic progenitor s (iMPCs), which, upon transplantation, integrate into dystrophic muscle and promote regeneration of -expressing myofibers. Similarly, combining MyoD overexpression with / editing in DMD mouse-derived iMPCs corrects mutations, enabling engraftment into host muscle and partial restoration of the , thus improving overall muscle . These methods leverage MyoD's role in myogenic commitment to bypass intrinsic dysfunction. Alterations in MyoD expression with aging, including higher basal levels but a blunted response to exercise, contribute to , the progressive loss of skeletal muscle mass and strength, by diminishing the regenerative potential of cells. Exercise serves as a key modulator, inducing MyoD mRNA expression in human to support , although the response is attenuated in older adults compared to younger individuals, highlighting age-specific impairments in muscle adaptation. Mouse models demonstrate that enhancing MyoD activity ameliorates DMD phenotypes. In mdx mice lacking MyoD, dystrophic symptoms worsen dramatically, with increased , , and reduced regeneration due to excessive satellite cell self-renewal at the expense of ; conversely, maintaining or boosting MyoD function preserves regenerative efficiency and mitigates severity.

Implications in Cancer

MyoD, a key myogenic regulatory factor, exhibits tumor-suppressive properties in certain malignancies, particularly by inhibiting processes that promote cancer progression. In gastric cancer, MyoD1 expression is significantly reduced in tumor tissues compared to normal , and its overexpression suppresses and through direct transcriptional inhibition of fucosyltransferase 4 (FUT4). Specifically, MyoD1 binds to the FUT4 promoter region, reducing FUT4-mediated fucosylation of glycoproteins that facilitate metastatic behaviors, thereby highlighting MyoD's role in restraining epithelial-mesenchymal transition ()-like phenotypes in this cancer type. In (), a of origin, MyoD dysregulation contributes to aggressive tumor biology. Mutations such as the L122R substitution in the MYOD1 gene are recurrent in spindle cell and sclerosing subtypes, leading to impaired myogenic and enhanced due to altered DNA binding and capabilities. These mutants drive uncontrolled by failing to induce terminal , resulting in poor clinical outcomes and resistance to therapy across age groups. Additionally, epigenetic silencing or functional inhibition of wild-type MyoD in fusion-negative sustains proliferative states by disrupting heterodimer formation with E-proteins, which normally switches cells toward . MyoD displays a dual role in cancer, acting as a pro-myogenic driver of in normal cells while potentially functioning as an in pathological contexts involving lineage . In , MyoD acts as a tumor suppressor, as its loss accelerates tumor development in mouse models. Conversely, in , MyoD sustains tumor survival by upregulating DNA methyltransferases that silence pro-apoptotic genes like CYLD, thereby enabling oncogenic and resistance to . Recent investigations have elucidated MyoD's interactions with repressors like SNAI2, which promote and oncogenesis in . In fusion-negative , SNAI2-MyoD complexes enhance tumor growth and by blocking myogenic and facilitating , with SNAI2 overexpression correlating to aggressive disease phenotypes. This interaction underscores MyoD's context-dependent contributions to progression, where it shifts from differentiation inducer to facilitator.

References

  1. [1]
  2. [2]
  3. [3]
  4. [4]
  5. [5]
    Expression of a single transfected cDNA converts fibroblasts to ...
    Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell. 1987 Dec 24;51(6):987-1000. doi: 10.1016/0092 ...
  6. [6]
    MyoD1: a nuclear phosphoprotein requiring a Myc homology region ...
    Oct 21, 1988 · MyoD1 is a phosphoprotein present in the nuclei of proliferating myoblasts and differentiated myotubes but not expressed in 10T1/2 fibroblasts or other ...Missing: 1987 | Show results with:1987
  7. [7]
    MyoD is a sequence-specific DNA binding protein requiring a region ...
    MyoD is a skeletal muscle-specific protein that is able to induce myogenesis in a wide variety of cell types. In this report, we show that MyoD is a DNA ...
