Retinoic acid enhances skeletal muscle progenitor formation and bypasses inhibition by bone morphogenetic protein 4 but not dominant negative beta-catenin

Karen A M Kennedy, Tammy Porter, Virja Mehta, Scott D Ryan, Feodor Price, Vian Peshdary, Christina Karamboulas, Josée Savage, Thomas A Drysdale, Shun-Cheng Li, Steffany A L Bennett, Ilona S Skerjanc, Karen A M Kennedy, Tammy Porter, Virja Mehta, Scott D Ryan, Feodor Price, Vian Peshdary, Christina Karamboulas, Josée Savage, Thomas A Drysdale, Shun-Cheng Li, Steffany A L Bennett, Ilona S Skerjanc

Abstract

Background: Understanding stem cell differentiation is essential for the future design of cell therapies. While retinoic acid (RA) is the most potent small molecule enhancer of skeletal myogenesis in stem cells, the stage and mechanism of its function has not yet been elucidated. Further, the intersection of RA with other signalling pathways that stimulate or inhibit myogenesis (such as Wnt and BMP4, respectively) is unknown. Thus, the purpose of this study is to examine the molecular mechanisms by which RA enhances skeletal myogenesis and interacts with Wnt and BMP4 signalling during P19 or mouse embryonic stem (ES) cell differentiation.

Results: Treatment of P19 or mouse ES cells with low levels of RA led to an enhancement of skeletal myogenesis by upregulating the expression of the mesodermal marker, Wnt3a, the skeletal muscle progenitor factors Pax3 and Meox1, and the myogenic regulatory factors (MRFs) MyoD and myogenin. By chromatin immunoprecipitation, RA receptors (RARs) bound directly to regulatory regions in the Wnt3a, Pax3, and Meox1 genes and RA activated a beta-catenin-responsive promoter in aggregated P19 cells. In the presence of a dominant negative beta-catenin/engrailed repressor fusion protein, RA could not bypass the inhibition of skeletal myogenesis nor upregulate Meox1 or MyoD. Thus, RA functions both upstream and downstream of Wnt signalling. In contrast, it functions downstream of BMP4, as it abrogates BMP4 inhibition of myogenesis and Meox1, Pax3, and MyoD expression. Furthermore, RA downregulated BMP4 expression and upregulated the BMP4 inhibitor, Tob1. Finally, RA inhibited cardiomyogenesis but not in the presence of BMP4.

Conclusion: RA can enhance skeletal myogenesis in stem cells at the muscle specification/progenitor stage by activating RARs bound directly to mesoderm and skeletal muscle progenitor genes, activating beta-catenin function and inhibiting bone morphogenetic protein (BMP) signalling. Thus, a signalling pathway can function at multiple levels to positively regulate a developmental program and can function by abrogating inhibitory pathways. Finally, since RA enhances skeletal muscle progenitor formation, it will be a valuable tool for designing future stem cell therapies.

Figures

Figure 1
Figure 1
Retinoic acid inhibits cardiomyogenesis and enhances skeletal myogenesis in P19 cells. P19 cells were aggregated with 0.8% dimethylsulfoxide (DMSO) in the presence and absence of 30 nM RA. Panel I: Cells were fixed on day 9 for immunofluorescence with MF20 antibody (A-D) and counter stained with Hoechst dye (E-H). Magnification is 160x. Panel II: Cardiac (n = 3) and skeletal (n = 4) myogenesis were quantified by counting the number of MHC+ve myocytes as a percentage of the total. Average +/- standard error of mean (SEM) is shown and statistics were Student's t-test, *P < 0.05. Panel III: Total RNA was harvested for northern blot analysis on the days indicated and hybridized to the cDNAs on the right. Lanes are spliced from the same autoradiogram. Panel IV: Quantitative polymerase chain reaction (PCR) analysis was performed on day 4 of differentiation for Pax3 and Wnt3a transcript levels (n = 2) and on day 9 for MyoD levels (n = 4). Results were expressed as fold change of transcript levels in the presence compared to the absence of RA treatment. Panel V: Chromatin immunoprecipitation experiments were performed on day 2 P19 aggregates treated with DMSO/retinoic acid and analysed by real-time PCR using primers for sites within regulatory regions of the genes indicated. Average +/-SEM is shown, relative to IgG, and statistics were Student's t-test of each region compared to IgG, n = 3-4, *P < 0.05. Panel VI: The position and conservation of the Meox1-2 and the Pax3-2 retinoic acid response elements are shown.
