#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

The PARN Deadenylase Targets a Discrete Set of mRNAs for Decay and Regulates Cell Motility in Mouse Myoblasts


PARN is one of several deadenylase enzymes present in mammalian cells, and as such the contribution it makes to the regulation of gene expression is unclear. To address this, we performed global mRNA expression and half-life analysis on mouse myoblasts depleted of PARN. PARN knockdown resulted in the stabilization of 40 mRNAs, including that encoding the mRNA decay factor ZFP36L2. Additional experiments demonstrated that PARN knockdown induced an increase in Zfp36l2 poly(A) tail length as well as increased translation. The elements responsible for PARN-dependent regulation lie within the 3′ UTR of the mRNA. Surprisingly, changes in mRNA stability showed an inverse correlation with mRNA abundance; stabilized transcripts showed either no change or a decrease in mRNA abundance. Moreover, we found that stabilized mRNAs had reduced accumulation of pre–mRNA, consistent with lower transcription rates. This presents compelling evidence for the coupling of mRNA decay and transcription to buffer mRNA abundances. Although PARN knockdown altered decay of relatively few mRNAs, there was a much larger effect on global gene expression. Many of the mRNAs whose abundance was reduced by PARN knockdown encode factors required for cell migration and adhesion. The biological relevance of this observation was demonstrated by the fact that PARN KD cells migrate faster in wound-healing assays. Collectively, these data indicate that PARN modulates decay of a defined set of mRNAs in mammalian cells and implicate this deadenylase in coordinating control of genes required for cell movement.


Vyšlo v časopise: The PARN Deadenylase Targets a Discrete Set of mRNAs for Decay and Regulates Cell Motility in Mouse Myoblasts. PLoS Genet 8(8): e32767. doi:10.1371/journal.pgen.1002901
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1002901

Souhrn

PARN is one of several deadenylase enzymes present in mammalian cells, and as such the contribution it makes to the regulation of gene expression is unclear. To address this, we performed global mRNA expression and half-life analysis on mouse myoblasts depleted of PARN. PARN knockdown resulted in the stabilization of 40 mRNAs, including that encoding the mRNA decay factor ZFP36L2. Additional experiments demonstrated that PARN knockdown induced an increase in Zfp36l2 poly(A) tail length as well as increased translation. The elements responsible for PARN-dependent regulation lie within the 3′ UTR of the mRNA. Surprisingly, changes in mRNA stability showed an inverse correlation with mRNA abundance; stabilized transcripts showed either no change or a decrease in mRNA abundance. Moreover, we found that stabilized mRNAs had reduced accumulation of pre–mRNA, consistent with lower transcription rates. This presents compelling evidence for the coupling of mRNA decay and transcription to buffer mRNA abundances. Although PARN knockdown altered decay of relatively few mRNAs, there was a much larger effect on global gene expression. Many of the mRNAs whose abundance was reduced by PARN knockdown encode factors required for cell migration and adhesion. The biological relevance of this observation was demonstrated by the fact that PARN KD cells migrate faster in wound-healing assays. Collectively, these data indicate that PARN modulates decay of a defined set of mRNAs in mammalian cells and implicate this deadenylase in coordinating control of genes required for cell movement.


Zdroje

1. WiederholdK, PassmoreLA (2010) Cytoplasmic deadenylation: regulation of mRNA fate. Biochem Soc Trans 38: 1531–1536 doi:10.1042/BST0381531.

2. GoldstrohmAC, WickensM (2008) Multifunctional deadenylase complexes diversify mRNA control. Nat Rev Mol Cell Biol 9: 337–344 doi:10.1038/nrm2370.

3. CollartMA, PanasenkoOO (2012) The Ccr4-Not complex. Gene 492: 42–53 doi:10.1016/j.gene.2011.09.033.

4. LauN-C, KolkmanA, van SchaikFMA, MulderKW, PijnappelWWMP, et al. (2009) Human Ccr4-Not complexes contain variable deadenylase subunits. Biochem J 422: 443–453 doi:10.1042/BJ20090500.

5. BraunJE, HuntzingerE, FauserM, IzaurraldeE (2011) GW182 proteins directly recruit cytoplasmic deadenylase complexes to miRNA targets. Mol Cell 44: 120–133 doi:10.1016/j.molcel.2011.09.007.

