Skip to main content
Log in

Cell-type-dependent histone demethylase specificity promotes meiotic chromosome condensation in Arabidopsis

  • Article
  • Published:

From Nature Plants

View current issue Submit your manuscript

An Author Correction to this article was published on 28 July 2020

This article has been updated

Abstract

Histone demethylation is crucial for proper chromatin structure and to ensure normal development, and requires the large family of Jumonji C (JmjC)-containing demethylases; however, the molecular mechanisms that regulate the substrate specificity of these JmjC-containing demethylases remain largely unknown. Here, we show that the substrate specificity of the Arabidopsis histone demethylase JMJ16 is broadened from Lys 4 of histone H3 (H3K4) alone in somatic cells to both H3K4 and H3K9 when it binds to the meiocyte-specific histone reader MMD1. Consistent with this, the JMJ16 catalytic domain exhibits both H3K4 and H3K9 demethylation activities. Moreover, the JMJ16 C-terminal FYR domain interacts with the JMJ16 catalytic domain and probably restricts its substrate specificity. By contrast, MMD1 can compete with the N-terminal catalytic domain of JMJ16 for binding to the FYR-C domain, thereby expanding the substrate specificity of JMJ16 by preventing the FYR domain from binding to the catalytic domain. We propose that MMD1 and JMJ16 together in male meiocytes promote gene expression in an H3K9me3-dependent manner and thereby contribute to meiotic chromosome condensation.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: JMJ16 interacts with MMD1 in vitro and in vivo.
Fig. 2: Demethylase activity assay of JMJ16.
Fig. 3: MMD1 promotes the substrate specificity of JMJ16 to H3K9me3.
Fig. 4: The effect of MMD1 on the substrate specificity of JMJ16.
Fig. 5: Structural modelling analysis of JMJ16 substrate specificity.
Fig. 6: JMJ16 promotes meiocyte gene expression and chromosome condensation in male meiocytes.
Fig. 7: A model of cell-type-dependent regulation of gene expression by the JMJ16–MMD1 module.

Similar content being viewed by others

Data availability

All data are available in the paper and its Supplementary Information. RNA-seq data of atjmj16 and atmmd1 were deposited to the SRA under accession numbers SRR3584322 (ref. 36) and PRJNA530269. Source data are provided with this paper.

Change history

References

  1. Klose, R. J. & Zhang, Y. Regulation of histone methylation by demethylimination and demethylation. Nat. Rev. Mol. Cell Biol. 8, 307–318 (2007).

    CAS  PubMed  Google Scholar 

  2. Shi, Y. et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119, 941–953 (2004).

    CAS  PubMed  Google Scholar 

  3. Tsukada, Y. et al. Histone demethylation by a family of JmjC domain-containing proteins. Nature 439, 811–816 (2006).

    CAS  PubMed  Google Scholar 

  4. Chen, X., Hu, Y. & Zhou, D. X. Epigenetic gene regulation by plant Jumonji group of histone demethylase. Biochim. Biophys. Acta 1809, 421–426 (2011).

    CAS  PubMed  Google Scholar 

  5. Liu, C., Lu, F., Cui, X. & Cao, X. Histone methylation in higher plants. Annu. Rev. Plant Biol. 61, 395–420 (2010).

    CAS  PubMed  Google Scholar 

  6. Kooistra, S. M. & Helin, K. Molecular mechanisms and potential functions of histone demethylases. Nat. Rev. Mol. Cell Biol. 13, 297–311 (2012).

    CAS  PubMed  Google Scholar 

  7. Ko, J. H. et al. Growth habit determination by the balance of histone methylation activities in Arabidopsis. EMBO J. 29, 3208–3215 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Lu, F., Cui, X., Zhang, S., Jenuwein, T. & Cao, X. Arabidopsis REF6 is a histone H3 lysine 27 demethylase. Nat. Genet. 43, 715–719 (2011).

    CAS  PubMed  Google Scholar 

  9. Yu, X. et al. Modulation of brassinosteroid-regulated gene expression by Jumonji domain-containing proteins ELF6 and REF6 in Arabidopsis. Proc. Natl Acad. Sci. USA 105, 7618–7623 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Lin, C. H. et al. Heterochromatin protein 1a stimulates histone H3 lysine 36 demethylation by the Drosophila KDM4A demethylase. Mol. Cell 32, 696–706 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Trojer, P. et al. Dynamic histone H1 isotype 4 methylation and demethylation by histone lysine methyltransferase G9a/KMT1C and the Jumonji domain-containing JMJD2/KDM4 proteins. J. Biol. Chem. 284, 8395–8405 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Whetstine, J. R. et al. Reversal of histone lysine trimethylation by the JMJD2 family of histone demethylases. Cell 125, 467–481 (2006).