Figure 2
Figure 2
Mouse embryonic stem (mES) cells differentiate into skeletal muscle in response to retinoic acid (RA). mES cells were aggregated in hanging drops for 2 days, cultured in suspension for a further 5 days with increasing concentrations of RA, and transferred to tissue culture dishes. Panel I: RNA was harvested from day 7 cultures and subjected to reverse transcriptase- polymerase chain reaction (PCR) followed by Southern blot analysis with the probes indicated on the left. Panel II: RNA was harvested from days 7 and 15 after differentiation with or without 25nM retinoic acid and examined by quantitative PCR analysis. The expression levels are expressed as fold increase in the presence, compared to the absence of RA, as the mean and standard error, n = 3. Statistics were Student's t-test, *P < 0.05. Panel III: On day 20 of differentiation, cells were fixed and reacted with Hoechst dye to detect nuclei (A and C) and with MF20 antibody to detect muscle (B and D). Magnification is 400×.
Figure 3
Figure 3
Retinoic acid (RA) cannot override the inhibition of skeletal myogenesis by β-Cat/EnR. P19[control] (A, B, E, F) and P19[β-Cat/EnR] (C, D, G, H) cells were aggregated in the presence of 0.8% dimethylsulfoxide (DMSO) with (E-H) and without (A-D) 10 nM RA. Cells were fixed on day 9 of differentiation for immunofluorescence with MF20 antibody (B, D, F, H) and counter stained with Hoechst dye (A, C, E, G). Magnification is 160×.
Figure 4
Figure 4
Retinoic acid (RA) enhances the expression of Pax3/7 but not MyoD or Meox1 in the presence of β-Cat/EnR. Panel I: P19[control] and P19[β-Cat/EnR] cultures were differentiated with 0.8% dimethylsulfoxide in the presence of 0, 3, and 10 nM RA. Total RNA was harvested and hybridized with the probes as indicated. Panel II: quantitative polymerase chain reaction analysis of Pax3 and Pax7 transcript levels, for each condition in Panel I, shown as one representative experiment performed in triplicate. Panel III: RA activates β-catenin in P19 aggregate but not monolayer cultures. P19 cells were transfected with the TOPFlash or FOPFlash reporter and treated with the compounds indicated in aggregated or monolayer cultures. Cells were harvested 24 hours later for luciferase assays (n = 2). Numbers represent the average +/- standard error of mean and statistics were Student's t-test, *P < 0.05.
Figure 5
Figure 5
PAX3/7 expression in P19 cultures treated with dimethylsulfoxide (DMSO) and 10 nM retinoic acid (RA) is indicative of skeletal myogenesis and not neurogenesis. Panel I: P19[control] cells, undifferentiated in monolayer cultures (control) or differentiated with 1% DMSO, 1% DMSO+ 10 nM RA, or 1% DMSO + 1 μM RA for 9 days, were immunolabelled with anti-TuJ1 (green) to detect terminally differentiated neurons and stained with the nuclear marker Hoechst (blue). Panel II: P19[β-Cat/EnR] cells were treated as in Panel I. Panel III: Quantitative analysis of the percentage of Tuj1-positive cells was established, expressed as the percentage of the total cell number (n = 6-10). Panel IV: Immunofluorescent staining of PAX3/7 protein (green), committed neuronal precursors (doublecortin-positive, red), and terminally differentiated neurons (TuJ1-positive, blue) in triple-labelled P19[control] cultures treated with DMSO + 10 nM RA, demonstrating that PAX3/7-positive cells (arrow) are not neuronal precursors or neurons. Panel V: Quantitative analysis indicated that the overwhelming majority of PAX3/7-positive cells in all treatments were non-neuronal. Statistics were analysis of variance, post-hoc Bonferroni, *P < 0.05, Scale bars, 50 μm.
Figure 6
Figure 6
BMP4 inhibits skeletal but not cardiac myogenesis. Panel I: P19[BMP4] and P19[control] cells were aggregated in the presence of 0.8% dimethylsulfoxide (DMSO). Cells were fixed on day 9, stained with MF20 antibody (A-D), and counter-stained with Hoechst dye to show the nuclei (E-H). Magnification is 400x. Panel II: The number of MHC+ve cells were counted and the average +/- standard error of mean shown. Statistics were Student's t-test, *P < 0.05, n = 3.
Figure 7
Figure 7
BMP4 inhibits skeletal muscle specification. Panels I and II: P19[BMP4] and P19[control] cells were aggregated in the presence of 0.8% dimethylsulfoxide (DMSO). P19[control] cells were also aggregated in the absence of DMSO to serve as negative controls. On days 0, 6 and 9 (Panel I) and days 1-5 (Panel II), total RNA was harvested for northern blot analysis and hybridized with the cDNAs indicated on the right. Panel III: P19 cells were transfected with the TOPFlash reporter, aggregated, and treated with the compounds indicated. Cells were harvested 24 hours later for luciferase assays (n = 2). Numbers represent the average +/- standard error of mean and statistics were Student's t-test, *P < 0.05.
