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:

MRG15 orchestrates rhythmic epigenomic remodelling and controls hepatic lipid metabolism

Abstract

The rhythmic regulation of transcriptional processes is intimately linked to lipid homeostasis, to anticipate daily changes in energy access. The Rev-erbα–HDAC3 complex was previously discovered to execute the rhythmic repression of lipid genes; however, the epigenetic switch that turns on these genes is less clear. Here, we show that genomic recruitment of MRG15, which is encoded by the mortality factor on chromosome 4 (MORF4)-related gene on chromosome 15, displays a significant diurnal rhythm and activates lipid genes in the mouse liver. RNA polymerase II (Pol II) recruitment and histone acetylation correspond to MRG15 binding, and the rhythm is impaired upon MRG15 depletion, establishing MRG15 as a key modulator in global rhythmic transcriptional regulation. MRG15 interacts with the nuclear receptor LRH-1, rather than with known core clock proteins, and is recruited to genomic loci near lipid genes via LRH-1. Blocking of MRG15 by CRISPR targeting or by the FDA-approved drug argatroban, which is an antagonist to MRG15, attenuates liver steatosis. This work highlights MRG15 as a targetable master regulator in the rhythmic regulation of hepatic lipid metabolism.

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: Rhythmic genomic recruitment of MRG15 in mouse liver.
Fig. 2: MRG15 regulates rhythmic RNA polymerase II recruitment in mouse liver.
Fig. 3: MRG15 regulates rhythmic histone acetylation and gene expression in mouse liver.
Fig. 4: LRH-1 mediates MRG15 recruitment to lipid regulatory genes.
Fig. 5: LRH-1 interacts with MRG15.
Fig. 6: Blocking of liver MRG15 by CRISPR targeting attenuates liver steatosis and improves metabolism.
Fig. 7: Treatment with the FDA-approved drug argatroban attenuates liver steatosis.
Fig. 8: Mechanistic model of the role of MRG15 in regulating liver lipid metabolism.

Similar content being viewed by others

Data availability

ChIP–seq and RNA-seq data have been deposited in the National Omics Data Encylopedia (http://www.biosino.org/node) under the accession code OEP000757.

References

  1. Zhang, R., Lahens, N. F., Ballance, H. I., Hughes, M. E. & Hogenesch, J. B. A circadian gene expression atlas in mammals: implications for biology and medicine. Proc. Natl Acad. Sci. US 111, 16219–16224 (2014).

    Article  CAS  Google Scholar 

  2. Takahashi, J. S. Transcriptional architecture of the mammalian circadian clock. Nat. Rev. Genet. 18, 164–179 (2017).

    Article  CAS  PubMed  Google Scholar 

  3. Feng, D. & Lazar, M. A. Clocks, metabolism, and the epigenome. Mol. Cell. 47, 158–167 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Tao, W. et al. SWItch/sucrose nonfermentable (SWI/SNF) complex subunit BAF60a integrates hepatic circadian clock and energy metabolism. Hepatology 54, 1410–1420 (2011).

    Article  CAS  PubMed  Google Scholar 

  5. Takahata, S. et al. Transactivation mechanisms of mouse clock transcription factors, mClock and mArnt3. Genes Cells 5, 739–747 (2000).

    Article  CAS  PubMed  Google Scholar 

  6. Etchegaray, J. P., Lee, C., Wade, P. A. & Reppert, S. M. Rhythmic histone acetylation underlies transcription in the mammalian circadian clock. Nature 421, 177–182 (2003).

    Article  CAS  PubMed  Google Scholar 

  7. Asher, G. et al. SIRT1 regulates circadian clock gene expression through PER2 deacetylation. Cell 134, 317–328 (2008).

