fbpx
Wikipedia

MyoD

MyoD, also known as myoblast determination protein 1,[5] is a protein in animals that plays a major role in regulating muscle differentiation. MyoD, which was discovered in the laboratory of Harold M. Weintraub,[6] belongs to a family of proteins known as myogenic regulatory factors (MRFs).[7] These bHLH (basic helix loop helix) transcription factors act sequentially in myogenic differentiation. Vertebrate MRF family members include MyoD1, Myf5, myogenin, and MRF4 (Myf6). In non-vertebrate animals, a single MyoD protein is typically found.

MYOD1
Available structures
PDBOrtholog search: PDBe RCSB
Identifiers
AliasesMYOD1, MYF3, MYOD, PUM, bHLHc1, myogenic differentiation 1, MYODRIF
External IDsOMIM: 159970 MGI: 97275 HomoloGene: 7857 GeneCards: MYOD1
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_002478

NM_010866

RefSeq (protein)

NP_002469

NP_034996

Location (UCSC)Chr 11: 17.72 – 17.72 MbChr 7: 46.03 – 46.03 Mb
PubMed search[3][4]
Wikidata
View/Edit HumanView/Edit Mouse

MyoD is one of the earliest markers of myogenic commitment. MyoD is expressed at extremely low and essentially undetectable levels in quiescent satellite cells, but expression of MyoD is activated in response to exercise or muscle tissue damage. The effect of MyoD on satellite cells is dose-dependent; high MyoD expression represses cell renewal, promotes terminal differentiation and can induce apoptosis. Although MyoD marks myoblast commitment, muscle development is not dramatically ablated in mouse mutants lacking the MyoD gene. This is likely due to functional redundancy from Myf5 and/or Mrf4. Nevertheless, the combination of MyoD and Myf5 is vital to the success of myogenesis.[8][9]

History edit

MyoD was cloned by a functional assay for muscle formation reported in Cell in 1987 by Davis, Weintraub, and Lassar. It was first described as a nuclear phosphoprotein in 1988 by Tapscott, Davis, Thayer, Cheng, Weintraub, and Lassar in Science. The researchers expressed the complementary DNA (cDNA) of the murine MyoD protein in a different cell lines (fibroblast and adipoblast) and found MyoD converted them to myogenic cells.[6][10] The following year, the same research team performed several tests to determine both the structure and function of the protein, confirming their initial proposal that the active site of the protein consisted of the helix loop helix (now referred to as basic helix loop helix) for dimerization and a basic site upstream of this bHLH region facilitated DNA binding only once it became a protein dimer.[11] MyoD has since been an active area of research as still relatively little is known concerning many aspects of its function.

Function edit

The function of MyoD in development is to commit mesoderm cells to a skeletal myoblast lineage, and then to regulate that continued state. MyoD may also regulate muscle repair. MyoD mRNA levels are also reported to be elevated in aging skeletal muscle.

One of the main actions of MyoD is to remove cells from the cell cycle (halt proliferation for terminal cell cycle arrest in differentiated myocytes) by enhancing the transcription of p21 and myogenin. MyoD is inhibited by cyclin dependent kinases (CDKs). CDKs are in turn inhibited by p21. Thus MyoD enhances its own activity in the cell in a feedforward manner.

Sustained MyoD expression is necessary for retaining the expression of muscle-related genes.[12]

MyoD is also an important effector for the fast-twitch muscle fiber (types IIA, IIX, and IIB) phenotype.[13][14]

Mechanisms edit

MyoD is a transcription factor and can also direct chromatin remodelling through binding to a DNA motif known as the E-box. MyoD is known to have binding interactions with hundreds of muscular gene promoters and to permit myoblast proliferation. While not completely understood, MyoD is now thought to function as a major myogenesis controller in an on/off switch association mediated by KAP1 (KRAB [Krüppel-like associated box]-associated protein 1) phosphorylation.[15] KAP1 is localized at muscle-related genes in myoblasts along with both MyoD and Mef2 (a myocyte transcription enhancer factor). Here, it serves as a scaffold and recruits the coactivators p300 and LSD1, in addition to several corepressors which include G9a and the Histone deacetylase HDAC1. The consequence of this coactivator/corepressor recruitment is silenced promoting regions on muscle genes. When the kinase MSK1 phosphorylates KAP1, the corepressors previously bound to the scaffold are released allowing MyoD and Mef2 to activate transcription.[16]

Once the "master controller" MyoD has become active, SETDB1 is required to maintain MyoD expression within the cell. Setdb1 appears to be necessary to maintain both MyoD expression and also genes that are specific to muscle tissues because reduction of Setdb1 expression results in a severe delay of myoblast differentiation and determination.[17] In Setdb1 depleted myoblasts that are treated with exogenous MyoD, myoblastic differentiation is successfully restored. In one model of Setdb1 action on MyoD, Setdb1 represses an inhibitor of MyoD. This unidentified inhibitor likely acts competitively against MyoD during typical cellular proliferation. Evidence for this model is that reduction of Setdb1 results in direct inhibition of myoblast differentiation which may be caused by the release of the unknown MyoD inhibitor.

 
Evidence suggests that Setdb1 inhibits a repressor of MyoD and this is the mechanism through which MyoD expression is retained in differentiated myoblasts.

MyoD has also been shown to function cooperatively with the tumor suppressor gene, Retinoblastoma (pRb) to cause cell cycle arrest in the terminally differentiated myoblasts.[18] This is done through regulation of the Cyclin, Cyclin D1. Cell cycle arrest (in which myoblasts would indicate the conclusion of myogenesis) is dependent on the continuous and stable repression of the D1 cyclin. Both MyoD and pRb are necessary for the repression of cyclin D1, but rather than acting directly on cyclin D1, they act on Fra-1 which is immediately early of cyclin D1. MyoD and pRb are both necessary for repressing Fra-1 (and thus cyclin D1) as either MyoD or pRb on its own is not sufficient alone to induce cyclin D1 repression and thus cell cycle arrest. In an intronic enhancer of Fra-1 there were two conserved MyoD binding sites discovered. There is cooperative action of MyoD and pRb at the Fra-1 intronic enhancer that suppresses the enhancer, therefore suppressing cyclin D1 and ultimately resulting in cell cycle arrest for terminally differentiated myoblasts.[19]

Wnt signalling can affect MyoD edit

Wnt signalling from adjacent tissues has been shown to induce cells in somites that receive these Wnt signals to express Pax3 and Pax7 in addition to myogenic regulatory factors, including Myf5 and MyoD. Specifically, Wnt3a can directly induce MyoD expression via cis-element interactions with a distal enhancer and Wnt response element.[20] Wnt1 from dorsal neural tube and Wnt6/Wnt7a from surface ectoderm have also been implicated in promoting myogenesis in the somite; the latter signals may act primarily through Myod.

