Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Decoupling of degradation from deadenylation reshapes poly(A) tail length in yeast meiosis

Abstract

Nascent messenger RNA is endowed with a poly(A) tail that is subject to gradual deadenylation and subsequent degradation in the cytoplasm. Deadenylation and degradation rates are typically correlated, rendering it difficult to dissect the determinants governing each of these processes and the mechanistic basis of their coupling. Here we developed an approach that allows systematic, robust and multiplexed quantification of poly(A) tails in Saccharomyces cerevisiae. Our results suggest that mRNA deadenylation and degradation rates are decoupled during meiosis, and that transcript length is a major determinant of deadenylation rates and a key contributor to reshaping of poly(A) tail lengths. Meiosis-specific decoupling also leads to unique positive associations between poly(A) tail length and gene expression. The decoupling is associated with a focal localization pattern of the RNA degradation factor Xrn1, and can be phenocopied by Xrn1 deletion under nonmeiotic conditions. Importantly, the association of transcript length with deadenylation rates is conserved across eukaryotes. Our study uncovers a factor that shapes deadenylation rate and reveals a unique context in which degradation is decoupled from deadenylation.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Meiotic poly(A) tail reshaping is associated with transcript length.
Fig. 2: Model for poly(A) tail length dynamics.
Fig. 3: Combination of modeling and perturbational assays shows decoupling of degradation from deadenylation in meiosis.
Fig. 4: Decoupling of degradation from deadenylation during meiosis triggers an association between gene expression and tail length.
Fig. 5: Xrn1 foci formation is connected to poly(A) tail–transcript length interaction.
Fig. 6: Transcript length impact in deadenylation is conserved in mammals.
Fig. 7: Poly(A) tail regulation in coupled and uncoupled regimes.

Similar content being viewed by others

Data availability

The datasets generated in this study for analyzing poly(A) tail length and RNA expression are available on Gene Expression Omnibus (GEO) under accession number GSE171329. Tail-seq and PAL-seq data were obtained from: GEO database (accession numbers GSE116355 and GSE134660). Meiotic RNA-seq and mass spectrometry data were obtained from GEO database (accession number GSE108778) and MassIVE (MSV000081874).

Code availability

Code for the analyses described in this paper is available from the corresponding author upon request.

References

  1. Lingner, J., Kellermann, J. & Keller, W. Cloning and expression of the essential gene for poly(A) polymerase from S. cerevisiae. Nature 354, 496–498 (1991).

    Article  CAS  PubMed  Google Scholar 

  2. Tucker, M., Staples, R. R., Valencia-Sanchez, M. A., Muhlrad, D. & Parker, R. Ccr4p is the catalytic subunit of a Ccr4p/Pop2p/Notp mRNA deadenylase complex in Saccharomyces cerevisiae. EMBO J. 21, 1427–1436 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. LaGrandeur, T. E. & Parker, R. Isolation and characterization of Dcp1p, the yeast mRNA decapping enzyme. EMBO J. 17, 1487–1496 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hsu, C. L. & Stevens, A. Yeast cells lacking 5′→3′ exoribonuclease 1 contain mRNA species that are poly(A) deficient and partially lack the 5′ cap structure. Mol. Cell. Biol. 13, 4826–4835 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Muhlrad, D., Decker, C. J. & Parker, R. Deadenylation of the unstable mRNA encoded by the yeast MFA2 gene leads to decapping followed by 5′→3′ digestion of the transcript. Genes Dev. 8, 855–866 (1994).

    Article  CAS  PubMed  Google Scholar 

  6. Larimer, F. W. & Stevens, A. Disruption of the gene XRN1, coding for a 5′→3′ exoribonuclease, restricts yeast cell growth. Gene 95, 85–90 (1990).

    Article  CAS  PubMed  Google Scholar 

  7. Goldstrohm, A. C. & Wickens, M. Multifunctional deadenylase complexes diversify mRNA control. Nat. Rev. Mol. Cell Biol. 9, 337–344 (2008).