  8. [8]
    Finding MyoD and lessons learned along the way - PubMed
    MyoD, a transcription factor, was identified in 1987 by Robert Davis, Hal Weintraub and Andrew B Lassar, and can reprogram fibroblasts into skeletal muscle ...Missing: subtractive hybridization
  9. [9]
    Myogenin, a factor regulating myogenesis, has a domain ... - PubMed
    Myogenin, a factor regulating myogenesis, has a domain homologous to MyoD. Cell. 1989 Feb 24;56(4):607-17. doi: 10.1016/0092-8674(89)90583-7 ...Missing: discovery paper
  10. [10]
    Inactivation of MyoD in mice leads to up-regulation of the myogenic ...
    Oct 30, 1992 · Our results indicate that MyoD is dispensable for skeletal muscle development in mice, revealing some degree of functional redundancy.Missing: 1990s paper
  11. [11]
    MyoD or Myf-5 is required for the formation of skeletal muscle
    Dec 31, 1993 · These observations suggest that either Myf-5 or MyoD is required for the determination of skeletal myoblasts, their propagation, or both during embryonic ...Missing: 1990s paper
  12. [12]
    MyoD Targets Chromatin Remodeling Complexes to the Myogenin ...
    MyoD-mediated gene activation is associated with chromatin remodeling in the regulatory regions of muscle-specific genes and depends on a cysteine-histidine- ...
  13. [13]
    Global and gene-specific analyses show distinct roles for Myod and ...
    These findings demonstrate that Myod and Myog function at sequential steps in regulating a set of genes: Myod is necessary to induce chromatin modifications at ...
  14. [14]
    Pbx Marks Genes for Activation by MyoD Indicating a Role for a ...
    This demonstrates a specific mechanism of targeting MyoD to loci in inactive chromatin and reveals a critical role of homeodomain proteins in marking specific ...Missing: papers | Show results with:papers
  15. [15]
  16. [16]
    Gene ResultMYOD1 myogenic differentiation 1 [ (human)] - NCBI
    Aug 19, 2025 · This gene encodes a nuclear protein that belongs to the basic helix-loop-helix family of transcription factors and the myogenic factors subfamily.Missing: organization chromosome exons PAX3/ 7
  17. [17]
  18. [18]
    MyoD1 promoter autoregulation is mediated by two proximal E-boxes
    We show that in mouse myoblasts the MyoD1 promoter is highly stimulated by MyoD1 expression, suggesting that it is controlled by a positive feedback loop.
  19. [19]
    Genome-Wide Identification of PAX3-FKHR Binding Sites in ...
    Genetic hierarchy analysis of skeletal muscle differentiation reveals that MYOD expression requires both PAX3 and MYF5 (27). Analysis of the MYOD core enhancer ...
  20. [20]
    Master control: transcriptional regulation of mammalian Myod
    Jul 12, 2019 · MYOD can also regulate its own expression in vitro via binding E-boxes, binding sites for bHLH factors, in the promoter region of Myod (Fig.Skeletal Muscle Development... · Mrfs And Pax3/pax7 · Srf, Mef2 And Cofactors
  21. [21]
    Myod1 myogenic differentiation 1 [ (house mouse)] - NCBI
    Sep 24, 2025 · In conclusion, these findings reveal mechanistic roles for the muscle specific transcription factor MYOD1 in the regulation of molecular ...Missing: vertebrates origin chordates
  22. [22]
    The single MyoD family gene of Ciona intestinalis encodes two ...
    The single MyoD family gene of Ciona intestinalis encodes two differentially expressed proteins: implications for the evolution of chordate muscle gene ...
  23. [23]
    Than One-to-Four via 2R: Evidence of an Independent Amphioxus ...
    Jun 10, 2020 · This level of conservation allows for comparative studies of MyoD orthologs across the bilaterians. ... chordate and vertebrate origins.
  24. [24]
    Differential regulation of multiple alternatively spliced transcripts of ...
    Transcript levels of TmyoD1-gamma only varied significantly in fast muscle and were 5-fold higher in adult compared to juvenile stages. Significant differences ...
  25. [25]
    MYOD1 - Myoblast determination protein 1 - Homo sapiens (Human)
    Oct 14, 2008 · ... gene promoter core region during myogenesis. Induces fibroblasts to ... Chromosome 11. Organism-specific databases. AGR · HGNC:7611 · HGNC.
  26. [26]
  27. [27]
    MyoD phosphorylation on multiple C terminal sites regulates ... - NIH
    Nov 3, 2016 · MyoD is a master regulator of myogenesis with a potent ability to redirect the cell fate of even terminally differentiated cells.