In typical adult muscles in a resting condition (absence of physiological stress) the specific Wnt family proteins that are expressed are Wnt5a, Wnt5b, Wnt7a and Wnt4. When a muscle becomes injured (thus requiring regeneration) Wnt5a, Wnt5b, and Wnt7a are increased in expression. As the muscle completes repair Wnt7b and Wnt3a are increased as well. This patterning of Wnt signalling expression in muscle cell repair induces the differentiation of the progenitor cells, which reduces the number of available satellite cells. Wnt plays a crucial role in satellite cell regulation and skeletal muscle aging and also regeneration. Wnts are known to active the expression of Myf5 and MyoD by Wnt1 and Wnt7a. Wnt4, Wnt5, and Wnt6 function to increase the expression of both of the regulatory factors but at a more subtle level. Additionally, MyoD increases Wnt3a when myoblasts undergo differentiation. Whether MyoD is activated by Wnt via cis-regulation direct targeting or through indirect physiological pathways remains to be elucidated.[21]

Coactivators and repressors edit

IFRD1 is a positive cofactor of MyoD, as it cooperates with MyoD at inducing the transcriptional activity of MEF2C (by displacing HDAC4 from MEF2C); moreover IFRD1 also represses the transcriptional activity of NF-κB, which is known to inhibit MyoD mRNA accumulation.[22][23]

NFATc1 is a transcription factor that regulates composition of fiber type and the fast-to-slow twitch transition resulting from aerobic exercise requires the expression of NFATc1. MyoD expression is a key transcription factor in fast twitch fibers which is inhibited by NFATc1 in oxidative fiber types. NFATc1 works to inhibit MyoD via a physical interaction with the MyoD N-terminal activation domain resulting in inhibited recruitment of the necessary transcriptional coactivator p300. NFATc1 physically disrupts the interaction between MyoD and p300. This establishes the molecular mechanism by which fiber types transition in vivo through exercise with opposing roles for NFATc1 and MyoD. NFATc1 controls this balance by physical inhibition of MyoD in slow-twitch muscle fiber types.[24]

 
MyoD works with a transient placeholder protein that functions to prevent other transcription factors from binding to the DNA and also retains an inactive conformation for the DNA. Once the placeholder is removed (or possibly deactivated) the necessary transcription factors are free to bind and initiate recruitment of RNA Polymerase II and initiate active RNA transcription.

The histone deacetyltransferase p300 functions with MyoD in an interaction that is essential for the myotube generation from fibroblasts that is mediated by MyoD. Recruitment of p300 is the rate-limiting process in the conversion of fibroblasts to myotubes.[25] In addition to p300, MyoD is also known to recruit Set7, H3K4me1, H3K27ac, and RNAP II to the enhancer that is bound with and this allows for the activation of muscle gene that is condition-specific and established by MyoD recruitment. Endogenous p300 though, is necessary for MyoD functioning by acting as an essential coactivator. MyoD associatively binds to the enhancer region in conjunction with a placeholding "putative pioneer factor" which helps to establish and maintain a both of them in a specific and inactive conformation. Upon the removal or inactivation on the placeholder protein bound to the enhancer, the recruitment of the additional group of transcription factors that help to positively regulate enhancer activity is permitted and this results in the MyoD-transcription factor-enhancer complex to assume a transcriptionally active state.

Interactions edit

MyoD has been shown to interact with:

References edit

  1. ^ a b c GRCh38: Ensembl release 89: ENSG00000129152 - Ensembl, May 2017
  2. ^ a b c GRCm38: Ensembl release 89: ENSMUSG00000009471 - Ensembl, May 2017
  3. ^ "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. ^ "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. ^ "P15172 (MYOD1_HUMAN)". UniProtKB. Retrieved 17 July 2019.
  6. ^ a b Davis RL, Weintraub H, Lassar AB (Dec 1987). "Expression of a single transfected cDNA converts fibroblasts to myoblasts". Cell. 51 (6): 987–1000. doi:10.1016/0092-8674(87)90585-X. PMID 3690668. S2CID 37741454.
  7. ^ "Entrez Gene: MYOD1 myogenic differentiation 1".
  8. ^ Rudnicki MA, Schnegelsberg PN, Stead RH, Braun T, Arnold HH, Jaenisch R (Dec 1993). "MyoD or Myf-5 is required for the formation of skeletal muscle". Cell. 75 (7): 1351–1359. doi:10.1016/0092-8674(93)90621-V. PMID 8269513. S2CID 27322641.
  9. ^ Hinits Y, Williams VC, Sweetman D, Donn TM, Ma TP, Moens CB, Hughes SM (Oct 2011). "Defective cranial skeletal development, larval lethality and haploinsufficiency in Myod mutant zebrafish". Dev. Biol. 358 (1): 102–112. doi:10.1016/j.ydbio.2011.07.015. PMC 3360969. PMID 21798255.
  10. ^ Tapscott SJ, Davis RL, Thayer MJ, Cheng PF, Weintraub H, Lassar AB (Oct 1988). "MyoD1: a nuclear phosphoprotein requiring a Myc homology region to convert fibroblasts to myoblasts". Science. 242 (4877): 405–511. Bibcode:1988Sci...242..405T. doi:10.1126/science.3175662. PMID 3175662.
  11. ^ Davis RL, Cheng PF, Lassar AB, Thayer M, Tapscott S, Weintraub H (1989). "MyoD and achaete-scute: 4-5 amino acids distinguishes myogenesis from neurogenesis". Princess Takamatsu Symposia. 20: 267–278. PMID 2562185.
  12. ^ Fong, A; Tapscott, S (October 2014). "Skeletal muscle programming and re-programming". Current Opinion in Genetics & Development. 23 (5): 568–573. doi:10.1016/j.gde.2013.05.002. PMC 3775946. PMID 23756045.
  13. ^ Hughes SM, Koishi K, Rudnicki M, Maggs AM (Jan 1997). "MyoD protein is differentially accumulated in fast and slow skeletal muscle fibres and required for normal fibre type balance in rodents". Mech Dev. 61 (1–2): 151–163. doi:10.1016/S0925-4773(96)00631-4. PMID 9076685. S2CID 17769090.
  14. ^ Ehlers ML, Celona B, Black BL (Sep 2014). "NFATc1 controls skeletal muscle fiber type and is a negative regulator of MyoD activity". Cell Reports. 8 (6): 1639–1648. doi:10.1016/j.celrep.2014.08.035. PMC 4180018. PMID 25242327.
  15. ^ Singh K, Cassano M, Planet E, Sebastian S, Jang SM, Sohi G, Faralli H, Choi J, Youn HD, Dilworth FJ, Trono D (Mar 2015). "A KAP1 phosphorylation switch controls MyoD function during skeletal muscle differentiation". Genes & Development. 29 (5): 513–525. doi:10.1101/gad.254532.114. PMC 4358404. PMID 25737281.
  16. ^ Buckingham, M; Rigby, P (February 2014). "Gene Regulatory Networks and Transcriptional Mechanisms that Control Myogenesis". Developmental Cell. 28 (3): 225–238. doi:10.1016/j.devcel.2013.12.020. PMID 24525185.
  17. ^ Song YJ, Choi JH, Lee H (Feb 2015). "Setdb1 Is Required for Myogenic Differentiation of C2C12 Myoblast Cells via Maintenance of MyoD Expression". Molecules and Cells. 38 (4): 362–372. doi:10.14348/molcells.2015.2291. PMC 4400312. PMID 25715926.
  18. ^ Rajabi HN, Takahashi C, Ewen ME (Aug 2014). "Retinoblastoma protein and MyoD function together to effect the repression of Fra-1 and in turn cyclin D1 during terminal cell cycle arrest associated with myogenesis". The Journal of Biological Chemistry. 289 (34): 23417–23427. doi:10.1074/jbc.M113.532572. PMC 4156083. PMID 25006242.
  19. ^ Milewska, M; Grabiec, K; Grzelkowska-Kowalczyk, K (May 2014). "[Interactions of proliferation and differentiation signaling pathways in myogenesis]". Postepy Hig Med Dosw. 68: 516–526. doi:10.5604/17322693.1101617. PMID 24864103.
  20. ^ Pan YC, Wang XW, Teng HF, Wu YJ, Chang HC, Chen SL (Feb 2015). "Wnt3a signal pathways activate MyoD expression by targeting cis-elements inside and outside its distal enhancer". Bioscience Reports. 35 (2): 1–12. doi:10.1042/BSR20140177. PMC 4370097. PMID 25651906.
  21. ^ Motohashi, N.; Asakura, Atsushi (January 2014). "Muscle satellite cell heterogeneity and self-renewal". Frontiers in Cell and Developmental Biology. 2 (1): 1. doi:10.3389/fcell.2014.00001. PMC 4206996. PMID 25364710.
  22. ^ Micheli L, Leonardi L, Conti F, Buanne P, Canu N, Caruso M, Tirone F (March 2005). "PC4 coactivates MyoD by relieving the histone deacetylase 4-mediated inhibition of myocyte enhancer factor 2C". Mol. Cell. Biol. 25 (6): 2242–59. doi:10.1128/MCB.25.6.2242-2259.2005. PMC 1061592. PMID 15743821.
  23. ^ Micheli L, Leonardi L, Conti F, Maresca G, Colazingari S, Mattei E, Lira SA, Farioli-Vecchioli S, Caruso M, Tirone F (February 2011). "PC4/Tis7/IFRD1 stimulates skeletal muscle regeneration and is involved in myoblast differentiation as a regulator of MyoD and NF-kappaB". J. Biol. Chem. 286 (7): 5691–707. doi:10.1074/jbc.M110.162842. PMC 3037682. PMID 21127072.
  24. ^ Ehlers ML, Celona B, Black BL (Sep 2014). "NFATc1 controls skeletal muscle fiber type and is a negative regulator of MyoD activity". Cell Reports. 8 (6): 1639–1648. doi:10.1016/j.celrep.2014.08.035. PMC 4180018. PMID 25242327.
  25. ^ Sartorelli, V; Huang, J; Hamamori, Y; Kedes, L (February 1997). "Molecular mechanisms of myogenic coactivation by p300: direct interaction with the activation domain of MyoD and with the MADS box of MEF2C". Molecular Cell Biology. 17 (2): 1010–1026. doi:10.1128/mcb.17.2.1010. PMC 231826. PMID 9001254.
  26. ^ Bengal E, Ransone L, Scharfmann R, Dwarki VJ, Tapscott SJ, Weintraub H, Verma IM (February 1992). "Functional antagonism between c-Jun and MyoD proteins: a direct physical association". Cell. 68 (3): 507–19. doi:10.1016/0092-8674(92)90187-h. PMID 1310896. S2CID 44966899.