    Article  CAS  PubMed  Google Scholar 

  8. Eisen, T. J. et al. The dynamics of cytoplasmic mRNA metabolism. Mol. Cell 77, 786–799.e10 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Brown, C. E. & Sachs, A. B. Poly(A) tail length control in Saccharomyces cerevisiae occurs by message-specific deadenylation. Mol. Cell. Biol. 18, 6548–6559 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Goldstrohm, A. C., Hook, B. A., Seay, D. J. & Wickens, M. PUF proteins bind Pop2p to regulate messenger RNAs. Nat. Struct. Mol. Biol. 13, 533–539 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Goldstrohm, A. C., Seay, D. J., Hook, B. A. & Wickens, M. PUF protein-mediated deadenylation is catalyzed by Ccr4p. J. Biol. Chem. 282, 109–114 (2007).

    Article  CAS  PubMed  Google Scholar 

  12. Webster, M. W., Stowell, J. A. & Passmore, L. A. RNA-binding proteins distinguish between similar sequence motifs to promote targeted deadenylation by Ccr4-Not. eLife 8, e40670 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Du, H. et al. YTHDF2 destabilizes m6A-containing RNA through direct recruitment of the CCR4–NOT deadenylase complex. Nat. Commun. 7, 12626 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Fabian, M. R. et al. miRNA-mediated deadenylation is orchestrated by GW182 through two conserved motifs that interact with CCR4–NOT. Nat. Struct. Mol. Biol. 18, 1211 (2011).

    Article  CAS  PubMed  Google Scholar 

  15. Chekulaeva, M. et al. miRNA repression involves GW182-mediated recruitment of CCR4–NOT through conserved W-containing motifs. Nat. Struct. Mol. Biol. 18, 1218 (2011).

    Article  CAS  PubMed  Google Scholar 

  16. Lim, J., Lee, M., Son, A., Chang, H. & Kim, V. N. MTAIL-seq reveals dynamic poly(A) tail regulation in oocyte-to-embryo development. Genes Dev. 30, 1671–1682 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Eichhorn, S. W. et al. mRNA poly(A)-tail changes specified by deadenylation broadly reshape translation in Drosophila oocytes and early embryos. eLife 5, e16955 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Kojima, S., Sher-Chen, E. L. & Green, C. B. Circadian control of mRNA polyadenylation dynamics regulates rhythmic protein expression. Genes Dev. 26, 2724–2736 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Yao, X., Kojima, S. & Chen, J. Critical role of deadenylation in regulating poly(A) rhythms and circadian gene expression. PLoS Comput. Biol. 16, e1007842 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Chen, C.-Y. A., Zhang, Y., Xiang, Y., Han, L. & Shyu, A.-B. Antagonistic actions of two human Pan3 isoforms on global mRNA turnover. RNA 23, 1404–1418 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Chen, C.-Y. A., Strouz, K., Huang, K.-L. & Shyu, A.-B. Tob2 phosphorylation regulates global mRNA turnover to reshape transcriptome and impact cell proliferation. RNA 26, 1143–1159 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yi, H. et al. PABP cooperates with the CCR4–NOT complex to promote mRNA deadenylation and block precocious decay. Mol. Cell 70, 1081–1088.e5 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Chu, S. The transcriptional program of sporulation in budding yeast. Science 282, 699–706 (1998).

    Article  CAS  PubMed  Google Scholar 

  24. Brar, G. A. et al. High-resolution view of the yeast meiotic program revealed by ribosome profiling. Science 335, 552–557 (2012).

    Article  CAS  PubMed  Google Scholar 

  25. Cheng, Z. et al. Pervasive, coordinated protein-level changes driven by transcript isoform switching during meiosis. Cell 172, 910–923.e16 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Lardenois, A. et al. Execution of the meiotic noncoding RNA expression program and the onset of gametogenesis in yeast require the conserved exosome subunit Rrp6. Proc. Natl Acad. Sci. USA 108, 1058–1063 (2011).