    Article  CAS  PubMed  Google Scholar 

  8. Nakahata, Y. et al. The NAD+-dependent deacetylase SIRT1 modulates CLOCK-mediated chromatin remodeling and circadian control. Cell 134, 329–340 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. DiTacchio, L. et al. Histone lysine demethylase JARID1a activates CLOCK-BMAL1 and influences the circadian clock. Science 333, 1881–1885 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Doi, M., Hirayama, J. & Sassone-Corsi, P. Circadian regulator CLOCK is a histone acetyltransferase. Cell 125, 497–508 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Nader, N., Chrousos, G. P. & Kino, T. Circadian rhythm transcription factor CLOCK regulates the transcriptional activity of the glucocorticoid receptor by acetylating its hinge region lysine cluster: potential physiological implications. FASEB J. 23, 1572–1583 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Feng, D. et al. A circadian rhythm orchestrated by histone deacetylase 3 controls hepatic lipid metabolism. Science 331, 1315–1319 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Zhang, Y. et al. Discrete functions of nuclear receptor Rev-erbα couple metabolism to the clock. Science 348, 1488–1492 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kim, Y. H. et al. Rev-erbα dynamically modulates chromatin looping to control circadian gene transcription. Science 359, 1274–1277 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Bertram, M. J. & Pereira-Smith, O. M. Conservation of the MORF4 related gene family: identification of a new chromo domain subfamily and novel protein motif. Gene 266, 111–121 (2001).

    Article  CAS  PubMed  Google Scholar 

  16. Chen, M., Pereira-Smith, O. M. & Tominaga, K. Loss of the chromatin regulator MRG15 limits neural stem/progenitor cell proliferation via increased expression of the p21 Cdk inhibitor. Stem Cell Res. 7, 75–88 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Chen, M., Takano-Maruyama, M., Pereira-Smith, O. M., Gaufo, G. O. & Tominaga, K. MRG15, a component of HAT and HDAC complexes, is essential for proliferation and differentiation of neural precursor cells. J. Neurosci. Res. 87, 1522–1531 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Garcia, S. N., Kirtane, B. M., Podlutsky, A. J., Pereira-Smith, O. M. & Tominaga, K. Mrg15 null and heterozygous mouse embryonic fibroblasts exhibit DNA-repair defects post exposure to gamma ionizing radiation. FEBS Lett. 581, 5275–5281 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Huang, C. et al. Mrg15 stimulates Ash1 H3K36 methyltransferase activity and facilitates Ash1 Trithorax group protein function in Drosophila. Nat. Commun. 8, 1649 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Iwamori, N. et al. MRG15 is required for pre-mRNA splicing and spermatogenesis. Proc. Natl Acad. Sci. USA 113, E5408–E5415 (2016).

    Article  CAS  PubMed  Google Scholar 

  21. Tominaga, K. et al. MRG15 regulates embryonic development and cell proliferation. Mol. Cell. Biol. 25, 2924–2937 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Pardo, P. S., Leung, J. K., Lucchesi, J. C. & Pereira-Smith, O. M. MRG15, a novel chromodomain protein, is present in two distinct multiprotein complexes involved in transcriptional activation. J. Biol. Chem. 277, 50860–50866 (2002).

    Article  CAS  PubMed  Google Scholar 

  23. Yochum, G. S. & Ayer, D. E. Role for the mortality factors MORF4, MRGX, and MRG15 in transcriptional repression via associations with Pf1, mSin3A, and Transducin-Like Enhancer of Split. Mol. Cell. Biol. 22, 7868–7876 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Cabianca, D. S. et al. Active chromatin marks drive spatial sequestration of heterochromatin in C. elegans nuclei. Nature 569, 734–739 (2019).

    Article  CAS  PubMed  Google Scholar 

  25. The Coronary Artery Disease (C4D) Genetics Consortium A genome-wide association study in Europeans and South Asians identifies five new loci for coronary artery disease. Nat. Genet. 43, 339–344 (2011).

  26. Reilly, M. P. et al. Identification of ADAMTS7 as a novel locus for coronary atherosclerosis and association of ABO with myocardial infarction in the presence of coronary atherosclerosis: two genome-wide association studies. Lancet 377, 383–392 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Schunkert, H. et al. Large-scale association analysis identifies 13 new susceptibility loci for coronary artery disease. Nat. Genet. 43, 333–338 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Bauer, R. C. et al. Knockout of Adamts7, a novel coronary artery disease locus in humans, reduces atherosclerosis in mice. Circulation 131, 1202–1213 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kessler, T. et al. ADAMTS-7 inhibits re-endothelialization of injured arteries and promotes vascular remodeling through cleavage of thrombospondin-1. Circulation 131, 1191–1201 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Koike, N. et al. Transcriptional architecture and chromatin landscape of the core circadian clock in mammals. Science 338, 349–354 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Le Martelot, G. et al. Genome-wide RNA polymerase II profiles and RNA accumulation reveal kinetics of transcription and associated epigenetic changes during diurnal cycles. PLoS Biol. 10, e1001442 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Zhu, J., Li, C., Gong, C. & Li, X. Regulation of Pol II pausing is involved in daily gene transcription in the mouse liver. J. Biol. Rhythms 33, 350–362 (2018).