  27. ^ Polesskaya A, Naguibneva I, Duquet A, Bengal E, Robin P, Harel-Bellan A (August 2001). "Interaction between acetylated MyoD and the bromodomain of CBP and/or p300". Mol. Cell. Biol. 21 (16): 5312–20. doi:10.1128/MCB.21.16.5312-5320.2001. PMC 87255. PMID 11463815.
  28. ^ a b Sartorelli V, Huang J, Hamamori Y, Kedes L (February 1997). "Molecular mechanisms of myogenic coactivation by p300: direct interaction with the activation domain of MyoD and with the MADS box of MEF2C". Mol. Cell. Biol. 17 (2): 1010–26. doi:10.1128/mcb.17.2.1010. PMC 231826. PMID 9001254.
  29. ^ Kong Y, Flick MJ, Kudla AJ, Konieczny SF (August 1997). "Muscle LIM protein promotes myogenesis by enhancing the activity of MyoD". Mol. Cell. Biol. 17 (8): 4750–60. doi:10.1128/mcb.17.8.4750. PMC 232327. PMID 9234731.
  30. ^ Zhang JM, Zhao X, Wei Q, Paterson BM (December 1999). "Direct inhibition of G(1) cdk kinase activity by MyoD promotes myoblast cell cycle withdrawal and terminal differentiation". EMBO J. 18 (24): 6983–93. doi:10.1093/emboj/18.24.6983. PMC 1171761. PMID 10601020.
  31. ^ Zhang JM, Wei Q, Zhao X, Paterson BM (February 1999). "Coupling of the cell cycle and myogenesis through the cyclin D1-dependent interaction of MyoD with cdk4". EMBO J. 18 (4): 926–33. doi:10.1093/emboj/18.4.926. PMC 1171185. PMID 10022835.
  32. ^ Reynaud EG, Leibovitch MP, Tintignac LA, Pelpel K, Guillier M, Leibovitch SA (June 2000). "Stabilization of MyoD by direct binding to p57(Kip2)". J. Biol. Chem. 275 (25): 18767–76. doi:10.1074/jbc.M907412199. PMID 10764802.
  33. ^ Lau P, Bailey P, Dowhan DH, Muscat GE (January 1999). "Exogenous expression of a dominant negative RORalpha1 vector in muscle cells impairs differentiation: RORalpha1 directly interacts with p300 and myoD". Nucleic Acids Res. 27 (2): 411–20. doi:10.1093/nar/27.2.411. PMC 148194. PMID 9862959.
  34. ^ Puri PL, Iezzi S, Stiegler P, Chen TT, Schiltz RL, Muscat GE, Giordano A, Kedes L, Wang JY, Sartorelli V (October 2001). "Class I histone deacetylases sequentially interact with MyoD and pRb during skeletal myogenesis". Mol. Cell. 8 (4): 885–97. doi:10.1016/s1097-2765(01)00373-2. PMID 11684023.
  35. ^ a b Mal A, Sturniolo M, Schiltz RL, Ghosh MK, Harter ML (April 2001). "A role for histone deacetylase HDAC1 in modulating the transcriptional activity of MyoD: inhibition of the myogenic program". EMBO J. 20 (7): 1739–53. doi:10.1093/emboj/20.7.1739. PMC 145490. PMID 11285237.
  36. ^ Garkavtsev I, Kozin SV, Chernova O, Xu L, Winkler F, Brown E, Barnett GH, Jain RK (March 2004). "The candidate tumour suppressor protein ING4 regulates brain tumour growth and angiogenesis". Nature. 428 (6980): 328–32. Bibcode:2004Natur.428..328G. doi:10.1038/nature02329. PMID 15029197. S2CID 4427531.
  37. ^ a b c Langlands K, Yin X, Anand G, Prochownik EV (August 1997). "Differential interactions of Id proteins with basic-helix-loop-helix transcription factors". J. Biol. Chem. 272 (32): 19785–93. doi:10.1074/jbc.272.32.19785. PMID 9242638.
  38. ^ Finkel T, Duc J, Fearon ER, Dang CV, Tomaselli GF (January 1993). "Detection and modulation in vivo of helix-loop-helix protein-protein interactions". J. Biol. Chem. 268 (1): 5–8. doi:10.1016/S0021-9258(18)54105-3. PMID 8380166.
  39. ^ Gupta K, Anand G, Yin X, Grove L, Prochownik EV (March 1998). "Mmip1: a novel leucine zipper protein that reverses the suppressive effects of Mad family members on c-myc". Oncogene. 16 (9): 1149–59. doi:10.1038/sj.onc.1201634. PMID 9528857.
  40. ^ McLoughlin P, Ehler E, Carlile G, Licht JD, Schäfer BW (October 2002). "The LIM-only protein DRAL/FHL2 interacts with and is a corepressor for the promyelocytic leukemia zinc finger protein". J. Biol. Chem. 277 (40): 37045–53. doi:10.1074/jbc.M203336200. PMID 12145280.
  41. ^ Ling MT, Chiu YT, Lee TK, Leung SC, Fung MK, Wang X, Wong KF, Wong YC (September 2008). "Id-1 induces proteasome-dependent degradation of the HBX protein". J. Mol. Biol. 382 (1): 34–43. doi:10.1016/j.jmb.2007.06.020. PMID 18674781.
  42. ^ Chen CM, Kraut N, Groudine M, Weintraub H (September 1996). "I-mf, a novel myogenic repressor, interacts with members of the MyoD family". Cell. 86 (5): 731–41. doi:10.1016/s0092-8674(00)80148-8. PMID 8797820. S2CID 16252710.
  43. ^ Lenormand JL, Benayoun B, Guillier M, Vandromme M, Leibovitch MP, Leibovitch SA (February 1997). "Mos activates myogenic differentiation by promoting heterodimerization of MyoD and E12 proteins". Mol. Cell. Biol. 17 (2): 584–93. doi:10.1128/mcb.17.2.584. PMC 231783. PMID 9001211.
  44. ^ Gu W, Schneider JW, Condorelli G, Kaushal S, Mahdavi V, Nadal-Ginard B (February 1993). "Interaction of myogenic factors and the retinoblastoma protein mediates muscle cell commitment and differentiation". Cell. 72 (3): 309–24. doi:10.1016/0092-8674(93)90110-c. PMID 8381715. S2CID 21581966.
  45. ^ Froeschlé A, Alric S, Kitzmann M, Carnac G, Auradé F, Rochette-Egly C, Bonnieu A (July 1998). "Retinoic acid receptors and muscle b-HLH proteins: partners in retinoid-induced myogenesis". Oncogene. 16 (26): 3369–78. doi:10.1038/sj.onc.1201894. PMID 9692544.
  46. ^ Kataoka Y, Matsumura I, Ezoe S, Nakata S, Takigawa E, Sato Y, Kawasaki A, Yokota T, Nakajima K, Felsani A, Kanakura Y (November 2003). "Reciprocal inhibition between MyoD and STAT3 in the regulation of growth and differentiation of myoblasts". J. Biol. Chem. 278 (45): 44178–87. doi:10.1074/jbc.M304884200. PMID 12947115.
  47. ^ Maleki SJ, Royer CA, Hurlburt BK (June 1997). "MyoD-E12 heterodimers and MyoD-MyoD homodimers are equally stable". Biochemistry. 36 (22): 6762–7. doi:10.1021/bi970262m. PMID 9184158.