    Article  CAS  PubMed  Google Scholar 

  27. Agarwala, S. D., Blitzblau, H. G., Hochwagen, A. & Fink, G. R. RNA methylation by the MIS complex regulates a cell fate decision in yeast. PLoS Genet. 8, e1002732 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Schwartz, S. et al. High-resolution mapping reveals a conserved, widespread, dynamic mRNA methylation program in yeast meiosis. Cell 155, 1409–1421 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Subtelny, A. O., Eichhorn, S. W., Chen, G. R., Sive, H. & Bartel, D. P. Poly(A)-tail profiling reveals an embryonic switch in translational control. Nature 508, 66–71 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Chang, H., Lim, J., Ha, M. & Kim, V. N. TAIL-seq: genome-wide determination of poly(A) tail length and 3′ end modifications. Mol. Cell 53, 1044–1052 (2014).

    Article  CAS  PubMed  Google Scholar 

  31. Legnini, I., Alles, J., Karaiskos, N., Ayoub, S. & Rajewsky, N. FLAM-seq: full-length mRNA sequencing reveals principles of poly(A) tail length control. Nat. Methods 16, 879–886 (2019).

    Article  CAS  PubMed  Google Scholar 

  32. Mattijssen, S., Iben, J. R., Li, T., Coon, S. L. & Maraia, R. J. Single molecule poly(A) tail-seq shows LARP4 opposes deadenylation throughout mRNA lifespan with most impact on short tails. eLife 9, e59186 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Workman, R. E. et al. Nanopore native RNA sequencing of a human poly(A) transcriptome. Nat. Methods 16, 1297–1305 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Long, Y., Jia, J., Mo, W., Jin, X. & Zhai, J. FLEP-seq: simultaneous detection of RNA polymerase II position, splicing status, polyadenylation site and poly(A) tail length at genome-wide scale by single-molecule nascent RNA sequencing. Nat. Protoc. 16, 4355–4381 (2021).

    Article  CAS  PubMed  Google Scholar 

  35. Dierks, D. et al. Multiplexed profiling facilitates robust m6A quantification at site, gene and sample resolution. Nat. Methods 18, 1060–1067 (2021).

    Article  CAS  PubMed  Google Scholar 

  36. van Werven, F. J. et al. Transcription of two long noncoding RNAs mediates mating-type control of gametogenesis in budding yeast. Cell 150, 1170–1181 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Simchen, G., Piñon, R. & Salts, Y. Sporulation in Saccharomyces cerevisiae: premeiotic DNA synthesis, readiness and commitment. Exp. Cell. Res. 75, 207–218 (1972).

    Article  CAS  PubMed  Google Scholar 

  38. Parker, R. RNA degradation in Saccharomyces cerevisae. Genetics 191, 671–702 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Nishimura, K., Fukagawa, T., Takisawa, H., Kakimoto, T. & Kanemaki, M. An auxin-based degron system for the rapid depletion of proteins in nonplant cells. Nat. Methods 6, 917–922 (2009).

    Article  CAS  PubMed  Google Scholar 

  40. Etcheverry, T. Induced expression using yeast copper metallothionein promoter. Methods Enzymol. 185, 319–329 (1990).

    Article  CAS  PubMed  Google Scholar 

  41. Lima, S. A. et al. Short poly(A) tails are a conserved feature of highly expressed genes. Nat. Struct. Mol. Biol. 24, 1057–1063 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sheth, U. & Parker, R. Decapping and decay of messenger RNA occur in cytoplasmic processing bodies. Science 300, 805–808 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Grousl, T., Opekarová, M., Stradalova, V., Hasek, J. & Malinsky, J. Evolutionarily conserved 5′−3′ exoribonuclease Xrn1 accumulates at plasma membrane-associated eisosomes in post-diauxic yeast. PLoS ONE 10, e0122770 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Zhang, B., Butler, A. M., Shi, Q., Xing, S. & Herman, P. K. P-body localization of the Hrr25/casein kinase 1 protein kinase is required for the completion of meiosis. Mol. Cell. Biol. 38, e00678-17 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Zhao, H. et al. A role for the respiratory chain in regulating meiosis initiation in Saccharomyces cerevisiae. Genetics 208, 1181–1194 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Jambhekar, A. & Amon, A. Control of meiosis by respiration. Curr. Biol. 18, 969–975 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Weidberg, H., Moretto, F., Spedale, G., Amon, A. & van Werven, F. J. Nutrient control of yeast gametogenesis is mediated by TORC1, PKA and energy availability. PLoS Genet. 12, e1006075 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Kim, J. H. & Richter, J. D. Opposing polymerase-deadenylase activities regulate cytoplasmic polyadenylation. Mol. Cell 24, 173–183 (2006).