  28. [28]
    Expression of two myogenic regulatory factors myogenin and MyoDl ...
    Sep 28, 1989 · MyoDl and myogenin are muscle-specific proteins which can convert non-myogenic cells in culture to differentiated muscle fibres, implicating them in myogenic ...Missing: discovery | Show results with:discovery
  29. [29]
    Differential activation of Myf5 and MyoD by different Wnts in explants ...
    In the mouse embryo, Myf5 is the first to be activated in the epaxial domain of newly formed somites at E8 following a rostrocaudal gradient. MyoD expression, ...
  30. [30]
    Early development of the myotome in the mouse - Venters - 1999
    Nov 15, 1999 · Myf-5 is the first MRF to be expressed in the mouse, with mRNA detectable at E8. 0 in the dorsomedial aspect of the epithelial somite (Ott et ...
  31. [31]
    MyoD expression marks the onset of skeletal myogenesis in Myf-5 ...
    Our initial studies suggested that Myf-5 is required for the development of early somitic myocytes (Braun et al., 1992). To explore in more detail the Myf-5 ...
  32. [32]
    Short Article Evidence for a Myotomal Hox/Myf Cascade Governing ...
    Apr 20, 2010 · Control of Rib Formation and Myf5/Myf6 Expression by Hox ... Six proteins regulate the activation of Myf5 expression in embryonic mouse limbs.
  33. [33]
    The Myogenic Regulatory Factors, Determinants of Muscle ...
    All these together, show that MyoD and Myf5 function as determination factors, whereas myogenin and MRF4 act to permit terminal differentiation (Figure 2).
  34. [34]
    Tissue-specific stem cells: Lessons from the skeletal muscle satellite ...
    In their quiescent state, adult satellite cells express, along with Pax7, the Myf5 transcript but lack Myod. During activation and lineage progression, ...
  35. [35]
    Stem cell activation in skeletal muscle regeneration - PMC - NIH
    Notch signaling level decreases almost immediately after muscle injury when satellite cells are activated [122], and increases again 4–5 days post-injury [127].
  36. [36]
    Stem cell activation in skeletal muscle regeneration
    Jan 9, 2015 · IGF-IR heterozygous mice display decreased the levels of MyoD expression and satellite cell activation [99], further confirming the importance ...
  37. [37]
    Myonuclear Domain Flexibility Challenges Rigid Assumptions on ...
    Satellite cell-mediated myonuclear accretion is thought to be required for skeletal muscle fiber hypertrophy, and even drive hypertrophy by preceding growth.
  38. [38]
    The multifaceted role of MyoD in adult skeletal muscle - PubMed - NIH
    Jul 1, 2025 · Single MyoD knockout analysis revealed that MyoD uniquely regulates adult muscle regeneration, considerably influencing delayed myogenic ...
  39. [39]
    MyoD protein is differentially accumulated in fast and slow skeletal ...
    MyoD mRNA levels differ between fast and slow muscles, suggesting that MyoD may regulate aspects of fibre type. Here we show that detectable MyoD protein ...
  40. [40]
    Muscle satellite cell heterogeneity and self-renewal - Frontiers
    Most Pax7(+)MyoD(+) activated satellite cells or myoblasts undergo Pax7(−)MyoD(+)Myogenin(+) myocyte differentiation, whereas a subset of Pax7(+)MyoD(+) ...
  41. [41]
    MyoD-expressing progenitors are essential for skeletal myogenesis ...
    Abstract. Skeletal myogenesis in the embryo is regulated by the coordinated expression of the MyoD family of muscle regulatory factors (MRFs).
  42. [42]
    Regulation of Satellite Cell Function in Sarcopenia - Frontiers
    This review discusses data pertaining to the satellite cell responses and function in aging skeletal muscle, and the impact that three compounds
  43. [43]
    Contribution of muscle satellite cells to sarcopenia - PMC
    MyoD can also induce expression of muscle-specific miRNAs (miR-206 and miR-133), downregulate Pax7, and activate the main regulator of differentiated myogenin ...