External links edit

  • MyoD+Protein at the U.S. National Library of Medicine Medical Subject Headings (MeSH)
  • Overview of all the structural information available in the PDB for UniProt: P10085 (Mouse Myoblast determination protein 1) at the PDBe-KB.

myod, confused, with, myd88, also, known, myoblast, determination, protein, protein, animals, that, plays, major, role, regulating, muscle, differentiation, which, discovered, laboratory, harold, weintraub, belongs, family, proteins, known, myogenic, regulator. Not to be confused with MyD88 MyoD also known as myoblast determination protein 1 5 is a protein in animals that plays a major role in regulating muscle differentiation MyoD which was discovered in the laboratory of Harold M Weintraub 6 belongs to a family of proteins known as myogenic regulatory factors MRFs 7 These bHLH basic helix loop helix transcription factors act sequentially in myogenic differentiation Vertebrate MRF family members include MyoD1 Myf5 myogenin and MRF4 Myf6 In non vertebrate animals a single MyoD protein is typically found MYOD1Available structuresPDBOrtholog search PDBe RCSBList of PDB id codes1MDYIdentifiersAliasesMYOD1 MYF3 MYOD PUM bHLHc1 myogenic differentiation 1 MYODRIFExternal IDsOMIM 159970 MGI 97275 HomoloGene 7857 GeneCards MYOD1Gene location Human Chr Chromosome 11 human 1 Band11p15 1Start17 719 571 bp 1 End17 722 136 bp 1 Gene location Mouse Chr Chromosome 7 mouse 2 Band7 B3 7 30 03 cMStart46 025 898 bp 2 End46 028 523 bp 2 RNA expression patternBgeeHumanMouse ortholog Top expressed intriceps brachii musclevastus lateralis musclegastrocnemius muscledeltoid muscletibialis anterior musclejejunumplacentaheadexocrine glandmouthTop expressed inwristtriceps brachii musclesternocleidomastoid muscletemporal muscleskeletal muscle tissueknee jointextensor digitorum longus muscleextraocular musclequadriceps femoris musclevastus lateralis muscleMore reference expression dataBioGPSMore reference expression dataGene ontologyMolecular functionDNA binding sequence specific DNA binding RNA polymerase II transcription regulatory region sequence specific DNA binding protein dimerization activity DNA binding transcription activator activity RNA polymerase II specific DNA binding transcription factor activity transcription factor binding chromatin binding RNA polymerase II cis regulatory region sequence specific DNA binding E box binding protein binding protein heterodimerization activity enzyme binding transcription factor activity RNA polymerase II distal enhancer sequence specific binding chromatin DNA binding ubiquitin protein ligase binding transcription coactivator activity DNA binding transcription factor activity RNA polymerase II specific promoter specific chromatin bindingCellular componenttranscription regulator complex nucleoplasm myofibril nucleus cytoplasm cytosol nuclear bodyBiological processcell differentiation myotube differentiation regulation of transcription DNA templated regulation of transcription by RNA polymerase II cellular response to starvation muscle cell fate commitment cellular response to estradiol stimulus skeletal muscle fiber adaptation muscle organ development regulation of alternative mRNA splicing via spliceosome myotube cell development positive regulation of skeletal muscle fiber development negative regulation of myoblast proliferation transcription DNA templated multicellular organism development positive regulation of transcription DNA templated protein phosphorylation regulation of RNA splicing positive regulation of myoblast fusion myoblast fate determination cellular response to glucocorticoid stimulus positive regulation of myoblast differentiation skeletal muscle fiber development histone H3 acetylation myotube differentiation involved in skeletal muscle regeneration histone H4 acetylation skeletal muscle cell differentiation positive regulation of skeletal muscle tissue regeneration regulation of gene expression striated muscle cell differentiation cellular response to oxygen levels myoblast fusion myoblast differentiation skeletal muscle tissue development positive regulation of transcription by RNA polymerase II transcription by RNA polymerase II positive regulation of muscle cell differentiation cellular response to tumor necrosis factor positive regulation of snRNA transcription by RNA polymerase IISources Amigo QuickGOOrthologsSpeciesHumanMouseEntrez465417927EnsemblENSG00000129152ENSMUSG00000009471UniProtP15172P10085RefSeq mRNA NM 002478NM 010866RefSeq protein NP 002469NP 034996Location UCSC Chr 11 17 72 17 72 MbChr 7 46 03 46 03 MbPubMed search 3 4 WikidataView Edit HumanView Edit MouseMyoD is one of the earliest markers of myogenic commitment MyoD is expressed at extremely low and essentially undetectable levels in quiescent satellite cells but expression of MyoD is activated in response to exercise or muscle tissue damage The effect of MyoD on satellite cells is dose dependent high MyoD expression represses cell renewal promotes terminal differentiation and can induce apoptosis Although MyoD marks myoblast commitment muscle development is not dramatically ablated in mouse mutants lacking the MyoD gene This is likely due to functional redundancy from Myf5 and or Mrf4 Nevertheless the combination of MyoD and Myf5 is vital to the success of myogenesis 8 9 Contents 1 History 2 Function 3 Mechanisms 4 Wnt signalling can affect MyoD 5 Coactivators and repressors 6 Interactions 7 References 8 External linksHistory editMyoD was cloned by a functional assay for muscle formation reported in Cell in 1987 by Davis Weintraub and Lassar It was first described as a nuclear phosphoprotein in 1988 by Tapscott Davis Thayer Cheng Weintraub and Lassar in Science The researchers expressed the complementary DNA cDNA of the murine MyoD protein in a different cell lines fibroblast and adipoblast and found MyoD converted them to myogenic cells 6 10 The following year the same research team performed several tests to determine both the structure and function of the protein confirming their initial proposal that the active site of the protein consisted of the helix loop helix now referred to as basic helix loop helix for dimerization and a basic site upstream of this bHLH region facilitated DNA binding only once it became a protein dimer 11 MyoD has since been an active area of research as still