    CAS  PubMed  Google Scholar 

  13. Walport, L. J. et al. Mechanistic and structural studies of KDM-catalysed demethylation of histone 1 isotype 4 at lysine 26. FEBS Lett. 592, 3264–3273 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Lu, F. et al. Comparative analysis of JmjC domain-containing proteins reveals the potential histone demethylases in Arabidopsis and rice. J. Integr. Plant Biol. 50, 886–896 (2008).

    CAS  PubMed  Google Scholar 

  15. Garcia-Alai, M. M., Allen, M. D., Joerger, A. C. & Bycroft, M. The structure of the FYR domain of transforming growth factor beta regulator 1. Protein Sci. 19, 1432–1438 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Zhou, X. & Ma, H. Evolutionary history of histone demethylase families: distinct evolutionary patterns suggest functional divergence. BMC Evol. Biol. 8, 294 (2008).

    PubMed  PubMed Central  Google Scholar 

  17. Lu, F., Cui, X., Zhang, S., Liu, C. & Cao, X. JMJ14 is an H3K4 demethylase regulating flowering time in Arabidopsis. Cell Res. 20, 387–390 (2010).

    PubMed  Google Scholar 

  18. Shen, Y. et al. Over-expression of histone H3K4 demethylase gene JMJ15 enhances salt tolerance in Arabidopsis. Front. Plant Sci. 5, 290 (2014).

    PubMed  PubMed Central  Google Scholar 

  19. Yang, H. et al. Overexpression of a histone H3K4 demethylase, JMJ15, accelerates flowering time in Arabidopsis. Plant Cell Rep. 31, 1297–1308 (2012).

    CAS  PubMed  Google Scholar 

  20. Liu, P. et al. The histone H3K4 demethylase JMJ16 represses leaf senescence in Arabidopsis. Plant Cell 31, 430–443 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Yang, H. et al. A companion cell-dominant and developmentally regulated H3K4 demethylase controls flowering time in Arabidopsis via the repression of FLC expression. PLoS Genet. 8, e1002664 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Chen, Q. et al. Structural basis of a histone H3 lysine 4 demethylase required for stem elongation in rice. PLoS Genet. 9, e1003239 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Cui, X. et al. Control of transposon activity by a histone H3K4 demethylase in rice. Proc. Natl Acad. Sci. USA 110, 1953–1958 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Yang, Z. et al. Structure of the Arabidopsis JMJ14-H3K4me3 complex provides insight into the substrate specificity of KDM5 subfamily histone demethylases. Plant Cell 30, 167–177 (2018).

    CAS  PubMed  Google Scholar 

  25. Becker, J. S., Nicetto, D. & Zaret, K. S. H3K9me3-dependent heterochromatin: barrier to cell fate changes. Trends Genet. 32, 29–41 (2016).

    CAS  PubMed  Google Scholar 

  26. Johnson, L. et al. Mass spectrometry analysis of Arabidopsis histone H3 reveals distinct combinations of post-translational modifications. Nucleic Acids Res. 32, 6511–6518 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Zhang, K., Sridhar, V. V., Zhu, J., Kapoor, A. & Zhu, J. K. Distinctive core histone post-translational modification patterns in Arabidopsis thaliana. PLoS ONE 2, e1210 (2007).

    PubMed  PubMed Central  Google Scholar 

  28. Turck, F. et al. Arabidopsis TFL2/LHP1 specifically associates with genes marked by trimethylation of histone H3 lysine 27. PLoS Genet. 3, e86 (2007).

    PubMed  PubMed Central  Google Scholar 

  29. Veiseth, S. V. et al. The SUVR4 histone lysine methyltransferase binds ubiquitin and converts H3K9me1 to H3K9me3 on transposon chromatin in Arabidopsis. PLoS Genet. 7, e1001325 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Zheng, S. et al. The Arabidopsis H3K27me3 demethylase JUMONJI 13 is a temperature and photoperiod dependent flowering repressor. Nat. Commun. 10, 1303 (2019).

