Skip to main content
The Journal of Cell Biology logoLink to The Journal of Cell Biology
. 1994 Dec 2;127(6):1755–1766. doi: 10.1083/jcb.127.6.1755

Bone morphogenetic protein-2 converts the differentiation pathway of C2C12 myoblasts into the osteoblast lineage [published erratum appears in J Cell Biol 1995 Feb;128(4):following 713]

PMCID: PMC2120318  PMID: 7798324

Abstract

The implantation of bone morphogenetic protein (BMP) into muscular tissues induces ectopic bone formation at the site of implantation. To investigate the mechanism underlying this process, we examined whether recombinant bone morphogenetic protein-2 (BMP-2) converts the differentiation pathway of the clonal myoblastic cell line, C2C12, into that of osteoblast lineage. Incubating the cells with 300 ng/ml of BMP- 2 for 6 d almost completely inhibited the formation of the multinucleated myotubes expressing troponin T and myosin heavy chain, and induced the appearance of numerous alkaline phosphatase (ALP)- positive cells. BMP-2 dose dependently induced ALP activity, parathyroid hormone (PTH)-dependent 3',5'-cAMP production, and osteocalcin production at concentrations above 100 ng/ml. The concentration of BMP-2 required to induce these osteoblastic phenotypes was the same as that required to almost completely inhibit myotube formation. Incubating primary muscle cells with 300 ng/ml of BMP-2 for 6 d also inhibited myotube formation, whereas induced ALP activity and osteocalcin production. Incubation with 300 ng/ml of BMP-2 suppressed the expression of mRNA for muscle creatine kinase within 6 h, whereas it induced mRNA expression for ALP, PTH/PTH-related protein (PTHrP) receptors, and osteocalcin within 24-48 h. BMP-2 completely inhibited the expression of myogenin mRNA by day 3. By day 3, BMP-2 also inhibited the expression of MyoD mRNA, but it was transiently stimulated 12 h after exposure to BMP-2. Expression of Id-1 mRNA was greatly stimulated by BMP-2. When C2C12 cells pretreated with BMP-2 for 6 d were transferred to a colony assay system in the absence of BMP-2, more than 84% of the colonies generated became troponin T-positive and ALP activity disappeared. TGF-beta 1 also inhibited myotube formation in C2C12 cells, and suppressed the expression of myogenin and MyoD mRNAs without inducing that of Id-1 mRNA. However, no osteoblastic phenotype was induced by TGF-beta 1 in C2C12 cells. TGF-beta 1 potentiated the inhibitory effect of BMP-2 on myotube formation, whereas TGF-beta 1 reduced ALP activity and osteocalcin production induced by BMP-2 in C2C12 cells. These results indicate that BMP-2 specifically converts the differentiation pathway of C2C12 myoblasts into that of osteoblast lineage cells, but that the conversion is not heritable.

Full Text

The Full Text of this article is available as a PDF (5.2 MB).

Selected References

These references are in PubMed. This may not be the complete list of references from this article.

  1. Abe H., Komiya T., Obinata T. Expression of multiple troponin T variants in neonatal chicken breast muscle. Dev Biol. 1986 Nov;118(1):42–51. doi: 10.1016/0012-1606(86)90071-0. [DOI] [PubMed] [Google Scholar]
  2. Bader D., Masaki T., Fischman D. A. Immunochemical analysis of myosin heavy chain during avian myogenesis in vivo and in vitro. J Cell Biol. 1982 Dec;95(3):763–770. doi: 10.1083/jcb.95.3.763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Benayahu D., Kletter Y., Zipori D., Wientroub S. Bone marrow-derived stromal cell line expressing osteoblastic phenotype in vitro and osteogenic capacity in vivo. J Cell Physiol. 1989 Jul;140(1):1–7. doi: 10.1002/jcp.1041400102. [DOI] [PubMed] [Google Scholar]