relatively little is known concerning many aspects of its function Function editThe function of MyoD in development is to commit mesoderm cells to a skeletal myoblast lineage and then to regulate that continued state MyoD may also regulate muscle repair MyoD mRNA levels are also reported to be elevated in aging skeletal muscle One of the main actions of MyoD is to remove cells from the cell cycle halt proliferation for terminal cell cycle arrest in differentiated myocytes by enhancing the transcription of p21 and myogenin MyoD is inhibited by cyclin dependent kinases CDKs CDKs are in turn inhibited by p21 Thus MyoD enhances its own activity in the cell in a feedforward manner Sustained MyoD expression is necessary for retaining the expression of muscle related genes 12 MyoD is also an important effector for the fast twitch muscle fiber types IIA IIX and IIB phenotype 13 14 Mechanisms editMyoD is a transcription factor and can also direct chromatin remodelling through binding to a DNA motif known as the E box MyoD is known to have binding interactions with hundreds of muscular gene promoters and to permit myoblast proliferation While not completely understood MyoD is now thought to function as a major myogenesis controller in an on off switch association mediated by KAP1 KRAB Kruppel like associated box associated protein 1 phosphorylation 15 KAP1 is localized at muscle related genes in myoblasts along with both MyoD and Mef2 a myocyte transcription enhancer factor Here it serves as a scaffold and recruits the coactivators p300 and LSD1 in addition to several corepressors which include G9a and the Histone deacetylase HDAC1 The consequence of this coactivator corepressor recruitment is silenced promoting regions on muscle genes When the kinase MSK1 phosphorylates KAP1 the corepressors previously bound to the scaffold are released allowing MyoD and Mef2 to activate transcription 16 Once the master controller MyoD has become active SETDB1 is required to maintain MyoD expression within the cell Setdb1 appears to be necessary to maintain both MyoD expression and also genes that are specific to muscle tissues because reduction of Setdb1 expression results in a severe delay of myoblast differentiation and determination 17 In Setdb1 depleted myoblasts that are treated with exogenous MyoD myoblastic differentiation is successfully restored In one model of Setdb1 action on MyoD Setdb1 represses an inhibitor of MyoD This unidentified inhibitor likely acts competitively against MyoD during typical cellular proliferation Evidence for this model is that reduction of Setdb1 results in direct inhibition of myoblast differentiation which may be caused by the release of the unknown MyoD inhibitor nbsp Evidence suggests that Setdb1 inhibits a repressor of MyoD and this is the mechanism through which MyoD expression is retained in differentiated myoblasts MyoD has also been shown to function cooperatively with the tumor suppressor gene Retinoblastoma pRb to cause cell cycle arrest in the terminally differentiated myoblasts 18 This is done through regulation of the Cyclin Cyclin D1 Cell cycle arrest in which myoblasts would indicate the conclusion of myogenesis is dependent on the continuous and stable repression of the D1 cyclin Both MyoD and pRb are necessary for the repression of cyclin D1 but rather than acting directly on cyclin D1 they act on Fra 1 which is immediately early of cyclin D1 MyoD and pRb are both necessary for repressing Fra 1 and thus cyclin D1 as either MyoD or pRb on its own is not sufficient alone to induce cyclin D1 repression and thus cell cycle arrest In an intronic enhancer of Fra 1 there were two conserved MyoD binding sites discovered There is cooperative action of MyoD and pRb at the Fra 1 intronic enhancer that suppresses the enhancer therefore suppressing cyclin D1 and ultimately resulting in cell cycle arrest for terminally differentiated myoblasts 19 Wnt signalling can affect MyoD editWnt signalling from adjacent tissues has been shown to induce cells in somites that receive these Wnt signals to express Pax3 and Pax7 in addition to myogenic regulatory factors including Myf5 and MyoD Specifically Wnt3a can directly induce MyoD expression via cis element interactions with a distal enhancer and Wnt response element 20 Wnt1 from dorsal neural tube and Wnt6 Wnt7a from surface ectoderm have also been implicated in promoting myogenesis in the somite the latter signals may act primarily through Myod In typical adult muscles in a resting condition absence of physiological stress the specific Wnt family proteins that are expressed are Wnt5a Wnt5b Wnt7a and Wnt4 When a muscle becomes injured thus requiring regeneration Wnt5a Wnt5b and Wnt7a are increased in expression As the muscle completes repair Wnt7b and Wnt3a are increased as well This patterning of Wnt signalling expression in muscle cell repair induces the differentiation of the progenitor cells which reduces the number of available satellite cells Wnt plays a crucial role in satellite cell regulation and skeletal muscle aging and also regeneration Wnts are known to active the expression of Myf5 and MyoD by Wnt1 and Wnt7a Wnt4 Wnt5 and Wnt6 function to increase the expression of both of the regulatory factors but at a more subtle level Additionally MyoD increases Wnt3a when myoblasts undergo differentiation Whether MyoD is activated by Wnt via cis regulation direct targeting or through indirect physiological pathways remains to be elucidated 21 Coactivators and repressors editIFRD1 is a positive cofactor of MyoD as it cooperates with MyoD at inducing the transcriptional activity of MEF2C by displacing HDAC4 from MEF2C moreover IFRD1 also represses the transcriptional activity of NF kB which is known to inhibit MyoD mRNA accumulation 22 23 NFATc1 is a transcription factor that regulates composition of fiber type and the fast to slow twitch transition resulting from aerobic exercise requires the expression of NFATc1 MyoD expression is a key transcription factor in fast twitch fibers which is inhibited by NFATc1 in oxidative fiber types NFATc1 works to inhibit MyoD via a physical interaction with the MyoD N terminal activation domain resulting in inhibited recruitment of the necessary transcriptional coactivator p300 NFATc1 physically disrupts the interaction between MyoD and p300 This