    Article  PubMed  CAS  Google Scholar 

  33. Platt, R. J. et al. CRISPR–Cas9 knockin mice for genome editing and cancer modeling. Cell 159, 440–455 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Pena, A. N., Tominaga, K. & Pereira-Smith, O. M. MRG15 activates the cdc2 promoter via histone acetylation in human cells. Exp. Cell Res. 317, 1534–1540 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Sun, Z. et al. Hepatic Hdac3 promotes gluconeogenesis by repressing lipid synthesis and sequestration. Nat. Med. 18, 934–942 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ueda, H. R. et al. System-level identification of transcriptional circuits underlying mammalian circadian clocks. Nat. Genet. 37, 187–192 (2005).

    Article  CAS  PubMed  Google Scholar 

  37. Miranda, D. A. et al. LRH-1 regulates hepatic lipid homeostasis and maintains arachidonoyl phospholipid pools critical for phospholipid diversity. JCI Insight 8, e96151 (2018).

    Article  Google Scholar 

  38. Oosterveer, M. H. et al. LRH-1-dependent glucose sensing determines intermediary metabolism in liver. J. Clin. Invest. 122, 2817–2826 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Stein, S. et al. Impaired SUMOylation of nuclear receptor LRH-1 promotes nonalcoholic fatty liver disease. J. Clin. Invest. 127, 583–592 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Stein, S. & Schoonjans, K. Molecular basis for the regulation of the nuclear receptor LRH-1. Curr. Opin Cell Biol. 33, 26–34 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. Zou, C. et al. Mortality factor 4 like 1 protein mediates epithelial cell death in a mouse model of pneumonia. Sci. Transl Med. 7, 311ra171 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Krylova, I. N. et al. Structural analyses reveal phosphatidyl inositols as ligands for the NR5 orphan receptors SF-1 and LRH-1. Cell 120, 343–355 (2005).

    Article  CAS  PubMed  Google Scholar 

  43. Ortlund, E. A. et al. Modulation of human nuclear receptor LRH-1 activity by phospholipids and SHP. Nat. Struct. Mol. Biol. 12, 357–363 (2005).

    Article  CAS  PubMed  Google Scholar 

  44. Wang, W. et al. The crystal structures of human steroidogenic factor-1 and liver receptor homologue-1. Proc. Natl Acad. Sci. USA 102, 7505–7510 (2005).

    Article  CAS  PubMed  Google Scholar 

  45. Li, S. et al. Genetic and chemical screenings identify HDAC3 as a key regulator in hepatic differentiation of human pluripotent stem cells. Stem Cell Reports 11, 22–31 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank K. Musunuru and Y. Chen for helpful discussions. We thank H. Feng and X.-F. Chen for assistance with data analysis, and Z.-G. Li for assistance with confocal analysis. This work was supported by grants from the National Key R&D Programme of China (2017YFA0102800 and 2017YFA0103700), the Strategic Priority Research Programme of the Chinese Academy of Sciences (XDA16030402), the National Natural Science Foundation of China (91957205, 31670829 and 31971063) and the China Postdoctoral Science Foundation (2019M661661).

Author information

Authors and Affiliations

Authors

Contributions

Y.W., C.T., Y.Z. and Q.D. designed experiments. Y.W., C.T., Y.Z., X.L., F.L., S.L., Y.C., Y.Q., Z.F., L.C, T.Z., X.R., C.F. and Y.L. carried out experiments and analysed data. X.L. performed bioinformatics analysis. Y.W., C.T., Y.Z., F.L., W.Y., H.Y. and Q.D. wrote the manuscript. Q.D. supervised the project.

Corresponding author

Correspondence to Qiurong Ding.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Primary Handling Editor: Pooja Jha.

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

Extended data

Extended Data Fig. 1 Rhythmic genomic recruitment of exogenous MRG15 in mouse liver.