  4. Benezra R., Davis R. L., Lockshon D., Turner D. L., Weintraub H. The protein Id: a negative regulator of helix-loop-helix DNA binding proteins. Cell. 1990 Apr 6;61(1):49–59. doi: 10.1016/0092-8674(90)90214-y. [DOI] [PubMed] [Google Scholar]
  5. Blau H. M., Chiu C. P., Webster C. Cytoplasmic activation of human nuclear genes in stable heterocaryons. Cell. 1983 Apr;32(4):1171–1180. doi: 10.1016/0092-8674(83)90300-8. [DOI] [PubMed] [Google Scholar]
  6. Braun T., Bober E., Winter B., Rosenthal N., Arnold H. H. Myf-6, a new member of the human gene family of myogenic determination factors: evidence for a gene cluster on chromosome 12. EMBO J. 1990 Mar;9(3):821–831. doi: 10.1002/j.1460-2075.1990.tb08179.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Braun T., Buschhausen-Denker G., Bober E., Tannich E., Arnold H. H. A novel human muscle factor related to but distinct from MyoD1 induces myogenic conversion in 10T1/2 fibroblasts. EMBO J. 1989 Mar;8(3):701–709. doi: 10.1002/j.1460-2075.1989.tb03429.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Braun T., Rudnicki M. A., Arnold H. H., Jaenisch R. Targeted inactivation of the muscle regulatory gene Myf-5 results in abnormal rib development and perinatal death. Cell. 1992 Oct 30;71(3):369–382. doi: 10.1016/0092-8674(92)90507-9. [DOI] [PubMed] [Google Scholar]
  9. Brennan T. J., Edmondson D. G., Li L., Olson E. N. Transforming growth factor beta represses the actions of myogenin through a mechanism independent of DNA binding. Proc Natl Acad Sci U S A. 1991 May 1;88(9):3822–3826. doi: 10.1073/pnas.88.9.3822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Brennan T. J., Olson E. N. Myogenin resides in the nucleus and acquires high affinity for a conserved enhancer element on heterodimerization. Genes Dev. 1990 Apr;4(4):582–595. doi: 10.1101/gad.4.4.582. [DOI] [PubMed] [Google Scholar]
  11. Brunetti A., Goldfine I. D. Role of myogenin in myoblast differentiation and its regulation by fibroblast growth factor. J Biol Chem. 1990 Apr 15;265(11):5960–5963. [PubMed] [Google Scholar]
  12. Celeste A. J., Iannazzi J. A., Taylor R. C., Hewick R. M., Rosen V., Wang E. A., Wozney J. M. Identification of transforming growth factor beta family members present in bone-inductive protein purified from bovine bone. Proc Natl Acad Sci U S A. 1990 Dec;87(24):9843–9847. doi: 10.1073/pnas.87.24.9843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Celeste A. J., Rosen V., Buecker J. L., Kriz R., Wang E. A., Wozney J. M. Isolation of the human gene for bone gla protein utilizing mouse and rat cDNA clones. EMBO J. 1986 Aug;5(8):1885–1890. doi: 10.1002/j.1460-2075.1986.tb04440.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Chomczynski P., Sacchi N. Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction. Anal Biochem. 1987 Apr;162(1):156–159. doi: 10.1006/abio.1987.9999. [DOI] [PubMed] [Google Scholar]
  15. Clegg C. H., Linkhart T. A., Olwin B. B., Hauschka S. D. Growth factor control of skeletal muscle differentiation: commitment to terminal differentiation occurs in G1 phase and is repressed by fibroblast growth factor. J Cell Biol. 1987 Aug;105(2):949–956. doi: 10.1083/jcb.105.2.949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Davis R. L., Cheng P. F., Lassar A. B., Weintraub H. The MyoD DNA binding domain contains a recognition code for muscle-specific gene activation. Cell. 1990 Mar 9;60(5):733–746. doi: 10.1016/0092-8674(90)90088-v. [DOI] [PubMed] [Google Scholar]
  17. Davis R. L., Weintraub H., Lassar A. B. Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell. 1987 Dec 24;51(6):987–1000. doi: 10.1016/0092-8674(87)90585-x. [DOI] [PubMed] [Google Scholar]