  44. [44]
    Co-dependent Activators Direct Myoblast Specific MyoD Transcription
    Recombinant Pax3 and Pax7 were found to bind the paired box probe, but not a mutant probe, although Pax3 binds with significantly higher affinity than Pax7 at ...Missing: MYOD1 | Show results with:MYOD1
  45. [45]
  46. [46]
    MyoD induces myogenic differentiation through cooperation of its NH2
    MyoD and Myf5 are basic helix-loop-helix transcription factors that play key but redundant roles in specifying myogenic progenitors during embryogenesis.
  47. [47]
  48. [48]
  49. [49]
    p53-Independent Expression of p21 Cip1 in Muscle and Other ...
    Although the muscle-specific transcription factor MyoD is sufficient to activate p21 expression in 10T1/2 cells, p21 was expressed in myogenic cells of mice ...
  50. [50]
    Cells that express MyoD mRNA in the epiblast are stably committed ...
    Aug 13, 2007 · We investigated whether MyoD-positive (MyoDpos) epiblast cells are stably committed to the skeletal muscle lineage or whether their fate can be ...
  51. [51]
    Inhibition of desmin expression blocks myoblast fusion ... - PubMed
    Inhibition of desmin expression blocks myoblast fusion and interferes with the myogenic regulators MyoD and myogenin.
  52. [52]
    MyoD Regulates Skeletal Muscle Oxidative Metabolism ... - NIH
    Oct 4, 2016 · Studies performed on the myosin heavy chain, skeletal α-actin, and troponin promoters demonstrated that MyoD can mediate transcriptional ...
  53. [53]
    Promoter-Specific Regulation of MyoD Binding and Signal ...
    MyoD regulates genes expressed at different times during myogenesis, and promoter-specific regulation of MyoD binding is a major mechanism of patterning gene ...
  54. [54]
    The cooperation of cis-elements during M-cadherin promoter ...
    Feb 26, 2021 · Stronger activation of M-cadherin promoter than that of muscle creatine kinase (MCK) by MyoD was observed regardless of culture conditions and ...
  55. [55]
    Myomaker, Regulated by MYOD, MYOG and miR-140-3p ... - PubMed
    Nov 2, 2015 · These findings confirm the essential roles of Myomaker in avian myoblast fusion and show that MYOD, MYOG and miR-140-3p can regulate Myomaker ...
  56. [56]
    Jamb and Jamc Are Essential for Vertebrate Myocyte Fusion
    Dec 13, 2011 · One pair, Jamb and Jamc, was selected because both genes are expressed by dividing myoblasts during primary myogenesis, but in distinct patterns ...
  57. [57]
    Regulation of skeletal muscle differentiation in fibroblasts ... - PubMed
    Myogenesis of fibroblasts incubated in the medium was activated by overexpression of MyoD, and the cells were accumulated and fused into multinucleated myotubes ...
  58. [58]
    Identification of MyoD-Responsive Transcripts Reveals a Novel ...
    Jun 6, 2017 · (A) Overexpression of MyoD caused the increased mRNA level of MyoG and MyHC gene. ... multinucleated myotubes. Proc Natl Acad Sci USA 87, 7988–92 ...<|control11|><|separator|>
  59. [59]
    MyoD is a sequence-specific DNA binding protein requiring a region ...
    Weintraub, A.B. Lassar. MyoD1: a nuclear phosphoprotein requiring a myc homology region to convert fibroblasts to myoblasts. Science, 242 (1988), pp. 405-411.
  60. [60]
    MyoD uses overlapping but distinct elements to bind E-box ... - NIH
    In this work, we identified elements in MyoD that bind E-box or tetraplex structures of promoter sequences of the muscle-specific genes α7 integrin and ...
  61. [61]
    Muscle-specific transcriptional activation by MyoD.
    The normal MyoD-activation domain maps within the amino-terminal 53 residues and can be functionally replaced by the activation domain of VP16. The activity ...
  62. [62]
    E47 phosphorylation by p38 MAPK promotes MyoD ... - EMBO Press
    Feb 17, 2005 · The bHLH domain of MRFs and E proteins is considered the minimum structure required for myogenic activity, with the basic region necessary for ...
  63. [63]
    TBP/TFIID-dependent activation of MyoD target genes in skeletal ...
    Feb 11, 2016 · The process of RNA polymerase II (RNAPII) recruitment to protein coding genes is part of the assembly of the Pre-Initiation Complex (PIC) ...