    PubMed  PubMed Central  Google Scholar 

  31. Sun, Q. & Zhou, D. X. Rice jmjC domain-containing gene JMJ706 encodes H3K9 demethylase required for floral organ development. Proc. Natl Acad. Sci. USA 105, 13679–13684 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Lee, K., Park, O. S. & Seo, P. J. JMJ30-mediated demethylation of H3K9me3 drives tissue identity changes to promote callus formation in Arabidopsis. Plant J. 95, 961–975 (2018).

    CAS  PubMed  Google Scholar 

  33. Reddy, T. V., Kaur, J., Agashe, B., Sundaresan, V. & Siddiqi, I. The DUET gene is necessary for chromosome organization and progression during male meiosis in Arabidopsis and encodes a PHD finger protein. Development 130, 5975–5987 (2003).

    CAS  PubMed  Google Scholar 

  34. Yang, X., Makaroff, C. A. & Ma, H. The Arabidopsis MALE MEIOCYTE DEATH1 gene encodes a PHD-finger protein that is required for male meiosis. Plant Cell 15, 1281–1295 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Andreuzza, S., Nishal, B., Singh, A. & Siddiqi, I. The chromatin protein DUET/MMD1 controls expression of the meiotic gene TDM1 during male meiosis in Arabidopsis. PLoS Genet. 11, e1005396 (2015).

    PubMed  PubMed Central  Google Scholar 

  36. Wang, J. et al. The PHD finger protein MMD1/DUET ensures the progression of male meiotic chromosome condensation and directly regulates the expression of the condensin gene CAP-D3. Plant Cell 28, 1894–1909 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Sanchez, R. & Zhou, M. M. The PHD finger: a versatile epigenome reader. Trends Biochem. Sci. 36, 364–372 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Kelley, L. A. & Sternberg, M. J. Protein structure prediction on the web: a case study using the Phyre server. Nat. Protoc. 4, 363–371 (2009).

    CAS  PubMed  Google Scholar 

  39. Ng, S. S. et al. Crystal structures of histone demethylase JMJD2A reveal basis for substrate specificity. Nature 448, 87–91 (2007).

    CAS  PubMed  Google Scholar 

  40. Kruidenier, L. et al. A selective Jumonji H3K27 demethylase inhibitor modulates the proinflammatory macrophage response. Nature 488, 404–408 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhang, Y. & Reinberg, D. Transcription regulation by histone methylation: interplay between different covalent modifications of the core histone tails. Genes Dev. 15, 2343–2360 (2001).

    CAS  PubMed  Google Scholar 

  42. Lauria, M. & Rossi, V. Epigenetic control of gene regulation in plants. Biochim. Biophys. Acta 1809, 369–378 (2011).

    CAS  PubMed  Google Scholar 

  43. Mohan, M., Herz, H. M. & Shilatifard, A. SnapShot: histone lysine methylase complexes. Cell 149, 498–498 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Horton, J. R. et al. Enzymatic and structural insights for substrate specificity of a family of Jumonji histone lysine demethylases. Nat. Struct. Mol. Biol. 17, 38–43 (2010).

    CAS  PubMed  Google Scholar 

  45. Yang, Y. et al. Structural insights into a dual-specificity histone demethylase ceKDM7A from Caenorhabditis elegans. Cell Res. 20, 886–898 (2010).

    CAS  PubMed  Google Scholar 

  46. Li, F. et al. Lid2 is required for coordinating H3K4 and H3K9 methylation of heterochromatin and euchromatin. Cell 135, 272–283 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Okada, Y., Scott, G., Ray, M. K., Mishina, Y. & Zhang, Y. Histone demethylase JHDM2A is critical for Tnp1 and Prm1 transcription and spermatogenesis. Nature 450, 119–123 (2007).

    CAS  PubMed  Google Scholar 

  48. Okada, Y., Tateishi, K. & Zhang, Y. Histone demethylase JHDM2A is involved in male infertility and obesity. J. Androl. 31, 75–78 (2010).

    CAS  PubMed  Google Scholar 

  49. Luo, M., Hung, F. Y., Yang, S. G., Liu, X. C. & Wu, K. Q. Histone lysine demethylases and their functions in plants. Plant Mol. Biol. Rep. 32, 558–565 (2014).

    CAS  Google Scholar 

  50. Hojfeldt, J. W., Agger, K. & Helin, K. Histone lysine demethylases as targets for anticancer therapy. Nat. Rev. Drug Discov. 12, 917–930 (2013).

    CAS  PubMed  Google Scholar 

  51. Yan, Y. et al. A MYB-domain protein EFM mediates flowering responses to environmental cues in Arabidopsis. Dev. Cell 30, 437–448 (2014).