Figure 8
Figure 8
Retinoic acid (RA) and BMP4 counteract each other's inhibition of skeletal myogenesis or cardiomyogenesis. P19 cells were mixed with P19[BMP4] or P19[control] cells in the presence of 1% dimethylsulfoxide (DMSO), with or without RA. Panel I: P19[BMP4] cultures treated with RA were fixed on day 9 for immunofluorescence with MF20 antibody (A, B) and counter stained with Hoechst dye (C, D). Magnification is 160x. Panel II: Skeletal myogenesis was quantified for each condition by counting the number of myosin heavy chain+ve bipolar skeletal myocytes, expressed as the percentage of total cells (white bars) and their standard errors (n = 3). MyoD transcript levels (black bars) were quantified by quantitative polymerase chain reaction and expressed relative to control cultures (n = 2), *P < 0.05. Panel III: Cardiomyogenesis was quantified as described for Panel II, n = 4. Panel IV: On days 6 and 9 total RNA was harvested for northern blot analysis and probed with the cDNAs indicated on the right. Panel V: A time course of P19[BMP4] and P19[control] cells aggregated in the presence of DMSO and RA. Total RNA was harvested for northern blot analysis on days 1-4, 6, and 9 and probed with the cDNAs indicated on the right.
Figure 9
Figure 9
Model of the intersection of retinoic acid (RA), Wnt, and BMP4 signalling during cardiac and skeletal muscle development. BMP4 upregulates Wnt/β-catenin during mesoderm induction (green arrow) and blocks skeletal myogenesis by downregulation of Meox1, Pax3 and myogenic regulatory factor expression (red inhibition arrow). This inhibition can be reversed by RA, which enhances Tob1, Wnt3a, Pax3 and Meox1 expression, activates β-catenin and inhibits BMP4 expression (green arrows). RA receptors bind directly to the Wnt3a, Meox1, and Pax3 regulatory regions (bold green arrows). RA inhibits GATA-4 expression and cardiomyogenesis, likely by inhibiting BMP4 expression and function. Grey arrows indicate previous work [references [6,8-10,35,38,40,44,102,103].

References

    1. Borycki AG, Brunk B, Tajbakhsh S, Buckingham M, Chiang C, Emerson CP., Jr Sonic hedgehog controls epaxial muscle determination through Myf5 activation. Development. 1999;126:4053–4063.
    1. Munsterberg AE, Kitajewski J, Bumcrot DA, McMahon AP, Lassar AB. Combinatorial signalling by sonic hedgehog and Wnt family members induces myogenic bHLH gene expression in the somite. Genes Dev. 1995;9:2911–2922. doi: 10.1101/gad.9.23.2911.
    1. Pourquie O, Fan CM, Coltey M, Hirsinger E, Watanabe Y, Breant C, Francis-West P, Brickell P, Tessier-Lavigne M, Le Douarin NM. Lateral and axial signals involved in avian somite patterning: a role for BMP4. Cell. 1996;84:461–471. doi: 10.1016/S0092-8674(00)81291-X.
    1. Tajbakhsh S, Borello U, Vivarelli E, Kelly R, Papkoff J, Duprez D, Buckingham M, Cossu G. Differential activation of Myf5 and MyoD by different Wnts in explants of mouse paraxial mesoderm and the later activation of myogenesis in the absence of Myf5. Development. 1998;125:4155–4162.
    1. Dietrich S, Schubert FR, Healy C, Sharpe PT, Lumsden A. Specification of the hypaxial musculature. Development. 1998;125:2235–2249.
    1. Williams BA, Ordahl CP. Pax-3 expression in segmental mesoderm marks early stages in myogenic cell specification. Development. 1994;120:785–796.
    1. McDermott A, Gustafsson M, Elsam T, Hui CC, Emerson CP, Jr, Borycki AG. Gli2 and Gli3 have redundant and context-dependent function in skeletal muscle formation. Development. 2005;132:345–357. doi: 10.1242/dev.01537.
    1. Petropoulos H, Skerjanc IS. Beta-catenin is essential and sufficient for skeletal myogenesis in P19 cells. J Biol Chem. 2002;277:15393–15399. doi: 10.1074/jbc.M112141200.
    1. Petropoulos H, Gianakopoulos PJ, Ridgeway AG, Skerjanc IS. Disruption of Meox or Gli activity ablates skeletal myogenesis in P19 cells. J Biol Chem. 2004;279:23874–23881. doi: 10.1074/jbc.M312612200.
    1. Ridgeway AG, Skerjanc IS. Pax3 is essential for skeletal myogenesis and the expression of Six1 and Eya2. J Biol Chem. 2001;276:19033–19039. doi: 10.1074/jbc.M011491200.
    1. Chanoine C, Della Gaspera B, Charbonnier F. Myogenic regulatory factors: redundant or specific functions? Lessons from Xenopus. Dev Dyn. 2004;231:662–670. doi: 10.1002/dvdy.20174.
    1. Tapscott SJ. The circuitry of a master switch: myod and the regulation of skeletal muscle gene transcription. Development. 2005;132:2685–2695. doi: 10.1242/dev.01874.
    1. Skerjanc IS. Cardiac and skeletal muscle development in P19 embryonal carcinoma cells. Trends Cardiovasc Med. 1999;9:139–143. doi: 10.1016/S1050-1738(99)00017-1.