  18. Edmondson D. G., Olson E. N. A gene with homology to the myc similarity region of MyoD1 is expressed during myogenesis and is sufficient to activate the muscle differentiation program. Genes Dev. 1989 May;3(5):628–640. doi: 10.1101/gad.3.5.628. [DOI] [PubMed] [Google Scholar]
  19. Edmondson D. G., Olson E. N. Helix-loop-helix proteins as regulators of muscle-specific transcription. J Biol Chem. 1993 Jan 15;268(2):755–758. [PubMed] [Google Scholar]
  20. Elford P. R., Guenther H. L., Felix R., Cecchini M. G., Fleisch H. Transforming growth factor-beta reduces the phenotypic expression of osteoblastic MC3T3-E1 cells in monolayer culture. Bone. 1987;8(4):259–262. doi: 10.1016/8756-3282(87)90174-8. [DOI] [PubMed] [Google Scholar]
  21. Florini J. R., Roberts A. B., Ewton D. Z., Falen S. L., Flanders K. C., Sporn M. B. Transforming growth factor-beta. A very potent inhibitor of myoblast differentiation, identical to the differentiation inhibitor secreted by Buffalo rat liver cells. J Biol Chem. 1986 Dec 15;261(35):16509–16513. [PubMed] [Google Scholar]
  22. Fujisawa-Sehara A., Nabeshima Y., Hosoda Y., Obinata T., Nabeshima Y. Myogenin contains two domains conserved among myogenic factors. J Biol Chem. 1990 Sep 5;265(25):15219–15223. [PubMed] [Google Scholar]
  23. Grigoriadis A. E., Heersche J. N., Aubin J. E. Continuously growing bipotential and monopotential myogenic, adipogenic, and chondrogenic subclones isolated from the multipotential RCJ 3.1 clonal cell line. Dev Biol. 1990 Dec;142(2):313–318. doi: 10.1016/0012-1606(90)90352-j. [DOI] [PubMed] [Google Scholar]
  24. Grigoriadis A. E., Heersche J. N., Aubin J. E. Differentiation of muscle, fat, cartilage, and bone from progenitor cells present in a bone-derived clonal cell population: effect of dexamethasone. J Cell Biol. 1988 Jun;106(6):2139–2151. doi: 10.1083/jcb.106.6.2139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Hara E., Yamaguchi T., Nojima H., Ide T., Campisi J., Okayama H., Oda K. Id-related genes encoding helix-loop-helix proteins are required for G1 progression and are repressed in senescent human fibroblasts. J Biol Chem. 1994 Jan 21;269(3):2139–2145. [PubMed] [Google Scholar]
  26. Hardy S., Kong Y., Konieczny S. F. Fibroblast growth factor inhibits MRF4 activity independently of the phosphorylation status of a conserved threonine residue within the DNA-binding domain. Mol Cell Biol. 1993 Oct;13(10):5943–5956. doi: 10.1128/mcb.13.10.5943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Hasty P., Bradley A., Morris J. H., Edmondson D. G., Venuti J. M., Olson E. N., Klein W. H. Muscle deficiency and neonatal death in mice with a targeted mutation in the myogenin gene. Nature. 1993 Aug 5;364(6437):501–506. doi: 10.1038/364501a0. [DOI] [PubMed] [Google Scholar]
  28. Heremans H., Billiau A., Cassiman J. J., Mulier J. C., de Somer P. In vitro cultivation of human tumor tissues. II. Morphological and virological characterization of three cell lines. Oncology. 1978;35(6):246–252. doi: 10.1159/000225298. [DOI] [PubMed] [Google Scholar]
  29. Jen Y., Weintraub H., Benezra R. Overexpression of Id protein inhibits the muscle differentiation program: in vivo association of Id with E2A proteins. Genes Dev. 1992 Aug;6(8):1466–1479. doi: 10.1101/gad.6.8.1466. [DOI] [PubMed] [Google Scholar]