    CAS  PubMed  Google Scholar 

  52. Gan, E. S. et al. Jumonji demethylases moderate precocious flowering at elevated temperature via regulation of FLC in Arabidopsis. Nat. Commun. 5, 5098 (2014).

    CAS  PubMed  Google Scholar 

  53. Loenarz, C. et al. PHF8, a gene associated with cleft lip/palate and mental retardation, encodes for an N ε-dimethyl lysine demethylase. Hum. Mol. Genet. 19, 217–222 (2010).

    CAS  PubMed  Google Scholar 

  54. Qi, H. H. et al. Histone H4K20/H3K9 demethylase PHF8 regulates zebrafish brain and craniofacial development. Nature 466, 503–507 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Klose, R. J. et al. Demethylation of histone H3K36 and H3K9 by Rph1: a vestige of an H3K9 methylation system in Saccharomyces cerevisiae. Mol. Cell. Biol. 27, 3951–3961 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Crevillen, P. et al. Epigenetic reprogramming that prevents transgenerational inheritance of the vernalized state. Nature 515, 587–590 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Lalonde, M. E., Cheng, X. & Cote, J. Histone target selection within chromatin: an exemplary case of teamwork. Genes Dev. 28, 1029–1041 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Qian, S., Wang, Y., Ma, H. & Zhang, L. Expansion and functional divergence of JumonjiC-containing histone demethylases: significance of duplications in ancestral angiosperms and vertebrates. Plant Physiol. 168, 1321–1337 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Naumann, K. et al. Pivotal role of AtSUVH2 in heterochromatic histone methylation and gene silencing in Arabidopsis. The EMBO J. 24, 1418–1429 (2005).

    CAS  PubMed  Google Scholar 

  60. Wang, Y., Cheng, Z., Lu, P., Timofejeva, L. & Ma, H. Molecular cell biology of male meiotic chromosomes and isolation of male meiocytes in Arabidopsis thaliana. Methods Mol. Biol. 1110, 217–230 (2014).

    CAS  PubMed  Google Scholar 

  61. Hu, C. D., Chinenov, Y. & Kerppola, T. K. Visualization of interactions among bZIP and Rel family proteins in living cells using bimolecular fluorescence complementation. Mol. Cell 9, 789–798 (2002).

    CAS  PubMed  Google Scholar 

  62. Yang, L., Liu, Z., Lu, F., Dong, A. & Huang, H. SERRATE is a novel nuclear regulator in primary microRNA processing in Arabidopsis. The Plant J. 47, 841–850 (2006).

    CAS  PubMed  Google Scholar 

  63. Robert, S., Zouhar, J., Carter, C. & Raikhel, N. Isolation of intact vacuoles from Arabidopsis rosette leaf-derived protoplasts. Nat. Protoc. 2, 259–262 (2007).

    CAS  PubMed  Google Scholar 

  64. Kelley, L. A., Mezulis, S., Yates, C. M., Wass, M. N. & Sternberg, M. J. The Phyre2 web portal for protein modeling, prediction and analysis. Nat. Protoc. 10, 845–858 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D 66, 486–501 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Gadella, T. W. Jr., van der Krogt, G. N. & Bisseling, T. GFP-based FRET microscopy in living plant cells. Trends Plant Sci. 4, 287–291 (1999).

    PubMed  Google Scholar 

  67. Hwang, W. L., Deindl, S., Harada, B. T. & Zhuang, X. Histone H4 tail mediates allosteric regulation of nucleosome remodelling by linker DNA. Nature 512, 213–217 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Yang, H., Lu, P., Wang, Y. & Ma, H. The transcriptome landscape of Arabidopsis male meiocytes from high-throughput sequencing: the complexity and evolution of the meiotic process. Plant J. 65, 503–516 (2011).

    CAS  PubMed  Google Scholar 

  69. Du, Z., Zhou, X., Ling, Y., Zhang, Z. & Su, Z. AgriGO: a GO analysis toolkit for the agricultural community. Nucleic Acids Res. 38, 64–70 (2010).