    1. Vidricaire G, Jardine K, McBurney MW. Expression of the Brachyury gene during mesoderm development in differentiating embryonal carcinoma cell cultures. Development. 1994;120:115–122.
    1. Rune Blomhoff HKB. Overview of retinoid metabolism and function. Journal of Neurobiology. 2006;66:606–630. doi: 10.1002/neu.20242.
    1. Edwards MK, McBurney MW. The concentration of retinoic acid determines the differentiated cell types formed by a teratocarcinoma cell line. Dev Biol. 1983;98:187–191. doi: 10.1016/0012-1606(83)90348-2.
    1. Halevy O, Lerman O. Retinoic acid induces adult muscle cell differentiation mediated by the retinoic acid receptor-alpha. J Cell Physiol. 1993;154:566–572. doi: 10.1002/jcp.1041540315.
    1. Albagli-Curiel O, Carnac G, Vandromme M, Vincent S, Crepieux P, Bonnieu A. Serum-induced inhibition of myogenesis is differentially relieved by retinoic acid and triiodothyronine in C2 murine muscle cells. Differentiation. 1993;52:201–210. doi: 10.1111/j.1432-0436.1993.tb00632.x.
    1. Momoi T, Miyagawa-Tomita S, Nakamura S, Kimura I, Momoi M. Retinoic acid ambivalently regulates the expression of MyoD1 in the myogenic cells in the limb buds of the early developmental stages. Biochem Biophys Res Commun. 1992;187:245–253. doi: 10.1016/S0006-291X(05)81484-6.
    1. Froeschle A, Alric S, Kitzmann M, Carnac G, Aurade F, Rochette-Egly C, Bonnieu A. Retinoic acid receptors and muscle b-HLH proteins: partners in retinoid- induced myogenesis. Oncogene. 1998;16:3369–3378. doi: 10.1038/sj.onc.1201894.
    1. Kessel M, Gruss P. Homeotic transformations of murine vertebrae and concomitant alteration of Hox codes induced by retinoic acid. Cell. 1991;67:89–104. doi: 10.1016/0092-8674(91)90574-I.
    1. Moreno TA, Kintner C. Regulation of segmental patterning by retinoic acid signalling during Xenopus somitogenesis. Dev Cell. 2004;6:205–218. doi: 10.1016/S1534-5807(04)00026-7.
    1. Vermot J, Pourquie O. Retinoic acid coordinates somitogenesis and left-right patterning in vertebrate embryos. Nature. 2005;435:215–220. doi: 10.1038/nature03488.
    1. Gordon MD, Nusse R. Wnt signalling: multiple pathways, multiple receptors, and multiple transcription factors. J Biol Chem. 2006;281:22429–22433. doi: 10.1074/jbc.R600015200.
    1. Brand-Saberi B. Genetic and epigenetic control of skeletal muscle development. Ann Anat. 2005;187:199–207. doi: 10.1016/j.aanat.2004.12.018.
    1. Schmidt M, Tanaka M, Munsterberg A. Expression of (beta)-catenin in the developing chick myotome is regulated by myogenic signals. Development. 2000;127:4105–4113.
    1. Capdevila J, Tabin C, Johnson RL. Control of dorsoventral somite patterning by Wnt-1 and beta-catenin. Dev Biol. 1998;193:182–194. doi: 10.1006/dbio.1997.8806.
    1. Borello U, Berarducci B, Murphy P, Bajard L, Buffa V, Piccolo S, Buckingham M, Cossu G. The Wnt/{beta}-catenin pathway regulates Gli-mediated Myf5 expression during somitogenesis. Development. 2006;133:3723–3732. doi: 10.1242/dev.02517.
    1. Massague J. Receptors for the TGF-beta family. Cell. 1992;69:1067–1070. doi: 10.1016/0092-8674(92)90627-O.
    1. Duprez DM, Coltey M, Amthor H, Brickell PM, Tickle C. Bone morphogenetic protein-2 (BMP-2) inhibits muscle development and promotes cartilage formation in chick limb bud cultures. Dev Biol. 1996;174:448–452. doi: 10.1006/dbio.1996.0087.
    1. Murray SS, Murray EJ, Glackin CA, Urist MR. Bone morphogenetic protein inhibits differentiation and affects expression of helix-loop-helix regulatory molecules in myoblastic cells. J Cell Biochem. 1993;53:51–60. doi: 10.1002/jcb.240530107.
    1. Marcelle C, Stark MR, Bronnerfraser M. Coordinate actions of Bmps, Wnts, Shh and noggin mediate patterning of the dorsal somite. Development. 1997;124:3955–3963.
    1. Streit A, Stern CD. Mesoderm patterning and somite formation during node regression: differential effects of chordin and noggin. Mech Dev. 1999;85:85–96. doi: 10.1016/S0925-4773(99)00085-4.