  30. Joyce M. E., Roberts A. B., Sporn M. B., Bolander M. E. Transforming growth factor-beta and the initiation of chondrogenesis and osteogenesis in the rat femur. J Cell Biol. 1990 Jun;110(6):2195–2207. doi: 10.1083/jcb.110.6.2195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Jüppner H., Abou-Samra A. B., Freeman M., Kong X. F., Schipani E., Richards J., Kolakowski L. F., Jr, Hock J., Potts J. T., Jr, Kronenberg H. M. A G protein-linked receptor for parathyroid hormone and parathyroid hormone-related peptide. Science. 1991 Nov 15;254(5034):1024–1026. doi: 10.1126/science.1658941. [DOI] [PubMed] [Google Scholar]
  32. Katagiri T., Lee T., Takeshima H., Suda T., Tanaka H., Omura S. Transforming growth factor-beta modulates proliferation and differentiation of mouse clonal osteoblastic MC3T3-E1 cells depending on their maturation stages. Bone Miner. 1990 Dec;11(3):285–293. doi: 10.1016/0169-6009(90)90025-b. [DOI] [PubMed] [Google Scholar]
  33. Katagiri T., Yamaguchi A., Ikeda T., Yoshiki S., Wozney J. M., Rosen V., Wang E. A., Tanaka H., Omura S., Suda T. The non-osteogenic mouse pluripotent cell line, C3H10T1/2, is induced to differentiate into osteoblastic cells by recombinant human bone morphogenetic protein-2. Biochem Biophys Res Commun. 1990 Oct 15;172(1):295–299. doi: 10.1016/s0006-291x(05)80208-6. [DOI] [PubMed] [Google Scholar]
  34. Kawaguchi N., DeLuca H. F., Noda M. Id gene expression and its suppression by 1,25-dihydroxyvitamin D3 in rat osteoblastic osteosarcoma cells. Proc Natl Acad Sci U S A. 1992 May 15;89(10):4569–4572. doi: 10.1073/pnas.89.10.4569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Lassar A. B., Buskin J. N., Lockshon D., Davis R. L., Apone S., Hauschka S. D., Weintraub H. MyoD is a sequence-specific DNA binding protein requiring a region of myc homology to bind to the muscle creatine kinase enhancer. Cell. 1989 Sep 8;58(5):823–831. doi: 10.1016/0092-8674(89)90935-5. [DOI] [PubMed] [Google Scholar]
  36. Li L., Zhou J., James G., Heller-Harrison R., Czech M. P., Olson E. N. FGF inactivates myogenic helix-loop-helix proteins through phosphorylation of a conserved protein kinase C site in their DNA-binding domains. Cell. 1992 Dec 24;71(7):1181–1194. doi: 10.1016/s0092-8674(05)80066-2. [DOI] [PubMed] [Google Scholar]
  37. Luyten F. P., Cunningham N. S., Ma S., Muthukumaran N., Hammonds R. G., Nevins W. B., Woods W. I., Reddi A. H. Purification and partial amino acid sequence of osteogenin, a protein initiating bone differentiation. J Biol Chem. 1989 Aug 15;264(23):13377–13380. [PubMed] [Google Scholar]
  38. Majeska R. J., Rodan S. B., Rodan G. A. Parathyroid hormone-responsive clonal cell lines from rat osteosarcoma. Endocrinology. 1980 Nov;107(5):1494–1503. doi: 10.1210/endo-107-5-1494. [DOI] [PubMed] [Google Scholar]
  39. Martin J. F., Li L., Olson E. N. Repression of myogenin function by TGF-beta 1 is targeted at the basic helix-loop-helix motif and is independent of E2A products. J Biol Chem. 1992 Jun 5;267(16):10956–10960. [PubMed] [Google Scholar]
  40. Massagué J., Cheifetz S., Endo T., Nadal-Ginard B. Type beta transforming growth factor is an inhibitor of myogenic differentiation. Proc Natl Acad Sci U S A. 1986 Nov;83(21):8206–8210. doi: 10.1073/pnas.83.21.8206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. McAllister R. M., Gardner M. B., Greene A. E., Bradt C., Nichols W. W., Landing B. H. Cultivation in vitro of cells derived from a human osteosarcoma. Cancer. 1971 Feb;27(2):397–402. doi: 10.1002/1097-0142(197102)27:2<397::aid-cncr2820270224>3.0.co;2-x. [DOI] [PubMed] [Google Scholar]
  42. Miner J. H., Wold B. Herculin, a fourth member of the MyoD family of myogenic regulatory genes. Proc Natl Acad Sci U S A. 1990 Feb;87(3):1089–1093. doi: 10.1073/pnas.87.3.1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Murray S. S., Glackin C. A., Winters K. A., Gazit D., Kahn A. J., Murray E. J. Expression of helix-loop-helix regulatory genes during differentiation of mouse osteoblastic cells. J Bone Miner Res. 1992 Oct;7(10):1131–1138. doi: 10.1002/jbmr.5650071004. [DOI] [PubMed] [Google Scholar]