    Google Scholar 

  70. Wang, Y. et al. The DNA replication factor RFC1 is required for interference-sensitive meiotic crossovers in Arabidopsis thaliana. PLoS Genet. 8, e1003039 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Saleh, A., Alvarez-Venegas, R. & Avramova, Z. An efficient chromatin immunoprecipitation (ChIP) protocol for studying histone modifications in Arabidopsis plants. Nat. Protoc. 3, 1018–1025 (2008).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank staff at the ABRC at Ohio State University for providing the Arabidopsis mutant seeds; J. Gu and Q. Yao at Fudan University for sharing experimental technology; and A. Dong at Fudan University for discussions. This research was supported by grants from the National Natural Science Foundation of China (nos. 31925005 and 31870293) the State Key Laboratory of Genetic Engineering and Fudan University.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: Y.W., H.M. and J.M. Methodology: J.W., C.Y., S.Z., J.Y., H.D., H.W., J.H. and X.C. Investigation: J.W., C.Y., S.Z., J.Y., H.D., H.W. and J.H. Writing, original draft: Y.W. and J.W. Review and editing, H.M., Y.W. and J.M. Supervision: H.M., Y.W., J.M. and X.C.

Corresponding authors

Correspondence to Jinbiao Ma, Hong Ma or Yingxiang Wang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information: Nature Plants thanks Jose Gutierrez-Marcos and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Interaction assays of MMD1 and JMJ16.

a, Self-activation detection of full length MMD1, PHD domain (601–680aa) and MMD1ΔPHD domain (1–600aa) with the blank AD. The blue growth yeast stains in QDO/X-gal/ABA (QDO/A/g) indicate self-activation. 3 replicates were done with the similar results. b, The functional enrichment analysis of the isolated proteins by the MMD1ΔPHD as a bait. c, Detection of interaction of MMD1 with JMJ14, JMJ18 and JMJ19 in tobacco leaves using bimolecular fluorescence complementation (BiFC). DAPI staining represents the nucleus. The merged panel (right panel) is the overlay of DAPI (left, blue) and YFP (middle, green). 12 cells were observed in each experiment. d, Showing the divide fragments for JMJ16 and MMD1 for the subsequent examination. e, Y2H assay of MMD1 truncations and FYR-C domain in the QDO/ABA (QDO/A) plates. The growth yeast indicates a positive interaction. The experiment was repeated for 3 times with similar results. f, Pull-down assay shows the interactions between FYR-C domain (978–1209aa) and MMD1-9 truncations staining by Coomassie blue. g, The MMD-9 interacts with the FYR-C domain by GST-pull down staining by Coomassie blue. The interaction between full-length MMD1 and JMJ16 proteins is a positive control, and between FYR-C and GST serves as a negative control. Recombinant His tagged full-length JMJ16 was purified from insect cells. GST tagged truncated MMD-9 and full length MMD1, and His tagged FYR-C were expressed in and purified from E. coli. The experiments were repeated three times. h, The core conserved domain in the MMD1ΔPHD regions and its homologues in plants was named the MMD domain. Alignment of MMD domain in land plants shows the conserved MMD domain highlighted in the red box. i, Comparison of FYR-C domain highlighted in green box in the PKDM7 family shows the similarity and diversity of FYR-C domains.

Source data

Extended Data Fig. 2 The co-localization of MMD1 and JMJ16 in male meiocytes, and purification of JMJ16 and MMD1 proteins.

a, MMD1 and JMJ16 partially co-localize in the pachytene chromosome of meiocytes isolated from transgenic plants JMJ16 fused HA driven by a native promoter. DAPI represents chromosome, green signals represent JMJ16 and red signals represent MMD1. Merged panel in the row 4 is the overlay of MMD1 and JMJ16. The co-localization regions of JMJ16 and MMD1 show color in yellow. JMJ16 and MMD1 are marked by HA and MMD1 primary antibodies and Alex Fluor 480 or 555 secondary antibody, respectively. Scale bar = 5 μm. b, The quantification of co-localization ratio of MMD1 and JMJ16 in by Image J in a (7 and 10 biologically independent leptotene and pachytene cells were used, respectively). The data was represented by mean ± SD and P-value (Pachytene/Leptotene) = 6.71 × 10−7. c, The HA-IP-purified JMJ16 protein from the JMJ16-YFP-HA transgenic Arabidopsis leaves was verified by western blot with anti-GFP antibody. d, His-purified full-length JMJ16- from insect cells (SF9) was stained by Coomassie (left) and verified by Western blot with anti-His antibody (right). e, MMD1-GST purified from E. coli (left panel) and after its cleavage (middle panel) was stained by Coomassie blue staining and verified by western blot with anti-MMD1 antibody (right panel). The Western blotting or Coomassie blue staining in each experiment was repeated for 3 times.