    1. McMahon JA, Takada S, Zimmerman LB, Fan CM, Harland RM, McMahon AP. Noggin-mediated antagonism of BMP signalling is required for growth and patterning of the neural tube and somite. Genes Dev. 1998;12:1438–1452. doi: 10.1101/gad.12.10.1438.
    1. Reshef R, Maroto M, Lassar AB. Regulation of dorsal somitic cell fates: BMPs and noggin control the timing and pattern of myogenic regulator expression. Genes Dev. 1998;12:290–303. doi: 10.1101/gad.12.3.290.
    1. Frasch M. Induction of visceral and cardiac mesoderm by ectodermal Dpp in the early Drosophila embryo. Nature. 1995;374:464–467. doi: 10.1038/374464a0.
    1. Schlange T, Andree B, Arnold HH, Brand T. BMP2 is required for early heart development during a distinct time period. Mech Dev. 2000;91:259–270. doi: 10.1016/S0925-4773(99)00311-1.
    1. Schultheiss TM, Burch JB, Lassar AB. A role for bone morphogenetic proteins in the induction of cardiac myogenesis. Genes Dev. 1997;11:451–462. doi: 10.1101/gad.11.4.451.
    1. Ladd AN, Yatskievych TA, Antin PB. Regulation of avian cardiac myogenesis by activin/TGFbeta and bone morphogenetic proteins. Dev Biol. 1998;204:407–419. doi: 10.1006/dbio.1998.9094.
    1. Monzen K, Shiojima I, Hiroi Y, Kudoh S, Oka T, Takimoto E, Hayashi D, Hosoda T, Habara-Ohkubo A, Nakaoka T, et al. Bone Morphogenetic Proteins Induce Cardiomyocyte differentiation through the mitogen-activated protein kinase kinase kinase TAK1 and cardiac transcription factors Csx/Nkx-2.5 and GATA-4. Mol Cell Biol. 1999;19:7096–7105.
    1. Schultheiss TM, Lassar AB. Induction of chick cardiac myogenesis by bone morphogenetic proteins. Cold Spring Harb Symp Quant Biol. 1997;62:413–419.
    1. Shi Y, Katsev S, Cai C, Evans S. BMP signalling is required for heart formation in vertebrates. Dev Biol. 2000;224:226–237. doi: 10.1006/dbio.2000.9802.
    1. Walters MJ, Wayman GA, Christian JL. Bone morphogenetic protein function is required for terminal differentiation of the heart but not for early expression of cardiac marker genes. Mech Dev. 2001;100:263–273. doi: 10.1016/S0925-4773(00)00535-9.
    1. Jamali M, Karamboulas C, Rogerson PJ, Skerjanc IS. BMP signalling regulates Nkx2-5 activity during cardiomyogenesis. FEBS Lett. 2001;509:126–130. doi: 10.1016/S0014-5793(01)03151-9.
    1. Behfar A, Zingman LV, Hodgson DM, Rauzier JM, Kane GC, Terzic A, Puceat M. Stem cell differentiation requires a paracrine pathway in the heart. Faseb J. 2002;16:1558–1566. doi: 10.1096/fj.02-0072com.
    1. Wobus AM, Kaomei G, Shan J, Wellner MC, Rohwedel J, Ji G, Fleischmann B, Katus HA, Hescheler J, Franz WM. Retinoic acid accelerates embryonic stem cell-derived cardiac differentiation and enhances development of ventricular cardiomyocytes. J Mol Cell Cardiol. 1997;29:1525–1539. doi: 10.1006/jmcc.1997.0433.
    1. Skerjanc IS, McBurney MW. The E box is essential for activity of the cardiac actin promoter in skeletal but not in cardiac muscle. Dev Biol. 1994;163:125–132. doi: 10.1006/dbio.1994.1128.
    1. Hidaka K, Lee JK, Kim HS, Ihm CH, Iio A, Ogawa M, Nishikawa S, Kodama I, Morisaki T. Chamber-specific differentiation of Nkx2.5-positive cardiac precursor cells from murine embryonic stem cells. Faseb J. 2003;17:740–742.
    1. Drysdale TA, Patterson KD, Saha M, Krieg PA. Retinoic acid can block differentiation of the myocardium after heart specification. Dev Biol. 1997;188:205–215. doi: 10.1006/dbio.1997.8623.
    1. Stainier DY, Fishman MC. Patterning the zebrafish heart tube: acquisition of anteroposterior polarity. Dev Biol. 1992;153:91–101. doi: 10.1016/0012-1606(92)90094-W.
    1. Osmond MK, Butler AJ, Voon FC, Bellairs R. The effects of retinoic acid on heart formation in the early chick embryo. Development. 1991;113:1405–1417.
    1. Yutzey KE, Rhee JT, Bader D. Expression of the atrial-specific myosin heavy chain AMHC1 and the establishment of anteroposterior polarity in the developing chicken heart. Development. 1994;120:871–883.
    1. Keegan BR, Feldman JL, Begemann G, Ingham PW, Yelon D. Retinoic acid signalling restricts the cardiac progenitor pool. Science. 2005;307:247–249. doi: 10.1126/science.1101573.