  44. Murray S. S., Murray E. J., Glackin C. A., Urist M. R. Bone morphogenetic protein inhibits differentiation and affects expression of helix-loop-helix regulatory molecules in myoblastic cells. J Cell Biochem. 1993 Sep;53(1):51–60. doi: 10.1002/jcb.240530107. [DOI] [PubMed] [Google Scholar]
  45. Murre C., McCaw P. S., Baltimore D. A new DNA binding and dimerization motif in immunoglobulin enhancer binding, daughterless, MyoD, and myc proteins. Cell. 1989 Mar 10;56(5):777–783. doi: 10.1016/0092-8674(89)90682-x. [DOI] [PubMed] [Google Scholar]
  46. Murre C., McCaw P. S., Vaessin H., Caudy M., Jan L. Y., Jan Y. N., Cabrera C. V., Buskin J. N., Hauschka S. D., Lassar A. B. Interactions between heterologous helix-loop-helix proteins generate complexes that bind specifically to a common DNA sequence. Cell. 1989 Aug 11;58(3):537–544. doi: 10.1016/0092-8674(89)90434-0. [DOI] [PubMed] [Google Scholar]
  47. Nabeshima Y., Hanaoka K., Hayasaka M., Esumi E., Li S., Nonaka I., Nabeshima Y. Myogenin gene disruption results in perinatal lethality because of severe muscle defect. Nature. 1993 Aug 5;364(6437):532–535. doi: 10.1038/364532a0. [DOI] [PubMed] [Google Scholar]
  48. Noda M., Camilliere J. J. In vivo stimulation of bone formation by transforming growth factor-beta. Endocrinology. 1989 Jun;124(6):2991–2994. doi: 10.1210/endo-124-6-2991. [DOI] [PubMed] [Google Scholar]
  49. Noda M., Rodan G. A. Type-beta transforming growth factor inhibits proliferation and expression of alkaline phosphatase in murine osteoblast-like cells. Biochem Biophys Res Commun. 1986 Oct 15;140(1):56–65. doi: 10.1016/0006-291x(86)91057-0. [DOI] [PubMed] [Google Scholar]
  50. Noda M. Transcriptional regulation of osteocalcin production by transforming growth factor-beta in rat osteoblast-like cells. Endocrinology. 1989 Feb;124(2):612–617. doi: 10.1210/endo-124-2-612. [DOI] [PubMed] [Google Scholar]
  51. Noda M., Yoon K., Thiede M., Buenaga R., Weiss M., Henthorn P., Harris H., Rodan G. A. cDNA cloning of alkaline phosphatase from rat osteosarcoma (ROS 17/2.8) cells. J Bone Miner Res. 1987 Apr;2(2):161–164. doi: 10.1002/jbmr.5650020212. [DOI] [PubMed] [Google Scholar]
  52. Ogata T., Wozney J. M., Benezra R., Noda M. Bone morphogenetic protein 2 transiently enhances expression of a gene, Id (inhibitor of differentiation), encoding a helix-loop-helix molecule in osteoblast-like cells. Proc Natl Acad Sci U S A. 1993 Oct 1;90(19):9219–9222. doi: 10.1073/pnas.90.19.9219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Olson E. N. MyoD family: a paradigm for development? Genes Dev. 1990 Sep;4(9):1454–1461. doi: 10.1101/gad.4.9.1454. [DOI] [PubMed] [Google Scholar]
  54. Owen M. Marrow stromal stem cells. J Cell Sci Suppl. 1988;10:63–76. doi: 10.1242/jcs.1988.supplement_10.5. [DOI] [PubMed] [Google Scholar]
  55. Ozkaynak E., Rueger D. C., Drier E. A., Corbett C., Ridge R. J., Sampath T. K., Oppermann H. OP-1 cDNA encodes an osteogenic protein in the TGF-beta family. EMBO J. 1990 Jul;9(7):2085–2093. doi: 10.1002/j.1460-2075.1990.tb07376.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Ozkaynak E., Schnegelsberg P. N., Jin D. F., Clifford G. M., Warren F. D., Drier E. A., Oppermann H. Osteogenic protein-2. A new member of the transforming growth factor-beta superfamily expressed early in embryogenesis. J Biol Chem. 1992 Dec 15;267(35):25220–25227. [PubMed] [Google Scholar]