    Article  CAS  PubMed  Google Scholar 

  49. Lim, J. et al. Uridylation by TUT4 and TUT7 marks mRNA for degradation. Cell 159, 1365–1376 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Lim, J. et al. Mixed tailing by TENT4A and TENT4B shields mRNA from rapid deadenylation. Science 361, 701–704 (2018).

    Article  CAS  PubMed  Google Scholar 

  51. Yamashita, A. et al. Concerted action of poly(A) nucleases and decapping enzyme in mammalian mRNA turnover. Nat. Struct. Mol. Biol. 12, 1054–1063 (2005).

    Article  CAS  PubMed  Google Scholar 

  52. Amrani, N., Ghosh, S., Mangus, D. A. & Jacobson, A. Translation factors promote the formation of two states of the closed-loop mRNP. Nature 453, 1276–1280 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Ripley, B. et al. MASS: support functions and datasets for Venables and Ripley’s MASS. R package version 7.3-54 https://cran.r-project.org/web/packages/MASS/index.html (2011).

  54. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    CAS  PubMed  Google Scholar 

  55. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  58. Nagalakshmi, U. et al. The transcriptional landscape of the yeast genome defined by RNA sequencing. Science 320, 1344–1349 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Al-Shahrour, F., Minguez, P., Vaquerizas, J. M., Conde, L. & Dopazo, J. BABELOMICS: a suite of web tools for functional annotation and analysis of groups of genes in high-throughput experiments. Nucleic Acids Res. 33, W460–W464 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Picard tools (Broad Institute, 2016).

Download references

Acknowledgements

S.S. received funding from the Israel Science Foundation (grant no. 543165), the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation programme (grant no. 714023 and ERC-POC 899122) and the Estate of Emile Mimran. S.S. is the incumbent of the Robert Edward and Roselyn Rich Manson Career Development Chair in Perpetuity. Y.A. is supported by the Israel Science Foundation (grant no. 1105/20). Y.A. is the incumbent of the Sygnet Career Development Chair for Bioinformatics.

Author information

Authors and Affiliations

Authors

Contributions

D.W. and S.S. conceived the project. D.W. conducted all experiments. D.W. and S.S. performed the data analysis. Y.A. developed the mathematical model with the input of D.W. and S.S. S.S. acquired funding. D.W., Y.A. and S.S. wrote the paper.

Corresponding authors

Correspondence to Yaron Antebi or Schraga Schwartz.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 mPAT-seq accurately measures poly(A) tail length.

A) Cumulative fraction plot showing quantification of the spike-ins adenosines homopolymeric region length in NextSeq (top) and MiSeq (bottom) platforms. B) Barplot of Spearman correlation between mPAT-seq replicas with different depth requirements across all of the samples obtained in 1 hr interval after meiosis induction. C) Boxplot of genes mean poly(A) tail length in Wt and deadenylases deletion strains. Two sided Wilcoxon test was performed over n = 1 biologically independent samples. The boxplot was generated like in Fig. 2f. D) Barplot of Spearman correlation between PC1 loadings and different mRNA properties. E) Scatterplot of the PC1 loadings and the log2 of transcript length per gene. The figure was generated like in Fig. 1d (p < 2.2e-16). F) Histogram showing FACS DNA zstaining quantification across the meiotic time course. G) Kinetic for meiotic nuclear divisions as assayed by DAPI DNA staining. H) Percentage of spores obtained in deadenylases and xrn1 deletions meiotic time course measured after 24Hrs in SPO.

Extended Data Fig. 2 Decoupling of metabolic conditions from the meiotic gene program.