    1. Kastner P, Grondona JM, Mark M, Gansmuller A, LeMeur M, Decimo D, Vonesch JL, Dolle P, Chambon P. Genetic analysis of RXR alpha developmental function: convergence of RXR and RAR signalling pathways in heart and eye morphogenesis. Cell. 1994;78:987–1003. doi: 10.1016/0092-8674(94)90274-7.
    1. Kastner P, Messaddeq N, Mark M, Wendling O, Grondona JM, Ward S, Ghyselinck N, Chambon P. Vitamin A deficiency and mutations of RXRalpha, RXRbeta and RARalpha lead to early differentiation of embryonic ventricular cardiomyocytes. Development. 1997;124:4749–4758.
    1. Ryckebusch L, Wang Z, Bertrand N, Lin SC, Chi X, Schwartz R, Zaffran S, Niederreither K. Retinoic acid deficiency alters second heart field formation. Proc Natl Acad Sci USA. 2008;105:2913–2918. doi: 10.1073/pnas.0712344105.
    1. Sirbu IO, Zhao X, Duester G. Retinoic acid controls heart anteroposterior patterning by down-regulating Isl1 through the Fgf8 pathway. Dev Dyn. 2008;237:1627–1635. doi: 10.1002/dvdy.21570.
    1. Wobus AM, Rohwedel J, Maltsev V, Hescheler J. In vitro differentiation of embryonic stem cells into cardiomyocytes or skeletal muscle cells is specifically modulated by retinoic acid. Roux's Arch Dev Biol. 1994;204:36–45. doi: 10.1007/BF00744871.
    1. Moss JB, Xavier-Neto J, Shapiro MD, Nayeem SM, McCaffery P, Drager UC, Rosenthal N. Dynamic patterns of retinoic acid synthesis and response in the developing mammalian heart. Dev Biol. 1998;199:55–71. doi: 10.1006/dbio.1998.8911.
    1. Grepin C, Dagnino L, Robitaille L, Haberstroh L, Antakly T, Nemer M. A hormone-encoding gene identifies a pathway for cardiac but not skeletal muscle gene transcription. Mol Cell Biol. 1994;14:3115–3129.
    1. Miller JB, Schaefer L, Dominov JA. Seeking muscle stem cells. Curr Top Dev Biol. 1999;43:191–219. full_text.
    1. Mic FA, Duester G. Patterning of forelimb bud myogenic precursor cells requires retinoic acid signalling initiated by Raldh2. Dev Biol. 2003;264:191–201. doi: 10.1016/S0012-1606(03)00403-2.
    1. Yoshida Y, von Bubnoff A, Ikematsu N, Blitz IL, Tsuzuku JK, Yoshida EH, Umemori H, Miyazono K, Yamamoto T, Cho KW. Tob proteins enhance inhibitory Smad-receptor interactions to repress BMP signalling. Mech Dev. 2003;120:629–637. doi: 10.1016/S0925-4773(03)00020-0.
    1. Matsuda S, Kawamura-Tsuzuku J, Ohsugi M, Yoshida M, Emi M, Nakamura Y, Onda M, Yoshida Y, Nishiyama A, Yamamoto T. Tob, a novel protein that interacts with p185erbB2, is associated with anti-proliferative activity. Oncogene. 1996;12:705–713.
    1. Gillespie RF, Gudas LJ. Retinoic acid receptor isotype specificity in F9 teratocarcinoma stem cells results from the differential recruitment of coregulators to retinoic response elements. J Biol Chem. 2007;282:33421–33434. doi: 10.1074/jbc.M704845200.
    1. DasGupta R, Kaykas A, Moon RT, Perrimon N. Functional genomic analysis of the Wnt-wingless signalling pathway. Science. 2005;308:826–833. doi: 10.1126/science.1109374.
    1. Easwaran V, Pishvaian M, Salimuddin , Byers S. Cross-regulation of beta-catenin-LEF/TCF and retinoid signalling pathways. Curr Biol. 1999;9:1415–1418. doi: 10.1016/S0960-9822(00)80088-3.
    1. Baker CV, Bronner-Fraser M. Establishing neuronal identity in vertebrate neurogenic placodes. Development. 2000;127:3045–3056.
    1. Koblar SA, Murphy M, Barrett GL, Underhill A, Gros P, Bartlett PF. Pax-3 regulates neurogenesis in neural crest-derived precursor cells. J Neurosci Res. 1999;56:518–530. doi: 10.1002/(SICI)1097-4547(19990601)56:5<518::AID-JNR7>;2-8.
    1. Pozniak CD, Pleasure SJ. A tale of two signals: Wnt and hedgehog in dentate neurogenesis. Sci STKE. 2006;2006:pe5. doi: 10.1126/stke.3192006pe5.