  57. Partridge N. C., Alcorn D., Michelangeli V. P., Ryan G., Martin T. J. Morphological and biochemical characterization of four clonal osteogenic sarcoma cell lines of rat origin. Cancer Res. 1983 Sep;43(9):4308–4314. [PubMed] [Google Scholar]
  58. Rhodes S. J., Konieczny S. F. Identification of MRF4: a new member of the muscle regulatory factor gene family. Genes Dev. 1989 Dec;3(12B):2050–2061. doi: 10.1101/gad.3.12b.2050. [DOI] [PubMed] [Google Scholar]
  59. Rosen D. M., Stempien S. A., Thompson A. Y., Seyedin S. M. Transforming growth factor-beta modulates the expression of osteoblast and chondroblast phenotypes in vitro. J Cell Physiol. 1988 Mar;134(3):337–346. doi: 10.1002/jcp.1041340304. [DOI] [PubMed] [Google Scholar]
  60. Rudnicki M. A., Braun T., Hinuma S., Jaenisch R. Inactivation of MyoD in mice leads to up-regulation of the myogenic HLH gene Myf-5 and results in apparently normal muscle development. Cell. 1992 Oct 30;71(3):383–390. doi: 10.1016/0092-8674(92)90508-a. [DOI] [PubMed] [Google Scholar]
  61. Rudnicki M. A., Schnegelsberg P. N., Stead R. H., Braun T., Arnold H. H., Jaenisch R. MyoD or Myf-5 is required for the formation of skeletal muscle. Cell. 1993 Dec 31;75(7):1351–1359. doi: 10.1016/0092-8674(93)90621-v. [DOI] [PubMed] [Google Scholar]
  62. Sampath T. K., Coughlin J. E., Whetstone R. M., Banach D., Corbett C., Ridge R. J., Ozkaynak E., Oppermann H., Rueger D. C. Bovine osteogenic protein is composed of dimers of OP-1 and BMP-2A, two members of the transforming growth factor-beta superfamily. J Biol Chem. 1990 Aug 5;265(22):13198–13205. [PubMed] [Google Scholar]
  63. Sampath T. K., Muthukumaran N., Reddi A. H. Isolation of osteogenin, an extracellular matrix-associated, bone-inductive protein, by heparin affinity chromatography. Proc Natl Acad Sci U S A. 1987 Oct;84(20):7109–7113. doi: 10.1073/pnas.84.20.7109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Spizz G., Hu J. S., Olson E. N. Inhibition of myogenic differentiation by fibroblast growth factor or type beta transforming growth factor does not require persistent c-myc expression. Dev Biol. 1987 Oct;123(2):500–507. doi: 10.1016/0012-1606(87)90408-8. [DOI] [PubMed] [Google Scholar]
  65. Spizz G., Roman D., Strauss A., Olson E. N. Serum and fibroblast growth factor inhibit myogenic differentiation through a mechanism dependent on protein synthesis and independent of cell proliferation. J Biol Chem. 1986 Jul 15;261(20):9483–9488. [PubMed] [Google Scholar]
  66. Stockdale F. E. Myogenic cell lineages. Dev Biol. 1992 Dec;154(2):284–298. doi: 10.1016/0012-1606(92)90068-r. [DOI] [PubMed] [Google Scholar]
  67. Sudo H., Kodama H. A., Amagai Y., Yamamoto S., Kasai S. In vitro differentiation and calcification in a new clonal osteogenic cell line derived from newborn mouse calvaria. J Cell Biol. 1983 Jan;96(1):191–198. doi: 10.1083/jcb.96.1.191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Takuwa Y., Ohse C., Wang E. A., Wozney J. M., Yamashita K. Bone morphogenetic protein-2 stimulates alkaline phosphatase activity and collagen synthesis in cultured osteoblastic cells, MC3T3-E1. Biochem Biophys Res Commun. 1991 Jan 15;174(1):96–101. doi: 10.1016/0006-291x(91)90490-x. [DOI] [PubMed] [Google Scholar]
  69. Tapscott S. J., Davis R. L., Thayer M. J., Cheng P. F., Weintraub H., Lassar A. B. MyoD1: a nuclear phosphoprotein requiring a Myc homology region to convert fibroblasts to myoblasts. Science. 1988 Oct 21;242(4877):405–411. doi: 10.1126/science.3175662. [DOI] [PubMed] [Google Scholar]