Adeno-associated virus 8 (AAV8) vectors either containing TBG-luciferase (AAV-Luci) or TBG-FLAG-Mrg15 (AAV-FLAG-MRG15) were administered via tail vein injection. Tissues were collected for analysis after virus injection for around 4 weeks. a, Western analysis of endogenous and FLAG-MRG15 levels in mouse liver tissues. Repeated twice with similar results. b, Average MRG15 signals from −5 kb to +5 kb surrounding the TSS and from −2 kb to +2 kb surrounding the gene bodies. c, Distribution of the MRG15 ZT22 binding sites in different genomic locations relative to known genes. d. ChIP–seq tracks of MRG15 signals at ZT10 and ZT22 in Acat2, Acly and Elovl5 gene loci, as indicated. e, Gene enrichment analysis of genes both bound by MRG15 as revealed in ChIP–seq analysis and displayed circadian rhythm at transcriptional levels (603 genes). Associated P values were determined according to the analysis in the GO database (Fisher’s exact P value).

Source data

Extended Data Fig. 2 Rhythmic genomic recruitment of endogenous MRG15 in mouse liver in Mrg15Flag-KI animals.

a, Scheme illustration of the generation of the Mrg15Flag-KI animals via knockin of a 3 × FLAG tag at the C-terminal of endogenous Mrg15 before the stop codon. b, Western analysis of the endogenous FLAG-MRG15 with both FLAG and MRG15 antibodies. Repeated twice with similar results. c, Relative MRG15 binding signals as assessed by ChIP–qPCR in animals at ZT10 and ZT22. n = 3 for Wild-type and 6 for Mrg15Flag-KI groups. d, The rhythm of MRG15 recruitment is reversed by daytime feeding. n = 3 for Wild-type, 4 for Mrg15Flag-KI ZT10 and 3 for Mrg15Flag-KI ZT22. Some of the values in the wild-type group are very low and were not detected by qPCR, therefore are not displayed. Data points were all from biologically independent samples (c, d). P values are shown for indicated comparisons by the two-tailed Mann–Whitney U tests. Values are mean ± s.e.m.

Source data

Extended Data Fig. 3 Liver-specific depletion of MRG15 via CRISPR technology.

a, Schematic illustration of the CRISPR–Cas9 KI mice and AAV constructs used to deplete genes in this study. b, In vivo bioluminescence imaging of AAV vectors (left) and endogenous liver MRG15 levels in animals administered AAV-Cre-2a-Luci (Control) or AAV-Cre-2a-Luci-Mrg15 sgRNA1 (Mrg15-sgRNA). Repeated twice with similar results.

Source data

Extended Data Fig. 4 MRG15 depletion in liver leads to reduced expression of lipid and cholesterol synthesis genes in NCD-fed animals.

Tissues were collected from animals under NCD after virus injection for 14 weeks for RNA-seq analysis. a, b, Representative genes downregulated in CRISPR-Mrg15 livers involved in triglyceride synthesis (a) and cholesterol synthesis (b). Values are indicated as log 2 [Fold change (CRISPR-Mrg15/Control)] in a and b. Genes that are direct targets of MRG15 as revealed in ChIP–seq analysis are labelled with an asterisk at upper right.

Extended Data Fig. 5 MRG15 binding sites show minimal overlap with binding loci of HDAC3, Rev-erbα or NCoR, at commonly bound genes.

Heatmap displaying localisation of ChIP signals relative to the TSS site (0 bp) near common target genes between MRG15 vs. HDAC3 a, MRG15 vs. Rev-erbα b, and MRG15 vs. NCoR c. The numbers of common target genes and overlapped peaks are indicated below, with common genes (peaks) showing in blue, and total bound genes (peaks) of HDAC3, Rev-erbα and NCoR are shown in black. Detailed information can also be found in Supplementary Table 1.

Extended Data Fig. 6 LRH-1 and MRG15 co-regulate lipid synthesis genes.

a, Venn diagram indicating overlapping genes in Lrh1/ and CRISPR-Mrg15 livers (left); and gene enrichment analysis of overlapped genes downregulated in Lrh1/ and CRISPR-Mrg15 livers (120 genes). b, Venn diagram indicating overlapping genes in LRH-1 K289R and CRISPR-Mrg15 livers (left); and gene enrichment analysis of overlapped genes upregulated in LRH-1 K289R livers and downregulated in CRISPR-Mrg15 livers (161 genes). Associated P values were determined according to the analysis in the GO database (Fisher’s exact P value) for a and b. c, Representative commonly regulating lipid synthesis genes between Lrh1−/− and CRISPR-Mrg15 livers, and LRH-1 K289R and CRISPR-Mrg15 livers.

Extended Data Fig. 7 mRNA expression levels of Mrg15 and Lrh-1 at ZT10 and ZT22.