    1. Lie DC, Colamarino SA, Song HJ, Desire L, Mira H, Consiglio A, Lein ES, Jessberger S, Lansford H, Dearie AR, et al. Wnt signalling regulates adult hippocampal neurogenesis. Nature. 2005;437:1370–1375. doi: 10.1038/nature04108.
    1. Bogoch Y, Linial M. Coordinated expression of cytoskeleton regulating genes in the accelerated neurite outgrowth of P19 embryonic carcinoma cells. Exp Cell Res. 2008;314:677–690. doi: 10.1016/j.yexcr.2007.12.003.
    1. Halilagic A, Ribes V, Ghyselinck NB, Zile MH, Dolle P, Studer M. Retinoids control anterior and dorsal properties in the developing forebrain. Developmental Biology. 2007;303:362–375. doi: 10.1016/j.ydbio.2006.11.021.
    1. Verani R, Cappuccio I, Spinsanti P, Gradini R, Caruso A, Magnotti MC, Motolese M, Nicoletti F, Melchiorri D. Expression of the Wnt inhibitor Dickkopf-1 is required for the induction of neural markers in mouse embryonic stem cells differentiating in response to retinoic acid. Journal of Neurochemistry. 2007;100:242–250. doi: 10.1111/j.1471-4159.2006.04207.x.
    1. zur Nieden NI, Price FD, Davis LA, Everitt RE, Rancourt DE. Gene profiling on mixed embryonic stem cell populations reveals a biphasic role for {beta}-catenin in osteogenic differentiation. doi:10.1210/me.2005-0438. Mol Endocrinol. 2007;21:674–685. doi: 10.1210/me.2005-0438.
    1. Jacobs S, Lie DC, DeCicco KL, Shi Y, DeLuca LM, Gage FH, Evans RM. Retinoic acid is required early during adult neurogenesis in the dentate gyrus. Proc Natl Acad Sci USA. 2006;103:3902–3907. doi: 10.1073/pnas.0511294103.
    1. Mercader N, Fischer S, Neumann CJ. Prdm1 acts downstream of a sequential RA, Wnt and Fgf signalling cascade during zebrafish forelimb induction 10.1242/dev.02455. Development. 2006;133:2805–2815. doi: 10.1242/dev.02455.
    1. Tice DA, Szeto W, Soloviev I, Rubinfeld B, Fong SE, Dugger DL, Winer J, Williams PM, Wieand D, Smith V, et al. Synergistic induction of tumor antigens by Wnt-1 signalling and retinoic acid revealed by gene expression profiling. J Biol Chem. 2002;277:14329–14335. doi: 10.1074/jbc.M200334200.
    1. Guo X, Wang XF. Signalling cross-talk between TGF-beta/BMP and other pathways. Cell Res. 2009;19:71–88. doi: 10.1038/cr.2008.302.
    1. Relaix F. Skeletal muscle progenitor cells: from embryo to adult. Cell Mol Life Sci. 2006;63:1221–1225. doi: 10.1007/s00018-006-6015-9.
    1. Katagiri T, Yamaguchi A, Komaki M, Abe E, Takahashi N, Ikeda T, Rosen V, Wozney JM, Fujisawa-Sehara A, Suda T. Bone morphogenetic protein-2 converts the differentiation pathway of C2C12 myoblasts into the osteoblast lineage. J Cell Biol. 1994;127:1755–1766. doi: 10.1083/jcb.127.6.1755.
    1. Collop AH, Broomfield JA, Chandraratna RA, Yong Z, Deimling SJ, Kolker SJ, Weeks DL, Drysdale TA. Retinoic acid signalling is essential for formation of the heart tube in Xenopus. Dev Biol. 2006;291:96–109. doi: 10.1016/j.ydbio.2005.12.018.
    1. van Wijk B, Moorman AFM, Hoff MJB van den. Role of bone morphogenetic proteins in cardiac differentiation. Cardiovascular Research. 2007;74:244–255. doi: 10.1016/j.cardiores.2006.11.022.
    1. Angello JC, Kaestner S, Welikson RE, Buskin JN, Hauschka SD. BMP induction of cardiogenesis in P19 cells requires prior cell-cell interaction(s) Dev Dyn. 2006;235:2122–33. doi: 10.1002/dvdy.20863.
    1. Gianakopoulos PJ, Skerjanc IS. Hedgehog signalling induces cardiomyogenesis in P19 cells. J Biol Chem. 2005;280:21022–21028. doi: 10.1074/jbc.M502977200.
    1. Shimo T, Koyama E, Sugito H, Wu C, Shimo S, Pacifici M. Retinoid signalling regulates CTGF expression in hypertrophic chondrocytes with differential involvement of MAP kinases. J Bone Miner Res. 2005;20:867–877. doi: 10.1359/JBMR.041235.
    1. Drissi MH, Li X, Sheu TJ, Zuscik MJ, Schwarz EM, Puzas JE, Rosier RN, O'Keefe RJ. Runx2/Cbfa1 stimulation by retinoic acid is potentiated by BMP2 signalling through interaction with Smad1 on the collagen X promoter in chondrocytes. J Cell Biochem. 2003;90:1287–1298. doi: 10.1002/jcb.10677.
    1. Li X, Schwarz EM, Zuscik MJ, Rosier RN, Ionescu AM, Puzas JE, Drissi H, Sheu TJ, O'Keefe RJ. Retinoic acid stimulates chondrocyte differentiation and enhances bone morphogenetic protein effects through induction of Smad1 and Smad5. Endocrinology. 2003;144:2514–2523. doi: 10.1210/en.2002-220969.
    1. Qin P, Haberbusch JM, Zhang Z, Soprano KJ, Soprano DR. Pre-B cell leukemia transcription factor (PBX) proteins are important mediators for retinoic acid-dependent endodermal and neuronal differentiation of mouse embryonal carcinoma P19 cells. J Biol Chem. 2004;279:16263–16271. doi: 10.1074/jbc.M313938200.
    1. Hallahan AR, Pritchard JI, Chandraratna RA, Ellenbogen RG, Geyer JR, Overland RP, Strand AD, Tapscott SJ, Olson JM. BMP-2 mediates retinoid-induced apoptosis in medulloblastoma cells through a paracrine effect. Nat Med. 2003;9:1033–1038. doi: 10.1038/nm904.
    1. Glozak MA, Rogers MB. Specific induction of apoptosis in P19 embryonal carcinoma cells by retinoic acid and Bmp2 or Bmp4. Developmental Biology. 1996;179:458–470. doi: 10.1006/dbio.1996.0275.
    1. Skerjanc IS, Petropoulos H, Ridgeway AG, Wilton S. Myocyte enhancer factor 2C and Nkx2-5 up-regulate each other's expression and initiate cardiomyogenesis in P19 cells. J Biol Chem. 1998;273:34904–34910. doi: 10.1074/jbc.273.52.34904.
    1. Rudnicki MA, McBurney MW. Cell culture methods and induction of differentiation of embryonal carcinoma cell lines. In: Robertson EJ, editor. Teratocarcinomas and embryonic stem cells A practical approach. Oxford: IRL Press; 1987. pp. 19–49.
    1. Ridgeway AG, Petropoulos H, Siu A, Ball JK, Skerjanc IS. Cloning, tissue distribution, subcellular localization and overexpression of murine histidine-rich Ca2+ binding protein. FEBS Lett. 1999;456:399–402. doi: 10.1016/S0014-5793(99)00993-X.
    1. Karamboulas C, Dakubo GD, Liu J, De Repentigny Y, Yutzey K, Wallace VA, Kothary R, Skerjanc IS. Disruption of MEF2 activity in cardiomyoblasts inhibits cardiomyogenesis. J Cell Sci. 2006;119:4315–4321. doi: 10.1242/jcs.03186.
    1. Bader D, Masaki T, Fischman DA. Immunochemical analysis of myosin heavy chain during avian myogenesis in vivo and in vitro. J Cell Biol. 1982;95:763–770. doi: 10.1083/jcb.95.3.763.
    1. Auffray C, Rougeon F. Purification of mouse immunoglobulin heavy-chain messenger RNAs from total myeloma tumor RNA. Eur J Biochem. 1980;107:303–314.
    1. Ridgeway AG, Wilton S, Skerjanc IS. Myocyte enhancer factor 2C and myogenin Up-regulate each other's expression and induce the development of skeletal muscle in P19 cells. J Biol Chem. 2000;275:41–46. doi: 10.1074/jbc.275.1.41.
    1. Karamboulas C, Swedani A, Ward C, Al-Madhoun AS, Wilton S, Boisvenue S, Ridgeway AG, Skerjanc IS. HDAC activity regulates entry of mesoderm cells into the cardiac muscle lineage. J Cell Sci. 2006;119:4305–4314. doi: 10.1242/jcs.03185.
    1. Savage J, Conley AJ, Blais A, Skerjanc IS. SOX15 and SOX7 differentially regulate the myogenic program in P19 cells. Stem Cells. 2009;27:1231–1243. doi: 10.1002/stem.57.
    1. Veeman MT, Slusarski DC, Kaykas A, Louie SH, Moon RT. Zebrafish prickle, a modulator of noncanonical Wnt/Fz signalling, regulates gastrulation movements. Curr Biol. 2003;13:680–685. doi: 10.1016/S0960-9822(03)00240-9.
    1. Grepin C, Nemer G, Nemer M. Enhanced cardiogenesis in embryonic stem cells overexpressing the GATA-4 transcription factor. Development. 1997;124:2387–2395.
    1. Mankoo BS, Skuntz S, Harrigan I, Grigorieva E, Candia A, Wright CV, Arnheiter H, Pachnis V. The concerted action of Meox homeobox genes is required upstream of genetic pathways essential for the formation, patterning and differentiation of somites. Development. 2003;130:4655–4664. doi: 10.1242/dev.00687.

Source: PubMed

3
Abonnieren