  70. Taylor S. M., Jones P. A. Multiple new phenotypes induced in 10T1/2 and 3T3 cells treated with 5-azacytidine. Cell. 1979 Aug;17(4):771–779. doi: 10.1016/0092-8674(79)90317-9. [DOI] [PubMed] [Google Scholar]
  71. Urist M. R. Bone: formation by autoinduction. Science. 1965 Nov 12;150(3698):893–899. doi: 10.1126/science.150.3698.893. [DOI] [PubMed] [Google Scholar]
  72. Urist M. R., Strates B. S. Bone morphogenetic protein. J Dent Res. 1971 Nov-Dec;50(6):1392–1406. doi: 10.1177/00220345710500060601. [DOI] [PubMed] [Google Scholar]
  73. Vaidya T. B., Rhodes S. J., Taparowsky E. J., Konieczny S. F. Fibroblast growth factor and transforming growth factor beta repress transcription of the myogenic regulatory gene MyoD1. Mol Cell Biol. 1989 Aug;9(8):3576–3579. doi: 10.1128/mcb.9.8.3576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Wang E. A., Rosen V., Cordes P., Hewick R. M., Kriz M. J., Luxenberg D. P., Sibley B. S., Wozney J. M. Purification and characterization of other distinct bone-inducing factors. Proc Natl Acad Sci U S A. 1988 Dec;85(24):9484–9488. doi: 10.1073/pnas.85.24.9484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Wang E. A., Rosen V., D'Alessandro J. S., Bauduy M., Cordes P., Harada T., Israel D. I., Hewick R. M., Kerns K. M., LaPan P. Recombinant human bone morphogenetic protein induces bone formation. Proc Natl Acad Sci U S A. 1990 Mar;87(6):2220–2224. doi: 10.1073/pnas.87.6.2220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Weintraub H., Davis R., Tapscott S., Thayer M., Krause M., Benezra R., Blackwell T. K., Turner D., Rupp R., Hollenberg S. The myoD gene family: nodal point during specification of the muscle cell lineage. Science. 1991 Feb 15;251(4995):761–766. doi: 10.1126/science.1846704. [DOI] [PubMed] [Google Scholar]
  77. Weintraub H. The MyoD family and myogenesis: redundancy, networks, and thresholds. Cell. 1993 Dec 31;75(7):1241–1244. doi: 10.1016/0092-8674(93)90610-3. [DOI] [PubMed] [Google Scholar]
  78. Wozney J. M., Rosen V., Celeste A. J., Mitsock L. M., Whitters M. J., Kriz R. W., Hewick R. M., Wang E. A. Novel regulators of bone formation: molecular clones and activities. Science. 1988 Dec 16;242(4885):1528–1534. doi: 10.1126/science.3201241. [DOI] [PubMed] [Google Scholar]
  79. Wright W. E. Muscle basic helix-loop-helix proteins and the regulation of myogenesis. Curr Opin Genet Dev. 1992 Apr;2(2):243–248. doi: 10.1016/s0959-437x(05)80280-1. [DOI] [PubMed] [Google Scholar]
  80. Wright W. E., Sassoon D. A., Lin V. K. Myogenin, a factor regulating myogenesis, has a domain homologous to MyoD. Cell. 1989 Feb 24;56(4):607–617. doi: 10.1016/0092-8674(89)90583-7. [DOI] [PubMed] [Google Scholar]
  81. Włodarski K. H. Properties and origin of osteoblasts. Clin Orthop Relat Res. 1990 Mar;(252):276–293. [PubMed] [Google Scholar]
  82. Yaffe D., Saxel O. Serial passaging and differentiation of myogenic cells isolated from dystrophic mouse muscle. Nature. 1977 Dec 22;270(5639):725–727. doi: 10.1038/270725a0. [DOI] [PubMed] [Google Scholar]
  83. Yamaguchi A., Kahn A. J. Clonal osteogenic cell lines express myogenic and adipocytic developmental potential. Calcif Tissue Int. 1991 Sep;49(3):221–225. doi: 10.1007/BF02556122. [DOI] [PubMed] [Google Scholar]
  84. Yamaguchi A., Katagiri T., Ikeda T., Wozney J. M., Rosen V., Wang E. A., Kahn A. J., Suda T., Yoshiki S. Recombinant human bone morphogenetic protein-2 stimulates osteoblastic maturation and inhibits myogenic differentiation in vitro. J Cell Biol. 1991 May;113(3):681–687. doi: 10.1083/jcb.113.3.681. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The Journal of Cell Biology are provided here courtesy of The Rockefeller University Press

RESOURCES