Relative mRNA expression of Mrg15 and Lrh-1 in liver at ZT10 and ZT22. n = 5 biologically independent samples. P values are shown for indicated comparisons by the two-tailed Mann–Whitney U tests. Values are mean ± s.e.m.

Source data

Extended Data Fig. 8 Liver depletion of MRG15 leads to reduced expression of lipid and cholesterol synthesis genes, and inflammatory genes in HFD animals.

Tissues were collected from animals under HFD after virus injection for 14 weeks for RNA-seq analysis. a, b, c, Representative genes downregulated in CRISPR-Mrg15 livers involved in triglyceride synthesis (a), cholesterol synthesis (b) and inflammatory response (c). Values are indicated as log 2 [Fold change (CRISPR-Mrg15/Control)] in a, b and c. Genes that are direct targets of MRG15 as revealed in ChIP–seq analysis in a and b are labelled with an asterisk at upper right. d, Gene enrichment analysis of downregulated genes in CRISPR-Mrg15 livers identified by RNA-seq (622 genes using a cut-off of P < 0.05). Associated P values were determined according to the analysis in the GO database (Fisher’s exact P value). e, Relative mRNA expression of representative lipid genes in liver in control and Mrg15-sgRNA treated animals under HFD. n = 9 biologically independent samples. P values are shown for indicated comparisons by the two-tailed Mann–Whitney U tests. Values are mean ± s.e.m.

Source data

Extended Data Fig. 9 CRISPR-Mrg15-sgRNA2 mice have improved metabolism.

a, Mrg15 targeting sequences of two sgRNAs. b, Western analysis of endogenous MRG15 levels after CRISPR treatment. Repeated twice with similar results. c, Body weight. d, Nuclear magnetic resonance analysis of fat mass. e, Nuclear magnetic resonance analysis of lean mass. Start, before virus injection. End, 19 weeks after virus injection. n = 10 (a-e). f, Blood triglyceride levels and total cholesterol levels (1 month: n = 11 and 13; 2 months: n = 13 and 15). g, GTT and ITT of animals after virus injection for 9–10 weeks. n = 11 for Control and 13 for Mrg15-sgRNA 2. h, Western analysis of endogenous MRG15 and LRH-1 levels after CRISPR treatment. Repeated twice with similar results. Data points were all from biologically independent samples (c-g). P values are shown for indicated comparisons by the two-tailed Mann–Whitney U tests (f) or were determined by multiple t-tests with fewer assumptions (do not assume consistent s.d.) for GTT and ITT analyses (g). Values are mean ± s.e.m.

Source data

Extended Data Fig. 10 MRG15 regulates expression of several core clock genes.

Relative mRNA levels of core clock genes in animal livers over a 24-h cycle. n = 4 biologically independent samples. Values are mean ± s.e.m.

Source data

Supplementary information

Supplementary Information

Supplementary Figures 1–4 and Supplementary Table 1

Reporting Summary

Supplementary Table 1

Statistical Source Data Supplementary Fig. 3

Source data

Source Data Fig. 1

Statistical Source Data

Source Data Fig. 2

Statistical Source Data

Source Data Fig. 3

Statistical Source Data

Source Data Fig. 4

Statistical Source Data

Source Data Fig. 5

Statistical Source Data

Source Data Fig. 5

Unprocessed Western Blots

Source Data Fig. 6

Statistical Source Data

Source Data Fig. 7

Statistical Source Data

Source Data Fig. 7

Unprocessed Western Blots

Source Data Extended Data Fig. 1

Unprocessed Western Blots

Source Data Extended Data Fig. 2

Statistical Source Data

Source Data Extended Data Fig. 2

Unprocessed Western Blots

Source Data Extended Data Fig. 3

Unprocessed Western Blots

Source Data Extended Data Fig. 7

Statistical Source Data

Source Data Extended Data Fig. 8

Statistical Source Data

Source Data Extended Data Fig. 9

Statistical Source Data,

Source Data Extended Data Fig. 9

Unprocessed Western Blots

Source Data Extended Data Fig. 10

Statistical Source Data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wei, Y., Tian, C., Zhao, Y. et al. MRG15 orchestrates rhythmic epigenomic remodelling and controls hepatic lipid metabolism. Nat Metab 2, 447–460 (2020). https://doi.org/10.1038/s42255-020-0203-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-020-0203-z

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