Source data

Extended Data Fig. 3 Immunostaining of specific histone modification in WT, mmd1 and jmj16 male meiocytes, and methylation assay of specific histone modification in WT, mmd1 and jmj16 inflorescences by WB.

ag, Immunostaining of specific histone modification (CENH3, H3K4me1, H3K4me2, H3K9me1, H3K9me2, H3K27me1 and H3K9Ac) on pachytene chromosome in WT mmd1 and jmj16. DAPI was observed in 340 nm and methylated histones were recorded in 555 nm. The merged panel (bottom) is the overlay of DAPI (above, blue) and methylated histones (middle, red). The sample sizes for each experiment were shown in Fig. 3b. h, i, H3 methylation levels in inflorescences by western blot (h) and their quantification in WT, mmd1 and jmj16 (i). Scale bar = 5μm. P-value (jmj16/WT) = 0.0369, * P < 0.05, ** P < 0.01, two-tailed Student’s t test with two-sided. Data are represented as mean ± SEM from three independent experiments.

Source data

Extended Data Fig. 4 Interaction examination of JMJ16-N and -C termini, and structural modeling of JMJ16 catalytic domain with zinc-finger and helical domain.

a, Illustration of FRET receptor bleaching in tobacco. The single-direction black arrow represents energy transfer in the top figure. We hypothesize that the closed distance or interaction between the JMJ16 N- and C-termini would result in energy transfer from CFP (donor) to YFP (receptor). Therefore, in the basal state, when YFP is inactivated using photo-bleaching, the energy will remain at CFP. If MMD1 can disrupt the self-interaction of JMJ16 in the bottom part, YFP should be always undetectable if the laser is only given to CFP. CFP will remain unchanged with the photo-bleaching of YFP. b, As the control to Fig. 4i, the laser was given to both CFP and YFP. Then, the YFP was inactivated by photo-bleaching, which demonstrates the normal function of the YFP protein in the presence of MMD1, and the photo-bleaching was also able to reduce the YFP activity. Four biological independent cells were used in the statistics analysis and the data was represented by mean ± SEM. c, The structure model of JMJ16 [catalytic domain Jumonji with C5HC2 zinc-finger and helical domain (HD)]. The Jumonji domain is colored in green, while the HD plus C5HC2-Zinc finger part is in marine. df, Structural comparison of JMJ16 model and peptide-bound Jumonji domains. d, JMJ14-H3K4me3 complex (PDB code: 5YKO): The Jumonji domain and HD -C5HC2 domain are colored in violet and yellow, respectively. e, Human JMJD2A-H3K9me3 complex (PDB code: 2OQ6): The Jumonji domain and the extended helixes region (294-355aa) are colored in wheat and gray, respectively. f, Mouse JMJD3-H3K27me3 complex (PDB code: 4EZH): The Jumonji domain and the C-helix and GATAL zinc-finger are colored in light blue and light pink, respectively. JMJ16 is colored in the same scheme as in Supplementary Fig. 4c. The H3 peptides are shown in space-filling representation.

Extended Data Fig. 5 Analyses of the target genes regulated by MMD1 and JMJ16.

a, The histogram shows up and down-expressed genes regulated by JMJ16 in meiocytes. b, Venn diagram shows genes co-regulated by both MMD1 and JMJ16. c, Validation of JMJ16 expression by anti-HA antibody in JMJ16-HA transgenic plants driven with native promoter. The experiment was repeated for 3 times with similar results. d, Analysis of condensin subunit gene expression in leaves of WT and jmj16. Genes with a reduction in jmj16 meiocytes as shown in Fig. 6b exhibit elevated expression or no significant alternation in leaves. P-values are 0.00686 for CAPE1, 1.91 × 10−4 for CAPE2, 9.36 × 10−4 for CAPG, 1.43 × 10−4 for CAPG2, 0.033 for CAPH and 0.031 for CAPC. e, f, Selected genes such as FLC show significantly increased expression in jmj16 leaves (e), but no significant changes in jmj16 meiocytes (f). The SMC3 gene is a control. P-values are 8.35 × 10−5 for FLC, 0.00144 for COL1, 0.0202 for UNE15, 0.021 for PPME1, 0.011 for PRF5, 0.013 for GRP16), 1.27 × 10−3 for IAA2, 9.37 × 10−4 for IAA7, 2.79 × 10−4 for IAA19, 0.044 for IAA30, 1.45 × 10−3 for AT3G38790, 0.0133 for AT3G28840, 0.0103 for AT3G28980 and 0.016 for AT3G28830 in Extended Data Fig. 5e; P-value (SMC3) = 0.0098 in Extended Data Fig. 5f. g, h, Detection of H3K4me3 (g) and H3K9me3 (h) occupancy in condensin genes in inflorescences of WT and mmd1. P-value (CAPH-P) = 1.94 × 10−4. * P < 0.05, ** P < 0.01, two-tailed Student’s t test with two-sided. Data are represented as mean ± SEM from three independent experiments.