establishes the molecular mechanism by which fiber types transition in vivo through exercise with opposing roles for NFATc1 and MyoD NFATc1 controls this balance by physical inhibition of MyoD in slow twitch muscle fiber types 24 nbsp MyoD works with a transient placeholder protein that functions to prevent other transcription factors from binding to the DNA and also retains an inactive conformation for the DNA Once the placeholder is removed or possibly deactivated the necessary transcription factors are free to bind and initiate recruitment of RNA Polymerase II and initiate active RNA transcription The histone deacetyltransferase p300 functions with MyoD in an interaction that is essential for the myotube generation from fibroblasts that is mediated by MyoD Recruitment of p300 is the rate limiting process in the conversion of fibroblasts to myotubes 25 In addition to p300 MyoD is also known to recruit Set7 H3K4me1 H3K27ac and RNAP II to the enhancer that is bound with and this allows for the activation of muscle gene that is condition specific and established by MyoD recruitment Endogenous p300 though is necessary for MyoD functioning by acting as an essential coactivator MyoD associatively binds to the enhancer region in conjunction with a placeholding putative pioneer factor which helps to establish and maintain a both of them in a specific and inactive conformation Upon the removal or inactivation on the placeholder protein bound to the enhancer the recruitment of the additional group of transcription factors that help to positively regulate enhancer activity is permitted and this results in the MyoD transcription factor enhancer complex to assume a transcriptionally active state Interactions editMyoD has been shown to interact with C jun 26 CREB binding protein 27 28 CSRP3 29 Cyclin dependent kinase 4 30 31 Cyclin dependent kinase inhibitor 1C 32 EP300 28 33 HDAC1 34 35 ID1 36 37 38 39 40 41 ID2 37 MDFI 42 MOS 43 Retinoblastoma protein 35 44 Retinoid X receptor alpha 45 STAT3 46 and TCF3 37 47 References edit a b c GRCh38 Ensembl release 89 ENSG00000129152 Ensembl May 2017 a b c GRCm38 Ensembl release 89 ENSMUSG00000009471 Ensembl May 2017 Human PubMed Reference National Center for Biotechnology Information U S National Library of Medicine Mouse PubMed Reference National Center for Biotechnology Information U S National Library of Medicine P15172 MYOD1 HUMAN UniProtKB Retrieved 17 July 2019 a b Davis RL Weintraub H Lassar AB Dec 1987 Expression of a single transfected cDNA converts fibroblasts to myoblasts Cell 51 6 987 1000 doi 10 1016 0092 8674 87 90585 X PMID 3690668 S2CID 37741454 Entrez Gene MYOD1 myogenic differentiation 1 Rudnicki MA Schnegelsberg PN Stead RH Braun T Arnold HH Jaenisch R Dec 1993 MyoD or Myf 5 is required for the formation of skeletal muscle Cell 75 7 1351 1359 doi 10 1016 0092 8674 93 90621 V PMID 8269513 S2CID 27322641 Hinits Y Williams VC Sweetman D Donn TM Ma TP Moens CB Hughes SM Oct 2011 Defective cranial skeletal development larval lethality and haploinsufficiency in Myod mutant zebrafish Dev Biol 358 1 102 112 doi 10 1016 j ydbio 2011 07 015 PMC 3360969 PMID 21798255 Tapscott SJ Davis RL Thayer MJ Cheng PF Weintraub H Lassar AB Oct 1988 MyoD1 a nuclear phosphoprotein requiring a Myc homology region to convert fibroblasts to myoblasts Science 242 4877 405 511 Bibcode 1988Sci 242 405T doi 10 1126 science 3175662 PMID 3175662 Davis RL Cheng PF Lassar AB Thayer M Tapscott S Weintraub H 1989 MyoD and achaete scute 4 5 amino acids distinguishes myogenesis from neurogenesis Princess Takamatsu Symposia 20 267 278 PMID 2562185 Fong A Tapscott S October 2014 Skeletal muscle programming and re programming Current Opinion in Genetics amp Development 23 5 568 573 doi 10 1016 j gde 2013 05 002 PMC 3775946 PMID 23756045 Hughes SM Koishi K Rudnicki M Maggs AM Jan 1997 MyoD protein is differentially accumulated in fast and slow skeletal muscle fibres and required for normal fibre type balance in rodents Mech Dev 61 1 2 151 163 doi 10 1016 S0925 4773 96 00631 4 PMID 9076685 S2CID 17769090 Ehlers ML Celona B Black BL Sep 2014 NFATc1 controls skeletal muscle fiber type and is a negative regulator of MyoD activity Cell Reports 8 6 1639 1648 doi 10 1016 j celrep 2014 08 035 PMC 4180018 PMID 25242327 Singh K Cassano M Planet E Sebastian S Jang SM Sohi G Faralli H Choi J Youn HD Dilworth FJ Trono D Mar 2015 A KAP1 phosphorylation switch controls MyoD function during skeletal muscle differentiation Genes amp Development 29 5 513 525 doi 10 1101 gad 254532 114 PMC 4358404 PMID 25737281 Buckingham M Rigby P February 2014 Gene Regulatory Networks and Transcriptional Mechanisms that Control Myogenesis Developmental Cell 28 3 225 238 doi 10 1016 j devcel 2013 12 020 PMID 24525185 Song YJ Choi JH Lee H Feb 2015 Setdb1 Is Required for Myogenic Differentiation of C2C12 Myoblast Cells via Maintenance of MyoD Expression Molecules and Cells 38 4 362 372 doi 10 14348 molcells 2015 2291 PMC 4400312 PMID 25715926 Rajabi HN Takahashi C Ewen ME Aug 2014 Retinoblastoma protein and MyoD function together to effect the repression of Fra 1 and in turn cyclin D1 during terminal cell cycle arrest associated with myogenesis The Journal of Biological Chemistry 289 34 23417 23427 doi 10 1074 jbc M113 532572 PMC 4156083 PMID 25006242 Milewska M Grabiec K Grzelkowska Kowalczyk K May 2014 Interactions of proliferation and differentiation signaling pathways in myogenesis Postepy Hig Med Dosw 68 516 526 doi 10 5604 17322693 1101617 PMID 24864103 Pan YC Wang XW Teng HF Wu YJ Chang HC Chen SL Feb 2015 Wnt3a signal pathways activate MyoD expression by targeting cis elements inside and outside its distal enhancer Bioscience Reports 35 2 1 12 doi 10 1042 BSR20140177 PMC 4370097 PMID 25651906 Motohashi N Asakura Atsushi January 2014 Muscle satellite cell heterogeneity and self renewal Frontiers in Cell and Developmental Biology 2 1 1 doi 10 3389 fcell 2014 00001 PMC 4206996 PMID 25364710 Micheli L Leonardi L Conti F Buanne P Canu N Caruso M Tirone F March 2005 PC4 coactivates MyoD by relieving the histone deacetylase 4 mediated inhibition of myocyte enhancer factor 2C Mol Cell Biol 25 6 2242 59 doi 10 1128 MCB 25 6 2242 2259 2005 PMC 1061592 PMID 15743821 Micheli L Leonardi L Conti F Maresca G Colazingari S Mattei E Lira SA Farioli Vecchioli S Caruso M Tirone F February 2011 