  64. [64]
    mef2c is activated directly by myogenic basic helix-loop-helix ...
    We show that mef2c is a direct transcriptional target of the MyoD family in vivo via an essential E box in the skeletal muscle enhancer of mef2c.
  65. [65]
    MyoD targets TAF3 /TRF3 to activate Myogenin transcription - NIH
    Skeletal muscle differentiation requires a cascade of transcriptional events to control the spatial and temporal expression of muscle-specific genes.Missing: TAD | Show results with:TAD
  66. [66]
    The circuitry of a master switch: Myod and the regulation of skeletal ...
    Jun 15, 2005 · Recent findings indicate that Myod functions in an instructive chromatin context and directly regulates genes that are expressed throughout the myogenic ...
  67. [67]
    CBP/p300 and muscle differentiation: no HAT, no ... - EMBO Press
    Our results clearly demonstrate that CBP/p300 HAT activity is critical for myogenic terminal differentiation. Furthermore, this requirement is restricted to a ...
  68. [68]
    Differential role of p300 and CBP acetyltransferase during myogenesis
    Studies in tissue culture cells have implicated p300 and CBP acetyltransferases in myogenic regulatory factor (MRF) mediated transcription and terminal ...
  69. [69]
    CBP and P300 regulate distinct gene networks required for human ...
    Aug 22, 2018 · We found that CBP and p300 play a critical role in the activation of the myogenic program and mostly regulate distinct gene sets to control several aspects of ...
  70. [70]
    Signal‐dependent incorporation of MyoD–BAF60c into Brg1‐based ...
    Nov 8, 2011 · We show that the muscle determination factor MyoD and the SWI/SNF subunit BAF60c interact on the regulatory elements of MyoD‐target genes in myoblasts.
  71. [71]
    The BAF60c-MyoD complex poises chromatin for rapid transcription
    Chromatin remodeling by the SWI/SNF complex is required to activate the transcription of myogenic-specific genes. Our work addressed the details of how SWI/SNF ...
  72. [72]
    MyoD is a 3D genome structure organizer for muscle cell identity
    Jan 11, 2022 · We show that MyoD functions as a “genome organizer” that specifies 3D genome architecture unique to muscle cell development.
  73. [73]
    WNT/β-catenin signaling plays a crucial role in myoblast fusion ...
    We show that WNT/β-catenin signaling is crucial for myoblast fusion through regulation of the nephrin (Nphs1) gene in the Myog-Cre-expressing myoblast ...
  74. [74]
    Canonical Wnt signaling induces BMP-4 to specify slow ...
    Mar 5, 2013 · It could suggest that canonical Wnt/β-catenin inhibits the proliferation and differentiation of myogenic cell lineages. This possibility ...<|separator|>
  75. [75]
    Bone Morphogenetic protein-2 Inhibits Terminal Differentiation of ...
    Feb 1, 1997 · Furthermore, BMP-2 inhibited the terminal differentiation of these subclonal 10T1/2 cells that stably expressed MyoD or myogenin into mature ...
  76. [76]
    FGF and Myogenesis - FGF Signalling in Vertebrate Development
    FGF initially represses myogenic differentiation, but in vivo, FGF activity promotes myogenic differentiation, regulating the balance between proliferation and ...
  77. [77]
    Hypoxia converts the myogenic action of insulin-like growth factors ...
    In this study, we observed that IGF activates p38 MAPK in the early stages of myogenesis. Pharmacological blockade of p38 MAPK inhibited the IGF-induced ...Sign Up For Pnas Alerts · Results · Hypoxia Alters Cellular...
  78. [78]
    p38 and Extracellular Signal-Regulated Kinases Regulate the ... - NIH
    Much evidence indicates that p38 is an activator of MyoD: (i) p38 kinase activity is required for the expression of MyoD-responsive genes, (ii) enforced ...Activation Of Mef2a And... · Dual Role Of Erk Activity... · P38 Mapks Positively...
  79. [79]
    The Notch signaling network in muscle stem cells during ...
    Apr 22, 2022 · Notch signalling acts in postmitotic avian myogenic cells to control MyoD ... Notch signaling rescues loss of satellite cells lacking Pax7 and ...
