#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

A Novel Role for the RNA–Binding Protein FXR1P in Myoblasts Cell-Cycle Progression by Modulating mRNA Stability


The Fragile X-Related 1 gene (FXR1) is a paralog of the Fragile X Mental Retardation 1 gene (FMR1), whose absence causes the Fragile X syndrome, the most common form of inherited intellectual disability. FXR1P plays an important role in normal muscle development, and its absence causes muscular abnormalities in mice, frog, and zebrafish. Seven alternatively spliced FXR1 transcripts have been identified and two of them are skeletal muscle-specific. A reduction of these isoforms is found in myoblasts from Facio-Scapulo Humeral Dystrophy (FSHD) patients. FXR1P is an RNA–binding protein involved in translational control; however, so far, no mRNA target of FXR1P has been linked to the drastic muscular phenotypes caused by its absence. In this study, gene expression profiling of C2C12 myoblasts reveals that transcripts involved in cell cycle and muscular development pathways are modulated by Fxr1-depletion. We observed an increase of p21—a regulator of cell-cycle progression—in Fxr1-knocked-down mouse C2C12 and FSHD human myoblasts. Rescue of this molecular phenotype is possible by re-expressing human FXR1P in Fxr1-depleted C2C12 cells. FXR1P muscle-specific isoforms bind p21 mRNA via direct interaction with a conserved G-quadruplex located in its 3′ untranslated region. The FXR1P/G-quadruplex complex reduces the half-life of p21 mRNA. In the absence of FXR1P, the upregulation of p21 mRNA determines the elevated level of its protein product that affects cell-cycle progression inducing a premature cell-cycle exit and generating a pool of cells blocked at G0. Our study describes a novel role of FXR1P that has crucial implications for the understanding of its role during myogenesis and muscle development, since we show here that in its absence a reduced number of myoblasts will be available for muscle formation/regeneration, shedding new light into the pathophysiology of FSHD.


Vyšlo v časopise: A Novel Role for the RNA–Binding Protein FXR1P in Myoblasts Cell-Cycle Progression by Modulating mRNA Stability. PLoS Genet 9(3): e32767. doi:10.1371/journal.pgen.1003367
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003367

Souhrn

The Fragile X-Related 1 gene (FXR1) is a paralog of the Fragile X Mental Retardation 1 gene (FMR1), whose absence causes the Fragile X syndrome, the most common form of inherited intellectual disability. FXR1P plays an important role in normal muscle development, and its absence causes muscular abnormalities in mice, frog, and zebrafish. Seven alternatively spliced FXR1 transcripts have been identified and two of them are skeletal muscle-specific. A reduction of these isoforms is found in myoblasts from Facio-Scapulo Humeral Dystrophy (FSHD) patients. FXR1P is an RNA–binding protein involved in translational control; however, so far, no mRNA target of FXR1P has been linked to the drastic muscular phenotypes caused by its absence. In this study, gene expression profiling of C2C12 myoblasts reveals that transcripts involved in cell cycle and muscular development pathways are modulated by Fxr1-depletion. We observed an increase of p21—a regulator of cell-cycle progression—in Fxr1-knocked-down mouse C2C12 and FSHD human myoblasts. Rescue of this molecular phenotype is possible by re-expressing human FXR1P in Fxr1-depleted C2C12 cells. FXR1P muscle-specific isoforms bind p21 mRNA via direct interaction with a conserved G-quadruplex located in its 3′ untranslated region. The FXR1P/G-quadruplex complex reduces the half-life of p21 mRNA. In the absence of FXR1P, the upregulation of p21 mRNA determines the elevated level of its protein product that affects cell-cycle progression inducing a premature cell-cycle exit and generating a pool of cells blocked at G0. Our study describes a novel role of FXR1P that has crucial implications for the understanding of its role during myogenesis and muscle development, since we show here that in its absence a reduced number of myoblasts will be available for muscle formation/regeneration, shedding new light into the pathophysiology of FSHD.


Zdroje

1. KhandjianEW (1999) Biology of the fragile X mental retardation protein, an RNA-binding protein. Biochem Cell Biol 77: 331–342.

2. SutherlandGR (1977) Fragile sites on human chromosomes: demonstration of their dependence on the type of tissue culture medium. Science 197: 265–266.

3. CoyJF, SedlacekZ, BachnerD, HameisterH, JoosS, et al. (1995) Highly conserved 3′ UTR and expression pattern of FXR1 points to a divergent gene regulation of FXR1 and FMR1. Hum Mol Genet 4: 2209–2218.

4. ZhangY, O'ConnorJP, SiomiMC, SrinivasanS, DutraA, et al. (1995) The fragile X mental retardation syndrome protein interacts with novel homologs FXR1 and FXR2. Embo J 14: 5358–5366.

5. PenagarikanoO, MulleJG, WarrenST (2007) The pathophysiology of fragile x syndrome. Annu Rev Genomics Hum Genet 8: 109–129.

6. KirkpatrickLL, McIlwainKA, NelsonDL (1999) Alternative splicing in the murine and human FXR1 genes. Genomics 59: 193–202.

7. DubéM, HuotME, KhandjianEW (2000) Muscle specific fragile X related protein 1 isoforms are sequestered in the nucleus of undifferentiated myoblast. BMC Genet 1: 4.

8. KhandjianEW, BardoniB, CorbinF, SittlerA, GirouxS, et al. (1998) Novel isoforms of the fragile X related protein FXR1P are expressed during myogenesis. Hum Mol Genet 7: 2121–2128.

9. DavidovicL, SacconiS, BecharaEG, DelplaceS, AllegraM, et al. (2008) Alteration of expression of muscle specific isoforms of the fragile X related protein 1 (FXR1P) in facioscapulohumeral muscular dystrophy patients. J Med Genet 45: 679–685.

10. DavidovicL, HuotME, KhandjianEW (2005) Lost once, the Fragile X Mental Retardation protein is now back onto brain polyribosomes. RNA Biol 2: 1–3.

11. BakkerCE, de Diego OteroY, BontekoeC, RaghoeP, LuteijnT, et al. (2000) Immunocytochemical and biochemical characterization of FMRP, FXR1P, and FXR2P in the mouse. Exp Cell Res 258: 162–170.

12. OrengoJP, ChambonP, MetzgerD, MosierDR, SnipesGJ, et al. (2008) Expanded CTG repeats within the DMPK 3′ UTR causes severe skeletal muscle wasting in an inducible mouse model for myotonic dystrophy. Proc Natl Acad Sci U S A 105: 2646–2651.

13. MientjesEJ, WillemsenR, KirkpatrickLL, NieuwenhuizenIM, Hoogeveen-WesterveldM, et al. (2004) Fxr1 knockout mice show a striated muscle phenotype: implications for Fxr1p function in vivo. Hum Mol Genet 13: 1291–1302.

14. HuotME, BissonN, DavidovicL, MazrouiR, LabelleY, et al. (2005) The RNA-binding protein fragile X-related 1 regulates somite formation in Xenopus laevis. Mol Biol Cell 16: 4350–4361.

15. Van't PadjeS, ChaudhryB, SeverijnenLA, van der LindeHC, MientjesEJ, et al. (2009) Reduction in fragile X related 1 protein causes cardiomyopathy and muscular dystrophy in zebrafish. J Exp Biol 212: 2564–2570.

16. SiomiH, SiomiMC, NussbaumRL, DreyfussG (1993) The protein product of the fragile X gene, FMR1, has characteristics of an RNA-binding protein. Cell 74: 291–298.

17. TamaniniF, BontekoeC, BakkerCE, van UnenL, AnarB, et al. (1999) Different targets for the fragile X-related proteins revealed by their distinct nuclear localizations. Hum Mol Genet 8: 863–869.

18. KhandjianEW, BecharaE, DavidovicL, BardoniB (2005) Fragile X Mental Retardation Protein: many partners and multiple targets for a promisuous function. Current Genomics 6: 515–522.

19. GarnonJ, LachanceC, Di MarcoS, HelZ, MarionD, et al. (2005) Fragile X-related protein FXR1P regulates proinflammatory cytokine tumor necrosis factor expression at the post-transcriptional level. J Biol Chem 280: 5750–5763.

20. VasudevanS, SteitzJA (2007) AU-rich-element-mediated upregulation of translation by FXR1 and Argonaute 2. Cell 128: 1105–1118.

21. BecharaE, DavidovicL, MelkoM, BensaidM, TremblayS, et al. (2007) Fragile X related protein 1 isoforms differentially modulate the affinity of fragile X mental retardation protein for G-quartet RNA structure. Nucleic Acids Res 35: 299–306.

22. WhitmanSA, CoverC, YuL, NelsonDL, ZarnescuDC, et al. (2011) Desmoplakin and talin2 are novel mRNA targets of fragile X-related protein-1 in cardiac muscle. Circ Res 109: 262–271.

23. MontarrasD, PinsetC, ChellyJ, KahnA, GrosF (1989) Expression of MyoD1 coincides with terminal differentiation in determined but inducible muscle cells. EMBO J 8: 2203–2207.

24. WatsonPA, Hanauske-AbelHH, FlintA, LalandeM (1991) Mimosine reversibly arrests cell cycle progression at the G1-S phase border. Cytometry 12: 242–246.

25. GongJ, TraganosF, DarzynkiewiczZ (1994) A selective procedure for DNA extraction from apoptotic cells applicable for gel electrophoresis and flow cytometry. Anal Biochem 218: 314–319.

26. KreipeH, HeidebrechtHJ, HansenS, RohlkW, KubbiesM, et al. (1993) A new proliferation-associated nuclear antigen detectable in paraffin-embedded tissues by the monoclonal antibody Ki-S1. Am J Pathol 142: 3–9.

27. BuckinghamM (1994) Muscle differentiation. Which myogenic factors make muscle? Curr Biol 4: 61–63.

28. HalevyO, NovitchBG, SpicerDB, SkapekSX, RheeJ, et al. (1995) Correlation of terminal cell cycle arrest of skeletal muscle with induction of p21 by MyoD. Science 267: 1018–1021.

29. SabourinLA, RudnickiMA (2000) The molecular regulation of myogenesis. Clin Genet 57: 16–25.

30. van der GiessenK, Di-MarcoS, ClairE, GallouziIE (2003) RNAi-mediated HuR depletion leads to the inhibition of muscle cell differentiation. J Biol Chem 278: 47119–47128.

31. SchaefferC, BardoniB, MandelJL, EhresmannB, EhresmannC, et al. (2001) The fragile X mental retardation protein binds specifically to its mRNA via a purine quartet motif. Embo J 20: 4803–4813.

32. UleJ, JensenK, MeleA, DarnellRB (2005) CLIP: a method for identifying protein-RNA interaction sites in living cells. Methods 37: 376–386.

33. KikinO, D'AntonioL, BaggaPS (2006) QGRS Mapper: a web-based server for predicting G-quadruplexes in nucleotide sequences. Nucleic Acids Res 34: W676–682.

34. YamaneA, AmanoO, SlavkinHC (2003) Insulin-like growth factors, hepatocyte growth factor and transforming growth factor-alpha in mouse tongue myogenesis. Dev Growth Differ 45: 1–6.

35. PhilippouA, MaridakiM, HalapasA, KoutsilierisM (2007) The role of the insulin-like growth factor 1 (IGF-1) in skeletal muscle physiology. In Vivo 21: 45–54.

36. GruberAR, FallmannJ, KratochvillF, KovarikP, HofackerIL (2011) AREsite: a database for the comprehensive investigation of AU-rich elements. Nucleic Acids Res 39: D66–69.

37. PuriPL, BalsanoC, BurgioVL, ChirilloP, NatoliG, et al. (1997) MyoD prevents cyclinA/cdk2 containing E2F complexes formation in terminally differentiated myocytes. Oncogene 14: 1171–1184.

38. PolesskayaA, RudnickiMA (2002) A MyoD-dependent differentiation checkpoint: ensuring genome integrity. Dev Cell 3: 757–758.

39. TamaniniF, KirkpatrickLL, SchonkerenJ, van UnenL, BontekoeC, et al. (2000) The fragile X-related proteins FXR1P and FXR2P contain a functional nucleolar-targeting signal equivalent to the HIV-1 regulatory proteins. Hum Mol Genet 9: 1487–1493.

40. CastetsM, SchaefferC, BecharaE, SchenckA, KhandjianEW, et al. (2005) FMRP interferes with the Rac1 pathway and controls actin cytoskeleton dynamics in murine fibroblasts. Hum Mol Genet 14: 835–844.

41. KwanKY, LamMM, JohnsonMB, DubeU, ShimS, et al. (2012) Species-dependent posttranscriptional regulation of NOS1 by FMRP in the developing cerebral cortex. Cell 149: 899–911.

42. SubramanianM, RageF, TabetR, FlatterE, MandelJL, et al. (2011) G-quadruplex RNA structure as a signal for neurite mRNA targeting. EMBO Rep 12: 697–704.

43. MelkoM, BardoniB (2010) The role of G-quadruplex in RNA metabolism: involvement of FMRP and FMR2P. Biochimie 92: 919–926.

44. KikinO, ZappalaZ, D'AntonioL, BaggaPS (2008) GRSDB2 and GRS_UTRdb: databases of quadruplex forming G-rich sequences in pre-mRNAs and mRNAs. Nucleic Acids Res 36: D141–148.

45. ArhinGK, BootsM, BaggaPS, MilcarekC, WiluszJ (2002) Downstream sequence elements with different affinities for the hnRNP H/H′ protein influence the processing efficiency of mammalian polyadenylation signals. Nucleic Acids Res 30: 1842–1850.

46. DanckwardtS, KaufmannI, GentzelM, FoerstnerKU, GantzertAS, et al. (2007) Splicing factors stimulate polyadenylation via USEs at non-canonical 3′ end formation signals. EMBO J 26: 2658–2669.

47. GagnonKT, CoreyDR (2012) Argonaute and the nuclear RNAs: new pathways for RNA-mediated control of gene expression. Nucleic Acid Ther 22: 3–16.

48. MortensenRD, SerraM, SteitzJA, VasudevanS (2011) Posttranscriptional activation of gene expression in Xenopus laevis oocytes by microRNA-protein complexes (microRNPs). Proc Natl Acad Sci U S A 108: 8281–8286.

49. CaudyAA, MyersM, HannonGJ, HammondSM (2002) Fragile X-related protein and VIG associate with the RNA interference machinery. Genes Dev 16: 2491–2496.

50. IshizukaA, SiomiMC, SiomiH (2002) A Drosophila fragile X protein interacts with components of RNAi and ribosomal proteins. Genes Dev 16: 2497–2508.

51. ChiSW, ZangJB, MeleA, DarnellRB (2009) Argonaute HITS-CLIP decodes microRNA-mRNA interaction maps. Nature 460: 479–86.

52. ChoSJ, ZhangJ, ChenX (2010) RNPC1 modulates the RNA-binding activity of, and cooperates with, HuR to regulate p21 mRNA stability. Nucleic Acids Res 38: 2256–2267.

53. BriataP, ForcalesSV, PonassiM, CorteG, ChenCY, et al. (2005) p38-dependent phosphorylation of the mRNA decay-promoting factor KSRP controls the stability of select myogenic transcripts. Mol Cell 20: 891–903.

54. WaggonerSA, JohannesGJ, LiebhaberSA (2009) Depletion of the poly(C)-binding proteins alphaCP1 and alphaCP2 from K562 cells leads to p53-independent induction of cyclin-dependent kinase inhibitor (CDKN1A) and G1 arrest. J Biol Chem 284: 9039–9049.

55. ScoumanneA, ChoSJ, ZhangJ, ChenX (2011) The cyclin-dependent kinase inhibitor p21 is regulated by RNA-binding protein PCBP4 via mRNA stability. Nucleic Acids Res 39: 213–224.

56. KitzmannM, FernandezA (2001) Crosstalk between cell cycle regulators and the myogenic factor MyoD in skeletal myoblasts. Cell Mol Life Sci 58: 571–579.

57. GuoK, WangJ, AndresV, SmithRC, WalshK (1995) MyoD-induced expression of p21 inhibits cyclin-dependent kinase activity upon myocyte terminal differentiation. Mol Cell Biol 15: 3823–3829.

58. WinokurST, BarrettK, MartinJH, ForresterJR, SimonM, et al. (2003) Facioscapulohumeral muscular dystrophy (FSHD) myoblasts demonstrate increased susceptibility to oxidative stress. Neuromuscul Disord 13: 322–333.

59. OsborneRJ, WelleS, VenanceSL, ThorntonCA, TawilR (2007) Expression profile of FSHD supports a link between retinal vasculopathy and muscular dystrophy. Neurology 68: 569–577.

60. HawkeTJ, MeesonAP, JiangN, GrahamS, HutchesonK, et al. (2003) p21 is essential for normal myogenic progenitor cell function in regenerating skeletal muscle. Am J Physiol Cell Physiol 285: C1019–1027.

61. YaffeD, SaxelO (1977) A myogenic cell line with altered serum requirements for differentiation. Differentiation 7: 159–166.

62. BlauHM, PavlathGK, HardemanEC, ChiuCP, SilbersteinL, et al. (1985) Plasticity of the differentiated state. Science 230: 758–766.

63. LivakKJ, SchmittgenTD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25: 402–408.

64. DavidovicL, BecharaE, GravelM, JaglinXH, TremblayS, et al. (2006) The nuclear microspherule protein 58 is a novel RNA-binding protein that interacts with fragile X mental retardation protein in polyribosomal mRNPs from neurons. Hum Mol Genet 15: 1525–1538.

65. DavidovicL, NavratilV, BonaccorsoCM, CataniaMV, BardoniB, et al. (2011) A metabolomic and systems biology perspective on the brain of the Fragile X syndrome mouse model. Genome Res 21: 2190–2202.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 3
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#