6. SandlerH, KrethJ, TimmersHTM, StoecklinG (2011) Not1 mediates recruitment of the deadenylase Caf1 to mRNAs targeted for degradation by tristetraprolin. Nucleic Acids Res 39: 4373–4386 doi:10.1093/nar/gkr011.

7. GaoM, FritzDT, FordLP, WiluszJ (2000) Interaction between a poly(A)-specific ribonuclease and the 5′ cap influences mRNA deadenylation rates in vitro. Mol Cell 5: 479–488.

8. DehlinE, WormingtonM, KörnerCG, WahleE (2000) Cap-dependent deadenylation of mRNA. EMBO J 19: 1079–1086 doi:10.1093/emboj/19.5.1079.

9. MartînezJ, RenYG, NilssonP, EhrenbergM, VirtanenA (2001) The mRNA cap structure stimulates rate of poly(A) removal and amplifies processivity of degradation. J Biol Chem 276: 27923–27929 doi:10.1074/jbc.M102270200.

10. CevherMA, ZhangX, FernandezS, KimS, BaqueroJ, et al. (2010) Nuclear deadenylation/polyadenylation factors regulate 3′ processing in response to DNA damage. EMBO J 29: 1674–1687 doi:10.1038/emboj.2010.59.

11. ReinhardtHC, HasskampP, SchmeddingI, MorandellS, van VugtMATM, et al. (2010) DNA damage activates a spatially distinct late cytoplasmic cell-cycle checkpoint network controlled by MK2-mediated RNA stabilization. Mol Cell 40: 34–49 doi:10.1016/j.molcel.2010.09.018.

12. KörnerCG, WormingtonM, MuckenthalerM, SchneiderS, DehlinE, et al. (1998) The deadenylating nuclease (DAN) is involved in poly(A) tail removal during the meiotic maturation of Xenopus oocytes. EMBO J 17: 5427–5437 doi:10.1093/emboj/17.18.5427.

13. BerndtH, HarnischC, RammeltC, StöhrN, ZirkelA, et al. (2012) Maturation of mammalian H/ACA box snoRNAs: PAPD5-dependent adenylation and PARN-dependent trimming. RNA 18: 958–972 doi:10.1261/rna.032292.112.

14. MoraesKCM, WiluszCJ, WiluszJ (2006) CUG-BP binds to RNA substrates and recruits PARN deadenylase. RNA 12: 1084–1091 doi:10.1261/rna.59606.

15. OtaR, KotaniT, YamashitaM (2011) Biochemical characterization of Pumilio1 and Pumilio2 in Xenopus oocytes. J Biol Chem 286: 2853–2863 doi:10.1074/jbc.M110.155523.

16. KimJH, RichterJD (2006) Opposing polymerase-deadenylase activities regulate cytoplasmic polyadenylation. Mol Cell 24: 173–183 doi:10.1016/j.molcel.2006.08.016.

17. LaiWS, KenningtonEA, BlackshearPJ (2003) Tristetraprolin and its family members can promote the cell-free deadenylation of AU-rich element-containing mRNAs by poly(A) ribonuclease. Mol Cell Biol 23: 3798–3812.

18. MittalS, AslamA, DoidgeR, MedicaR, WinklerGS (2011) The Ccr4a (CNOT6) and Ccr4b (CNOT6L) deadenylase subunits of the human Ccr4-Not complex contribute to the prevention of cell death and senescence. Mol Biol Cell 22: 748–758 doi:10.1091/mbc.E10-11-0898.

19. ShalemO, GroismanB, ChoderM, DahanO, PilpelY (2011) Transcriptome kinetics is governed by a genome-wide coupling of mRNA production and degradation: a role for RNA Pol II. PLoS Genet 7: e1002273 doi:10.1371/journal.pgen.1002273.

20. BregmanA, Avraham-KelbertM, BarkaiO, DuekL, GutermanA, et al. (2011) Promoter elements regulate cytoplasmic mRNA decay. Cell 147: 1473–1483 doi:10.1016/j.cell.2011.12.005.

21. TrcekT, LarsonDR, MoldónA, QueryCC, SingerRH (2011) Single-molecule mRNA decay measurements reveal promoter- regulated mRNA stability in yeast. Cell 147: 1484–1497 doi:10.1016/j.cell.2011.11.051.

22. Dori-BachashM, ShemaE, TiroshI (2011) Coupled evolution of transcription and mRNA degradation. PLoS Biol 9: e1001106 doi:10.1371/journal.pbio.1001106.