  80. [80]
    Oscillations of Delta-like1 regulate the balance between ... - Nature
    Feb 26, 2021 · We conclude that the oscillatory Dll1 input into Notch signaling ensures the equilibrium between self-renewal and differentiation in myogenic ...
  81. [81]
    TRANSCRIPTIONAL CONTROL OF MUSCLE DEVELOPMENT BY ...
    Synergistic activation of myogenesis by MyoD and MEF2 can also be observed with MEF2 mutants that fail to bind DNA, indicating that this type of ...<|control11|><|separator|>
  82. [82]
    Molecular mechanisms of myogenic coactivation by p300 - PubMed
    Coactivation by p300 involves novel physical interactions between p300 and the amino-terminal activation domain of MyoD. In particular, disruption of the FYD ...
  83. [83]
    Control of muscle development by dueling HATs and HDACs
    The MyoD and MEF2 families of transcription factors act cooperatively to regulate the expression of skeletal muscl …
  84. [84]
    Interaction between SNAI2 and MYOD enhances oncogenesis and ...
    Jan 8, 2021 · SNAI2 is known to repress the transcription of target genes by specifically binding the consensus motif CANNTG (i.e., E-box), and there are only ...
  85. [85]
    FOXP1 Interacts with MyoD to Repress its Transcription and ...
    We provide a model in which FOXP1 displaces ID and E12/E47 to repress MyoD during the proliferative phase of myoblast differentiation.
  86. [86]
    TRIM32 Regulates Skeletal Muscle Stem Cell Differentiation and Is ...
    Moreover we provide evidence that TRIM32 induces the differentiation of skeletal muscle progenitor cells through the degradation of c-Myc via the ubiquitination ...
  87. [87]
    Tripartite Motif-Containing Protein 32 (TRIM32): What Does It Do for ...
    Aug 19, 2023 · TRIM32 promotes the ubiquitination and degradation of skeletal ... c-Myc degradation released c-Myc inhibition on MyoD and myogenin ...
  88. [88]
    Functional redundancy of the muscle-specific transcription factors ...
    Feb 29, 1996 · The myogenic basic helix–loop–helix transcription factors, Myf5, MyoD, myogenin and MRF4, play key roles in skeletal muscle development.Missing: discovery | Show results with:discovery
  89. [89]
    p38-γ–dependent gene silencing restricts entry into the myogenic ...
    The regenerative capacity of muscle is regulated by p38-γ, which phosphorylates MyoD and leads to formation of a complex that represses myogenin transcript.
  90. [90]
    The absence of MyoD in regenerating skeletal muscle affects the ...
    The absence of MyoD in regenerating skeletal muscle affects the expression pattern of basement membrane, interstitial matrix and integrin molecules that is ...
  91. [91]
    Promyogenic function of Integrin/FAK signaling is mediated by Cdo ...
    The Integrin/FAK signaling pathway is required for myoblast differentiation by regulating the expression of the promyogenic factors, Cdo, MyoD and Cdc42.
  92. [92]
    Retinoblastoma Protein and MyoD Function Together to Effect the ...
    The findings reveal a link between MyoD, pRb, Fra-1, and cyclin D1, the latter three of which are often deregulated in cancer.Missing: synergy | Show results with:synergy
  93. [93]
    Regulation of MyoD function in the dividing myoblast - ScienceDirect
    Feb 16, 2001 · Their findings indicate that Rb promotes myogenesis by inhibiting cell cycle progression and cooperating with MyoD to activate the ...
  94. [94]
    PC4 Coactivates MyoD by Relieving the Histone Deacetylase 4 ...
    Histone deacetylase 4 (HDAC4) negatively regulates skeletal myogenesis by associating with the myocyte enhancer factor 2 (MEF2) transcription factors.Missing: hypertrophy | Show results with:hypertrophy
  95. [95]
    MyoD protein is differentially accumulated in fast and slow skeletal ...
    MyoD is a muscle-specific transcription factor involved in commitment of cells to myogenesis. MyoD mRNA levels differ between fast and slow muscles, ...
  96. [96]
    MyoD Regulates Skeletal Muscle Oxidative Metabolism ... - PubMed
    Oct 4, 2016 · MyoD is a key regulator of skeletal myogenesis that directs contractile protein synthesis, but whether this transcription factor also regulates skeletal muscle ...