Source data

Extended Data Fig. 6 The Arabidopsis jmj16 mutant shows defects in fertility and meiotic chromosome condensation.

a, Illustration of the gene, protein organizations and T-DNA insertional positions of JMJ16. b, The JMJ16 expression level was analyzed by RT-qPCR in WT and jmj16 mutants. P1-4 represents primers’ position on JMJ16 marked in a. Data are represented as mean ± SEM from three independent experiments. c, d, Morphology of fertility was examined and homozygous mutants show partial sterility with abnormally short siliques, scale bar = 5 cm and inviable pollen grains (red pollens are alive and dark/green pollens are dead) by Alexzander staining of anthers, scale bar = 10μm. Independent plants for WT (n = 45), jmj16-1(n = 40), jmj16-2 (n = 25) and JMJ16 complementation transgenic lines (n = 20) were used with similar results. Twenty independent anthers for each genotype were used with similar results. e, Genetic analyses of meiotic chromosome condensation defects in jmj16, mmd1 and mmd1 jmj16 double mutants. Diplotene cells for WT (n = 21), mmd1 (n = 20), jmj16-1 (n = 25) and mmd1 jmj16-1 (n = 30) were used with similar results. Diakinesis cells for WT (n = 25), mmd1 (n = 30), jmj16-1 (n = 31) and mmd1 jmj16-1 (n = 20) were used with similar results; Metaphase I cells for WT (n = 20), mmd1 (n = 24), jmj16-1 (n = 30) and mmd1 jmj16-1 (n = 20) were used with similar results. f, The estimation of metaphase I chromatin areas (circled by yellow lines) in WT (n = 17), mmd1 (n = 11), jmj16-1 (n = 12) and mmd1 jmj16-1 double mutants (n = 16). Data are represented as mean ± SEM. Scale bar = 5μm. P-values are 1.34 × 10−13 (mmd1/ WT), 1.26 × 10−8 (jmj16-1/WT), 5.65 × 10−15 (mmd1 jmj16-1/WT), 1.18 × 10−4 (mmd1/jmj16-1), 1.88 × 10−6 (jmj16-1/mmd1 jmj16-1) and 0.296 (mmd1/mmd1 jmj16-1). * P < 0.05, ** P < 0.01, two-tailed Student’s t test.

Source data

Extended Data Fig. 7 The expression of synthetic JMJ16-N with MMD PHD domain partially recues meiotic chromosome condensation defect in mmd1 in vivo.