23. LeeJE, LeeJY, WiluszJ, TianB, WiluszCJ (2010) Systematic analysis of cis-elements in unstable mRNAs demonstrates that CUGBP1 is a key regulator of mRNA decay in muscle cells. PLoS ONE 5: e11201 doi:10.1371/journal.pone.0011201.

24. HuangDW, ShermanBT, LempickiRA (2009) Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4: 44–57 doi:10.1038/nprot.2008.211.

25. HorwitzR, WebbD (2003) Cell migration. Current Biology 13: R756–R759 doi:10.1016/j.cub.2003.09.014.

26. FriedlP (2004) Prespecification and plasticity: shifting mechanisms of cell migration. Current Opinion in Cell Biology 16: 14–23 doi:10.1016/j.ceb.2003.11.001.

27. PichotCS, ArvanitisC, HartigSM, JensenSA, BechillJ, et al. (2010) Cdc42-interacting protein 4 promotes breast cancer cell invasion and formation of invadopodia through activation of N-WASp. Cancer Res 70: 8347–8356 doi:10.1158/0008-5472.CAN-09-4149.

28. MonciniS, SalviA, ZuccottiP, VieroG, QuattroneA, et al. (2011) The role of miR-103 and miR-107 in regulation of CDK5R1 expression and in cellular migration. PLoS ONE 6: e20038 doi:10.1371/journal.pone.0020038.

29. SahlgrenCM, MikhailovA, VaittinenS, PallariH-M, KalimoH, et al. (2003) Cdk5 regulates the organization of Nestin and its association with p35. Mol Cell Biol 23: 5090–5106.

30. MonciniS, BevilacquaA, VenturinM, FalliniC, RattiA, et al. (2007) The 3′ untranslated region of human Cyclin-Dependent Kinase 5 Regulatory subunit 1 contains regulatory elements affecting transcript stability. BMC Mol Biol 8: 111 doi:10.1186/1471-2199-8-111.

31. BoudoukhaS, CuvellierS, PolesskayaA (2010) Role of the RNA-binding protein IMP-2 in muscle cell motility. Mol Cell Biol 30: 5710–5725 doi:10.1128/MCB.00665-10.

32. GuW, KatzZ, WuB, ParkHY, LiD, et al. (2012) Regulation of local expression of cell adhesion and motility-related mRNAs in breast cancer cells by IMP1/ZBP1. J Cell Sci 125: 81–91 doi:10.1242/jcs.086132.

33. GrimaDP, SullivanM, ZabolotskayaMV, BrowneC, SeagoJ, et al. (2008) The 5′-3′ exoribonuclease pacman is required for epithelial sheet sealing in Drosophila and genetically interacts with the phosphatase puckered. Biol Cell 100: 687–701 doi:10.1042/BC20080049.

34. ZhangL, LeeJE, WiluszJ, WiluszCJ (2008) The RNA-binding protein CUGBP1 regulates stability of tumor necrosis factor mRNA in muscle cells: implications for myotonic dystrophy. J Biol Chem 283: 22457–22463 doi:10.1074/jbc.M802803200.

35. StewartSA, DykxhoornDM, PalliserD, MizunoH, YuEY, et al. (2003) Lentivirus-delivered stable gene silencing by RNAi in primary cells. RNA 9: 493–501.

36. WangY, LiuCL, StoreyJD, TibshiraniRJ, HerschlagD, et al. (2002) Precision and functional specificity in mRNA decay. Proc Natl Acad Sci USA 99: 5860–5865 doi:10.1073/pnas.092538799.

37. GarneauNL, SokoloskiKJ, OpyrchalM, NeffCP, WiluszCJ, et al. (2008) The 3′ untranslated region of sindbis virus represses deadenylation of viral transcripts in mosquito and Mammalian cells. J Virol 82: 880–892 doi:10.1128/JVI.01205-07.

38. GarneauNL, WiluszCJ, WiluszJ (2008) Chapter 5. In vivo analysis of the decay of transcripts generated by cytoplasmic RNA viruses. Meth Enzymol 449: 97–123 doi:10.1016/S0076-6879(08)02405-1.

39. WindhagerL, BonfertT, BurgerK, RuzsicsZ, KrebsS, et al. (2012) Ultra short and progressive 4sU-tagging reveals key characteristics of RNA processing at nucleotide resolution. Genome research Available:http://www.ncbi.nlm.nih.gov/pubmed/22539649. Accessed 15 May 2012.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2012 Číslo 8
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#