  97. [97]
    Altered muscle satellite cell activation following 16 wk of resistance ...
    Jan 1, 2017 · Following 16 wk of RT, MyoD mRNA expression increased from basal to 24 h after the single bout of exercise (P < 0.05); this change was not ...
  98. [98]
    A new hypoglycemic mechanism of catalpol revealed by enhancing ...
    Sep 15, 2018 · Improvement of insulin sensitivity by enhancing MyoD/MyoG-mediated myogenesis may constitute a new therapeutic approach for treating type 2 ...
  99. [99]
    Mechanisms regulating GLUT4 glucose transporter expression and ...
    GLUT4 gene transcription is regulated by a number of factors in skeletal muscle that include MEF2, MyoD myogenic proteins, thyroid hormone receptors.
  100. [100]
    MyoD is required for myogenic stem cell function in adult skeletal ...
    Newborn mice deficient for both MyoD and. Myf-5 are totally devoid of skeletal myoblasts and mus- cle (Rudnicki et al. 1993). Mice lacking myogenin die at birth ...
  101. [101]
    MyoD is required for myogenic stem cell function in adult skeletal ...
    We propose that the failure of MyoD-deficient muscle to regenerate efficiently is not caused by a reduction in numbers of satellite cells, the stem cells of ...
  102. [102]
    Post-transcriptional regulation of satellite cell quiescence by TTP ...
    Mar 27, 2015 · Upon muscle injury, SCs activate and then enter S-phase to generate a proliferating myoblast population that differentiates, repairs skeletal ...P38α/β Mapk Inhibition... · Ttp Binds The Myod 3′ Utr... · The Myod 3′ Utr Ttp...
  103. [103]
    Loss of full-length dystrophin expression results in major cell ... - eLife
    Sep 27, 2022 · In Dmdmdx myoblasts lacking full-length dystrophin, the expression of 170 genes was significantly altered. Myod1 and key genes controlled by ...
  104. [104]
  105. [105]
    Exercise-induced MyoD mRNA Expression in Young and Older ...
    May 3, 2025 · ... Exercise-induced MyoD mRNA Expression in Young and Older Human Skeletal Muscle: A Systematic Review and Meta-Analysis. Sports Med 55 ...
  106. [106]
    MyoD is required for myogenic stem cell function in adult skeletal ...
    Therefore, we propose that the failure of MyoD-deficient muscle to regenerate efficiently is not caused by a reduction in numbers of satellite cells, the stem ...
  107. [107]
    MyoD1 suppresses cell migration and invasion by inhibiting FUT4 ...
    Dec 12, 2019 · MyoD1 suppresses cell migration and invasion by inhibiting FUT4 transcription in human gastric cancer cells. Cancer Gene Ther 27, 773–784 (2020) ...
  108. [108]
    MYOD1 (L122R) mutations are associated with spindle cell ... - Nature
    Aug 26, 2016 · Recently, two independent studies demonstrated presence of MYODI (L122R) mutations as the basis to re-classify a spindle cell rhabdomyosarcoma, ...Missing: silencing | Show results with:silencing
  109. [109]
    MYOD1-Mutant Spindle Cell and Sclerosing Rhabdomyosarcoma
    Sep 4, 2018 · MYOD1 mutation defines an aggressive rhabdomyosarcoma subset, with poor outcome and response to therapy, irrespective of age.Missing: silencing | Show results with:silencing
  110. [110]
    MyoD and E-protein heterodimers switch rhabdomyosarcoma cells ...
    A forced heterodimer between MyoD and the full-length E12 robustly restores differentiation in rhabdomyosarcoma cells and broadly suppresses multiple ...
  111. [111]
    MyoD is a tumor suppressor gene in Medulloblastoma - PMC - NIH
    Findings in a variety of cancers implicate MyoD as a possible tumor suppressor. ... These results directly support our finding of a tumor suppressor role of MyoD ...
  112. [112]
    MyoD is essential in rhabdomyosarcoma by promoting survival ...
    Aug 15, 2025 · Results showed that MyoDWT rescued death in RH30-SR MyoDΔ cells (Figure 2H), but a similar rescue was not achieved with the MyoD mutant, MYOD1 ...