PC4 Tis7 IFRD1 stimulates skeletal muscle regeneration and is involved in myoblast differentiation as a regulator of MyoD and NF kappaB J Biol Chem 286 7 5691 707 doi 10 1074 jbc M110 162842 PMC 3037682 PMID 21127072 Ehlers ML Celona B Black BL Sep 2014 NFATc1 controls skeletal muscle fiber type and is a negative regulator of MyoD activity Cell Reports 8 6 1639 1648 doi 10 1016 j celrep 2014 08 035 PMC 4180018 PMID 25242327 Sartorelli V Huang J Hamamori Y Kedes L February 1997 Molecular mechanisms of myogenic coactivation by p300 direct interaction with the activation domain of MyoD and with the MADS box of MEF2C Molecular Cell Biology 17 2 1010 1026 doi 10 1128 mcb 17 2 1010 PMC 231826 PMID 9001254 Bengal E Ransone L Scharfmann R Dwarki VJ Tapscott SJ Weintraub H Verma IM February 1992 Functional antagonism between c Jun and MyoD proteins a direct physical association Cell 68 3 507 19 doi 10 1016 0092 8674 92 90187 h PMID 1310896 S2CID 44966899 Polesskaya A Naguibneva I Duquet A Bengal E Robin P Harel Bellan A August 2001 Interaction between acetylated MyoD and the bromodomain of CBP and or p300 Mol Cell Biol 21 16 5312 20 doi 10 1128 MCB 21 16 5312 5320 2001 PMC 87255 PMID 11463815 a b Sartorelli V Huang J Hamamori Y Kedes L February 1997 Molecular mechanisms of myogenic coactivation by p300 direct interaction with the activation domain of MyoD and with the MADS box of MEF2C Mol Cell Biol 17 2 1010 26 doi 10 1128 mcb 17 2 1010 PMC 231826 PMID 9001254 Kong Y Flick MJ Kudla AJ Konieczny SF August 1997 Muscle LIM protein promotes myogenesis by enhancing the activity of MyoD Mol Cell Biol 17 8 4750 60 doi 10 1128 mcb 17 8 4750 PMC 232327 PMID 9234731 Zhang JM Zhao X Wei Q Paterson BM December 1999 Direct inhibition of G 1 cdk kinase activity by MyoD promotes myoblast cell cycle withdrawal and terminal differentiation EMBO J 18 24 6983 93 doi 10 1093 emboj 18 24 6983 PMC 1171761 PMID 10601020 Zhang JM Wei Q Zhao X Paterson BM February 1999 Coupling of the cell cycle and myogenesis through the cyclin D1 dependent interaction of MyoD with cdk4 EMBO J 18 4 926 33 doi 10 1093 emboj 18 4 926 PMC 1171185 PMID 10022835 Reynaud EG Leibovitch MP Tintignac LA Pelpel K Guillier M Leibovitch SA June 2000 Stabilization of MyoD by direct binding to p57 Kip2 J Biol Chem 275 25 18767 76 doi 10 1074 jbc M907412199 PMID 10764802 Lau P Bailey P Dowhan DH Muscat GE January 1999 Exogenous expression of a dominant negative RORalpha1 vector in muscle cells impairs differentiation RORalpha1 directly interacts with p300 and myoD Nucleic Acids Res 27 2 411 20 doi 10 1093 nar 27 2 411 PMC 148194 PMID 9862959 Puri PL Iezzi S Stiegler P Chen TT Schiltz RL Muscat GE Giordano A Kedes L Wang JY Sartorelli V October 2001 Class I histone deacetylases sequentially interact with MyoD and pRb during skeletal myogenesis Mol Cell 8 4 885 97 doi 10 1016 s1097 2765 01 00373 2 PMID 11684023 a b Mal A Sturniolo M Schiltz RL Ghosh MK Harter ML April 2001 A role for histone deacetylase HDAC1 in modulating the transcriptional activity of MyoD inhibition of the myogenic program EMBO J 20 7 1739 53 doi 10 1093 emboj 20 7 1739 PMC 145490 PMID 11285237 Garkavtsev I Kozin SV Chernova O Xu L Winkler F Brown E Barnett GH Jain RK March 2004 The candidate tumour suppressor protein ING4 regulates brain tumour growth and angiogenesis Nature 428 6980 328 32 Bibcode 2004Natur 428 328G doi 10 1038 nature02329 PMID 15029197 S2CID 4427531 a b c Langlands K Yin X Anand G Prochownik EV August 1997 Differential interactions of Id proteins with basic helix loop helix transcription factors J Biol Chem 272 32 19785 93 doi 10 1074 jbc 272 32 19785 PMID 9242638 Finkel T Duc J Fearon ER Dang CV Tomaselli GF January 1993 Detection and modulation in vivo of helix loop helix protein protein interactions J Biol Chem 268 1 5 8 doi 10 1016 S0021 9258 18 54105 3 PMID 8380166 Gupta K Anand G Yin X Grove L Prochownik EV March 1998 Mmip1 a novel leucine zipper protein that reverses the suppressive effects of Mad family members on c myc Oncogene 16 9 1149 59 doi 10 1038 sj onc 1201634 PMID 9528857 McLoughlin P Ehler E Carlile G Licht JD Schafer BW October 2002 The LIM only protein DRAL FHL2 interacts with and is a corepressor for the promyelocytic leukemia zinc finger protein J Biol Chem 277 40 37045 53 doi 10 1074 jbc M203336200 PMID 12145280 Ling MT Chiu YT Lee TK Leung SC Fung MK Wang X Wong KF Wong YC September 2008 Id 1 induces proteasome dependent degradation of the HBX protein J Mol Biol 382 1 34 43 doi 10 1016 j jmb 2007 06 020 PMID 18674781 Chen CM Kraut N Groudine M Weintraub H September 1996 I mf a novel myogenic repressor interacts with members of the MyoD family Cell 86 5 731 41 doi 10 1016 s0092 8674 00 80148 8 PMID 8797820 S2CID 16252710 Lenormand JL Benayoun B Guillier M Vandromme M Leibovitch MP Leibovitch SA February 1997 Mos activates myogenic differentiation by promoting heterodimerization of MyoD and E12 proteins Mol Cell Biol 17 2 584 93 doi 10 1128 mcb 17 2 584 PMC 231783 PMID 9001211 Gu W Schneider JW Condorelli G Kaushal S Mahdavi V Nadal Ginard B February 1993 Interaction of myogenic factors and the retinoblastoma protein mediates muscle cell commitment and differentiation Cell 72 3 309 24 doi 10 1016 0092 8674 93 90110 c PMID 8381715 S2CID 21581966 Froeschle A Alric S Kitzmann M Carnac G Aurade F Rochette Egly C Bonnieu A July 1998 Retinoic acid receptors and muscle b HLH proteins partners in retinoid induced myogenesis Oncogene 16 26 3369 78 doi 10 1038 sj onc 1201894 PMID 9692544 Kataoka Y Matsumura I Ezoe S Nakata S Takigawa E Sato Y Kawasaki A Yokota T Nakajima K Felsani A Kanakura Y November 2003 Reciprocal inhibition between MyoD and STAT3 in the regulation of growth and differentiation of myoblasts J Biol Chem 278 45 44178 87 doi 10 1074 jbc M304884200 PMID 12947115 Maleki SJ Royer CA Hurlburt BK June 1997 MyoD E12 heterodimers and MyoD MyoD homodimers are equally stable Biochemistry 36 22 6762 7 doi 10 1021 bi970262m PMID 9184158 External links editMyoD Protein at the U S National Library of Medicine Medical Subject Headings MeSH Overview of all the structural information available in the PDB for UniProt P10085 Mouse Myoblast determination protein 1 at the PDBe KB Retrieved from https en wikipedia org w index php title MyoD amp oldid 1120413401, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.