A) Barplot of Spearman correlation between gene mean poly(A) tail length and log2 of transcript length in the A/A background. B) Schematic representation of the return to growth (RtG) experiment, circles indicate when each of the samples was taken. C) Percentage of spores obtained in RtG experiments measured after 24Hrs in SPO. D) Barplot showing the correlation between poly(A) tail length and transcript length after transfer to YPD following 1Hr or 7Hrs of SPO incubation. E) Heatmap of the correlations of the RtG poly(A) tail profiles. Color code corresponds to the strength of Spearman correlation between each sample.

Extended Data Fig. 3 Poly(A) tail model is robust to deviation in the two key assumptions.

A) Simulated tail distribution of.genes with increasing variance in Amax and Amin between genes. B) Pooled distribution of the different simulations. Color code corresponds to the standard deviation of Amax and Amin distributions. C) Scatterplot showing the correlation between mean poly(A) tail and the fraction of short tail of the simulated distributions. D) Equation describing the linear interaction between poly(A) tail length and deadenylation rate (β) (top). Tail distributions under different rates of tail impact on deadenylation rate represented as b. Color code corresponds to this value (middle). Scatterplot showing the correlation between mean poly(A) tail and the fraction of short tail of the simulated distributions (bottom). E) Similar to D but for an exponential relationship between poly(A) tail length and the deadenylation rate.

Extended Data Fig. 4 Regulatory modules for poly(A) tail length dynamics.

A) A heatmap of calculated poly(A) tail lengths across 3-orders of magnitude variations in deadenylation and degradation rates, demonstrating the dependency of mean poly(A) tail lengths on the ratio between degradation and deadenylation rates. B) Scatterplot of the short tails fraction vs the mean poly(A) tail length, both in vegetative growth (left) and after 5Hrs in SPO (right). The number of genes in the analysis is specified in the figure. Spearman correlation and significance of the correlation coefficient p-value (Vegetative growth, p < 2.2e-16; Meiosis, p < 2.2e-16) using a t-distribution with n – 2 degrees of freedom were calculated. C) Transcript length (Ltx) potential regulatory modules over deadenylation (β) and degradation rates (γ). The first row consists of a simple linear relationship of transcript length over one of the rates (Or none in 1). The second row consists of the modules where transcript length affects both of the rates independently. The third and fourth rows contain modules where transcript length impacts one of the rates and in a dependent manner affects the other.

Extended Data Fig. 5 Disruption of production and degradation rates.

A) Scatterplot of the short tail fraction of xrn1Δ/Δ in vegetative growth condition vs the Wt, both in vegetative growth (left) and after 5Hrs in SPO (right). Genes were color coded by transcript length. Spearman correlation and significance of the correlation coefficient p-value (Vegetative growth, p = 5.6e-07; Meiosis, p < 2.2e-19) using a t-distribution with n – 2 degrees of freedom were calculated, and the number of analyzed transcripts specified. B) Xrn1 downregulation using the degron system. Left: boxplot of genes mean poly(A) tail length in Wt, before and after 30 min auxin addition. Middle: Spearman correlation between genes mean poly(A) tail length and log2 of transcript length. Right: Boxplot of the fraction of tails under 20nt per genes and plotted by transcript length groups indicating the number of genes per box. The boxplot was generated like in Fig. 2f. C) Boxplot showing the fraction of short tail per gene in the Wt and deadenylases deletion strains over n = 1 biologically independent samples. The boxplot was generated like in Fig. 2f. D) Tail length distributions of mRNA molecules of the Wt and pop2Δ/Δ in vegetative growth. A dashed line was drawn at 20nt. E) Barplot showing the relative induction of Ccr4 after induction measured by RT-qPCR.F) Barplot showing Spearman correlation between genes mean poly(A) tail length and log2 of transcript length of the Wt and Ccr4 inducible strain before and after induction. G) Boxplot of the fraction of tails under 20nt per gene and plotted by transcript length groups. The number of genes per group is indicated with the corresponding specific color. The boxplot was generated like in Fig. 2f.

Extended Data Fig. 6 The negative correlation between poly(A) tail and gene expression in vegetative growth conditions is driven by a subset of highly expressed genes.