a, Illustration of synthetic constructs between the PHD domain of MMD1 (PHDMMD1) and the JMJ16-N without (Construct A) or with (Construct B) ZF (zinc finger domain). b, The expression level of JMJ16 in transgenic plant meiocytes was analyzed by RT-qPCR using the primers in the JJMJ16-N. The data are represented by mean ± SEM by three replicates with similar results. P-values are 1.51 × 10−4 (Construct A-1/WT), 1.19 × 10−5 (Construct A-2/WT), 4.54 × 10−4 (Construct B-1/WT) and 4.39 × 10−5 (Construct B-2/WT). c, Meiotic prophase chromosome behaviors of WT, mmd1 and constructs A and B transgenic plants are stained by DAPI in zygotene, pachytene, diakinesis and anaphase II, respectively. Zygotene cells for WT (n = 20), mmd1 (n = 20), Construct A-1 (n = 22), Construct A-2 (n = 21), Construct B-1 (n = 21) and Construct B-2 (n = 20) were used with similar results; Pachytene cells for WT (n = 15), mmd1 (n = 14), Construct A-1 (n = 14), Construct A-2 (n = 10), Construct B-1 (n = 12) and Construct B-2 (n = 20) were used with similar results; Diakinesis cells for WT (n = 20), mmd1 (n = 21), Construct A-1 (n = 20), Construct A-2 (n = 15), Construct B-1 (n = 21) and Construct B-2 (n = 19) were used with similar results; Anaphase II cells for WT (n = 20), mmd1 (n = 24), Construct A-1 (n = 22), Construct A-2 (n = 20), Construct B-1 (n = 12) and Construct B-2 (n = 20) were used with similar results. d, Estimation of chromatin areas of metaphase I chromosome in male meiocytes in Fig. 6i. The areas of chromatins in construct A and construct B were defined by circle drawing tools and area detection by Image J software and the number of cells is labeled in the column. We used cm2 as the area unit that is calculated by the pictures enlarged by the microscope in the centi-meter level, however, the real size of chromatins is micro-meter. P-values are 3.17 ×10−9 (mmd1/WT), 7.7 × 10−11 (Construct B-1/WT) and 5.33 × 10−19 (Construct B-2/WT). The data are represented as mean ± SD. e, RT-qPCR analyses of the CAP-D3 expression in meiocytes of WT, mmd1 and transgenic lines. The data are represented as mean ± SEM with three biological independent replicates. P-values are 3.09 ×10−4 (mmd1/WT), 7.37 × 10−5 (Construct B-1/WT) and 7.74 × 10−7 (Construct B-2/WT). f, The binding affinity of construct A and B. Well 1 and 2 are controls from WT to show H3K4me3 and H3K9me3. FLAG IP was conducted in WT (Well 3), A (Well 4) and B (Well 5) with anti-FLAG. The IP lysates were detected by H3K4me3, H3K9me3 and FLAG antibodies. Construct B only binds H3K4me3 and Construct A binds to both H3K9me3 and H3K4me3. Two replicates for Western blotting were performed. gi, The complementation of full length MMD1 and JMJ16 in mmd1 mutants. ACTIN::MMD1-FLAG and ACTIN::JMJ16-HA are transformed into mmd1 heterozygotes. g, In the offspring of mmd1 homozygotes, MMD1 and JMJ16 were detected by MMD1 and HA antibodies. h, The siliques used for WT (n = 120), mmd1 (n = 145) and JMJ16 + MMD1(mmd1) (n = 168). i, Meiotic metaphase I chromosome phenotypes used for WT (n = 24), mmd1 (n = 20) and JMJ16 + MMD1(mmd1) (n = 27). Scale bar = 5μm. * P < 0.05, ** P < 0.01, two-tailed Student’s t test.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1–3. Supplementary Table 1: summary of JmjC demethylases towards multiple substrate specificities in plants and animals. JmjC-containing demethylates in plants and animals showed multiple demethylation activities for different histone modifications. Supplementary Table 2: peptides used in this study. Peptides used in the MALDI–TOF-MS and FP are listed in this table with amino-acid sequences, catalogue numbers and providers. Supplementary Table 3: primers used in this study. Primer names, sequences and applications were noted in this table.

Reporting Summary

Supplementary Data

Genes with downregulated and upregulated expression in jmj16 meiocytes. Sheet 1: 1,633 upregulated genes in jmj16 male meiocytes (log2[fdc] ≥ 1). Sheet 2: 4,626 downregulated genes in jmj16 male meiocytes (log2[fdc] ≤ −1). Sheet 3: 543 coregulated genes with upregulated expression in mmd1 (stage 4–7 anthers) (log2[fdc] ≥ 1) and jmj16 male meiocytes (log2[fdc] ≥ 1). Sheet 4: 802 coregulated genes with downregulated expression in mmd1 (stage 4–7 anthers) (log2[fdc] ≤ −1) and jmj16 male meiocytes (log2[fdc] ≤ −1). Sheet 5: biological processes annotation in upregulated genes or downregulated genes in jmj16 meiocytes. The transcriptome data were obtained from two replicates. For each replicate, about 20,000 male meiocytes were isolated from anthers (see Methods). The statistical significance (P value) of the differential expression of genes was calculated using a two-tailed Student’s t-test; FDR-adjusted P ≤ 0.05 was considered to be significant. The GO enrichment analysis was conducted by AgriGO (http://bioinfo.cau.edu.cn/agriGO/analysis.php), and statistics were compiled by FDR correction; FDR-adjusted P ≤ 0.05 was considered to be significant.

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed gels.

Source Data Extended Data Fig. 2

Unprocessed western blots and gels.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 7

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, J., Yu, C., Zhang, S. et al. Cell-type-dependent histone demethylase specificity promotes meiotic chromosome condensation in Arabidopsis. Nat. Plants 6, 823–837 (2020). https://doi.org/10.1038/s41477-020-0697-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41477-020-0697-0

  • Springer Nature Limited

This article is cited by

Navigation