Scatterplots of the poly(A) tail length vs log2 of normalized gene expression in the Wt in vegetative growth (Left) meiosis (Center) and xrn1Δ/Δ in vegetative growth (Right). Highly expressed genes were excluded from the analysis retaining the following percentages of genes: 100% (up), 90% (middle), and 75% (bottom). The figure was generated like in Fig. 1d (100%:Vegetative growth, p < 2.2e-16; Meiosis, p < 2.2e-16; xrn1Δ/Δ, p < 2.2e-16–90%:Vegetative growth, p < 2.2e-16; Meiosis, p < 2.2e-16; xrn1Δ/Δ, p < 2.2e-16–75%:Vegetative growth, p < 2.5e-07; Meiosis, p < 2.2e-16; xrn1Δ/Δ, p < 2.2e-16).

Extended Data Fig. 7 Expression levels of the degradation and deadenylation machinery related genes do not undergo major changes in meiosis.

Mass spectrometry (blue) and RNA-seq (red) results from Cheng et25 were normalized to vegetative control and plotted. The dashed line is drawn at a 2-fold change. Meiosis related genes were selected as control.

Extended Data Fig. 8 Initial poly(A) tail length and decapping rate are not associated with transcript length.

A) Scatterplot of log2 of the initial poly(A) tail (nt) and log2 of transcript length.Spearman correlation and significance of the correlation coefficient p-value(p = 1.2e-05) using a t-distribution with n – 2 degrees of freedom were calculated, and the number of analysed transcripts specified. B) Scatterplot of log2 of the degradation rate and log2 of transcript length. Spearman correlation and significance of the correlation coefficient p-value (p = 9.1e-12) using a t-distribution with n – 2 degrees of freedom were calculated, and the number of analysed transcripts specified.

Extended Data Fig. 9 Biological implications behind decoupling.

A) Scatterplot of the poly(A) tail differences between the ccr4Δ/Δ (up)/pop2Δ/Δ(down) and the Wt vs the log2 of transcript length, both in vegetative growth (left) and after 5Hrs in SPO (right). The figure was generated like in Fig. 1d (ccr4Δ/Δ:Vegetative growth, p < 2.2e-16; Meiosis, p < 2.2e-16-pop2Δ/Δ:Vegetative growth, p < 2.2e-16; Meiosis, p < 2.2e-16). B) Barplot showing the standard deviation of the log2 normalized gene expression of the Wt (Vegetative growth and meiosis) and the xrn1Δ/Δ strain in vegetative growth conditions. B) Similar to A but for cells uncommitted (1Hr in SPO) and committed (7Hrs in SPO) to meiosis, before and after transfer to YPD. C) Relevant groups of genes for mitosis and meiosis undergo tail length changes across meiosis. Gene ontology terms associated with elongated and shortened tails across the meiotic time course.

Extended Data Fig. 10 Partially degraded mRNA molecules are not detectable with RiboZero RNA-seq.

A) Sample normalized coverage across normalized transcript length generated with Picard tools60 in the Wt (Vegetative growth and meiosis) and xrn1Δ/Δ strains. B) Violin plots showing the gene specific 3′ Bias calculated either as the ratio of the last 100nt coverage divided by the first 100nt coverage (Left) or as the ratio of the last 10%nt coverage divided by the first 10%nt coverage (Right). C) Scatterplots of both 3′bias metrics vs the log2 of transcript length. Spearman correlation was calculated and significance of the correlation coefficient p-value (100nt-Vegetative growth, p = 0.44; Meiosis, p = 0.54; xrn1Δ/Δ, p = 0.8–10%:Vegetative growth, p = 0.81; Meiosis, p = 0.93; xrn1Δ/Δ, p = 0.39) using a t-distribution with n – 2 degrees of freedom were calculated, and the number of analysed transcripts specified.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wiener, D., Antebi, Y. & Schwartz, S. Decoupling of degradation from deadenylation reshapes poly(A) tail length in yeast meiosis. Nat Struct Mol Biol 28, 1038–1049 (2021). https://doi.org/10.1038/s41594-021-00694-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-021-00694-3

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing